Network


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

Hotspot


Dive into the research topics where Weihe Zhang is active.

Publication


Featured researches published by Weihe Zhang.


Journal of Clinical Investigation | 2013

MERTK receptor tyrosine kinase is a therapeutic target in melanoma

Jennifer Schlegel; Maria J. Sambade; Susan Sather; Stergios J. Moschos; Aik Choon Tan; Amanda Winges; Deborah DeRyckere; Craig Carson; Dimitri G. Trembath; John J. Tentler; S. Gail Eckhardt; Pei Fen Kuan; Ronald L. Hamilton; Lyn M. Duncan; C. Ryan Miller; Nana Nikolaishvili-Feinberg; Bentley R. Midkiff; Jing Liu; Weihe Zhang; Chao Yang; Xiaodong Wang; Stephen V. Frye; H. Shelton Earp; Janiel M. Shields; Douglas K. Graham

Metastatic melanoma is one of the most aggressive forms of cutaneous cancers. Although recent therapeutic advances have prolonged patient survival, the prognosis remains dismal. C-MER proto-oncogene tyrosine kinase (MERTK) is a receptor tyrosine kinase with oncogenic properties that is often overexpressed or activated in various malignancies. Using both protein immunohistochemistry and microarray analyses, we demonstrate that MERTK expression correlates with disease progression. MERTK expression was highest in metastatic melanomas, followed by primary melanomas, while the lowest expression was observed in nevi. Additionally, over half of melanoma cell lines overexpressed MERTK compared with normal human melanocytes; however, overexpression did not correlate with mutations in BRAF or RAS. Stimulation of melanoma cells with the MERTK ligand GAS6 resulted in the activation of several downstream signaling pathways including MAPK/ERK, PI3K/AKT, and JAK/STAT. MERTK inhibition via shRNA reduced MERTK-mediated downstream signaling, reduced colony formation by up to 59%, and diminished tumor volume by 60% in a human melanoma murine xenograft model. Treatment of melanoma cells with UNC1062, a novel MERTK-selective small-molecule tyrosine kinase inhibitor, reduced activation of MERTK-mediated downstream signaling, induced apoptosis in culture, reduced colony formation in soft agar, and inhibited invasion of melanoma cells. This work establishes MERTK as a therapeutic target in melanoma and provides a rationale for the continued development of MERTK-targeted therapies.


Journal of Medicinal Chemistry | 2014

UNC2025, a Potent and Orally Bioavailable MER/FLT3 Dual Inhibitor

Weihe Zhang; Deborah DeRyckere; Debra Hunter; Jing Liu; Michael A. Stashko; Katherine A. Minson; Christopher T. Cummings; Trevor G. Glaros; Dianne Newton; Susan Sather; Dehui Zhang; Dmitri Kireev; William P. Janzen; H. Shelton Earp; Douglas K. Graham; Stephen V. Frye; Xiaodong Wang

We previously reported a potent small molecule Mer tyrosine kinase inhibitor UNC1062. However, its poor PK properties prevented further assessment in vivo. We report here the sequential modification of UNC1062 to address DMPK properties and yield a new potent and highly orally bioavailable Mer inhibitor, 11, capable of inhibiting Mer phosphorylation in vivo, following oral dosing as demonstrated by pharmaco-dynamic (PD) studies examining phospho-Mer in leukemic blasts from mouse bone marrow. Kinome profiling versus more than 300 kinases in vitro and cellular selectivity assessments demonstrate that 11 has similar subnanomolar activity against Flt3, an additional important target in acute myelogenous leukemia (AML), with pharmacologically useful selectivity versus other kinases examined.


European Journal of Medicinal Chemistry | 2013

UNC1062, a new and potent Mer inhibitor.

Jing Liu; Weihe Zhang; Michael A. Stashko; Deborah DeRyckere; Christopher T. Cummings; Debra Hunter; Chao Yang; Chatura N. Jayakody; Nancy Cheng; Catherine Simpson; Jacqueline Norris-Drouin; Susan Sather; Dmitri Kireev; William P. Janzen; H. Shelton Earp; Douglas K. Graham; Stephen V. Frye; Xiaodong Wang

Abnormal activation of Mer kinase has been implicated in the oncogenesis of many human cancers including acute lymphoblastic and myeloid leukemia, non-small cell lung cancer, and glioblastoma. We have discovered a new family of small molecule Mer inhibitors, pyrazolopyrimidine sulfonamides, that potently inhibit the kinase activity of Mer. Importantly, these compounds do not demonstrate significant hERG activity in the PatchXpress assay. Through structure-activity relationship studies, 35 (UNC1062) was identified as a potent (IC50 = 1.1 nM) and selective Mer inhibitor. When applied to live tumor cells, UNC1062 inhibited Mer phosphorylation and colony formation in soft agar. Given the potential of Mer as a therapeutic target, UNC1062 is a promising candidate for further drug development.


Journal of Medicinal Chemistry | 2013

Discovery of Mer Specific Tyrosine Kinase Inhibitors for the Treatment and Prevention of Thrombosis

Weihe Zhang; Andrew L. McIver; Michael A. Stashko; Deborah DeRyckere; Brian R. Branchford; Debra Hunter; Dmitri Kireev; Michael J. Miley; Jacqueline Norris-Drouin; Wendy M. Stewart; Susan Sather; Yingqiu Zhou; Jorge Di Paola; Mischa Machius; William P. Janzen; H. Shelton Earp; Douglas K. Graham; Stephen V. Frye; Xiaodong Wang

The role of Mer kinase in regulating the second phase of platelet activation generates an opportunity to use Mer inhibitors for preventing thrombosis with diminished likelihood for bleeding as compared to current therapies. Toward this end, we have discovered a novel, Mer kinase specific substituted-pyrimidine scaffold using a structure-based drug design and a pseudo ring replacement strategy. The cocrystal structure of Mer with two compounds (7 and 22) possessing distinct activity have been determined. Subsequent SAR studies identified compound 23 (UNC2881) as a lead compound for in vivo evaluation. When applied to live cells, 23 inhibits steady-state Mer kinase phosphorylation with an IC50 value of 22 nM. Treatment with 23 is also sufficient to block EGF-mediated stimulation of a chimeric receptor containing the intracellular domain of Mer fused to the extracellular domain of EGFR. In addition, 23 potently inhibits collagen-induced platelet aggregation, suggesting that this class of inhibitors may have utility for prevention and/or treatment of pathologic thrombosis.


Journal of Medicinal Chemistry | 2013

Pseudo-Cyclization through Intramolecular Hydrogen Bond Enables Discovery of Pyridine Substituted Pyrimidines as New Mer Kinase Inhibitors

Weihe Zhang; Dehui Zhang; Michael A. Stashko; Deborah DeRyckere; Debra Hunter; Dmitri Kireev; Michael J. Miley; Christopher T. Cummings; Jacqueline Norris-Drouin; Wendy M. Stewart; Susan Sather; Yingqiu Zhou; Gregory Kirkpatrick; Mischa Machius; William P. Janzen; H. Shelton Earp; Douglas K. Graham; Stephen V. Frye; Xiaodong Wang

Abnormal activation or overexpression of Mer receptor tyrosine kinase has been implicated in survival signaling and chemoresistance in many human cancers. Consequently, Mer is a promising novel cancer therapeutic target. A structure-based drug design approach using a pseudo-ring replacement strategy was developed and validated to discover a new family of pyridinepyrimidine analogues as potent Mer inhibitors. Through SAR studies, 10 (UNC2250) was identified as the lead compound for further investigation based on high selectivity against other kinases and good pharmacokinetic properties. When applied to live cells, 10 inhibited steady-state phosphorylation of endogenous Mer with an IC50 of 9.8 nM and blocked ligand-stimulated activation of a chimeric EGFR-Mer protein. Treatment with 10 also resulted in decreased colony-forming potential in rhabdoid and NSCLC tumor cells, thereby demonstrating functional antitumor activity. The results provide a rationale for further investigation of this compound for therapeutic application in patients with cancer.


Molecular Cancer Therapeutics | 2015

Small molecule inhibition of MERTK is efficacious in non-small cell lung cancer models independent of driver oncogene status

Christopher T. Cummings; Weihe Zhang; Kurtis D. Davies; Gregory D. Kirkpatrick; Dehui Zhang; Deborah DeRyckere; Xiaodong Wang; Stephen V. Frye; H. Shelton Earp; Douglas K. Graham

Treatment of non–small cell lung cancer (NSCLC) has been transformed by targeted therapies directed against molecular aberrations specifically activated within an individual patients tumor. However, such therapies are currently only available against a small number of such aberrations, and new targets and therapeutics are needed. Our laboratory has previously identified the MERTK receptor tyrosine kinase (RTK) as a potential drug target in multiple cancer types, including NSCLC. We have recently developed UNC2025—the first-in-class small molecule inhibitor targeting MERTK with pharmacokinetic properties sufficient for clinical translation. Here, we utilize this compound to further validate the important emerging biologic functions of MERTK in lung cancer pathogenesis, to establish that MERTK can be effectively targeted by a clinically translatable agent, and to demonstrate that inhibition of MERTK is a valid treatment strategy in a wide variety of NSCLC lines independent of their driver oncogene status, including in lines with an EGFR mutation, a KRAS/NRAS mutation, an RTK fusion, or another or unknown driver oncogene. Biochemically, we report the selectivity of UNC2025 for MERTK, and its inhibition of oncogenic downstream signaling. Functionally, we demonstrate that UNC2025 induces apoptosis of MERTK-dependent NSCLC cell lines, while decreasing colony formation in vitro and tumor xenograft growth in vivo in murine models. These findings provide further evidence for the importance of MERTK in NSCLC, and demonstrate that MERTK inhibition by UNC2025 is a feasible, clinically relevant treatment strategy in a wide variety of NSCLC subtypes, which warrants further investigation in clinical trials. Mol Cancer Ther; 14(9); 2014–22. ©2015 AACR.


Clinical Cancer Research | 2017

UNC2025, a MerTK small molecule inhibitor, is therapeutically effective alone and in combination with methotrexate in leukemia models

Deborah DeRyckere; Alisa B. Lee-Sherick; Madeline G. Huey; Amanda A. Hill; Jeffrey W. Tyner; Kristen M. Jacobsen; Lauren S. Page; Gregory G. Kirkpatrick; Fatma Eryildiz; Stephanie A. Montgomery; Weihe Zhang; Xiaodong Wang; Stephen V. Frye; H. Shelton Earp; Douglas K. Graham

Purpose: MERTK tyrosine kinase is ectopically expressed in 30% to 50% of acute lymphoblastic leukemias (ALL) and more than 80% of acute myeloid leukemias (AML) and is a potential therapeutic target. Here, we evaluated the utility of UNC2025, a MERTK tyrosine kinase inhibitor, for treatment of acute leukemia. Experimental Design: Preclinical in vitro and in vivo assays using cell lines and primary leukemia patient samples were used to evaluate antileukemic effects of UNC2025. Results: UNC2025 potently inhibited prosurvival signaling, induced apoptosis, and reduced proliferation and colony formation in MERTK-expressing ALL and AML cell lines and patient samples. Approximately 30% of primary leukemia patient samples (78 of 261 total) were sensitive to UNC2025. Sensitive samples were most prevalent in the AML, T-ALL, and minimally differentiated (M0) AML subsets. UNC2025 inhibited MERTK in bone marrow leukemia cells and had significant therapeutic effects in xenograft models, with dose-dependent decreases in tumor burden and consistent two-fold increases in median survival, irrespective of starting disease burden. In a patient-derived AML xenograft model, treatment with UNC2025 induced disease regression. In addition, UNC2025 increased sensitivity to methotrexate in vivo, suggesting that addition of MERTK-targeted therapy to current cytotoxic regimens may be particularly effective and/or allow for chemotherapy dose reduction. Conclusions: The broad-spectrum activity mediated by UNC2025 in leukemia patient samples and xenograft models, alone or in combination with cytotoxic chemotherapy, supports continued development of MERTK inhibitors for treatment of leukemia. Clin Cancer Res; 23(6); 1481–92. ©2016 AACR.


JCI insight | 2016

The MERTK/FLT3 inhibitor MRX-2843 overcomes resistance-conferring FLT3 mutations in acute myeloid leukemia

Katherine A. Minson; Catherine C. Smith; Deborah DeRyckere; Clara Libbrecht; Alisa B. Lee-Sherick; Madeline G. Huey; Elisabeth Lasater; Gregory Kirkpatrick; Michael A. Stashko; Weihe Zhang; Craig T. Jordan; Dmitri Kireev; Xiaodong Wang; Stephen V. Frye; H. Shelton Earp; Neil P. Shah; Douglas K. Graham

FMS-like tyrosine kinase 3-targeted (FLT3-targeted) therapies have shown initial promise for the treatment of acute myeloid leukemia (AML) expressing FLT3-activating mutations; however, resistance emerges rapidly. Furthermore, limited options exist for the treatment of FLT3-independent AML, demonstrating the need for novel therapies that reduce toxicity and improve survival. MERTK receptor tyrosine kinase is overexpressed in 80% to 90% of AMLs and contributes to leukemogenesis. Here, we describe MRX-2843, a type 1 small-molecule tyrosine kinase inhibitor that abrogates activation of both MERTK and FLT3 and their downstream effectors. MRX-2843 treatment induces apoptosis and inhibits colony formation in AML cell lines and primary patient samples expressing MERTK and/or FLT3-ITD, with a wide therapeutic window compared with that of normal human cord blood cells. In murine orthotopic xenograft models, once-daily oral therapy prolonged survival 2- to 3-fold over that of vehicle-treated controls. Additionally, MRX-2843 retained activity against quizartinib-resistant FLT3-ITD-mutant proteins with clinically relevant alterations at the D835 or F691 loci and prolonged survival in xenograft models of quizartinib-resistant AML. Together, these observations validate MRX-2843 as a translational agent and support its clinical development for the treatment of AML.


ChemMedChem | 2017

Discovery of Macrocyclic Pyrimidines as MerTK-Specific Inhibitors

Andrew L. McIver; Weihe Zhang; Qingyang Liu; Xinpeng Jiang; Michael A. Stashko; James Nichols; Michael J. Miley; Jacqueline Norris-Drouin; Mischa Machius; Deborah DeRyckere; Edgar R. Wood; Douglas K. Graham; H. Shelton Earp; Dmitri Kireev; Stephen V. Frye; Xiaodong Wang

Macrocycles have attracted significant attention in drug discovery recently. In fact, a few de novo designed macrocyclic kinase inhibitors are currently in clinical trials with good potency and selectivity for their intended target. In this study, we successfully engaged a structure‐based drug design approach to discover macrocyclic pyrimidines as potent Mer tyrosine kinase (MerTK)‐specific inhibitors. An enzyme‐linked immunosorbent assay (ELISA) in 384‐well format was employed to evaluate the inhibitory activity of macrocycles in a cell‐based assay assessing tyrosine phosphorylation of MerTK. Through structure–activity relationship (SAR) studies, analogue 11 [UNC2541; (S)‐7‐amino‐N‐(4‐fluorobenzyl)‐8‐oxo‐2,9,16‐triaza‐1(2,4)‐pyrimidinacyclohexadecaphane‐1‐carboxamide] was identified as a potent and MerTK‐specific inhibitor that exhibits sub‐micromolar inhibitory activity in the cell‐based ELISA. In addition, an X‐ray structure of MerTK protein in complex with 11 was resolved to show that these macrocycles bind in the MerTK ATP pocket.


Cancer Research | 2014

Abstract 1736: A novel Mer tyrosine kinase inhibitor mediates increased cell killing in combination with FGFR inhibition

Timothy P Newton; Christopher T. Cummings; Gregory Kirkpatrick; Trista K. Hinz; Deborah DeRyckere; Weihe Zhang; Xiaodong Wang; Stephen V. Frye; H. Shelton Earp; Lynn E. Heasley; Douglas K. Graham

Purpose of Study: Although therapies targeting recently identified oncogenic drivers of non-small cell lung cancer (NSCLC) are in clinical use, a significant proportion of patients still lack a molecularly-targeted therapeutic option. Therefore, there is a continued need for development of new therapeutic strategies. We recently demonstrated oncogenic roles for Mer receptor tyrosine kinase in NSCLC. More specifically, we showed aberrant expression of Mer in approximately 70% of NSCLC patient samples compared to normal lung. Additionally, shRNA-mediated Mer inhibition resulted in increased cell death, decreased colony formation in clonogenic assays, increased chemosensitivity, and decreased tumorigenesis in murine xenografts. These data validate Mer as a potential therapeutic target in NSCLC. Fibroblast growth factor receptors (FGFR) are another class of tyrosine kinases that are aberrantly expressed and function to promote tumorigenesis in NSCLC. FGFR inhibition has been validated as a therapeutic strategy in preclinical NSCLC models and several FGFR inhibitors are currently in clinical development for treatment of NSCLC. Although Mer can signal through both the MEK/ERK and PI3K/AKT pathways, in NSCLC Mer signals mainly through PI3K/AKT. Because Mer and FGFR signal primarily through complementary pathways that mediate survival and proliferation (PI3K/AKT and MEK/ERK, respectively), we hypothesized that dual inhibition of FGFR and Mer may provide a therapeutic advantage relative to inhibition of either kinase alone. In this study we investigated the interaction between a novel Mer-selective small molecule tyrosine kinase inhibitor (TKI) and AZD-4547, an FGFR TKI, in NSCLC cell lines. Methods Used: Colo699 (Mer+, FGFR+) and H226 (Mer+, FGFR+) NSCLC cells were cultured for 14 days in soft agar in the presence of Mer TKI and/or AZD-4547, alone or in combination, and colonies were stained and counted. Changes in the activity of downstream signaling pathways, including PI3K/AKT, MEK/ERK, and STAT proteins were evaluated by immunoblotting. Results and Conclusions: In the soft agar assay, Colo699 and H226 colony formation was inhibited in the presence of Mer TKI and AZD-4547, both as single agents and in combination. Importantly, concurrent treatment with Mer TKI and AZD-4547 resulted in a greater decrease in colony-formation relative to either single agent. Immunoblotting revealed increased inhibition of pro-survival signaling in cells treated with both inhibitors relative to the single agents. Taken together, these data suggest that combination therapies targeting Mer kinase and FGFR may be effective for treatment of NSCLC and indicate biochemical mechanisms by which the combination therapy may mediate increased anti-tumor activity. Citation Format: Timothy P. Newton, Christopher T. Cummings, Gregory D. Kirkpatrick, Trista K. Hinz, Deborah DeRyckere, Weihe Zhang, Xiaodong Wang, Stephen Frye, H. Shelton Earp, Lynn Heasley, Douglas K. Graham. A novel Mer tyrosine kinase inhibitor mediates increased cell killing in combination with FGFR inhibition. [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 1736. doi:10.1158/1538-7445.AM2014-1736

Collaboration


Dive into the Weihe Zhang's collaboration.

Top Co-Authors

Avatar

Xiaodong Wang

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Stephen V. Frye

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Douglas K. Graham

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Deborah DeRyckere

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

H. Shelton Earp

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Dmitri Kireev

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Michael A. Stashko

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jing Liu

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Susan Sather

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Jacqueline Norris-Drouin

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge