Network


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

Hotspot


Dive into the research topics where Charles Duke is active.

Publication


Featured researches published by Charles Duke.


Pharmaceutical Research | 2009

Micellar Delivery of Bicalutamide and Embelin for Treating Prostate Cancer

Michael Danquah; Feng Li; Charles Duke; Duane D. Miller; Ram I. Mahato

PurposeTo examine the effect of bicalutamide and embelin on the growth of prostate cancer cells in vitro and in vivoMethodsCell viability was determined by MTT assay. Micelles were fabricated with polyethylene glycol-b-polylactic acid (PEG-PLA) copolymer and characterized in terms of particle size, micellar solubilization and drug loading, followed by evaluation in nude mice bearing LNCaP xenografts.ResultsEmbelin induced caspase 3 and 9 activation in LNCaP and C4–2 cells by decreasing XIAP expression and was more potent than bicalutamide in killing prostate tumor cells irrespective of their androgen status. As analyzed by isobologram analysis the combination of bicalutamide and embelin was synergistic for C4–2 but additive and slightly antagonistic for LNCaP cells. Micellar formulation resulted in at least 60-fold increase in the aqueous solubility of bicalutamide and embelin. Tumor growth was effectively regressed upon treatment with bicalutamide, but the extent of tumor regression was significantly higher when bicalutamide was formulated in micelles. However, tumor response to bicalutamide stopped after prolonged treatment and began to grow. Sequential treatment with XIAP inhibitor embelin resulted in regression of these hormone refractory tumors.ConclusionCombined treatment with bicalutamide and embelin may be an effective strategy for treating hormone refractory prostate cancer.


Journal of Medicinal Chemistry | 2011

Unexpected binding orientation of bulky-B-ring anti-androgens and implications for future drug targets.

Charles Duke; Amanda Jones; Casey E. Bohl; James T. Dalton; Duane D. Miller

Several new androgen receptor antagonists were synthesized and found to have varying activities across typically anti-androgen resistant mutants (Thr877 → Ala and Trp741 → Leu) and markedly improved potency over previously reported pan-antagonists. X-ray crystallography of a new anti-androgen in an androgen receptor mutant (Thr877 → Ala) shows that the receptor can accommodate the added bulk presented by phenyl to naphthyl substitution, casting doubt on previous reports of predicted binding orientation and the causes of antagonism in bulky-B-ring antagonists.


Expert Opinion on Therapeutic Patents | 2012

Androgen receptor antagonists: A patent review (2008 - 2011)

Michael L. Mohler; Christopher C. Coss; Charles Duke; Shivaputra Patil; Duane D. Miller; James T. Dalton

Introduction: Androgen receptor (AR) antagonists are predominantly used as chemical castration to treat prostate cancer (i.e., in conjunction with androgen deprivation therapy (ADT)). Unfortunately, castration-resistant prostate cancer (CRPC) typically develops that is refractory to targeted therapy. Insights into CRPC biology have led to the emergence of a promising clinical candidate MDV3100 (Figure 2) and a resurgence in this field. A pipeline of preclinical competitive (C-terminally directed) antagonists was discovered using a variety of innovative screening paradigms. Some inhibit nuclear translocation, selectively downregulate or degrade AR (SARD), antagonize wild-type and escape mutant AR (pan-antagonists) and/or antagonize AR target organs in vivo. Separately, the N-terminal domain has emerged as a promising novel target for noncompetitive antagonists. Areas covered: AR antagonists whose patents published between 2008 and 2011 are reviewed. Antagonists are organized based on the screening paradigm reported as discussed above. Expert opinion: Novel mechanisms provide a more informed basis for selecting a competitive antagonist; however, high potency and favorable in vivo properties remain paramount. Noncompetitive antagonists have theoretical advantages suggestive of improved clinical efficacy, but no clinical proof of concept as of yet.


Journal of Pharmacology and Experimental Therapeutics | 2010

Nonsteroidal Selective Androgen Receptor Modulators Enhance Female Sexual Motivation

Amanda Jones; Dong Jin Hwang; Charles Duke; Yali He; Anjaiah Siddam; Duane D. Miller; James T. Dalton

Women experience a decline in estrogen and androgen levels after natural or surgically induced menopause, effects that are associated with a loss of sexual desire and bone mineral density. Studies in our laboratories have shown the beneficial effects of selective androgen receptor modulators (SARMs) in the treatment of osteoporosis and muscle wasting in animal models. A series of S-3-(phenoxy)-2-hydroxy-2-methyl-N-(4-cyano-3-trifluoromethyl-phenyl)-propionamide analogs was synthesized to evaluate the effects of B-ring substitutions on in vitro and in vivo pharmacologic activity, especially female sexual motivation. The androgen receptor (AR) relative binding affinities ranged from 0.1 to 26.5% (relative to dihydrotestosterone) and demonstrated a range of agonist activity at 100 nM. In vivo pharmacologic activity was first assessed by using male rats. Structural modifications to the B-ring significantly affected the selectivity of the SARMs, demonstrating that single-atom substitutions can dramatically and unexpectedly influence activity in androgenic (i.e., prostate) and anabolic (i.e., muscle) tissues. (S)-N-(4-cyano-3-trifluoromethyl-phenyl)-3-(3-fluoro,4-chlorophenoxy)-2-hydroxy-2-methyl-propanamide (S-23) displayed full agonist activity in androgenic and anabolic tissues; however, the remaining SARMs were more prostate-sparing, selectively maintaining the size of the levator ani muscle in castrated rats. The partner-preference paradigm was used to evaluate the effects of SARMs on female sexual motivation. With the exception of two four-halo substituted analogs, the SARMs increased sexual motivation in ovariectomized rats, with potency and efficacy comparable with testosterone propionate. These results indicate that the AR is important in regulating female libido given the nonaromatizable nature of SARMs and it could be a superior alternative to steroidal testosterone preparations in the treatment of hypoactive sexual desire disorder.


Molecular Cancer Therapeutics | 2010

I-387, a Novel Antimitotic Indole, Displays a Potent In vitro and In vivo Antitumor Activity with Less Neurotoxicity

Sunjoo Ahn; Charles Duke; Christina M. Barrett; Dong Jin Hwang; Chien-Ming Li; Duane D. Miller; James T. Dalton

(3-(1H-indol-2-yl)phenyl)(3,4,5-trimethoxyphenyl)methanone (I-387) is a novel synthetic compound that inhibits tubulin action and exhibits potent antitumor activity in various preclinical models. I-387 inhibited the in vitro growth of several human cancer cell lines with IC50 values in the range of 15 to 39 nmol/L. Nanomolar concentrations of the compound induced apoptosis and caused phosphorylation of the antiapoptotic protein Bcl-2. I-387 induced a strong and concentration-dependent G2-M arrest in PC-3 cells by constitutive activation of Cdc2/cyclin B1 complex and destabilized polymerization of purified tubulin in vitro by binding to the colchicine-binding site. In vivo, I-387 treatment effectively inhibited tumor growth in mice bearing PC-3 tumor xenografts. In vitro studies of nerve growth factor–dependent neurite outgrowth in PC12 pheochromocytoma cells and in vivo studies of mouse behavior showed that I-387 was less neurotoxic than vinblastine and vincristine, tubulin destabilizers with known neurotoxicity. Interestingly, multidrug-resistant cell lines that overexpressed P-glycoprotein (P-gp), multidrug resistance–associated proteins, and breast cancer resistance protein were rendered resistant to docetaxel, vinblastine, SN-38, and doxorubicin, but not to I-387. I-387 dosed at 10 mg/kg was equally effective with 76% tumor growth inhibition in xenograft models using MES-SA uterine sarcoma cells and MES-SA/DX5 cells overexpressing P-gp. In contrast, docetaxel and vinblastine were not effective in MES-SA/DX5 xenograft models. The potent in vitro and in vivo antitumor activity of I-387 suggests that it may represent a new antimitotic agent for management of various malignancies, particularly for patients with drug-resistant cancer. Mol Cancer Ther; 9(11); 2859–68. ©2010 AACR.


Drug Metabolism and Disposition | 2011

Biotransformation of a novel antimitotic agent, I-387, by mouse, rat, dog, monkey, and human liver microsomes and in vivo pharmacokinetics in mice.

Sunjoo Ahn; Jeffrey D. Kearbey; Chien Ming Li; Charles Duke; Duane D. Miller; James T. Dalton

3-(1H-Indol-2-yl)phenyl)(3,4,5-trimethoxyphenyl)methanone (I-387) is a novel indole compound with antitubulin action and potent antitumor activity in various preclinical models. I-387 avoids drug resistance mediated by P-glycoprotein and showed less neurotoxicity than vinca alkaloids during in vivo studies. We examined the pharmacokinetics and metabolism of I-387 in mice as a component of our preclinical development of this compound and continued interest in structure-activity relationships for antitubulin agents. After a 1 mg/kg intravenous dose, noncompartmental pharmacokinetic analysis in plasma showed that clearance (CL), volume of distribution at steady state (Vdss), and terminal half-life (t1/2) of I-387 were 27 ml per min/kg, 5.3 l/kg, and 7 h, respectively. In the in vitro metabolic stability study, half-lives of I-387 were between 10 and 54 min by mouse, rat, dog, monkey, and human liver microsomes in the presence of NADPH, demonstrating interspecies variability. I-387 was most stable in rat liver microsomes and degraded quickly in monkey liver microsomes. Liquid chromatography-tandem mass spectrometry was used to identify phase I metabolites. Hydroxylation, reduction of a ketone group, and O-demethylation were the major metabolites formed by the liver microsomes of the five species. The carbonyl group of I-387 was reduced and identified as the most labile site in human liver microsomes. The results of these drug metabolism and pharmacokinetic studies provide the foundation for future structural modification of this pharmacophore to improve stability of drugs with potent anticancer effects in cancer patients.


Pharmaceutical Research | 2012

Combination Therapy of Antiandrogen and XIAP Inhibitor for Treating Advanced Prostate Cancer

Michael Danquah; Charles Duke; Renukadevi Patil; Duane D. Miller; Ram I. Mahato


Journal of Travel Medicine | 2016

Older age, chronic medical conditions and polypharmacy in Himalayan trekkers in Nepal: an epidemiologic survey and case series

Linda E. Keyes; Luke Mather; Charles Duke; Nirajan Regmi; Benoit Phelan; Sushil Pant; Jennifer Starling; Matthew McElwee; Devlin Cole; Theodore McConnell; Purshotam Paudel; T. Douglas Sallade; Alison Sheets; David Twillman; David S. Young; Buddha Basnyat


Wilderness & Environmental Medicine | 2016

Relationship of Blood Pressure and Hypertension to Acute Mountain Sickness

Jennifer Starling; Linda E. Keyes; Sushil Patel; Nirajam Regmi; Devlin Cole; Charles Duke; Luke Mather; Theodore McConnell; Matthew McElwee; Purshotam Paudel; Benoit Phelan; Douglas Sallade; Alison Sheets; David Twillmann; David T. Young; Buddha Basnyat


Wilderness & Environmental Medicine | 2016

Ambulatory Blood Pressure Monitoring Reveals Increased Sleeping Blood Pressure in Hypertensive Individuals at High Altitude

David S. Young; Linda E. Keyes; Luke Mather; Charles Duke; Nirajam Rejmi; Benoit Phelan; Sushil Pant; Jennifer Starling; Matthew McElwee; Devlin Cole; Theodore McConnell; Purshotam Paudel; Douglas Sallade; Allison L. Sheets; David R. Twillman; Buddha Basynat

Collaboration


Dive into the Charles Duke's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jennifer Starling

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Luke Mather

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Matthew McElwee

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Devlin Cole

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Linda E. Keyes

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge