Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Immunity ; 46(2): 197-204, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228279

ABSTRACT

Response to immune checkpoint blockade in mesenchymal tumors is poorly characterized, but immunogenomic dissection of these cancers could inform immunotherapy mediators. We identified a treatment-naive patient who has metastatic uterine leiomyosarcoma and has experienced complete tumor remission for >2 years on anti-PD-1 (pembrolizumab) monotherapy. We analyzed the primary tumor, the sole treatment-resistant metastasis, and germline tissue to explore mechanisms of immunotherapy sensitivity and resistance. Both tumors stained diffusely for PD-L2 and showed sparse PD-L1 staining. PD-1+ cell infiltration significantly decreased in the resistant tumor (p = 0.039). Genomically, the treatment-resistant tumor uniquely harbored biallelic PTEN loss and had reduced expression of two neoantigens that demonstrated strong immunoreactivity with patient T cells in vitro, suggesting long-lasting immunological memory. In this near-complete response to PD-1 blockade in a mesenchymal tumor, we identified PTEN mutations and reduced expression of genes encoding neoantigens as potential mediators of resistance to immune checkpoint therapy.


Subject(s)
Drug Resistance, Neoplasm/genetics , Leiomyosarcoma/pathology , PTEN Phosphohydrolase/genetics , Uterine Neoplasms/pathology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , DNA Mutational Analysis , Female , Gene Expression Profiling , Humans , Leiomyosarcoma/drug therapy , Leiomyosarcoma/genetics , Middle Aged , Mutation , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Transcriptome , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics
2.
Nature ; 540(7631): 114-118, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27905446

ABSTRACT

Germ-cell tumours (GCTs) are derived from germ cells and occur most frequently in the testes. GCTs are histologically heterogeneous and distinctly curable with chemotherapy. Gains of chromosome arm 12p and aneuploidy are nearly universal in GCTs, but specific somatic genomic features driving tumour initiation, chemosensitivity and progression are incompletely characterized. Here, using clinical whole-exome and transcriptome sequencing of precursor, primary (testicular and mediastinal) and chemoresistant metastatic human GCTs, we show that the primary somatic feature of GCTs is highly recurrent chromosome arm level amplifications and reciprocal deletions (reciprocal loss of heterozygosity), variations that are significantly enriched in GCTs compared to 19 other cancer types. These tumours also acquire KRAS mutations during the development from precursor to primary disease, and primary testicular GCTs (TGCTs) are uniformly wild type for TP53. In addition, by functional measurement of apoptotic signalling (BH3 profiling) of fresh tumour and adjacent tissue, we find that primary TGCTs have high mitochondrial priming that facilitates chemotherapy-induced apoptosis. Finally, by phylogenetic analysis of serial TGCTs that emerge with chemotherapy resistance, we show how TGCTs gain additional reciprocal loss of heterozygosity and that this is associated with loss of pluripotency markers (NANOG and POU5F1) in chemoresistant teratomas or transformed carcinomas. Our results demonstrate the distinct genomic features underlying the origins of this disease and associated with the chemosensitivity phenotype, as well as the rare progression to chemoresistance. These results identify the convergence of cancer genomics, mitochondrial priming and GCT evolution, and may provide insights into chemosensitivity and resistance in other cancers.


Subject(s)
Drug Resistance, Neoplasm , Genome, Human/genetics , Neoplasms, Germ Cell and Embryonal/drug therapy , Neoplasms, Germ Cell and Embryonal/genetics , Apoptosis , Disease Progression , Evolution, Molecular , Exome/genetics , Genomics , Humans , Loss of Heterozygosity , Male , Mitochondria/metabolism , Mutation , Nanog Homeobox Protein/deficiency , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasms, Germ Cell and Embryonal/metabolism , Neoplasms, Germ Cell and Embryonal/pathology , Octamer Transcription Factor-3/deficiency , Phylogeny , Proto-Oncogene Proteins p21(ras)/genetics , Teratoma/genetics , Testicular Neoplasms/drug therapy , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology , Transcriptome/genetics , Tumor Suppressor Protein p53/genetics
3.
N Engl J Med ; 371(15): 1426-33, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25295501

