Try a new search

Format these results:

Searched for:

person:dsh279

in-biosketch:yes

Total Results:

29


InterMEL: An international biorepository and clinical database to uncover predictors of survival in early-stage melanoma

Orlow, Irene; Sadeghi, Keimya D; Edmiston, Sharon N; Kenney, Jessica M; Lezcano, Cecilia; Wilmott, James S; Cust, Anne E; Scolyer, Richard A; Mann, Graham J; Lee, Tim K; Burke, Hazel; Jakrot, Valerie; Shang, Ping; Ferguson, Peter M; Boyce, Tawny W; Ko, Jennifer S; Ngo, Peter; Funchain, Pauline; Rees, Judy R; O'Connell, Kelli; Hao, Honglin; Parrish, Eloise; Conway, Kathleen; Googe, Paul B; Ollila, David W; Moschos, Stergios J; Hernando, Eva; Hanniford, Douglas; Argibay, Diana; Amos, Christopher I; Lee, Jeffrey E; Osman, Iman; Luo, Li; Kuan, Pei-Fen; Aurora, Arshi; Gould Rothberg, Bonnie E; Bosenberg, Marcus W; Gerstenblith, Meg R; Thompson, Cheryl; Bogner, Paul N; Gorlov, Ivan P; Holmen, Sheri L; Brunsgaard, Elise K; Saenger, Yvonne M; Shen, Ronglai; Seshan, Venkatraman; Nagore, Eduardo; Ernstoff, Marc S; Busam, Klaus J; Begg, Colin B; Thomas, Nancy E; Berwick, Marianne
INTRODUCTION:We are conducting a multicenter study to identify classifiers predictive of disease-specific survival in patients with primary melanomas. Here we delineate the unique aspects, challenges, and best practices for optimizing a study of generally small-sized pigmented tumor samples including primary melanomas of at least 1.05mm from AJTCC TNM stage IIA-IIID patients. We also evaluated tissue-derived predictors of extracted nucleic acids' quality and success in downstream testing. This ongoing study will target 1,000 melanomas within the international InterMEL consortium. METHODS:Following a pre-established protocol, participating centers ship formalin-fixed paraffin embedded (FFPE) tissue sections to Memorial Sloan Kettering Cancer Center for the centralized handling, dermatopathology review and histology-guided coextraction of RNA and DNA. Samples are distributed for evaluation of somatic mutations using next gen sequencing (NGS) with the MSK-IMPACTTM assay, methylation-profiling (Infinium MethylationEPIC arrays), and miRNA expression (Nanostring nCounter Human v3 miRNA Expression Assay). RESULTS:Sufficient material was obtained for screening of miRNA expression in 683/685 (99%) eligible melanomas, methylation in 467 (68%), and somatic mutations in 560 (82%). In 446/685 (65%) cases, aliquots of RNA/DNA were sufficient for testing with all three platforms. Among samples evaluated by the time of this analysis, the mean NGS coverage was 249x, 59 (18.6%) samples had coverage below 100x, and 41/414 (10%) failed methylation QC due to low intensity probes or insufficient Meta-Mixed Interquartile (BMIQ)- and single sample (ss)- Noob normalizations. Six of 683 RNAs (1%) failed Nanostring QC due to the low proportion of probes above the minimum threshold. Age of the FFPE tissue blocks (p<0.001) and time elapsed from sectioning to co-extraction (p = 0.002) were associated with methylation screening failures. Melanin reduced the ability to amplify fragments of 200bp or greater (absent/lightly pigmented vs heavily pigmented, p<0.003). Conversely, heavily pigmented tumors rendered greater amounts of RNA (p<0.001), and of RNA above 200 nucleotides (p<0.001). CONCLUSION:Our experience with many archival tissues demonstrates that with careful management of tissue processing and quality control it is possible to conduct multi-omic studies in a complex multi-institutional setting for investigations involving minute quantities of FFPE tumors, as in studies of early-stage melanoma. The study describes, for the first time, the optimal strategy for obtaining archival and limited tumor tissue, the characteristics of the nucleic acids co-extracted from a unique cell lysate, and success rate in downstream applications. In addition, our findings provide an estimate of the anticipated attrition that will guide other large multicenter research and consortia.
PMCID:10069769
PMID: 37011054
ISSN: 1932-6203
CID: 5463642

The State of Melanoma: Emergent Challenges and Opportunities

Atkins, Michael B; Curiel-Lewandrowski, Clara; Fisher, David E; Swetter, Susan M; Tsao, Hensin; Aguirre-Ghiso, Julio A; Soengas, Maria S; Weeraratna, Ashani T; Flaherty, Keith T; Herlyn, Meenhard; Sosman, Jeffrey A; Tawbi, Hussein A; Pavlick, Anna C; Cassidy, Pamela B; Chandra, Sunandana; Chapman, Paul B; Daud, Adil; Eroglu, Zeynep; Ferris, Laura K; Fox, Bernard A; Gerhsenwald, Jeffrey E; Gibney, Geoffrey T; Grossman, Douglas; Hanks, Brent A; Hanniford, Douglas; Hernando, Eva; Jeter, Joanne M; Johnson, Douglas B; Khleif, Samir N; Kirkwood, John M; Leachman, Sancy A; Mays, Darren; Nelson, Kelly C; Sondak, Vernon K; Sullivan, Ryan J; Merlino, Glenn
Five years ago, the Melanoma Research Foundation (MRF) conducted an assessment of the challenges and opportunities facing the melanoma research community and patients with melanoma. Since then, remarkable progress has been made on both the basic and clinical research fronts. However, the incidence, recurrence and death rates for melanoma remain unacceptably high and significant challenges remain. Hence, the MRF Scientific Advisory Council and Breakthrough Consortium, a group that includes clinicians and scientists, reconvened to facilitate intensive discussions on thematic areas essential to melanoma researchers and patients alike - prevention, detection, diagnosis, metastatic dormancy and progression, response and resistance to targeted and immune-based therapy, and the clinical consequences of COVID-19 for melanoma patients and providers. These extensive discussions helped to crystalize our understanding of the challenges and opportunities facing the broader melanoma community today. In this report, we discuss the progress made since the last MRF assessment, comment on what remains to be overcome and offer recommendations for the best path forward.
PMID: 33414132
ISSN: 1557-3265
CID: 4739332

Functional analysis of RPS27 mutations and expression in melanoma

Floristán, Alfredo; Morales, Leah; Hanniford, Douglas; Martinez, Carlos; Castellano-Sanz, Elena; Dolgalev, Igor; Ulloa-Morales, Alejandro; Vega-Saenz de Miera, Eleazar; Moran, Una; Darvishian, Farbod; Osman, Iman; Kirchhoff, Tomas; Hernando, Eva
Next-generation sequencing has enabled genetic and genomic characterization of melanoma to an unprecedent depth. However, the high mutational background plus the limited deep-coverage whole-genome sequencing performed on cutaneous melanoma samples, make difficult the identification of novel driver mutations. We sought to explore the somatic mutation portfolio in exonic and gene regulatory regions in human melanoma samples, for which we performed targeted sequencing of tumors and matched germline DNA samples from 89 melanoma patients, identifying known and novel recurrent mutations. Two recurrent mutations found in the RPS27 promoter associated with decreased RPS27 mRNA levels in vitro. Data mining and IHC analyses revealed a bimodal pattern of RPS27 expression in melanoma, with RPS27-low patients displaying worse prognosis. In vitro characterization of RPS27-high and -low melanoma cell lines, as well as loss-of-function experiments, demonstrated that high RPS27 status provides increased proliferative and invasive capacities, while low RPS27 confers survival advantage in low-attachment and resistance to therapy. Additionally, we demonstrate that 10 other cancer types harbor bimodal RPS27 expression and in those, similarly to melanoma, RPS27-low expression associates with worse clinical outcomes. RPS27 promoter mutation could thus represent a mechanism of gene expression modulation in melanoma patients, which may have prognostic and predictive implications.
PMID: 31663663
ISSN: 1755-148x
CID: 4162282

circSamd4 represses myogenic transcriptional activity of PUR proteins

Pandey, Poonam R; Yang, Jen-Hao; Tsitsipatis, Dimitrios; Panda, Amaresh C; Noh, Ji Heon; Kim, Kyoung Mi; Munk, Rachel; Nicholson, Thomas; Hanniford, Douglas; Argibay, Diana; Yang, Xiaoling; Martindale, Jennifer L; Chang, Ming-Wen; Jones, Simon W; Hernando, Eva; Sen, Payel; De, Supriyo; Abdelmohsen, Kotb; Gorospe, Myriam
By interacting with proteins and nucleic acids, the vast family of mammalian circRNAs is proposed to influence many biological processes. Here, RNA sequencing analysis of circRNAs differentially expressed during myogenesis revealed that circSamd4 expression increased robustly in mouse C2C12 myoblasts differentiating into myotubes. Moreover, silencing circSamd4, which is conserved between human and mouse, delayed myogenesis and lowered the expression of myogenic markers in cultured myoblasts from both species. Affinity pulldown followed by mass spectrometry revealed that circSamd4 associated with PURA and PURB, two repressors of myogenesis that inhibit transcription of the myosin heavy chain (MHC) protein family. Supporting the hypothesis that circSamd4 might complex with PUR proteins and thereby prevent their interaction with DNA, silencing circSamd4 enhanced the association of PUR proteins with the Mhc promoter, while overexpressing circSamd4 interfered with the binding of PUR proteins to the Mhc promoter. These effects were abrogated when using a mutant circSamd4 lacking the PUR binding site. Our results indicate that the association of PUR proteins with circSamd4 enhances myogenesis by contributing to the derepression of MHC transcription.
PMID: 31980816
ISSN: 1362-4962
CID: 4274142

Epigenetic Silencing of CDR1as Drives IGF2BP3-Mediated Melanoma Invasion and Metastasis

Hanniford, Douglas; Ulloa-Morales, Alejandro; Karz, Alcida; Berzoti-Coelho, Maria Gabriela; Moubarak, Rana S; Sánchez-Sendra, Beatriz; Kloetgen, Andreas; Davalos, Veronica; Imig, Jochen; Wu, Pamela; Vasudevaraja, Varshini; Argibay, Diana; Lilja, Karin; Tabaglio, Tommaso; Monteagudo, Carlos; Guccione, Ernesto; Tsirigos, Aristotelis; Osman, Iman; Aifantis, Iannis; Hernando, Eva
Metastasis is the primary cause of death of cancer patients. Dissecting mechanisms governing metastatic spread may uncover important tumor biology and/or yield promising therapeutic insights. Here, we investigated the role of circular RNAs (circRNA) in metastasis, using melanoma as a model aggressive tumor. We identified silencing of cerebellar degeneration-related 1 antisense (CDR1as), a regulator of miR-7, as a hallmark of melanoma progression. CDR1as depletion results from epigenetic silencing of LINC00632, its originating long non-coding RNA (lncRNA) and promotes invasion in vitro and metastasis in vivo through a miR-7-independent, IGF2BP3-mediated mechanism. Moreover, CDR1as levels reflect cellular states associated with distinct therapeutic responses. Our study reveals functional, prognostic, and predictive roles for CDR1as and expose circRNAs as key players in metastasis.
PMID: 31935372
ISSN: 1878-3686
CID: 4263262

PRC2-mediated silencing of circRNA CDR1as drives miR-7- independent melanoma metastasis [Meeting Abstract]

Hanniford, D; Moubarak, R; Imig, J; Ulloa, A; Sendra, B S; Karz, A; Osman, I; Aifantis, I; Hernando, E
Circular RNAs are a novel class of non-coding RNAs with functions that remain poorly characterized in normal and pathological conditions. CDR1as is a non-canonical circRNA observed to act as a sponge for miR-7 in brain tissues. Analysis of RNA-seq data of melanocytes and melanoma cell lines and short-term cultures revealed loss of CDR1as expression as a hallmark of melanoma cells. We confirmed silencing of CDR1as in melanoma cells and tissues by RT-qPCR using divergent primers. Clinically, we observed CDR1as loss associated with metastatic progression and poor patient outcomes in a cohort of fresh-frozen melanoma tissue samples. Depletion of CDR1as in melanoma cell lines enhanced invasion in vitro and lung metastasis in vivo, demonstrating functional significance of CDR1as silencing. Surprisingly, CDR1as depletion had no clear effect on miR-7 activity in melanoma cells, and miR-7 inhibition was insufficient to rescue CDR1as silencing-induced invasion. Moreover, GSEA analyses of proteomic profiling of melanoma cells depleted of CDR1as revealed reductions of proteins involved in oxidative phosphorylation (OXPHOS) and mitochondrial function, suggesting CDR1as loss may alter metabolism of melanoma cells. Mining of CLIP-Seq data sets and subsequent RIP-PCR revealed direct interactions of CDR1as with the IGF2BP family of proteins and TAR
EMBASE:618565797
ISSN: 1538-7445
CID: 2752502

Mutation burden as a potential prognostic marker of melanoma progression and survival [Meeting Abstract]

Simpson, D; Ferguson, R; Martinez, C N; Kazlow, E; Moran, U; Heguy, A; Hanniford, D; Hernando, E; Osman, I; Kirchhoff, T
Background: Recently, tumor mutation burden (TMB) has been shown to increase the presentation of neoantigens that stimulate immune tumor recognition, resulting in improved immunotherapy (IT) outcomes in melanoma and other cancers. As melanoma is highly immunogenic, here we tested whether TMB associates with immune recognition during tumor progression, hence impacting melanoma overall survival (OS), independently of IT treatment. Methods: We have generated somatic mutation data from 314 IT-naive metastatic melanomas from The Cancer Genome Atlas (TCGA). In the TCGA cohort, TMB has been calculated for 210 genes (200GS) previously established from TMB studies of anti-CTLA4 and anti-PD1/PD-L1 IT. For validation, we have sequenced exonic regions of 20 genes (20GS) with the highest TMB among 200GS in 89 IT-naive metastatic melanomas ascertained at New York University Langone Medical Center. The TMB was defined using total number of somatic, non-synonymous mutations in either 200GS (TCGA discovery) or 20GS (validation), respectively. For discovery and validation cohorts, OS from primary diagnosis of samples with high TMB was compared against low TMB, using thresholds established in previous studies. Results: We found that total TMB predicts better OS (p = 0.03, HR = 2.64) in TCGA melanomas. Restricting the analysis only to the established 200GS, this association became more significant in all patients (p = 0.01, HR = 2.67) as well as in patients without IT (p = 0.01, HR = 2.67). In the validation stage of 89 melanomas without prior IT treatment, a high TMB in a subset of 20GS accurately determined favorable OS (p = 0.02, HR = 2.69) and confirmed TCGA observations from the 200GS. Conclusions: Here we show, for the first time, that in addition to IT, high TMB predicts more favorable OS in patients that never received IT, potentially serving as a novel marker of prognosis of melanoma and likely other immunogenic tumors at early stages. In addition, our study suggests that TMB test can be robust when applied to only a small subset of genes that trigger significantly higher immunogenicity. This may also eventually assist with accurate sub-selection of early stage patients likely to respond to IT regimens
EMBASE:617435426
ISSN: 0732-183x
CID: 2651092

MicroRNA-125a promotes resistance to BRAF inhibitors through suppression of the intrinsic apoptotic pathway

Koetz-Ploch, Lisa; Hanniford, Douglas; Dolgalev, Igor; Sokolova, Elena; Zhong, Judy; Diaz-Martinez, Marta; Bernstein, Emily; Darvishian, Farbod; Flaherty, Keith T; Chapman, Paul B; Tawbi, Hussein; Hernando, Eva
Melanoma patients with BRAFV600E -mutant tumors display striking responses to BRAF inhibitors (BRAFi); however, almost all invariably relapse with drug-resistant disease. Here we report that microRNA-125a (miR-125a) expression is upregulated in human melanoma cells and patient tissues upon acquisition of BRAFi resistance. We show that miR-125a induction confers resistance to BRAFV600E melanoma cells to BRAFi by directly suppressing pro-apoptotic components of the intrinsic apoptosis pathway, including BAK1 and MLK3. Apoptotic suppression and prolonged survival favor reactivation of the MAPK and AKT pathways by drug-resistant melanoma cells. We demonstrate that miR-125a inhibition suppresses the emergence of resistance to BRAFi and, in a subset of resistant melanoma cell lines, leads to partial drug re-sensitization. Finally, we show that miR-125a upregulation is mediated by TGFbeta signaling. In conclusion, the identification of this novel role for miR-125a in BRAFi resistance exposes clinically relevant mechanisms of melanoma cell survival that can be exploited therapeutically
PMCID:5411293
PMID: 28140520
ISSN: 1755-148x
CID: 2425092

A TGFbeta-miR-182-BRCA1 axis controls the mammary differentiation hierarchy

Martinez-Ruiz, Haydeliz; Illa-Bochaca, Irineu; Omene, Coral; Hanniford, Douglas; Liu, Qi; Hernando, Eva; Barcellos-Hoff, Mary Helen
Maintenance of mammary functional capacity during cycles of proliferation and regression depends on appropriate cell fate decisions of mammary progenitor cells to populate an epithelium consisting of secretory luminal cells and contractile myoepithelial cells. It is well established that transforming growth factor-beta (TGFbeta) restricts mammary epithelial cell proliferation and that sensitivity to TGFbeta is decreased in breast cancer. We show that TGFbeta also exerts control of mammary progenitor self-renewal and lineage commitment decisions by stringent regulation of breast cancer associated 1 (BRCA1), which controls stem cell self-renewal and lineage commitment. Either genetic depletion of Tgfb1 or transient blockade of TGFbeta increased self-renewal of mammary progenitor cells in mice, cultured primary mammary epithelial cells, and also skewed lineage commitment toward the myoepithelial fate. TGFbeta stabilized the abundance of BRCA1 by reducing the abundance of microRNA-182 (miR-182). Ectopic expression of BRCA1 or antagonism of miR-182 in cultured TGFbeta-deficient mammary epithelial cells restored luminal lineage commitment. These findings reveal that TGFbeta modulation of BRCA1 directs mammary epithelial cell fate and, because stem or progenitor cells are thought to be the cell of origin for aggressive breast cancer subtypes, suggest that TGFbeta dysregulation during tumorigenesis may promote distinct breast cancer subtypes.
PMCID:5619986
PMID: 27923913
ISSN: 1937-9145
CID: 2353502

SPROUTY-2 represses the epithelial phenotype of colon carcinoma cells via upregulation of ZEB1 mediated by ETS1 and miR-200/miR-150

Barbachano, A; Fernandez-Barral, A; Pereira, F; Segura, M F; Ordonez-Moran, P; Carrillo-de Santa Pau, E; Gonzalez-Sancho, J M; Hanniford, D; Martinez, N; Costales-Carrera, A; Real, F X; Palmer, H G; Rojas, J M; Hernando, E; Munoz, A
SPROUTY-2 (SPRY2) is a modulator of tyrosine kinase receptor signaling with receptor- and cell type-dependent inhibitory or enhancing effects. Studies on the action of SPRY2 in major cancers are conflicting and its role remains unclear. Here we have dissected SPRY2 action in human colon cancer. Global transcriptomic analyses show that SPRY2 downregulates genes encoding tight junction proteins such as claudin-7 and occludin and other cell-to-cell and cell-to-matrix adhesion molecules in human SW480-ADH colon carcinoma cells. Moreover, SPRY2 represses LLGL2/HUGL2, PATJ1/INADL and ST14, main regulators of the polarized epithelial phenotype, and ESRP1, an epithelial-to-mesenchymal transition (EMT) inhibitor. A key action of SPRY2 is the upregulation of the major EMT inducer ZEB1, as these effects are reversed by ZEB1 knock-down by means of RNA interference. Consistently, we found an inverse correlation between the expression level of claudin-7 and those of SPRY2 and ZEB1 in human colon tumors. Mechanistically, ZEB1 upregulation by SPRY2 results from the combined induction of ETS1 transcription factor and the repression of microRNAs (miR-200 family, miR-150) that target ZEB1 RNA. Moreover, SPRY2 increased AKT activation by epidermal growth factor, whereas AKT and also Src inhibition reduced the induction of ZEB1. Altogether, these data suggest that AKT and Src are implicated in SPRY2 action. Collectively, these results show a tumorigenic role of SPRY2 in colon cancer that is based on the dysregulation of tight junction and epithelial polarity master genes via upregulation of ZEB1. The dissection of the mechanism of action of SPRY2 in colon cancer cells is important to understand the upregulation of this gene in a subset of patients with this neoplasia that have poor prognosis.Oncogene advance online publication, 12 October 2015; doi:10.1038/onc.2015.366.
PMID: 26455323
ISSN: 1476-5594
CID: 1803562