Network


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

Hotspot


Dive into the research topics where Christopher T. Gubish is active.

Publication


Featured researches published by Christopher T. Gubish.


Clinical Cancer Research | 2009

HGF and c-Met Participate in Paracrine Tumorigenic Pathways in Head and Neck Squamous Cell Cancer

Lynn M. Knowles; Laura P. Stabile; Ann Marie Egloff; Mary E. Rothstein; Sufi M. Thomas; Christopher T. Gubish; Edwina C. Lerner; Raja R. Seethala; Shinsuke Suzuki; Kelly Quesnelle; Sarah Morgan; Robert L. Ferris; Jennifer R. Grandis; Jill M. Siegfried

Purpose: We determined hepatocyte growth factor (HGF) and c-Met expression and signaling in human head and neck squamous cell carcinoma (HNSCC) cells and primary tissues and tested the ability of c-Met tyrosine kinase inhibitors (TKI) to block HGF-induced biological signaling. Experimental Design: Expression and signaling were determined using immunoblotting, ELISA, and immunohistochemistry. Biological end points included wound healing, cell proliferation, and invasion. c-Met TKIs were tested for their ability to block HGF-induced signaling and biological effects in vitro and in xenografts established in nude mice. Results: c-Met was expressed and functional in HNSCC cells. HGF was secreted by HNSCC tumor-derived fibroblasts, but not by HNSCC cells. Activation of c-Met promoted phosphorylation of AKT and mitogen-activated protein kinase as well as release of the inflammatory cytokine interleukin-8. Cell growth and wound healing were also stimulated by HGF. c-Met TKIs blocked HGF-induced signaling, interleukin-8 release, and wound healing. Enhanced invasion of HNSCC cells induced by the presence of tumor-derived fibroblasts was completely blocked with a HGF-neutralizing antibody. PF-2341066, a c-Met TKI, caused a 50% inhibition of HNSCC tumor growth in vivo with decreased proliferation and increased apoptosis within the tumors. In HNSCC tumor tissues, both HGF and c-Met protein were increased compared with expression in normal mucosa. Conclusions: These results show that HGF acts mainly as a paracrine factor in HNSCC cells, the HGF/c-Met pathway is frequently up-regulated and functional in HNSCC, and a clinically relevant c-Met TKI shows antitumor activity in vivo. Blocking the HGF/c-Met pathway may be clinically useful for the treatment of HNSCC.


The Annals of Thoracic Surgery | 1998

The clinical significance of hepatocyte growth factor for non–small cell lung cancer

Jill M. Siegfried; Lisa A. Weissfeld; James D. Luketich; Robert J. Weyant; Christopher T. Gubish; Rodney J. Landreneau

BACKGROUND Hepatocyte growth factor (HGF) is a cytokine that is released after injury. It is a paracrine factor that is produced by mesenchymal cells; epithelial and endothelial cells respond to HGF through its receptor, the c-met protein. Hepatocyte growth factor induces cell growth and cell movement and is also highly angiogenic. Evidence from breast cancer patients suggests that HGF is a negative prognostic indicator for breast cancer and is associated with invasive disease. METHODS We measured the HGF content in tumor tissue from 56 non-small cell lung cancer patients using the Western blot technique. The amount of HGF in tumor extracts was quantitated by densitometry after transfer of proteins to nitrocellulose and exposure to antibodies. Survival curves were generated based on clinical information obtained for each patient. RESULTS Our data indicate that HGF is also a negative prognostic indicator in lung cancer. As in the study of breast cancer patients, HGF was associated with recurrence and poor survival; the relative risk was seen to increase with increasing HGF tumor content. At levels of HGF greater than 100 units, the relative risk was 10, compared with that in patients with an HGF level of 1 unit. Node-negative patients with an elevated HGF tumor content had a significantly poorer outcome than node-positive patients with a low HGF tumor content. The same relationship was observed if the patients were stratified by stage: elevated HGF was associated with stage I patients whose disease recurred and who died of their disease, and stage I patients with elevated HGF had a worse survival than higher stage patients with a low level of HGF. CONCLUSIONS These results suggest that elevated HGF may predict a more aggressive biology in non-small cell lung cancer patients. The level of HGF may be useful as an indicator of high risk in early stage lung cancer patients.


Oncogene | 2011

HGF-independent Potentiation of EGFR Action by c-Met

Austin M. Dulak; Christopher T. Gubish; Laura P. Stabile; Cassandra Henry; Jill M. Siegfried

The c-Met receptor is a potential therapeutic target for non-small cell lung cancer (NSCLC). Signaling interactions between c-Met and the mutant epidermal growth factor receptor (EGFR) have been studied extensively, but signaling intermediates and biological consequences of lateral signaling to c-Met in EGFR wild-type tumors are minimally understood. Our observations indicate that delayed c-Met activation in NSCLC cell lines is initiated by wild-type EGFR, the receptor most often found in NSCLC tumors. EGFR ligands induce accumulation of activated c-Met, which begins at 8 h and continues for 48 h. This effect is accompanied by an increase in c-Met expression and phosphorylation of critical c-Met tyrosine residues without activation of mitogen-activated protein kinase (MAPK) or Akt. Gene transcription is required for delayed c-Met activation; however, phosphorylation of c-Met by EGFR occurs without production of hepatocyte growth factor (HGF) or another secreted factor, supporting a ligand-independent mechanism. Lateral signaling is blocked by two selective c-Met tyrosine kinase inhibitors (TKIs), PF2341066 and SU11274, or with gefitinib, an EGFR TKI, suggesting kinase activity of both receptors is required for this effect. Prolonged c-Src phosphorylation is observed, and c-Src pathway is essential for EGFR to c-Met communication. Pretreatment with pan-Src family kinase inhibitors, PP2 and dasatinib, abolishes delayed c-Met phosphorylation. A c-Src dominant-negative construct reduces EGF-induced c-Met phosphorylation compared with control, further confirming a c-Src requirement. Inhibition of c-Met with PF2341066 and siRNA decreases EGF-induced phenotypes of invasion by ∼86% and motility by ∼81%, suggesting that a novel form of c-Met activation is utilized by EGFR to maximize these biological effects. Combined targeting of c-Met and EGFR leads to increased xenograft antitumor activity, demonstrating that inhibition of downstream and lateral signaling from the EGFR-c-Src–c-Met axis might be effective in treatment of NSCLC.


Clinical Cancer Research | 2011

Dual blockade of EGFR and c-Met abrogates redundant signaling and proliferation in head and neck carcinoma cells

Hai Xu; Laura P. Stabile; Christopher T. Gubish; William E. Gooding; Jennifer R. Grandis; Jill M. Siegfried

Purpose: Head and neck squamous cell carcinoma (HNSCC) is usually fatal, and innovative approaches targeting growth pathways are necessary to effectively treat this disease. Both the epidermal growth factor receptor (EGFR) and the hepatocyte growth factor (HGF)/c-Met pathways are overexpressed in HNSCC and initiate similar downstream signaling pathways. c-Met may act in consort with EGFR and/or be activated as a compensatory pathway in the presence of EGFR blockade. Experimental Design: Expression levels of EGFR and c-Met were determined by Western analysis in HNSCC cell lines and correlated with antitumor responses to inhibitors of these pathways. Results: Combining the c-Met inhibitor PF2341066 with the EGFR inhibitor gefitinib abrogated HNSCC cell proliferation, invasion, and wound healing significantly more than inhibition of each pathway alone in HNSCC cell lines. When both HGF and the EGFR ligand, TGF-α, were present in vitro, P-AKT and P-MAPK expression were maximally inhibited by targeting both EGFR and c-Met pathways, suggesting that c-Met or EGFR can compensate when phosphorylation of the other receptor is inhibited. We also showed that TGF-α can induce phosphorylation of c-Met over sixfold by 8 hours in the absence of HGF, supporting a ligand-independent mechanism. Combined targeting of c-Met and EGFR resulted in an enhanced inhibition of tumor volumes accompanied by a decreased number of proliferating cells and increased apoptosis compared with single agent treatment in vivo. Conclusions: Together, these results suggest that dual blockade of c-Met and EGFR may be a promising clinical therapeutic strategy for treating HNSCC. Clin Cancer Res; 17(13); 4425–38. ©2011 AACR.


Molecular Cancer Therapeutics | 2007

Antitumor mechanisms of combined gastrin-releasing peptide receptor and epidermal growth factor receptor targeting in head and neck cancer

Qing Zhang; Neil E. Bhola; Vivian Wai Yan Lui; Doris R. Siwak; Sufi M. Thomas; Christopher T. Gubish; Jill M. Siegfried; Gordon B. Mills; Dong M. Shin; Jennifer R. Grandis

Head and neck squamous cell carcinoma (HNSCC) is characterized by epidermal growth factor receptor (EGFR) overexpression, where EGFR levels correlate with survival. To date, EGFR targeting has shown limited antitumor effects in head and neck cancer when administrated as monotherapy. We previously identified a gastrin-releasing peptide/gastrin-releasing peptide receptor (GRP/GRPR) aurocrine regulatory pathway in HNSCC, where GRP stimulates Src-dependent cleavage of EGFR proligands with subsequent EGFR phosphorylation and mitogen-activated protein kinase (MAPK) activation. To determine whether GRPR targeting can enhance the antitumor efficacy of EGFR inhibition, we investigated the effects of a GRPR antagonist (PD176252) in conjunction with an EGFR tyrosine kinase inhibitor (erlotinib). Combined blockade of GRPR and EGFR pathways significantly inhibited HNSCC, but not immortalized mucosal epithelial cell, proliferation, invasion, and colony formation. In addition, the percentage of apoptotic cells increased upon combined inhibition. The enhanced antitumor efficacy was accompanied by increased expression of cleaved poly(ADP-ribose) polymerase (PARP) and decreased phospho-EGFR, phospho-MAPK, and proliferating cell nuclear antigen (PCNA). Using reverse-phase protein microarray (RPPA), we further detected decreased expression of phospho–c-Jun, phospho-p70S6K, and phospho-p38 with combined targeting. Cumulatively, these results suggest that GRPR targeting can enhance the antitumor effects of EGFR inhibitors in head and neck cancer. [Mol Cancer Ther 2007;6(4):1414–24]


Clinical Cancer Research | 2013

c-Src Activation Mediates Erlotinib Resistance in Head and Neck Cancer by Stimulating c-Met

Laura P. Stabile; Guoqing He; Vivian Wai Yan Lui; Cassandra Henry; Christopher T. Gubish; Sonali Joyce; Kelly Quesnelle; Jill M. Siegfried; Jennifer R. Grandis

Purpose: Strategies to inhibit the EGF receptor (EGFR) using the tyrosine kinase inhibitor erlotinib have been associated with limited clinical efficacy in head and neck squamous cell carcinoma (HNSCC). Co-activation of alternative kinases may contribute to erlotinib resistance. Experimental Design: We generated HNSCC cells expressing dominant-active c-Src (DA-Src) to determine the contribution of c-Src activation to erlotinib response. Results: Expression of DA-Src conferred resistance to erlotinib in vitro and in vivo compared with vector-transfected control cells. Phospho-Met was strongly upregulated by DA-Src, and DA-Src cells did not produce hepatocyte growth factor (HGF). Knockdown of c-Met enhanced sensitivity to erlotinib in DA-Src cells in vitro, as did combining a c-Met or c-Src inhibitor with erlotinib. Inhibiting EGFR resulted in minimal reduction of phospho-Met in DA-Src cells, whereas complete phospho-Met inhibition was achieved by inhibiting c-Src. A c-Met inhibitor significantly sensitized DA-Src tumors to erlotinib in vivo, resulting in reduced Ki67 labeling and increased apoptosis. In parental cells, knockdown of endogenous c-Src enhanced sensitivity to erlotinib, whereas treatment with HGF to directly induce phospho-Met resulted in erlotinib resistance. The level of endogenous phospho-c-Src in HNSCC cell lines was also significantly correlated with erlotinib resistance. Conclusions: Ligand-independent activation of c-Met contributes specifically to erlotinib resistance, not cetuximab resistance, in HNSCC with activated c-Src, where c-Met activation is more dependent on c-Src than on EGFR, providing an alternate survival pathway. Addition of a c-Met or c-Src inhibitor to erlotinib may increase efficacy of EGFR inhibition in patients with activated c-Src. Clin Cancer Res; 19(2); 380–92. ©2012 AACR.


Molecular Pharmacology | 2007

Signaling pathways involved in cyclooxygenase-2 induction by hepatocyte growth factor in non small-cell lung cancer.

Jill M. Siegfried; Christopher T. Gubish; Mary E. Rothstein; Pierre E. Queiroz de Oliveira; Laura P. Stabile

Many studies have suggested a role for the hepatocyte growth factor (HGF)/c-Met pathway in tumorigenesis. Some actions of HGF are believed to be mediated by cyclooxygenase-2 (COX-2), resulting in the production of prostaglandin E2 (PGE2). We examined four c-Met-positive non–small-cell lung cancer (NSCLC) cell lines for effects of HGF on COX-2. HGF increased COX-2 protein expression 3-fold over basal levels. Induction of COX-2 occurred through both the extracellular signal-regulated kinase 1/2 and p38 pathways. HGF treatment caused activation of the activator protein-1, CCAAT/enhancer-binding protein, and cAMP response element-binding protein transcription factors, and COX-2 induction was blocked by actinomycin D. The half-life of COX-2 mRNA was also increased by HGF. HGF stimulation resulted in a 4-fold increase in PGE2 secretion, and treatment of NSCLC cells with exogenous PGE2 significantly increased cell proliferation. The addition of PGE2 to NSCLC cells also led to rapid phosphorylation of c-Met in the absence of HGF, which was blocked by epidermal growth factor receptor (EGFR) inhibition. EGFR ligands were released in response to PGE2. This suggests that secretion of PGE2 induced by HGF/c-Met pathway activation can further activate the c-Met pathway via EGFR in a reinforcing loop that is independent of HGF. HGF and PGE2 each significantly stimulated invasion in NSCLC cells. Cells transiently transfected with c-Met antisense plasmid showed a significant decrease in HGF- or PGE2-induced invasion. PGE2-induced invasion was EGFR-dependent, confirming a link between PGE2, EGFR, and c-Met. Targeting of both the HGF/c-Met and PGE2 pathways with a neutralizing antibody to HGF and celecoxib resulted in enhanced anti-invasion effects in response to HGF.


Journal of Thoracic Oncology | 2012

Combining the multitargeted tyrosine kinase inhibitor vandetanib with the antiestrogen fulvestrant enhances its antitumor effect in non-small cell lung cancer.

Jill M. Siegfried; Christopher T. Gubish; Mary E. Rothstein; Cassandra Henry; Laura P. Stabile

Introduction: Estrogen is known to promote proliferation and to activate the epidermal growth factor receptor (EGFR) in non-small cell lung cancer (NSCLC). Vascular endothelial growth factor (VEGF) is a known estrogen responsive gene in breast cancer. We sought to determine whether the VEGF pathway is also regulated by estrogen in lung cancer cells, and whether combining an inhibitor of the ER pathway with a dual vascular endothelial growth factor receptor (VEGFR)/EGFR inhibitor would show enhanced antitumor effects. Methods: We examined activation of EGFR and expression of VEGF in response to &bgr;-estradiol, and the antitumor activity of the multitargeted VEGFR/EGFR/RET inhibitor, vandetanib, when combined with the antiestrogen fulvestrant both in vitro and in vivo. Results: NSCLC cells expressed VEGFR-3 and EGFR. Vandetanib treatment of NSCLC cells resulted in inhibition of EGFR and VEGFR-3 and inhibition of &bgr;-estradiol-induced P-MAPK activation, demonstrating that vandetanib blocks &bgr;-estradiol-induced EGFR signaling. Treatment with &bgr;-estradiol stimulated VEGFA mRNA and protein (p < 0.0001 over baseline), suggesting estrogenic signaling causes heightened VEGFA pathway activation. This estrogenic induction of VEGFA mRNA seems largely dependent on cross-talk with EGFR. Long-term vandetanib treatment also significantly increased ER&bgr; protein expression. The combination of vandetanib with fulvestrant maximally inhibited cell growth compared with single agents (p < 0.0001) and decreased tumor xenograft volume by 64%, compared with 51% for vandetanib (p < 0.05) and 23% for fulvestrant (p < 0.005). Antitumor effects of combination therapy were accompanied by a significant increase in apoptotic cells compared with single agents. Conclusions: Fulvestrant may enhance effects of vandetanib in NSCLC by blocking estrogen-driven activation of the EGFR pathway.


Journal of Thoracic Oncology | 2014

Co-targeting c-Met and COX-2 leads to enhanced inhibition of lung tumorigenesis in a murine model with heightened airway HGF.

Laura P. Stabile; Mary E. Rothstein; Christopher T. Gubish; Diana E. Cunningham; Nathan Lee; Jill M. Siegfried

