Network


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

Hotspot


Dive into the research topics where Sunil R. Hingorani is active.

Publication


Featured researches published by Sunil R. Hingorani.


Science | 2009

Inhibition of Hedgehog Signaling Enhances Delivery of Chemotherapy in a Mouse Model of Pancreatic Cancer

Kenneth P. Olive; Michael A. Jacobetz; Christian J. Davidson; Aarthi Gopinathan; Dominick J.O. McIntyre; Davina Jean Honess; Basetti Madhu; Mae Goldgraben; Meredith E. Caldwell; David Allard; Kristopher K. Frese; Gina M. DeNicola; Christine Feig; Chelsea Combs; Stephen P. Winter; Heather Ireland-Zecchini; Stefanie Reichelt; William J. Howat; Alex R. Chang; Mousumi Dhara; Lifu Wang; Felix Rückert; Robert Grützmann; Christian Pilarsky; Kamel Izeradjene; Sunil R. Hingorani; Pearl S. Huang; Susan E. Davies; William Plunkett; Merrill J. Egorin

Its All in the Delivery Pancreatic cancer is almost universally associated with a poor prognosis, in part because the tumors are resistant to chemotherapeutic drugs. Working with a mouse tumor model that displays many features of the human disease, Olive et al. (p. 1457, published online 21 May; see the Perspective by Olson and Hanahan) found that the tumors were poorly vascularized, a factor likely to impede drug delivery. Treatment of the mice with the chemotherapeutic drug gemcitabine in combination with a drug that depletes tumor-associated stromal tissue led to an increase in tumor vasculature, enhanced delivery of gemcitabine, and a delay in disease progression. Thus, drugs targeting the tumor stroma may merit investigation as a way to enhance the efficacy of conventional chemotherapy for pancreatic cancer. Pancreatic tumors are unresponsive to chemotherapy because their limited vasculature precludes efficient drug delivery. Pancreatic ductal adenocarcinoma (PDA) is among the most lethal human cancers in part because it is insensitive to many chemotherapeutic drugs. Studying a mouse model of PDA that is refractory to the clinically used drug gemcitabine, we found that the tumors in this model were poorly perfused and poorly vascularized, properties that are shared with human PDA. We tested whether the delivery and efficacy of gemcitabine in the mice could be improved by coadministration of IPI-926, a drug that depletes tumor-associated stromal tissue by inhibition of the Hedgehog cellular signaling pathway. The combination therapy produced a transient increase in intratumoral vascular density and intratumoral concentration of gemcitabine, leading to transient stabilization of disease. Thus, inefficient drug delivery may be an important contributor to chemoresistance in pancreatic cancer.


Cancer Cell | 2003

Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse

Sunil R. Hingorani; Emanuel F. Petricoin; Anirban Maitra; Vinodh N. Rajapakse; Catrina King; Michael A. Jacobetz; Sally Ross; Thomas P. Conrads; Timothey D. Veenstra; Ben A. Hitt; Yoshiya Kawaguchi; Don Johann; Lance A. Liotta; Howard C. Crawford; Mary E. Putt; Tyler Jacks; Christopher V.E. Wright; Ralph H. Hruban; Andrew M. Lowy; David A. Tuveson

To evaluate the role of oncogenic RAS mutations in pancreatic tumorigenesis, we directed endogenous expression of KRAS(G12D) to progenitor cells of the mouse pancreas. We find that physiological levels of Kras(G12D) induce ductal lesions that recapitulate the full spectrum of human pancreatic intraepithelial neoplasias (PanINs), putative precursors to invasive pancreatic cancer. The PanINs are highly proliferative, show evidence of histological progression, and activate signaling pathways normally quiescent in ductal epithelium, suggesting potential therapeutic and chemopreventive targets for the cognate human condition. At low frequency, these lesions also progress spontaneously to invasive and metastatic adenocarcinomas, establishing PanINs as definitive precursors to the invasive disease. Finally, mice with PanINs have an identifiable serum proteomic signature, suggesting a means of detecting the preinvasive state in patients.


Cancer Cell | 2012

Enzymatic Targeting of the Stroma Ablates Physical Barriers to Treatment of Pancreatic Ductal Adenocarcinoma

Paolo P. Provenzano; Carlos Cuevas; Amy E. Chang; Vikas K. Goel; Daniel D. Von Hoff; Sunil R. Hingorani

Pancreatic ductal adenocarcinomas (PDAs) are characterized by a robust fibroinflammatory response. We show here that this desmoplastic reaction generates inordinately high interstitial fluid pressures (IFPs), exceeding those previously measured or theorized for solid tumors, and induces vascular collapse, while presenting substantial barriers to perfusion, diffusion, and convection of small molecule therapeutics. We identify hyaluronan, or hyaluronic acid (HA), as the primary matrix determinant of these barriers and show that systemic administration of an enzymatic agent can ablate stromal HA from autochthonous murine PDA, normalize IFP, and re-expand the microvasculature. In combination with the standard chemotherapeutic, gemcitabine, the treatment permanently remodels the tumor microenvironment and consistently achieves objective tumor responses, resulting in a near doubling of overall survival.


Cancer Cell | 2004

Endogenous oncogenic K-rasG12D stimulates proliferation and widespread neoplastic and developmental defects

David A. Tuveson; Alice T. Shaw; Nicholas A. Willis; Daniel P. Silver; Erica L. Jackson; Sandy Chang; Kim L. Mercer; Rebecca Grochow; Hanno Hock; Denise Crowley; Sunil R. Hingorani; Tal Z. Zaks; Catrina King; Michael A. Jacobetz; Lifu Wang; Roderick T. Bronson; Stuart H. Orkin; Ronald A. DePinho; Tyler Jacks

Activating mutations in the ras oncogene are not considered sufficient to induce abnormal cellular proliferation in the absence of cooperating oncogenes. We demonstrate that the conditional expression of an endogenous K-ras(G12D) allele in murine embryonic fibroblasts causes enhanced proliferation and partial transformation in the absence of further genetic abnormalities. Interestingly, K-ras(G12D)-expressing fibroblasts demonstrate attenuation and altered regulation of canonical Ras effector signaling pathways. Widespread expression of endogenous K-ras(G12D) is not tolerated during embryonic development, and directed expression in the lung and GI tract induces preneoplastic epithelial hyperplasias. Our results suggest that endogenous oncogenic ras is sufficient to initiate transformation by stimulating proliferation, while further genetic lesions may be necessary for progression to frank malignancy.


Journal of Clinical Oncology | 2009

Consensus report of the national cancer institute clinical trials planning meeting on pancreas cancer treatment

Philip A. Philip; Margaret Mooney; Deborah Jaffe; Gail Eckhardt; Malcolm J. Moore; Neal J. Meropol; Leisha A. Emens; Eileen Mary O'Reilly; Murray Korc; Lee M. Ellis; Jacqueline Benedetti; Mace L. Rothenberg; Christopher G. Willett; Margaret A. Tempero; Andrew M. Lowy; James L. Abbruzzese; Diane M. Simeone; Sunil R. Hingorani; Jordan Berlin; Joel E. Tepper

Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer mortality, despite significant improvements in diagnostic imaging and operative mortality rates. The 5-year survival rate remains less than 5% because of microscopic or gross metastatic disease at time of diagnosis. The Clinical Trials Planning Meeting in pancreatic cancer was convened by the National Cancer Institutes Gastrointestinal Cancer Steering Committee to discuss the integration of basic and clinical knowledge in the design of clinical trials in PDAC. Major emphasis was placed on the enhancement of research to identify and validate the relevant targets and molecular pathways in PDAC, cancer stem cells, and the microenvironment. Emphasis was also placed on developing rational combinations of targeted agents and the development of predictive biomarkers to assist selection of patient subsets. The development of preclinical tumor models that are better predictive of human PDAC must be supported with wider availability to the research community. Phase III clinical trials should be implemented only if there is a meaningful clinical signal of efficacy and safety in the phase II setting. The emphasis must therefore be on performing well-designed phase II studies with uniform sets of basic entry and evaluation criteria with survival as a primary endpoint. Patients with either metastatic or locally advanced PDAC must be studied separately.


Molecular and Cellular Biology | 2003

Targeted Deletion Reveals an Essential Function for the Telomere Length Regulator Trf1

Jan Karlseder; Leili Kachatrian; Hiroyuki Takai; Kim L. Mercer; Sunil R. Hingorani; Tyler Jacks; Titia de Lange

ABSTRACT The human telomeric DNA binding factor TRF1 (hTRF1) and its interacting proteins TIN2, tankyrase 1 and 2, and PINX1 have been implicated in the regulation of telomerase-dependent telomere length maintenance. Here we show that targeted deletion of exon 1 of the mouse gene encoding Trf1 causes early (day 5 to 6 postcoitus) embryonic lethality. The absence of telomerase did not alter the Terf1ex1Δ/ex1Δ lethality, indicating that the phenotype was not due to inappropriate telomere elongation by telomerase. Terf1ex1Δ/ex1Δ blastocysts had a severe growth defect of the inner cell mass that was accompanied by apoptosis. However, no evidence was found for telomere uncapping causing this cell death; chromosome spreads of Terf1ex1Δ/ex1Δ blastocysts did not reveal chromosome end-to-end fusions, and p53 deficiency only briefly delayed Terf1ex1Δ/ex1Δ lethality. These data suggest that murine Trf1 has an essential function that is independent of telomere length regulation.


Clinical Cancer Research | 2008

A Phase I Trial of the Oral, Multikinase Inhibitor Sorafenib in Combination with Carboplatin and Paclitaxel

Keith T. Flaherty; Joan H. Schiller; Lynn M. Schuchter; Glenn Liu; David A. Tuveson; Maryann Redlinger; Chetan Lathia; Chenghua Xia; Oana Petrenciuc; Sunil R. Hingorani; Michael A. Jacobetz; Patricia Van Belle; David E. Elder; Marcia S. Brose; Barbara L. Weber; Mark R. Albertini; Peter J. O'Dwyer

Purpose: This study evaluated the safety, maximum tolerated dose, pharmacokinetics, and antitumor activity of sorafenib, a multikinase inhibitor, combined with paclitaxel and carboplatin in patients with solid tumors. Patients and Methods: Thirty-nine patients with advanced cancer (24 with melanoma) received oral sorafenib 100, 200, or 400 mg twice daily on days 2 to 19 of a 21-day cycle. All patients received carboplatin corresponding to AUC6 and 225 mg/m2 paclitaxel on day 1. Pharmacokinetic analyses were done for sorafenib on days 2 and 19 of cycle 1 and for paclitaxel on day 1 of cycles 1 and 2. Pretreatment tumor samples from 17 melanoma patients were analyzed for BRAF mutations. Results: Sorafenib was well tolerated at the doses evaluated. The most frequent severe adverse events were hematologic toxicities (grade 3 or 4 in 33 patients, 85%). Twenty-seven (69%) patients had sorafenib-related adverse events, the most frequent of which were dermatologic events (26 patients, 67%). Exposure to paclitaxel was not altered by intervening treatment with sorafenib. Treatment with sorafenib, paclitaxel, and carboplatin resulted in one complete response and nine partial responses, all among patients with melanoma. There was no correlation between BRAF mutational status and treatment responses in patients with melanoma. Conclusions: The recommended phase II doses are oral 400 mg twice daily sorafenib, carboplatin at an AUC6 dose, and 225 mg/m2 paclitaxel. The tumor responses observed with this combined regimen in patients with melanoma warrant further investigation.


British Journal of Cancer | 2013

Hyaluronan, fluid pressure, and stromal resistance in pancreas cancer

Paolo P. Provenzano; Sunil R. Hingorani

Pancreatic ductal adenocarcinomas (PDAs) are notoriously aggressive and resistant to treatment. They distinguish themselves further by their robust fibroinflammatory, or desmoplastic, stromal reaction and degree of hypovascularity. Recent findings have revealed multiple mechanisms of stromal complicity in disease pathogenesis and resistance. In this review, we focus on altered physicomechanics as one mechanism of what we term as ‘stromal resistance’ in PDA. Extremely high interstitial fluid pressures and a dense extracellular matrix combine to limit the delivery and distribution of therapeutic agents. We discuss the genesis and consequences of altered fluid dynamics in PDA and strategies to restore them.


Clinical Cancer Research | 2016

Phase Ib Study of PEGylated Recombinant Human Hyaluronidase and Gemcitabine in Patients with Advanced Pancreatic Cancer

Sunil R. Hingorani; William Proctor Harris; J. Thaddeus Beck; Boris A. Berdov; Stephanie Ann Wagner; Eduard M. Pshevlotsky; Sergei Tjulandin; Oleg Gladkov; Randall F. Holcombe; Ronald L. Korn; Natarajan Raghunand; Samuel S. Dychter; Ping Jiang; H. Michael Shepard; Craig Devoe

Purpose: This phase Ib study evaluated the safety and tolerability of PEGylated human recombinant hyaluronidase (PEGPH20) in combination with gemcitabine (Gem), and established a phase II dose for patients with untreated stage IV metastatic pancreatic ductal adenocarcinoma (PDA). Objective response rate and treatment efficacy using biomarker and imaging measurements were also evaluated. Experimental Design: Patients received escalating intravenous doses of PEGPH20 in combination with Gem using a standard 3+3 dose-escalation design. In cycle 1 (8 weeks), PEGPH20 was administrated twice weekly for 4 weeks, then once weekly for 3 weeks; Gem was administrated once weekly for 7 weeks, followed by 1 week off treatment. In each subsequent 4-week cycle, PEGPH20 and Gem were administered once weekly for 3 weeks, followed by 1 week off. Dexamethasone (8 mg) was given pre- and post-PEGPH20 administration. Several safety parameters were evaluated. Results: Twenty-eight patients were enrolled and received PEGPH20 at 1.0 (n = 4), 1.6 (n = 4), or 3.0 μg/kg (n = 20), respectively. The most common PEGPH20-related adverse events were musculoskeletal and extremity pain, peripheral edema, and fatigue. The incidence of thromboembolic events was 29%. Median progression-free survival (PFS) and overall survival (OS) rates were 5.0 and 6.6 months, respectively. In 17 patients evaluated for pretreatment tissue hyaluronan (HA) levels, median PFS and OS rates were 7.2 and 13.0 months for “high”-HA patients (n = 6), and 3.5 and 5.7 months for “low”-HA patients (n = 11), respectively. Conclusions: PEGPH20 in combination with Gem was well tolerated and may have therapeutic benefit in patients with advanced PDA, especially in those with high HA tumors. Clin Cancer Res; 22(12); 2848–54. ©2016 AACR.


Gut | 2014

Targeted depletion of an MDSC subset unmasks pancreatic ductal adenocarcinoma to adaptive immunity

Ingunn M. Stromnes; J. Scott Brockenbrough; Kamel Izeradjene; Markus A. Carlson; Carlos Cuevas; Randi M Simmons; Philip D. Greenberg; Sunil R. Hingorani

Background Pancreatic ductal adenocarcinoma (PDA) is characterised by a robust desmoplasia, including the notable accumulation of immunosuppressive cells that shield neoplastic cells from immune detection. Immune evasion may be further enhanced if the malignant cells fail to express high levels of antigens that are sufficiently immunogenic to engender an effector T cell response. Objective To investigate the predominant subsets of immunosuppressive cancer-conditioned myeloid cells that chronicle and shape the progression of pancreas cancer. We show that selective depletion of one subset of myeloid-derived suppressor cells (MDSC) in an autochthonous, genetically engineered mouse model (GEMM) of PDA unmasks the ability of the adaptive immune response to engage and target tumour epithelial cells. Methods A combination of in vivo and in vitro studies were performed employing a GEMM that faithfully recapitulates the cardinal features of human PDA. The predominant cancer-conditioned myeloid cell subpopulation was specifically targeted in vivo and the biological outcomes determined. Results PDA orchestrates the induction of distinct subsets of cancer-associated myeloid cells through the production of factors known to influence myelopoiesis. These immature myeloid cells inhibit the proliferation and induce apoptosis of activated T cells. Targeted depletion of granulocytic MDSC (Gr-MDSC) in autochthonous PDA increases the intratumoral accumulation of activated CD8 T cells and apoptosis of tumour epithelial cells and also remodels the tumour stroma. Conclusions Neoplastic ductal cells of the pancreas induce distinct myeloid cell subsets that promote tumour cell survival and accumulation. Targeted depletion of a single myeloid subset, the Gr-MDSC, can unmask an endogenous T cell response, disclosing an unexpected latent immunity and invoking targeting of Gr-MDSC as a potential strategy to exploit for treating this highly lethal disease.

Collaboration


Dive into the Sunil R. Hingorani's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

David A. Tuveson

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Ingunn M. Stromnes

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Philip D. Greenberg

Fred Hutchinson Cancer Research 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

Markus A. Carlson

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Kathleen E. DelGiorno

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Martin C. Whittle

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge