Network


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

Hotspot


Dive into the research topics where Andrew Capen is active.

Publication


Featured researches published by Andrew Capen.


Cancer Research | 2005

The Protein Kinase Cβ–Selective Inhibitor, Enzastaurin (LY317615.HCl), Suppresses Signaling through the AKT Pathway, Induces Apoptosis, and Suppresses Growth of Human Colon Cancer and Glioblastoma Xenografts

Jeremy R. Graff; Ann M. McNulty; Kimberly R. Hanna; Bruce W. Konicek; Rebecca L. Lynch; Spring N. Bailey; Crystal Banks; Andrew Capen; Robin L. Goode; Jason E. Lewis; Lillian Sams; Karen L. Huss; Robert M. Campbell; Philip W. Iversen; Blake Lee Neubauer; Thomas J. Brown; Luna Musib; Sandaruwan Geeganage; Donald Thornton

Activation of protein kinase Cbeta (PKCbeta) has been repeatedly implicated in tumor-induced angiogenesis. The PKCbeta-selective inhibitor, Enzastaurin (LY317615.HCl), suppresses angiogenesis and was advanced for clinical development based upon this antiangiogenic activity. Activation of PKCbeta has now also been implicated in tumor cell proliferation, apoptosis, and tumor invasiveness. Herein, we show that Enzastaurin has a direct effect on human tumor cells, inducing apoptosis and suppressing the proliferation of cultured tumor cells. Enzastaurin treatment also suppresses the phosphorylation of GSK3betaser9, ribosomal protein S6(S240/244), and AKT(Thr308). Oral dosing with Enzastaurin to yield plasma concentrations similar to those achieved in clinical trials significantly suppresses the growth of human glioblastoma and colon carcinoma xenografts. As in cultured tumor cells, Enzastaurin treatment suppresses the phosphorylation of GSK3beta in these xenograft tumor tissues. Enzastaurin treatment also suppresses GSK3beta phosphorylation to a similar extent in peripheral blood mononuclear cells (PBMCs) from these treated mice. These data show that Enzastaurin has a direct antitumor effect and that Enzastaurin treatment suppresses GSK3beta phosphorylation in both tumor tissue and in PBMCs, suggesting that GSK3beta phosphorylation may serve as a reliable pharmacodynamic marker for Enzastaurin activity. With previously published reports, these data support the notion that Enzastaurin suppresses tumor growth through multiple mechanisms: direct suppression of tumor cell proliferation and the induction of tumor cell death coupled to the indirect effect of suppressing tumor-induced angiogenesis.


Journal of Clinical Investigation | 2007

Therapeutic suppression of translation initiation factor eIF4E expression reduces tumor growth without toxicity

Jeremy R. Graff; Bruce W. Konicek; Thomas M. Vincent; Rebecca L. Lynch; David Monteith; Spring Weir; Phil Schwier; Andrew Capen; Robin L. Goode; Michele Dowless; Yuefeng Chen; Hong Zhang; Sean Sissons; Karen Cox; Ann M. McNulty; Stephen Parsons; Tao Wang; Lillian Sams; Sandaruwan Geeganage; Larry E. Douglass; Blake Lee Neubauer; Nicholas M. Dean; Kerry Blanchard; Jianyong Shou; Louis Stancato; Julia H. Carter; Eric G. Marcusson

Expression of eukaryotic translation initiation factor 4E (eIF4E) is commonly elevated in human and experimental cancers, promoting angiogenesis and tumor growth. Elevated eIF4E levels selectively increase translation of growth factors important in malignancy (e.g., VEGF, cyclin D1) and is thereby an attractive anticancer therapeutic target. Yet to date, no eIF4E-specific therapy has been developed. Herein we report development of eIF4E-specific antisense oligonucleotides (ASOs) designed to have the necessary tissue stability and nuclease resistance required for systemic anticancer therapy. In mammalian cultured cells, these ASOs specifically targeted the eIF4E mRNA for destruction, repressing expression of eIF4E-regulated proteins (e.g., VEGF, cyclin D1, survivin, c-myc, Bcl-2), inducing apoptosis, and preventing endothelial cells from forming vessel-like structures. Most importantly, intravenous ASO administration selectively and significantly reduced eIF4E expression in human tumor xenografts, significantly suppressing tumor growth. Because these ASOs also target murine eIF4E, we assessed the impact of eIF4E reduction in normal tissues. Despite reducing eIF4E levels by 80% in mouse liver, eIF4E-specific ASO administration did not affect body weight, organ weight, or liver transaminase levels, thereby providing the first in vivo evidence that cancers may be more susceptible to eIF4E inhibition than normal tissues. These data have prompted eIF4E-specific ASO clinical trials for the treatment of human cancers.


Cancer Research | 2011

Abstract 2819: Identification and characterization of a novel smoothened antagonist for the treatment of cancer with deregulated hedgehog signaling

Mark H. Bender; Philip Arthur Hipskind; Andrew Capen; Michael D. Cockman; Kelly M. Credille; Hong Gao; Jolie Anne Bastian; Julia M. Clay; Karen Lynn Lobb; Daniel Jon Sall; Michelle Lee Thompson; Takako Wilson; Graham N. Wishart; Bharvin Patel

The Hedgehog (Hh) pathway is a highly conserved signaling system that plays an important role in embryonic development and tissue homeostasis through regulation of cell differentiation and proliferation, and deregulated Hh signaling has been implicated in variety of cancers. Two distinct mechanisms are responsible for inappropriate and uncontrolled Hh pathway activation in human malignancies: ligand-dependent, due to over-expression of Hh ligand, and ligand-independent, resulting from genetic mutations in pathway components such as Patched (Ptch) and Smoothened (Smo). Smo, a member of the class F G-protein coupled receptor family, is a key regulator of Hh signaling pathway, and therefore is an attractive target for pathway modulation. We have identified a potent and selective small molecule antagonist of Smo. This novel molecule (LY2940680) binds to the Smo receptor and potently inhibits Hh signaling in Daoy, a human medulloblastoma tumor cell line, and C3H10T½, a mouse mesenchymal cell line. Importantly, LY2940680 binds to and inhibits the functional activity of resistant Smo mutant (D473H) produced by treatment with GDC-0449 (a Smo antagonist from Genentech). LY2940680 also has excellent pharmacokinetic properties in rodent and non-rodent species. Treatment of Ptch +/− p53 −/− transgenic mice, which spontaneously develop medulloblastoma, with oral administration of LY2940680 produced remarkable efficacy and significantly improved their survival. Magnetic resonance imaging of these mice revealed rapid kinetics of anti-tumor activity. Immunohistochemistry analysis of medulloblastoma tumors showed that LY2940680 treatment induced Caspase-3 activity and reduced proliferation. LY2940680 inhibited Hh regulated gene expression in the subcutaneous xenograft tumor stroma and produced significant anti-tumor activity. In summary, we have characterized an orally bio-available small molecule Smo antagonist that may provide therapeutic benefit to cancer patients with deregulated Hh signaling. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 2819. doi:10.1158/1538-7445.AM2011-2819


PLOS ONE | 2015

Activating the Wnt/β-Catenin Pathway for the Treatment of Melanoma – Application of LY2090314, a Novel Selective Inhibitor of Glycogen Synthase Kinase-3

Jennifer M. Atkinson; Kenneth B. Rank; Yi Zeng; Andrew Capen; Vipin Yadav; Jason Manro; Thomas A. Engler; Marcio Chedid

It has previously been observed that a loss of β-catenin expression occurs with melanoma progression and that nuclear β-catenin levels are inversely proportional to cellular proliferation, suggesting that activation of the Wnt/β-catenin pathway may provide benefit for melanoma patients. In order to further probe this concept we tested LY2090314, a potent and selective small-molecule inhibitor with activity against GSK3α and GSK3β isoforms. In a panel of melanoma cell lines, nM concentrations of LY2090314 stimulated TCF/LEF TOPFlash reporter activity, stabilized β-catenin and elevated the expression of Axin2, a Wnt responsive gene and marker of pathway activation. Cytotoxicity assays revealed that melanoma cell lines are very sensitive to LY2090314 in vitro (IC50 ~10nM after 72hr of treatment) in contrast to other solid tumor cell lines (IC50 >10uM) as evidenced by caspase activation and PARP cleavage. Cell lines harboring mutant B-RAF or N-RAS were equally sensitive to LY2090314 as were those with acquired resistance to the BRAF inhibitor Vemurafenib. shRNA studies demonstrated that β-catenin stabilization is required for apoptosis following treatment with the GSK3 inhibitor since the sensitivity of melanoma cell lines to LY290314 could be overcome by β-catenin knockdown. We further demonstrate that in vivo, LY2090314 elevates Axin2 gene expression after a single dose and produces tumor growth delay in A375 melanoma xenografts with repeat dosing. The activity of LY2090314 in preclinical models suggests that the role of Wnt activators for the treatment of melanoma should be further explored.


European Journal of Cancer | 2014

A phase I trial of LY2584702 tosylate, a p70 S6 kinase inhibitor, in patients with advanced solid tumours

Anthony W. Tolcher; Jonathan W. Goldman; Amita Patnaik; Kyriakos P. Papadopoulos; Paul Westwood; Claudia S. Kelly; William Bumgardner; Lillian Sams; Sandaruwan Geeganage; Tao Wang; Andrew Capen; Jianping Huang; Sajan Joseph; Joel Miller; Karim A. Benhadji; Les H. Brail; Lee S. Rosen

BACKGROUND LY2584702 tosylate (hereafter referred to as LY2584702) is a potent, highly selective adenosine triphosphate (ATP) competitive inhibitor against p70 S6 kinase, a downstream component of the phosphatidylinositol-3-kinase signalling pathway which regulates cell proliferation and survival. LY2584702 exhibited anti-tumour activity in preclinical analysis. METHODS Patients with advanced solid tumours were treated with LY2584702 orally on a 28-day cycle until the criteria for maximum tolerated dose (MTD) were met. Skin biopsies were collected for pharmacodynamic analysis, and levels of phospho-S6 protein were examined. The primary objective was to determine a phase II dose and schedule with secondary objectives of observing safety and tolerability. Dose escalation was based upon Common Terminology Criteria for Adverse Events Version 3.0. RESULTS Thirty-four patients were enrolled onto this phase I study and treated with LY2584702 on a QD (once-daily) or BID (twice-daily) dosing schedule. Part A dose escalation (n=22) began with 300 mg BID (n=2). Due to toxicity, this was scaled back to doses of 25mg (n=3), 50 mg (n=8), 100mg (n=3), and 200 mg (n=6) QD. Part B dose escalation (n=12) included 50 mg (n=3), 75 mg (n=3), and 100 mg (n=6) BID. Seven patients experienced dose-limiting toxicity (DLT). All DLTs were Grade 3 and included vomiting, increased lipase, nausea, hypophosphataemia, fatigue and pancreatitis. CONCLUSION The MTD was determined to be 75 mg BID or 100mg QD. No responses were observed at these levels. Pharmacokinetic analysis revealed substantial variability in exposure and determined that LY2584702 treatment was not dose proportional with increasing dose.


Molecular Cancer Therapeutics | 2015

Myostatin Neutralization Results in Preservation of Muscle Mass and Strength in Preclinical Models of Tumor Induced Muscle Wasting

Rosamund C. Smith; Martin S. Cramer; Pamela Jean Mitchell; Andrew Capen; Lysiane Huber; Rong Wang; Laura Myers; Bryan Edward Jones; Brian J. Eastwood; Darryl Ballard; Jeffrey C. Hanson; Kelly M. Credille; Victor J. Wroblewski; Boris Lin; Josef G. Heuer

Skeletal muscle wasting occurs in a great majority of cancer patients with advanced disease and is associated with a poor prognosis and decreased survival. Myostatin functions as a negative regulator of skeletal muscle mass and has recently become a therapeutic target for reducing the loss of skeletal muscle and strength associated with clinical myopathies. We generated neutralizing antibodies to myostatin to test their potential use as therapeutic agents to attenuate the skeletal muscle wasting due to cancer. We show that our neutralizing antimyostatin antibodies significantly increase body weight, skeletal muscle mass, and strength in non–tumor-bearing mice with a concomitant increase in mean myofiber area. The administration of these neutralizing antibodies in two preclinical models of cancer-induced muscle wasting (C26 colon adenocarcinoma and PC3 prostate carcinoma) resulted in a significant attenuation of the loss of muscle mass and strength with no effect on tumor growth. We also show that the skeletal muscle mass– and strength-preserving effect of the antibodies is not affected by the coadministration of gemcitabine, a common chemotherapeutic agent, in both non–tumor-bearing mice and mice bearing C26 tumors. In addition, we show that myostatin neutralization with these antibodies results in the preservation of skeletal muscle mass following reduced caloric intake, a common comorbidity associated with advanced cancer. Our findings support the use of neutralizing antimyostatin antibodies as potential therapeutics for cancer-induced muscle wasting. Mol Cancer Ther; 14(7); 1661–70. ©2015 AACR.


Oncotarget | 2018

Merestinib (LY2801653) inhibits neurotrophic receptor kinase (NTRK) and suppresses growth of NTRK fusion bearing tumors

Bruce W. Konicek; Andrew Capen; Kelly M. Credille; Philip J. Ebert; Beverly L. Falcon; Gary L. Heady; Bharvin Patel; Victoria L. Peek; Jennifer R. Stephens; Julie Stewart; Stephanie L. Stout; David E. Timm; Suzane L. Um; Melinda D. Willard; Isabella H. Wulur; Yi Zeng; Yong Wang; Richard A. Walgren; Sau-Chi Betty Yan

Merestinib is an oral multi-kinase inhibitor targeting a limited number of oncokinases including MET, AXL, RON and MKNK1/2. Here, we report that merestinib inhibits neurotrophic receptor tyrosine kinases NTRK1/2/3 which are oncogenic drivers in tumors bearing NTRK fusion resulting from chromosomal rearrangements. Merestinib is shown to be a type II NTRK1 kinase inhibitor as determined by x-ray crystallography. In KM-12 cells harboring TPM3-NTRK1 fusion, merestinib exhibits potent p-NTRK1 inhibition in vitro by western blot and elicits an anti-proliferative response in two- and three-dimensional growth. Merestinib treatment demonstrated profound tumor growth inhibition in in vivo cancer models harboring either a TPM3-NTRK1 or an ETV6-NTRK3 gene fusion. To recapitulate resistance observed from type I NTRK kinase inhibitors entrectinib and larotrectinib, we generated NIH-3T3 cells exogenously expressing TPM3-NTRK1 wild-type, or acquired mutations G595R and G667C in vitro and in vivo. Merestinib blocks tumor growth of both wild-type and mutant G667C TPM3-NTRK1 expressing NIH-3T3 cell-derived tumors. These preclinical data support the clinical evaluation of merestinib, a type II NTRK kinase inhibitor (NCT02920996), both in treatment naïve patients and in patients progressed on type I NTRK kinase inhibitors with acquired secondary G667C mutation in NTRK fusion bearing tumors.


Cancer Research | 2013

Abstract 1131: Novel inhibitor of Notch signaling for the treatment of cancer.

Mark H. Bender; Hong Gao; Andrew Capen; Julia M. Clay; Philip Arthur Hipskind; Jon K. Reel; Jason Manro; Karim A. Benhadji; Bharvin Patel

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC The Notch pathway is a highly conserved signaling system that plays an important role in development and tissue homeostasis. While Notch mutations are well characterized and implicated in hematological malignancies such as T-cell acute lymphoblastic leukemia, mutations in solid tumors were not reported until recently. With the whole genomic deep sequencing of a large number of samples, deregulated Notch signaling has been implicated in a small percentage of solid tumors such as ovarian, lung, and triple negative breast cancer due to genomic alterations including mutations, amplification, and fusion of Notch pathway components. Inhibition of Notch signaling may provide an attractive targeted cancer therapeutic strategy. We have identified and characterized LY3039478 a novel small molecule that is an exquisitely potent inhibitor of Notch-1 intracellular domain (N1ICD) cleavage with an IC50 of ∼1nM in most of the tumor cell lines tested. We also demonstrate that LY3039478 potently inhibits mutant Notch receptor activity. In a xenograft tumor model, LY3039478 inhibited N1ICD cleavage and expression of Notch-regulated genes in the tumor microenvironment. The inhibition of Notch cleavage also resulted in the induction of apoptosis in a Notch-dependent xenograft model. Using extensive PK/PD data we determined the strength and duration of N1ICD cleavage required for anti-tumor activity which was observed in several xenograft tumors including patient derived tumors representing colon, lung, ovarian, gastric, and breast cancer and glioblastoma. To mitigate the mucoid gasteroentropathy caused by Notch inhibition, PK/PD data were incorporated in devising dosing strategies that identified an optimal intermittent dosing schedule without negatively impacting efficacy. Furthermore, the mucoid gastroentropathy was also mitigated by the prophylactic administration of dexamethasone without negatively impacting the Notch inhibitor mediated efficacy. Mechanistic studies revealed that dexamethasone does not interfere with LY3039478-mediated inhibition of N1ICD cleavage and gene expression but alters the expression of stem cell gene expression in GI tract. In summary, we have characterized an orally bio-available small molecule Notch inhibitor that may provide therapeutic benefit to cancer patients with deregulated Notch signaling. LY3039478 is specifically designed to potently inhibit Notch signaling and is being investigated in Phase I. Citation Format: Mark H. Bender, Hong Gao, Andrew R. Capen, Julia M. Clay, Philip A. Hipskind, Jon K. Reel, Maciej J. Zamek-Gliszczynski, Jason R. Manro, Karim Benhadji, Bharvin K. R. Patel. Novel inhibitor of Notch signaling for the treatment of cancer. [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 1131. doi:10.1158/1538-7445.AM2013-1131


Cancer Research | 2017

Abstract 583: The CDK4/6 inhibitor abemaciclib induces synergistic immune activation and antitumor efficacy in combination with PD-L1 blockade

Jack Dempsey; Lysiane Huber; Amelie Forest; Jennifer R. Stephens; Thompson N. Doman; Jason Manro; Andrew Capen; Robert Flack; Gregory P. Donoho; Sean Buchanan; Alfonso De Dios; Kyla Driscoll; Michael Kalos; Ruslan D. Novosiadly; Richard P. Beckmann; David Schaer

Targeting cyclin dependent kinases 4 and 6 (CDK4/6) with inhibitors such as abemaciclib has shown promise in early and late phase clinical trials in both breast cancer and NSCLC. While there is evidence that patients benefit from single-agent abemaciclib, combination strategies leveraging this compound together with immunotherapy are of interest for the treatment of these and other cancers. Consequently, it is important to understand if and how a cell cycle inhibitor can be combined with immunotherapy. However, because most preclinical studies have been performed using xenograft tumors in immune-compromised mice, the potential immunomodulatory effects of abemaciclib have not been adequately ascertained. To investigate the immune combinatorial potential of abemaciclib, we studied the effects of treatment alone and in combination with checkpoint immunotherapy in a murine syngeneic tumor model sensitive to abemaciclib using immuno-competent mice. Abemaciclib monotherapy of established murine CT26 tumors, which harbor KRAS G12C mutation and CDKN2A deletion, caused a dose-dependent delay in tumor growth. Surprisingly, gene expression analysis showed that treatment was associated with an increase in intra-tumor immune inflammation without major alteration in immune subset frequencies. Testing of various dosing regimens in this preclinical model found that monotherapy abemaciclib pretreatment followed by combination with anti-PD-L1 antibody therapy, induced an enhanced anti-tumor response compared to abemaciclib and anti-PD-L1 monotherapies. Optimal combination therapy exhibited superior anti-tumor efficacy, resulting in complete tumor regression (CR) in 50-60% of mice in a setting where anti-PD-L1 monotherapy showed little or no efficacy (0% CRs). Mice which maintained CRs after cessation of combination therapy were able to resist later CT26 rechallenge, demonstrating that abemaciclib in combination with anti-PD-L1 enabled the generation of an immunologic memory. Examination of intra-tumor gene expression during treatment found that combination therapy further amplified the immune/T cell activation signature compared to both monotherapies. Intra-tumoral suppression of cell cycle genes, which are indicative of inhibition of CDK4/6, was also greater during the combination therapy, suggesting that the effects anti-PD-L1 therapy may augment the cell cycle arrest induced by abemaciclib. Although it was uncertain if agents that inhibit cell proliferation could be combined with immunotherapy, these preclinical results demonstrate that it is possible to combine CDK4/6 inhibition by abemaciclib with checkpoint immunotherapy to improve tumor efficacy. The synergistic responses observed in terms of tumor efficacy, immune activation, and cell cycle control provides support for the clinical investigation of this combination. Citation Format: Jack Dempsey, Lysiane Huber, Amelie Forest, Jennifer R. Stephens, Thompson N. Doman, Jason Manro, Andrew Capen, Robert S. Flack, Gregory P. Donoho, Sean Buchanan, Alfonso De Dios, Kyla Driscoll, Michael Kalos, Ruslan Novosiadly, Richard P. Beckmann, David A. Schaer. The CDK4/6 inhibitor abemaciclib induces synergistic immune activation and antitumor efficacy in combination with PD-L1 blockade [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 583. doi:10.1158/1538-7445.AM2017-583


Cancer Research | 2016

Abstract 2647: Merestinib (LY2801653), targeting several oncokinases including NTRK1/2/3, shows potent anti-tumor effect in colorectal cell line- and patient-derived xenograft (PDX) model bearing TPM3-NTRK1 fusion

Bruce W. Konicek; Steve M. Bray; Andrew Capen; John N. Calley; Kelly M. Credille; Philip J. Ebert; Gary L. Heady; Bharvin K. Patel; Victoria L. Peek; Jennifer R. Stephens; Suzane L. Um; Melinda D. Willard; Isabella H. Wulur; Yi Zeng; Richard A. Walgren; Sau-Chi Betty Yan

In cancer, the formation of chimeric gene fusions by genomic rearrangement causes aberrant receptor tyrosine kinase activation resulting in sustained oncogenic signaling driving tumorigenesis. Neurotrophic tyrosine receptor kinase 1 (NTRK1), the cognate receptor for nerve growth factor (NGF), has been reported in 7 tumor types as a NTRK1 kinase domain fused with several reported partners including the 5’ coiled-coil domain of the tropomysin TPM3 gene. The resultant NTRK1 fusion protein is present in about 1.5% of colorectal cancer (CRC), 3% of lung and 12% of papillary thyroid cancers. In addition, gene fusions involving NTRK2 and NTRK3 are present in about 19 different tumor types. Thus pharmacologically targeting NTRK kinase in cancers bearing NTRK fusions may provide treatment options to patients who otherwise might be resistant to standard oncolytic regimens. Merestinib (LY2801653) is an orally bioavailable small molecule inhibitor of several oncokinases, including MET, AXL, ROS1 and MKNK1/2. Merestinib and its two primary metabolites, M1 (LSN2800870) and M2 (LSN2887652) were shown in scanMaxSM kinase binding assays to inhibit all three NTRKs with an IC50 ranging from 15-320 nM, and in the cell-based PathHunter® NTRK1 assay with an IC50 ranging from 12-92 nM. Merestinib, M1 and M2 were evaluated in vitro in TPM3-NTRK1 fusion bearing CRC cells (KM-12). Merestinib, M1 and M2 reduced p-NTRK1 levels, cell proliferation (IC50 of 11 nM, 18 nM and 100 nM respectively) and anchorage independent growth (IC50 of 45 nM, 79 nM and 206 nM respectively). Crizotinib previously reported (Nat Med. 2013;19:1469-72) to have moderate activity against NTRK1, was used to treat a patient with NTRK1 fusion resulted with transient response. Crizotinib was shown here to also reduce p-NTRK1 levels, cell proliferation (IC50 = 88nM) and anchorage independent growth (IC50 = 276nM) in vitro in KM-12 cells. Merestinib treatment at 24 mg/kg once daily arrested tumor growth (T/C = 4%) in KM-12 xenograft tumor bearing mice. Crizotinib administered at 25 mg/kg twice daily in this same model did not result in tumor growth arrest (T/C = 39.5%). Merestinib treatment at 24 mg/kg once daily led to tumor regression in a CRC PDX xenograft model (EL1989) bearing the TPM3-NTRK1 fusion. Crizotinib treatment at 25 mg/kg twice daily in this model did not show tumor regression. Further pre-clinical studies of Merestinib inhibition of NTRK2 and NTRK3 gene fusion are ongoing. These data support the clinical evaluation of Merestinib in patients with tumors harboring NTRK fusion. Merestinib is currently being studied clinically in advanced cancers (NCT01285037). Citation Format: Bruce W. Konicek, Steve M. Bray, Andrew R. Capen, John N. Calley, Kelly M. Credille, Philip J. Ebert, Gary Heady, Bharvin K. Patel, Victoria L. Peek, Jennifer R. Stephens, Suzane L. Um, Melinda D. Willard, Isabella H. Wulur, Yi Zeng, Richard A. Walgren, Sau-Chi Betty Yan. Merestinib (LY2801653), targeting several oncokinases including NTRK1/2/3, shows potent anti-tumor effect in colorectal cell line- and patient-derived xenograft (PDX) model bearing TPM3-NTRK1 fusion. [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 2647.

Collaboration


Dive into the Andrew Capen'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

David Schaer

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge