Network


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

Hotspot


Dive into the research topics where Bryan M. Lewis is active.

Publication


Featured researches published by Bryan M. Lewis.


Biochemistry | 2010

Eribulin Binds at Microtubule Ends to a Single Site on Tubulin to Suppress Dynamic Instability

Jennifer A. Smith; Leslie Wilson; Olga Azarenko; Xiaojie Zhu; Bryan M. Lewis; Bruce A. Littlefield; Mary Ann Jordan

Eribulin mesylate (E7389), a synthetic analogue of the marine natural product halichondrin B, is in phase III clinical trials for the treatment of cancer. Eribulin targets microtubules, suppressing dynamic instability at microtubule plus ends through an inhibition of microtubule growth with little or no effect on shortening [Jordan, M. A., et al. (2005) Mol. Cancer Ther. 4, 1086-1095]. Using [(3)H]eribulin, we found that eribulin binds soluble tubulin at a single site; however, this binding is complex with an overall K(d) of 46 microM, but also showing a real or apparent very high affinity (K(d) = 0.4 microM) for a subset of 25% of the tubulin. Eribulin also binds microtubules with a maximum stoichiometry of 14.7 +/- 1.3 molecules per microtubule (K(d) = 3.5 microM), strongly suggesting the presence of a relatively high-affinity binding site at microtubule ends. At 100 nM, the concentration that inhibits microtubule plus end growth by 50%, we found that one molecule of eribulin is bound per two microtubules, indicating that the binding of a single eribulin molecule at a microtubule end can potently inhibit its growth. Eribulin does not suppress dynamic instability at microtubule minus ends. Preincubation of microtubules with 2 or 4 microM vinblastine induced additional lower-affinity eribulin binding sites, most likely at splayed microtubule ends. Overall, our results indicate that eribulin binds with high affinity to microtubule plus ends and thereby suppresses dynamic instability.


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.


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.


Synlett | 2013

Commercial Manufacture of Halaven®: Chemoselective Transformations En Route to Structurally Complex Macrocyclic Ketones

Brian Austad; Trevor L. Calkins; Charles E. Chase; Francis G. Fang; Thomas E. Horstmann; Yongbo Hu; Bryan M. Lewis; Xiang Niu; Thomas A. Noland; John D. Orr; Matthew J. Schnaderbeck; Huiming Zhang; Naoki Asakawa; Naoki Asai; Hiroyuki Chiba; Takashi Hasebe; Yorihisa Hoshino; Hiroyuki Ishizuka; Takashi Kajima; Akio Kayano; Yuki Komatsu; Manabu Kubota; Hirofumi Kuroda; Mamoru Miyazawa; Katsuya Tagami; Tomohiro Watanabe


Synlett | 2013

Process Development of Halaven®: Synthesis of the C1–C13 Fragment from d-(–)-Gulono-1,4-lactone

Charles E. Chase; Francis G. Fang; Bryan M. Lewis; Gordon D. Wilkie; Matthew J. Schnaderbeck; Xiaojie Zhu


Synlett | 2013

Process Development of Halaven®: Synthesis of the C14–C35 Fragment via Iterative Nozaki–Hiyama–Kishi Reaction–Williamson Ether Cyclization

Brian Austad; Farid Benayoud; Trevor L. Calkins; Silvio Campagna; Charles E. Chase; Hyeong-wook Choi; William J. Christ; Robert Costanzo; James Cutter; Atsushi Endo; Francis G. Fang; Yongbo Hu; Bryan M. Lewis; Michael D. Lewis; Shawn McKenna; Thomas A. Noland; John D. Orr; Marc Pesant; Matthew J. Schnaderbeck; Gordon D. Wilkie; Taichi Abe; Naoki Asai; Yumi Asai; Akio Kayano; Yuichi Kimoto; Yuki Komatsu; Manabu Kubota; Hirofumi Kuroda; Masanori Mizuno; Taiju Nakamura


Archive | 2004

Reagents and methods for labeling terminal olefins

Thomas E. Horstmann; Bryan M. Lewis


Archive | 2014

Methods useful in the synthesis of halichondrin b analogs

Bryan M. Lewis; Yongbo Hu; Huiming Zhang; Hiroyuki Chiba; Yuki Komatsu

Collaboration


Dive into the Bryan M. Lewis's collaboration.

Researchain Logo
Decentralizing Knowledge