Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Thyroid ; 30(9): 1384-1389, 2020 09.
Article in English | MEDLINE | ID: mdl-32292131

ABSTRACT

Background: Anaplastic thyroid carcinoma (ATC) remains one of the most challenging malignancies to treat in the modern era. Most patients present with or develop recurrent/metastatic incurable disease with poor response rates to conventional chemotherapy, and life expectancy is short. Next-generation sequencing (NGS) can be leveraged in ATC to identify oncogenic alterations that can be targeted with molecularly specific therapy, offering new effective treatment options to a subset of patients. Patient Findings: A 73-year-old man presenting with locally advanced papillary thyroid carcinoma containing a minor component of ATC was treated with surgery and iodine-131. He developed biopsy-confirmed ATC distant metastases that progressed on cytotoxic chemotherapy. NGS revealed several alterations, including a CCDC6-RET gene fusion. The patient enrolled in LIBRETTO-001, a phase I/II trial of the potent and specific RET inhibitor, LOXO-292. The patient tolerated LOXO-292 well and experienced a deep and durable partial response, ongoing beyond 19 months. Conclusion: This clinically significant response achieved with LOXO-292 in a patient with a CCDC6-RET fusion-positive ATC who had exhausted conventional treatment options highlights the importance of conducting tumor genomic profiling in patients with ATC to identify uncommon but actionable genomic alterations, such as RET gene fusions.


Subject(s)
Proto-Oncogene Proteins c-ret/metabolism , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Carcinoma, Anaplastic/pathology , Aged , Antineoplastic Agents/pharmacology , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , High-Throughput Nucleotide Sequencing , Humans , Male , Neoplasm Metastasis , Oncogenes , Positron Emission Tomography Computed Tomography , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins c-ret/genetics , Pyrazoles/pharmacology , Pyridines/pharmacology , Thyroid Cancer, Papillary/genetics , Thyroid Carcinoma, Anaplastic/drug therapy , Thyroid Neoplasms/pathology , Treatment Outcome
2.
Nat Genet ; 52(4): 371-377, 2020 04.
Article in English | MEDLINE | ID: mdl-32203465

ABSTRACT

Brain metastases from lung adenocarcinoma (BM-LUAD) frequently cause patient mortality. To identify genomic alterations that promote brain metastases, we performed whole-exome sequencing of 73 BM-LUAD cases. Using case-control analyses, we discovered candidate drivers of brain metastasis by identifying genes with more frequent copy-number aberrations in BM-LUAD compared to 503 primary LUADs. We identified three regions with significantly higher amplification frequencies in BM-LUAD, including MYC (12 versus 6%), YAP1 (7 versus 0.8%) and MMP13 (10 versus 0.6%), and significantly more frequent deletions in CDKN2A/B (27 versus 13%). We confirmed that the amplification frequencies of MYC, YAP1 and MMP13 were elevated in an independent cohort of 105 patients with BM-LUAD. Functional assessment in patient-derived xenograft mouse models validated the notion that MYC, YAP1 or MMP13 overexpression increased the incidence of brain metastasis. These results demonstrate that somatic alterations contribute to brain metastases and that genomic sequencing of a sufficient number of metastatic tumors can reveal previously unknown metastatic drivers.


Subject(s)
Adenocarcinoma of Lung/genetics , Brain Neoplasms/genetics , Lung Neoplasms/genetics , Neoplasm Metastasis/genetics , Adenocarcinoma of Lung/pathology , Animals , Brain Neoplasms/pathology , Case-Control Studies , Cell Line , DNA Copy Number Variations/genetics , Female , Genes, myc/genetics , Genomics/methods , HEK293 Cells , Humans , Lung Neoplasms/pathology , Male , Matrix Metalloproteinase 13/genetics , Mice , Mice, Nude , Mutation/genetics , Neoplasm Metastasis/pathology , Transcription Factors/genetics , Exome Sequencing
3.
J Mol Diagn ; 21(4): 632-645, 2019 07.
Article in English | MEDLINE | ID: mdl-31026600

ABSTRACT

The use of liquid biopsies to identify driver mutations in patients with solid tumors holds great promise for performing targeted therapy selection, monitoring disease progression, and detecting treatment resistance mechanisms. We describe herein the development and clinical validation of a 28-gene cell-free DNA panel that targets the most common genetic alterations in solid tumors. Bioinformatic and variant filtering solutions were developed to improve test sensitivity and specificity. The panel and these tools were used to analyze commercially available controls, allowing establishment of a limit of detection allele fraction cutoff of 0.25%, with 100% (95% CI, 81.5%-100%) specificity and 89.8% (95% CI, 81.0%-94.9%) sensitivity. In addition, we analyzed a total of 163 blood samples from patients with metastatic cancer (n = 123) and demonstrated a >90% sensitivity for detecting previously identified expected mutations. Longitudinal monitoring of patients revealed a strong correlation of variant allele frequency changes and clinical outcome. Additional clinically relevant information included identification of resistance mutations in patients receiving targeted treatment and detection of complex patterns of mutational heterogeneity. Achieving lower limits of detection will require additional improvements to molecular barcoding; however, these data strongly support clinical implementation of cell-free DNA panels in advanced cancer patients.


Subject(s)
Biomarkers, Tumor , Cell-Free Nucleic Acids , Circulating Tumor DNA , Genetic Testing , Liquid Biopsy , Neoplasms/diagnosis , Neoplasms/genetics , Adult , Aged , Aged, 80 and over , DNA Copy Number Variations , Disease Progression , Female , Genetic Testing/methods , Genetic Testing/standards , Humans , In Situ Hybridization, Fluorescence , Liquid Biopsy/methods , Liquid Biopsy/standards , Male , Middle Aged , Neoplasm Staging , Reproducibility of Results
4.
Oncologist ; 24(10): 1356-1367, 2019 10.
Article in English | MEDLINE | ID: mdl-30926674

ABSTRACT

BACKGROUND: Adenoid cystic carcinoma (ACC) is an aggressive salivary gland malignancy without effective systemic therapies. Delineation of molecular profiles in ACC has led to an increased number of biomarker-stratified clinical trials; however, the clinical utility and U.S.-centric financial sustainability of integrated next-generation sequencing (NGS) in routine practice has, to our knowledge, not been assessed. MATERIALS AND METHODS: In our practice, NGS genotyping was implemented at the discretion of the primary clinician. We combined NGS-based mutation and fusion detection, with MYB break-apart fluorescent in situ hybridization (FISH) and MYB immunohistochemistry. Utility was defined as the fraction of patients with tumors harboring alterations that are potentially amenable to targeted therapies. Financial sustainability was assessed using the fraction of global reimbursement. RESULTS: Among 181 consecutive ACC cases (2011-2018), prospective genotyping was performed in 11% (n = 20/181; n = 8 nonresectable). Testing identified 5/20 (25%) NOTCH1 aberrations, 6/20 (30%) MYB-NFIB fusions (all confirmed by FISH), and 2/20 (10%) MYBL1-NFIB fusions. Overall, these three alterations (MYB/MYBL1/NOTCH1) made up 65% of patients, and this subset had a more aggressive course with significantly shorter progression-free survival. In 75% (n = 6/8) of nonresectable patients, we detected potentially actionable alterations. Financial analysis of the global charges, including NGS codes, indicated 63% reimbursement, which is in line with national (U.S.-based) and international levels of reimbursement. CONCLUSION: Prospective routine clinical genotyping in ACC can identify clinically relevant subsets of patients and is approaching financial sustainability. Demonstrating clinical utility and financial sustainability in an orphan disease (ACC) requires a multiyear and multidimensional program. IMPLICATIONS FOR PRACTICE: Delineation of molecular profiles in adenoid cystic carcinoma (ACC) has been accomplished in the research setting; however, the ability to identify relevant patient subsets in clinical practice has not been assessed. This work presents an approach to perform integrated molecular genotyping of patients with ACC with nonresectable, recurrent, or systemic disease. It was determined that 75% of nonresectable patients harbor potentially actionable alterations and that 63% of charges are reimbursed. This report outlines that orphan diseases such as ACC require a multiyear, multidimensional program to demonstrate utility in clinical practice.


Subject(s)
Carcinoma, Adenoid Cystic/diagnosis , High-Throughput Nucleotide Sequencing/methods , Molecular Diagnostic Techniques/methods , Female , Humans , Male , Prospective Studies , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...