Background: The hepatocyte growth factor (HGF)/c-Met pathway is often dysregulated in non–small-cell lung cancer (NSCLC). HGF activation of c-Met induces cyclooxygenase-2 (COX-2), resulting in downstream stimulation by prostaglandin E2 of additional pathways. Targeting both c-Met and COX-2 might lead to enhanced antitumor effects by blocking signaling upstream and downstream of c-Met. Methods: Effects of crizotinib or celecoxib alone or in combination were tested in NSCLC cells in vitro and in mice transgenic for airway expression of human HGF. Results: Proliferation and invasion of NSCLC cells treated with a combination of crizotinib and celecoxib were significantly lower compared with single treatments. Transgenic mice showed enhanced COX-2 expression localized to preneoplastic areas following exposure to the tobacco carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone, which was not present without carcinogen exposure. This shows that COX-2 activity is present during lung tumor development in a high HGF environment. After 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone treatment, a significant decrease in the number of lung tumors per animal was observed after 13-week treatments of crizotinib, celecoxib, or the combination compared with placebo (p < 0.001). With combination treatment, the number of tumors was also significantly lower than single agent treatment (p < 0.001). In the resulting lung tumors, P-c-Met, COX-2, prostaglandin E2, and P-MAPK were significantly downmodulated by combination treatment compared with single treatment. Expression of the epithelial-mesenchymal transition markers E-cadherin and snail was also modulated by combination treatment. Conclusions: In the presence of high HGF, dual inhibition of c-Met and COX-2 may enhance antitumor effects. This combination may have clinical potential in NSCLCs with high HGF/c-Met expression or epithelial-mesenchymal transition phenotype.


Cancer Research | 2013

Abstract 2026: Co-targeting c-Met and COX-2 leads to enhanced inhibition of lung tumorigenesis in a murine model with heightened HGF signaling in the airways.

Laura P. Stabile; Mary E. Rothstein; Christopher T. Gubish; Nathan Lee; Diana E. Cunningham; Jill M. Siegfried

The hepatocyte growth factor (HGF)/c-Met signaling pathway is often dysregulated in lung cancer. We previously showed that HGF activation of c-Met in non-small cell lung cancer (NSCLC) cells led to a significant induction of cyclooxygenase-2 (COX-2) followed by secretion of PGE 2 . PGE 2 caused secretion of TGFα by NSCLC cells, followed by both phosphorylation of EGFR and later, delayed phosphorylation of c-Met that does not require HGF. Secretion of PGE 2 also appears to boost c-Met pathway signaling downstream via cross-activation by EGFR, in a reinforcing loop that is independent of HGF. We hypothesized that targeting of both c-Met and COX-2 might lead to enhanced anti-tumor effects by blocking both upstream and downstream pro-tumor events. Maximum effects on cell proliferation, invasion and EMT markers were observed in vitro in NSCLC cell lines with combination celecoxib and crizotinib treatment. We next tested whether targeting c-Met with the tyrosine kinase inhibitor crizotinib combined with celecoxib to target COX-2 would result in enhanced anti-tumor effects in an animal model with heightened HGF/c-Met pathway signaling in the airways. Mice transgenic for airway expression of human HGF were treated with the tobacco carcinogen, 4-(methylnitroso-amino)-1-(3-pyridyl)-1-butanone, from weeks 1-4. Crizotinib (40mg/kg), celecoxib (50mg/kg), combined crizotinib and celecoxib or placebo control was administered by oral gavage daily from week 3 until week 15, at which time lung tumors were evaluated. A highly significant decrease in the number of lung tumors per animal was observed with crizotinib treatment (mean 3.6, range 2-6), celecoxib treatment (mean 4.3, range 3-6) and combination treatment (mean 1.5, range 0-3) compared to placebo control (mean 9.9, range 6-13; P 2 ), celecoxib (mean 0.22mm 2 ) and combination (mean 0.15mm 2 ) compared to placebo (mean 0.38mm 2 , P 2 , and P-MAPK) were optimally down-modulated by combination treatment, compared to single treatment. Additionally, E-cadherin expression was up-regulated while the E-cadherin repressor, snail, was down-regulated with dual treatment. In a pulmonary environment with heightened HGF/c-Met pathway activity, dual inhibition of the c-Met and COX-2 pathways may enhance anti-tumor effects by targeting reinforcing up- and downstream signaling. Such a combination has potential clinical benefit for lung cancer patients with dysregulated c-Met/COX-2 pathways. Supported by R01 CA79882 and P50 CA090440. Citation Format: Laura P. Stabile, Mary E. Rothstein, Christopher T. Gubish, Nathan Lee, Diana E. Cunningham, Jill M. Siegfried. Co-targeting c-Met and COX-2 leads to enhanced inhibition of lung tumorigenesis in a murine model with heightened HGF signaling in the airways. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 2026. doi:10.1158/1538-7445.AM2013-2026

Collaboration


Dive into the Christopher T. Gubish's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vivian Wai Yan Lui

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge