Network


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

Hotspot


Dive into the research topics where Wanjun Zheng is active.

Publication


Featured researches published by Wanjun Zheng.


Cancer Research | 2011

Eribulin Induces Irreversible Mitotic Blockade: Implications of Cell-Based Pharmacodynamics for In vivo Efficacy under Intermittent Dosing Conditions

Murray J. Towle; Kathleen A. Salvato; Bruce F. Wels; Kimberley K. Aalfs; Wanjun Zheng; Boris M. Seletsky; Xiaojie Zhu; Bryan M. Lewis; Yoshito Kishi; Melvin J. Yu; Bruce A. Littlefield

Eribulin (E7389), a mechanistically unique microtubule inhibitor in phase III clinical trials for cancer, exhibits superior efficacy in vivo relative to the more potent compound ER-076349, a fact not explained by different pharmacokinetic properties. A cell-based pharmacodynamic explanation was suggested by observations that mitotic blockade induced by eribulin, but not ER-076349, is irreversible as measured by a flow cytometric mitotic block reversibility assay employing full dose/response treatment. Cell viability 5 days after drug washout established relationships between mitotic block reversibility and long-term cell survival. Similar results occurred in U937, Jurkat, HL-60, and HeLa cells, ruling out cell type-specific effects. Studies with other tubulin agents suggest that mitotic block reversibility is a quantifiable, compound-specific characteristic of antimitotic agents in general. Bcl-2 phosphorylation patterns parallel eribulin and ER-076349 mitotic block reversibility patterns, suggesting persistent Bcl-2 phosphorylation contributes to long-term cell-viability loss after eribulins irreversible blockade. Drug uptake and washout/retention studies show that [3H]eribulin accumulates to lower intracellular levels than [3H]ER-076349, yet is retained longer and at higher levels. Similar findings occurred with irreversible vincristine and reversible vinblastine, pointing to persistent cellular retention as a component of irreversibility. Our results suggest that eribulins in vivo superiority derives from its ability to induce irreversible mitotic blockade, which appears related to persistent drug retention and sustained Bcl-2 phosphorylation. More broadly, our results suggest that compound-specific reversibility characteristics of antimitotic agents contribute to interactions between cell-based pharmacodynamics and in vivo pharmacokinetics that define antitumor efficacy under intermittent dosing conditions.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Therapeutical targeting of nucleic acid-sensing Toll-like receptors prevents experimental cerebral malaria

Bernardo S. Franklin; Sally T. Ishizaka; Marc Lamphier; Fabian Gusovsky; Hans Hansen; Jeffrey M. Rose; Wanjun Zheng; Marco Antonio Ataide; Rosane B. de Oliveira; Douglas T. Golenbock; Ricardo T. Gazzinelli

Excessive release of proinflammatory cytokines by innate immune cells is an important component of the pathogenic basis of malaria. Proinflammatory cytokines are a direct output of Toll-like receptor (TLR) activation during microbial infection. Thus, interference with TLR function is likely to render a better clinical outcome by preventing their aberrant activation and the excessive release of inflammatory mediators. Herein, we describe the protective effect and mechanism of action of E6446, a synthetic antagonist of nucleic acid-sensing TLRs, on experimental cerebral malaria (ECM) induced by Plasmodium berghei ANKA. We show that in vitro, low doses of E6446 specifically inhibited the activation of human and mouse TLR9. Tenfold higher concentrations of this compound also inhibited the human TLR8 response to single-stranded RNA. In vivo, therapy with E6446 diminished the activation of TLR9 and prevented the exacerbated cytokine response observed during acute Plasmodium infection. Furthermore, severe signs of ECM, such as limb paralysis, brain vascular leak, and death, were all prevented by oral treatment with E6446. Hence, we provide evidence that supports the involvement of nucleic acid-sensing TLRs in malaria pathogenesis and that interference with the activation of these receptors is a promising strategy to prevent deleterious inflammatory responses that mediate pathogenesis and severity of malaria.


Molecular Pharmacology | 2014

Novel small molecule inhibitors of TLR7 and TLR9: mechanism of action and efficacy in vivo

Marc Lamphier; Wanjun Zheng; Eicke Latz; Mark Spyvee; Hans Hansen; Jeffrey Rose; Melinda Genest; Hua Yang; Christina J. Shaffer; Yan Zhao; Yongchun Shen; Carrie Liu; Diana Liu; Thorsten R. Mempel; Christopher Rowbottom; Jesse Chow; Natalie C. Twine; Melvin J. Yu; Fabian Gusovsky; Sally T. Ishizaka

The discovery that circulating nucleic acid-containing complexes in the serum of autoimmune lupus patients can stimulate B cells and plasmacytoid dendritic cells via Toll-like receptors 7 and 9 suggested that agents that block these receptors might be useful therapeutics. We identified two compounds, AT791 {3-[4-(6-(3-(dimethylamino)propoxy)benzo[d]oxazol-2-yl)phenoxy]-N,N-dimethylpropan-1-amine} and E6446 {6-[3-(pyrrolidin-1-yl)propoxy)-2-(4-(3-(pyrrolidin-1-yl)propoxy)phenyl]benzo[d]oxazole}, that inhibit Toll-like receptor (TLR)7 and 9 signaling in a variety of human and mouse cell types and inhibit DNA-TLR9 interaction in vitro. When administered to mice, these compounds suppress responses to challenge doses of cytidine-phosphate-guanidine (CpG)–containing DNA, which stimulates TLR9. When given chronically in spontaneous mouse lupus models, E6446 slowed development of circulating antinuclear antibodies and had a modest effect on anti–double-stranded DNA titers but showed no observable impact on proteinuria or mortality. We discovered that the ability of AT791 and E6446 to inhibit TLR7 and 9 signaling depends on two properties: weak interaction with nucleic acids and high accumulation in the intracellular acidic compartments where TLR7 and 9 reside. Binding of the compounds to DNA prevents DNA-TLR9 interaction in vitro and modulates signaling in vivo. Our data also confirm an earlier report that this same mechanism may explain inhibition of TLR7 and 9 signaling by hydroxychloroquine (Plaquenil; Sanofi-Aventis, Bridgewater, NJ), a drug commonly prescribed to treat lupus. Thus, very different structural classes of molecules can inhibit endosomal TLRs by essentially identical mechanisms of action, suggesting a general mechanism for targeting this group of TLRs.


Annual Reports in Medicinal Chemistry | 2011

Case History: Discovery of Eribulin (HALAVEN™), a Halichondrin B Analogue That Prolongs Overall Survival in Patients with Metastatic Breast Cancer

Melvin J. Yu; Wanjun Zheng; Boris M. Seletsky; Bruce A. Littlefield; Yoshito Kishi

Publisher Summary This chapter focuses on the discovery and development of eribulin. Eribulin mesylate (HALAVEN) is an antitubulin antimitotic agent with distinct microtubule end-binding properties that result in inhibition of microtubule dynamics in ways that differ from those of vinblastine and paclitaxel. This agent was recently approved by the U.S. Food and Drug Administration (FDA) for use in patients with metastatic breast cancer who have previously received at least two chemotherapeutic regimens for the treatment of metastatic disease. HALAVEN represents a new and exciting treatment option that for the first time has been shown to improve overall survival in heavily pretreated women with late stage breast cancer. The path leading to the discovery and development of eribulin included close three-way collaboration among the Kishi group at Harvard University, the National Cancer Institute (NCI), and Eisai. At many points along the drug discovery path, setbacks and roadblocks arose that threatened to derail the program. Nevertheless, each of the problems was solved in turn, thereby demonstrating that a structurally complex molecule could be optimized through total synthesis to successfully deliver a marketed drug that meets all pharmacological, toxicological, and physicochemical requirements.


Bioorganic & Medicinal Chemistry Letters | 2011

Novel second generation analogs of eribulin. Part II: Orally available and active against resistant tumors in vivo.

Sridhar Narayan; Eric M. Carlson; Hongsheng Cheng; Krista Condon; Hong Du; Sean Eckley; Yongbo Hu; Yimin Jiang; Vipul Kumar; Bryan M. Lewis; Philip Saxton; Edgar Schuck; Boris M. Seletsky; Karen TenDyke; Huiming Zhang; Wanjun Zheng; Bruce A. Littlefield; Murray J. Towle; Melvin J. Yu

Eribulin mesylate is a newly approved treatment for locally advanced and metastatic breast cancer. We targeted oral bioavailability and efficacy against multidrug resistant (MDR) tumors for further work. The design, synthesis and evaluation of novel amine-containing analogs of eribulin mesylate are described in this part. Attenuation of basicity of the amino group(s) in the C32 side-chain region led to compounds with low susceptibility to PgP-mediated drug efflux. These compounds were active against MDR tumor cell lines in vitro and in xenograft models in vivo, in addition to being orally bioavailable.


Bioorganic & Medicinal Chemistry Letters | 2011

Novel second generation analogs of eribulin. Part I: Compounds containing a lipophilic C32 side chain overcome P-glycoprotein susceptibility

Sridhar Narayan; Eric M. Carlson; Hongsheng Cheng; Hong Du; Yongbo Hu; Yimin Jiang; Bryan M. Lewis; Boris M. Seletsky; Karen TenDyke; Huiming Zhang; Wanjun Zheng; Bruce A. Littlefield; Murray J. Towle; Melvin J. Yu

Eribulin mesylate (Halaven™), a totally synthetic analog of the marine polyether macrolide halichondrin B, has recently been approved in the United States as a treatment for breast cancer. It is also currently under regulatory review in Japan and the European Union. Our continuing medicinal chemistry efforts on this scaffold have focused on oral bioavailability, brain penetration and efficacy against multidrug resistant (MDR) tumors by lowering the susceptibility of these compounds to P-glycoprotein (P-gp)-mediated drug efflux. Replacement of the 1,2-amino alcohol C32 side chain of eribulin with fragments neutral at physiologic pH led to the identification of analogs with significantly lower P-gp susceptibility. The analogs maintained low- to sub-nM potency in vitro against both sensitive and MDR cell lines. Within this series, increasing lipophilicity generally led to decreased P-gp susceptibility. In addition to potency in cell culture, these compounds showed in vivo activity in mouse xenograft models.


Bioorganic & Medicinal Chemistry Letters | 2011

Novel second generation analogs of eribulin. Part III: Blood-brain barrier permeability and in vivo activity in a brain tumor model.

Sridhar Narayan; Eric M. Carlson; Hongsheng Cheng; Krista Condon; Hong Du; Sean Eckley; Yongbo Hu; Yimin Jiang; Vipul Kumar; Bryan M. Lewis; Philip Saxton; Edgar Schuck; Boris M. Seletsky; Karen TenDyke; Huiming Zhang; Wanjun Zheng; Bruce A. Littlefield; Murray J. Towle; Melvin J. Yu

Novel second generation analogs of eribulin mesylate, a tubulin agent recently approved for the treatment of breast cancer, are reported. Our recent efforts have focused on expanding the target indications for this class of compounds to other tumor types. Herein, we describe the design, synthesis and evaluation of eribulin analogs active against brain tumor cell lines in vitro and corresponding brain tumor models in mice. Attenuation of basicity of the amino group(s) in the C32 side-chain region led to compounds with lower susceptibility to P-gp mediated drug efflux, allowing these compounds to permeate through the blood-brain barrier. In preclinical in vivo studies, these compounds showed significantly higher levels in the brain and cerebrospinal fluid as compared to eribulin. In addition, analogs within this series showed antitumor activity in an orthotopic murine model of human glioblastoma.


Cancer Biology & Therapy | 2015

Small molecule schweinfurthins selectively inhibit cancer cell proliferation and mTOR/AKT signaling by interfering with trans-Golgi-network trafficking

Xingfeng Bao; Wanjun Zheng; Naoko Hata Sugi; Kishan Agarwala; Qunli Xu; Zichun Wang; Karen TenDyke; Winnie Lee; Lana Parent; Wei Li; Hongsheng Cheng; Yongchun Shen; Noel Taylor; Zoltan Dezso; Hong Du; Yoshihiko Kotake; Nanding Zhao; John Wang; Maarten Postema; Mary Woodall-Jappe; Yasutaka Takase; Toshimitsu Uenaka; David G. I. Kingston; Kenichi Nomoto

Natural compound schweinfurthins are of considerable interest for novel therapy development because of their selective anti-proliferative activity against human cancer cells. We previously reported the isolation of highly active schweinfurthins E-H, and in the present study, mechanisms of the potent and selective anti-proliferation were investigated. We found that schweinfurthins preferentially inhibited the proliferation of PTEN deficient cancer cells by indirect inhibition of AKT phosphorylation. Mechanistically, schweinfurthins and their analogs arrested trans-Golgi-network trafficking, an intracellular vesicular trafficking system, resulting in the induction of endoplasmic reticulum stress and the suppression of both lipid raft-mediated PI3K activation and mTOR/RheB complex formation, which collectively led to an effective inhibition of mTOR/AKT signaling. The trans-Golgi-network traffic arresting effect of schweinfurthins was associated with their in vitro binding activity to oxysterol-binding proteins that are known to regulate intracellular vesicular trafficking. Moreover, schweinfurthins were found to be highly toxic toward PTEN-deficient B cell lymphoma cells, and displayed 2 orders of magnitude lower activity toward normal human peripheral blood mononuclear cells and primary fibroblasts in vitro. These results revealed a previously unrecognized role of schweinfurthins in regulating trans-Golgi-network trafficking, and linked mechanistically this cellular effect with mTOR/AKT signaling and with cancer cell survival and growth. Our findings suggest the schweinfurthin class of compounds as a novel approach to modulate oncogenic mTOR/AKT signaling for cancer treatment.


Bioorganic & Medicinal Chemistry Letters | 2012

Atom-based enumeration: new eribulin analogues with low susceptibility to P-glycoprotein-mediated drug efflux.

Melvin J. Yu; Wanjun Zheng; Karen TenDyke

A series of eribulin analogues was evolved in silico through iterative atom-based enumeration employing a genetic algorithm-derived survival function to minimize predicted PgP-mediated drug efflux. Representatives of the virtual series were subsequently synthesized in the laboratory and tested in vitro for PgP-susceptibility. These new computer-inspired derivatives were found to exhibit high cell growth inhibitory activity and to be among the least sensitive to P-glycoprotein-mediated drug efflux in the eribulin series, thereby validating this approach to in silico molecular design.


OncoImmunology | 2017

EP4 Antagonism by E7046 diminishes Myeloid immunosuppression and synergizes with Treg-reducing IL-2-Diphtheria toxin fusion protein in restoring anti-tumor immunity

Diana I. Albu; Zichun Wang; Kuan-Chun Huang; Jiayi Wu; Natalie C. Twine; Sarah Leacu; Christy Ingersoll; Lana Parent; Winnie Lee; Diana Liu; Renee Wright-Michaud; Namita Kumar; Galina Kuznetsov; Qian Chen; Wanjun Zheng; Kenichi Nomoto; Mary Woodall-Jappe; Xingfeng Bao

ABSTRACT Reprogramming of immunosuppressive tumor microenvironment (TME) by targeting alternatively activated tumor associated macrophages (M2TAM), myeloid-derived suppressor cells (MDSC), and regulatory T cells (Tregs), represents a promising strategy for developing novel cancer immunotherapy. Prostaglandin E2 (PGE2), an arachidonic acid pathway metabolite and mediator of chronic inflammation, has emerged as a powerful immunosuppressor in the TME through engagement with one or more of its 4 receptors (EP1-EP4). We have developed E7046, an orally bioavailable EP4-specific antagonist and show here that E7046 has specific and potent inhibitory activity on PGE2-mediated pro-tumor myeloid cell differentiation and activation. E7046 treatment reduced the growth or even rejected established tumors in vivo in a manner dependent on both myeloid and CD8+ T cells. Furthermore, co-administration of E7046 and E7777, an IL-2-diphtheria toxin fusion protein that preferentially kills Tregs, synergistically disrupted the myeloid and Treg immunosuppressive networks, resulting in effective and durable anti-tumor immune responses in mouse tumor models. In the TME, E7046 and E7777 markedly increased ratios of CD8+granzymeB+ cytotoxic T cells (CTLs)/live Tregs and of M1-like/M2TAM, and converted a chronic inflammation phenotype into acute inflammation, shown by substantial induction of STAT1/IRF-1 and IFNγ-controlled genes. Notably, E7046 also showed synergistic anti-tumor activity when combined with anti-CTLA-4 antibodies, which have been reported to diminish intratumoral Tregs. Our studies thus reveal a specific myeloid cell differentiation-modifying activity by EP4 blockade and a novel combination of E7046 and E7777 as a means to synergistically mitigate both myeloid and Treg-derived immunosuppression for cancer treatment in preclinical models.

Collaboration


Dive into the Wanjun Zheng's collaboration.

Researchain Logo
Decentralizing Knowledge