Network


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

Hotspot


Dive into the research topics where Jonathan Tetreault is active.

Publication


Featured researches published by Jonathan Tetreault.


Clinical Cancer Research | 2014

LY2875358, a Neutralizing and Internalizing Anti-MET Bivalent Antibody, Inhibits HGF-Dependent and HGF-Independent MET Activation and Tumor Growth

Ling Liu; Wei Zeng; Mark Wortinger; S. Betty Yan; Paul Cornwell; Victoria L. Peek; Jennifer R. Stephens; Jonathan Tetreault; Jinqi Xia; Jason Manro; Kelly M. Credille; Darryl Ballard; Patricia Brown-Augsburger; Volker Wacheck; Chi-Kin Chow; Lihua Huang; Yong Wang; Irene Denning; Julian Davies; Ying Tang; Peter Edward Vaillancourt; Jirong Lu

Purpose: MET, the receptor for hepatocyte growth factor (HGF), has been implicated in driving tumor proliferation and metastasis. High MET expression is correlated with poor prognosis in multiple cancers. Activation of MET can be induced either by HGF-independent mechanisms such as gene amplification, specific genetic mutations, and transcriptional upregulation or by HGF-dependent autocrine or paracrine mechanisms. Experimental Design/Results: Here, we report on LY2875358, a novel humanized bivalent anti-MET antibody that has high neutralization and internalization activities, resulting in inhibition of both HGF-dependent and HGF-independent MET pathway activation and tumor growth. In contrast to other bivalent MET antibodies, LY2875358 exhibits no functional agonist activity and does not stimulate biologic activities such as cell proliferation, scattering, invasion, tubulogenesis, or apoptosis protection in various HGF-responsive cells and no evidence of inducing proliferation in vivo in a monkey toxicity study. LY2875358 blocks HGF binding to MET and HGF-induced MET phosphorylation and cell proliferation. In contrast to the humanized one-armed 5D5 anti-MET antibody, LY2875358 induces internalization and degradation of MET that inhibits cell proliferation and tumor growth in models where MET is constitutively activated. Moreover, LY2875358 has potent antitumor activity in both HGF-dependent and HGF-independent (MET-amplified) xenograft tumor models. Together, these findings indicate that the mechanism of action of LY2875358 is different from that of the one-armed MET antibody. Conclusions: LY2875358 may provide a promising therapeutic strategy for patients whose tumors are driven by both HGF-dependent and HGF-independent MET activation. LY2875358 is currently being investigated in multiple clinical studies. Clin Cancer Res; 20(23); 6059–70. ©2014 AACR.


Journal of Inflammation Research | 2016

Generation and characterization of ixekizumab, a humanized monoclonal antibody that neutralizes interleukin-17A

Ling Liu; Jirong Lu; Barrett Allan; Ying Tang; Jonathan Tetreault; Chi-Kin Chow; Barbra Barmettler; James Nelson; Holly Bina; Lihua Huang; Victor J. Wroblewski; Kristine Kay Kikly

Interleukin (IL)-17A exists as a homodimer (A/A) or as a heterodimer (A/F) with IL-17F. IL-17A is expressed by a subset of T-cells, called Th17 cells, at inflammatory sites. Most cell types can respond to the local production of IL-17A because of the near ubiquitous expression of IL-17A receptors, IL-17RA and IL-17RC. IL-17A stimulates the release of cytokines and chemokines designed to recruit and activate both neutrophils and memory T-cells to the site of injury or inflammation and maintain a proinflammatory state. IL-17A-producing pathogenic T-cells contribute to the pathogenesis of autoimmune diseases, including psoriasis, psoriatic arthritis, rheumatoid arthritis, and ankylosing spondylitis. This study describes the generation and characterization of ixekizumab, a humanized IgG4 variant IL-17A-neutralizing antibody. Ixekizumab binds human and cynomolgus monkey IL-17A with high affinity and binds rabbit IL-17A weakly but does not bind to rodent IL-17A or other IL-17 family members. Ixekizumab effectively inhibits the interaction between IL-17A and its receptor in binding assays and potently blocks IL-17A-induced GRO or KC secretion in cell-based assays. In an in vivo mouse pharmcodynamic model, ixekizumab blocks human IL-17A-induced mouse KC secretion. These data provide a comprehensive preclinical characterization of ixekizumab, for which the efficacy and safety have been demonstrated in human clinical trials in psoriasis and psoriatic arthritis.


Cancer Research | 2012

Abstract 2738: c-Met antibody LY2875358 (LA480) has pre-clinical enhanced efficacy with gastric cancer standard-of-care in vitro and in vivo

Mark Wortinger; Victoria L. Peek; Wei Zeng; Lei Yan; Jonathan Tetreault; Jinqi Xia; Lu Jirong; Chi-Kin Chow; Jason Manro; Jennifer R. Stephens; Farhana F. Merzoug; Peter Edward Vaillancourt; S. Betty Yan; Ling Liu

cMet is a member of the receptor tyrosine kinase family and is the receptor for hepatocyte growth factor (HGF). cMet has been implicated in the initiation and progression of cancer due to the range of activities that cMet stimulates including proliferation, migration, morphogenesis, and survival. Inappropriate activation of c-Met can be induced by ligand-independent mechanisms such as gene amplification, specific genetic mutations, and transcriptional upregulation, or by ligand-dependent autocrine or paracrine mechanisms. Indeed, amplification of the c-Met gene, with consequent protein overexpression and constitutive kinase activation, has been reported in a number of human cancers, including gastric, esophageal and non-small-cell lung carcinomas. It has been reported that ∼10-20% of gastric tumors have increased copy numbers of the MET gene and overexpression of c-Met significantly correlates with poor prognosis in gastric cancer. c-Met antibody LY2875358 treatment reduces proliferation of gastric cancer cell lines with ligand-independent activation of c-Met resulting from gene amplification. The ability of LY2875358 to internalize and deplete cell surface c-Met is implicated in its activity against ligand-independent driven gastric cell lines. Here, we demonstrate that the pre-clinical combination of c-Met antibody LY2875358 with gastric cancer standard-of-care treatment has better efficacy than either treatment alone, both in vitro and in vivo. These data suggest that LY2875358 in combination with standard-of-care may be a promising treatment for gastric cancer. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 2738. doi:1538-7445.AM2012-2738


Cancer Research | 2012

Abstract 2734: c-Met antibody LY2875358 (LA480) shows differential antitumor effects in non-small cell lung cancer

Wei Zeng; Lei Yan; Victoria L. Peek; Mark Wortinger; Jonathan Tetreault; Jinqi Xia; Chi-Kin Chow; Jason Manro; Jennifer R. Stephens; Spring Weir; Ying Tang; Peter Edward Vaillancourt; Jirong Lu; Betty Yan; Ling Liu

c-Met is a member of the receptor tyrosine kinase family and is the receptor for hepatocyte growth factor (HGF). c-Met is involved in many mechanisms of cancer proliferation and metastasis. Inappropriate activation of c-Met can be induced by ligand-independent mechanisms such as gene amplification, specific genetic mutations, and transcriptional up-regulation, or by ligand-dependent autocrine or paracrine mechanisms. Lung cancer is the leading cause of cancer death worldwide. Despite the successful development of EGFR- or EML4-ALK-targetd therapies, treatment options remain limited for patients with advanced lung cancer, making the identification of new therapeutic targets essential. c-Met expression was reported in 41-72% non-small cell lung cancer (NSCLC), amplification of c-Met occurs in 5-10 % of patients, and c-Met mutations have been detected in 8-13% of patients. We have developed a bi-valent c-Met antibody LY2875358 (LA480), which blocks ligand-dependent and ligand-independent c-Met activations. It is currently in clinical development. Here, we have demonstrated that LY2875358 alone or in combination with standard-of-care (SOC) affected cell proliferation, migration and signal transduction in NSCLC cells with c-Met gene amplification, mutations and overexpression. In vitro, LY2875358 induces wild type and mutant c-Met internalization and degradation. In vivo, LY2875358 alone shows a marked antitumor activity in Met amplification NSCLC xenograft models. The combination of LY2875358 with SOC chemotherapeutics treatments has better efficacy than either treatment alone, both in vitro and in vivo. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 2734. doi:1538-7445.AM2012-2734


Cancer Research | 2013

Abstract 5465: LY2875358, a bivalent MET antibody with anti-tumor activity through blocking HGF as well as inducing degradation of MET, differentiates from a one-armed 5D5 MET antibody.

Wei Zeng; Victoria L. Peek; Mark Wortinger; Jonathan Tetreault; Jinqi Xia; Jennifer R. Stephens; Kelly M. Credille; Darryl Ballard; Trish Brown-Augsburger; Jirong Lu; Chi-Kin Chow; Peter Edward Vaillancourt; Ying Tang; Sau-Chi B. Yan; Ling Liu

MET is involved in many mechanisms of cancer proliferation and metastasis. Inappropriate activation of MET can be induced by HGF-independent mechanisms such as gene amplification, specific genetic mutations, and transcriptional up-regulation, or by HGF-dependent autocrine or paracrine mechanisms. LY2875358 is a novel humanized bivalent MET antibody currently in phase I clinical testing (trial NCT01287546). LY2875358 has high neutralization and internalization activities against MET for inhibiting HGF-dependent and HGF-independent MET pathway activation and tumor growth. In HGF-dependent MET activation, LY2875358 blocks HGF binding to MET, HGF-induced MET phosphorylation and tumor growth both in cell culture and in mouse xenograft models, resembling activities of a humanized one-armed 5D5 MET antibody (monovalent antibody similar to Onartuzumab). In tumors with HGF-independent MET activation through MET gene amplification, LY2875358 induces internalization and degradation of MET, which results in decreased pMET and total MET, inhibition of cell proliferation and tumor growth in MKN45 and SNU5 gastric tumor lines and EBC-1 and H1993 NSCLC tumor lines. Moreover, LY2875358 enhances antitumor activity in combination with cisplatin or 5-FU in vitro and in vivo in MET amplified tumor cells. However, under the same ligand-independent conditions, the one-armed 5D5 antibody did not have anti-tumor activities. When HGF is added to tumor cells with high MET gene amplification, LY2875358 decreases cell proliferation, while the one-armed 5D5 antibody does not. In contrast to other bivalent MET antibodies, LY2875358 has no or otherwise negligible agonist activity and does not stimulate biological activities such as cell proliferation, scattering, invasion, tubulogenesis, apoptosis protection or angiogenesis in various HGF responsive cells. These findings indicate that LY2875358 has a different mechanism of action from the humanized one-armed 5D5 MET antibody. LY2875358 may be a promising therapy for treatment of patients whose tumors are driven by HGF-dependent and HGF-independent MET activation. Citation Format: Wei Zeng, Victoria Peek, Mark Wortinger, Jonathan Tetreault, Jinqi Xia, Jennifer Stephens, Kelly Credille, Darryl Ballard, Trish Brown-Augsburger, Jirong Lu, Chi-Kin Chow, Peter Vaillancourt, Ying Tang, Sau-Chi B. Yan, Ling Liu. LY2875358, a bivalent MET antibody with anti-tumor activity through blocking HGF as well as inducing degradation of MET, differentiates from a one-armed 5D5 MET antibody. [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 5465. doi:10.1158/1538-7445.AM2013-5465


Cancer Research | 2017

Abstract 3090: LY3207447, a tetravalent bispecific antibody targeting VEGFR2 and angiopoietin-2, provides a more efficient anti-angiogenic therapy and an alternative for combination

Jonathan Tetreault; Sudhakar Chintharlapalli; Donmienne Doen Mun Leung; Damien Gerald; Linda Lee; Rowena Almonte-Baldonado; Lysiane Huber; Jianghuai Xu; Bharathi Ramamurthy; Jennifer Pereira; Johnny E. Croy; Jirong Lu; Ling Liu

Angiopoietin-2 (Ang-2) is released from endothelial cells only in response to stimulus (e.g. wound healing, tumor growth) and facilitates blood vessel sprouting and inhibits pericyte-endothelial cell interaction via Tie2 signaling. In tumors, Ang-2 is up-regulated and acts together with the VEGF/VEGFR2 pathway to stimulate tumor angiogenesis and metastasis. While therapeutic intervention using antagonists to the VEGF/VEGFR2 pathway has proven to be successful in limiting disease progression in a number of different clinical settings, there is an obvious need for an improved response. In various preclinical mouse angiogenesis or xenograft models, the combination treatment with anti-Ang2 antibody and the VEGF/VEGFR blocker provided additional benefit over inhibiting the individual pathway. Here as an alternative to combo therapy, we have engineered a tetravalent IgG-scFv bispecific antibody, LY3207447. LY3207447 is an immunoglobulin G4 (IgG4) antibody, comprising of VEGFR2 antibody derived from ramucirumab and a C-terminally fused single-chain variable fragment (scFv) targeting Ang2. LY3207447 binds to both the extracellular domain of VEGFR2 and soluble Ang2 with high affinity and blocks binding of Ang2 to Tie2 and VEGF to VEGFR2, and therefore inhibits signaling. We have shown that LY3207447 blocks binding of human Ang-2 to human Tie2-Fc by an ELISA assay and neutralizes Ang-2 induced phospho-Tie-2, but not Ang-1 induced phospho-Tie-2 in CHO cells overexpressing Tie-2 receptor. Moreover, LY3207447 neutralizes human VEGF165-induced phospho-VEGFR2 stimulation, cord formation and cell proliferation in human endothelial colony forming cells (ECFCs) and human dermal microvascular endothelial cells (HMVEC-d). LY3207447 blocks human and cyno VEGFR2, but not rodent VEGFR2. For anti-Ang2 arm, LY3207447 blocks human, cyno and mouse Ang2. Pre-clinical evaluation of LY3207447 in mouse retinal angiogenesis model resulted in an abrogation of angiogenesis. Combination studies using a rodent-specific surrogate VEGFR2 blocking antibody, DC101, with parental Ang2 antibody from which the scFv was derived from, inhibited both tumor growth and metastasis, resulting in increased survival compared to monotherapies in mouse xenograft model. These data establish VEGFR2/Ang2 bispecific antibodies as a promising anti-angiogenic, anti-metastatic and anti-tumor agent for the treatment of cancer in combination with other therapies. Citation Format: Jonathan Tetreault, Sudhakar Chintharlapalli, Donmienne Leung, Damien Gerald, Linda Lee, Rowena Almonte-Baldonado, Lysiane Huber, Jianghuai Xu, Bharathi Ramamurthy, Jennifer Pereira, Johnny Croy, Jirong Lu, Ling Liu. LY3207447, a tetravalent bispecific antibody targeting VEGFR2 and angiopoietin-2, provides a more efficient anti-angiogenic therapy and an alternative for combination [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3090. doi:10.1158/1538-7445.AM2017-3090


Cancer Research | 2016

Abstract 3259: LY3127804, a novel anti-Angiopoietin-2 antibody in combination with an anti-VEGFR2 antibody potently inhibits angiogenesis, tumor growth and metastasis

Sudhakar Chintharlapalli; Johnny E. Croy; Donmienne Doen Mun Leung; Damien Gerald; Jirong Lu; Philip W. Iversen; Linda N. Lee; Lysiane Huber; Jonathan Tetreault; Rowena Almonte-Baldonado; Jianghuai Xu; Bharathi Ramamurthy; Jennifer Pereira; Chi-Kin Chow; Axel-Rainer Hanauske; Volker Wacheck; Laura E. Benjamin; Ling Liu

Angiopoeitin-2 (Ang2) is released from endothelial cells only in response to stimulus (e.g. wound healing, tumor growth) and facilitates blood vessel sprouting and inhibits pericyte-endothelial cell interaction via Tie2 signaling. Combination of an anti-Ang2 antibody and aflibercept, a VEGF trap, has been shown to inhibit tumor growth and decrease tumor vascularity in mouse xenograft tumor models (Daly et al., Cancer Res (2013) 73(1):108). Multiple investigational anti-Ang2 antibody therapies are currently in clinical trials. LY3127804 is a humanized and engineered IgG4 isotype antibody that selectively binds to Ang2 with high affinity and neutralizes Ang2 induced phospho-Tie2. LY3127804 inhibits sprouting angiogenesis and increases pericyte coverage in a mouse developmental retinal angiogenesis model and in mice bearing PC3 xenograft tumors. Combination of LY3127804 and DC101, a potent anti-VEGFR2 antibody, exhibits enhanced efficacy when compared to monotherapy in multiple patient derived xenograft models including NSCLC and ovarian cancers. Anti-Ang2 antibody monotherapy alone resulted in marginal reduction of tumor growth and improved overall survival, while DC101monotherapy had greater reduction in tumor volume with no survival benefit in MDA-MB-231 breast orthotopic model. Combination of anti-Ang2 antibody with anti-VEGFR2 antibody shows reduction in tumor volume and improved overall survival. This robust pre-clinical evidence supports testing the combination of anti-Ang2 and anti-VEGFR2 antibodies in the clinic. LY3127804 is currently in Phase 1 clinical trials (NCT02597036) Citation Format: Sudhakar R. Chintharlapalli, Johnny E. Croy, Donmienne Leung, Damien Gerald, Jirong Lu, Philip W. Iversen, Linda N. Lee, Lysiane Huber, Jonathan Tetreault, Rowena Almonte-Baldonado, Jianghuai Xu, Bharathi Ramamurthy, Jennifer A. Pereira, Chi-Kin Chow, Axel-Rainer Hanauske, Volker Wacheck, Laura Benjamin, Ling Liu. LY3127804, a novel anti-Angiopoietin-2 antibody in combination with an anti-VEGFR2 antibody potently inhibits angiogenesis, tumor growth and metastasis. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3259.


Cancer Research | 2010

Abstract 2430: Gastric cancer cell lines with c-Met gene amplification are sensitive to growth inhibition by the bivalent c-Met antibody LA480

Jonathan Tetreault; Jinqi Xia; Wei Zeng; Mark Wortinger; Jason Cunningham; Farhana Mohammed Feroze; Kun Yu; Chi-Kin Chow; Jirong Lu; Irene Denning; Peter Edward Vaillancourt; Julian Davies; Ling Liu

Gastric cancer is common in Asia, and advanced disease is typically incurable. Development of effective treatments for gastric cancer is thus a high priority to address this highly unmet clinical need. c-Met gene amplification resulting in c-Met overexpression and pathway activation has been highly linked to tumor progression and metastasis. It has been reported that approximately 10-20% of gastric tumors have increased copy numbers of the MET gene. c-Met- amplified gastric cancer cell lines that are dependent on an activated c-Met pathway for cell growth have been reported to be highly sensitive to growth inhibition by small molecule inhibitors of c-Met. We report that LA480, a humanized monoclonal antibody specific for c-Met, can directly inhibit HGF-independent in vitro cell proliferation of three c-Met- amplified Asian gastric cell lines (MKN45, SNU-5, and NUGC-4), in a dose-dependent manner. After 48 hours of treatment with LA480, 60-70% percent inhibition of 3 H-thymidine incorporation was observed. For one of these gastric lines (NUGC-4), sensitivity to LA480 increases with continuous passage of the culture. This increased sensitivity to LA480 over time appears to be associated with increased expression of cell surface c-Met and increased constitutive phospho-Met expression. In addition to inhibiting proliferation of c-Met- amplified Asian gastric lines, LA480 also directly inhibited proliferation of a c-Met- amplified gastric cell line (Hs 746T) from a Caucasian patient. These data suggest that targeting c-Met amplification with LA480 may be a promising treatment for gastric cancer. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2430.


Archive | 2006

Anti-il-17 antibodies

Barrett Allan; Chi-Kin Chow; Lihua Huang; Ling Liu; Jirong Lu; Kingman Ng; Jonathan Tetreault; Andrew Gordon Werner


Blood | 2003

Selective enhancement of multipotential hematopoietic progenitors in vitro and in vivo by IL-20.

Ling Liu; Chunjin Ding; Wei Zeng; Josef G. Heuer; Jonathan Tetreault; Timothy Wayne Noblitt; Giao Hangoc; Scott Cooper; Kellie A. Brune; Ganesh R. Sharma; Niles Fox; Scott William Rowlinson; Danise Paige Rogers; Derrick Ryan Witcher; Peter K. Lambooy; Victor J. Wroblewski; James R. Miller; Hal E. Broxmeyer

Collaboration


Dive into the Jonathan Tetreault's collaboration.

Top Co-Authors

Avatar

Ling Liu

Eli Lilly and Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jirong Lu

Eli Lilly and Company

View shared research outputs
Top Co-Authors

Avatar

Wei Zeng

Eli Lilly and Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jinqi Xia

Eli Lilly and Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge