Purpose:

Melanoma is a biologically heterogeneous disease composed of distinct clinicopathologic subtypes that frequently resist treatment. To explore the evolution of treatment resistance and metastasis, we used a combination of temporal and multilesional tumor sampling in conjunction with whole-exome sequencing of 110 tumors collected from 7 patients with cutaneous (n = 3), uveal (n = 2), and acral (n = 2) melanoma subtypes.

Experimental Design:

Primary tumors, metastases collected longitudinally, and autopsy tissues were interrogated. All but 1 patient died because of melanoma progression.

Results:

For each patient, we generated phylogenies and quantified the extent of genetic diversity among tumors, specifically among putative somatic alterations affecting therapeutic resistance.

Conclusions:

In 4 patients who received immunotherapy, we found 1–3 putative acquired and intrinsic resistance mechanisms coexisting in the same patient, including mechanisms that were shared by all tumors within each patient, suggesting that future therapies directed at overcoming intrinsic resistance mechanisms may be broadly effective.

Translational Relevance

Melanoma often resists therapy, creating a challenge for treating metastatic disease. We analyzed multilesion sequencing data from cutaneous, uveal, and acral melanomas and found that somatic mutations related to treatment resistance often occur across metastases. These findings have implications for developing more effective therapies targeting all lesions within a patient.

Tumor evolution begins with a single cell initiating a clonal expansion that can eventually lead to metastasis and, in some cases, therapeutic resistance. Melanoma exemplifies this evolutionary paradigm, originating with a normal melanocyte that transforms into a malignancy via molecular alterations (1). Melanoma subtypes differ in anatomic origin, pathogenesis (including canonical driver gene mutations) and therapeutic response (including immune checkpoint inhibitors and mutation-targeted therapies). Metastasis represents a lethal stage of melanoma across these subtypes. Although the treatment options for patients with metastatic melanoma have greatly improved because of recent therapeutic advances, patient outcomes differ widely due to treatment resistance associated with heterogeneity of driver gene mutations and treatment resistance mechanisms among histologic subtypes (2).

Acquired resistance occurs when a melanoma evolves in response to treatment. For example, some tumors develop somatic mutations (e.g., in NRAS or MEK) or amplifications that drive altered signaling in response to RAF kinase inhibitors (3, 4). In these patients, initial or mixed responses may be seen followed by progression months later. In contrast, intrinsic resistance occurs when genetic mutations that confer a high degree of fitness and hence drug tolerance are present in tumor cells prior to administration of therapy. In these cases, melanomas manifest early progression despite treatment (5). Mechanisms of melanoma intrinsic resistance are diverse and include somatic mutations in driver genes that current therapies do not directly target, such as NF1 and GNAQ mutations that mediate resistance to RAF inhibition, defects in immune signaling or antigen presentation, and a number of other biological factors (6–11). What is currently unknown is the extent to which acquired and intrinsic resistance mechanisms coexist in the same patient during metastatic progression, if they are shared across multiple metastases versus arise independently at each site, and if there are organ site–specific differences in acquired resistance. These issues represent an unmet clinical need to maximize the utility of immunotherapy and mutation-targeted therapies in patients with melanoma, among whom only approximately 50% derive long-term benefit from existing therapies. Furthermore, these therapies are more active against tumors of cutaneous versus acral or uveal origin, largely due to divergent genomic profiles among these distinct melanoma subtypes.

Studies have leveraged multilesional sequencing of primary tumors and metastases in cutaneous, uveal, and acral melanomas (12–22). However, in those studies, the number of sequenced metastases was generally <5 per patient, and the extent of genetic heterogeneity varied among the studies. Therefore, in a single study, we leveraged extended multilesional sampling of disseminated metastatic disease in 7 patients with metastatic melanoma coming to autopsy, representing cutaneous, acral, and uveal subtypes. Such an approach permits near “sampling to completion” to comprehensively define the genomic evolution of a patient's disease (23), thereby more accurately accounting for genetic heterogeneity of metastases across time and anatomic space.

Patient selection and tissue processing

This study was approved by the Institutional Review Boards of Johns Hopkins Medicine (Baltimore, MD) and Memorial Sloan Kettering Cancer Center (New York, NY), and was conducted in accordance with the Declaration of Helsinki. Informed written consent was obtained from each subject or each subject's guardian. Tumor samples and matched normal tissues were collected from 7 patients with pathologic confirmation of melanoma. Across these 7 patients, the therapeutic interventions received included surgery, immunotherapies, radiation, chemotherapies, and mutation-targeted therapies (Fig. 1; Supplementary Table S1). The timeline from primary diagnosis to death for patient MEL6 was 13 months, while the remaining 6 patients ranged 2–5 years. Tumor specimens collected premortem, including the primary melanoma lesion and metastases (formalin-fixed, paraffin-embedded; FFPE), were retrieved from pathology archives. Autopsies were conducted and tissues were collected within 2–44 hours after death. Following collection, each tumor and normal tissue was immediately frozen in liquid nitrogen and stored at −80°C, while a matching aliquot of each tissue was also preserved in 10% buffered formalin. Each frozen tissue sample was embedded in Tissue-Tek optimum cutting temperature compound and sectioned at 5–10 μm on a Leica Cryostat to create a hematoxylin and eosin–stained slide for review. Each FFPE sample was similarly sectioned using a Leica Microtome. Upon pathologic review, each tumor sample was macrodissected from sectioned slides to remove nonneoplastic components. The neoplastic component of each tumor underwent genomic DNA extraction using a Qiagen DNAeasy Blood and Tissue Kit and protocol, while the FFPE samples were extracted using a QIAamp DNA FFPE Tissue Kit and protocol.

Figure 1.

Clinical timelines and tumors collected for 7 patients with melanoma. The left side shows the legend with clinical events and samples collected. The right side shows the example timeline on top, followed by timelines for cutaneous melanomas MEL1, MEL2, and MEL3; uveal melanomas MEL4 and MEL5; and acral melanomas MEL6 and MEL7. Each timeline is shown from diagnosis to death (from left to right), with time measured in months. Each treatment is indicated where appropriate, with numbers indicating months since diagnosis (t = 0). The treatment types include immunotherapy (blue), mutation-targeted therapy (green: MEL5, cobimetinib; MEL6, dabrafenib + trametinib), chemotherapy (red), and radiation (orange). Clinically observed disease progression (+), defined by RECIST v1.1, is also annotated. Tumor collections are indicated with a gray box with a dashed outline. Premortem tumors are triangles above each timeline, while postmortem tumors are circles to the right of each timeline (i.e., after death). For premortem tumors, the number indicates when each tumor was collected. The anatomic site of each tumor is depicted by color: each primary tumor is indicated in black, each locoregional tumor in gray, and distant metastatic sites are colored by organ site (see Supplementary Table S1 for details). For MEL6′s treatments, the box on the right shows an enhanced view of months 5–13.

Figure 1.

Clinical timelines and tumors collected for 7 patients with melanoma. The left side shows the legend with clinical events and samples collected. The right side shows the example timeline on top, followed by timelines for cutaneous melanomas MEL1, MEL2, and MEL3; uveal melanomas MEL4 and MEL5; and acral melanomas MEL6 and MEL7. Each timeline is shown from diagnosis to death (from left to right), with time measured in months. Each treatment is indicated where appropriate, with numbers indicating months since diagnosis (t = 0). The treatment types include immunotherapy (blue), mutation-targeted therapy (green: MEL5, cobimetinib; MEL6, dabrafenib + trametinib), chemotherapy (red), and radiation (orange). Clinically observed disease progression (+), defined by RECIST v1.1, is also annotated. Tumor collections are indicated with a gray box with a dashed outline. Premortem tumors are triangles above each timeline, while postmortem tumors are circles to the right of each timeline (i.e., after death). For premortem tumors, the number indicates when each tumor was collected. The anatomic site of each tumor is depicted by color: each primary tumor is indicated in black, each locoregional tumor in gray, and distant metastatic sites are colored by organ site (see Supplementary Table S1 for details). For MEL6′s treatments, the box on the right shows an enhanced view of months 5–13.

Close modal

Whole-exome sequencing and alignment

DNA quantification, library preparation, whole-exome sequencing (WES), and bioinformatic analysis were performed by the Integrated Genomics Operation and the Bioinformatics Core at Memorial Sloan Kettering Cancer Center (New York, NY). After genomic DNA quantification using Qubit, sequencing libraries were created with the Agilent SureSelect XT Human All Exon V4 and WES was performed on a Illumina HiSeq 2500 in a 100 bp paired-end run using the Illumina HiSeq SBS Kit v4, to a target coverage of 150× for each tumor and 70× for each patient matched normal. The average depth of coverage achieved was 209× and 131× for the tumor and matched normal samples, respectively. Sequencing data were aligned to the hg19 reference human genome using Burrows-Wheeler Aligner (24), and read deduplication, recalibration, and sequence realignment were completed using the Picard toolkit and GATK (25).

Point mutations

For all 7 patients, after analysis with MuTect (ref. 26; v1.1.7) for single-nucleotide variants (SNV) and HaplotypeCaller (25) for small insertions and deletions (INDEL), we evaluated the putative somatic mutations to identify true positives while limiting false negatives, false positives, and sequencing artifacts. Each mutation required at least a 5% variant allele frequency with 20× coverage in at least one tumor sample, and less than 2% of the reads (or two reads total) of the matched normal sample with a coverage of 10×. We also removed passenger mutations found in multiple patients at >2% variant allele frequency or in ExAC at >0.04% (27). We restricted our analysis to missense, nonsense, frameshift INDELs, in frame INDELs, nonstop, silent, translation start site, and splice site mutations that affected <5 bp. In combination with manual review, across the 7 patients' tumors, this resulted in a total 4,680 unique somatic SNVs and INDELs.

Somatic copy-number alterations

Allele-specific somatic copy-number alterations (SCNA) were detected with FACETS (28). Allelic losses were defined as total copy number (tcn) = 0 and lower copy number (lcn) = 0, and gains were defined as tcn ≥ 10. For each tumor sample, we calculated the fraction of the genome altered (FGA) by somatic copy-number alterations (Supplementary Fig. S1). For the three cutaneous melanomas, the median FGA was 84%, although several tumor samples in MEL3 exhibited lower than 50%. For the two uveal melanomas, the median FGA was 64%. The two acral melanomas, MEL6 and MEL7, were categorically different from one another: MEL6 had a median FGA of 41%, whereas MEL7 had a median FGA of 100%. Supplementary Table S3 has the SCNAs we detected across the seven cases.

The FGA can also serve as a sequencing-based metric for tumor purity calculated by FACETS. From this analysis, three tumor samples indicated relatively low purities (less than 25% tumor cells): MEL2-M3, MEL3-M13, and MEL7-M4. These three tumors also exhibited the lowest SNV and INDEL burdens among the samples from each respective patient. As an orthogonal metric, we used Treeomics to estimate tumor purity for each sample using the SNV and INDEL data (29). These results agreed that the three tumor samples exhibited relatively low purities that were not due to differences in sequencing coverage, formalin fixation, or treatment status of the samples (Supplementary Fig. S1; Supplementary Table S1). Histologic review also showed low cellularity and viability across the samples. Given the concordance between the two metrics, and that low purity samples complicate phylogenetic inference, these three samples were removed from further analysis. For the remaining 107 tumor samples, the median purity estimates were 70% and 67% for the FACETS and Treeomics metrics, respectively.

Whole-genome duplication

For each tumor sample, whole-genome duplication (WGD) was assessed (Supplementary Fig. S1). WGD was called if the major copy number was greater than or equal to ≥ 2 and overall ploidy was ≥ 2.5, with > 50% of the autosomal genome affected.

Ultraviolet mutation signature

All SNVs with at least 5% variant allele frequency in the respective sample and a C>T transition at a dipyrimidine site (YC>T) were considered consistent with UV-induced DNA damage. In addition, mutation signatures were analyzed using Palimpsest (30).

Evolutionary analysis

We used Treeomics v. 1.7.4 to infer the phylogeny for each patient's tumor based on the SNVs and INDELs (29). In cases with more than 10 tumor samples, a consensus phylogeny was inferred by consolidating Treeomics phylogenies derived from sample subsets. We used Treeomics to quantify the Jaccard similarity coefficients for each intrapatient pair of tumor samples and to infer phylogenies after removing somatic mutations consistent with UV induced DNA damage [C>T transition at a dipyrimidine site (YC>T)].

Somatic mutation annotation

Each SNV and INDEL underwent driver gene mutation (31) analysis to determine the statistical likelihood of the specific mutation either activating an oncogene or inactivating a tumor suppressor gene (TSG). A mutation was considered a driver if the mutation was (i) nonsilent, (ii) CHASMplus assigned a TSG q-value or a gwCHASMplus q-value of ≤ 0.1, and (iii) the gene was defined as a driver by two or more previously published melanoma whole genomic or exomic datasets (Supplementary Table S4). Hotspot mutations in TERT were reviewed manually. Mutations were also analyzed for relevance to therapeutic resistance, including targeted therapies and immunotherapies (Supplementary Table S4). The mutations in Supplementary Table S5 underwent analysis by the Variant Effect Scoring Tool (VEST; ref. 32). SCNAs were annotated manually based on the same published melanoma datasets that were used for the SNVs and INDELs.

Neoantigen analysis

HLA typing was performed using a sequencing-based method to validate each patient's loci. Given the HLA status of each patient, each missense SNV was translated into a 17-mer peptide sequence that centered on the mutated amino acid. Each 17-mer was used to create 9-mer peptides using a sliding window approach to quantify putative MHC class I binding affinities via NetMHC v4.0. Neopeptides with a MT.Score of < 2, as defined by patient-matched specific HLAs, underwent further analysis.

Data availability

Sequence data have been deposited at the European Genomephenome Archive (EGA), which is hosted by the European Bioinformatics Institute and the Centre for Genomic Regulation, under accession number EGAS00001003582. Further information about EGA can be found at https://ega-archive.org and "The European Genome-phenome Archive of human data consented for biomedical research" (http://www.nature.com/ng/journal/v47/n7/full/ng.3312.html).

Whole-exome sequencing and driver mutations

We analyzed 7 patients who were diagnosed with cutaneous, uveal, or acral melanoma, and developed metastatic disease to multiple anatomic sites. Patient MEL1 was the subject of our previous analysis (33), while patients MEL2-MEL7 were newly analyzed in this study. Figure 1 shows the clinical timeline for each patient (Supplementary Table S1). Patient MEL1′s cutaneous metastases exhibited a mixed response to immunotherapy at the time of death, characterized by distinct gene expression profiles in responding versus progressing lesions (33). Metastases from patients MEL2-MEL7 were progressing at the time of death, in some cases demonstrating resistance to multiple therapeutic interventions (e.g., patient MEL6). Patient MEL4 (uveal melanoma) received local therapy to the primary tumor site but did not receive systemic treatment for metastatic disease. Upon death, each patient underwent a research autopsy to sample multiple metastatic sites (110 total tumor samples, range 11–26 per patient). We also recovered premortem tumor specimens from the pathology archives when available, including the primary tumors from 6 of 7 patients, and longitudinal biopsies of metastatic lesions. After performing WES of DNA collected from each tumor, we identified SNVs and INDELs (Fig. 2; Supplementary Table S2). We also analyzed the percentage of the genome altered by SCNAs, as well as WGD in each case (Supplementary Fig. S1; Supplementary Table S3). In cases MEL1, MEL2, MEL5, and MEL7, we detected WGD in nearly all tumor samples within each case, indicating that WGD in these cases was a relatively early event in the evolution of these tumors. Interestingly, in MEL3 (cutaneous) and MEL4 (uveal), we observed heterogeneity for WGD, indicating that WGD occurred relatively late in tumor evolution in these two cases. We did not detect any evidence for WGD in the acral melanoma, MEL6.

Figure 2.

Somatic mutation analysis and phylogenetics. See Fig. 1 for sample legend. For the histograms on the top of the figure, when applicable the primary tumor is the first column, followed by the tumors ranked by the number of somatic mutations (≥5% variant allele frequency). The proportion of SNVs consistent with the UV signature YC>T are also shown. For the phylogenies, the trunk and branch lengths are proportional to the number of somatic SNVs and INDELs as depicted in the top left by the scale bar for “100 mutations” (for the seven phylogenies with no outlined boxes) and the scale bar for “10 mutations” (for the four phylogenies within the outlined boxes). For patients MEL4 to MEL7, each corresponding box represents the higher-resolution phylogeny. A dotted line appears when the branch has been artificially extended for visualization. Putative driver genes, immunotherapy related genes, and mutation-targeted therapy genes affected by mutations are shown in blue. For tumors, P: primary, R: recurrence, M: metastasis.

Figure 2.

Somatic mutation analysis and phylogenetics. See Fig. 1 for sample legend. For the histograms on the top of the figure, when applicable the primary tumor is the first column, followed by the tumors ranked by the number of somatic mutations (≥5% variant allele frequency). The proportion of SNVs consistent with the UV signature YC>T are also shown. For the phylogenies, the trunk and branch lengths are proportional to the number of somatic SNVs and INDELs as depicted in the top left by the scale bar for “100 mutations” (for the seven phylogenies with no outlined boxes) and the scale bar for “10 mutations” (for the four phylogenies within the outlined boxes). For patients MEL4 to MEL7, each corresponding box represents the higher-resolution phylogeny. A dotted line appears when the branch has been artificially extended for visualization. Putative driver genes, immunotherapy related genes, and mutation-targeted therapy genes affected by mutations are shown in blue. For tumors, P: primary, R: recurrence, M: metastasis.

Close modal

Recent sequencing studies have demonstrated that cutaneous, uveal, and acral melanomas acquire distinct driver gene mutations during tumor evolution (1, 2, 34–37). The driver gene mutations we observed reflected the respective melanoma subtype of each case (Supplementary Tables S4 and S5). Specifically, the cutaneous melanomas harbored mutations in ARID2, BRCA1, CDKN2A, EZH2, KIT, MDM2, NF1, NRAS, RAC1, SMARCA4, and TERT; the uveal melanomas had mutations in BAP1, CDKN2A, GNA11, and GNAQ; and the acral melanomas had mutations affecting BRAF, CDKN2A, and NRAS. With the exception of a single SNV in CDKN2A in patient MEL3, the driver gene mutations were found to be present in all metastases within each case (Fig. 2). This implies that these common melanoma driver mutations occurred prior to systemic dissemination.

Cutaneous melanoma is one of most frequently mutated human cancers (34, 35, 37). UV light is known to cause a specific mutation signature (C>T at dypyrimidine sites) that is common in cutaneous melanomas (34, 35), but not in uveal or acral melanomas. The importance of UV damage in other melanoma subtypes has been a subject of scientific debate, with studies supporting (17) or refuting (38) a significant role. The cutaneous melanomas in our study exhibited a higher tumor mutational burden (TMB; Fig. 2, SNV median = 1,033, range 665–1,233) compared with uveal and acral melanomas (SNV median = 21, range 15–39). Upon analyzing these SNVs for mutational signatures, the three cutaneous melanomas exhibited an abundant UV-damage signature, whereas the uveal and acral melanomas did not (Fig. 2; Supplementary Figs. S2–S4; Supplementary Table S6). We also observed a high proportion of C>T mutations at dipyrimidine sites as compared with nondipyrimidine sites in the cutaneous melanomas (Supplementary Fig. S2). In the uveal and acral melanomas, we observed somatic SNVs that correlate with a UV signature, albeit at a lower proportion compared with the cutaneous melanomas. We found that the proportions of C>T mutations in the uveal and acral melanomas were similar between dipyrimidine and nondypyrimidine sites, as were the percentages of other substitutions at these sites (Supplementary Fig. S2; ref. 38). Overall, this indicated that uveal and acral melanomas did not exhibit a pronounced UV signature to the same extent as found in cutaneous melanomas. This was confirmed in a second mutation signature analysis using Palimpsest (30, 39), in which we observed that six of 12 mutation signatures were predominantly C>T transitions (Supplementary Figs. S3 and S4; Supplementary Table S6). Interestingly, a seventh signature represented a putative indirect effect of UV exposure (see Palimp_10; ref. 39). We identified one predominant mutation signature in each of the uveal and acral melanomas (Supplementary Fig. S3; Supplementary Table S6), each of which was dissimilar to the aging signatures previously described in these tumor types (37, 40), suggesting that the mutational processes of uveal and acral melanomas require further elucidation.

Phylogenetic analysis

Phylogenies based on the somatic SNVs and INDELs (Fig. 2) indicated that the cumulative lengths of cutaneous melanoma trunks and branches were significantly longer than those of the acral and uveal melanomas. In part, this was due to an approximate 40-fold higher mutational burden in cutaneous melanomas (P = 0.02, t test), predominantly due to longer trunks rather than longer branches or leaves. We further quantified this observation using the Jaccard similarity coefficient (29), and found that the cutaneous melanomas had significantly higher Jaccard similarity coefficients among all tumors in an individual patient, than the uveal or acral melanomas (P < 0.00001, t test; Supplementary Fig. S5; Supplementary Table S7). This difference was not due to differences in tumor purities as assessed by the SNVs and INDELs (P = 0.93, t test; Supplementary Table S1). Overall, these results suggest that tumors from an individual patient with cutaneous melanoma exhibited higher genetic similarity than those found in patients with uveal or acral melanoma, although there were some exceptions (e.g., sample MEL1-M4). These findings indicate that in addition to having a higher TMB, cutaneous melanomas also exhibit different phylogenetic structures, typified by longer trunk lengths (i.e., a greater frequency of common mutations among all lesions), compared with uveal or acral melanomas. Thus, intrinsic treatment resistance may be more likely to occur in uveal and acral melanomas, which display more genetic diversity among different tumor lesions in an individual patient, compared with cutaneous melanomas. To determine the extent to which UV signature mutations specifically contribute to the differences in TMB or phylogenetic relationships, we recomputed the phylogenies after removal of all mutations corresponding to this signature from all cases (Supplementary Figs. S5 and S6). Trunks of the cutaneous phylogenies remained approximately 4× longer than those of the uveal or acral phylogenies, which suggests two features of cutaneous melanoma: first, UV mutations alone do not fully account for the large differences in mutational burden among these subtypes, and second, the TMB contributed by UV damage in cutaneous melanoma mostly occurs prior to metastatic dissemination.

Evolution of somatic mutations associated with treatment resistance

We next explored somatic alterations that may have contributed to treatment resistance, given that genomic mechanisms have been implicated in intrinsic or acquired resistance to mutation-targeted therapies and immune checkpoint blockade in melanoma. Six patients received immunotherapy with checkpoint blocking antibodies (anti-PD-1, anti-CTLA-4), including 1 patient (MEL2) who also received IL2 and IL15; 2 of these patients also received BRAF and/or MEK inhibitors (cobimetinib, dabrafenib, and trametinib; Fig. 1). Thus, we analyzed each patient's SNVs, INDELs, and SCNAs for somatic mutations in genes curated from >70 melanoma studies (Supplementary Tables S4 and S5). One patient with ocular melanoma (MEL4) did not receive any systemic therapy and was not included in this analysis.

In 4 of 6 patients who received immunotherapy, we identified multiple somatic mutations that may have contributed to treatment resistance: an MDM2 gain and two point mutations in JAK2 in MEL1, a loss of heterozygosity in B2M and a SNV in EZH2 in MEL2, a B2M SNV in MEL3, and a PAK1 gain in MEL7 (Supplementary Table S5; refs. 8, 10, 11, 41, 42). These somatic mutations were detected on the trunk of each patient's respective phylogeny, except for the JAK2 point mutations in MEL1 and the B2M SNV in MEL3 (Fig. 2). In MEL1, one JAK2 mutation (L1001*) was detected in a subset of the cutaneous metastases that were progressing or regressing under immunotherapy (Supplementary Table S1), while a second frameshift mutation in JAK2 (E268Gfs*5) occurred in one regressing metastasis (MEL1-M15). Both JAK2 mutations were predicted to have a high pathogenicity impact by VEST (E268Gfs*5 P = 0.012, L1001* P = 0.002; ref. 32). Thus, both mutations could have resulted in a loss of function of JAK2. The two JAK2 mutations may have thus represented emerging resistant subclones that did not yet manifest as fully resistant to immunotherapy. The B2M mutation in MEL3 occurred in two lung metastases, MEL3-M17 and MEL3-M19. We did not identify any somatic mutation associated with immunotherapy resistance in patients MEL5 or MEL6. In summary, we identified 1–3 putative resistance mechanisms to immunotherapy in the cutaneous melanomas MEL1, MEL2, and MEL3, and in acral melanoma MEL7.

Two patients, MEL5 (uveal) and MEL6 (acral), received therapies specific to their tumor driver mutations, in addition to receiving immunotherapy. Patient MEL5 received cobimetinib, and patient MEL6 received combination therapy with dabrafenib + trametinib. Whereas we did not identify any known mechanisms of intrinsic resistance in MEL5, we identified a single therapeutically relevant mutation, a SCNA deleting CDKN2A on the trunk of the phylogeny, that may have contributed to resistance of combined BRAF and MEK therapeutic targeting in MEL6 (43).

Neoantigen evolution

To further explore genetic mechanisms potentially related to endogenous antimelanoma immunity and immunotherapeutic response, we analyzed the somatic SNVs and INDELs for putative neoantigens (Fig. 3; Supplementary Table S8). Each of the seven cases exhibited multiple putative tumor neoantigens predicted to bind to autologous MHC class I molecules (HLA-A, HLA-B, HLA-C) for potential immune recognition (Fig. 3), although these mutations may not have been expressed in RNA and peptides may not have existed on the cell surface, or may have been unrecognizable by immune cells within each patient. Expectedly, the number of neoantigens in the cutaneous melanomas was higher than in uveal and acral melanomas, reflecting the higher mutational burdens found in the former. Interestingly, regardless of melanoma subtype, approximately half of the somatic mutations in each melanoma generated at least one predicted neoantigen (Fig. 3; range 44%–59%). Likewise, the proportion of mutations that yielded single versus multiple neoantigens did not widely vary across cases (Fig. 3). This demonstrates that, despite widely divergent mutational burdens, these seven melanomas exhibited similar proportions of somatic mutations that may have generated neoantigens recognizable by the immune system.

Figure 3.

Analysis of neoantigens. In the top row, the histograms show the number of neopeptides predicted from somatic mutations with ≥5% variant allele frequency in each sample. Cutaneous melanoma: patients MEL1, MEL2, and MEL3; uveal melanoma: patients MEL4 and MEL5; acral melanoma: patients MEL6 and MEL7. In the bottom left, the somatic mutations of each patient are categorized into three columns of bar charts. The first column indicates the proportion of somatic mutations that were (i) not predicted to generate a neoantigen (gray), (ii) predicted to generate only weakly binding neoantigens (pink), and (iii) predicted to generate at least one strongly binding neoantigen (dark pink), defined by the predicted patient-matched HLA binding affinity (weak = 0.5–1.99, strong < 0.5). The second column indicates the proportion of somatic mutations that generated single (pink) versus multiple (red) neoantigens with a predicted patient-matched HLA binding affinity (ranging 0–1.99). The third column indicates the proportion of somatic mutations that did or did not generate at least one neoantigen, distinguished by trunk or branch position on the respective phylogeny [neoantigens (Neos) shown in red and pink, mutations not predicted to generate a neoantigen shown in black and gray]. In the bottom right, the MEL4 phylogeny is shown, with the trunk and branches of each sample indicating the number of mutations that did not generate a neoantigen (gray) and the number of putative neoantigen:HLA binding predictions that were weak (0.5–1.99, pink) or strong (<0.5, dark pink). The legend and scale bar are shown in the bottom right (scale bar length = 10 putative neoantigen:HLA binding predictions). The histogram below the phylogeny indicates the proportion of none, weak or strong neoantigen:HLA predictions on the trunk and private to each sample (excluding samples M5, M9, and M7 that did not have private mutations).

Figure 3.

Analysis of neoantigens. In the top row, the histograms show the number of neopeptides predicted from somatic mutations with ≥5% variant allele frequency in each sample. Cutaneous melanoma: patients MEL1, MEL2, and MEL3; uveal melanoma: patients MEL4 and MEL5; acral melanoma: patients MEL6 and MEL7. In the bottom left, the somatic mutations of each patient are categorized into three columns of bar charts. The first column indicates the proportion of somatic mutations that were (i) not predicted to generate a neoantigen (gray), (ii) predicted to generate only weakly binding neoantigens (pink), and (iii) predicted to generate at least one strongly binding neoantigen (dark pink), defined by the predicted patient-matched HLA binding affinity (weak = 0.5–1.99, strong < 0.5). The second column indicates the proportion of somatic mutations that generated single (pink) versus multiple (red) neoantigens with a predicted patient-matched HLA binding affinity (ranging 0–1.99). The third column indicates the proportion of somatic mutations that did or did not generate at least one neoantigen, distinguished by trunk or branch position on the respective phylogeny [neoantigens (Neos) shown in red and pink, mutations not predicted to generate a neoantigen shown in black and gray]. In the bottom right, the MEL4 phylogeny is shown, with the trunk and branches of each sample indicating the number of mutations that did not generate a neoantigen (gray) and the number of putative neoantigen:HLA binding predictions that were weak (0.5–1.99, pink) or strong (<0.5, dark pink). The legend and scale bar are shown in the bottom right (scale bar length = 10 putative neoantigen:HLA binding predictions). The histogram below the phylogeny indicates the proportion of none, weak or strong neoantigen:HLA predictions on the trunk and private to each sample (excluding samples M5, M9, and M7 that did not have private mutations).

Close modal

We also investigated the evolutionary patterns of neoantigens. We found that the proportion of neoantigens on the phylogenetic trunk varied among the cases; however, these proportions simply recapitulated the overall mutational pattern (Fig. 3). Thus, each case acquired neoantigens that were shared by all samples as well as neoantigens that were private to certain melanoma lesions. For example, in patient MEL4, the GNA11 p.Q209L mutation was a driver mutation that was shared by all tumors and also generated three putative neopeptides, one of which was predicted to bind to multiple HLA molecules in this patient (Supplementary Table S8). This GNA11 mutation exemplifies the possibility that driver gene mutations may create neoantigens, thus providing targets for immunotherapy that will be present across a patient population (44). Patient MEL4 also acquired a private mutation in the OGN gene in a liver metastasis (MEL4-M2; Fig. 3), a small deletion that was predicted to generate 52 unique neopeptides with binding affinities ranging from weak to strong (Supplementary Table S8). This illustrates that a private mutation, although restricted to a single metastasis, may nonetheless generate multiple neopeptides that could be targeted by the immune system. As another example of this principle, we observed private mutations generating neopeptides among metastases from patient MEL6 (Fig. 3).

Previous multilesion studies of melanoma genomic evolution have most often focused on a single subtype—cutaneous (12–14, 22), uveal (15, 16), or acral (17, 18)—although some studies have included both cutaneous and acral melanomas (19, 20). Birkeland and colleagues sequenced cutaneous, uveal, and acral melanomas (21); however, only the cutaneous melanomas underwent multilesion analysis. To our knowledge, our study is unique in utilizing WES to survey an extensive set of tumors representing these three melanoma subtypes. This is important because the dataset we analyzed here was normalized for sequencing approach, mutational calling, and evolutionary analysis, thus providing an opportunity to compare evolutionary patterns across patients with melanoma.

With improvements in the utility of immune checkpoint blockade and mutation-targeted therapies against metastatic melanoma comes the need to better understand the mechanisms of treatment resistance. In this study, we uncover several aspects of the genetic evolution of such resistant tumors across three main melanoma subtypes.

In 5 of 6 patients in this study who received systemic immune-based and mutation-targeted therapies (cobimetinib, dabrafenib, and trametinib), we found 1–3 genetic alterations that may represent functionally nonredundant resistance mechanisms coexisting in the same patient. We also found that more than half of these resistance mechanisms (5/8) were shared by all metastases within a given patient. Notable exceptions that may represent clonal or organ-site specific differences among metastases were found in JAK2 in MEL1 and B2M in MEL3. Several studies in melanoma have described putative molecular markers of therapeutic resistance, including concordant driver genes (6), discordant driver genes (45), heterogeneously acquired resistance (43, 46), and transcriptional changes (33, 47). Our results indicate that these melanomas harbored genetic changes that may have contributed to therapeutic resistance, many of which were shared by all tumor samples within an individual patient (12, 14, 16, 21, 36). This indicates that future interventions to circumvent intrinsic resistance hold the potential to successfully target most (if not all) metastases within a patient. We did not identify any somatic mutations associated with immunotherapy resistance in patients MEL5 or MEL6. One explanation for this could be that the immunotherapy resistance in these cases was due to nongenetic factors (33), or that unknown genetic factors may confer immunotherapy resistance that are specific to uveal or acral melanomas.

Our results showed that the evolution of predicted MHC I–restricted neoantigens largely recapitulated the broader evolutionary patterns of somatic mutations. However, we found multiple examples of somatic mutations that generated an exceptional number of putative neopeptides. Such mutations were likely passenger gene mutations, a finding that has potential clinical implications. For example, MEL4 was diagnosed with a uveal melanoma, a tumor type that typically has a relatively low mutational burden. However, the single OGN mutation in liver metastasis MEL4-M2 was predicted to generate multiple neoantigens that may have served as targets for the immune system. This example illustrates that a single passenger gene mutation may generate multiple targets for immune cells, even in tumors with a low mutational burden. This finding is important because it provides optimism for treating low TMB metastases, as private mutations isolated to a single lesion may nonetheless provide additional molecular targets for patients receiving immunotherapies. Future studies will be needed to elucidate the extent to which such private mutations result in beneficial response to immunotherapy.

Melanomas arise from melanocytes that inhabit tissues at select organ sites throughout the body and are thus subject to diverse microenvironments and mutagens, resulting in several genetically and phenotypically distinct melanoma subtypes (2). For example, DNA damage from UV radiation represents a major mutagenic process in cutaneous melanoma, but not in uveal or acral melanomas [although UV radiation has been implicated in uveal melanomas arising from the iris (40)]. We found that the high mutational burden in the cutaneous melanomas was associated with a high genetic similarity among multiple tumor lesions in an individual patient. In contrast, the lower mutational burden in the uveal and acral melanomas was associated with a lower genetic similarity among tumor deposits. This finding aligns with the notion that UV-induced mutagenesis occurs prior to metastatic clone dissemination (48); however, the relatively high genetic similarities of cutaneous melanoma metastases did not depend solely on the UV mutational signature. This is consistent with three possible scenarios: (i) the three main melanoma subtypes (cutaneous, uveal, acral) have distinct cells of origin; (ii) the differentiation state of the origin cell differs among subtypes, possibly affecting mutation rates and DNA repair; and/or (iii) the origin cells across the subtypes experience diverse microenvironments.

The phylogenetic analyses described herein also have implications for the relative timing of metastatic dissemination in these cases. In all seven cases, the genomes of multiple metastatic clones were highly similar or identical despite inhabiting distant organs, implying that there was not sufficient time for DNA damage to accumulate among these clones during dissemination. This is surprising because several premortem tumors were nonetheless found to be highly similar to metastases collected years later (e.g., patients MEL1 and MEL2; Fig. 1). A relatively low degree of genetic heterogeneity has also been observed in pancreatic and lung cancers, in contrast to other tumors types with high genetic heterogeneity (e.g., kidney and endometrial carcinomas), although further investigation is needed to define the extent to which underlying tumor biology and clinical interventions influence the genetic heterogeneity observed (49).

Our study has several limitations. Although we analyzed a large number of specimens, they were derived from a limited number of cases (7 patients) with melanomas that were progressing at the time of death, and it is unclear how our results would generalize to a larger patient population. Our results suggest that the relative infrequency of acquired resistance to immunotherapy among patients with cutaneous melanoma may be due to a relatively high genetic similarity among multiple tumors within an individual patient, as exhibited by the cutaneous melanomas here. A second limitation of our study was that with the exception of MEL1, we did not analyze patients whose melanomas responded to therapy. A third limitation was that we only focused on somatic mutations, and did not analyze other types of molecular variation, or tumor microenvironmental and immune cell factors that are posited to contribute to melanoma pathogenesis and treatment resistance (50). Regardless of the molecular mechanism, our results quantify the extent that putative innate and acquired resistance mechanisms occur throughout the genome during melanoma progression and metastasis.

A.P. Makohon-Moore reports grants from NCI during the conduct of the study. E.J. Lipson reports grants from Melanoma Research Alliance, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Barney Family Foundation, Moving for Melanoma of Delaware, and Laverna Hahn Charitable Trust during the conduct of the study, as well as grants and personal fees from Bristol-Myers Squibb, Merck, and Regeneron and personal fees from Array BioPharma, EMD Serono, MacroGenics, Novartis, and Sanofi Genzyme outside the submitted work. J.E. Hooper reports grants from NIH/NCI during the conduct of the study and other from Springer Publishing outside the submitted work. V. Makarov reports a patent for EP3090066A2, Determinants of cancer response to immunotherapy issued and with royalties paid. N. Riaz reports grants from BMS and Pfizer, and grants and personal fees from REPARE Therapeutics outside the submitted work. M.A. Postow reports grants and personal fees from BMS, Merck, and Array BioPharma; personal fees from Eisai; and grants from Novartis, RGenix, Infinity, and AstraZeneca outside the submitted work. D.B. Solit reports personal fees from Pfizer, Loxo Oncology, Lilly Oncology, BridgeBio, Illumina, Vividion Therapeutics, and Scorpion Therapeutics outside the submitted work. T.A. Chan reports other from Gritstone Oncology; grants from AstraZeneca, BMS, and Pfizer; and grants and personal fees from Illumina outside the submitted work. B.S. Taylor reports grants and personal fees from Genentech, Inc. and personal fees from Boehringer Ingelheim and Loxo Oncology at Lilly outside the submitted work. S.L. Topalian reports grants from Melanoma Research Alliance, Bloomberg-Kimmel Institute for Cancer Immunotherapy, and NCI R01 CA142779 and other from Barney Family Foundation, Moving for Melanoma of Delaware, and Laverna Hahn Charitable Trust during the conduct of the study, as well as other from Aduro Biotech, Dracen Pharmaceuticals, Jounce Therapeutics, Tizona LLC, and Trieza Therapeutics; personal fees from Amgen, AstraZeneca, Bayer, Dynavax Technologies, Immunocore, Immunomic Therapeutics, Janssen Pharmaceuticals, and Merck; grants from Bristol-Myers Squibb and Compugen; personal fees and other from DNAtrix, Dragonfly Therapeutics, Ervaxx Ltd., Five Prime Therapeutics, RAPT, and WindMIL; and grants and other from Potentza Therapeutics outside the submitted work; in addition, S.L. Topalian has a patent for Aduro Biotech licensed, Arbor Pharmaceuticals licensed, Bristol-Myers Squibb with royalties paid, Immunomic Therapeutics with royalties paid, NexImmune licensed, and WindMIL licensed. C.A. Iacobuzio-Donahue reports other from Bristol Myers Squibb outside the submitted work. No disclosures were reported by the other authors.

A.P. Makohon-Moore: Conceptualization, resources, data curation, software, formal analysis, supervision, funding acquisition, validation, investigation, visualization, methodology, writing-original draft, project administration, writing-review and editing. E.J. Lipson: Conceptualization, resources, data curation, software, formal analysis, supervision, funding acquisition, validation, investigation, visualization, methodology, writing-original draft, project administration, writing-review and editing. J.E. Hooper: Resources, data curation, investigation, methodology, writing-original draft, project administration. A. Zucker: Resources, data curation, investigation, methodology, writing-original draft, project administration. J. Hong: Data curation, software, formal analysis, investigation, methodology, writing-original draft, project administration. C.M. Bielski: Data curation, software, formal analysis, investigation, methodology, writing-original draft, project administration. A. Hayashi: Data curation, software, formal analysis, investigation, methodology, writing-original draft, project administration. C. Tokheim: Data curation, software, formal analysis, investigation, methodology, writing-original draft, project administration. P. Baez: Resources, data curation, investigation, methodology, writing-original draft, project administration. R. Kappagantula: Resources, data curation, investigation, methodology, writing-original draft, project administration. Z. Kohutek: Resources, data curation, investigation, methodology, writing-original draft, project administration. V. Makarov: Data curation, software, formal analysis, investigation, methodology, writing-original draft, project administration. N. Riaz: Data curation, software, formal analysis, investigation, methodology, writing-original draft, project administration. M.A. Postow: Resources, data curation, investigation, methodology, writing-original draft, project administration. P.B. Chapman: Resources, data curation, investigation, methodology, writing-original draft, project administration. R. Karchin: Data curation, software, formal analysis, investigation, methodology, writing-original draft, project administration. N.D. Socci: Data curation, software, formal analysis, investigation, methodology, writing-original draft, project administration. D.B. Solit: Resources, data curation, formal analysis, investigation, methodology, writing-original draft, project administration. T.A. Chan: Data curation, software, formal analysis, investigation, methodology, writing-original draft, project administration. B.S. Taylor: Data curation, software, formal analysis, investigation, methodology, writing-original draft, project administration. S.L. Topalian: Conceptualization, resources, supervision, funding acquisition, visualization, methodology, writing-original draft, project administration, writing-review and editing. C.A. Iacobuzio-Donahue: Conceptualization, resources, data curation, software, formal analysis, supervision, funding acquisition, validation, investigation, visualization, methodology, writing-original draft, project administration, writing-review and editing.

The authors thank the patients and families who generously participated in this study. They thank Richard White for critical comments on the article, and Annamalai Kumar (MSKCC) for assisting with HLA genotyping. This study was supported by the Melanoma Research Alliance (to E.J. Lipson, C.A. Iacobuzio-Donahue, and S.L. Topalian), the Bloomberg-Kimmel Institute for Cancer Immunotherapy (to E.J. Lipson and S.L. Topalian), the Barney Family Foundation (to E.J. Lipson and S.L. Topalian), Moving for Melanoma of Delaware (to E.J. Lipson and S.L. Topalian), the Laverna Hahn Charitable Trust (to E.J. Lipson and S.L. Topalian), the NCI (R01 CA142779, to S.L. Topalian), and the MSKCC TROT fellowship (T32 CA160001-06 and K99 CA229979 to A.P. Makohon-Moore). The Legacy Gift Rapid Autopsy Program at Johns Hopkins is supported by an NIH Cancer Clinical Core Support grant P30 CA006973 and a grant from the Sol Goldman Pancreatic Research Center. This research was also funded in part through the NIH/NCI Cancer Center Support Grant P30 CA008748. The authors also acknowledge funding sources, including Pershing Square Sohn Cancer Research grant (to T.A. Chan), the PaineWebber Chair (to T.A. Chan), NIH R01 CA205426 (to T.A. Chan), NIH R35 CA232097 (to T.A. Chan), and the STARR Cancer Consortium (to T.A. Chan).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Shain
AH
,
Bastian
BC
. 
From melanocytes to melanomas
.
Nat Rev Cancer
2016
;
16
:
345
58
.
2.
Bastian
BC
. 
The molecular pathology of melanoma: an integrated taxonomy of melanocytic neoplasia
.
Annu Rev Pathol
2014
;
9
:
239
71
.
3.
Shi
H
,
Moriceau
G
,
Kong
X
,
Lee
M-K
,
Lee
H
,
Koya
RC
, et al
Melanoma whole-exome sequencing identifies (V600E)B-RAF amplification-mediated acquired B-RAF inhibitor resistance
.
Nat Commun
2012
;
3
:
724
.
4.
Nazarian
R
,
Shi
H
,
Wang
Q
,
Kong
X
,
Koya
RC
,
Lee
H
, et al
Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation
.
Nature
2010
;
468
:
973
7
.
5.
Kluger
HM
,
Tawbi
HA
,
Ascierto
ML
,
Bowden
M
,
Callahan
MK
,
Cha
E
, et al
Defining tumor resistance to PD-1 pathway blockade: recommendations from the first meeting of the SITC Immunotherapy Resistance Taskforce
.
J Immunother Cancer
2020
;
8
:
e000398
.
6.
Turajlic
S
,
Furney
SJ
,
Stamp
G
,
Rana
S
,
Ricken
G
,
Oduko
Y
, et al
Whole-genome sequencing reveals complex mechanisms of intrinsic resistance to BRAF inhibition
.
Ann Oncol
2014
;
25
:
959
67
.
7.
Whittaker
SR
,
Theurillat
J-P
,
Van Allen
E
,
Wagle
N
,
Hsiao
J
,
Cowley
GS
, et al
A genome-scale RNA interference screen implicates NF1 loss in resistance to RAF inhibition
.
Cancer Discov
2013
;
3
:
350
62
.
8.
Miao
D
,
Margolis
CA
,
Vokes
NI
,
Liu
D
,
Taylor-Weiner
A
,
Wankowicz
SM
, et al
Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors
.
Nat Genet
2018
;
50
:
1271
81
.
9.
Gao
J
,
Shi
LZ
,
Zhao
H
,
Chen
J
,
Xiong
L
,
He
Q
, et al
Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy
.
Cell
2016
;
167
:
397
404
.
10.
Zaretsky
JM
,
Garcia-Diaz
A
,
Shin
DS
,
Escuin-Ordinas
H
,
Hugo
W
,
Hu-Lieskovan
S
, et al
Mutations associated with acquired resistance to PD-1 blockade in melanoma
.
N Engl J Med
2016
;
375
:
819
29
.
11.
Sade-Feldman
M
,
Jiao
YJ
,
Chen
JH
,
Rooney
MS
,
Barzily-Rokni
M
,
Eliane
JP
, et al
Resistance to checkpoint blockade therapy through inactivation of antigen presentation
.
Nat Commun
2017
;
8
:
1136
.
12.
Gartner
JJ
,
Davis
S
,
Wei
X
,
Lin
JC
,
Trivedi
NS
,
Teer
JK
, et al
Comparative exome sequencing of metastatic lesions provides insights into the mutational progression of melanoma
.
BMC Genomics
2012
;
13
:
505
.
13.
Ding
L
,
Kim
M
,
Kanchi
KL
,
Dees
ND
,
Lu
C
,
Griffith
M
, et al
Clonal architectures and driver mutations in metastatic melanomas
.
PLoS One
2014
;
9
:
e111153
.
14.
Manca
A
,
Paliogiannis
P
,
Colombino
M
,
Casula
M
,
Lissia
A
,
Botti
G
, et al
Mutational concordance between primary and metastatic melanoma: a next-generation sequencing approach
.
J Transl Med
2019
;
17
:
289
.
15.
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
30
.
16.
Rodrigues
M
,
Mobuchon
L
,
Houy
A
,
Alsafadi
S
,
Baulande
S
,
Mariani
O
, et al
Evolutionary routes in metastatic uveal melanomas depend on MBD4 alterations
.
Clin Cancer Res
2019
;
25
:
5513
24
.
17.
Turajlic
S
,
Furney
SJ
,
Lambros
MB
,
Mitsopoulos
C
,
Kozarewa
I
,
Geyer
FC
, et al
Whole genome sequencing of matched primary and metastatic acral melanomas
.
Genome Res
2012
;
22
:
196
207
.
18.
Sharma
G
,
Lian
CG
,
Lin
WM
,
Amin-Mansour
A
,
Jané-Valbuena
J
,
Garraway
L
, et al
Distinct genetic profiles of extracranial and intracranial acral melanoma metastases
.
J Cutan Pathol
2016
;
43
:
884
91
.
19.
Harbst
K
,
Lauss
M
,
Cirenajwis
H
,
Winter
C
,
Howlin
J
,
Törngren
T
, et al
Molecular and genetic diversity in the metastatic process of melanoma
.
J Pathol
2014
;
233
:
39
50
.
20.
Sanborn
JZ
,
Chung
J
,
Purdom
E
,
Wang
NJ
,
Kakavand
H
,
Wilmott
JS
, et al
Phylogenetic analyses of melanoma reveal complex patterns of metastatic dissemination
.
Proc Natl Acad Sci U S A
2015
;
112
:
10995
1000
.
21.
Birkeland
E
,
Zhang
S
,
Poduval
D
,
Geisler
J
,
Nakken
S
,
Vodak
D
, et al
Patterns of genomic evolution in advanced melanoma
.
Nat Commun
2018
;
9
:
2665
.
22.
Sanna
A
,
Harbst
K
,
Johansson
I
,
Christensen
G
,
Lauss
M
,
Mitra
S
, et al
Tumor genetic heterogeneity analysis of chronic sun-damaged melanoma
.
Pigment Cell Melanoma Res
2020
;
33
:
480
9
.
23.
Iacobuzio-Donahue
CA
,
Michael
C
,
Baez
P
,
Kappagantula
R
,
Hooper
JE
,
Hollman
TJ
. 
Cancer biology as revealed by the research autopsy
.
Nat Rev Cancer
2019
;
19
:
686
97
.
24.
Li
H
,
Durbin
R
. 
Fast and accurate short read alignment with Burrows-Wheeler transform
.
Bioinformatics
2009
;
25
:
1754
60
.
25.
DePristo
MA
,
Banks
E
,
Poplin
R
,
Garimella
KV
,
Maguire
JR
,
Hartl
C
, et al
A framework for variation discovery and genotyping using next-generation DNA sequencing data
.
Nat Genet
2011
;
43
:
491
8
.
26.
Cibulskis
K
,
Lawrence
MS
,
Carter
SL
,
Sivachenko
A
,
Jaffe
D
,
Sougnez
C
, et al
Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples
.
Nat Biotechnol
2013
;
31
:
213
9
.
27.
Lek
M
,
Karczewski
KJ
,
Minikel
EV
,
Samocha
KE
,
Banks
E
,
Fennell
T
, et al
Analysis of protein-coding genetic variation in 60,706 humans
.
Nature
2016
;
536
:
285
91
.
28.
Shen
R
,
Seshan
VE
. 
FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing
.
Nucleic Acids Res
2016
;
44
:
e131
.
29.
Reiter
JG
,
Makohon-Moore
AP
,
Gerold
JM
,
Bozic
I
,
Chatterjee
K
,
Iacobuzio-Donahue
CA
, et al
Reconstructing metastatic seeding patterns of human cancers
.
Nat Commun
2017
;
8
:
14114
.
30.
Shinde
J
,
Bayard
Q
,
Imbeaud
S
,
Hirsch
T
,
Liu
F
,
Renault
V
, et al
Palimpsest: an R package for studying mutational and structural variant signatures along clonal evolution in cancer
.
Bioinformatics
2018
;
34
:
3380
1
.
31.
Tokheim
C
,
Karchin
R
. 
CHASMplus reveals the scope of somatic missense mutations driving human cancers
.
Cell Syst
2019
;
9
:
9
23
.
32.
Carter
H
,
Douville
C
,
Stenson
PD
,
Cooper
DN
,
Karchin
R
. 
Identifying Mendelian disease genes with the variant effect scoring tool
.
BMC Genomics
2013
;
14
:
S3
.
33.
Ascierto
ML
,
Makohon-Moore
A
,
Lipson
EJ
,
Taube
JM
,
McMiller
TL
,
Berger
AE
, et al
Transcriptional mechanisms of resistance to anti-PD-1 therapy
.
Clin Cancer Res
2017
;
23
:
3168
80
.
34.
Akbani
R
,
Akdemir
KC
,
Aksoy
BA
,
Albert
M
,
Ally
A
,
Amin
SB
, et al
Genomic classification of cutaneous melanoma
.
Cell
2015
;
161
:
1681
96
.
35.
Hodis
E
,
Watson
IR
,
Kryukov
GV
,
Arold
ST
,
Imielinski
M
,
Theurillat
J-P
, et al
A landscape of driver mutations in melanoma
.
Cell
2012
;
150
:
251
63
.
36.
Field
MG
,
Durante
MA
,
Anbunathan
H
,
Cai
LZ
,
Decatur
CL
,
Bowcock
AM
, et al
Punctuated evolution of canonical genomic aberrations in uveal melanoma
.
Nat Commun
2018
;
9
:
116
.
37.
Hayward
NK
,
Wilmott
JS
,
Waddell
N
,
Johansson
PA
,
Field
MA
,
Nones
K
, et al
Whole-genome landscapes of major melanoma subtypes
.
Nature
2017
;
545
:
175
80
.
38.
Krauthammer
M
,
Kong
Y
,
Ha
BH
,
Evans
P
,
Bacchiocchi
A
,
McCusker
JP
, et al
Exome sequencing identifies recurrent somatic RAC1 mutations in melanoma
.
Nat Genet
2012
;
44
:
1006
14
.
39.
Alexandrov
LB
,
Kim
J
,
Haradhvala
NJ
,
Huang
MN
,
Tian Ng
AW
,
Wu
Y
, et al
The repertoire of mutational signatures in human cancer
.
Nature
2020
;
578
:
94
101
.
40.
Johansson
PA
,
Brooks
K
,
Newell
F
,
Palmer
JM
,
Wilmott
JS
,
Pritchard
AL
, et al
Whole genome landscapes of uveal melanoma show an ultraviolet radiation signature in iris tumours
.
Nat Commun
2020
;
11
2408
.
41.
Shin
DS
,
Zaretsky
JM
,
Escuin-Ordinas
H
,
Garcia-Diaz
A
,
Hu-Lieskovan
S
,
Kalbasi
A
, et al
Primary resistance to PD-1 blockade mediated by JAK1/2 mutations
.
Cancer Discov
2017
;
7
:
188
201
.
42.
Zingg
D
,
Arenas-Ramirez
N
,
Sahin
D
,
Rosalia
RA
,
Antunes
AT
,
Haeusel
J
, et al
The histone methyltransferase Ezh2 controls mechanisms of adaptive resistance to tumor immunotherapy
.
Cell Rep
2017
;
20
:
854
67
.
43.
Moriceau
G
,
Hugo
W
,
Hong
A
,
Shi
H
,
Kong
X
,
Yu
CC
, et al
Tunable-combinatorial mechanisms of acquired resistance limit the efficacy of BRAF/MEK cotargeting but result in melanoma drug addiction
.
Cancer Cell
2015
;
27
:
240
56
.
44.
Shao
XM
,
Bhattacharya
R
,
Huang
J
,
Sivakumar
IKA
,
Tokheim
C
,
Zheng
L
, et al
High-throughput prediction of MHC class I and II neoantigens with MHCnuggets
.
Cancer Immunol Res
2020
;
8
:
396
408
.
45.
Yancovitz
M
,
Litterman
A
,
Yoon
J
,
Ng
E
,
Shapiro
RL
,
Berman
RS
, et al
Intra- and inter-tumor heterogeneity of BRAF(V600E))mutations in primary and metastatic melanoma
.
PLoS One
2012
;
7
:
e29336
.
46.
Van Allen
EM
,
Wagle
N
,
Sucker
A
,
Treacy
DJ
,
Johannessen
CM
,
Goetz
EM
, et al
The genetic landscape of clinical resistance to RAF inhibition in metastatic melanoma
.
Cancer Discov
2014
;
4
:
94
109
.
47.
Song
C
,
Piva
M
,
Sun
L
,
Hong
A
,
Moriceau
G
,
Kong
X
, et al
Recurrent tumor cell-intrinsic and -extrinsic alterations during MAPKi-induced melanoma regression and early adaptation
.
Cancer Discov
2017
;
7
:
1248
65
.
48.
Shain
AH
,
Yeh
I
,
Kovalyshyn
I
,
Sriharan
A
,
Talevich
E
,
Gagnon
A
, et al
The genetic evolution of melanoma from precursor lesions
.
N Engl J Med
2015
;
373
:
1926
36
.
49.
Iacobuzio-Donahue
CA
,
Litchfield
K
,
Swanton
C
. 
Intratumor heterogeneity reflects clinical disease course
.
Nat Cancer
2020
;
1
:
3
6
.
50.
Conway
JR
,
Kofman
E
,
Mo
SS
,
Elmarakeby
H
,
Van Allen
E
. 
Genomics of response to immune checkpoint therapies for cancer: implications for precision medicine
.
Genome Med
2018
;
10
:
93
.