Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Sci Rep ; 12(1): 1862, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35115586

ABSTRACT

In recurrent epithelial ovarian cancer (EOC) most patients develop platinum-resistance. On molecular level the NRF2 pathway, a cellular defense mechanism against reactive oxygen species, is induced. In this study, we investigate AKR1C1/2, target of NRF2, in a well-established EOC collective by immunohistochemistry and in a panel of ovarian cancer cell lines including platinum-resistant clones. The therapeutic effect of carboplatin and MPA as monotherapy or in combination was assessed by functional assays, using OV90 and OV90cp cells. Molecular mechanisms of action of MPA were investigated by NRF2 silencing and AKR activity measurements. Immunohistochemical analysis revealed that AKR1C1/2 is a key player in the development of chemoresistance and an independent indicator for short PFS (23.5 vs. 49.6 months, p = 0.013). Inhibition of AKR1C1/2 by MPA led to a concentration- and time-dependent decline of OV90 viability and to an increased response to CP in vitro. By NRF2 silencing, however, the effects of MPA treatment were reduced. Concludingly, our data suggest that a combination therapy of carboplatin and MPA might be a promising therapeutic approach to increase response rates of EOC patients, which should be explored in clinical context.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carboplatin/pharmacology , Carcinoma, Ovarian Epithelial/drug therapy , Enzyme Inhibitors/pharmacology , Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Medroxyprogesterone Acetate/pharmacology , Ovarian Neoplasms/drug therapy , 20-Hydroxysteroid Dehydrogenases/metabolism , Carcinoma, Ovarian Epithelial/enzymology , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm , Female , Humans , Hydroxysteroid Dehydrogenases/metabolism , Middle Aged , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology
2.
Chem Biol Interact ; 351: 109746, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34780792

ABSTRACT

Human aldo-keto reductase family 1C1 (AKR1C1) is an important enzyme involved in human hormone metabolism, which is mainly responsible for the metabolism of progesterone in the human body. AKR1C1 is highly expressed and has an important relationship with the occurrence and development of various diseases, especially some cancers related to hormone metabolism. Nowadays, many inhibitors against AKR1C1 have been discovered, including some synthetic compounds and natural products, which have certain inhibitory activity against AKR1C1 at the target level. Here we briefly reviewed the physiological and pathological functions of AKR1C1 and the relationship with the disease, and then summarized the development of AKR1C1 inhibitors, elucidated the interaction between inhibitors and AKR1C1 through molecular docking results and existing co-crystal structures. Finally, we discussed the design ideals of selective AKR1C1 inhibitors from the perspective of AKR1C1 structure, discussed the prospects of AKR1C1 in the treatment of human diseases in terms of biomarkers, pre-receptor regulation and single nucleotide polymorphisms, aiming to provide new ideas for drug research targeting AKR1C1.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 20-Hydroxysteroid Dehydrogenases/physiology , Enzyme Inhibitors/pharmacology , 20-Hydroxysteroid Dehydrogenases/chemistry , 20-Hydroxysteroid Dehydrogenases/metabolism , Animals , Catalytic Domain , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Molecular Docking Simulation , Protein Binding
3.
J Enzyme Inhib Med Chem ; 36(1): 1500-1508, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34227437

ABSTRACT

Enzymes AKR1C regulate the action of oestrogens, androgens, and progesterone at the pre-receptor level and are also associated with chemo-resistance. The activities of these oestrone halides were investigated on recombinant AKR1C enzymes. The oestrone halides with halogen atoms at both C-2 and C-4 positions (13ß-, 13α-methyl-17-keto halogen derivatives) were the most potent inhibitors of AKR1C1. The lowest IC50 values were for the 13α-epimers 2_2I,4Br and 2_2I,4Cl (IC50, 0.7 µM, 0.8 µM, respectively), both of which selectively inhibited the AKR1C1 isoform. The 13α-methyl-17-keto halogen derivatives 2_2Br and 2_4Cl were the most potent inhibitors of AKR1C2 (IC50, 1.5 µM, 1.8 µM, respectively), with high selectivity for the AKR1C2 isoform. Compound 1_2Cl,4Cl showed the best AKR1C3 inhibition, and it also inhibited AKR1C1 (Ki: AKR1C1, 0.69 µM; AKR1C3, 1.43 µM). These data show that halogenated derivatives of oestrone represent a new class of potent and selective AKR1C inhibitors as lead compounds for further optimisations.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Estrone/pharmacology , 20-Hydroxysteroid Dehydrogenases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Estrone/analogs & derivatives , Estrone/chemistry , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship
4.
Mol Cell Biochem ; 476(8): 2975-2987, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33770316

ABSTRACT

Aldo-keto reductase 1C1 (AKR1C1) is a hydroxysteroid dehydrogenase, known to inactivate the biologically active progesterone into its corresponding 20 α-hydroxyprogesterone. Increased expression of the AKR1C1 gene in oncogenesis is linked with resistance to various anticancer agents and hence it is considered as an emerging drug target for the design and developing the novel anticancer drugs. We have performed QSAR pharmacophore modeling for AKR1C1 inhibitors followed by a virtual screening of ~ 59,000 compounds present at the Maybridge database. The screened compounds were refined using drug-like filters of Lipinski rule, ADMET plot, molecular docking and scoring and subsequently top 20 hits were selected. Selected compounds were subjected to the in vitro for AKR1C1 inhibition assay and best seven compounds bearing excellent binding affinity to the AKR1C1 were finally selected. The identified compounds may be exploited in hit-to-lead development and may also prove as an interventional strategy in preventing a pre-term birth due to declining levels of progesterone.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Breast Neoplasms/drug therapy , Drug Design , Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Breast Neoplasms/pathology , Cell Proliferation , Female , Humans , Models, Molecular , Molecular Docking Simulation , Structure-Activity Relationship , Tumor Cells, Cultured
5.
Br J Cancer ; 118(7): 985-994, 2018 04.
Article in English | MEDLINE | ID: mdl-29515258

ABSTRACT

BACKGROUND: Despite chemotherapy intensification, a subgroup of high-risk paediatric T-cell acute lymphoblastic leukemia (T-ALL) patients still experience treatment failure. In this context, we hypothesised that therapy resistance in T-ALL might involve aldo-keto reductase 1C (AKR1C) enzymes as previously reported for solid tumors. METHODS: Expression of NRF2-AKR1C signaling components has been analysed in paediatric T-ALL samples endowed with different treatment outcomes as well as in patient-derived xenografts of T-ALL. The effects of AKR1C enzyme modulation has been investigated in T-ALL cell lines and primary cultures by combining AKR1C inhibition, overexpression, and gene silencing approaches. RESULTS: We show that T-ALL cells overexpress AKR1C1-3 enzymes in therapy-resistant patients. We report that AKR1C1-3 enzymes play a role in the response to vincristine (VCR) treatment, also ex vivo in patient-derived xenografts. Moreover, we demonstrate that the modulation of AKR1C1-3 levels is sufficient to sensitise T-ALL cells to VCR. Finally, we show that T-ALL chemotherapeutics induce overactivation of AKR1C enzymes independent of therapy resistance, thus establishing a potential resistance loop during T-ALL combination treatment. CONCLUSIONS: Here, we demonstrate that expression and activity of AKR1C enzymes correlate with response to chemotherapeutics in T-ALL, posing AKR1C1-3 as potential targets for combination treatments during T-ALL therapy.


Subject(s)
Aldo-Keto Reductases/physiology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , 20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 20-Hydroxysteroid Dehydrogenases/physiology , Age of Onset , Aldo-Keto Reductase Family 1 Member C3/antagonists & inhibitors , Aldo-Keto Reductase Family 1 Member C3/physiology , Aldo-Keto Reductases/antagonists & inhibitors , Animals , Child , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Leukemic/drug effects , Humans , Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Hydroxysteroid Dehydrogenases/physiology , Isoenzymes/physiology , Medroxyprogesterone Acetate/administration & dosage , Mice , Mice, Inbred NOD , Mice, SCID , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/physiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/epidemiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Cells, Cultured , Vincristine/administration & dosage , Xenograft Model Antitumor Assays
6.
Oncol Rep ; 37(4): 2025-2032, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28259989

ABSTRACT

Resistance to anticancer medications often leads to poor outcomes. The present study explored an effective approach for enhancing chemotherapy targeted against human cancer cells. Real-time quantitative real-time polymerase chain reaction (qRT-PCR) analysis revealed overexpression of members of aldo-keto reductase (AKR) 1C family, AKR1C1, AKR1C2, AKR1C3, and AKR1C4, in cisplatin, cis-diamminedichloroplatinum (II) (CDDP)-resistant human cancer cell lines, HeLa (cervical cancer cells) and Sa3 (oral squamous cell carcinoma cells). The genes were downregulated using small-interfering RNA (siRNA) transfection, and the sensitivity to CDDP or 5-fluorouracil (5-FU) was investigated. When the genes were knocked down, sensitivity to CDDP and 5-FU was restored. Furthermore, we found that administration of mefenamic acid, a widely used non-steroidal anti-inflammatory drug (NSAID) and a known inhibitor of AKR1Cs, enhanced sensitivity to CDDP and 5-FU. The present study suggests that AKR1C family is closely associated with drug resistance to CDDP and 5-FU, and mefenamic acid enhances their sensitivity through its inhibitory activity in drug-resistant human cancer cells. Thus, the use of mefenamic acid to control biological function of AKR1C may lead to effective clinical outcomes by overcoming anticancer drug resistance.


Subject(s)
20-Hydroxysteroid Dehydrogenases/biosynthesis , 3-Hydroxysteroid Dehydrogenases/biosynthesis , Hydroxyprostaglandin Dehydrogenases/biosynthesis , Hydroxysteroid Dehydrogenases/biosynthesis , Mefenamic Acid/administration & dosage , Neoplasms/drug therapy , 20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 20-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 3-Hydroxysteroid Dehydrogenases/genetics , Aldo-Keto Reductase Family 1 Member C3 , Cisplatin/administration & dosage , Drug Resistance, Neoplasm/drug effects , Fluorouracil/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Hydroxysteroid Dehydrogenases/genetics , Neoplasms/genetics , Neoplasms/pathology , Oxidoreductases
7.
Chem Biol Interact ; 240: 310-5, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26362498

ABSTRACT

Recent epidemiological studies show conflicting data for the first-line anti-diabetic sulphonylureas drugs in treating cancer progression in type II diabetes patients. How sulphonylureas promote or diminish tumor growth is not fully understood. Here, we report that seven sulphonylureas exhibit different in vitro inhibition towards AKR1Cs (AKR1C1, AKR1C2, AKR1C3), which are critical steroid hormone metabolism enzymes that are related to prostate cancer, breast cancer and endometrial diseases. Interactions of the sulphonylureas and AKR1Cs were analyzed by X-ray crystallography.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Models, Molecular , Sulfonylurea Compounds/classification , Sulfonylurea Compounds/pharmacology , Aldo-Keto Reductase Family 1 Member C3 , Binding Sites , Humans , Inhibitory Concentration 50 , Molecular Structure , Sulfonylurea Compounds/chemistry
8.
Chem Biol Interact ; 234: 349-59, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25446855

ABSTRACT

The human aldo-keto reductases (AKRs) from the 1C subfamily are important targets for the development of new drugs. In this study, we have investigated the possible interactions between the recombinant AKR1C enzymes AKR1C1-AKR1C3 and ruthenium(II) complexes; in particular, we were interested in the potential inhibitory actions. Five novel ruthenium complexes (1a, 1b, 2a, 2b, 2c), two precursor ruthenium compounds (P1, P2), and three ligands (a, b, c) were prepared and included in this study. Two different types of novel ruthenium(II) complexes were synthesized. First, bearing the sulphur macrocycle [9]aneS3, S-bonded dimethylsulphoxide (dmso-S), and an N,N-donor ligand, with the general formula of [Ru([9]aneS3)(dmso)(N,N-ligand)](PF6)2 (1a, 1b), and second, with the general formula of [(η(6)-p-cymene)RuCl(N,N-ligand)]Cl (2a, 2b, 2c). All of these synthesized compounds were characterized by high-resolution NMR spectroscopy, X-ray crystallography (compounds a, b, c, 1a, 1b) and other standard physicochemical methods. To evaluate the potential inhibitory actions of these compounds on the AKR1C enzymes, we followed enzymatically catalyzed oxidation of the substrate 1-acenaphthenol by NAD(+) in the absence and presence of various micromolar concentrations of the individual compounds. Among 10 compounds, one ruthenium complex (2b) and two precursor ruthenium compounds (P1, P2) inhibited all three AKR1C enzymes, and one ruthenium complex (2a) inhibited only AKR1C3. Ligands a, b and c revealed no inhibition of the AKR1C enzymes. All four of the active compounds showed multiple binding with the AKR1C enzymes that was characterized by an initial instantaneous inhibition followed by a slow quasi-irreversible step. To the best of our knowledge, this is the first study that has examined interactions between these AKR1C enzymes and ruthenium(II) complexes.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Aldehyde Reductase/antagonists & inhibitors , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Ruthenium/pharmacology , Aldo-Keto Reductase Family 1 Member C3 , Aldo-Keto Reductases , Crystallography, X-Ray/methods , Humans , Ligands , Magnetic Resonance Spectroscopy/methods , Oxidation-Reduction
9.
Chem Biol Interact ; 202(1-3): 204-9, 2013 Feb 25.
Article in English | MEDLINE | ID: mdl-23261716

ABSTRACT

The imbalance in expression of the human aldo-keto reductases AKR1C1-AKR1C3 is related to different hormone dependent and independent cancers and some other diseases. The AKR1C1-3 enzymes thus represent emerging targets for the development of new drugs. Currently, various enzymatic assays are used in the search for AKR1C inhibitors, and consequently the results of different research groups are not necessarily comparable. During our recent search for AKR1C inhibitors, we found a cyclopentanol derivative (2-(4-chlorobenzylidene)cyclopentanol, CBCP-ol) and its respective cyclopentanone counterpart (2-(4-chlorobenzylidene)cyclopentanone, CBCP-one) that acted as AKR1C substrates. We determined the kinetic parameters KM, kcat and kcat/KM for oxidation of CBCP-ol and reduction of CBCP-one by AKR1C enzymes in the presence of NAD(+)/NADP(+) and NADH/NADPH, respectively. The catalytic efficiencies for the oxidation of CBCP-ol in the presence of NAD(+) or NADP(+) were in general higher when compared to the catalytic efficiencies for reduction of CBCP-one in the presence of NADH or NADPH. When NADPH was used, as compared to NADH, the reductions of CBCP-one by AKR1C1, AKR1C2 and AKR1C3 were 14-, 51- and 31-fold more efficient, respectively. When comparing to oxidations of the well-known artificial substrates, 1-acenaphthenol and S-tetralol, we observed similar catalytic efficiencies as for CBCP-ol oxidation with NAD(+) and NADP(+). The comparison of CBCP-one reduction with NADPH to reductions of physiological substrates revealed in general higher efficiencies, except for reduction of 9-cis-retinaldehyde by AKR1C3. This NADPH-dependent reduction of CBCP-one was then used to re-evaluate inhibitory potencies of the known inhibitors of the target AKR1C3 and the anti-target AKR1C2, medroxyprogesterone acetate and ursodeoxycholic acid, respectively, showing Ki constants similar to the reported values. Our data thus confirm that the new enzymatic assays with two cyclopentane substrates CBP-ol and CBP-one, and especially reduction of CBCP-one with NADPH, are appropriate for the evaluation of AKR1C inhibitors.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 20-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 3-Hydroxysteroid Dehydrogenases/metabolism , Enzyme Assays/methods , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Hydroxyprostaglandin Dehydrogenases/metabolism , Alcohol Oxidoreductases/metabolism , Aldehyde Reductase , Aldo-Keto Reductase Family 1 Member C3 , Aldo-Keto Reductases , Catalysis , Cyclopentanes/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Medroxyprogesterone Acetate/pharmacology , NAD/metabolism , NADP/metabolism , Oxidation-Reduction , Ursodeoxycholic Acid/pharmacology
10.
J Med Chem ; 55(17): 7417-24, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-22881866

ABSTRACT

Human aldo-keto reductases 1C1-1C4 (AKR1C1-AKR1C4) function in vivo as 3-keto-, 17-keto-, and 20-ketosteroid reductases and regulate the activity of androgens, estrogens, and progesterone and the occupancy and transactivation of their corresponding receptors. Aberrant expression and action of AKR1C enzymes can lead to different pathophysiological conditions. AKR1C enzymes thus represent important targets for development of new drugs. We performed a virtual high-throughput screen of a fragment library that was followed by biochemical evaluation on AKR1C1-AKR1C4 enzymes. Twenty-four structurally diverse compounds were discovered with low µM K(i) values for AKR1C1, AKR1C3, or both. Two structural series included the salicylates and the N-phenylanthranilic acids, and additionally a series of inhibitors with completely novel scaffolds was discovered. Two of the best selective AKR1C3 inhibitors had K(i) values of 0.1 and 2.7 µM, exceeding expected activity for fragments. The compounds identified represent an excellent starting point for further hit-to-lead development.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , 20-Hydroxysteroid Dehydrogenases/chemistry , 3-Hydroxysteroid Dehydrogenases/chemistry , Aldo-Keto Reductase Family 1 Member C3 , Hydroxyprostaglandin Dehydrogenases/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Salicylic Acid/chemistry
11.
J Med Chem ; 55(5): 2311-23, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22263837