ABSTRACT

Everolimus, an inhibitor of the mammalian target of rapamycin (mTOR), is effective in treating tumors harboring alterations in the mTOR pathway. Mechanisms of resistance to everolimus remain undefined. Resistance developed in a patient with metastatic anaplastic thyroid carcinoma after an extraordinary 18-month response. Whole-exome sequencing of pretreatment and drug-resistant tumors revealed a nonsense mutation in TSC2, a negative regulator of mTOR, suggesting a mechanism for exquisite sensitivity to everolimus. The resistant tumor also harbored a mutation in MTOR that confers resistance to allosteric mTOR inhibition. The mutation remains sensitive to mTOR kinase inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/genetics , Thyroid Neoplasms/therapy , Tumor Suppressor Proteins/genetics , Combined Modality Therapy , Everolimus , Female , Humans , Lymphatic Metastasis/pathology , Middle Aged , Mutation , Neoplasm Recurrence, Local/diagnostic imaging , Neoplasm Recurrence, Local/pathology , Protein Conformation , Radiography , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/chemistry , Thyroid Carcinoma, Anaplastic , Thyroid Neoplasms/diagnostic imaging , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Tuberous Sclerosis Complex 2 Protein
4.
Genet Med ; 19(7): 787-795, 2017 07.
Article in English | MEDLINE | ID: mdl-28125075

ABSTRACT

PURPOSE: Implementing cancer precision medicine in the clinic requires assessing the therapeutic relevance of genomic alterations. A main challenge is the systematic interpretation of whole-exome sequencing (WES) data for clinical care. METHODS: One hundred sixty-five adults with metastatic colorectal and lung adenocarcinomas were prospectively enrolled in the CanSeq study. WES was performed on DNA extracted from formalin-fixed paraffin-embedded tumor biopsy samples and matched blood samples. Somatic and germ-line alterations were ranked according to therapeutic or clinical relevance. Results were interpreted using an integrated somatic and germ-line framework and returned in accordance with patient preferences. RESULTS: At the time of this analysis, WES had been performed and results returned to the clinical team for 165 participants. Of 768 curated somatic alterations, only 31% were associated with clinical evidence and 69% with preclinical or inferential evidence. Of 806 curated germ-line variants, 5% were clinically relevant and 56% were classified as variants of unknown significance. The variant review and decision-making processes were effective when the process was changed from that of a Molecular Tumor Board to a protocol-based approach. CONCLUSION: The development of novel interpretive and decision-support tools that draw from scientific and clinical evidence will be crucial for the success of cancer precision medicine in WES studies.Genet Med advance online publication 26 January 2017.


Subject(s)
Exome Sequencing/methods , Exome/genetics , Precision Medicine/methods , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Adult , Colorectal Neoplasms/genetics , Databases, Genetic , Genomics/methods , Germ-Line Mutation/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Lung Neoplasms/genetics , Mutation/genetics , Prospective Studies , Sequence Analysis, DNA/methods
5.
J Cutan Pathol ; 43(10): 884-91, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27251777

ABSTRACT

BACKGROUND: There is limited knowledge of the genetic alterations in acral melanoma metastases at different anatomic sites. Here, we characterized the genetic abnormalities of metastases in a 51-year-old man with stage IIIC heel melanoma who developed concomitant brain and cutaneous metastases in spite of multiple treatment modalities. METHODS: Melanoma cells were isolated following palliative resection of the patient's cortical tumor and biopsy of cutaneous thigh metastasis. Mutational analysis using polymerase chain reaction amplification and BLAST, as well as exome sequencing (160 Mb coverage) was performed on the tumors, cell lines generated thereof and normal lymph nodes. RESULTS: All specimens had neuroblastoma RAS viral oncogene homolog Q61K mutations. There was a 40-fold higher somatic mutation frequency in the brain metastasis compared to the cutaneous metastasis. The former showed truncations of DNA mismatch repair genes (MLH1 and MSH2), and non-canonical BRAF (v-raf murine sarcoma viral oncogene homolog B1), PIK3CA and NF-1 mutations not observed in the extracranial lesion. Genomic profiling of each cell line was concordant with the respective original tumor tissue. CONCLUSIONS: We present the mutational differences between brain and cutaneous acral melanoma metastases in a patient with concomitant lesions. Further genetic and functional studies are needed to understand the biology of metastatic disease appearing at different sites.


Subject(s)
Brain Neoplasms , Melanoma , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Skin Neoplasms , Biopsy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Class I Phosphatidylinositol 3-Kinases , Humans , Male , Melanoma/genetics , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Neurofibromin 1/genetics , Phosphatidylinositol 3-Kinases/genetics , Skin Neoplasms/genetics , Skin Neoplasms/metabolism
6.
Nat Cancer ; 5(3): 481-499, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38233483

ABSTRACT

Activating mutations in GNAQ/GNA11 occur in over 90% of uveal melanomas (UMs), the most lethal melanoma subtype; however, targeting these oncogenes has proven challenging and inhibiting their downstream effectors show limited clinical efficacy. Here, we performed genome-scale CRISPR screens along with computational analyses of cancer dependency and gene expression datasets to identify the inositol-metabolizing phosphatase INPP5A as a selective dependency in GNAQ/11-mutant UM cells in vitro and in vivo. Mutant cells intrinsically produce high levels of the second messenger inositol 1,4,5 trisphosphate (IP3) that accumulate upon suppression of INPP5A, resulting in hyperactivation of IP3-receptor signaling, increased cytosolic calcium and p53-dependent apoptosis. Finally, we show that GNAQ/11-mutant UM cells and patients' tumors exhibit elevated levels of IP4, a biomarker of enhanced IP3 production; these high levels are abolished by GNAQ/11 inhibition and correlate with sensitivity to INPP5A depletion. Our findings uncover INPP5A as a synthetic lethal vulnerability and a potential therapeutic target for GNAQ/11-mutant-driven cancers.


Subject(s)
Melanoma , Humans , Melanoma/drug therapy , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/therapeutic use , Mutation , Signal Transduction , Inositol Polyphosphate 5-Phosphatases/genetics
7.
Clin Cancer Res ; 25(16): 5135-5142, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31164371

ABSTRACT

PURPOSE: Leiomyosarcoma and liposarcoma are common subtypes of soft tissue sarcoma (STS). Patients with metastatic leiomyosarcoma or dedifferentiated liposarcoma (DDLPS) typically have worse outcomes compared with localized leiomyosarcoma or well-differentiated liposarcoma (WDLPS). A better understanding of genetic changes between primary/metastatic leiomyosarcoma and between WDLPS/DDLPS may provide insight into their genetic evolution. EXPERIMENTAL DESIGN: We interrogated whole-exome sequencing (WES) from "trios" of normal tissue, primary tumor, and metastatic tumor from individual patients with leiomyosarcoma (n = 9), and trios of normal tissue, well-differentiated tumor, and dedifferentiated tumor from individual patients with liposarcoma (n = 19). Specifically, we performed mutational, copy number, and tumor evolution analyses on these cohorts and compared patterns among leiomyosarcoma and liposarcoma trios. RESULTS: Leiomyosarcoma cases harbored shared drivers through a typical parent/child relationship where the metastatic tumor was derived from the primary tumor. In contrast, while all liposarcoma cases shared the characteristic focal chromosome 12 amplicon, most paired liposarcoma cases did not share additional mutations, suggesting a divergent evolutionary pattern from a common precursor. No highly recurrent genomic alterations from WES were identified that could be implicated as driving the progression of disease in either sarcoma subtype. CONCLUSIONS: From a genomic perspective, leiomyosarcoma metastases contain genetic alterations that are also found in primary tumors. WDLPS and DDLPS, however, appear to divergently evolve from a common precursor harboring 12q amplification, rather than as a transformation to a higher-grade tumor. Further efforts to identify specific drivers of these distinct evolutionary patterns may inform future translational and clinical research in STS.


Subject(s)
Cell Transformation, Neoplastic/genetics , Genetic Predisposition to Disease , Genomics , Leiomyosarcoma/genetics , Leiomyosarcoma/pathology , Liposarcoma/genetics , Liposarcoma/pathology , Adult , Aged , Biopsy , Female , Gene Expression Profiling , Genomics/methods , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Retrospective Studies , Exome Sequencing
8.
Clin Cancer Res ; 23(12): 3214-3222, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-27852700

ABSTRACT

Purpose: Squamous cell carcinoma of the anal canal (ASCC) accounts for 2% to 4% of gastrointestinal malignancies in the United States and is increasing in incidence; however, genomic features of ASCC are incompletely characterized. Primary treatment of ASCC involves concurrent chemotherapy and radiation (CRT), but the mutational landscape of resistance to CRT is unknown. Here, we aim to compare mutational features of ASCC in the pre- and post-CRT setting.Experimental Design: We perform whole-exome sequencing of primary (n = 31) and recurrent (n = 30) ASCCs and correlate findings with clinical data. We compare genomic features of matched pre- and post-CRT tumors to identify genomic features of CRT response. Finally, we investigate the mutational underpinnings of an extraordinary ASCC response to immunotherapy.Results: We find that both primary and recurrent ASCC tumors harbor mutations in genes, such as PIK3CA and FBXW7, that are also mutated in other HPV-associated cancers. Overall mutational burden was not significantly different in pre- versus post-CRT tumors, and several examples of shared clonal driver mutations were identified. In two cases, clonally related pre- and post-CRT tumors harbored distinct oncogenic driver mutations in the same cancer gene (KRAS or FBXW7). A patient with recurrent disease achieved an exceptional response to anti-programmed death (PD-1) therapy, and genomic dissection revealed high mutational burden and predicted neoantigen load.Conclusions: We perform comprehensive mutational analysis of ASCC and characterize mutational features associated with CRT. Although many primary and recurrent tumors share driver events, we identify several unique examples of clonal evolution in response to treatment. Clin Cancer Res; 23(12); 3214-22. ©2016 AACR.


Subject(s)
Anus Neoplasms/genetics , Carcinoma, Squamous Cell/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , F-Box-WD Repeat-Containing Protein 7/genetics , Adult , Aged , Aged, 80 and over , Anus Neoplasms/drug therapy , Anus Neoplasms/pathology , Anus Neoplasms/radiotherapy , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/radiotherapy , Chemoradiotherapy/adverse effects , Disease-Free Survival , Drug Resistance, Neoplasm/genetics , Evolution, Molecular , Female , Genome, Human/drug effects , Genome, Human/radiation effects , Humans , Male , Middle Aged , Mutation/drug effects , Mutation/radiation effects , Neoplasm Recurrence, Local , Radiation Tolerance/genetics
9.
PLoS One ; 12(6): e0178189, 2017.
Article in English | MEDLINE | ID: mdl-28594900

ABSTRACT

To further our understanding of the somatic genetic basis of uveal melanoma, we sequenced the protein-coding regions of 52 primary tumors and 3 liver metastases together with paired normal DNA. Known recurrent mutations were identified in GNAQ, GNA11, BAP1, EIF1AX, and SF3B1. The role of mutated EIF1AX was tested using loss of function approaches including viability and translational efficiency assays. Knockdown of both wild type and mutant EIF1AX was lethal to uveal melanoma cells. We probed the function of N-terminal tail EIF1AX mutations by performing RNA sequencing of polysome-associated transcripts in cells expressing endogenous wild type or mutant EIF1AX. Ribosome occupancy of the global translational apparatus was sensitive to suppression of wild type but not mutant EIF1AX. Together, these studies suggest that cells expressing mutant EIF1AX may exhibit aberrant translational regulation, which may provide clonal selective advantage in the subset of uveal melanoma that harbors this mutation.


Subject(s)
Genome, Human , Melanoma/genetics , Protein Biosynthesis/genetics , Uveal Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Eukaryotic Initiation Factor-1/genetics , Female , Humans , Male , Melanoma/pathology , Middle Aged , Mutation , Uveal Neoplasms/pathology , Young Adult
10.
Cancer Discov ; 7(9): 973-983, 2017 09.
Article in English | MEDLINE | ID: mdl-28515055

ABSTRACT

African-American men have the highest incidence of and mortality from prostate cancer. Whether a biological basis exists for this disparity remains unclear. Exome sequencing (n = 102) and targeted validation (n = 90) of localized primary hormone-naïve prostate cancer in African-American men identified several gene mutations not previously observed in this context, including recurrent loss-of-function mutations in ERF, an ETS transcriptional repressor, in 5% of cases. Analysis of existing prostate cancer cohorts revealed ERF deletions in 3% of primary prostate cancers and mutations or deletions in ERF in 3% to 5% of lethal castration-resistant prostate cancers. Knockdown of ERF confers increased anchorage-independent growth and generates a gene expression signature associated with oncogenic ETS activation and androgen signaling. Together, these results suggest that ERF is a prostate cancer tumor-suppressor gene. More generally, our findings support the application of systematic cancer genomic characterization in settings of broader ancestral diversity to enhance discovery and, eventually, therapeutic applications.Significance: Systematic genomic sequencing of prostate cancer in African-American men revealed new insights into prostate cancer, including the identification of ERF as a prostate cancer gene; somatic copy-number alteration differences; and uncommon PIK3CA and PTEN alterations. This study highlights the importance of inclusion of underrepresented minorities in cancer sequencing studies. Cancer Discov; 7(9); 973-83. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 920.


Subject(s)
Prostatic Neoplasms/genetics , Repressor Proteins/genetics , Black or African American/genetics , Animals , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , Exome , Humans , Male , Mice , Mutation , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/pathology , Exome Sequencing
11.
Genome Med ; 8(1): 79, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27460824

ABSTRACT

BACKGROUND: The diversity of clinical tumor profiling approaches (small panels to whole exomes with matched or unmatched germline analysis) may engender uncertainty about their benefits and liabilities, particularly in light of reported germline false positives in tumor-only profiling and use of global mutational and/or neoantigen data. The goal of this study was to determine the impact of genomic analysis strategies on error rates and data interpretation across contexts and ancestries. METHODS: We modeled common tumor profiling modalities-large (n = 300 genes), medium (n = 48 genes), and small (n = 15 genes) panels-using clinical whole exomes (WES) from 157 patients with lung or colon adenocarcinoma. We created a tumor-only analysis algorithm to assess germline false positive rates, the impact of patient ancestry on tumor-only results, and neoantigen detection. RESULTS: After optimizing a germline filtering strategy, the germline false positive rate with tumor-only large panel sequencing was 14 % (144/1012 variants). For patients whose tumor-only results underwent molecular pathologist review (n = 91), 50/54 (93 %) false positives were correctly interpreted as uncertain variants. Increased germline false positives were observed in tumor-only sequencing of non-European compared with European ancestry patients (p < 0.001; Fisher's exact) when basic germline filtering approaches were used; however, the ExAC database (60,706 germline exomes) mitigated this disparity (p = 0.53). Matched and unmatched large panel mutational load correlated with WES mutational load (r(2) = 0.99 and 0.93, respectively; p < 0.001). Neoantigen load also correlated (r(2) = 0.80; p < 0.001), though WES identified a broader spectrum of neoantigens. Small panels did not predict mutational or neoantigen load. CONCLUSIONS: Large tumor-only targeted panels are sufficient for most somatic variant identification and mutational load prediction if paired with expanded germline analysis strategies and molecular pathologist review. Paired germline sequencing reduced overall false positive mutation calls and WES provided the most neoantigens. Without patient-matched germline data, large germline databases are needed to minimize false positive mutation calling and mitigate ethnic disparities.


Subject(s)
Adenocarcinoma/genetics , Antigens, Neoplasm/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Precision Medicine , Adenocarcinoma/diagnosis , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Colonic Neoplasms/diagnosis , Colonic Neoplasms/pathology , Databases, Genetic , Exome , False Positive Reactions , Gene Expression Profiling , Genomics/methods , Germ-Line Mutation , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/pathology , Mutation Rate , Pedigree , Sequence Analysis, DNA
12.
Cell Rep ; 15(4): 857-865, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27149842

ABSTRACT

Large-scale genomic characterization of tumors from prospective cohort studies may yield new insights into cancer pathogenesis. We performed whole-exome sequencing of 619 incident colorectal cancers (CRCs) and integrated the results with tumor immunity, pathology, and survival data. We identified recurrently mutated genes in CRC, such as BCL9L, RBM10, CTCF, and KLF5, that were not previously appreciated in this disease. Furthermore, we investigated the genomic correlates of immune-cell infiltration and found that higher neoantigen load was positively associated with overall lymphocytic infiltration, tumor-infiltrating lymphocytes (TILs), memory T cells, and CRC-specific survival. The association with TILs was evident even within microsatellite-stable tumors. We also found positive selection of mutations in HLA genes and other components of the antigen-processing machinery in TIL-rich tumors. These results may inform immunotherapeutic approaches in CRC. More generally, this study demonstrates a framework for future integrative molecular epidemiology research in colorectal and other malignancies.

13.
JAMA Oncol ; 2(8): 1056-64, 2016 Aug 01.
Article in English | MEDLINE | ID: mdl-27124486

ABSTRACT

IMPORTANCE: Combined treatment with dabrafenib and trametinib (CombiDT) achieves clinical responses in only about 15% of patients with BRAF inhibitor (BRAFi)-refractory metastatic melanoma in contrast to the higher response rate observed in BRAFi-naïve patients. Identifying correlates of response and mechanisms of resistance in this population will facilitate clinical management and rational therapeutic development. OBJECTIVE: To determine correlates of benefit from CombiDT therapy in patients with BRAFi-refractory metastatic melanoma. DESIGN, SETTING, AND PARTICIPANTS: Single-center, single-arm, open-label phase 2 trial of CombiDT treatment in patients with BRAF V600 metastatic melanoma resistant to BRAFi monotherapy conducted between September 2012 and October 2014 at the University of Texas MD Anderson Cancer Center. Key eligibility criteria for participants included BRAF V600 metastatic melanoma, prior BRAFi monotherapy, measurable disease (RECIST 1.1), and tumor accessible for biopsy. INTERVENTIONS: Patients were treated with dabrafenib (150 mg, twice daily) and trametinib (2 mg/d) continuously until disease progression or intolerance. All participants underwent a mandatory baseline biopsy, and optional biopsy specimens were obtained on treatment and at disease progression. Whole-exome sequencing, reverse transcription polymerase chain reaction analysis for BRAF splicing, RNA sequencing, and immunohistochemical analysis were performed on tumor samples, and blood was analyzed for levels of circulating BRAF V600. MAIN OUTCOMES AND MEASURES: The primary end point was overall response rate (ORR). Progression-free survival (PFS) and overall survival (OS) were secondary clinical end points. RESULTS: A total of 28 patients were screened, and 23 enrolled. Among evaluable patients, the confirmed ORR was 10%; disease control rate (DCR) was 45%, and median PFS was 13 weeks. Clinical benefit was associated with duration of prior BRAFi therapy greater than 6 months (DCR, 73% vs 11% for ≤6 months; P = .02) and decrease in circulating BRAF V600 at day 8 of cycle 1 (DCR, 75% vs 18% for no decrease; P = .02) but not with pretreatment mitogen-activated protein kinase (MAPK) pathway mutations or activation. Biopsy specimens obtained during treatment demonstrated that CombiDT therapy failed to achieve significant MAPK pathway inhibition or immune infiltration in most patients. CONCLUSIONS AND RELEVANCE: The baseline presence of MAPK pathway alterations was not associated with benefit from CombiDT in patients with BRAFi-refractory metastatic melanoma. Failure to inhibit the MAPK pathway provides a likely explanation for the limited clinical benefit of CombiDT in this setting. Circulating BRAF V600 is a promising early biomarker of clinical response. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT01619774.


Subject(s)
Antineoplastic Agents/therapeutic use , MAP Kinase Signaling System/genetics , Melanoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Skin Neoplasms/drug therapy , Adaptor Proteins, Signal Transducing/metabolism , Adult , B7-H1 Antigen/metabolism , CD8 Antigens/metabolism , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Humans , Imidazoles/administration & dosage , Immunohistochemistry , Male , Melanoma/genetics , Melanoma/immunology , Melanoma/secondary , Middle Aged , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Oximes/administration & dosage , Phosphorylation , Prospective Studies , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Ribosomal Protein S6/metabolism , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Treatment Outcome
14.
Nat Biotechnol ; 32(7): 644-52, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24952901

ABSTRACT

Molecular profiling of tumors promises to advance the clinical management of cancer, but the benefits of integrating molecular data with traditional clinical variables have not been systematically studied. Here we retrospectively predict patient survival using diverse molecular data (somatic copy-number alteration, DNA methylation and mRNA, microRNA and protein expression) from 953 samples of four cancer types from The Cancer Genome Atlas project. We find that incorporating molecular data with clinical variables yields statistically significantly improved predictions (FDR < 0.05) for three cancers but those quantitative gains were limited (2.2-23.9%). Additional analyses revealed little predictive power across tumor types except for one case. In clinically relevant genes, we identified 10,281 somatic alterations across 12 cancer types in 2,928 of 3,277 patients (89.4%), many of which would not be revealed in single-tumor analyses. Our study provides a starting point and resources, including an open-access model evaluation platform, for building reliable prognostic and therapeutic strategies that incorporate molecular data.


Subject(s)
Biomarkers, Tumor/genetics , DNA, Neoplasm/genetics , Genetic Predisposition to Disease/genetics , Neoplasms/genetics , Neoplasms/mortality , Proteome/genetics , Survival Analysis , Databases, Genetic , Genetic Markers/genetics , Genetic Predisposition to Disease/epidemiology , Humans , Neoplasms/classification , Prevalence , Risk Assessment
SELECTION OF CITATIONS
SEARCH DETAIL