Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Cell Rep ; 39(1): 110641, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35385746

ABSTRACT

CDC42 family GTPases (RHOJ, RHOQ, CDC42) are upregulated but rarely mutated in cancer and control both the ability of tumor cells to invade surrounding tissues and the ability of endothelial cells to vascularize tumors. Here, we use computer-aided drug design to discover a chemical entity (ARN22089) that has broad activity against a panel of cancer cell lines, inhibits S6 phosphorylation and MAPK activation, activates pro-inflammatory and apoptotic signaling, and blocks tumor growth and angiogenesis in 3D vascularized microtumor models (VMT) in vitro. Additionally, ARN22089 has a favorable pharmacokinetic profile and can inhibit the growth of BRAF mutant mouse melanomas and patient-derived xenografts in vivo. ARN22089 selectively blocks CDC42 effector interactions without affecting the binding between closely related GTPases and their downstream effectors. Taken together, we identify a class of therapeutic agents that influence tumor growth by modulating CDC42 signaling in both the tumor cell and its microenvironment.


Subject(s)
Endothelial Cells , Neoplasms , Animals , Endothelial Cells/metabolism , Humans , Mice , Neoplasms/drug therapy , Neovascularization, Pathologic , Signal Transduction , Tumor Microenvironment , cdc42 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism
3.
J Med Chem ; 63(21): 12873-12886, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33079544

ABSTRACT

We disclose a novel class of 6-amino-tetrahydroquinazoline derivatives that inhibit human topoisomerase II (topoII), a validated target of anticancer drugs. In contrast to topoII-targeted drugs currently in clinical use, these compounds do not act as topoII poisons that enhance enzyme-mediated DNA cleavage, a mechanism that is linked to the development of secondary leukemias. Instead, these tetrahydroquinazolines block the topoII function with no evidence of DNA intercalation. We identified a potent lead compound [compound 14 (ARN-21934) IC50 = 2 µM for inhibition of DNA relaxation, as compared to an IC50 = 120 µM for the anticancer drug etoposide] with excellent metabolic stability and solubility. This new compound also shows ~100-fold selectivity for topoIIα over topoß, a broad antiproliferative activity toward cultured human cancer cells, a favorable in vivo pharmacokinetic profile, and the ability to penetrate the blood-brain barrier. Thus, ARN-21934 is a highly promising lead for the development of novel and potentially safer topoII-targeted anticancer drugs.


Subject(s)
DNA Topoisomerases, Type II/chemistry , Quinidine/analogs & derivatives , Topoisomerase II Inhibitors/chemistry , Animals , Cell Line, Tumor , Cell Survival/drug effects , DNA/chemistry , DNA/metabolism , DNA Cleavage , DNA Topoisomerases, Type II/metabolism , Drug Screening Assays, Antitumor , Half-Life , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Mice , Quinidine/chemistry , Quinidine/metabolism , Quinidine/pharmacology , Topoisomerase II Inhibitors/metabolism , Topoisomerase II Inhibitors/pharmacology
4.
Eur J Med Chem ; 202: 112504, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32712536

ABSTRACT

Drugs targeting human topoisomerase II (topoII) are used in clinical practice since decades. Nevertheless, there is an urgent need for new and safer topoII inhibitors due to the emergence of secondary malignancies and the appearance of resistance mechanisms upon treatment with topoII-targeted anticancer drugs. In the present investigation, we report the discovery of a new topoII inhibitor, whose design was based on the structure of the natural product trypthantrin, a natural alkaloid containing a basic indoloquinazoline moiety. This new topoII inhibitor, here numbered compound 5, is found to inhibit topoII with an IC50 of 26.6 ± 4.7 µM. Notably, compound 5 is more potent than the template compound trypthantrin, and even than the widely used topoII-targeted clinical drug etoposide. In addition, compound 5 also exhibits high water solubility, and a promising antiproliferative activity on different tumor cell lines such as acute leukemia, colon, and breast cancer. In light of these results, compound 5 represents a promising lead for developing new topoII inhibitors as anti-cancer therapeutic agents.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Topoisomerases, Type II/metabolism , Drug Discovery , Topoisomerase II Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistry , Tumor Cells, Cultured
5.
J Med Chem ; 63(7): 3508-3521, 2020 04 09.
Article in English | MEDLINE | ID: mdl-32196342

ABSTRACT

We previously reported a first set of hybrid topoisomerase II (topoII) poisons whose chemical core merges key pharmacophoric elements of etoposide and merbarone, which are two well-known topoII blockers. Here, we report on the expansion of this hybrid molecular scaffold and present 16 more hybrid derivatives that have been designed, synthesized, and characterized for their ability to block topoII and for their overall drug-like profile. Some of these compounds act as topoII poison and exhibit good solubility, metabolic (microsomal) stability, and promising cytotoxicity in three cancer cell lines (DU145, HeLa, A549). Compound 3f (ARN24139) is the most promising drug-like candidate, with a good pharmacokinetics profile in vivo. Our results indicate that this hybrid new chemical class of topoII poisons deserves further exploration and that 3f is a favorable lead candidate as a topoII poison, meriting future studies to test its efficacy in in vivo tumor models.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Topoisomerase II Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/metabolism , Drug Design , Drug Screening Assays, Antitumor , Humans , Mice , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Poly-ADP-Ribose Binding Proteins/chemistry , Poly-ADP-Ribose Binding Proteins/metabolism , Protein Binding , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/metabolism , Topoisomerase II Inhibitors/pharmacokinetics
6.
J Med Chem ; 61(3): 1375-1379, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29077404

ABSTRACT

We used a pharmacophore hybridization strategy to combine key structural elements of merbarone and etoposide and generated new type II topoisomerase (topoII) poisons. This first set of hybrid topoII poisons shows promising antiproliferative activity on human cancer cells, endorsing their further exploration for anticancer drug discovery.


Subject(s)
DNA Topoisomerases, Type II/metabolism , Drug Design , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Topoisomerases, Type II/chemistry , Humans , Molecular Docking Simulation , Protein Conformation , Topoisomerase II Inhibitors/metabolism
7.
J Med Chem ; 60(13): 5800-5815, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28603987

ABSTRACT

Acid ceramidase (AC) hydrolyzes ceramides, which are central lipid messengers for metabolism and signaling of sphingolipids. A growing body of evidence links deregulation of sphingolipids to several diseases, including cancer. Indeed, AC expression is abnormally high in melanoma cells. AC inhibition may thus be key to treating malignant melanoma. Here, we have used a systematic scaffold exploration to design a general pharmacophore for AC inhibition. This pharmacophore comprises a 6 + 5 fused ring heterocycle linked to an aliphatic substituent via a urea moiety. We have thus identified the novel benzimidazole derivatives 10, 21, 27, and 30, which are highly potent AC inhibitors. Their chemical and metabolic stabilities are comparable or superior to those of previously reported AC inhibitors. Moreover, they are potent against endogenous AC in intact melanoma cells. These novel inhibitors merit further characterization and can serve as a promising starting point for the discovery of new antimelanoma therapeutics.


Subject(s)
Acid Ceramidase/antagonists & inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Acid Ceramidase/metabolism , Animals , Antineoplastic Agents/blood , Benzimidazoles/blood , Cell Line, Tumor , Cell Survival/drug effects , Drug Stability , Enzyme Inhibitors/blood , HEK293 Cells , Humans , Melanoma/drug therapy , Melanoma/metabolism , Mice
8.
ACS Chem Biol ; 12(2): 564-573, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28045490

ABSTRACT

There is a current and pressing need for improved cancer therapies. The use of small molecule kinase inhibitors and their application in combinatorial regimens represent an approach to personalized targeted cancer therapy. A number of AGC kinases, including atypical Protein Kinase C enzymes (PKCs), are validated drug targets for cancer treatment. Most drug development programs for protein kinases focus on the development of drugs that bind at the ATP-binding site. Alternatively, allosteric drugs have great potential for the development of future innovative drugs. However, the rational development of allosteric drugs poses important challenges because the compounds not only must bind to a given site but also must stabilize forms of the protein with a desired effect at a distant site. Here we describe the development of a new class of compounds targeting a regulatory site (PIF-pocket) present in the kinase domain and provide biochemical and crystallographic data showing that these compounds allosterically inhibit the activity of atypical PKCs. PS432, a representative compound, decreased the rate of proliferation of non-small cell lung cancer cells more potently than aurothiomalate, an atypical PKCι inhibitor currently under evaluation in clinical trials, and significantly reduced tumor growth without side effects in a mouse xenograft model. The druglike chemical class provides ample possibilities for the synthesis of derivative compounds, with the potential to allosterically modulate the activity of atypical PKCs and other kinases.


Subject(s)
Isoenzymes/metabolism , Protein Kinase C/metabolism , Allosteric Regulation , Animals , Cell Line, Tumor , Female , Heterografts , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Mice, Nude
9.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(5): 441-451, 2017 May.
Article in English | MEDLINE | ID: mdl-28088576

ABSTRACT

Human monoacylglycerol lipase (MAGL) is a membrane-interacting enzyme that generates pro-inflammatory signaling molecules. For this reason, MAGL inhibition is a promising strategy to treat pain, cancer, and neuroinflammatory diseases. MAGL can hydrolyze monoacylglycerols bearing an acyl chain of different lengths and degrees of unsaturation, cleaving primarily the endocannabinoid 2-arachidonoylglycerol. Importantly, the enzymatic binding site of MAGL is confined by a 75-amino-acid-long, flexible cap domain, named 'lid domain', which is structurally similar to that found in several other lipases. However, it is unclear how lid domain plasticity affects catalysis in MAGL. By integrating extensive molecular dynamics simulations and free-energy calculations with mutagenesis and kinetic experiments, we here define a lid-domain-mediated mechanism for substrate selection and binding in MAGL catalysis. In particular, we clarify the key role of Phe159 and Ile179, two conserved residues within the lid domain, in regulating substrate specificity in MAGL. We conclude by proposing that other structurally related lipases may share this lid-domain-mediated mechanism for substrate specificity.


Subject(s)
Monoacylglycerol Lipases/chemistry , Monoacylglycerol Lipases/metabolism , Monoglycerides/chemistry , Catalysis , Enzyme Inhibitors/chemistry , Humans , Kinetics , Molecular Dynamics Simulation , Monoacylglycerol Lipases/genetics , Monoglycerides/metabolism , Protein Binding , Protein Domains , Substrate Specificity
10.
Cancer Res ; 75(15): 3147-54, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26239478

ABSTRACT

iRGD is a derivative of the integrin-binding peptide RGD, which selectively increases the penetrability of tumor tissue to various coadministered substances in several preclinical models. In this study, we investigated the ability of iRGD to improve the delivery of sorafenib and doxorubicin therapy in hepatocellular carcinoma (HCC) using established mouse models of the disease. A contrast-enhanced MRI method was developed in parallel to assess the in vivo effects of iRGD in this setting. We found that iRGD improved the delivery of marker substances to the tumors of HCC-bearing mice about three-fold without a parallel increase in normal tissues. Control peptides lacking the critical CendR motif had no effect. Similarly, iRGD also selectively increased the signal intensity from tumors in Gd-DTPA-enhanced MRI. In terms of antitumor efficacy, iRGD coadministration significantly augmented the individual inhibitory effects of sorafenib and doxorubicin without increasing systemic toxicity. Overall, our results offered a preclinical proof of concept for the use of iRGD coadministration as a strategy to widen the therapeutic window for HCC chemotherapy, as monitored by Gd-DTPA-enhanced MRI as a noninvasive, clinically applicable method to identify iRGD-reactive tumors.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Doxorubicin/administration & dosage , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Oligopeptides/administration & dosage , Phenylurea Compounds/administration & dosage , Administration, Intravenous , Amino Acid Motifs , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Doxorubicin/pharmacokinetics , Drug Delivery Systems , Evans Blue/administration & dosage , Gadolinium DTPA , Hep G2 Cells/drug effects , Humans , Magnetic Resonance Imaging , Male , Mice, Nude , Mice, Transgenic , Niacinamide/administration & dosage , Oligopeptides/chemistry , Sorafenib , Tissue Distribution
11.
Chem Biol ; 21(6): 754-65, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24836908

ABSTRACT

Protein kinases play important regulatory roles in cells and organisms. Therefore, they are subject to specific and tight mechanisms of regulation that ultimately converge on the catalytic domain and allow the kinases to be activated or inhibited only upon the appropriate stimuli. AGC protein kinases have a pocket in the catalytic domain, the PDK1-interacting fragment (PIF)-pocket, which is a key mediator of the activation. We show here that helix αC within the PIF-pocket of atypical protein kinase C (aPKC) is the target of the interaction with its inhibitory N-terminal domains. We also provide structural evidence that the small compound PS315 is an allosteric inhibitor that binds to the PIF-pocket of aPKC. PS315 exploits the physiological dynamics of helix αC for its binding and allosteric inhibition. The results will support research on allosteric mechanisms and selective drug development efforts against PKC isoforms.


Subject(s)
Biphenyl Compounds/pharmacology , Cinnamates/pharmacology , Protein Kinase C/chemistry , Protein Kinase C/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Allosteric Regulation/drug effects , Biphenyl Compounds/chemistry , Cinnamates/chemistry , Humans , Models, Molecular , Molecular Structure , Protein Kinase C/antagonists & inhibitors , Protein Structure, Tertiary/drug effects , Structure-Activity Relationship
12.
Biochim Biophys Acta ; 1834(7): 1302-21, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23524293

ABSTRACT

The group of AGC protein kinases includes more than 60 protein kinases in the human genome, classified into 14 families: PDK1, AKT/PKB, SGK, PKA, PKG, PKC, PKN/PRK, RSK, NDR, MAST, YANK, DMPK, GRK and SGK494. This group is also widely represented in other eukaryotes, including causative organisms of human infectious diseases. AGC kinases are involved in diverse cellular functions and are potential targets for the treatment of human diseases such as cancer, diabetes, obesity, neurological disorders, inflammation and viral infections. Small molecule inhibitors of AGC kinases may also have potential as novel therapeutic approaches against infectious organisms. Fundamental in the regulation of many AGC kinases is a regulatory site termed the "PIF-pocket" that serves as a docking site for substrates of PDK1. This site is also essential to the mechanism of activation of AGC kinases by phosphorylation and is involved in the allosteric regulation of N-terminal domains of several AGC kinases, such as PKN/PRKs and atypical PKCs. In addition, the C-terminal tail and its interaction with the PIF-pocket are involved in the dimerization of the DMPK family of kinases and may explain the molecular mechanism of allosteric activation of GRKs by GPCR substrates. In this review, we briefly introduce the AGC kinases and their known roles in physiology and disease and the discovery of the PIF-pocket as a regulatory site in AGC kinases. Finally, we summarize the current status and future therapeutic potential of small molecules directed to the PIF-pocket; these molecules can allosterically activate or inhibit the kinase as well as act as substrate-selective inhibitors. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases (2012).


Subject(s)
Models, Molecular , Protein Kinases/chemistry , Protein Kinases/metabolism , Protein Structure, Tertiary , Allosteric Regulation , Candida albicans/drug effects , Candida albicans/enzymology , Candidiasis/drug therapy , Candidiasis/microbiology , Humans , Neoplasms/drug therapy , Neoplasms/enzymology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinases/classification
13.
Chem Biol ; 19(9): 1152-63, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22999883

ABSTRACT

The PIF-pocket of AGC protein kinases participates in the physiologic mechanism of regulation by acting as a docking site for substrates and as a switch for the transduction of the conformational changes needed for activation or inhibition. We describe the effects of compounds that bind to the PIF-pocket of PDK1. In vitro, PS210 is a potent activator of PDK1, and the crystal structure of the PDK1-ATP-PS210 complex shows that PS210 stimulates the closure of the kinase domain. However, in cells, the prodrug of PS210 (PS423) acts as a substrate-selective inhibitor of PDK1, inhibiting the phosphorylation and activation of S6K, which requires docking to the PIF-pocket, but not affecting PKB/Akt. This work describes a tool to study the dynamics of PDK1 activity and a potential approach for drug discovery.


Subject(s)
Allosteric Site/drug effects , Chalcones/pharmacology , Dicarboxylic Acids/pharmacology , Prodrugs/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line , Chalcones/chemistry , Dicarboxylic Acids/chemistry , HEK293 Cells , Humans , Mice , Models, Biological , Models, Molecular , Molecular Structure , Molecular Weight , Prodrugs/chemistry , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Structure-Activity Relationship , Substrate Specificity
14.
Int J Oncol ; 34(2): 457-63, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19148481

ABSTRACT

Prostate cancer is one of the most frequent malignancies in the Western world. The identification of additional molecular markers is needed to refine the diagnosis of prostate cancer and to develop more effective therapies. In order to identify molecular abnormalities involved in prostate cancer progression, we performed gene expression analysis of prostate cancer samples compared to matched normal tissue from the same patient using a cancer-related microarray. Amplified RNA was hybridized to a cDNA microarray containing 6386 genes and tissue microarrays were used to study protein expression levels. Using significance analysis of microarrays, we identified >1300 genes differentially expressed in prostate cancer compared to normal tissue. Forty-two of these genes were highly upregulated in prostate cancer while 169 were highly repressed. We found that the gene coding for tspan13 was upregulated >2-fold in 75% of the samples analyzed. Immunohistochemistry analysis of prostate cancer tissue microarrays showed that tspan13 is overexpressed in 80% of prostate cancer samples analyzed. We found that tspan13 expression inversely correlates with Gleason score (p=0.01) and PSA preoperative levels (p=0.11) and directly correlates with presence of prostatic intraepithelial neoplasia in tumor tissue (p=0.04). Moreover, we detected tspan13 expression in low-grade prostatic intraepithelial neoplasia. Thus, our results show that tspan13 is overexpressed in prostate cancer and its expression correlates with factors of favourable outcome. Therefore we suggest that tspan13 may have an important role in the progression of prostate cancer.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Prostatic Neoplasms/genetics , DNA, Complementary/genetics , DNA, Neoplasm/genetics , Humans , Immunohistochemistry , Male , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Tetraspanins , Treatment Outcome , Up-Regulation
15.
Int J Oncol ; 27(6): 1617-23, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16273218

ABSTRACT

Doxazosin is a quinazoline-based compound acting as an alpha-1-adrenergic inhibitor shown to induce apoptosis in prostate cancer cell lines via an alpha-1-adrenergic receptor-independent mechanism. To better understand the mechanism of doxazosin-induced apoptosis in prostate cancer, we performed cDNA microarray to analyze gene expression changes produced by doxazosin in the androgen-dependent human prostate cancer cell line, LNCaP. We found that 70 and 92 genes were deregulated after 8 and 24 h of doxazosin treatment, respectively. These genes are involved in several cellular processes such as cell-cycle regulation, cell adhesion and signal transduction pathways. Strikingly, we found that doxazosin induces deregulation of genes implicated in DNA replication and repair, such as GADD45A, XRCC5 and PRKDC. These facts, together with the demonstration of the ability of doxazosin to bind DNA, allowed us to propose a novel mechanism of action for doxazosin in prostate cancer cells that implies DNA-damage mediated apoptosis by down-regulation of XRCC5 and PRKDC genes.


Subject(s)
Apoptosis/drug effects , DNA, Neoplasm/metabolism , DNA-Activated Protein Kinase/genetics , Doxazosin/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Binding, Competitive , Cell Line, Tumor , Cell Proliferation/drug effects , DNA, Superhelical/chemistry , DNA, Superhelical/genetics , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/genetics , Doxazosin/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Nucleic Acid Conformation/drug effects , Oligonucleotide Array Sequence Analysis/methods , Plasmids/chemistry , Plasmids/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction
16.
Anticancer Drugs ; 13(9): 949-56, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12394258

ABSTRACT

The receptors for luteinizing hormone-releasing hormone (LHRH) are found in 80% of human ovarian carcinomas. These receptors can be used for targeted chemotherapy with cytotoxic analogs of LHRH, such as AN-207, consisting of 2-pyrrolinodoxorubicin (AN-201) linked to [D-Lys ]LHRH. We investigated the effects of AN-207 and AN-201 on the growth of LHRH receptor-positive ES-2 human ovarian cancers. The effects of the treatment on mRNA and protein levels of human epidermal growth factor (EGF) receptors (EGFR and HER-2) in ovarian tumors were determined by RT-PCR and immunoblotting. In Experiment 1, nude mice bearing ES-2 ovarian tumors were injected i.v. with 250 nmol/kg doses of AN-207, AN-201, the carrier [D-Lys ]LHRH, an unconjugated mixture of AN-201 and [D-Lys ]LHRH or vehicle. AN-207 caused a significant ( <0.01) 59.5% inhibition in tumor growth while its components were ineffective. In Experiment 2, mice with large ES-2 tumors were treated with AN-207 or AN-201 at 250 nmol/kg. Again, AN-207, but not AN-201, inhibited tumor growth. In Experiment 3, the site of action of AN-207 was investigated. The blockade of LHRH receptors with Cetrorelix partially suppressed the antitumor effect of AN-207. Treatment with AN-207 significantly ( <0.01) decreased the expression of mRNA for EGFR, and HER-2 by 27 and 34%, respectively, as compared to controls and reduced the receptor protein levels of EGFR and HER-2 by 35 and 36%, respectively ( <0.05). The results indicate that cytotoxic LHRH analog AN-207 could be considered for chemotherapy of ovarian cancers expressing LHRH receptors.


Subject(s)
Antineoplastic Agents/therapeutic use , Doxorubicin/analogs & derivatives , Doxorubicin/therapeutic use , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/therapeutic use , Ovarian Neoplasms/drug therapy , Receptors, LHRH/biosynthesis , Animals , Drug Evaluation, Preclinical , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/biosynthesis , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/metabolism , Pyrroles/therapeutic use , RNA, Messenger/analysis , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/biosynthesis , Receptors, LHRH/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Treatment Outcome , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...