Network


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

Hotspot


Dive into the research topics where Murray J. Towle is active.

Publication


Featured researches published by Murray J. Towle.


Cancer Research | 2004

Induction of Morphological and Biochemical Apoptosis following Prolonged Mitotic Blockage by Halichondrin B Macrocyclic Ketone Analog E7389

Galina Kuznetsov; Murray J. Towle; Hongsheng Cheng; Takanori Kawamura; Karen TenDyke; Diana Liu; Yoshito Kishi; Melvin J. Yu; Bruce A. Littlefield

E7389, a macrocyclic ketone analog of the marine natural product halichondrin B, currently is undergoing clinical trials for cancer. This fully synthetic agent exerts its highly potent in vitro and in vivo anticancer effects via tubulin-based antimitotic mechanisms, which are similar or identical to those of parental halichondrin B. In an attempt to understand the impressive potency of E7389 in animal models of human cancer, its ability to induce apoptosis following prolonged mitotic blockage was evaluated. Treatment of U937 human histiocytic lymphoma cells with E7389 led to time-dependent collection of cells in the G2-M phase of the cell cycle, beginning as early as 2 h and becoming maximal by 12 h. Increased numbers of hypodiploid events were seen beginning at 12 h, suggesting initiation of apoptosis after prolonged E7389-induced mitotic blockage. The identity of hypodiploid events as apoptotic cells under these conditions was confirmed by two additional morphologic criteria: green to orange/yellow shifts on acridine orange/ethidium bromide staining, and cell surface annexin V binding as assessed by flow cytometry. Several biochemical correlates of apoptosis also were seen following E7389 treatment, including phosphorylation of the antiapoptotic protein Bcl-2, cytochrome c release from mitochondria, proteolytic activation of caspase-3 and -9, and cleavage of the caspase-3 substrate poly(ADP-ribose) polymerase (PARP). In LNCaP human prostate cancer cells, treatment with E7389 also led to generation of hypodiploid cells, activation of caspase-3 and -9, and appearance of cleaved PARP, indicating that E7389 can activate cellular apoptosis pathways under anchorage-independent and -dependent cell culture conditions. These results show that prolonged mitotic blockage by E7389 can lead to apoptotic cell death of human cancer cells in vitro and can provide a mechanistic basis for the significant in vivo anticancer efficacy of E7389.


Cancer Research | 2011

Comparison of Neuropathy-Inducing Effects of Eribulin Mesylate, Paclitaxel, and Ixabepilone in Mice

Krystyna M. Wozniak; Kenichi Nomoto; Rena G. Lapidus; Ying Wu; Valentina Alda Carozzi; Guido Cavaletti; Kazuhiro Hayakawa; Satoru Hosokawa; Murray J. Towle; Bruce A. Littlefield; Barbara S. Slusher

Chemotherapy-induced neurotoxicity is a significant problem associated with successful treatment of many cancers. Tubulin is a well-established target of antineoplastic therapy; however, tubulin-targeting agents, such as paclitaxel and the newer epothilones, induce significant neurotoxicity. Eribulin mesylate, a novel microtubule-targeting analogue of the marine natural product halichondrin B, has recently shown antineoplastic activity, with relatively low incidence and severity of neuropathy, in metastatic breast cancer patients. The mechanism of chemotherapy-induced neuropathy is not well understood. One of the main underlying reasons is incomplete characterization of pathology of peripheral nerves from treated subjects, either from patients or preclinically from animals. The current study was conducted to directly compare, in mice, the neuropathy-inducing propensity of three drugs: paclitaxel, ixabepilone, and eribulin mesylate. Because these drugs have different potencies and pharmacokinetics, we compared them on the basis of a maximum tolerated dose (MTD). Effects of each drug on caudal and digital nerve conduction velocity, nerve amplitude, and sciatic nerve and dorsal root ganglion morphology at 0.25 × MTD, 0.5 × MTD, 0.75 × MTD, and MTD were compared. Paclitaxel and ixabepilone, at their respective MTDs, produced significant deficits in caudal nerve conduction velocity, caudal amplitude and digital nerve amplitudes, as well as moderate to severe degenerative pathologic changes in dorsal root ganglia and sciatic nerve. In contrast, eribulin mesylate produced no significant deleterious effects on any nerve conduction parameter measured and caused milder, less frequent effects on morphology. Overall, our findings indicate that eribulin mesylate induces less neuropathy in mice than paclitaxel or ixabepilone at equivalent MTD-based doses.


Molecular Cancer Therapeutics | 2009

Potent in vitro and in vivo anticancer activities of des-methyl, des-amino pateamine A, a synthetic analogue of marine natural product pateamine A

Galina Kuznetsov; Qunli Xu; Lori Rudolph-Owen; Karen TenDyke; Junke Liu; Murray J. Towle; Nanding Zhao; Joanne Marsh; Sergei Agoulnik; Natalie C. Twine; Lana Parent; Zhihong Chen; Jue-lon Shie; Yimin Jiang; Huiming Zhang; Hong Du; Roch Boivin; Yuan Wang; Daniel Romo; Bruce A. Littlefield

We report here that des-methyl, des-amino pateamine A (DMDA-PatA), a structurally simplified analogue of the marine natural product pateamine A, has potent antiproliferative activity against a wide variety of human cancer cell lines while showing relatively low cytotoxicity against nonproliferating, quiescent human fibroblasts. DMDA-PatA retains almost full in vitro potency in P-glycoprotein-overexpressing MES-SA/Dx5-Rx1 human uterine sarcoma cells that are significantly resistant to paclitaxel, suggesting that DMDA-PatA is not a substrate for P-glycoprotein-mediated drug efflux. Treatment of proliferating cells with DMDA-PatA leads to rapid shutdown of DNA synthesis in the S phase of the cell cycle. Cell-free studies show that DMDA-PatA directly inhibits DNA polymerases α and γ in vitro albeit at concentrations considerably higher than those that inhibit cell proliferation. DMDA-PatA shows potent anticancer activity in several human cancer xenograft models in nude mice, including significant regressions observed in the LOX and MDA-MB-435 melanoma models. DMDA-PatA thus represents a promising natural product-based anticancer agent that warrants further investigation.[Mol Cancer Ther 2009;8(5):1250–60]


Bioorganic & Medicinal Chemistry | 1993

The synthesis of three 4-substitutedbenzo[b]thiophene-2-carboxamidines as potent and selective inhibitors of urokinase

Alexander J. Bridges; Arthur Lee; C.Eric Schwartz; Murray J. Towle; Bruce A. Littlefield

Efficient synthesis of structurally novel 4-substituted benzo[b]thiophene-2-carboxamidines 1-3, which selectively inhibit urokinase-type plasminogen activator (uPA) with IC50 values of 70-320 nM, are described. The key intermediate, methyl 4-iodobenzo[b]thiophene-2-carboxylate (7), is prepared from 3-fluoroiodobenzene in two steps in 70% overall yield via fluorine-directed metalation/formylation and subsequent thiophene annulation. Amidination of ester 7 gives the 320 nM inhibitor 1. Palladium-catalyzed arylacetylene and vinyl stannane couplings with ester 7 or 4-iodobenzo[b]thiophene-2-carbonitrile (16, derived from 7), respectively, followed by amidination leads to the more potent inhibitors 2 (IC50 = 133 nM) and 3 (IC50 = 70 nM). These compounds represent an important new class of synthetic uPA inhibitors, with carboxamidine 3 being the most potent selective inhibitor currently described in the literature.


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.


Anticancer Research | 2018

Broad-spectrum Preclinical Antitumor Activity of Eribulin (Halaven®): Combination with Anticancer Agents of Differing Mechanisms

Makoto Asano; Junji Matsui; Murray J. Towle; Jiayi Wu; Sharon McGonigle; Marc Hillairet De Boisferon; Toshimitsu Uenaka; Kenichi Nomoto; Bruce A. Littlefield

Background: Eribulin is used in many countries to treat patients with advanced breast cancer or liposarcoma and exerts in vivo anticancer activity under monotherapy conditions against various human tumor xenograft models. Here, eribulin in combination with mechanistically different anticancer agents was evaluated. Materials and Methods: Eribulin was combined with cytotoxic agents (capecitabine, carboplatin, cisplatin, doxorubicin, gemcitabine) or targeted agents (bevacizumab, BKM-120, E7449, erlotinib, everolimus, lenvatinib, palbociclib) in tumor xenograft models of breast cancer, melanoma, non-small cell lung cancer (NSCLC), and ovarian cancer. Results: Across nearly all models, eribulin with either cytotoxic or targeted agents demonstrated combination activity, defined as the activity demonstrably greater than that of either agent alone. Combination activity was absent only with doxorubicin (MDA-MB-435 model) and with lenvatinib (NCI-H1975 model), both of which responded to the agents as monotherapy. Conclusion: Eribulin has combination activity with multiple agents from different mechanistic classes in several human cancer models, including breast, NSCLC, ovarian, and melanoma.


Cancer Research | 2010

Abstract 4438: Comparison of neuropathy-inducing effects of eribulin versus paclitaxel in mice

Krystyna M. Wozniak; Rena G. Lapidus; Ying Wu; Valentina Alda Carozzi; Guido Cavalleti; Kazuhiro Hayakawa; Satoru Hosokawa; Murray J. Towle; Bruce A. Littlefield; Kenichi Nomoto; Barbara S. Slusher

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC Eribulin (referred to as E7389 in the mesylate salt form) is a structurally simplified analog of the potent anticancer agent halichondrin B. Eribulin inhibits microtubule dynamics via a mechanism that is distinct from that of other tubulin-targeting drugs. Eribulin has recently shown activity in Phase III trials for advanced metastatic breast cancer. Current standard treatment for breast cancer includes paclitaxel, whose major dose-limiting toxicity is peripheral neuropathy. This study was conducted to directly compare the neuropathy-producing effects of paclitaxel versus eribulin in mice. Maximum tolerated dose (MTD) levels using a 3x/week intravenous regimen (Q2D x 3) for 2 weeks, were determined for each compound. Neuropathy studies were then conducted in mice dosed at the respective MTDs or fractions thereof (0.75, 0.5 and 0.25 of the MTD), or with vehicle control. Mice were evaluated for nerve conduction velocity (NCV) and amplitude in caudal and digital nerves 24 hr after their last treatment. Sciatic nerves and dorsal root ganglia (DRG) were subsequently removed for pathological assessment. After 2 weeks dosing with paclitaxel, clear dose-dependent decreases in caudal NCV and amplitude were evident. At MTD (30 mg/kg), paclitaxel caused a significant reduction in caudal NCV compared to control (18 +/− 3.8%; p<0.01), while both MTD and 0.75 MTD doses caused significant reduction in caudal amplitude compared to control (63.5 +/− 4.8% and 48.8 +/− 4.2%, respectively; p<0.001). Both MTD and 0.75 MTD paclitaxel also caused significant reduction in digital nerve amplitude (62.8 +/− 4.1 % and 35.2 +/− 4.3 %, p<0.001 and p<0.05 respectively). In contrast, after 2 weeks dosing with eribulin, there were no decreases in caudal or digital NCV or amplitude at any dose, including the MTD (1.75 mg/kg). Microscopic examination revealed that paclitaxel induced moderate to severe dose-dependent pathological changes in sciatic nerve and DRG at doses of 0.5 MTD or higher. Eribulin produced some mild pathological changes in DRG neurons and sciatic nerve, which although less severe, were clearly evident at the 0.75 MTD and MTD doses. In conclusion, eribulin administered to mice intravenously at doses up to and including the MTD, during an intensive two week regimen induced no significant reduction in nerve conduction velocity or amplitude in caudal and digital nerves, findings that contrast markedly with the significant decreases in nerve conduction velocity and amplitude induced by paclitaxel. In addition, eribulin caused less severe morphological changes in DRG and sciatic nerves at equivalent MTD-based doses. Overall, these preclinical findings suggest that eribulin may be less likely to induce neuropathy than paclitaxel. Note: This abstract was not presented at the AACR 101st Annual Meeting 2010 because the presenter was unable to attend. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4438.


Cancer Research | 2001

In Vitro and In Vivo Anticancer Activities of Synthetic Macrocyclic Ketone Analogues of Halichondrin B

Murray J. Towle; Kathleen A. Salvato; Jacqueline Budrow; Bruce F. Wels; Galina Kuznetsov; Kimberley K. Aalfs; Susan Welsh; Wanjun Zheng; Boris M. Seletsky; Monica H. Palme; Gregory J. Habgood; Lori A. Singer; Lucian Dipietro; Yuan Wang; Jack J. Chen; David A. Quincy; Ashley Davis; Kentaro Yoshimatsu; Yoshito Kishi; Melvin J. Yu; Bruce A. Littlefield

Collaboration


Dive into the Murray J. Towle's collaboration.

Researchain Logo
Decentralizing Knowledge