93 research outputs found

    TC--A Perennial Athletic Power

    Get PDF

    Comprehensive serial molecular profiling of an “N of 1” exceptional non-responder with metastatic prostate cancer progressing to small cell carcinoma on treatment

    Get PDF
    Abstract Importance Small cell carcinoma/neuroendocrine prostate cancer (NePC) is a lethal, poorly understood prostate cancer (PCa) subtype. Controversy exists about the origin of NePC in this setting. Objective To molecularly profile archived biopsy specimens from a case of early-onset PCa that rapidly progressed to NePC to identify drivers of the aggressive course and mechanisms of NePC origin and progression. Design, setting, and participants A 47-year-old patient presented with metastatic prostatic adenocarcinoma (Gleason score 9). After a 6-month response to androgen deprivation therapy, the patient developed jaundice and liver biopsy revealed exclusively NePC. Targeted next generation sequencing (NGS) from formalin-fixed paraffin-embedded (FFPE)-isolated DNA was performed from the diagnostic prostate biopsy and the liver biopsy at progression. Intervention Androgen deprivation therapy for adenocarcinoma followed by multiagent chemotherapy for NePC. Main outcomes and measures Identification of the mutational landscape in primary adenocarcinoma and NePC liver metastasis. Whether the NePC arose independently or was derived from the primary adenocarcinoma was considered based on mutational profiles. Results A deleterious somatic SMAD4 L535fs variant was present in both prostate and liver specimens; however, a TP53 R282W mutation was exclusively enriched in the liver specimen. Copy number analysis identified concordant, low-level alterations in both specimens, with focal MYCL amplification and homozygous PTEN, RB1, and MAP2K4 losses identified exclusively in the NePC specimen. Integration with published genomic profiles identified MYCL as a recurrently amplified in NePC. Conclusions and relevance NGS of routine biopsy samples from an exceptional non-responder identified SMAD4 as a driver of the aggressive course and supports derivation of NePC from primary adenocarcinoma (transdifferentiation).http://deepblue.lib.umich.edu/bitstream/2027.42/113670/1/13045_2015_Article_204.pd

    Rapid, ultra low coverage copy number profiling of cell-free DNA as a precision oncology screening strategy.

    Get PDF
    Current cell-free DNA (cfDNA) next generation sequencing (NGS) precision oncology workflows are typically limited to targeted and/or disease-specific applications. In advanced cancer, disease burden and cfDNA tumor content are often elevated, yielding unique precision oncology opportunities. We sought to demonstrate the utility of a pan-cancer, rapid, inexpensive, whole genome NGS of cfDNA approach (PRINCe) as a precision oncology screening strategy via ultra-low coverage (~0.01x) tumor content determination through genome-wide copy number alteration (CNA) profiling. We applied PRINCe to a retrospective cohort of 124 cfDNA samples from 100 patients with advanced cancers, including 76 men with metastatic castration-resistant prostate cancer (mCRPC), enabling cfDNA tumor content approximation and actionable focal CNA detection, while facilitating concordance analyses between cfDNA and tissue-based NGS profiles and assessment of cfDNA alteration associations with mCRPC treatment outcomes. Therapeutically relevant focal CNAs were present in 42 (34%) cfDNA samples, including 36 of 93 (39%) mCRPC patient samples harboring AR amplification. PRINCe identified pre-treatment cfDNA CNA profiles facilitating disease monitoring. Combining PRINCe with routine targeted NGS of cfDNA enabled mutation and CNA assessment with coverages tuned to cfDNA tumor content. In mCRPC, genome-wide PRINCe cfDNA and matched tissue CNA profiles showed high concordance (median Pearson correlation = 0.87), and PRINCe detectable AR amplifications predicted reduced time on therapy, independent of therapy type (Kaplan-Meier log-rank test, chi-square = 24.9, p < 0.0001). Our screening approach enables robust, broadly applicable cfDNA-based precision oncology for patients with advanced cancer through scalable identification of therapeutically relevant CNAs and pre-/post-treatment genomic profiles, enabling cfDNA- or tissue-based precision oncology workflow optimization

    Discordant and heterogeneous clinically relevant genomic alterations in circulating tumor cells vs plasma DNA from men with metastatic castration resistant prostate cancer

    Full text link
    Circulating tumor cell (CTC) and cellĂą free (cf) DNAĂą based genomic alterations are increasingly being used for clinical decisionĂą making in oncology. However, the concordance and discordance between paired CTC and cfDNA genomic profiles remain largely unknown. We performed comparative genomic hybridization (CGH) on CTCs and cfDNA, and lowĂą pass whole genome sequencing (lpWGS) on cfDNA to characterize genomic alterations (CNA) and tumor content in two independent prospective studies of 93 men with mCRPC treated with enzalutamide/abiraterone, or radiumĂą 223. Comprehensive analysis of 69 patient CTCs and 72 cfDNA samples from 93 men with mCRPC, including 64 paired samples, identified common concordant gains in FOXA1, AR, and MYC, and losses in BRCA1, PTEN, and RB1 between CTCs and cfDNA. Concordant PTEN loss and discordant BRCA2 gain were associated with significantly worse outcomes in Epic ARĂą V7 negative men with mCRPC treated with abiraterone/enzalutamide. We identified and externally validated CTCĂą specific genomic alternations that were discordant in paired cfDNA, even in samples with high tumor content. These CTC/cfDNAĂą discordant regions included key genomic regulators of lineage plasticity, osteomimicry, and cellular differentiation, including MYCN gain in CTCs (31%) that was rarely detected in cfDNA. CTC MYCN gain was associated with poor clinical outcomes in ARĂą V7 negative men and small cell transformation. In conclusion, we demonstrated concordance of multiple genomic alterations across CTC and cfDNA platforms; however, some genomic alterations displayed substantial discordance between CTC DNA and cfDNA despite the use of identical copy number analysis methods, suggesting tumor heterogeneity and divergent evolution associated with poor clinical outcomes.Peer Reviewedhttps://deepblue.lib.umich.edu/bitstream/2027.42/153751/1/gcc22824.pdfhttps://deepblue.lib.umich.edu/bitstream/2027.42/153751/2/gcc22824_am.pd

    The molecular and cellular origin of human prostate cancer

    Get PDF
    Prostate cancer is the most commonly diagnosed male malignancy. Despite compelling epidemiology, there are no definitive aetiological clues linking development to frequency. Pre-malignancies such as proliferative inflammatory atrophy (PIA) and prostatic intraepithelial neoplasia (PIN) yield insights into the initiating events of prostate cancer, as they supply a background "field" for further transformation. An inflammatory aetiology, linked to recurrent prostatitis, and heterologous signalling from reactive stroma and infiltrating immune cells may result in cytokine addiction of cancer cells, including a tumour-initiating population also known as cancer stem cells (CSCs). In prostate tumours, the background mutational rate is rarely exceeded, but genetic change via profound sporadic chromosomal rearrangements results in copy number variations and aberrant gene expression. In cancer, dysfunctional differentiation is imposed upon the normal epithelial lineage, with disruption/disappearance of the basement membrane, loss of the contiguous basal cell layer and expansion of the luminal population. An initiating role for androgen receptor (AR) is attractive, due to the luminal phenotype of the tumours, but alternatively a pool of CSCs, which express little or no AR, has also been demonstrated. Indolent and aggressive tumours may also arise from different stem or progenitor cells. Castrate resistant prostate cancer (CRPC) remains the inevitable final stage of disease following treatment. Time-limited effectiveness of second-generation anti-androgens, and the appearance of an AR-neuroendocrine phenotype imply that metastatic disease is reliant upon the plasticity of the CSC population, and indeed CSC gene expression profiles are most closely related to those identified in CRPCs

    Crizotinib in patients with tumors harboring ALK or ROS1 rearrangements in the NCI-MATCH trial.

    Get PDF
    The NCI-MATCH was designed to characterize the efficacy of targeted therapies in histology-agnostic driver mutation-positive malignancies. Sub-protocols F and G were developed to evaluate the role of crizotinib in rare tumors that harbored either ALK or ROS1 rearrangements. Patients with malignancies that progressed following at least one prior systemic therapy were accrued to the NCI-MATCH for molecular profiling, and those with actionable ALK or ROS1 rearrangements were offered participation in sub-protocols F or G, respectively. There were five patients who enrolled on Arm F (ALK) and four patients on Arm G (ROS1). Few grade 3 or 4 toxicities were noted, including liver test abnormalities, and acute kidney injury. For sub-protocol F (ALK), the response rate was 50% (90% CI 9.8-90.2%) with one complete response among the 4 eligible patients. The median PFS was 3.8 months, and median OS was 4.3 months. For sub-protocol G (ROS1) the response rate was 25% (90% CI 1.3-75.1%). The median PFS was 4.3 months, and median OS 6.2 months. Data from 3 commercial vendors showed that the prevalence of ALK and ROS1 rearrangements in histologies other than non-small cell lung cancer and lymphoma was rare (0.1% and 0.4% respectively). We observed responses to crizotinib which met the primary endpoint for ALK fusions, albeit in a small number of patients. Despite the limited accrual, some of the patients with these oncogenic fusions can respond to crizotinib which may have a therapeutic role in this setting

    Precision Oncology Opportunities And Disease Insights From Next-Generation-Sequencing Profiling Of Routine Clinical Biospecimens

    Full text link
    Rapid technological developments in next-generation sequencing (NGS) and inter-institutional collaborations including The Cancer Genome Atlas (TCGA) have enabled comprehensive characterization of the genomic, transcriptomic, and epigenetic landscapes from bulk tissue specimens in a wide range of cancers. Emerging work has focused on scaling NGS-based profiling strategies to guide precision medicine approaches in clinical oncology using routine clinical biospecimens such as formalin-fixed, paraffin-embedded (FFPE) tissue or less-invasive liquid (e.g., blood or urine) samples. Technical challenges associated with limited tumor lesion size, low nucleic acid quantities, disease-specificity applications, and disease and histological heterogeneity present hurdles to widespread adoption and utility of extant NGS-based precision oncology approaches. Here, several analytical advances are described supporting democratization of precision oncology approaches from clinical tissue and liquid biospecimens, while revealing disease insights and important clinical considerations in the context of both localized and advanced (including multifocal and/or heterogeneous) disease. First, development and validation of a targeted DNA and RNA NGS assay compatible with small quantities of DNA and RNA isolated from routine, archived FFPE tissue specimens is described. This assay, targeting recurrently mutated oncogenic hotspots, tumor suppressors, copy-number-altered genes, and recurrent gene fusions is applied to a cohort of >300 FFPE tissue samples, revealing high sensitivity with orthogonal molecular diagnostic assays for BRAF, KRAS, and EGFR oncogenic alterations. Second, I describe a rapid, inexpensive, low-pass cell-free DNA (cfDNA) whole-genome sequencing (WGS) copy-number profiling approach, including a novel heuristic tumor content approximation method, capable of establishing genome-wide copy-number profiles from 0.01-0.1x sequencing coverage. Application of our approach in plasma samples from patients with advanced cancer with matched comprehensive tissue NGS revealed high concordance with tissue-based molecular profiles, while highlighting important areas of potential utility from noninvasive profiling of overall disease burden. Third, I describe the systematic assessment of expression-based molecular subtypes in histologically heterogenous bladder cancers, revealing robust identification of basal/luminal molecular subtypes in a cohort of >100 bladder cancer cell lines and tumor tissue specimens, and recapitulation of basal/luminal subtypes in >400 samples profiled by TCGA using selected marker subsets. Importantly, I describe divergent expression profiles in the context of shared genomic alterations for individual histologically divergent tumor components from the same tumor, confounding proposed clinical utility of expression-based subtypes for disease prediction and prognosis. Fourth, I describe the development of a targeted RNAseq panel capable of assessing major transcriptional programs and disease biomarkers across the full spectrum of prostate cancer disease, while deriving commercially available prognostic scores that show limited robustness to disease multifocality. Lastly, I describe extensions of our cfDNA WGS approach to urine cfDNA samples from patients with advanced cancer, while exploring the potential utility of pairing described analytic tools with existing and emerging molecular profiling strategies to improve our understanding of disease biology and maximize clinical utility.PHDBioinformaticsUniversity of Michigan, Horace H. Rackham School of Graduate Studieshttps://deepblue.lib.umich.edu/bitstream/2027.42/140890/1/hovelson_1.pd
    • 

    corecore