Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cancers (Basel) ; 11(11)2019 Oct 24.
Article in English | MEDLINE | ID: mdl-31653008

ABSTRACT

These studies compared the efficacies of our clinical agent galeterone (Gal) and the FDA-approved prostate cancer drug, enzalutamide (ENZ) with two lead next generation galeterone analogs (NGGAs), VNPP414 and VNPP433-3ß, using prostate cancer (PC) in vitro and in vivo models. Antitumor activities of orally administered agents were also assessed in CWR22Rv1 tumor-bearing mice. We demonstrated that Gal and NGGAs degraded AR/AR-V7 and Mnk1/2; blocked cell cycle progression and proliferation of human PC cells; induced apoptosis; inhibited cell migration, invasion, and putative stem cell markers; and reversed the expression of epithelial-to-mesenchymal transition (EMT). In addition, Gal/NGGAs (alone or in combination) also inhibited the growth of ENZ-, docetaxel-, and mitoxantrone-resistant human PC cell lines. The NGGAs exhibited improved pharmacokinetic profiles over Gal in mice. Importantly, in vivo testing showed that VNPP433-3ß (at 7.53-fold lower equimolar dose than Gal) markedly suppressed (84% vs. Gal, 47%; p < 0.01) the growth of castration-resistant PC (CRPC) CWR22Rv1 xenograft tumors, with no apparent host toxicity. ENZ was ineffective in this CRPC xenograft model. In summary, our findings show that targeting AR/AR-V7 and Mnk1/2 for degradation represents an effective therapeutic strategy for PC/CRPC treatment and supports further development of VNPP433-3ß towards clinical investigation.

2.
Cancers (Basel) ; 11(3)2019 Mar 03.
Article in English | MEDLINE | ID: mdl-30832411

ABSTRACT

Currently, there are no effective therapies for patients with triple-negative breast cancer (TNBC), an aggressive and highly metastatic disease. Activation of eukaryotic initiation factor 4E (eIF4E) by mitogen-activated protein kinase (MAPK)-interacting kinases 1 and 2 (Mnk1/2) play a critical role in the development, progression and metastasis of TNBC. Herein, we undertook a comprehensive study to evaluate the activity of a first-in-class Mnk1/2 protein degraders, racemic VNLG-152R and its two enantiomers (VNLG-152E1 and VNLG-152E2) in in vitro and in vivo models of TNBC. These studies enabled us to identify racemic VNLG-152R as the most efficacious Mnk1/2 degrader, superior to its pure enantiomers. By targeting Mnk1/2 protein degradation (activity), VNLG-152R potently inhibited both Mnk-eIF4E and mTORC1 signaling pathways and strongly regulated downstream factors involved in cell cycle regulation, apoptosis, pro-inflammatory cytokines/chemokines secretion, epithelial-mesenchymal transition (EMT) and metastasis. Most importantly, orally bioavailable VNLG-152R exhibited remarkable antitumor (91 to 100% growth inhibition) and antimetastatic (~80% inhibition) activities against cell line and patient-derived TNBC xenograft models, with no apparent host toxicity. Collectively, these studies demonstrate that targeting Mnk-eIF4E/mTORC1 signaling with a potent Mnk1/2 degrader, VNLG-152R, is a novel therapeutic strategy that can be developed as monotherapy for the effective treatment of patients with primary/metastatic TNBC.

3.
J Nutr ; 144(5): 734-42, 2014 May.
Article in English | MEDLINE | ID: mdl-24717362

ABSTRACT

We identified normal vs. abnormal 25-hydroxyvitamin D [25(OH)D] concentrations by examining the relation of 25(OH)D to non-bone-related measures (plasma glucose, insulin resistance, lipids, blood pressure, fitness, obesity, and regional adiposity) and asking whether there is a 25(OH)D concentration above and below which the relation between 25(OH)D and outcome changes. We examined the relation between 25(OH)D and outcome by race to see whether race-specific normal ranges are needed, and we examined the role of insulin-like growth factor-1 (IGF-1) in modulating the relation between 25(OH)D and outcome. In a cross-sectional study of 239 overweight and obese, sedentary postmenopausal women without diabetes (83 black, 156 white), outcome measures included plasma lipids, glucose, insulin, homeostasis model assessment of insulin resistance (HOMA-IR), IGF-1, parathyroid hormone (PTH), aerobic fitness, body composition, subcutaneous abdominal and visceral fat, and blood pressure. We identified threshold effects in the association between 25(OH)D and these variables using piecewise linear regressions. We found that 25(OH)D was inversely related to fasting glucose, fasting and 2-h insulin, HOMA-IR, visceral abdominal fat, percentage fat, PTH, and triglycerides. Evidence for a threshold effect of 25(OH)D was found for 2-h glucose, 2-h insulin, fasting insulin, and HOMA-IR. There was no evidence suggesting the need for race-specific normal 25(OH)D concentrations. IGF-1 modulated the relation between 25(OH)D and outcome but only below, and not above, a threshold 25(OH)D concentration. Our findings suggest a threshold effect of 25(OH)D on glucose-insulin metabolism such that 25(OH)D ≥ ∼26 µg/L (65.0 pmol/L) supports normal glucose homeostasis and that the same cut point defining normal 25(OH)D concentration can be used in black and white women. This study was registered at clinicaltrials.gov as NCT01798030.


Subject(s)
Black People/statistics & numerical data , Glucose Intolerance/drug therapy , Glucose Intolerance/ethnology , Vitamin D/analogs & derivatives , White People/statistics & numerical data , Adult , Aged , Body Composition/drug effects , Body Composition/physiology , Cross-Sectional Studies , Female , Homeostasis/drug effects , Homeostasis/physiology , Humans , Hyperlipidemias/drug therapy , Hyperlipidemias/ethnology , Insulin Resistance/physiology , Insulin-Like Growth Factor I/metabolism , Linear Models , Middle Aged , Obesity/drug therapy , Obesity/ethnology , Physical Fitness/physiology , Postmenopause/drug effects , Postmenopause/physiology , Risk Factors , Vitamin D/administration & dosage , Vitamins/administration & dosage
4.
J Med Chem ; 56(12): 4880-98, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23713567

ABSTRACT

As part of our program to explore the influence of small structural modifications of our drug candidate 3ß-(hydroxy)-17-(1H-benzimidazol-1-yl)androsta-5,16-diene (galeterone, 5) on the modulation of the androgen receptor (AR), we have prepared and evaluated a series of novel C-3, C-16, and C-17 analogues. Using structure activity analysis, we established that the benzimidazole moiety at C-17 is essential and optimal and also that hydrophilic and heteroaromatic groups at C-3 enhance both antiproliferative (AP) and AR degrading (ARD) activities. The most potent antiproliferative compounds were 3ß-(1H-imidazole-1-carboxylate)-17-(1H-benzimidazol-1-yl)androsta-5,16-diene (47), 3-((EZ)-hydroximino)-17-(1H-benzimidazol-1-yl)androsta-4,16-diene (36), and 3ß-(pyridine-4-carboxylate)-17-(1H-benzimidazol-1-yl)androsta-5,16-diene (43), with GI50 values of 0.87, 1.91, and 2.57 µM, respectively. Compared to 5, compound 47 was 4- and 8-fold more potent with respect to AP and ARD activities, respectively. Importantly, we also discovered that our compounds, including 5, 36, 43, and 47, could degrade both full-length and truncated ARs in CWR22rv1 human prostate cancer cells. With these activities, they have potential for development as new drugs for the treatment of all forms of prostate cancer.


Subject(s)
Androstadienes/chemistry , Androstadienes/pharmacology , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Down-Regulation/drug effects , Drug Design , Molecular Targeted Therapy , Prostatic Neoplasms/drug therapy , Receptors, Androgen/metabolism , Androstadienes/therapeutic use , Benzimidazoles/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Prostatic Neoplasms/pathology , Proteolysis/drug effects , Receptors, Androgen/genetics , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Transcriptional Activation/drug effects
5.
J Health Care Poor Underserved ; 24(1 Suppl): 36-46, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23395943

ABSTRACT

There is growing interest in the diverse signaling pathways that regulate and affect breast tumorigenesis, including the role of phytochemicals and the emerging role of microRNAs (miRNAs). Recent studies demonstrate that miRNAs regulate fundamental cellular and developmental processes at the transcriptional and translational level under normal and disease conditions. While there is growing evidence to support the role of phytoalexin-mediated miRNA regulation of cancer, few reports address this role in breast cancer. Recent reports by our group and others demonstrate that natural products, including stilbenes, curcumin, and glyceollins, could alter the expression of specific miRNAs, which may lead to increased sensitivity of cancer cells to conventional anti-cancer agents and, therefore, hormone-dependent and hormone-independent tumor growth inhibition. This review will discuss how dietary intake of natural products, by regulating specific miRNAs, contribute to the prevention and treatment of breast cancer.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/drug effects , Sesquiterpenes/pharmacology , Female , Humans , Phytoalexins
6.
Steroids ; 76(12): 1268-79, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21729712

ABSTRACT

In a continuing study of our clinical candidate 5 VN/124-1 (TOK-001) and analogs as potential agents for prostate cancer therapy, putative metabolites (10, 15 and 18) of compound 5 were rationally designed and synthesized. However, none of these agents were as efficacious as 5 in several in vitro studies. Using western blot analysis, we have generated a preliminary structure-activity relationship (SAR) of 5 and related analogs as androgen receptor ablative agents (ARAAs). In vivo using the androgen-dependent LAPC-4 prostate cancer xenograft model, we demonstrated for the first time that 5 is more efficacious than the 17-lyase inhibitor 3 (abiraterone)/4 (abiraterone acetate) that is currently in phase III clinical trials. In our desire to optimize the potency of 5, compounds 6 (3ξ-fluoro-) and 9 (3ß-sulfamate-) designed to increase the stability and oral bioavailability of 5, respectively were evaluated in vivo. We showed, that on equimolar basis, compound 6 was ∼2-fold more efficacious versus LAPC-4 xenografts than 5, but the toxicity observed with 6 is of concern. These studies further demonstrate the efficacy of 5 in a clinically relevant prostate cancer model and justify its current clinical development as a potential treatment of prostate cancer.


Subject(s)
Androstadienes/chemistry , Androstadienes/therapeutic use , Androstenols/chemistry , Androstenols/therapeutic use , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Benzimidazoles/chemistry , Benzimidazoles/therapeutic use , Prostatic Neoplasms/drug therapy , Androstenes , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Clinical Trials, Phase III as Topic , Drug Combinations , Estradiol/analogs & derivatives , Estradiol/pharmacology , Humans , Male , Mice , Mice, SCID , Norethindrone/pharmacology , Receptors, Androgen/metabolism , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Structure-Activity Relationship , Testosterone/analogs & derivatives , Testosterone/pharmacology , Xenograft Model Antitumor Assays
7.
J Steroid Biochem Mol Biol ; 125(1-2): 23-31, 2011 May.
Article in English | MEDLINE | ID: mdl-21092758

ABSTRACT

Prostate cancer (PC) is now the second most prevalent cause of death in men in the USA and Europe. At present, the major treatment options include surgical or medical castration. These strategies cause ablation of the production of testosterone (T), dihydrotestosterone (DHT) and related androgens by the testes. However, because these procedures do not affect adrenal, prostate and other tissues' androgen production, they are often combined with androgen receptor antagonists to block their action. Indeed, recent studies have unequivocally established that in castration-resistant prostate cancer (CRPC) many androgen-regulated genes become re-expressed and tissue androgen levels increase despite low serum levels. Clearly, inhibition of the key enzyme which catalyzes the biosynthesis of androgens from pregnane precursors, 17α-hydroxy/17,20-lyase (hereafter referred to as CYP17) could prevent androgen production from all sources. Thus, total ablation of androgen production by potent CYP17 inhibitors may provide effective treatment of prostate cancer patients. This review highlights the role of androgen biosynthesis in the progression of prostate cancer and the impact of CYP17 inhibitors, such as ketoconazole, abiraterone acetate, VN/124-1 (TOK-001) and TAK-700 in the clinic and in clinical development. Article from the special issue on Targeted Inhibitors.


Subject(s)
Androgens/biosynthesis , Enzyme Inhibitors/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Steroid 17-alpha-Hydroxylase/metabolism , Androgens/chemistry , Clinical Trials as Topic , Disease Progression , Humans , Male , Molecular Structure , Receptors, Androgen/metabolism , Steroid 17-alpha-Hydroxylase/chemistry , Structure-Activity Relationship
8.
Future Med Chem ; 2(4): 667-80, 2010 Apr.
Article in English | MEDLINE | ID: mdl-21426013

ABSTRACT

The beneficial effect of androgen ablation has been well established in prostate cancer therapy. Despite the initial response, patients typically relapse with a more aggressive form described as castration-resistant prostate cancer (CRCP), driven by continued androgen receptor (AR) signaling. This review details the current state of anti-androgen therapy, mainly for CRPC, with major emphasis on the most potent and promising compounds under development. Anti-androgen failure has been linked to elevated AR expression, increased expression of coactivator proteins, AR mutations, ligand-independent AR activation and persistent intraprostatic androgens. MDV3100, BMS-641988 and VN/124-1 were developed to overcome these mechanisms. In CRCP, prostate cancer cells still rely on intracellular androgens and, to a greater extent, on active AR for growth and survival. Therefore, potent anti-androgens that efficiently disrupt the functions (signaling) of AR are envisioned to be effective drugs for all types of prostate cancers.


Subject(s)
Androgen Antagonists/chemistry , Androgen Antagonists/therapeutic use , Prostatic Neoplasms/drug therapy , Androgen Antagonists/chemical synthesis , Humans , Male , Molecular Structure , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction/physiology
9.
Mol Cancer Ther ; 7(8): 2348-57, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18723482

ABSTRACT

We previously reported that our novel compound 3beta-hydroxy-17-(1H-benzimidazole-1-yl)androsta-5,16-diene (VN/124-1) is a potent 17alpha-hydroxylase/17,20-lyase (CYP17) inhibitor/antiandrogen and strongly inhibits the formation and proliferation of human prostate cancer LAPC4 tumor xenografts in severe combined immunodeficient mice. In this study, we report that VN/124-1 and other novel CYP17 inhibitors also cause down-regulation of androgen receptor (AR) protein expression in vitro and in vivo. This mechanism of action seems to contribute to their antitumor efficacy. We compared the in vivo antitumor efficacy of VN/124-1 with that of castration and a clinically used antiandrogen, Casodex, and show that VN/124-1 is more potent than castration in the LAPC4 xenograft model. Treatment with VN/124-1 (0.13 mmol/kg twice daily) was also very effective in preventing the formation of LAPC4 tumors (6.94 versus 2410.28 mm(3) in control group). VN/124-1 (0.13 mmol/kg twice daily) and VN/124-1 (0.13 mmol/kg twice daily) + castration induced regression of LAPC4 tumor xenografts by 26.55% and 60.67%, respectively. Treatments with Casodex (0.13 mmol/kg twice daily) or castration caused significant tumor suppression compared with control. Furthermore, treatment with VN/124-1 caused marked down-regulation of AR protein expression, in contrast to treatments with Casodex or castration that caused significant up-regulation of AR protein expression. The results suggest that VN/124-1 acts by several mechanisms (CYP17 inhibition, competitive inhibition, and down-regulation of the AR). These actions contribute to inhibition of the formation of LAPC4 tumors and cause regression of growth of established tumors. VN/124-1 is more efficacious than castration in the LAPC4 xenograft model, suggesting that the compound has potential for the treatment of prostate cancer.


Subject(s)
Androgen Receptor Antagonists , Androstadienes/therapeutic use , Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Enzyme Inhibitors/therapeutic use , Prostatic Neoplasms/drug therapy , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Androstadienes/pharmacology , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Binding, Competitive , Cell Line, Tumor , Down-Regulation , Enzyme Inhibitors/pharmacology , Humans , Male , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism
10.
Steroids ; 73(12): 1217-27, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18582482

ABSTRACT

We have exploited the reaction of 1,1'-carbonylbis(2-methylimidazole) (CBMI) with several 17beta-hydroxy androstanes to synthesize a series of novel C17 steroidal carbamates. Structural elucidation features have been provided for the final compounds based on 1D and 2D NMR techniques, IR spectroscopy, and related literature. The new compounds were tested for inhibition of human cytochrome 17alpha-hydroxylase-C17,20-lyase (CYP17) and androgen receptor (AR) binding and function effects. Their inhibitory potential against PC-3 cell proliferation was also evaluated. Compounds 11 and 23 were found to inhibit CYP17 with IC50 values of 17.1 and 11.5 microM, respectively. The carbamate moiety at C17 allowed tight binding of the synthesized compounds to both wild-type (wt-) and mutated AR. When bound to the mutated AR, the compounds were found to have a dual effect, stimulating transcription at low concentrations while almost fully blocking it at the higher concentrations tested, in the presence of the natural androgen dihydrotestosterone (DHT). Compounds 8 and 12 were the most active against PC-3 cell proliferation with EC50 values of 2.2 and 0.2 microM, respectively.


Subject(s)
Carbamates/chemical synthesis , Cell Division/physiology , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Steroid 17-alpha-Hydroxylase/metabolism , Steroids/chemical synthesis , Cell Line, Tumor , Humans , Magnetic Resonance Spectroscopy , Male , Prostatic Neoplasms/enzymology , Receptors, Androgen/physiology , Spectrometry, Mass, Electrospray Ionization
11.
Bioorg Med Chem ; 16(7): 3519-29, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18316193

ABSTRACT

The search for novel androgen receptor (AR) down-regulating agents by catalyst HipHop pharmacophore modeling led to the discovery of some lead molecules. Unexpectedly, the effect of these leads on human prostate cancer LNCaP cell viability did not correlate with the ability of the compounds to cause down-regulation of AR protein expression. Through rational synthetic optimization of the lead compound (BTB01434), we have discovered a series of novel substituted diaryl molecules as potent anti-prostate cancer agents. Some compounds (1-6) were shown to be extremely potent inhibitors of LNCaP cell viability with GI(50) values in the nanomolar range (1.45-83 nM). The most potent compound (4-methylphenyl)[(4-methylphenyl)sulfonyl]amine (5) with a GI(50) value of 1.45 nM is 27,000 times more potent than our lead compound BTB01434 (GI(50)=39.8 microM). In addition, some of the compounds exhibited modest anti-androgenic activities and one was also a potent inhibitor (GI(50)=850 nM) of PC-3 (AR-null) cell growth. A clear structure-activity relationship (SAR) has been established for activity against LNCaP cells, where potent molecules possess two substituted/unsubstituted aromatic rings connected through a sulfonamide linker. These novel compounds are strong candidates for development for the treatment of hormone-sensitive and importantly hormone-refractory prostate cancers in humans.


Subject(s)
Androgen Receptor Antagonists , Antineoplastic Agents, Hormonal/chemical synthesis , Antineoplastic Agents, Hormonal/pharmacology , Down-Regulation , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Amines/chemical synthesis , Amines/chemistry , Androgens/metabolism , Antineoplastic Agents, Hormonal/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Humans , Imines/chemical synthesis , Imines/chemistry , Male , Models, Molecular , Molecular Structure , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Structure-Activity Relationship , Transcription, Genetic/genetics
12.
Steroids ; 72(14): 939-48, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17884122

ABSTRACT

A series of novel 1H- and 2H-indazole derivatives of the commercially available dehydroepiandrosterone acetate have been synthesized and tested for inhibition of human cytochrome 17alpha-hydroxylase-C(17,20)-lyase (CYP17), androgen receptor (AR) binding affinity, and cytotoxic potential against three prostate cancer (PC) cell lines.


Subject(s)
Dehydroepiandrosterone/analogs & derivatives , Dehydroepiandrosterone/chemical synthesis , Dehydroepiandrosterone/pharmacology , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Binding, Competitive/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dehydroepiandrosterone/chemistry , Dehydroepiandrosterone/toxicity , Humans , Male , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Steroid 17-alpha-Hydroxylase/metabolism
13.
Bioorg Med Chem ; 15(10): 3413-21, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17383188

ABSTRACT

A qualitative 3D pharmacophore model (a common feature based model or Catalyst HipHop algorithm) was developed for well-known natural product androgen receptor down-regulating agents (ARDAs). The four common chemical features identified included: one hydrophobic group, one ring aromatic group, and two hydrogen bond acceptors. This model served as a template in virtual screening of the Maybridge and NCI databases that resulted in identification of six new ARDAs (EC(50) values 17.5-212 microM). Five of these molecules strongly inhibited the growth of human prostate LNCaP cells. These novel compounds may be used as leads to develop other novel anti-prostate cancer agents.


Subject(s)
Androgen Antagonists/chemical synthesis , Androgen Antagonists/pharmacology , Androgen Receptor Antagonists , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Prostatic Neoplasms/drug therapy , Blotting, Western , Catalysis , Cell Line, Tumor , Computational Biology , Databases, Factual , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Humans , Male , Models, Molecular , Neoplasm Proteins/chemical synthesis , Neoplasm Proteins/pharmacology , Prostatic Neoplasms/pathology , Structure-Activity Relationship , Tetrazolium Salts , Thiazoles
14.
Bioorg Med Chem ; 14(13): 4323-40, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16530416

ABSTRACT

The naturally occurring retinoids and their synthetic analogs play a key role in differentiation, proliferation, and apoptosis, and their use/potential in oncology, dermatology and a variety of diseases are well documented. This review focuses on the role of all-trans-retinoic acid (ATRA), the principal endogenous metabolite of vitamin A (retinol) and its metabolism in oncology and dermatology. ATRA has been used successfully in differentiated therapy of acute promyelocytic leukemia, skin cancer, Kaposi's sarcoma, and cutaneous T-cell lymphoma, and also in the treatment of acne and psoriasis. However, its usefulness is limited by the rapid emergence of acquired ATRA resistance involving multifactoral mechanisms. A key mechanism of resistance involves ATRA-induced catabolism of ATRA. Thus, a novel strategy to overcome the limitation associated with exogenous ATRA therapy has been to modulate and/or increase the levels of endogenous ATRA by inhibiting the cytochrome P450-dependent ATRA-4-hydroxylase enzymes (particularly CYP26s) responsible for ATRA metabolism. These inhibitors are also referred to as retinoic acid metabolism blocking agents (RAMBAs). This review highlights development in the design, synthesis, and evaluation of RAMBAs. Major emphasis is given to liarozole, the most studied and only RAMBA in clinical use and also the new RAMBAs in development and with clinical potential.


Subject(s)
Antineoplastic Agents/chemistry , Cytochrome P-450 Enzyme Inhibitors , Dermatologic Agents/chemistry , Neoplasms/drug therapy , Skin Diseases/drug therapy , Tretinoin/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cytochrome P-450 Enzyme System/metabolism , Dermatologic Agents/therapeutic use , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Humans , Neoplasms/enzymology , Retinoic Acid 4-Hydroxylase , Skin Diseases/enzymology , Tretinoin/therapeutic use
15.
J Med Chem ; 48(8): 2972-84, 2005 Apr 21.
Article in English | MEDLINE | ID: mdl-15828836

ABSTRACT

New chemical entities, steroidal C-17 benzoazoles (5, 6, 9 and 10) and pyrazines (14 and 15) were rationally designed and synthesized. The key reaction for synthesis of the benzoazoles involved the nucleophilic vinylic "addition-elimination" substitution reaction of 3beta-acetoxy-17-chloro-16-formylandrosta-5,16-diene (2) and benzoazole nucleophiles, while that for synthesis of pyrazines involved palladium-catalyzed cross-coupling reaction of 17-iodoandrosta-5,16-dien-3beta-ol (13) with tributylstannyl diazines. Some of the compounds were shown to be potent inhibitors of human CYP17 enzyme as well as potent antagonist of both wild type and mutant androgen receptors (AR). The most potent CYP17 inhibitors were 3beta-hydroxy-17-(1H-benzimidazole-1-yl)androsta-5,16-diene (5, code named VN/124-1), 3beta-hydroxy-17-(5(1)-pyrimidyl)androsta-5,16-diene (15) and 17-(1H-benzimidazole-1-yl)androsta-4,16-dien-3-one (6), with IC(50) values of 300, 500 and 915 nM, respectively. Compounds 5, 6, 14 and 15 were effective at preventing binding of (3)H-R1881 (methyltrienolone, a stable synthetic androgen) to both the mutant LNCaP AR and the wild-type AR, but with a 2.2- to 5-fold higher binding efficiency to the latter. Compounds 5 and 6 were also shown to be potent pure AR antagonists. The cell growth studies showed that 5 and 6 inhibit the growth of DHT-stimulated LNCaP and LAPC4 prostate cancer cells with IC(50) values in the low micromolar range (i.e., <10 microM). Their inhibitory potencies were comparable to that of casodex but remarkably superior to that of flutamide. The pharmacokinetics of compounds 5 and 6 in mice were investigated. Following s.c. administration of 50 mg/kg of 5 and 6, peak plasma levels of 16.82 and 5.15 ng/mL, respectively, occurred after 30 to 60 min, both compounds were cleared rapidly from plasma (terminal half-lives of 44.17 and 39.93 min, respectively), and neither was detectable at 8 h. Remarkably, compound 5 was rapidly converted into a metabolite tentatively identified as 17-(1H-benzimidazol-1-yl)androsta-3-one. When tested in vivo, 5 proved to be very effective at inhibiting the growth of androgen-dependent LAPC4 human prostate tumor xenograft, while 6 was ineffective. Compound 5 (50 mg/kg/twice daily) resulted in a 93.8% reduction (P = 0.00065) in the mean final tumor volume compared with controls, and it was also significantly more effective than castration. To our knowledge, this is the first example of an antihormonal agent (an inhibitor of androgen synthesis (CYP17 inhibitor)/antiandrogen) that is significantly more effective than castration in suppression of androgen-dependent prostate tumor growth. In view of these impressive anticancer properties, compound 5 is a strong candidate for development for the treatment of human prostate cancer.


Subject(s)
Androgen Antagonists/chemical synthesis , Androstadienes/chemical synthesis , Antineoplastic Agents/chemical synthesis , Azoles/chemical synthesis , Benzimidazoles/chemical synthesis , Prostatic Neoplasms/drug therapy , Pyrazines/chemical synthesis , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , 5-alpha Reductase Inhibitors , Androgen Antagonists/chemistry , Androgen Antagonists/pharmacology , Androstadienes/pharmacokinetics , Androstadienes/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Azoles/pharmacokinetics , Azoles/pharmacology , Benzimidazoles/pharmacokinetics , Benzimidazoles/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Isoenzymes/antagonists & inhibitors , Male , Mice , Mice, SCID , Mutation , Pyrazines/pharmacokinetics , Pyrazines/pharmacology , Radioligand Assay , Receptors, Androgen/drug effects , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Structure-Activity Relationship , Tissue Distribution , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Xenograft Model Antitumor Assays
16.
J Biol Chem ; 279(17): 17319-28, 2004 Apr 23.
Article in English | MEDLINE | ID: mdl-14966121

ABSTRACT

Androgen receptor (AR) signaling pathways mediate critical events in normal and neoplastic prostate growth. Shortening of the polymorphic N-terminal polyglutamine (poly(Q)) tract of the AR gene leads to transcriptional hyperactivity and has been correlated with an increased risk of prostate cancer. The underlying mechanisms for these effects are poorly understood. We show here that androgen-dependent cellular proliferation and transcription in prostate cancer cells is inversely correlated to the length of the AR poly(Q) region. We further show that AR proteins containing a shortened poly(Q) region functionally respond to lower concentrations of androgens than wild type AR. Whereas DNA binding activity is relatively unaffected by AR poly(Q) variation, we found that ligand binding affinity and the ligand-induced NH(2)- to COOH-terminal intramolecular interaction is enhanced when the poly(Q) region is shortened. Importantly, we show that AR proteins containing a shortened poly(Q) region associate in vivo with higher levels of specific p160 coactivators and components of the SWI/SNF chromatin remodeling complex as compared with the wild type AR. Collectively, our findings suggest that the AR transcriptional hyperactivity associated with shortened poly(Q) length stems from altered ligand-induced conformational changes that enhance coactivator recruitment.


Subject(s)
Peptides/chemistry , Prostatic Neoplasms/metabolism , Receptors, Androgen/chemistry , Transcription, Genetic , Blotting, Western , Cell Division , Cell Line , Cell Line, Tumor , DNA/chemistry , Humans , Immunoblotting , Ligands , Male , Models, Genetic , Plasmids/metabolism , Precipitin Tests , Protein Conformation , Protein Structure, Tertiary , Signal Transduction , Time Factors , Transcriptional Activation , Transfection , Two-Hybrid System Techniques
17.
J Med Chem ; 46(12): 2345-51, 2003 Jun 05.
Article in English | MEDLINE | ID: mdl-12773039

ABSTRACT

We report here a molecular modeling investigation of steroidal and nonsteroidal inhibitors of human cytochrome P450 17alpha-hydroxylase-17,20-lyase (CYP17). Using the pharmacophore perception technique, we have generated common-feature pharmacophore model(s) to explain the putative binding requirements for two classes of human CYP17 inhibitors. Common chemical features in the steroid and nonsteroid human CYP17 enzyme inhibitors, as deduced by the Catalyst/HipHop program, are one to two hydrogen bond acceptors (HBAs) and three hydrophobic groups. For azole-steroidal ligands, the 3beta-OH group of ring A and the N-3 of the azole ring attached to ring D at C-17 act as hydrogen bond acceptors. A model that permits hydrogen bond interaction between the azole functionality on ring D and the enzyme is consistent with experimental deductions for type II CYP17 inhibitors where a sixth ligating atom interacts with Fe(II) of heme. In general, pharmacophore models derived for steroid and nonsteroidal compounds bear striking similarities to all azole sites mapping the HBA functionality and to three hydrophobic features describing the hydrophobic interactions between the ligands and the enzyme. Using the pharmacophore model derived for azole-steroidal inhibitors as a 3D search query against several 3D multiconformational Catalyst formatted databases, we identified several steroidal compounds with potential inhibition of this enzyme. Biological testing of some of these compounds show low to high inhibitory potency against the human CYP17 enzyme. This shows the potential of our pharmacophore model in identifying new and potent CYP17 inhibitors. Further refinement of the model is in progress with a view to identifying and optimizing new leads.


Subject(s)
Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Steroids/chemistry , Algorithms , Azoles/chemistry , Binding Sites , Humans , Hydrogen Bonding , Male , Models, Molecular , Prostatic Neoplasms/drug therapy , Protein Binding , Quantitative Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...