ABSTRACT

Aldo-keto reductase 1C3 (AKR1C3; type 5 17ß-hydroxysteroid dehydrogenase) is overexpressed in castration resistant prostate cancer (CRPC) and is implicated in the intratumoral biosynthesis of testosterone and 5α-dihydrotestosterone. Selective AKR1C3 inhibitors are required because compounds should not inhibit the highly related AKR1C1 and AKR1C2 isoforms which are involved in the inactivation of 5α-dihydrotestosterone. NSAIDs, N-phenylanthranilates in particular, are potent but nonselective AKR1C3 inhibitors. Using flufenamic acid, 2-{[3-(trifluoromethyl)phenyl]amino}benzoic acid, as lead compound, five classes of structural analogues were synthesized and evaluated for AKR1C3 inhibitory potency and selectivity. Structure-activity relationship (SAR) studies revealed that a meta-carboxylic acid group relative to the amine conferred pronounced AKR1C3 selectivity without loss of potency, while electron withdrawing groups on the phenylamino B-ring were optimal for AKR1C3 inhibition. Lead compounds did not inhibit COX-1 or COX-2 but blocked the AKR1C3 mediated production of testosterone in LNCaP-AKR1C3 cells. These compounds offer promising leads toward new therapeutics for CRPC.


Subject(s)
3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Fenamates/chemical synthesis , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , 20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Aldo-Keto Reductase Family 1 Member C3 , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/chemical synthesis , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/pharmacology , Fenamates/chemistry , Fenamates/pharmacology , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxyprostaglandin Dehydrogenases/metabolism , Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Isoenzymes/antagonists & inhibitors , Male , Prostatic Neoplasms/drug therapy , Structure-Activity Relationship , Testosterone/antagonists & inhibitors , Testosterone/biosynthesis
12.
Curr Med Chem ; 18(17): 2554-65, 2011.
Article in English | MEDLINE | ID: mdl-21568892

ABSTRACT

The AKR1C aldo-keto reductases (AKR1C1-AKR1C4) are enzymes that interconvert steroidal hormones between their active and inactive forms. In this manner, they can regulate the occupancy and trans-activation of the androgen, estrogen and progesterone receptors. The AKR1C isoforms also have important roles in the production and inactivation of neurosteroids and prostaglandins, and in the metabolism of xenobiotics. They thus represent important emerging drug targets for the development of agents for the treatment of hormone-dependent forms of cancer, like breast, prostate and endometrial cancers, and other diseases, like premenstrual syndrome, endometriosis, catamenial epilepsy and depressive disorders. We present here the physiological roles of these enzymes, along with their structural properties and an overview of the recent developments regarding their inhibitors. The most important strategies of inhibitor design are described, which include the screening of banks of natural compounds (like cinnamic acids, flavonoids, jasmonates, and related compounds), the screening of and structural modifications to non-steroidal anti-inflammatory drugs, the substrate-inspired design of steroidal and nonsteroidal inhibitors, and computer-assisted structure-based inhibitor design.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , 20-Hydroxysteroid Dehydrogenases/physiology , Amino Acid Sequence , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Catalytic Domain , Cinnamates/pharmacology , Drug Design , Drug Discovery , Flavonoids/pharmacology , Gonadal Steroid Hormones/metabolism , Humans , Models, Molecular , Neurotransmitter Agents/metabolism , Protein Structure, Secondary , Salicylates/pharmacology , Sequence Alignment
13.
Bioorg Med Chem Lett ; 21(8): 2564-7, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21414777

ABSTRACT

Human 20α-hydroxysteroid dehydrogenase (AKR1C1) is an important drug target due to its role in the development of lung and endometrial cancers, premature birth and neuronal disorders. We report the crystal structure of AKR1C1 complexed with the first structure-based designed inhibitor 3-chloro-5-phenylsalicylic acid (K(i)=0.86 nM) bound in the active site. The binding of 3-chloro-5-phenylsalicylic acid to AKR1C1 resulted in a conformational change in the side chain of Phe311 to accommodate the bulky phenyl ring substituent at the 5-position of the inhibitor. The contributions of the nonconserved residues Leu54, Leu306, Leu308 and Phe311 to the binding were further investigated by site-directed mutagenesis, and the effects of the mutations on the K(i) value were determined. The Leu54Val and Leu306Ala mutations resulted in 6- and 81-fold increases, respectively, in K(i) values compared to the wild-type enzyme, while the remaining mutations had little or no effects.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 20-Hydroxysteroid Dehydrogenases/genetics , 20-Hydroxysteroid Dehydrogenases/metabolism , Binding Sites , Crystallography, X-Ray , Humans , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Salicylates/chemistry
14.
Chem Biol Interact ; 191(1-3): 227-33, 2011 May 30.
Article in English | MEDLINE | ID: mdl-21182831

ABSTRACT

The human aldo-keto reductases 1C1 and 1C3 (AKR1C1 and AKR1C3) are important 20-ketosteroid reductases in pre-receptor regulation of progesterone action. Both AKR1C1 and AKR1C3 convert progesterone to the less potent metabolite 20α-hydroxyprogesterone, although AKR1C1 has a higher catalytic efficiency than AKR1C3. Recently, we reported significant up-regulation of AKR1C1 and AKR1C3 in ovarian endometriosis, a complex estrogen-dependent disease. The typical characteristics of endometriosis are increased formation of estradiol, which stimulates proliferation of endometriotic tissue, and disturbed action of the protective progesterone. Although progestins have been used for treatment of endometriosis since the 1960s, their detailed mechanisms of action are still not completely understood. In the present study, we evaluated the potential inhibitory effects of progestins on the pre-receptor regulatory enzymes AKR1C1 and AKR1C3. We examined the following progestins as inhibitors of progesterone reduction catalyzed by recombinant AKR1C1 and AKR1C3: progesterone derivatives (dydrogesterone, its metabolite, 20α-hydroxydydrogesterone; and medroxyprogesterone acetate), 19-nortestosterone derivatives (desogestrel, norethinodrone and levonorgestrel), and the androgen danazol. Dydrogesterone, medroxyprogesterone acetate, 20α-hydroxydydrogesterone and norethinodrone inhibited AKR1C1 and AKR1C3 with K(i) values of 1.9 µM, 7.9 µM, 20.8 µM and 48.0 µM, and of 0.5 µM, 1.4 µM, 18.2 µM and 6.6 µM, respectively. Levonorgestrel and desogestrel preferentially inhibited AKR1C3 with K(i) values of 5.6µM and 39.1µM, respectively. Our data thus show that dydrogesterone, medroxyprogesterone acetate, 20α-hydroxydydrogesterone and norethinodrone inhibit AKR1C1 and AKR1C3 in vitro, although their physiological inhibitory effects still need to be evaluated further. Additionally, we investigated whether progestin dydrogesterone can be metabolized to its active 20α-hydroxymetabolite by AKR1C1 and AKR1C3. AKR1C1 converted dydrogesterone with a high catalytic efficiency while AKR1C3 was less active, which suggests that in vivo dydrogesterone is metabolized mainly by AKR1C1. Docking simulations of dydrogesterone into AKR1C1 and AKR1C3 also support these experimental data.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Progestins/pharmacology , 20-Hydroxysteroid Dehydrogenases/chemistry , 20-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxysteroid Dehydrogenases/chemistry , 3-Hydroxysteroid Dehydrogenases/metabolism , Aldo-Keto Reductase Family 1 Member C3 , Biocatalysis , Drug Evaluation, Preclinical , Dydrogesterone/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Hydroxyprostaglandin Dehydrogenases/chemistry , Hydroxyprostaglandin Dehydrogenases/metabolism , Models, Molecular , Oxidation-Reduction/drug effects , Progestins/chemistry , Progestins/metabolism , Protein Conformation
15.
J Steroid Biochem Mol Biol ; 125(1-2): 105-11, 2011 May.
Article in English | MEDLINE | ID: mdl-21050889

ABSTRACT

Human 20α-hydroxysteroid dehydrogenase (AKR1C1), a member of the aldo-keto reductase (AKR) superfamily, is one of four isoforms (with >84% amino acid sequence identity) existing in human tissues. AKR1C1 most efficiently reduces biologically active progesterone and 5α-pregnan-3α-ol-20-one into their corresponding 20α-hydroxysteroids among the isoforms. The enzyme also accepts endogenous and xenobiotic non-steroidal carbonyl compounds as the substrates. In addition to the up-regulation of the AKR1C1 gene in cancer cells, the enzyme's over-expression in the cells of lung, ovary, uterine cervix, skin and colon carcinomas was reported to be associated with resistance against several anticancer agents. Thus, AKR1C1 may be a marker of the above cancers and a target of poor prognosis in cancer therapy. The recently determined X-ray crystal structures of AKR1C1/NADP(+)/20α-hydroxyprogesterone and AKR1C1/NADP(+)/3,5-dichlorosalicylic acid ternary complexes have provided a strong foundation for structure-based design methods to improve inhibitor selectivity and potency. In this review we provide an overview of the different types of AKR1C1 inhibitors and an update on the design of potent and selective inhibitors based on the crystal structure of the enzyme-inhibitor complex. Article from the Special issue on Targeted Inhibitors.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Enzyme Inhibitors/metabolism , Isoenzymes/antagonists & inhibitors , 20-Hydroxysteroid Dehydrogenases/chemistry , 20-Hydroxysteroid Dehydrogenases/metabolism , Animals , Enzyme Inhibitors/chemistry , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Molecular Structure
16.
Eur J Med Chem ; 45(11): 5309-17, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20850205

ABSTRACT

The tertiary structure of the Leu308Val mutant of human 20α-hydroxysteroid dehydrogenase (AKR1C1) in complex with the inhibitor 3,5-dichlorosalicylic acid (DCL) has been determined. Structures and kinetic properties of the wild-type and mutant enzymes indicate that Leu308 is a selectivity determinant for inhibitor binding. The Leu308Val mutation resulted in 13-fold and 3-fold reductions in the inhibitory potencies of DCL and 3-bromo-5-phenylsalicylic acid (BPSA), respectively. The replacement of Leu308 with an alanine resulted in 473-fold and 27-fold reductions in the potencies for DCL and BPSA, respectively. In our attempts to optimize inhibitor potency and selectivity we synthesized 5-substituted 3-chlorosalicylic acid derivatives, of which the most potent compound, 3-chloro-5-phenylsalicylic acid (K(i) = 0.86 nM), was 24-fold more selective for AKR1C1 relative to the structurally similar 3α-hydroxysteroid dehydrogenase (AKR1C2). Furthermore, the compound inhibited the metabolism of progesterone in AKR1C1-overexpressed cells with an IC(50) value equal to 100 nM.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Salicylic Acid/chemistry , Salicylic Acid/pharmacology , 20-Hydroxysteroid Dehydrogenases/genetics , Animals , Cattle , Cells, Cultured , Crystallization , Humans , Inhibitory Concentration 50 , Kinetics , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Structure , Mutagenesis, Site-Directed , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship
17.
Biochim Biophys Acta ; 1801(11): 1221-31, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20673851

ABSTRACT

Cerebral 3α-hydroxysteroid dehydrogenase (3α-HSD) activity was suggested to be responsible for the local directed formation of neuroactive 5α,3α-tetrahydrosteroids (5α,3α-THSs) from 5α-dihydrosteroids. We show for the first time that within human brain tissue 5α-dihydroprogesterone and 5α-dihydrotestosterone are converted via non-stereo-selective 3-ketosteroid reductase activity to produce the respective 5α,3α-THSs and 5α,3ß-THSs. Apart from this, we prove that within the human temporal lobe and limbic system cytochrome P450c17 and 3ß-HSD/Δ(5-4) ketosteroid isomerase are not expressed. Thus, it appears that these brain regions are unable to conduct de novo biosynthesis of Δ(4)-3-ketosteroids from Δ(5)-3ß-hydroxysteroids. Consequently, the local formation of THSs will depend on the uptake of circulating Δ(4)-3-ketosteroids such as progesterone and testosterone. 3α- and 3ß-HSD activity were (i) equally enriched in the cytosol, (ii) showed equal distribution between cerebral neocortex and subcortical white matter without sex- or age-dependency, (iii) demonstrated a strong and significant positive correlation when comparing 46 different specimens and (iv) exhibited similar sensitivities to different inhibitors of enzyme activity. These findings led to the assumption that cerebral 3-ketosteroid reductase activity might be catalyzed by a single enzyme and is possibly attributed to the expression of a soluble AKR1C aldo-keto reductase. AKR1Cs are known to act as non-stereo-selective 3-ketosteroid reductases; low AKR1C mRNA expression was detected. However, the cerebral 3-ketosteroid reductase was clearly refractory to inhibition by AKR1C inhibitors indicating the expression of a currently unidentified enzyme. Its lack of stereo-selectivity is of physiological significance, since only 5α,3α-THSs enhance the effect of GABA on the GABA(A) receptor, whereas 5α,3ß-THSs are antagonists.


Subject(s)
20-Hydroxysteroid Dehydrogenases/genetics , Brain/metabolism , Gene Expression Regulation, Enzymologic , 20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 3-Hydroxysteroid Dehydrogenases/metabolism , Adult , Aged , Brain/pathology , Cell Line, Tumor , Chromatography, Thin Layer/methods , Female , Gas Chromatography-Mass Spectrometry/methods , Humans , Infant , Middle Aged , Steroid 17-alpha-Hydroxylase/genetics , Steroids/chemistry , Temporal Lobe/pathology
18.
Cancer Res ; 69(11): 4769-75, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19487289

ABSTRACT

Members of the aldo-keto reductase (AKR) superfamily, particularly the AKR1C subfamily, are emerging as important mediators of the pathology of cancer. Agents that inhibit these enzymes may provide novel agents for either the chemoprevention or treatment of diverse malignancies. Recently, jasmonates, a family of plant stress hormones that bear a structural resemblance to prostaglandins, have been shown to elicit anticancer activities both in vitro and in vivo. In this study, we show that jasmonic acid (JA) and methyl jasmonate (MeJ) are capable of inhibiting all four human AKR1C isoforms. Although JA is the more potent inhibitor of recombinant AKR1C proteins, including the in vitro prostaglandin F synthase activity of AKR1C3, MeJ displayed greater potency in cellular systems that was, at least in part, due to increased cellular uptake of MeJ. Moreover, using the acute myelogenous leukemia cell lines HL-60 and KG1a, we found that although both jasmonates were able to induce high levels of reactive oxygen species in a dose-dependent fashion, only MeJ was able to induce high levels of mitochondrial superoxide (MSO), possibly as an epiphenomenon of mitochondrial damage. There was a strong correlation observed between MSO formation at 24 hours and reduced cellularity at day 5. In conclusion, we have identified AKR1C isoforms as a novel target of jasmonates in cancer cells and provide further evidence of the promise of these compounds, or derivatives thereof, as adjunctive therapies in the treatment of cancer.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Acetates/pharmacology , Cyclopentanes/pharmacology , Drug Delivery Systems , Mitochondria/drug effects , Oxylipins/pharmacology , 20-Hydroxysteroid Dehydrogenases/metabolism , 20-Hydroxysteroid Dehydrogenases/physiology , Acetates/pharmacokinetics , Cell Survival/drug effects , Cells, Cultured , Cyclopentanes/pharmacokinetics , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , HL-60 Cells , Humans , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Hydroxyprostaglandin Dehydrogenases/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Isoenzymes/physiology , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Mitochondria/physiology , Models, Biological , Oxylipins/pharmacokinetics , Prostaglandin D2/metabolism , Reactive Oxygen Species/metabolism
19.
J Med Chem ; 52(10): 3259-64, 2009 May 28.
Article in English | MEDLINE | ID: mdl-19397269

ABSTRACT

The first design, synthesis, and evaluation of human 20alpha-hydroxysteroid dehydrogenase (AKR1C1) inhibitors based on the recently published crystal structure of its ternary complex with inhibitor are reported. While the enzyme-inhibitor interactions observed in the crystal structure remain conserved with the newly designed inhibitors, the additional phenyl group of the most potent compound, 3-bromo-5-phenylsalicylic acid, targets a nonconserved hydrophobic pocket in the active site of AKR1C1 resulting in 21-fold improved potency (K(i) = 4 nM) over the structurally similar 3alpha-hydroxysteroid dehydrogenase isoform (AKR1C2). The compound is hydrogen bonded to Tyr55, His117, and His222, and the phenyl ring forms additional van der Waals interactions with residues Leu308, Phe311, and the nonconserved Leu54 (Val in AKR1C2). Additionally, the metabolism of progesterone in AKR1C1-overexpressed cells was potently inhibited by 3-bromo-5-phenylsalicylic acid, which was effective from 10 nM with an IC(50) value equal to 460 nM.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/chemical synthesis , Salicylic Acid/pharmacology , Catalytic Domain , Cell Line , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Bonding , Progesterone/metabolism , Protein Binding , Salicylates , Salicylic Acid/chemical synthesis , Salicylic Acid/chemistry , Structure-Activity Relationship
20.
Eur J Med Chem ; 44(6): 2563-71, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19237229

ABSTRACT

A series of cyclopentane derivatives was synthesized and evaluated for inhibition of the steroid metabolizing enzymes AKR1C1 and AKR1C3. Selective inhibitors that are active in the low micromolar range were identified. These compounds represent promising starting points in the development of new anticancer agents for the treatment of hormone-dependent forms of cancer and other diseases where AKR1C1 and AKR1C3 are involved.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Cyclopentanes/pharmacology , Enzyme Inhibitors/pharmacology , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Aldo-Keto Reductase Family 1 Member C3 , Computer Simulation , Cyclopentanes/chemical synthesis , Cyclopentanes/chemistry , Dose-Response Relationship, Drug , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Models, Chemical , Molecular Conformation , Stereoisomerism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...