Network


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

Hotspot


Dive into the research topics where Harold Frucht is active.

Publication


Featured researches published by Harold Frucht.


Gastroenterology | 2013

Mice That Express Human Interleukin-8 Have Increased Mobilization of Immature Myeloid Cells, Which Exacerbates Inflammation and Accelerates Colon Carcinogenesis

Samuel Asfaha; Alexander Dubeykovskiy; Hiroyuki Tomita; Xiangdong Yang; Sarah Stokes; Wataru Shibata; Richard A. Friedman; Hiroshi Ariyama; Zinaida A. Dubeykovskaya; Sureshkumar Muthupalani; Russell Ericksen; Harold Frucht; James G. Fox; Timothy C. Wang

BACKGROUND & AIMSnInterleukin (IL)-8 has an important role in initiating inflammation in humans, attracting immune cells such as neutrophils through their receptors CXCR1 and CXCR2. IL-8 has been proposed to contribute to chronic inflammation and cancer. However, mice do not have the IL-8 gene, so human cancer cell lines and xenograft studies have been used to study the role of IL-8 in colon and gastric carcinogenesis. We generated mice that carry a bacterial artificial chromosome that encompasses the entire human IL-8 gene, including its regulatory elements (IL-8Tg mice).nnnMETHODSnWe studied the effects of IL-8 expression in APCmin(+/-) mice and IL-8Tg mice given azoxymethane and dextran sodium sulfate (DSS). We also examined the effects of IL-8 expression in gastric cancer in INS-GAS mice that overexpress gastrin and IL-8Tg mice infected with Helicobacter felis.nnnRESULTSnIn IL-8Tg mice, expression of human IL-8 was controlled by its own regulatory elements, with virtually no messenger RNA or protein detectable under basal conditions. IL-8 was strongly up-regulated on systemic or local inflammatory stimulation, increasing mobilization of immature CD11b(+)Gr-1(+) myeloid cells (IMCs) with thioglycolate-induced peritonitis, DSS-induced colitis, and H. felis-induced gastritis. IL-8 was increased in colorectal tumors from patients and IL-8Tg mice compared with nontumor tissues. IL-8Tg mice developed more tumors than wild-type mice following administration of azoxymethane and DSS. Expression of IL-8 increased tumorigenesis in APCmin(+/-) mice compared with APCmin(+/-) mice that lack IL-8; this was associated with increased numbers of IMCs and angiogenesis in the tumors.nnnCONCLUSIONSnIL-8 contributes to gastrointestinal carcinogenesis by mobilizing IMCs and might be a therapeutic target for gastrointestinal cancers.


World Journal of Gastroenterology | 2014

Pancreatic ductal adenocarcinoma: Risk factors, screening, and early detection

Andrew E. Becker; Yasmin Hernandez; Harold Frucht; Aimee L. Lucas

Pancreatic cancer is the fourth most common cause of cancer-related deaths in the United States, with over 38000 deaths in 2013. The opportunity to detect pancreatic cancer while it is still curable is dependent on our ability to identify and screen high-risk populations before their symptoms arise. Risk factors for developing pancreatic cancer include multiple genetic syndromes as well as modifiable risk factors. Genetic conditions include hereditary breast and ovarian cancer syndrome, Lynch Syndrome, familial adenomatous polyposis, Peutz-Jeghers Syndrome, familial atypical multiple mole melanoma syndrome, hereditary pancreatitis, cystic fibrosis, and ataxia-telangiectasia; having a genetic predisposition can raise the risk of developing pancreatic cancer up to 132-fold over the general population. Modifiable risk factors, which include tobacco exposure, alcohol use, chronic pancreatitis, diet, obesity, diabetes mellitus, as well as certain abdominal surgeries and infections, have also been shown to increase the risk of pancreatic cancer development. Several large-volume centers have initiated such screening protocols, and consensus-based guidelines for screening high-risk groups have recently been published. The focus of this review will be both the genetic and modifiable risk factors implicated in pancreatic cancer, as well as a review of screening strategies and their diagnostic yields.


Clinical Cancer Research | 2013

High Prevalence of BRCA1 and BRCA2 Germline Mutations With Loss of Heterozygosity In a Series of Resected Pancreatic Adenocarcinoma and Other Neoplastic Lesions

Aimee L. Lucas; Reena Shakya; Marla Lipsyc; Elana B. Mitchel; Sheila Kumar; Caroline Hwang; Liyong Deng; Catherine Devoe; John A. Chabot; Matthias Szabolcs; Thomas Ludwig; Wendy K. Chung; Harold Frucht

PURPOSEnPancreatic ductal adenocarcinoma (PDAC) is associated with the breast ovarian cancer syndrome (BRCA1/BRCA2) mutations. It is unknown if this association is causal.nnnEXPERIMENTAL DESIGNnThis is a single-site study of patients who underwent surgical pancreatic tumor resection and self-identified as Ashkenazi Jewish. DNA from normal pancreatic tissue was genotyped for the three Ashkenazi Jewish BRCA1/2 founder mutations BRCA1 185delAG, BRCA1 5382insC, and BRCA2 6174delT, and loss of heterozygosity (LOH) was determined by sequencing DNA from microdissected tumor. When additional tumor tissue was available, p53 immunohistochemistry (IHC) was conducted.nnnRESULTSnThirty-seven patients underwent surgery for PDAC, seven for intraductal papillary mucinous neoplasm (IPMN), and 19 for other diseases. A high prevalence of BRCA1/2 mutations was found in the surgical cohort (12/63; 19.0%; P < 0.001), PDAC cohort (8/37; 21.6%; P < 0.001), and IPMN cohort (2/7; 28.6%; P = .01) compared with published control mutation frequency. A high prevalence of BRCA1 185delAG (8.1%; P < 0.001) and BRCA2 6174delT (10.8%; P < 0.001) in Ashkenazi Jewish patients with PDAC was shown. BRCA1/2 LOH was found in 2 of 4 BRCA1-associated PDACs and 3 of 4 BRCA2-associated PDACs. Positive p53 IHC was found in 5 of 8 BRCA1/2 PDACs.nnnCONCLUSIONSnWe show a high prevalence of BRCA1/2 mutations with LOH in an Ashkenazi Jewish cohort of surgically resected PDAC and neoplastic lesions, suggesting that these germline mutations are causal in selected individuals.


Biochimica et Biophysica Acta | 2002

Functional interaction of lithocholic acid conjugates with M3 muscarinic receptors on a human colon cancer cell line.

Kunrong Cheng; Ying Chen; Piotr Zimniak; Jean-Pierre Raufman; Yinghua Xiao; Harold Frucht

Lithocholic acid (LA) conjugates interact with M3 receptors, the muscarinic receptor subtype that modulates colon cancer cell proliferation. This observation prompted us to examine the action of bile acids on two human colon cancer cell lines: H508, which expresses M3 receptors, and SNU-C4, which does not. Cellular proliferation was determined using a colorimetric assay. Interaction with muscarinic receptors was determined by measuring inhibition of muscarinic radioligand binding and changes in cellular inositol phosphate (IP) formation. Lithocholyltaurine (LCT) caused a dose-dependent increase in H508 cell proliferation that was not observed in SNU-C4 cells. After a 6-day incubation with 300 microM LCT, H508 cell proliferation increased by 200% compared to control. Moreover, in H508 cells, LCT caused a dose-dependent inhibition of radioligand binding and an increase in IP formation. LCT did not alter the rate of apoptosis in H508 or SNU-C4 cells. These data indicate that, at concentrations achievable in the gut, LA derivatives interact with M3 muscarinic receptors on H508 human colon cancer cells, thereby causing an increase in IP formation and cell proliferation. This suggests a mechanism whereby alterations in intestinal bile acids may affect the growth of colon cancer cells.


Head and Neck-journal for The Sciences and Specialties of The Head and Neck | 2009

Head and neck squamous cell carcinoma in FAMMM syndrome.

Vladimir Vinarsky; Robert L. Fine; Adel Assaad; Ying Qian; John A. Chabot; Gloria H. Su; Harold Frucht

Germline mutations at the INK4a/p16 locus are implicated in several human cancer syndromes, including familial atypical multiple mole melanoma (FAMMM) syndrome, FAMMM‐pancreatic cancer (FAMMM‐PC) syndrome, and in familial head and neck cancer syndrome.


Cancer | 2014

BRCA1 and BRCA2 germline mutations are frequently demonstrated in both high‐risk pancreatic cancer screening and pancreatic cancer cohorts

Aimee L. Lucas; Laura E. Frado; Caroline Hwang; Sheila Kumar; Lauren G. Khanna; Elana Levinson; John A. Chabot; Wendy K. Chung; Harold Frucht

Approximately 10% of pancreatic ductal adenocarcinoma (PDAC) is due to a genetic predisposition, including the breast and ovarian cancer syndrome germline mutations BRCA1 and BRCA2. Knowledge of specific genetic mutations predisposing to PDAC may enable risk stratification, early detection, and the development of effective screening and surveillance programs. In the current study, the authors attempted to determine the diagnostic yield of testing for BRCA1/2 germline mutations in a PDAC screening cohort and a PDAC cohort referred for genetic testing.


Pancreas | 2013

Evidence for treatment and survival disparities by age in pancreatic adenocarcinoma: a population-based analysis.

Sunil Amin; Aimee L. Lucas; Harold Frucht

Objectives Studies demonstrate safety and survival benefits of surgical resection in older individuals with pancreatic adenocarcinoma. We investigated treatment disparities by age. Methods The Surveillance, Epidemiology, and End Results database for survival and treatment of pancreatic adenocarcinoma between 1983 and 2007 stratified by age: younger than 50 years, between 50 and 70 years, or older than 70 years. Kaplan-Meier curves and Cox proportional hazards models were used for survival differences, and logistic regression models were used for treatment disparities and the decision to refuse surgery. Results A total of 45,509 patients had microscopically confirmed pancreatic adenocarcinoma. Of these, 7374 (16%) received surgery and 9842 (22%) received radiation. Younger patients were more likely to receive both surgery and radiation. The prevalence of surgery decreased from 21% for those younger than 50 years to 19% for those between 50 and 70 years to 13% for those older than 70 years (P < 0.001). Radiation decreased from 28% to 25% to 17% (P < 0.001). Overall survival decreased with increasing age at diagnosis, 10.4 months (age <50 years) to 9.1 months (age 50–70 years) to 6.4 months (age >70 years) controlling for stage, sex, race, radiation, and surgery (P < 0.001). Increasing age negatively predicted the odds of receiving both surgery and radiation and increased the likelihood of refusing surgery. Conclusions Treatment disparities exist by age despite advances in radiation and surgical treatment. Increased treatment in the elderly will increase overall survival from pancreatic adenocarcinoma.


Cancer | 2012

Incidence of subsequent pancreatic adenocarcinoma in patients with a history of nonpancreatic primary cancers.

Sunil Amin; Russell McBride; Jennie Kline; Elana B. Mitchel; Aimee L. Lucas; Alfred I. Neugut; Harold Frucht

Several environmental risk factors are known to predispose individuals to pancreatic cancer, and up to 15% of pancreatic cancers have an inherited component. Understanding metachronous cancer associations can modify pancreas cancer risk. The objective of this study was to investigate the association of nonpancreatic cancers with subsequent pancreatic adenocarcinoma.


Pancreas | 2014

Absence of pancreatic intraepithelial neoplasia predicts poor survival after resection of pancreatic cancer.

Benjamin G. Hassid; Aimee L. Lucas; Marcela Salomao; Chunhua Weng; Feng Liu; Lauren G. Khanna; Sheila Kumar; Caroline Hwang; John A. Chabot; Harold Frucht

Objectives Pancreatic intraepithelial neoplasia (PanIN), thought to represent the dominant precursor of pancreatic adenocarcinoma (PDAC), is often found synchronously adjacent to resected PDAC tumors. However, its prognostic significance on outcome after PDAC resection is unknown. Methods A total of 342 patients who underwent resection for PDAC between 2005 and 2010 at a single institution were identified and stratified according to highest grade of PanIN demonstrated surrounding the tumor. Clinical and pathologic characteristics of each patient and tissue were recorded and analyzed. The primary outcome was length of survival after resection. Results An absence of PanIN lesions was identified in 32 patients (9%), low grade PanIN without synchronous high grade lesions was identified in 52 patients (15%), and high grade PanIN was found in 258 patients (75%). Median survival were 12.8 months for the non-PanIN group, 26.3 months for the low-grade PanIN group, and 23.8 months for the high-grade PanIN groups (P = 0.043). In multivariable analysis, absence of PanIN was independently associated with poor survival (P = 0.002). Conclusions The patients who demonstrate an absence of PanIN in the pancreatic tissue adjacent to the resected PDAC tumor have shorter postresection survival compared with those who demonstrate a PanIN lesion.


Abdominal Imaging | 2011

Physician knowledge and appropriate utilization of computed tomographic colonography in colorectal cancer screening

Matthew S. Chang; Jessica Shah; Sunil Amin; Susana Gonzalez; Joan C. Prowda; Joan M. Cheng; Elizabeth C. Verna; Don C. Rockey; Harold Frucht

GoalsTo assess physician understanding of computed tomographic colonography (CTC) in colorectal cancer (CRC) screening guidelines in a pilot study.BackgroundCTC is a sensitive and specific method of detecting colorectal polyps and cancer. However, several factors have limited its clinical availability, and CRC screening guidelines have issued conflicting recommendations.StudyA web-based survey was administered to physicians at two institutions with and without routine CTC availability.Results398 of 1655 (24%) participants completed the survey, 59% was from the institution with routine CTC availability, 52% self-identified as trainees, and 15% as gastroenterologists. 78% had no personal experience with CTC. Only 12% was aware of any current CRC screening guidelines that included CTC. In a multiple regression model, gastroenterologists had greater odds of being aware of guidelines (OR 3.49, CI 1.67–7.26), as did physicians with prior CTC experience (OR 4.81, CI 2.39–9.68), controlling for institution, level of training, sex, and practice type. Based on guidelines that recommend CTC, when given a clinical scenario, 96% of physicians was unable to select the appropriate follow-up after a CTC, which was unaffected by institution.ConclusionsMost physicians have limited experience with CTC and are unaware of recent recommendations concerning CTC in CRC screening.

Collaboration


Dive into the Harold Frucht's collaboration.

Top Co-Authors

Avatar

Aimee L. Lucas

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Caroline Hwang

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lauren G. Khanna

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elizabeth C. Verna

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Gloria H. Su

Columbia University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge