Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Richard F. Dunne is active.

Publication


Featured researches published by Richard F. Dunne.


Genes & Development | 2008

Methylation of RUNX1 by PRMT1 abrogates SIN3A binding and potentiates its transcriptional activity

Xinyang Zhao; Vladimir Jankovic; Alexander Gural; Gang Huang; Animesh Pardanani; Silvia Menendez; Jin Zhang; Richard F. Dunne; Andrew Xiao; Hediye Erdjument-Bromage; C. David Allis; Paul Tempst; Stephen D. Nimer

RUNX1/AML1 is required for the development of definitive hematopoiesis, and its activity is altered by mutations, deletions, and chromosome translocations in human acute leukemia. RUNX1 function can be regulated by post-translational modifications and protein-protein interactions. We show that RUNX1 is arginine-methylated in vivo by the arginine methyltransferase PRMT1, and that PRMT1 serves as a transcriptional coactivator for RUNX1 function. Using mass spectrometry, and a methyl-arginine-specific antibody, we identified two arginine residues (R206 and R210) within the region of RUNX1 that interact with the corepressor SIN3A and are methylated by PRMT1. PRMT1- dependent methylation of RUNX1 at these arginine residues abrogates its association with SIN3A, whereas shRNA against PRMT1 (or use of a methyltransferase inhibitor) enhances this association. We find arginine-methylated RUNX1 on the promoters of two bona fide RUNX1 target genes, CD41 and PU.1 and show that shRNA against PRMT1 or RUNX1 down-regulates their expression. These arginine methylation sites and the dynamic regulation of corepressor binding are lost in the leukemia-associated RUNX1-ETO fusion protein, which likely contributes to its dominant inhibitory activity.


Hematology-oncology Clinics of North America | 2015

Genetics and Biology of Pancreatic Ductal Adenocarcinoma

Richard F. Dunne

Pancreatic ductal adenocarcinoma remains a clinical challenge. Thus far, enlightenment on the downstream activities of Kras, the tumors unique metabolic needs, and how the stroma and immune system affect it have remained untranslated to the clinical practice. Given the numbers of diverse therapies in development and a growing knowledge about how to evaluate these systems preclinically and clinically, this is expected to change significantly and for the better over the next 5 years.


Cancer Investigation | 2012

Diabetes mellitus is associated with the presence of metastatic spread at disease presentation in hepatocellular carcinoma.

Gregory C. Connolly; Saman Safadjou; Rui Chen; Afamefuna Nduaguba; Richard F. Dunne; Alok A. Khorana

Purpose: Diabetes mellitus (DM) is identified as a negative prognostic indicator in hepatocellular carcinoma (HCC). Methods: A retrospective review of HCC patients was conducted to assess the effect of DM on clinical variables. Results: Ninety-seven of 265 (34%) patients had DM at the time of diagnosis. Distant metastasis was found in 33% (30/91) of patients with DM compared with only 9.7% (17/174) of those without DM (OR: 4.5, 95% CI: 2.3–8.8, p < .0001). This difference remained significant when adjusting for other clinical variables (OR: 10.0, 95% CI: 3.9–25.7, p < .0001). Conclusions: DM is associated with the presence of metastatic disease among a single institution cohort of HCC patients.


Rare Tumors | 2014

A Case Series of Transformation of Teratoma to Primitive Neuroectodermal Tumor: Evolving Management of a Rare Malignancy

Richard F. Dunne; Deepak M. Sahasrabudhe; Edward M. Messing; Jerome Jean-Gilles; Chunkit Fung

Primitive neuroectodermal tumor (PNET) is a pathologic diagnosis that encompasses several different tumor types, including central nervous system tumors and Ewing’s sarcomas. Teratoma, a common element of germ cell tumor (GCT), has the ability to transform to malignant PNET in a small number of patients. Making a definitive diagnosis of PNET is difficult given its deviation from elements of GCT and its non-specific pathologic findings. Establishing the diagnosis is crucial as PNETs respond poorly to standard platinum-based chemotherapy used for treatment of GCT. Primary treatment for PNET is surgical, though this is often not feasible in many patients due to extensive disease at diagnosis. As an alternative, chemotherapy regimens traditionally used for Ewing’s sarcoma, such as vincristine, doxorubicin and cyclophosphamide alternating with ifosfamide and etoposide, have shown limited efficacy in the neoadjuvant, adjuvant, and palliative settings. Future research should delineate the genetic underpinnings of PNET and develop therapeutic options accordingly.


JAMA Oncology | 2017

Association of Alterations in Main Driver Genes With Outcomes of Patients With Resected Pancreatic Ductal Adenocarcinoma

Zhi Rong Qian; Douglas A. Rubinson; Jonathan A. Nowak; Vicente Morales-Oyarvide; Richard F. Dunne; Margaret M. Kozak; Marisa W. Welch; Lauren K. Brais; Annacarolina da Silva; Tingting Li; Wanwan Li; Atsuhiro Masuda; Juhong Yang; Yan Shi; Mancang Gu; Yohei Masugi; Justin L. Bui; Caitlin Zellers; Chen Yuan; Ana Babic; Natalia Khalaf; Andrew J. Aguirre; Kimmie Ng; Rebecca A. Miksad; Andrea J. Bullock; Daniel T. Chang; Jennifer F. Tseng; Thomas E. Clancy; David C. Linehan; Jennifer J. Findeis-Hosey

Importance Although patients with resected pancreatic adenocarcinoma are at high risk for disease recurrence, few biomarkers are available to inform patient outcomes. Objective To evaluate the alterations of the 4 main driver genes in pancreatic adenocarcinoma and patient outcomes after cancer resection. Design, Setting, and Participants This study analyzed protein expression and DNA alterations for the KRAS, CDKN2A, SMAD4, and TP53 genes by immunohistochemistry and next-generation sequencing in formalin-fixed, paraffin-embedded tumors in 356 patients with resected pancreatic adenocarcinoma who were treated at the Dana-Farber/Brigham and Women’s Cancer Center (October 26, 2002, to May 21, 2012), University of Rochester Medical Center (March 1, 2006, to November 1, 2013), or Stanford Cancer Institute (September 26, 1995, to May 22, 2013). Associations of driver gene alterations with disease-free survival (DFS) and overall survival (OS) were evaluated using Cox proportional hazards regression with estimation of hazard ratios (HRs) and 95% CIs and adjustment for age, sex, tumor characteristics, institution, and perioperative treatment. Data were collected September 9, 2012, to June 28, 2016, and analyzed December 17, 2016, to March 14, 2017. Main Outcomes and Measures The DFS and OS among patients with resected pancreatic adenocarcinoma. Results Of the 356 patients studied, 191 (53.7%) were men and 165 (46.3%) were women, with a median (interquartile range [IQR]) age of 67 (59.0-73.5) years. Patients with KRAS mutant tumors had worse DFS (median [IQR], 12.3 [6.7 -27.2] months) and OS (20.3 [11.3-38.3] months) compared with patients with KRAS wild-type tumors (DFS, 16.2 [8.9-30.5] months; OS, 38.6 [16.6-63.1] months) and had 5-year OS of 13.0% vs 30.2%. Particularly poor outcomes were identified in patients with KRAS G12D-mutant tumors, who had a median (IQR) OS of 15.3 (9.8-32.7) months. Patients whose tumors lacked CDKN2A expression had worse DFS (median, 11.5 [IQR, 6.2-24.5] months) and OS (19.7 [10.9-37.1] months) compared with patients who had intact CDKN2A (DFS, 14.8 [8.2-30.5] months; OS, 24.6 [14.1-44.6] months). The molecular status of SMAD4 was not associated with DFS or OS, whereas TP53 status was associated only with shorter DFS (HR, 1.33; 95% CI, 1.02-1.75; P = .04). Patients had worse DFS and OS if they had a greater number of altered driver genes. Compared with patients with 0 to 2 altered genes, those with 4 altered genes had worse DFS (HR, 1.79 [95% CI, 1.24-2.59; P = .002]) and OS (HR, 1.38 [95% CI, 0.98-1.94; P = .06]). Five-year OS was 18.4% for patients with 0 to 2 gene alterations, 14.1% for those with 3 alterations, and 8.2% for those with 4 alterations. Conclusions and Relevance Patient outcomes are associated with alterations of the 4 main driver genes in resected pancreatic adenocarcinoma.


Human Pathology | 2015

Down-regulation of cytoplasmic PLZF correlates with high tumor grade and tumor aggression in non-small cell lung carcinoma.

Guang-Qian Xiao; Faqian Li; Jennifer J. Findeis-Hosey; Ollivier Hyrien; Pamela D. Unger; Lu Xiao; Richard F. Dunne; Eric S. Kim; Qi Yang; Loralee McMahon; David E. Burstein

There are currently no effective prognostic biomarkers for lung cancer. Promyelocytic leukemia zinc finger (PLZF), a transcriptional repressor, has a role in cell cycle progression and tumorigenicity in various cancers. The expression and value of PLZF in lung carcinoma, particularly in the subclass of non-small cell lung carcinoma (NSCLC), has not been studied. Our aim was to study the immunohistochemical expression of PLZF in lung adenocarcinoma and squamous cell carcinoma and correlate the alteration of PLZF expression with tumor differentiation, lymph node metastasis, tumor stage, and overall survival. A total of 296 NSCLCs being mounted on tissue microarray (181 adenocarcinomas and 91 squamous cell carcinomas) were investigated. Moderate to strong expression of PLZF was found in the cytoplasm of all the nonneoplastic respiratory epithelium and most (89.9%) well-differentiated adenocarcinoma. The proportions of moderately differentiated, poorly differentiated adenocarcinoma, and paired lymph node adenocarcinoma metastases that demonstrated negative or only weak PLZF reactivity were 75.6%, 97.2%, and 89.9%, respectively. The expression of PLZF in squamous cell carcinoma was mostly weak or absent and significantly lower than that in adenocarcinoma of the same grade (P < .0005). The loss of cytoplasmic PLZF strongly correlated with high tumor grade and lymph node metastasis in both squamous carcinoma and adenocarcinoma (P < .0001). Down-regulation of PLZF also correlated with higher tumor stage and shorter overall survival (P < .05). These results support a prognostic value for loss of cytoplasmic PLZF expression in the stratification of NSCLC and a possible role of cytoplasmic shift and down-regulation of PLZF in the pathogenesis of NSCLC.


Genetics in Medicine | 2018

Germline cancer susceptibility gene variants, somatic second hits, and survival outcomes in patients with resected pancreatic cancer

Matthew B. Yurgelun; Anu Chittenden; Vicente Morales-Oyarvide; Douglas A. Rubinson; Richard F. Dunne; Margaret M. Kozak; Zhi Rong Qian; Marisa W. Welch; Lauren K. Brais; Annacarolina da Silva; Justin L. Bui; Chen Yuan; Tingting Li; Wanwan Li; Atsuhiro Masuda; Mancang Gu; Andrea J. Bullock; Daniel T. Chang; Thomas E. Clancy; David C. Linehan; Jennifer J. Findeis-Hosey; Leona A. Doyle; Aaron R. Thorner; Matthew Ducar; Bruce M. Wollison; Natalia Khalaf; Kimberly Perez; Sapna Syngal; Andrew J. Aguirre; William C. Hahn

PurposeGermline variants in double-strand DNA damage repair (dsDDR) genes (e.g., BRCA1/2) predispose to pancreatic adenocarcinoma (PDAC) and may predict sensitivity to platinum-based chemotherapy and poly(ADP) ribose polymerase (PARP) inhibitors. We sought to determine the prevalence and significance of germline cancer susceptibility gene variants in PDAC with paired somatic and survival analyses.MethodsUsing a customized next-generation sequencing panel, germline/somatic DNA was analyzed from 289 patients with resected PDAC ascertained without preselection for high-risk features (e.g., young age, personal/family history). All identified variants were assessed for pathogenicity. Outcomes were analyzed using multivariable-adjusted Cox proportional hazards regression.ResultsWe found that 28/289 (9.7%; 95% confidence interval [CI] 6.5–13.7%) patients carried pathogenic/likely pathogenic germline variants, including 21 (7.3%) dsDDR gene variants (3 BRCA1, 4 BRCA2, 14 other dsDDR genes [ATM, BRIP1, CHEK2, NBN, PALB2, RAD50, RAD51C]), 3 Lynch syndrome, and 4 other genes (APC p.I1307K, CDKN2A, TP53). Somatic sequencing and immunohistochemistry identified second hits in the tumor in 12/27 (44.4%) patients with germline variants (1 failed sequencing). Compared with noncarriers, patients with germline dsDDR gene variants had superior overall survival (hazard ratio [HR] 0.54; 95% CI 0.30–0.99; P = 0.05).ConclusionNearly 10% of PDAC patients harbor germline variants, although the majority lack somatic second hits, the therapeutic significance of which warrants further study.


Current Opinion in Supportive and Palliative Care | 2017

Research priorities in cancer cachexia: The University of Rochester Cancer Center NCI Community Oncology Research Program Research Base Symposium on Cancer Cachexia and Sarcopenia

Richard F. Dunne; Karen M. Mustian; Jose M. Garcia; William Dale; Reid Hayward; Breton Roussel; Mary M. Buschmann; Bette J. Caan; Calvin Cole; Fergal J. Fleming; Joe V. Chakkalakal; David C. Linehan; Supriya G. Mohile

Purpose of review Cancer cachexia remains understudied and there are no standard treatments available despite the publication of an international consensus definition and the completion of several large phase III intervention trials in the past 6 years. In September 2015, The University of Rochester Cancer Center NCORP Research Base led a Symposium on Cancer Cachexia and Sarcopenia with goals of reviewing the state of the science, identifying knowledge gaps, and formulating research priorities in cancer cachexia through active discussion and consensus. Recent findings Research priorities that emerged from the discussion included the implementation of morphometrics into clinical decision making, establishing specific diagnostic criteria for the stages of cachexia, expanding patient selection in intervention trials, identifying clinically meaningful trial endpoints, and the investigation of exercise as an intervention for cancer cachexia. Summary Standardizing how we define and measure cancer cachexia, targeting its complex biologic mechanisms, enrolling patients early in their disease course, and evaluating exercise, either alone or in combination, were proposed as initiatives that may ultimately result in the improved design of cancer cachexia therapeutic trials.


British Journal of Cancer | 2017

Lymph node metastases in resected pancreatic ductal adenocarcinoma: predictors of disease recurrence and survival.

Vicente Morales-Oyarvide; Douglas A. Rubinson; Richard F. Dunne; Margaret M. Kozak; Justin L. Bui; Chen Yuan; Zhi Rong Qian; Ana Babic; Annacarolina da Silva; Jonathan A. Nowak; Natalia Khalaf; Lauren K. Brais; Marisa W. Welch; Caitlin Zellers; Kimmie Ng; Daniel T. Chang; Rebecca A. Miksad; Andrea J. Bullock; Jennifer F. Tseng; Richard Swanson; Thomas E. Clancy; David C. Linehan; Jennifer J. Findeis-Hosey; Leona A. Doyle; Jason L. Hornick; Shuji Ogino; Charles S. Fuchs; Albert C. Koong; Brian M. Wolpin

Background:Few studies have simultaneously assessed the prognostic value of the multiple classification systems for lymph node (LN) metastases in resected pancreatic ductal adenocarcinoma (PDAC).Methods:In 600 patients with resected PDAC, we examined the association of LN parameters (AJCC 7th and 8th editions, LN ratio (LNR), and log odds of metastatic LN (LODDS)) with pattern of recurrence and patient survival using logistic regression and Cox proportional hazards regression, respectively. Regression models adjusted for age, sex, margin status, tumour grade, and perioperative therapy.Results:Lymph node metastases classified by AJCC 7th and 8th editions, LNR, and LODDS were associated with worse disease free-survival (DFS) and overall survival (OS) (all Ptrend<0.01). American Joint Committee on Cancer 8th edition effectively predicted DFS and OS, while minimising model complexity. Lymph node metastases had weaker prognostic value in patients with positive margins and distal resections (both Pinteraction<0.03). Lymph node metastases by AJCC 7th and 8th editions did not predict the likelihood of local disease as the first site of recurrence.Conclusions:American Joint Committee on Cancer 8th edition LN classification is an effective and practical tool to predict outcomes in patients with resected PDAC. However, the prognostic value of LN metastases is attenuated in patients with positive resection margins and distal pancreatectomies.


Journal of gastrointestinal oncology | 2018

Beyond microsatellite testing: assessment of tumor mutational burden identifies subsets of colorectal cancer who may respond to immune checkpoint inhibition

David Fabrizio; Thomas J. George; Richard F. Dunne; Garrett Michael Frampton; James Sun; Kyle Gowen; Mark Kennedy; Joel Greenbowe; Alexa B. Schrock; Jeffrey S. Ross; P.J. Stephens; Siraj M. Ali; Vincent A. Miller; Marwan Fakih; Samuel J. Klempner

Background The clinical application of PD1/PD-L1 targeting checkpoint inhibitors in colorectal cancer (CRC) has largely focused on a subset of microsatellite instable (MSI-high) patients. However, the proposed genotype that sensitizes these patients to immunotherapy is not captured by MSI status alone. Estimation of tumor mutational burden (TMB) from comprehensive genomic profiling is validated against whole exome sequencing and linked to checkpoint response in metastatic melanoma, urothelial bladder cancer and non-small cell lung carcinoma. We sought to explore the subset of microsatellite stable (MSS) CRC patients with high TMB, and identify the specific genomic signatures associated with this phenotype. Furthermore, we explore the ability to quantify TMB as a potential predictive biomarker of PD1/PD-L1 therapy in CRC. Methods Formalin-fixed, paraffin embedded tissue sections from 6,004 cases of CRC were sequenced with a CLIA-approved CGP assay. MSI and TMB statuses were computationally determined using validated methods. The cutoff for TMB-high was defined according to the lower bound value that satisfied the 90% probability interval based on the TMB distribution across all MSI-High patients. Results MSS tumors were observed in 5,702 of 6,004 (95.0%) cases and MSI-H tumors were observed in 302 (5.0%) cases. All but one (99.7%) MSI-H cases were TMB-high (range, 6.3-746.9 mut/Mb) and 5,538 of 5,702 (97.0%) MSS cases were TMB-low (range, 0.0-10.8 mut/Mb). Consequently, 164 of 5,702 (2.9%) MSS cases were confirmed as TMB-high (range, 11.7-707.2 mut/Mb), representing an increase in the target population that may respond to checkpoint inhibitor therapy by 54% (466 vs. 302, respectively). Response to PD-1 inhibitor is demonstrated in MSS/TMB-high cases. Conclusions Concurrent TMB assessment accurately classifies MSI tumors as TMB-high and simultaneously identifies nearly 3% or CRC as MSS/TMB-high. This subgroup may expand the population of CRC who may benefit from immune checkpoint inhibitor based therapeutic approaches.

Collaboration


Dive into the Richard F. Dunne's collaboration.

Top Co-Authors

Avatar

David C. Linehan

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jennifer J. Findeis-Hosey

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar

Calvin Cole

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar

Karen M. Mustian

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar

Chunkit Fung

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar

Andrea J. Bullock

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge