Network


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

Hotspot


Dive into the research topics where Ray Camphausen is active.

Publication


Featured researches published by Ray Camphausen.


mAbs | 2011

A fibronectin scaffold approach to bispecific inhibitors of epidermal growth factor receptor and insulin-like growth factor-I receptor.

Emanuel Sl; Engle Lj; Chao G; Zhu Rr; Cao C; Lin Z; Yamniuk Ap; Hosbach J; Brown J; Fitzpatrick E; Jochem Gokemeijer; Morin P; Brent Morse; Irvith M. Carvajal; Fabrizio D; Martin C. Wright; Das Gupta R; Michael L. Gosselin; Cataldo D; Ryseck Rp; Doyle Ml; Wong Tw; Ray Camphausen; Cload St; Marsh Hn; Gottardis Mm; Eric Furfine

Engineered domains of human fibronectin (Adnectins™) were used to generate a bispecific Adnectin targeting epidermal growth factor receptor (EGFR) and insulin-like growth factor-I receptor (IGF-IR), two transmembrane receptors that mediate proliferative and survival cell signaling in cancer. Single-domain Adnectins that specifically bind EGFR or IGF-IR were generated using mRNA display with a library containing as many as 1013 Adnectin variants. mRNA display was also used to optimize lead Adnectin affinities, resulting in clones that inhibited EGFR phosphorylation at 7 to 38 nM compared to 2.6 μM for the parental clone. Individual, optimized, Adnectins specific for blocking either EGFR or IGF-IR signaling were engineered into a single protein (EI-Tandem Adnectin). The EI-Tandems inhibited phosphorylation of EGFR and IGF-IR, induced receptor degradation, and inhibited down-stream cell signaling and proliferation of human cancer cell lines (A431, H292, BxPC3 and RH41) with IC50 values ranging from 0.1 to 113 nM. Although Adnectins bound to EGFR at a site distinct from those of anti-EGFR antibodies cetuximab, panitumumab and nimotuzumab, like the antibodies, the anti-EGFR Adnectins blocked the binding of EGF to EGFR. PEGylated EI-Tandem inhibited the growth of both EGFR and IGF-IR driven human tumor xenografts, induced degradation of EGFR, and reduced EGFR phosphorylation in tumors. These results demonstrate efficient engineering of bispecific Adnectins with high potency and desired specificity. The bispecificity may improve biological activity compared to monospecific biologics as tumor growth is driven by multiple growth factors. Our results illustrate a technological advancement for constructing multi-specific biologics in cancer therapy.


mAbs | 2010

Anti-tumor effect of CT-322 as an Adnectin inhibitor of vascular endothelial growth factor receptor-2

Roni Mamluk; Irvith M. Carvajal; Brent Morse; Henry K Wong; Janette Abramowitz; Sharon Aslanian; Ai-Ching Lim; Jochem Gokemeijer; Michael J. Storek; Joonsoo Lee; Michael L. Gosselin; Martin C. Wright; Ray Camphausen; Jack Wang; Yan Chen; Kathy D. Miller; Kerry Sanders; Sarah Short; Jeff Sperinde; Gargi Prasad; Stephen Williams; Robert S. Kerbel; John M.L. Ebos; Anthony J. Mutsaers; John Mendlein; Alan S. Harris; Eric Furfine

CT-322 is a new anti-angiogenic therapeutic agent based on an engineered variant of the tenth type III domain of human fibronectin, i.e., an AdnectinTM, designed to inhibit vascular endothelial growth factor receptor (VEGFR)-2. This PEGylated Adnectin was developed using an mRNA display technology. CT-322 bound human VEGFR-2 with high affinity (KD, 11 nM), but did not bind VEGFR-1 or VEGFR-3 at concentrations up to 100 nM, as determined by surface plasmon resonance studies. Western blot analysis showed that CT-322 blocked VEGF-induced phosphorylation of VEGFR-2 and mitogen-activated protein kinase in human umbilical vascular endothelial cells. CT-322 significantly inhibited the growth of human tumor xenograft models of colon carcinoma and glioblastoma at doses of 15-60 mg/kg administered 3 times/week. Anti-tumor effects of CT-322 were comparable to those of sorafenib or sunitinib, which inhibit multiple kinases, in a colon carcinoma xenograft model, although CT-322 caused less overt adverse effects than the kinase inhibitors. CT-322 also enhanced the anti-tumor activity of the chemotherapeutic agent temsirolimus in the colon carcinoma model. The high affinity and specificity of CT-322 binding to VEGFR-2 and its anti-tumor activities establish CT-322 as a promising anti-angiogenic therapeutic agent. Our results further suggest that Adnectins are an important new class of targeted biologics that can be developed as potential treatments for a wide variety of diseases.


Cancer Research | 2010

Abstract 2586: Adnectins as a platform for multi-specific targeted biologics: A novel bispecific inhibitor of EGFR and IGF-IR growth factor receptors

Stuart Emanuel; Linda Engle; Carolyn Cao; Ginger Chao; Zheng Lin; Rong-Rong Zhu; Aaron P. Yamniuk; Jennifer Hosbach; Jennifer S. Brown; Elizabeth Fitzpatrick; Jochem Gokemeijer; Paul E. Morin; Brent Morse; Irvith M. Carvajal; David Fabrizio; Martin C. Wright; Ruchira Dasgupta; Mike Gosselin; Rolf Ryseck; Michael L. Doyle; Tai W. Wong; Ray Camphausen; Sharon T. Cload; Nick Marsh; Eric Furfine; Marco M. Gottardis

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC The epidermal growth factor receptor (EGFR) and insulin-like growth factor receptor-1 (IGFR) are transmembrane receptor tyrosine kinases that mediate proliferative and invasive cell signaling in cancer. Inhibition of either receptor reduces tumor growth in both mouse models and in human clinical studies. Blocking the EGFR pathway can induce compensatory activation of the IGFR pathway to drive tumor growth and IGFR inhibition can result in activation of EGFR signaling in preclinical models. Therefore, blocking both receptors simultaneously may achieve superior efficacy to blocking either pathway alone. We developed individual optimized Adnectins™ specific for blocking either EGFR or IGFR signaling and engineered them into a single protein that linked both Adnectins together to construct a bi-specific Adnectin targeting the EGFR and IGFR (EI-tandem). The bifunctional molecule blocked activation of EGFR and IGFR, inhibited both EGF and IGF-induced down-stream cell signaling (MAPK and AKT pathways) and was antiproliferative in human cancer cell lines. Potency of the EI-tandem was comparable to anti-EGFR and anti-IGFR antibodies. The EI-tandem demonstrated a synergistic inhibition of IGFR phosphorylation and down-stream cell signaling compared to Adnectins specific for only EGFR or IGFR alone. Although Adnectins bound to the EGFR at a site distinct from the clinically approved anti-EGFR antibodies cetuximab, panitumumab and nimotuzumab, they still blocked binding of EGF to the EGFR. PEGylated EI-tandem inhibited the growth of human tumor xenografts driven by both EGFR and IGFR signaling, degraded EGFR and IGFR, and reduced phosphorylation of EGFR in tumors. Treatment of mice with EI-tandem caused increases in levels of the circulating ligands TGFα and IGF1 resulting from blockade of their respective receptors and provided convenient soluble biomarkers of target suppression. These results show that a bifunctional Adnectin can confer improved biological activity compared to monospecific biologics in tumors where growth is driven by multiple growth factors. 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 2586.


Archive | 2011

Fibronectin based scaffold domain proteins that bind PCSK9

Ray Camphausen; Jonathan Davis; Sharon T. Cload; Fabienne M. Denhez; Amna Saeed-Kothe; Dasa Lipovsek; Ching-Hsiung Frederick Lo; Chee Meng Low; Bowman Miao; Tracy S. Mitchell; Rex A. Parker; Ginger Chao Rakestraw; Katie A. Russo; Doree Sitkoff


Archive | 2011

Serum albumin binding molecules

Michael L. Gosselin; David Fabrizio; Joanna Swain; Tracy S. Mitchell; Ray Camphausen; Sharon T. Cload; Eric Furfine; Paul E. Morin; Ranjan Mukherjee; Simeon I. Taylor


Archive | 2009

Multivalent fibronectin based scaffold domain proteins

Ray Camphausen; Eric Furfine; Irvith M. Carvajal; H. Nicholas Marsh; Marco M. Gottardis; Joan M. Carboni; Ricardo M. Attar


Archive | 2009

Bispecific egfr/igfir binding molecules

Stuart Emanuel; Linda Engle; Ray Camphausen; Martin C. Wright; Ginger Chao Rakestraw; Marco M. Gottardis; Joan M. Carboni


Archive | 2007

Targeted therapeutics based on engineered proteins for tyrosine kinases receptors, including IGF-IR

Ray Camphausen; David Fabrizio; Martin C. Wright; Patrick Gage; John Mendlein


Archive | 2009

Targeted therapeutics based on engineered proteins that bind egfr

Ray Camphausen; Brent Morse; Stuart Emanuel; David Fabrizio


Archive | 2012

Fc fusion proteins comprising novel linkers or arrangements

Ray Camphausen; Amna Saeed-Kothe; Jonathan Davis; Tracy S. Mitchell

Collaboration


Dive into the Ray Camphausen'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge