Network


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

Hotspot


Dive into the research topics where Brian E. Fink is active.

Publication


Featured researches published by Brian E. Fink.


Clinical Cancer Research | 2006

Preclinical antitumor activity of BMS-599626, a pan-HER kinase inhibitor that inhibits HER1/HER2 homodimer and heterodimer signaling.

Tai W. Wong; Francis Y. Lee; Chiang Yu; Feng R. Luo; Simone Oppenheimer; Hongjian Zhang; Richard Smykla; Harold Mastalerz; Brian E. Fink; John T. Hunt; Ashvinikumar V. Gavai; Gregory D. Vite

Purpose: The studies described here are intended to characterize the ability of BMS-599626, a small-molecule inhibitor of the human epidermal growth factor receptor (HER) kinase family, to modulate signaling and growth of tumor cells that depend on HER1 and/or HER2. Experimental Design: The potency and selectivity of BMS-599626 were assessed in biochemical assays using recombinant protein kinases, as well as in cell proliferation assays using tumor cell lines with varying degrees of dependence on HER1 or HER2 signaling. Modulation of receptor signaling was determined in cell assays by Western blot analyses of receptor autophosphorylation and downstream signaling. The ability of BMS-599626 to inhibit receptor heterodimer signaling in tumor cells was studied by receptor coimmunoprecipitation. Antitumor activity of BMS-599626 was evaluated using a number of different xenograft models that represent a spectrum of human tumors with HER1 or HER2 overexpression. Results: BMS-599626 inhibited HER1 and HER2 with IC50 of 20 and 30 nmol/L, respectively, and was highly selective when tested against a broad panel of diverse protein kinases. Biochemical studies suggested that BMS-599626 inhibited HER1 and HER2 through distinct mechanisms. BMS-599626 abrogated HER1 and HER2 signaling and inhibited the proliferation of tumor cell lines that are dependent on these receptors, with IC50 in the range of 0.24 to 1 μmol/L. BMS-599626 was highly selective for tumor cells that depend on HER1/HER2 and had no effect on the proliferation of cell lines that do not express these receptors. In tumor cells that are capable of forming HER1/HER2 heterodimers, BMS-599626 inhibited heterodimerization and downstream signaling. BMS-599626 had antitumor activity in models that overexpress HER1 (GEO), as well as in models that have HER2 gene amplification (KPL4) or overexpression (Sal2), and there was good correlation between the inhibition of receptor signaling and antitumor activity. Conclusions: BMS-599626 is a highly selective and potent inhibitor of HER1 and HER2 kinases and inhibits tumor cell proliferation through modulation of receptor signaling. BMS-599626 inhibits HER1/HER2 receptor heterodimerization and provides an additional mechanism of inhibiting tumors in which receptor coexpression and heterodimerization play a major role in driving tumor growth. The preclinical data support the advancement of BMS-599626 into clinical development for the treatment of cancer.


Clinical Cancer Research | 2011

Antitumor and Antiangiogenic Activities of BMS-690514, an Inhibitor of Human EGF and VEGF Receptor Kinase Families

Tai Wai Wong; Francis Lee; Stuart Emanuel; Craig R. Fairchild; Joseph Fargnoli; Brian E. Fink; Ashvinikumar V. Gavai; Amy Hammell; Benjamin Henley; Christine Hilt; John T. Hunt; Bala Krishnan; Daniel Kukral; Anne Lewin; Harold Malone; Derek J. Norris; Simone Oppenheimer; Gregory D. Vite; Chiang Yu

Purpose: The extensive involvement of the HER kinases in epithelial cancer suggests that kinase inhibitors targeting this receptor family have the potential for broad spectrum antitumor activity. BMS-690514 potently inhibits all three HER kinases, and the VEGF receptor kinases. This report summarizes data from biochemical and cellular pharmacology studies, as well as antitumor activity of BMS-690514. Experimental Design: The potency and selectivity of BMS-690514 was evaluated by using an extensive array of enzymatic and binding assays, as well as cellular assays that measure proliferation and receptor signaling. Antitumor activity was evaluated by using multiple xenograft models that depend on HER kinase signaling. The antiangiogenic properties of BMS-690514 were assessed in a matrigel plug assay, and effect on tumor blood flow was measured by dynamic contrast-enhanced MRI. Results: BMS-690514 is a potent and selective inhibitor of epidermal growth factor receptor (EGFR), HER2, and HER4, as well as the VEGF receptor kinases. It inhibits proliferation of tumor cells with potency that correlates with inhibition of receptor signaling, and induces apoptosis in lung tumor cells that have an activating mutation in EGFR. Antitumor activity was observed with BMS-690514 at multiple doses that are well tolerated in mice. There was evidence of suppression of tumor angiogenesis and endothelial function by BMS-690514, which may contribute to its efficacy. Conclusions: By combining inhibition of two receptor kinase families, BMS-690524 is a novel targeted agent that disrupts signaling in the tumor and its vasculature. Clin Cancer Res; 17(12); 4031–41. ©2011 AACR.


Journal of Medicinal Chemistry | 2009

Discovery and preclinical evaluation of [4-[[1-(3-fluorophenyl)methyl]-1H-indazol-5-ylamino]-5-methylpyrrolo[2,1-f][1,2,4]triazin-6-yl]carbamic acid, (3S)-3-morpholinylmethyl ester (BMS-599626), a selective and orally efficacious inhibitor of human epidermal growth factor receptor 1 and 2 kinases.

Ashvinikumar V. Gavai; Brian E. Fink; David J. Fairfax; Gregory Scott Martin; Lana M. Rossiter; Christian L. Holst; Soong-Hoon Kim; Kenneth J. Leavitt; Harold Mastalerz; Wen-Ching Han; Derek J. Norris; Bindu Goyal; Shankar Swaminathan; Bharat P. Patel; Arvind Mathur; Dolatrai M. Vyas; John S. Tokarski; Chiang Yu; Simone Oppenheimer; Hongjian Zhang; Punit Marathe; Joseph Fargnoli; Francis Y. Lee; Tai W. Wong; Gregory D. Vite

Structure-activity relationships in a series of 4-[1H-indazol-5-ylamino]pyrrolo[2,1-f][1,2,4]triazine-6-carbamates identified dual human epidermal growth factor receptor (HER)1/HER2 kinase inhibitors with excellent biochemical potency and kinase selectivity. On the basis of its favorable pharmacokinetic profile and robust in vivo activity in HER1 and HER2 driven tumor models, 13 (BMS-599626) was selected as a clinical candidate for treatment of solid tumors.


Bioorganic & Medicinal Chemistry Letters | 2011

Novel pyrrolo[2,1-f][1,2,4]triazin-4-amines: Dual inhibitors of EGFR and HER2 protein tyrosine kinases

Brian E. Fink; Derek J. Norris; Harold Mastalerz; Ping Chen; Bindu Goyal; Yufen Zhao; Soong-Hoon Kim; Gregory D. Vite; Francis Y. Lee; Hongjian Zhang; Simone Oppenheimer; John S. Tokarski; Tai W. Wong; Ashvinikumar V. Gavai

A novel series of 5-((4-aminopiperidin-1-yl)methyl)-pyrrolo[2,1-f][1,2,4]triazin-4-amines with small aniline substituents at the C4 position were optimized for dual EGFR and HER2 protein tyrosine kinase inhibition. Compound 8l exhibited promising oral efficacy in both EGFR and HER2-driven human tumor xenograft models.


Molecular Cancer Therapeutics | 2006

Constitutively active receptor tyrosine kinases as oncogenes in preclinical models for cancer therapeutics

Kristen A. Kellar; Matthew V. Lorenzi; Ching Ping Ho; Dan You; Mei-Li Wen; Rolf Peter Ryseck; Simone Oppenheimer; Brian E. Fink; Gregory D. Vite; Bruce R. Rowley; Chiang Yu; David K. Bol; Francis Y. Lee; Tai W. Wong

Receptor tyrosine kinases (RTK) remain an area of therapeutic interest because of their role in epithelial tumors, and experimental models specific to these targets are highly desirable. Chimeric receptors were prepared by in-frame fusion of the CD8 extracellular sequence with the cytoplasmic sequences of RTKs. A CD8HER2 fusion protein was shown to form disulfide-mediated homodimers and to transform fibroblasts and epithelial cells. CD8RTK fusion proteins transform rat kidney epithelial cells and impart phenotypes that may reflect signaling specificity inherent in the native receptors. Transgenic expression of CD8HER2 and CD8Met in mice resulted in the formation of salivary and mammary gland tumors. The transgenic tumors allow the derivation of allograft tumors and cell lines that are sensitive to inhibition by small molecule kinase inhibitors. This approach provides excellent cell and tumor models for the characterization of signaling properties of diverse RTKs and for the evaluation of rationally designed antagonists targeting these kinases. [Mol Cancer Ther 2006;5(6):1571–6]


ACS Medicinal Chemistry Letters | 2018

Discovery of 4-Azaindole Inhibitors of TGFβRI as Immuno-oncology Agents

Yong Zhang; Yufen Zhao; Andrew J. Tebben; Steven Sheriff; Max Ruzanov; Mark P. Fereshteh; Yi Fan; Jonathan Lippy; Jesse Swanson; Ching-Ping Ho; Barri Wautlet; Anne Rose; Karen Parrish; Zheng Yang; Andrew F. Donnell; Liping Zhang; Brian E. Fink; Gregory D. Vite; Karen Augustine-Rauch; Joseph Fargnoli; Robert M. Borzilleri

The multifunctional cytokine TGFβ plays a central role in regulating antitumor immunity. It has been postulated that inhibition of TGFβ signaling in concert with checkpoint blockade will provide improved and durable immune response against tumors. Herein, we describe a novel series of 4-azaindole TGFβ receptor kinase inhibitors with excellent selectivity for TGFβ receptor 1 kinase. The combination of compound 3f and an antimouse-PD-1 antibody demonstrated significantly improved antitumor efficacy compared to either treatment alone in a murine tumor model.


Cancer Research | 2015

Abstract 5395: Anti-tumor activity of BMS-595, a novel CK2 kinase inhibitor

Brent A. Rupnow; Chiang Yu; Jonathan G. Pabalan; Urvashi V. Roongta; Jonathan Lippy; Ashok Dongre; Mary T. Obermeier; Aberra Fura; Paul A. Elzinga; Benjamin Henley; Joseph Fargnoli; Francis Y. Lee; William R. Foster; Christine M. Tarby; Brian E. Fink; John S. Tokarski; Ashvinikumar V. Gavai; Tai W. Wong; John T. Hunt; Gregory D. Vite; Ashok V. Purandare

The CK2 protein kinases are a small family of two highly related serine/threonine kinases composed of two catalytic subunits, α and α’, and a single β subunit. Numerous substrates have been reported for CK2 and these proteins are known to participate in diverse cellular processes, including cell signaling, transcription, DNA repair, apoptosis regulation and tumor suppression. Elevated CK2 expression and kinase activity has been observed in many cancer types. Further, mRNA knockdown and enzyme inhibition studies have demonstrated that many cancer cell lines are dependent on CK2 for growth and survival. To further evaluate CK2 kinases as targets for therapeutic intervention in cancer, we identified BMS-595, a potent and selective, ATP-competitive CK2 inhibitor. BMS-595 inhibits the in vitro proliferation of human colorectal and lung cancer cell lines with IC50s ranging from less than 10 nM to greater than 1 μM. In sensitive cell lines, anti-proliferative effects of BMS-595 and structurally related analogs strongly correlated with cellular CK2 kinase inhibition. Oral administration of BMS-595 to mice bearing colorectal cancer and lung cancer xenografts demonstrated pharmacodynamic effects and robust efficacy at tolerated doses. These studies confirm the dependence of a subset of human colon and lung cancer cell lines on CK2 activity for growth and demonstrate that pharmacologic inhibition of CK2 can produce anti-tumor efficacy at tolerated doses. Citation Format: Brent A. Rupnow, Chiang Yu, Jonathan G. Pabalan, Urvashi V. Roongta, Jonathan S. Lippy, Ashok R. Dongre, Mary T. Obermeier, Aberra Fura, Paul A. Elzinga, Benjamin J. Henley, Joseph Fargnoli, Francis Y. Lee, William R. Foster, Christine M. Tarby, Brian E. Fink, John S. Tokarski, Ashvinikumar V. Gavai, Tai W. Wong, John T. Hunt, Gregory D. Vite, Ashok V. Purandare. Anti-tumor activity of BMS-595, a novel CK2 kinase inhibitor. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5395. doi:10.1158/1538-7445.AM2015-5395


Cancer Research | 2015

Abstract 5417: The identification of BMS-595, an orally active imidazo[1,2-b]pyridazine CK2 inhibitor with in vivo anti-tumor activity

Christine M. Tarby; Liqi He; Brian E. Fink; Andrew Nation; Yufen Zhao; Soong-Hoon Kim; Libing Chen; John S. Tokarski; Chiang Yu; Jonathan G. Pabalan; Urvashi V. Roongta; Jonathan Lippy; Mary T. Obermeier; Paul A. Elzinga; Aberra Fura; Benjamin Henley; Joseph Fargnoli; William R. Foster; Ashvinikumar V. Gavai; Tai W. Wong; John T. Hunt; Gregory D. Vite; Ashok V. Purandare; Brent A. Rupnow

CK2 is a highly conserved, and constitutively active family of serine/threonine kinases abnormally elevated in a wide variety of cancers and linked to poor prognosis and disease progression. The enzymes form as hetero-tetrameric complexes comprised of two highly related catalytic subunits (α or α´) with two regulatory β subunits in various combinations and distributions, depending on cell type. While CK2 plays a role in normal growth and development, deregulation of the enzymes has been shown to promote and maintain a malignant phenotype through mechanisms in both the anti-apoptotic and the pro-proliferative signaling pathways. CK2 has been reported to modulate the activity of several oncogenic transcription factors including CREB, Myc, Jun and Fos. Studies with RNAi and small molecule compounds have demonstrated tumor cell dependence on CK2. We sought to identify potent CK2 inhibitors to probe the function of CK2 in cancer-linked pathways and for evaluation in CK2 dependent tumor xenograft models. Herein we report SAR studies in the imidazo[1,2-b]pyridazine chemotype leading to the discovery of BMS-595, a highly potent and selective ATP-competitive CK2 inhibitor with a commensurate level of cellular potency. BMS-595 demonstrates strong PK/PD correlations and robust, oral anti-tumor efficacy in CK2-driven xenograft models at tolerated doses. Citation Format: Christine M. Tarby, Liqi He, Brian E. Fink, Andrew Nation, Yufen Zhao, Soong-Hoon Kim, Libing Chen, John S. Tokarski, Chiang Yu, Jonathan G. Pabalan, Urvashi V. Roongta, Jonathan Lippy, Mary Obermeier, Paul A. Elzinga, Aberra Fura, Benjamin Henley, Joseph J. Fargnoli, William R. Foster, Ashvinikumar V. Gavai, Tai W. Wong, John T. Hunt, Gregory D. Vite, Ashok V. Purandare, Brent A. Rupnow. The identification of BMS-595, an orally active imidazo[1,2-b]pyridazine CK2 inhibitor with in vivo anti-tumor activity. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5417. doi:10.1158/1538-7445.AM2015-5417


Cancer Research | 2014

Abstract 1643: BMS-983970, an oral pan-Notch inhibitor for the treatment of cancer

Ashvinikumar V. Gavai; Yufen Zhao; Daniel O'Malley; Brian E. Fink; Claude A. Quesnelle; Derek J. Norris; Libing Chen; Soong-Hoon Kim; Wen-Ching Han; Patrice Gill; Weifang Shan; Aaron Balog; Andrew J. Tebben; Richard Rampulla; Dauh-Rurng Wu; Yingru Zhang; Arvind Mathur; Haiqing Wang; Zheng Yang; Qian Ruan; Robin Moore; David Rodrigues; Asoka Ranasinghe; Celia D'Arienzo; Ching Kim Tye; Ching Su; Gerry Everlof; Melissa Yarde; Mary Ellen Cvijic; Krista Menard

Deregulation of the Notch pathway has been shown to be oncogenic in numerous tissue types including T-cell acute lymphoblastic leukemia (T-ALL), breast cancer, non-small cell lung cancer, and colorectal carcinoma. Notch signal activation can cause uncontrolled proliferation, restrict differentiation leading to increased self-renewal capacity, evasion of apoptosis, and enhancement of angiogenesis and metastasis. There is increasing evidence that Notch plays a role in the maintenance and survival of cancer stem cells. γ-Secretase mediates the Notch signaling pathway by releasing the Notch intracellular domain (NICD) which translocates to the nucleus and binds to the transcription factor CSL to activate transcription of various target genes. BMS-906024 is a potent pan-Notch inhibitor that demonstrated robust anti-tumor activity at tolerated doses in multiple tumor xenograft models. It is being evaluated in Phase 1 clinical studies. BMS-906024 is being administered IV (once weekly) in the clinic and the projected human efficacious dose is 4 - 6 mg. Based on the preclinical data, the projected human half-life of BMS-906024 is in the 37 h - 124 h range. This presentation will describe further structure-activity relationships in the 1,4-benzodiazepinone series that culminated in the identification of BMS-983970 as an oral-pan-Notch inhibitor. Pharmacokinetic properties and in vivo evaluation of BMS-983970 in T-ALL and solid tumor xenograft models will be presented. Citation Format: Ashvinikumar V. Gavai, Yufen Zhao, Daniel O9Malley, Brian Fink, Claude Quesnelle, Derek Norris, Libing Chen, Soong-Hoon Kim, Wen-Ching Han, Patrice Gill, Weifang Shan, Aaron Balog, Andrew Tebben, Richard Rampulla, Dauh-Rurng Wu, Yingru Zhang, Arvind Mathur, Haiqing Wang, Zheng Yang, Qian Ruan, Robin Moore, David Rodrigues, Asoka Ranasinghe, Celia D9Arienzo, Ching Kim Tye, Ching Su, Gerry Everlof, Melissa Yarde, Mary Ellen Cvijic, Krista Menard, Mei-Li Wen, George Trainor, Bruce Fischer, John Hunt, Gregory Vite, Richard Westhouse, Francis Lee. BMS-983970, an oral pan-Notch inhibitor for the treatment of cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1643. doi:10.1158/1538-7445.AM2014-1643


Molecular Cancer Therapeutics | 2013

Abstract B284: Discovery of imidazopyridazinecarbonitriles as potent, selective inhibitors of CK2.

Brian E. Fink; Ashvinikumar V. Gavai; Soong-Hoon Kim; Yufen Zhao; Ashok V. Purandare; Gregory D. Vite; John S. Tokarski; Chiang Yu; Benjamin Henley; Joseph Fargnoli; Heshani Desilva; Petra Ross-Macdonald; Brent A. Rupnow; Tai W. Wong

Casein kinase 2 (CK2) is a serine/threonine kinase that has been implicated in the regulation of a number of oncogenic or tumor suppressor proteins. CK2 activity has been shown to be elevated in numerous studies in a variety of cancer types. CK2 has been shown to phosphorylate numerous cellular proteins. Among the proteins regulated directly or indirectly by CK2 phosphorylation are oncogenes and tumor suppressor proteins including beta-catenin, c-Myc, PML, and PTEN as well as proteins directly involved in cell cycle, apoptosis, and transcriptional regulation. The plethora of CK2 substrates and their participation in various cellular processes is a major confounding factor in understanding the role of CK2 in oncogenesis. Unlike other kinase targets that participate in relatively linear growth factor signaling pathways, CK2 appears to function more “laterally,” across many important signaling pathways to promote growth and survival of cancer cells. It is clear from numerous studies using siRNA as well as small molecule CK2 inhibitors that cancer cells are highly dependent upon CK2 for growth and survival. As a result, new small molecule inhibitors of CK2 may provide useful tools for probing CK2 biology and may also provide therapeutic benefits against several cancer types. Herein, we report our efforts toward the identification of CK2 inhibitors based on an imidazopyridazine carbonitrile scaffold. Lead compounds from this series demonstrate low nanomolar CK2 biochemical potency, while achieving excellent selectivity versus the majority of kinases in the human kinome. The development of structure-activity relationships and the establishment of a strong correlation between biochemical potency, inhibition of cellular protein target phosphorylation and anti-proliferative effects in targeted cancer cell lines will be presented. In addition, the optimization of pharmacokinetic properties resulting in compounds with excellent in vivo exposure has allowed for the investigation of CK2 inhibition in a pharmacodynamic model. Finally, our efforts to elucidate pathway effects mediated by CK2 in model colon cancer cell lines, including gene expression profiling using advanced small molecules leads, will be disclosed. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):B284. Citation Format: Brian Fink, Ashvinikumar Gavai, Soong-Hoon Kim, Yufen Zhao, Ashok Purandare, Gregory Vite, John Tokarski, Chiang Yu, Benjamin Henley, Joseph Fargnoli, Heshani Desilva, Petra Ross-MacDonald, Brent Rupnow, Tai W. Wong. Discovery of imidazopyridazinecarbonitriles as potent, selective inhibitors of CK2. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr B284.

Collaboration


Dive into the Brian E. Fink'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