Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.098
Filter
2.
Horm Metab Res ; 46(1): 21-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23839655

ABSTRACT

Stromal cells strictly modulate the differentiation of the normal prostate epithelium. In benign prostatic hyperplasia (BPH) tissue, the ratio of stromal to epithelial cells reaches a 5:1 ratio. In this study, we evaluated the effects of crossover conditioned media (CM) of stromal and epithelial prostate cells before and after treatment with LHRH antagonist Cetrorelix. WPMY-1 human prostate stromal cells and BPH-1 human benign prostatic hyperplasia cells were cultured in vitro and the effects of crossover conditioned media (CM) from those cells were studied. We evaluated the effect of Cetrorelix on the expression of PCNA and p53 in those cells. We then studied the effect of Cetrorelix on BPH-1 cells cultured with the CM from WPMY-1 cells, as well as the mechanisms which govern these interactions. CM from WPMY-1 cells strongly stimulated the proliferation of BPH-1 cells in a dose dependent manner, while CM from BPH-1 cells only slightly increased the proliferation of WPMY-1 cells. Cetrorelix inhibited the proliferation of both cell lines and the expression of PCNA, while the expression of p53 was increased. Cetrorelix also inhibited the proliferation of BPH-1 cells stimulated with the CM from WPMY-1 cells. In the crossover experiment, conditioned media from WPMY-1 and BPH-1 cells increased the expression of phosphorylated ERK1/2 and STAT3. Our results support previous observations on the bidirectional stromal-epithelial interactions in prostate gland and shed more light on the mechanistic action of those effects. Our study strongly supports the hypothesis that LHRH antagonists may be beneficial for BPH prevention and treatment.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/drug effects , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Prostate/cytology , Cell Line , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Enzyme Activation/drug effects , Epithelial Cells/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Humans , Male , Phosphorylation/drug effects , Proliferating Cell Nuclear Antigen/metabolism , Receptors, LHRH/metabolism , STAT3 Transcription Factor/metabolism , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/enzymology , Tumor Suppressor Protein p53/metabolism
3.
Oncogene ; 33(45): 5251-61, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-24186207

ABSTRACT

Prostate cancer is the most commonly diagnosed malignancy among men in industrialized countries, accounting for the second leading cause of cancer-related deaths. Although we now know that the androgen receptor (AR) is important for progression to the deadly advanced stages of the disease, it is poorly understood what AR-regulated processes drive this pathology. Here we demonstrate that AR regulates prostate cancer cell growth via the metabolic sensor 5'-AMP-activated protein kinase (AMPK), a kinase that classically regulates cellular energy homeostasis. In patients, activation of AMPK correlated with prostate cancer progression. Using a combination of radiolabeled assays and emerging metabolomic approaches, we also show that prostate cancer cells respond to androgen treatment by increasing not only rates of glycolysis, as is commonly seen in many cancers, but also glucose and fatty acid oxidation. Importantly, this effect was dependent on androgen-mediated AMPK activity. Our results further indicate that the AMPK-mediated metabolic changes increased intracellular ATP levels and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α)-mediated mitochondrial biogenesis, affording distinct growth advantages to the prostate cancer cells. Correspondingly, we used outlier analysis to determine that PGC-1α is overexpressed in a subpopulation of clinical cancer samples. This was in contrast to what was observed in immortalized benign human prostate cells and a testosterone-induced rat model of benign prostatic hyperplasia. Taken together, our findings converge to demonstrate that androgens can co-opt the AMPK-PGC-1α signaling cascade, a known homeostatic mechanism, to increase prostate cancer cell growth. The current study points to the potential utility of developing metabolic-targeted therapies directed toward the AMPK-PGC-1α signaling axis for the treatment of prostate cancer.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Androgens/pharmacology , Cell Proliferation/drug effects , Prostatic Neoplasms/metabolism , Transcription Factors/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Glycolysis/drug effects , Humans , Kaplan-Meier Estimate , Male , Metribolone/pharmacology , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/ultrastructure , Oxidative Phosphorylation/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA Interference , Rats, Wistar , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factors/genetics
4.
Oncol Rep ; 30(1): 413-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23624870

ABSTRACT

In preclinical studies, antagonists of growth hormone-releasing hormone (GHRH) have demonstrated inhibitory effects on the growth of various types of cancers expressing the pituitary type of GHRH receptors (pGHRH-R) and/or its active splice variant 1 (SV1). In this study, we investigated the effectiveness of the treatment of MDA-MB-231 human triple-negative breast cancer (TNBC) with GHRH antagonist JMR-132 alone or in combination with docetaxel. Receptor expression in the MDA-MB-231 human breast cancer cell line was evaluated by reverse transcription-polymerase chain reaction (RT-PCR). Cell viability assays were performed on MDA-MB-231 cells treated with JMR-132, docetaxel or in combination. For studies in vivo, a subcutaneous nude mouse xenograft model was used. JMR-132 was administered s.c. at a dose of 10 µg/day and docetaxel at a dose of 10 mg/kg i.p. given on day 1 and 5. Similar regimens were used for the combination of both substances. At the end of the experiment, an mRNA-based human cancer pathway array including 84 major genes was performed on the tumor tissue of mice treated with JMR-132 to elucidate the mechanism of action of GHRH antagonists in vivo. The in vitro proliferation studies revealed that JMR-132 and docetaxel decreased the cell viability in a dose-dependent manner. The combination of both treatments produced a significantly greater inhibition of cell viability compared to the single agents. Treatment of nude mice bearing MDA-MB-231 xenografts with JMR-132 and docetaxel significantly (p<0.05) inhibited tumor growth by 46 and 50%, respectively. Treatment with the combination of JMR-132 and docetaxel led to an inhibition of tumor volume by 71.6% (p<0.001). Polymerase chain reaction array analysis revealed that JMR-132 interacts with signal transduction pathways involved in proliferation, apoptosis and angiogenesis. Our results suggest that GHRH antagonists in combination with taxanes may enhance the efficacy of treatment for patients with TNBC expressing the SV1 and/or the pGHRH receptor.


Subject(s)
Antineoplastic Agents/therapeutic use , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Sermorelin/analogs & derivatives , Taxoids/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Docetaxel , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Sermorelin/therapeutic use , Triple Negative Breast Neoplasms/genetics , Xenograft Model Antitumor Assays
5.
Mol Cell Endocrinol ; 371(1-2): 189-94, 2013 May 22.
Article in English | MEDLINE | ID: mdl-23267837

ABSTRACT

Pheochromocytoma is a rare but potentially lethal chromaffin cell tumor with currently no effective treatment. Peptide hormone receptors are frequently overexpressed on endocrine tumor cells and can be specifically targeted by various anti-tumor peptide analogs. The present study carried out on mouse pheochromocytoma cells (MPCs) and a more aggressive mouse tumor tissue-derived (MTT) cell line revealed that these cells are characterized by pronounced expression of the somatostatin receptor 2 (sst2), growth hormone-releasing hormone (GHRH) receptor and the luteinizing hormone-releasing hormone (LHRH) receptor. We further demonstrated significant anti-tumor effects mediated by cytotoxic somatostatin analogs, AN-162 and AN-238, by LHRH antagonist, Cetrorelix, by the cytotoxic LHRH analog, AN-152, and by recently developed GHRH antagonist, MIA-602, on MPC and for AN-152 and MIA-602 on MTT cells. Studies of novel anti-tumor compounds on these mouse cell lines serve as an important basis for mouse models of metastatic pheochromocytoma, which we are currently establishing.


Subject(s)
Adrenal Gland Neoplasms/drug therapy , Pheochromocytoma/drug therapy , Receptors, Neuropeptide/drug effects , 2-Hydroxyphenethylamine/analogs & derivatives , 2-Hydroxyphenethylamine/pharmacology , Aniline Compounds/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Gonadotropin-Releasing Hormone/pharmacology , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Mice , Pyrroles/pharmacology , Receptors, LHRH/biosynthesis , Receptors, LHRH/drug effects , Receptors, LHRH/metabolism , Receptors, Neuropeptide/biosynthesis , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/biosynthesis , Receptors, Pituitary Hormone-Regulating Hormone/drug effects , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Receptors, Somatostatin/biosynthesis , Receptors, Somatostatin/drug effects , Receptors, Somatostatin/metabolism , Sermorelin/analogs & derivatives , Sermorelin/pharmacology , Somatostatin/analogs & derivatives
6.
Arch Gynecol Obstet ; 286(2): 437-42, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22555802

ABSTRACT

Receptors luteinizing hormone-releasing hormone (LHRH) are expressed in about 80 % of human endometrial and ovarian cancers and account for more than 50 % of breast cancers including triple negative breast cancers. Apart from the pituitary and reproductive organs, no other organs or hematopoietic stem cells express LHRH (GnRH) receptors. Thus, these receptors can be regarded as an ideal target for a personalized medicine approach in cancer therapy. AEZS-108 (formerly known as AN-152) in which doxorubin is linked to the LHRH agonist [D: -Lys(6)]LHRH, appears to be the most advanced compound in late stage clinical development. Results of phase I and phase II clinical trials in patients with gynecological cancers demonstrated anticancer activity without any cardiotoxicity even in highly pretreated patients. AEZS-108 is therefore being considered for phase II trials in triple negative breast cancers and phase III studies in advanced endometrial cancers positive for LHRH-receptor. EP-100 is a membrane-disrupting peptide targeted to LHRH receptors, which is undergoing early clinical studies in ovarian cancer patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Doxorubicin/analogs & derivatives , Endometrial Neoplasms/drug therapy , Gonadotropin-Releasing Hormone/analogs & derivatives , Molecular Targeted Therapy/methods , Ovarian Neoplasms/drug therapy , Animals , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Dogs , Doxorubicin/therapeutic use , Female , Gonadotropin-Releasing Hormone/therapeutic use , Humans , Mice , Precision Medicine/methods , Rats
7.
Behav Brain Res ; 228(2): 388-91, 2012 Mar 17.
Article in English | MEDLINE | ID: mdl-22197299

ABSTRACT

MZ-4-71 is an antagonist of growth hormone-releasing hormone (GH-RH) which suppresses the secretion of GH-RH. It has been shown that MZ-4-71 has antidepressive-like effects in a modified forced swimming test (FST) in mice, exerts anxiolytic effects in an elevated plus maze test, improves memory consolidation in passive avoidance learning, and corrects the impairment of memory consolidation caused by ß-amyloid 25-35 in mice. However, little is known about the mechanisms of action of MZ-4-71 on brain functions. The involvement of the adrenergic, serotonergic, cholinergic, dopaminergic or GABA-ergic receptors in the antidepressant-like action of MZ-4-71 (1.0 µg/2 µl, intracerebroventricular (i.c.v.)) was studied in a modified mouse forced swimming test (FST). Mice were pretreated with a non-selective α-adrenergic receptor antagonist, phenoxybenzamine, an α1/α2ß-adrenergic receptor antagonist prazosin, an α2-adrenergic receptor antagonist, yohimbine, a ß-adrenergic receptor antagonist, propranolol, a mixed 5-HT1/5-HT2 serotonergic receptor antagonist methysergide, a non-selective 5-HT2 serotonergic receptor antagonist, cyproheptadine, a non-selective muscarinic acetylcholine receptor antagonist, atropine, a D2, D3, D4 dopamine receptor antagonist, haloperidol or a γ-aminobutyric acid subunit A (GABA-A) receptor antagonist bicuculline. Phenoxybenzamine, prazosin, methysergide, cyproheptadine and atropine prevented the effects of MZ-4-71 on the immobility, the climbing and the swimming times. Yohimbine, propranolol, haloperidol and bicuculline did not change the effects of MZ-4-71. The results demonstrated that the antidepressant-like effects of MZ-4-71 in this modified mouse FST are mediated, at least in part, by the an interaction of the α1-adrenergic, 5-HT1/5-HT2 serotonergic, and muscarinic acetylcholine receptors.


Subject(s)
Antidepressive Agents/therapeutic use , Depression/drug therapy , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Neurotransmitter Agents/metabolism , Sermorelin/analogs & derivatives , Analysis of Variance , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Immobility Response, Tonic/drug effects , Injections, Intraventricular , Male , Mice , Muscle Strength/drug effects , Neurotransmitter Agents/therapeutic use , Sermorelin/therapeutic use , Swimming/psychology
8.
Horm Metab Res ; 43(11): 816-20, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22009378

ABSTRACT

Epithelial ovarian carcinoma is the leading cause of cancer-related deaths among women with gynecologic malignancies. Antagonists of the growth hormone-releasing hormone (GHRH) have been shown to inhibit growth of various cancers through endocrine, autocrine, and paracrine mechanisms. In this study, we have investigated the effects of GHRH antagonists (GHRHa) in ES-2 human clear cell ovarian cancer and in UCI-107 human serous ovarian cancer in vitro and in vivo. We evaluated the expression of mRNA for GHRH receptor, the binding to GHRH receptors, in specimens of ES-2 ovarian cancer. We evaluated also the in vitro effects of GHRHa on ES-2 cells and the in vivo effect of 2 different GHRHa on ES-2 and UCI-107 tumors. Nude mice bearing xenografts on ES-2 and UCI-107 ovarian cancer were treated with JMR-132 and MZ-J-7-118, respectively. Tumor growth was compared to control. ES-2 cells expressed mRNA for the functional splice variant SV1 of the GHRH receptor. JMR-132 inhibited cell proliferation in vitro by 42% and 18% at 10 and 1 µM concentration, respectively. Specific high affinity receptors for GHRH were detected in ES-2 cancer samples. In vivo daily subcutaneous injections of GHRHa significantly reduced tumor growth compared to a control group in both animal models. Our results indicate that GHRHa such as JMR-132 and MZ-J-7-118 can inhibit the growth of human ovarian cancer. The efficacy of GHRHa in ovarian cancer should be assessed in clinical trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/therapeutic use , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Hormone Antagonists/metabolism , Hormone Antagonists/pharmacology , Humans , Mice , Mice, Nude , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Random Allocation , Sermorelin/analogs & derivatives , Sermorelin/pharmacology , Sermorelin/therapeutic use , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
Horm Metab Res ; 42(11): 781-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20665426

ABSTRACT

Glioblastoma multiforme is the most common and most aggressive type of high grade tumor with a poor prognosis upon discovery. Based on earlier promising results earned with AN-162, a doxorubicin molecule linked to somatostatin (SST) analogue RC-160, it was our aim to determine the effect of AN-162 on DBTRG-05 glioblastoma cell line, and to test its efficacy in experimental brain tumors. We detected the expression of mRNA for somatostatin receptor (SSTR) subtypes 2 and 3 in DBTRG-05 cells with RT-PCR. Using ligand competition assay, specific high affinity receptors for somatostatin were found. The MTT assay showed that both AN-162 and doxorubicin (DOX) significantly inhibited cell proliferation and that there was no significant difference between the effects in vitro. Nude mice were xenografted with DBTRG-05 glioblastoma tumors. AN-162 showed a significant inhibition of tumor growth compared with the control group and the groups treated with equimolar doses of doxorubicin, somatostatin analogue RC-160, or the unconjugated mixture of doxorubicin plus RC-160. The tumor doubling time in the group of animals treated with AN-162 was extended and was significantly different from doubling times in the control group and in the other treatment groups. Our study clearly demonstrates a potent inhibitory effect of AN-162 in experimental glioblastoma, thus suggesting the possibility of its utilization in patients suffering from malignant brain cancer.


Subject(s)
2-Hydroxyphenethylamine/analogs & derivatives , Aniline Compounds/pharmacology , Glioblastoma/pathology , Somatostatin/analogs & derivatives , 2-Hydroxyphenethylamine/pharmacology , 2-Hydroxyphenethylamine/therapeutic use , Aniline Compounds/therapeutic use , Animals , Biological Assay , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/chemically induced , Glioblastoma/drug therapy , Glioblastoma/genetics , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Reverse Transcriptase Polymerase Chain Reaction
10.
Regul Pept ; 159(1-3): 142-7, 2010 Jan 08.
Article in English | MEDLINE | ID: mdl-19706309

ABSTRACT

The decapeptide LHRH antagonist, Cetrorelix, inhibits gonadotropin and sex-steroid secretion. Cetrorelix is used for IVF-ET procedures and for the treatment of benign prostatic hyperplasia, endometriosis and leiomyomas. However little is known about the effects of Cetrorelix on brain functions. Previously we have tested Cetrorelix in mice on the impairment of the consolidation of a passive avoidance behavior caused by beta-amyloid 25-35, anxiolytic action in the plus-maze, antidepressive action in a forced swimming test, tail suspension and open-field behavior following its administration into the lateral brain ventricle. In the present study we repeated and extended the experiments in rats in order to determine whether there are species differences in the action of Cetrorelix between mice and rats. The effects of Cetrorelix evaluated included the methods used in mice without tail suspension test and extended by measuring core temperature. Cetrorelix fully blocked the impairment of the consolidation of passive avoidance learning when given icv 30 min following administration of beta-amyloid 25-35. If beta-amyloid 25-35 and Cetrorelix were given simultaneously, Cetrorelix was ineffective. Cetrorelix elicited slight anxiogenic and stronger anxiolytic action in the plus-maze, depending on the dose used. In the forced swimming tests, Cetrorelix showed antidepressive-like action. In open-field behavior tests Cetrorelix displayed a U-type action on locomotion with 0.5 and 2 microg increasing locomotion, and increase rearing but and had no effect on grooming at 0.5-2 microg. Cetrorelix had no action on core temperature. Our findings demonstrate that Cetrorelix is able to correct the impairment of the memory consolidation caused by beta-amyloid 25-35. Cetrorelix elicits anxiolytic and antidepressive action, slightly increases locomotion and rearing in open field, but it does not influence the core temperature. The results obtained in rats are similar to those reported previously by us in mice. Collectively our findings confirm the effects of Cetrorelix on brain function in two species and suggest the possible merit of a clinical trial with Cetrorelix in patients with anxiety, depression and Alzheimer's disease.


Subject(s)
Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/pharmacology , Maze Learning/drug effects , Animals , Dose-Response Relationship, Drug , Gonadotropin-Releasing Hormone/pharmacology , Male , Mice , Rats , Rats, Wistar
11.
Proc Natl Acad Sci U S A ; 106(37): 15879-84, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19717419

ABSTRACT

Peptide analogues targeting various neuropeptide receptors have been used effectively in cancer therapy. A hallmark of adrenocortical tumor formation is the aberrant expression of peptide receptors relating to uncontrolled cell proliferation and hormone overproduction. Our microarray results have also demonstrated a differential expression of neuropeptide hormone receptors in tumor subtypes of human pheochromocytoma. In light of these findings, we performed a comprehensive analysis of relevant receptors in both human adrenomedullary and adrenocortical tumors and tested the antiproliferative effects of peptide analogues targeting these receptors. Specifically, we examined the receptor expression of somatostatin-type-2 receptor, growth hormone-releasing hormone (GHRH) receptor or GHRH receptor splice variant-1 (SV-1) and luteinizing hormone-releasing hormone (LHRH) receptor at the mRNA and protein levels in normal human adrenal tissues, adrenocortical and adrenomedullary tumors, and cell lines. Cytotoxic derivatives of somatostatin AN-238 and, to a lesser extent, AN-162, reduced cell numbers of uninduced and NGF-induced adrenomedullary pheochromocytoma cells and adrenocortical cancer cells. Both the splice variant of GHRH receptor SV-1 and the LHRH receptor were also expressed in adrenocortical cancer cell lines but not in the pheochromocytoma cell line. The GHRH receptor antagonist MZ-4-71 and LHRH antagonist Cetrorelix both significantly reduced cell growth in the adrenocortical cancer cell line. In conclusion, the expression of receptors for somatostatin, GHRH, and LHRH in the normal human adrenal and in adrenal tumors, combined with the growth-inhibitory effects of the antitumor peptide analogues, may make possible improved treatment approaches to adrenal tumors.


Subject(s)
Adrenal Gland Neoplasms/drug therapy , Adrenal Gland Neoplasms/metabolism , Neuropeptides/pharmacology , Receptors, Neuropeptide/metabolism , 2-Hydroxyphenethylamine/analogs & derivatives , 2-Hydroxyphenethylamine/pharmacology , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/pathology , Adrenal Glands/metabolism , Aniline Compounds/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cytostatic Agents/pharmacology , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Gene Expression , Humans , Oligonucleotide Array Sequence Analysis , PC12 Cells , Pyrroles/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Receptors, Neuropeptide/genetics , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Somatostatin/analogs & derivatives , Somatostatin/pharmacology
12.
Br J Cancer ; 98(11): 1790-6, 2008 Jun 03.
Article in English | MEDLINE | ID: mdl-18506184

ABSTRACT

Splice Variant 1 (SV-1) of growth hormone-releasing hormone (GHRH) receptor, found in a wide range of human cancers and established human cancer cell lines, is a functional receptor with ligand-dependent and independent activity. In the present study, we demonstrated by western blots the presence of the SV1 of GHRH receptor and the production of GHRH in MDA-MB-468, MDA-MB-435S and T47D human breast cancer cell lines, LNCaP prostate cancer cell line as well as in NCI H838 non-small cell lung carcinoma. We have also shown that GHRH produced in the conditioned media of these cell lines is biologically active. We then inhibited the intrinsic production of GHRH in these cancer cell lines using si-RNA, specially designed for human GHRH. The knocking down of the GHRH gene expression suppressed the proliferation of T47D, MDA-MB-435S, MDA-MB-468 breast cancer, LNCaP prostate cancer and NCI H838 non-SCLC cell lines in vitro. However, the replacement of the knocked down GHRH expression by exogenous GHRH (1-29)NH(2) re-established the proliferation of the silenced cancer cell lines. Furthermore, the proliferation rate of untransfected cancer cell lines could be stimulated by GHRH (1-29)NH(2) and inhibited by GHRH antagonists MZ-5-156, MZ-4-71 and JMR-132. These results extend previous findings on the critical function of GHRH in tumorigenesis and support the role of GHRH as a tumour growth factor.


Subject(s)
Growth Hormone-Releasing Hormone/antagonists & inhibitors , Neoplasms/therapy , Cell Line, Tumor , Cell Proliferation , Female , Growth Hormone-Releasing Hormone/analysis , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/physiology , Humans , Male , Neoplasms/pathology , RNA, Small Interfering/genetics , Radioimmunoassay , Sermorelin/analogs & derivatives , Sermorelin/pharmacology
14.
Horm Metab Res ; 40(5): 315-22, 2008 May.
Article in English | MEDLINE | ID: mdl-18491250

ABSTRACT

The discovery of hypothalamic hormones was briefly reviewed. The development of new hormonal methods for the therapy of various cancers based on analogues of hypothalmic hormones is then presented. My group isolated luteininzing hormone-releasing hormone (LH-RH), also known as Gn-RH, from pig hypothalmi, elucidated its amino acid sequence, and synthesized it in 1971. The interest in medical applications of LH-RH led to the synthesis of LH-RH analogues by various groups. LH-RH agonists substituted in positions 6 or 10 including Decapeptyl, Leuprolide and Zoladex are much more active than LH-RH and on continuous administration produce inhibition of pituitary and gonads. Chronic administration of LH-RH agonists is being utilized for the treatment of prostate and breast cancer. Octapeptide analogues of somatostatin have various applications in Oncology. In 1980 we developed a new endocrine therapy for advanced prostate cancer based on agonists of LH-RH, which is now preferred by 70-90% of prostate cancer patients for primary treatment. LH-RH antagonists such as Cetrorelix can be used for therapy of BPH. On the basis of the presence of specific receptors for hypothalamic peptides on human cancers, we developed targeted cytotoxic analogues of LH-RH, somatostatin, and bombesin/GRP linked to doxorubicin or 2-pyrrolinodoxorubicin. These analogues inhibit the growth of experimental human prostate, breast, ovarian and endometrial cancer, renal cell carcinoma, pancreatic, colorectal and gastric cancers, small cell lung carcinoma (SCLC) and non-SCLC, brain tumors, melanomas, and lymphomas. Cytotoxic LH-RH analogues are now in clinical trials. Recently we demonstrated that growth hormone-releasing hormone (GH-RH) also serves as an autocrine growth factor in many cancers. Antagonistic analogues of GH-RH synthesized in our laboratory inhibit the growth of diverse tumors. The discovery of LH-RH and somatostatin has led to clinical use of their analogues in the field of cancer treatment and GH-RH antagonists also show a great promise.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Peptide Hormones/therapeutic use , Peptides/therapeutic use , Animals , Female , Humans , Male , Swine
15.
Regul Pept ; 136(1-3): 105-8, 2006 Sep 11.
Article in English | MEDLINE | ID: mdl-16781787

ABSTRACT

Growth hormone-releasing hormone (GHRH) stimulates the production and release of growth hormone in the pituitary and induces cell proliferation in a variety of peripheral tissues and tumors. These extrapituitary effects of GHRH are in many cases mediated by a splice variant of GHRH receptor designated SV1 that differs from the pituitary GHRH receptor in a small portion of its amino-terminal region. While SV1 has been detected in several primary tumors and many cancer cell lines its expression in normal tissues remains unclear. In this study we report the results of an immunohistochemical analysis for SV1 and GHRH expression in normal mouse tissues. For the detection of SV1 immunoreactivity we used a polyclonal antiserum against segments 1-25 of the SV1 receptor protein. Mouse heart, colon, lungs, small intestine, stomach and kidneys exhibited increased SV1 immunoreactivity. These tissues were also positive for GHRH expression, however, tissues such as the endometrium were positive only for GHRH and not for SV1 expression. On the contrary, testis were positive for SV1 and not for GHRH expression. These results indicate that SV1 may play a role in normal physiology.


Subject(s)
Alternative Splicing , Gene Expression Regulation , Growth Hormone-Releasing Hormone/biosynthesis , Receptors, Neuropeptide/biosynthesis , Receptors, Neuropeptide/genetics , Receptors, Pituitary Hormone-Regulating Hormone/biosynthesis , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Animals , Female , Immunohistochemistry , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , RNA, Messenger/metabolism , Tissue Distribution
16.
Verh Dtsch Ges Pathol ; 90: 186-92, 2006.
Article in German | MEDLINE | ID: mdl-17867596

ABSTRACT

AIMS: Cytotoxic luteinizing hormone releasing hormone (LHRH) analogues AN-152 and AN-207 consist of [D-Lys6] LHRH linked to doxorubicin or its hyperactive derivate AN-201 and bind with high affinity to LHRH receptors. We evaluated the use of AN-207 and AN-201 in a nude mice model. In order to provide a rationale for the possible use of cytotoxic LHRH analogues in different malignancies we investigated the expression of LHRH-R in human renal cell carcinoma (RCC), melanoma and non Hodgkin's Lymphoma (NHL). METHODS: The expression of LHRH-R was examined in surgically removed human specimens of primary tumours and metastases from 37 RCC, 19 melanomas and 17 NHLs. In addition, human tumour cell lines expressing LHRH receptors were transplanted into nude mice and anti-tumour efficacy and systemic toxicity of AN-207 and its cytotoxic radical AN-201 were compared in various experiments. RESULTS: Positive staining for LHRH receptors was found in all of the RCC (37/37) and the melanoma specimens (19/19) as well as in 100% (10/10) of the NHLs. In in vivo experiments AN-207 significantly inhibited tumour growth while the cytotoxic radical alone was ineffective. Furthermore, side effects were reduced with targeted therapy. CONCLUSIONS: LHRH receptor expression was found to be very high in melanomas, RCCs and NHLs. Therefore targeted therapy with cytotoxic LHRH analogues may be a promising, novel therapy for advanced stages of these tumours. A first clinical trial with AN-152 was initiated recently in breast cancer patients.


Subject(s)
Kidney Neoplasms/pathology , Lymphoma, Non-Hodgkin/pathology , Melanoma/pathology , Animals , Antineoplastic Agents/therapeutic use , Disease Models, Animal , Doxorubicin/therapeutic use , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/genetics , Melanoma/drug therapy , Melanoma/genetics , Neoplasm Metastasis , Receptors, LHRH/analysis , Receptors, LHRH/antagonists & inhibitors
17.
J Urol ; 173(6): 2154-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15879878

ABSTRACT

PURPOSE: Gastrin releasing peptide (GRP) is a growth factor for renal cell carcinoma (RCC) and it has vasoactive properties. Blockade of GRP receptor inhibits the growth of GRP receptor positive and negative tumors in nude mice, suggesting GRP effects other than those related to tumor epithelium. Therefore, in this study we analyzed the effects of GRP receptor blockade on neoangiogenesis in RCC. MATERIALS AND METHODS: GRP receptor expression was determined in human RCC and corresponding normal tissue by real-time reverse transcriptase-polymerase chain reaction, immunohistochemistry and confocal laser scanning microscopy. Multicellular spheroids of the A498 RCC line were implanted into dorsal skin fold chambers of athymic nude mice. Neoangiogenesis was measured by intravital microscopy after blockade of GRP receptors by the GRP antagonist RC-3095. The influence of GRP on vascular endothelial growth factor secretion in A498 cells was studied in vitro. RESULTS: GRP receptor expression was immunolocalized in tumor cells and microvessels. Implanted tumor cell spheroids and spheroid microvessels of the chamber also expressed GRP receptors. Spheroid neoangiogenesis was significantly inhibited by RC-3095 when given immediately after spheroid implantation. Vascular endothelial growth factor secretion of A498 cells was not affected by GRP. CONCLUSIONS: RCC angiogenesis is sensitive to GRP receptor blockade. Therefore, GRP receptors may not only stimulate tumor cell proliferation, but also affect tumor microcirculation.


Subject(s)
Carcinoma, Renal Cell/blood supply , Kidney Neoplasms/blood supply , Neovascularization, Pathologic/genetics , Receptors, Bombesin/genetics , Animals , Carcinoma, Renal Cell/pathology , Cell Division/genetics , Cell Line, Tumor , Endothelium, Vascular/pathology , Gene Expression Regulation, Neoplastic/physiology , Humans , Kidney Neoplasms/pathology , Male , Mice , Mice, Nude , Microcirculation/pathology , Microscopy, Confocal , Neoplasm Staging , Neoplasm Transplantation , Polymerase Chain Reaction
18.
Curr Pharm Des ; 11(9): 1167-80, 2005.
Article in English | MEDLINE | ID: mdl-15853664

ABSTRACT

Chemotherapy is one of the main modalities in the therapy of cancer. However, an improvement in the efficacy and a reduction in the toxicity of chemotherapeutic agents remains a great challenge to oncologists. A specific delivery of cytotoxic drugs to cancerous cells may help improving both aspects. Peptide hormones, for which receptors have been found in various human cancers, can serve as carriers for a local delivery of cytotoxic agents or radiopharmaceuticals to the tumors, as demonstrated by the successful clinical use of radiolabeled somatostatin analog Octreoscan for the detection and treatment of some somatostatin receptor-positive tumors. Thus, in recent years we developed a series of cytotoxic peptide hormone conjugates based on derivatives of hypothalamic hormones such as somatostatin and luteinizing hormone-releasing hormone (LHRH), and the brain-gut hormone bombesin. To create targeted conjugates with high cytotoxic activity, a derivative of doxorubicin (DOX), 2-pyrrolino-DOX (AN-201), which is 500-1, 000 times more active than its parent compound, was developed. This agent was coupled to somatostatin octapeptide RC-121 to form cytotoxic conjugate AN-238, and to [D-Lys6]LHRH carrier to produce analog AN-207. Cytotoxic bombesin hybrid AN-215 also contains AN-201. DOX was likewise linked to [D-Lys6]LHRH to form AN-152. A comprehensive testing of these cytotoxic conjugates in experimental models of various human and rodent cancers led to their selection as candidates for clinical trials.


Subject(s)
Bombesin/toxicity , Drug Delivery Systems/methods , Gonadotropin-Releasing Hormone/toxicity , Neoplasms/drug therapy , Neoplasms/metabolism , Receptors, Drug/metabolism , Somatostatin/toxicity , Animals , Bombesin/administration & dosage , Bombesin/analogs & derivatives , Bombesin/metabolism , Gonadotropin-Releasing Hormone/administration & dosage , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/metabolism , Humans , Neoplasms/pathology , Receptors, Bombesin/metabolism , Receptors, LHRH/metabolism , Receptors, Somatostatin/metabolism , Somatostatin/administration & dosage , Somatostatin/analogs & derivatives , Somatostatin/metabolism
19.
Cell Mol Life Sci ; 61(9): 1042-68, 2004 May.
Article in English | MEDLINE | ID: mdl-15112052

ABSTRACT

Cancers of the stomach, colon and exocrine pancreas are major international health problems and result in more than a million deaths worldwide each year. The therapies for these malignancies must be improved. The effects of gastrointestinal (GI) hormonal peptides and endogenous growth factors on these cancers were reviewed. Some GI peptides, including gastrin and gastrin-releasing peptide (GRP) (mammalian bombesin), appear to be involved in the growth of neoplasms of the GI tract. Certain growth factors such as insulin-like growth factor (IGF)-I, IGF-II and epidermal growth factor and their receptors that regulate cell proliferation are also implicated in the development and progression of GI cancers. Experimental investigations on gastric, colorectal and pancreatic cancers with analogs of somatostatin, antagonists of bombesin/GRP, antagonists of growth hormone-releasing hormone as well as cytotoxic peptides that can be targeted to peptide receptors on tumors were summarized. Clinical trials on peptide analogs in patients with gastric, colorectal and pancreatic cancers were reviewed and analyzed. It may be possible to develop new approaches to hormonal therapy of GI malignancies based on various peptide analogs.


Subject(s)
Colonic Neoplasms/drug therapy , Oligopeptides/pharmacology , Pancreatic Neoplasms/drug therapy , Stomach Neoplasms/drug therapy , Animals , Bombesin/analogs & derivatives , Bombesin/pharmacology , Humans , Somatostatin/analogs & derivatives , Somatostatin/pharmacology
20.
Eur J Cancer ; 40(3): 436-44, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14746863

ABSTRACT

In the present study, we investigated whether the growth hormone-releasing hormone (GH-RH) antagonist JV-1-38 could enhance the effects of androgen deprivation produced by the anti-androgen Flutamide and luteinising hormone-releasing hormone (LH-RH) agonist Decapeptyl in an experimental model of human androgen-sensitive MDA PCa 2b prostate carcinoma implanted subcutaneously (s.c.) into nude mice. We also evaluated by reverse transcriptase-polymerase chain reaction (RT-PCR) the effects of combined treatment on the mRNA expression for prostate-specific antigen (PSA) and measured serum PSA levels. In experiment 1, GH-RH antagonist JV-1-38 greatly inhibited tumour growth in combination with Decapeptyl, but was ineffective when given alone. Thus, combined therapy with JV-1-38 at 20 microg/day and Decapeptyl microcapsules releasing 12.5 microg/day for 29 days inhibited significantly (P<0.01) MDA PCa 2b tumour growth by 65%, compared with controls. Combined treatment also significantly (P<0.05) decreased serum PSA levels by 52% and reduced tumour weight by 54% vs. controls. In experiment 2, GH-RH antagonist JV-1-38 at 20 microg/day likewise showed powerful growth inhibitory effects when combined with Flutamide (25 mg/kg/day) for 21 days. Combined treatment with JV-1-38 and slow-release pellets of Flutamide significantly (P<0.001) inhibited tumour growth by 61% versus controls, and was significantly (P<0.05) more effective than Flutamide or JV-1-38 alone. Combination therapy also reduced significantly (P<0.001) tumour weight and serum PSA levels by 59 and 47%, respectively. The mRNA expression for PSA in MDA PCa 2b tumours was not changed by JV-1-38, Decapeptyl and Flutamide alone or in their respective combinations. Our findings suggest that GH-RH antagonists could enhance the tumour inhibitory effects of androgen deprivation for the primary therapy of patients with advanced prostate carcinoma.


Subject(s)
Androgen Antagonists/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Flutamide/therapeutic use , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Growth Hormone-Releasing Hormone/therapeutic use , Prostatic Neoplasms/drug therapy , Triptorelin Pamoate/therapeutic use , Animals , Cell Division , Cell Line, Tumor , Drug Interactions , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/pathology , Random Allocation
SELECTION OF CITATIONS
SEARCH DETAIL
...