1
|
Gesmundo I, Pedrolli F, Cai R, Sha W, Schally AV, Granata R. Growth hormone-releasing hormone and cancer. Rev Endocr Metab Disord 2024:10.1007/s11154-024-09919-4. [PMID: 39422787 DOI: 10.1007/s11154-024-09919-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
The hypothalamic hormone growth hormone-releasing hormone (GHRH), in addition to promoting the synthesis and release of growth hormone (GH), stimulates the proliferation of human normal and malignant cells by binding to GHRH-receptor (GHRH-R) and its main splice variant, SV1. Both GHRH and GHRH-Rs are expressed in various cancers, forming a stimulatory pathway for cancer cell growth; additionally, SV1 possesses ligand independent proliferative effects. Therefore, targeting GHRH-Rs pharmacologically has been proposed for the treatment of cancer. Various classes of synthetic GHRH antagonists have been developed, endowed with strong anticancer activity in vitro and in vivo, in addition to displaying anti-inflammatory, antioxidant and immune-modulatory functions. GHRH antagonists exert indirect effects by blocking the pituitary GH/hepatic insulin-like growth factor I (IGF-I) axis, or directly inhibiting the binding of GHRH on tumor GHRH-Rs. Additionally, GHRH antagonists block the mitogenic functions of SV1 in tumor cells. This review illustrates the main findings on the antitumor effects of GHRH antagonists in experimental human cancers, along with their underlying mechanisms. The development of GHRH antagonists, with reduced toxicity and high stability, could lead to novel therapeutic agents for the treatment of cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Iacopo Gesmundo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Renzhi Cai
- Veterans Affairs Medical Center, Endocrine, Polypeptide and Cancer Institute, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Wei Sha
- Veterans Affairs Medical Center, Endocrine, Polypeptide and Cancer Institute, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Pathology, School of Medicine and Sylvester Comprehensive Cancer Center, University of Miami Miller, Miami, FL, USA
| | - Andrew V Schally
- Veterans Affairs Medical Center, Endocrine, Polypeptide and Cancer Institute, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Pathology, School of Medicine and Sylvester Comprehensive Cancer Center, University of Miami Miller, Miami, FL, USA
| | - Riccarda Granata
- Department of Medical Sciences, University of Turin, Turin, Italy.
| |
Collapse
|
2
|
Szabo Z, Juhasz E, Schally AV, Dezso B, Huga S, Hernadi Z, Halmos G, Kiss C. Expression of Growth Hormone-Releasing Hormone and Its Receptor Splice Variants in Primary Human Endometrial Carcinomas: Novel Therapeutic Approaches. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27092671. [PMID: 35566020 PMCID: PMC9101386 DOI: 10.3390/molecules27092671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 11/16/2022]
Abstract
Antagonists of growth hormone-releasing hormone (GHRH) inhibit the growth of various tumors, including endometrial carcinomas (EC). However, tumoral receptors that mediate the antiproliferative effects of GHRH antagonists in human ECs have not been fully characterized. In this study, we investigated the expression of mRNA for GHRH and splice variants (SVs) of GHRH receptors (GHRH-R) in 39 human ECs and in 7 normal endometrial tissue samples using RT-PCR. Primers designed for the PCR amplification of mRNA for the full length GHRH-R and SVs were utilized. The PCR products were sequenced, and their specificity was confirmed. Nine ECs cancers (23%) expressed mRNA for SV1, three (7.7%) showed SV2 and eight (20.5%) revealed mRNA for SV4. The presence of SVs for GHRH-Rs could not be detected in any of the normal endometrial tissue specimens. The presence of specific, high affinity GHRH-Rs was also demonstrated in EC specimens using radioligand binding studies. Twenty-four of the investigated thirty-nine tumor samples (61.5%) and three of the seven corresponding normal endometrial tissues (42.9%) expressed mRNA for GHRH ligand. Our findings suggest the possible existence of an autocrine loop in EC based on GHRH and its tumoral SV receptors. The antiproliferative effects of GHRH antagonists on EC are likely to be exerted in part by the local SVs and GHRH system.
Collapse
Affiliation(s)
- Zsuzsanna Szabo
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (G.H.)
| | - Eva Juhasz
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Andrew V. Schally
- Veterans Affairs Medical Center, Endocrine, Polypeptide and Cancer Institute, Miami, FL 33125, USA;
- Department of Pathology, Department of Medicine, Divisions of Hematology-Oncology and Endocrinology, Miller School of Medicine, University of Miami, Miami, FL 33101, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Balazs Dezso
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Sandor Huga
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (S.H.); (Z.H.)
| | - Zoltan Hernadi
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (S.H.); (Z.H.)
| | - Gabor Halmos
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (G.H.)
- Veterans Affairs Medical Center, Endocrine, Polypeptide and Cancer Institute, Miami, FL 33125, USA;
| | - Csongor Kiss
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
- Correspondence: ; Tel.: +36-52-452-747; Fax: +36-52-255-893
| |
Collapse
|
3
|
Zheng Y, Karnoub AE. Endocrine regulation of cancer stem cell compartments in breast tumors. Mol Cell Endocrinol 2021; 535:111374. [PMID: 34242715 DOI: 10.1016/j.mce.2021.111374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 10/20/2022]
Abstract
Cancer cells within breast tumors exist within a hierarchy in which only a small and rare subset of cells is able to regenerate growths with the heterogeneity of the original tumor. These highly malignant cancer cells, which behave like stem cells for new cancers and are called "cancer stem cells" or CSCs, have also been shown to possess increased resistance to therapeutics, and represent the root cause underlying therapy failures, persistence of residual disease, and relapse. As >90% of cancer deaths are due to refractory tumors, identification of critical molecular drivers of the CSC-state would reveal vulnerabilities that can be leveraged in designing therapeutics that eradicate advanced disease and improve patient survival outcomes. An expanding and complex body of work has now described the exquisite susceptibility of CSC pools to the regulatory influences of local and systemic hormones. Indeed, breast CSCs express a plethora of hormonal receptors, which funnel hormonal influences over every aspect of breast neoplasia - be it tumor onset, growth, survival, invasion, metastasis, or therapy resistance - via directly impacting CSC behavior. This article is intended to shed light on this active area of investigation by attempting to provide a systematic and comprehensive overview of the available evidence directly linking hormones to breast CSC biology.
Collapse
Affiliation(s)
- Yurong Zheng
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Antoine E Karnoub
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Harvard Stem Cell Institute, Cambridge, MA, 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
4
|
Verigos J, Karakaidos P, Kordias D, Papoudou-Bai A, Evangelou Z, Harissis HV, Klinakis A, Magklara A. The Histone Demethylase LSD1/ΚDM1A Mediates Chemoresistance in Breast Cancer via Regulation of a Stem Cell Program. Cancers (Basel) 2019; 11:cancers11101585. [PMID: 31627418 PMCID: PMC6827056 DOI: 10.3390/cancers11101585] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/13/2019] [Accepted: 10/15/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the leading cause of cancer death in the female population, despite advances in diagnosis and treatment. The highly heterogeneous nature of the disease represents a major obstacle to successful therapy and results in a significant number of patients developing drug resistance and, eventually, suffering from tumor relapse. Cancer stem cells (CSCs) are a small subset of tumor cells characterized by self-renewal, increased tumor-initiation capacity, and resistance to conventional therapies. As such, they have been implicated in the etiology of tumor recurrence and have emerged as promising targets for the development of novel therapies. Here, we show that the histone demethylase lysine-specific demethylase 1 (LSD1) plays an important role in the chemoresistance of breast cancer cells. Our data, from a series of in vitro and in vivo assays, advocate for LSD1 being critical in maintaining a pool of tumor-initiating cells that may contribute to the development of drug resistance. Combinatory administration of LSD1 inhibitors and anti-cancer drugs is more efficacious than monotherapy alone in eliminating all tumor cells in a 3D spheroid system. In conclusion, we provide compelling evidence that LSD1 is a key regulator of breast cancer stemness and a potential target for the design of future combination therapies.
Collapse
Affiliation(s)
- John Verigos
- Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology, 45110 Ioannina, Greece.
- Department of Clinical Chemistry, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece.
| | - Panagiotis Karakaidos
- Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology, 45110 Ioannina, Greece.
- Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece.
| | - Dimitris Kordias
- Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology, 45110 Ioannina, Greece.
- Department of Clinical Chemistry, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece.
| | | | - Zoi Evangelou
- Department of Pathology, University Hospital of Ioannina, 45500 Ioannina, Greece.
| | | | | | - Angeliki Magklara
- Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology, 45110 Ioannina, Greece.
- Department of Clinical Chemistry, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece.
| |
Collapse
|
5
|
Kővári B, Vranic S, Marchio C, Sapino A, Cserni G. The expression of GHRH and its receptors in breast carcinomas with apocrine differentiation-further evidence of the presence of a GHRH pathway in these tumors. Hum Pathol 2017; 64:164-170. [PMID: 28438614 DOI: 10.1016/j.humpath.2017.03.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/14/2017] [Accepted: 03/29/2017] [Indexed: 02/07/2023]
Abstract
Apocrine breast carcinomas were evaluated for the expression of components of the growth hormone-releasing hormone (GHRH) autocrine/paracrine pathway: GHRH and its receptors (GHRH-R), as mammary apocrine carcinomas and epithelium seemed to be uniformly positive for GHRH-R in a pilot study. The apocrine phenotype was determined on the basis of hematoxylin-eosin morphology and a congruent immunohistochemical profile (estrogen receptor negativity, androgen receptor and gross cystic disease fluid protein-15 positivity). Thirty-five formalin-fixed, paraffin-embedded apocrine breast cancers in tissue microarrays and 24 cases using whole-tissue sections were evaluated for GHRH-R and GHRH expression by immunohistochemistry using polyclonal antibodies raised against various domains of GHRH-R and one polyclonal antibody specific for GHRH. GHRH-R positivity was detected in the overwhelming majority (ranging from 90% to 100%) of apocrine breast carcinomas with all but one of the antibodies applied. The expression was usually diffuse with only isolated cases showing positivity in less than 50% of tumor cells. With the PA5-33583 antibody, GHRH-R positivity was seen only in 73% of the cases in at least 50% of the tumor cells. GHRH expression was also present in all but one case tested, with more than 50% of the cells expressing it in 30/34 cases. These results support a high rate of GHRH-R and GHRH expression in apocrine breast carcinomas. Whether these findings can be exploited for the targeted treatment of apocrine breast carcinomas with GHRH antagonists requires further study.
Collapse
Affiliation(s)
- Bence Kővári
- Department of Pathology, University of Szeged, 6720 Szeged, Hungary.
| | - Semir Vranic
- Department of Pathology, Clinical Center of the University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina; School of Medicine, University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina
| | - Caterina Marchio
- Department of Medical Sciences, University of Turin, Pathology Unit, 10126 Turin, Italy
| | - Anna Sapino
- Department of Medical Sciences, University of Turin, Pathology Unit, 10126 Turin, Italy; Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO), IRCCS, 10060, Candiolo (To), Italy
| | - Gábor Cserni
- Department of Pathology, University of Szeged, 6720 Szeged, Hungary; Department of Pathology, Bács-Kiskun County Teaching Hospital, 6000 Kecskemét, Hungary
| |
Collapse
|
6
|
Moscona JC, Peters MN, Schally AV, Srivastav S, Delafontaine P, Irimpen A. The effects of a growth hormone-releasing hormone antagonist and a gastrin-releasing peptide antagonist on intimal hyperplasia of the carotid artery after balloon injury in a diabetic rat model☆. Artery Res 2017. [DOI: 10.1016/j.artres.2017.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
7
|
Wootten D, Miller LJ, Koole C, Christopoulos A, Sexton PM. Allostery and Biased Agonism at Class B G Protein-Coupled Receptors. Chem Rev 2016; 117:111-138. [PMID: 27040440 DOI: 10.1021/acs.chemrev.6b00049] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Class B G protein-coupled receptors (GPCRs) respond to paracrine or endocrine peptide hormones involved in control of bone homeostasis, glucose regulation, satiety, and gastro-intestinal function, as well as pain transmission. These receptors are targets for existing drugs that treat osteoporosis, hypercalcaemia, Paget's disease, type II diabetes, and obesity and are being actively pursued as targets for numerous other diseases. Exploitation of class B receptors has been limited by difficulties with small molecule drug discovery and development and an under appreciation of factors governing optimal therapeutic efficacy. Recently, there has been increasing awareness of novel attributes of GPCR function that offer new opportunity for drug development. These include the presence of allosteric binding sites on the receptor that can be exploited as drug binding pockets and the ability of individual drugs to enrich subpopulations of receptor conformations to selectively control signaling, a phenomenon termed biased agonism. In this review, current knowledge of biased signaling and small molecule allostery within class B GPCRs is discussed, highlighting areas that have progressed significantly over the past decade, in addition to those that remain largely unexplored with respect to these phenomena.
Collapse
Affiliation(s)
- Denise Wootten
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville 3052, Victoria, Australia
| | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic , Scottsdale, Arizona 85259, United States
| | - Cassandra Koole
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville 3052, Victoria, Australia.,Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University , New York, New York 10065, United States
| | - Arthur Christopoulos
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville 3052, Victoria, Australia
| | - Patrick M Sexton
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville 3052, Victoria, Australia
| |
Collapse
|
8
|
Schally AV, Block NL, Rick FG. New therapies for relapsed castration-resistant prostate cancer based on peptide analogs of hypothalamic hormones. Asian J Androl 2015; 17:925-8. [PMID: 26112478 PMCID: PMC4814950 DOI: 10.4103/1008-682x.152819] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
It is a pleasure to contribute our presentation at the International Prostate Forum of the Annual Meeting of the American Urological Association (AUA) to this special issue of the Asian Journal of Andrology.
Collapse
Affiliation(s)
- Andrew V Schally
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL 33125, USA
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, Division of Hematology/Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, Division of Endocrinology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Norman L Block
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL 33125, USA
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, Division of Hematology/Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ferenc G Rick
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL 33125, USA
- Department of Urology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33174, USA
| |
Collapse
|
9
|
Kővári B, Rusz O, Schally AV, Kahán Z, Cserni G. Differential immunostaining of various types of breast carcinomas for growth hormone-releasing hormone receptor - Apocrine epithelium and carcinomas emerging as uniformly positive. APMIS 2014; 122:824-31. [PMID: 24479854 DOI: 10.1111/apm.12224] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 11/04/2013] [Indexed: 12/14/2022]
Abstract
Different classes of breast cancers were explored for their positivity for growth hormone-releasing hormone receptors (GHRH-R) in this pilot study, as no systematic evaluation of such tumors has been performed to date. Seventy-two small primary breast carcinomas were evaluated for GHRH-R expression by immunohistochemistry using a polyclonal antibody and a cutoff value of 10% staining. GHRH-R positivity was detected in 58% of all cases, 20/23 (87%) of invasive lobular carcinomas (ILC) and 22/46 (48%) of invasive ductal carcinomas (IDC). GHRH-R positivity was more frequent in grade 2 tumors (86%), as compared to grade 1 (18%) or grade 3 (47%) cancers. GHRH-R expression was not associated with mitotic scores, the Ki-67 labeling indices or nodal status. IDCs with casting-type calcifications on the mammogram showed positivity for GHRH-R in 9/12 (75%) cases. Most importantly, apocrine epithelium, and all 10 apocrine carcinomas added later to the study were GHRH-R-positive. These preliminary results suggest a greater than average GHRH-R expression in ILCs and IDCs associated with casting-type calcifications on the mammogram. Apocrine carcinomas seem uniformly positive for GHRH-R. Whether these findings could indicate a potential role of GHRH-antagonists in targeted treatment of these types of breast cancer requires further studies.
Collapse
Affiliation(s)
- Bence Kővári
- Department of Pathology, University of Szeged, Szeged, Hungary
| | | | | | | | | |
Collapse
|
10
|
Possible predictors of histopathological response to neoadjuvant chemoradiotherapy for rectal cancer. J Cancer Res Clin Oncol 2011; 138:387-95. [DOI: 10.1007/s00432-011-1110-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 11/24/2011] [Indexed: 01/04/2023]
|
11
|
Samineni S, Glackin C, Shively JE. Role of CEACAM1, ECM, and mesenchymal stem cells in an orthotopic model of human breast cancer. Int J Breast Cancer 2010; 2011:381080. [PMID: 22332010 PMCID: PMC3276115 DOI: 10.4061/2011/381080] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Accepted: 09/06/2010] [Indexed: 12/29/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule-1 (CEACAM1) is a morphogen in an in vitro model for lumen formation and plays a similar role in breast epithelial cells implanted in humanized mammary fat pads in NOD-SCID mice. Although extra cellular matrix alone is sufficient to stimulate lumen formation in CEACAM1 transfected MCF-7 cells grown in 3D culture, there is an additional requirement for stromal or mesenchymal cells (MSCs) for these cells to form xenografts with glandular structures in an orthotopic site. We demonstrate that optimal in vitro conditions include both Matrigel and MSCs and that the inclusion of collagen I inhibits xenograft differentiation. Additionally, there is no need to remove the nascent murine mammary gland. The previously observed difference in gland development between the long and short cytoplasmic domain isoforms of CEACAM1 is no longer observed in pregnant NOD/SCID mice suggesting that stimulation of the mammary fat pad by pregnancy critically affects xenograft differentiation.
Collapse
Affiliation(s)
- Sridhar Samineni
- Irell & Manella City of Hope Graduate School of Biological Sciences, Duarte, CA 91010, USA
| | | | | |
Collapse
|
12
|
Guo J, Schally AV, Zarandi M, Varga J, Leung PCK. Antiproliferative effect of growth hormone-releasing hormone (GHRH) antagonist on ovarian cancer cells through the EGFR-Akt pathway. Reprod Biol Endocrinol 2010; 8:54. [PMID: 20509930 PMCID: PMC2891788 DOI: 10.1186/1477-7827-8-54] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 05/28/2010] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Antagonists of growth hormone-releasing hormone (GHRH) are being developed for the treatment of various human cancers. METHODS MTT assay was used to test the proliferation of SKOV3 and CaOV3. The splice variant expression of GHRH receptors was examined by RT-PCR. The expression of protein in signal pathway was examined by Western blotting. siRNA was used to block the effect of EGFR. RESULTS In this study, we investigated the effects of a new GHRH antagonist JMR-132, in ovarian cancer cell lines SKOV3 and CaOV3 expressing splice variant (SV)1 of GHRH receptors. MTT assay showed that JMR-132 had strong antiproliferative effects on SKOV3 and CaOV3 cells in both a time-dependent and dose-dependent fashion. JMR-132 also induced the activation and increased cleaved caspase3 in a time- and dose-dependent manner in both cell lines. In addition, JMR-132 treatments decreased significantly the epidermal growth factor receptor (EGFR) level and the phosphorylation of Akt (p-Akt), suggesting that JMR-132 inhibits the EGFR-Akt pathway in ovarian cancer cells. More importantly, treatment of SKOV3 and CaOV3 cells with 100 nM JMR-132 attenuated proliferation and the antiapoptotic effect induced by EGF in both cell lines. After the knockdown of the expression of EGFR by siRNA, the antiproliferative effect of JMR-132 was abolished in SKOV3 and CaOV3 cells. CONCLUSIONS The present study demonstrates that the inhibitory effect of the GHRH antagonist JMR-132 on proliferation is due, in part, to an interference with the EGFR-Akt pathway in ovarian cancer cells.
Collapse
Affiliation(s)
- Jian Guo
- Department of Obstetrics & Gynaecology, Child and Family Research Institute, UBC, Vancouver, Canada
- School of Preclinical Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Andrew V Schally
- Veterans Affairs Medical Center and Departments of Pathology and Medicine, Division of Hematology/Oncology, University of Miami Miller School of Medicine, Miami, FL 33125, USA
| | - Marta Zarandi
- Veterans Affairs Medical Center and Departments of Pathology and Medicine, Division of Hematology/Oncology, University of Miami Miller School of Medicine, Miami, FL 33125, USA
| | - Jozsef Varga
- Veterans Affairs Medical Center and Departments of Pathology and Medicine, Division of Hematology/Oncology, University of Miami Miller School of Medicine, Miami, FL 33125, USA
| | - Peter CK Leung
- Department of Obstetrics & Gynaecology, Child and Family Research Institute, UBC, Vancouver, Canada
| |
Collapse
|
13
|
GHRH antagonists reduce the invasive and metastatic potential of human cancer cell lines in vitro. Cancer Lett 2010; 293:31-40. [PMID: 20064686 DOI: 10.1016/j.canlet.2009.12.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2009] [Revised: 12/13/2009] [Accepted: 12/17/2009] [Indexed: 12/21/2022]
Abstract
We investigated the effect of a GHRH antagonist, MIA-602on the metastatic cascade in vitro of three human cancers, DBTRG-05 glioblastoma, MDA-MB-468 estrogen-independent breast, and ES-2 clear cell ovarian cancer. GHRH receptors and their main splice variant, SV1 were detected on all three cell lines. After treatment with MIA-602, the cell viability decreased significantly, significant inhibition of cell invasion was observed and the release of MMPs was significantly decreased. The attachment of cancer cells to fibronectin and matrigel was severely hindered. Wound-healing experiments demonstrated a reduced cellular motility in all three cell lines. The upregulation of caveolin-1 and E-cadherin,and thepowerful downregulation of NF-kappaB and beta-catenin was detected. Our study suggests that the clinical application of highly potent GHRH antagonists in cancer therapy would be desirable since they inhibit proliferation and metastasis development as well.
Collapse
|
14
|
Markovic D, Challiss RAJ. Alternative splicing of G protein-coupled receptors: physiology and pathophysiology. Cell Mol Life Sci 2009; 66:3337-52. [PMID: 19629391 PMCID: PMC11115665 DOI: 10.1007/s00018-009-0093-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 06/30/2009] [Accepted: 07/03/2009] [Indexed: 12/16/2022]
Abstract
The G protein-coupled receptors (GPCRs) are a superfamily of transmembrane receptors that have a broad distribution and can collectively recognise a diverse array of ligands. Activation or inhibition of GPCR signalling can affect many (patho)physiological processes, and consequently they are a major target for existing and emerging drug therapies. A common observation has been that the pharmacological, signalling and regulatory properties of GPCRs can differ in a cell- and tissue-specific manner. Such "phenotypic" diversity might be attributable to post-translational modifications and/or association of GPCRs with accessory proteins, however, post-transcriptional mechanisms are also likely to contribute. Although approximately 50% of GPCR genes are intronless, those that possess introns can undergo alternative splicing, generating GPCR subtype isoforms that may differ in their pharmacological, signalling and regulatory properties. In this review we shall highlight recent research into GPCR splice variation and discuss the potential consequences this might have for GPCR function in health and disease.
Collapse
Affiliation(s)
- Danijela Markovic
- Department of Cell Physiology and Pharmacology, University of Leicester, Henry Wellcome Building, Leicester, UK.
| | | |
Collapse
|
15
|
Fu L, Osuga Y, Yano T, Takemura Y, Morimoto C, Hirota Y, Schally AV, Taketani Y. Expression and possible implication of growth hormone-releasing hormone receptor splice variant 1 in endometriosis. Fertil Steril 2008; 92:47-53. [PMID: 18684444 DOI: 10.1016/j.fertnstert.2008.04.048] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2008] [Revised: 04/21/2008] [Accepted: 04/21/2008] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To determine possible involvement of splice variant 1 (SV1), a variant of the pituitary growth hormone-releasing hormone (GHRH) receptor, in the development of endometriosis. DESIGN Comparative and laboratory study. SETTING University teaching hospital reproductive endocrinology and infertility practice. PATIENT(S) Eutopic and ectopic endometrial tissues, and peritoneal bone marrow-derived cells were collected from women with or without endometriosis. Normal ovarian tissues were collected from women without endometriosis. INTERVENTION(S) Ectopic endometrial stromal cells (ESC) were isolated and cultured with or without GHRH. MAIN OUTCOME MEASURE(S) Gene expression of GHRH and SV1 in the sample tissues was determined by reverse transcriptase (RT) nested polymerase chain reaction (PCR). Cyclic adenosine monophosphate (cAMP) production and 5-bromo-2'-deoxyuridine (BrdU) incorporation in ESC were measured using specific assay systems. RESULT(S) We detected SV1 messenger RNA (mRNA) in 17 out of 27 (63%) ectopic endometrial tissues, which was statistically significantly higher than that detected in eutopic endometrial tissues (2 out of 47, 4%) and normal ovarian tissues (0 out of 14). A relatively low rate of GHRH mRNA was detected in ectopic endometrial tissues (6 out of 27, 24%) and in eutopic endometrial tissues (12 out of 47, 26%). In contrast, relatively high rates were detected in normal ovarian tissues (14 out of 14, 100%) and peritoneal bone marrow-derived cells (13 out of 16, 81%). We found that GHRH stimulated the production of cAMP and the incorporation of BrdU in SV1-expressing ESC. CONCLUSION(S) GHRH and SV1 may play a role in promoting the development of endometriosis.
Collapse
Affiliation(s)
- Li Fu
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Köster F, Engel JB, Schally AV, Hönig A, Schröer A, Seitz S, Hohla F, Ortmann O, Diedrich K, Buchholz S. Triple-negative breast cancers express receptors for growth hormone-releasing hormone (GHRH) and respond to GHRH antagonists with growth inhibition. Breast Cancer Res Treat 2008; 116:273-9. [DOI: 10.1007/s10549-008-0120-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Accepted: 07/01/2008] [Indexed: 11/24/2022]
|
17
|
Affiliation(s)
- Michael J Waters
- Institute for Molecular Bioscience, University of Queensland, St. Lucia 4072, Australia.
| | | |
Collapse
|
18
|
Buchholz S, Schally AV, Engel JB, Hohla F, Heinrich E, Koester F, Varga JL, Halmos G. Potentiation of mammary cancer inhibition by combination of antagonists of growth hormone-releasing hormone with docetaxel. Proc Natl Acad Sci U S A 2007; 104:1943-6. [PMID: 17261802 PMCID: PMC1794297 DOI: 10.1073/pnas.0610860104] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Antagonists of growth hormone-releasing hormone (GHRH) are being developed for the treatment of various cancers. In this study, we investigated the effectiveness of treatment with GHRH antagonist JMR-132 alone and in combination with docetaxel chemotherapy in nude mice bearing MX-1 human breast cancers. Specific high-affinity binding sites for GHRH were found on MX-1 tumor membranes using ligand competition assays with (125)I-labeled GHRH antagonist JV-1-42. JMR-132 displaced radiolabeled JV-1-42 with an IC(50) of 0.14 nM, indicating a high affinity of JMR-132 to GHRH receptors. Treatment of nude mice bearing xenografts of MX-1 with JMR-132 at 10 microg per day s.c. for 22 days significantly (P < 0.05) inhibited tumor volume by 62.9% and tumor weight by 47.8%. Docetaxel given twice at a dose of 20 mg/kg i.p. significantly reduced tumor volume and weight by 74.1% and 58.6%, respectively. Combination treatment with JMR-132 (10 microg/day) and docetaxel (20 mg/kg i.p.) led to growth arrest of most tumors as shown by an inhibition of tumor volume and weight by 97.7% and 95.6%, respectively (P < 0.001). Because no vital cancer cells were detected in some of the excised tumors, a total regression of the tumors was achieved in some cases. Treatment with JMR-132 also strongly reduced the concentration of EGF receptors in MX-1 tumors. Our results demonstrate that GHRH antagonists might provide a therapy for breast cancer and could be combined with docetaxel chemotherapy to enhance the efficacy of treatment.
Collapse
Affiliation(s)
- Stefan Buchholz
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
- Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Universität Regensburg, 93051 Regensbug, Germany
| | - Andrew V. Schally
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
- Veterans Affairs Medical Center, South Florida Veterans Affairs Foundation for Research and Education, Miami, FL 33125
- University of Miami Miller School of Medicine, Miami, FL 33101
- To whom correspondence should be addressed at:
VA Medical Center, 1201 Northwest 16th Street, Research (151), Room 2A103C, Miami, FL 33125. E-mail:
| | - Jörg B. Engel
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
- Universität Würzburg, Frauenklinik, 97080 Würzburg, Germany; and
| | - Florian Hohla
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
- **Department of Internal Medicine, Hospital Oberndorf, 5100 Oberndorf, Austria
| | - Elmar Heinrich
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
| | - Frank Koester
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
| | - Jozsef L. Varga
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
| | - Gabor Halmos
- *Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
- Veterans Affairs Medical Center, South Florida Veterans Affairs Foundation for Research and Education, Miami, FL 33125
- University of Miami Miller School of Medicine, Miami, FL 33101
| |
Collapse
|
19
|
Insulin-like growth factors and breast cancer therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 608:101-12. [PMID: 17993235 DOI: 10.1007/978-0-387-74039-3_7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Despite improvements in breast cancer therapy in recent years, additional therapies need to be developed. New therapies may have activity by themselves or may have utility in combination with other agents. Population, preclinical, and basic data suggest the insulin-like growth factor (IGF) system functions to maintain the malignant phenotype in breast cancer. Since the IGFs act via transmembrane tyrosine kinase receptors, targeting of the key receptors could provide a new pathway in breast cancer. In addition, IGF action enhances cell survival, so combination of anti-IGF therapy with conventional cytotoxic drugs could lead to synergistic effects. In this review, we will discuss the rationale for targeting the IGF system, potential methods to disrupt IGF signaling, and identify potential interactions between IGF inhibitors and other anti-tumor strategies. We will also identify important issues to consider when designing clinical trials.
Collapse
|
20
|
Schulz S, Röcken C, Schulz S. Immunocytochemical localisation of plasma membrane GHRH receptors in human tumours using a novel anti-peptide antibody. Eur J Cancer 2006; 42:2390-6. [PMID: 16904887 DOI: 10.1016/j.ejca.2006.03.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2005] [Revised: 03/13/2006] [Accepted: 03/15/2006] [Indexed: 12/23/2022]
Abstract
Antagonists of growth hormone releasing hormone (GHRH) directly inhibit the growth of a variety of human neoplasms. However, the plasma membrane receptor mediating these effects has not been immunocytochemically visualised in primary tumour cells. Given that previous attempts using an antibody to the amino-terminal region did not result in the visualisation of plasma membrane receptors, we have developed and characterised an anti-peptide antibody to the carboxy-terminal region 403-422 of the human pituitary GHRH receptor. This sequence is identical to residues 339-358 of splice variant 1 (SV1) of tumoural GHRH receptors. Specificity of the antibody was demonstrated by (1) immunocytochemical staining of GHRH receptor-transfected cells, (2) detection of a broad glycosylated protein band migrating at Mr 50,000-60,000 in Western blots of membranes from human pituitary, and (3) abolition of tissue immunostaining by preadsorbtion of the antibody with its immunising peptide. The distribution of GHRH receptors was investigated in 69 formalin-fixed, paraffin-embedded human tumours showing that GHRH receptors were frequently expressed in breast, ovarian and prostate carcinomas. Immunoreactive GHRH receptors were clearly confined to the plasma membrane and uniformly present on nearly all tumour cells. In Western blots of membranes prepared from human tumours, the anti-GHRH receptor antibody detected a non-glycosylated protein band migrating at Mr 40,000, which corresponds to the expected molecular weight of splice variant 1 of tumoural GHRH receptors. Together, our findings provide direct evidence for the presence of GHRH receptor protein on the plasma membrane of primary human tumour cells. The GHRH receptor visualisation could be of value for a rapid immunohistochemical identification of those tumours which could be a target for diagnostic or therapeutic intervention using GHRH analogues.
Collapse
Affiliation(s)
- Solveig Schulz
- Department of Obstetrics and Gynecology, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | | | | |
Collapse
|
21
|
Gable KL, Maddux BA, Penaranda C, Zavodovskaya M, Campbell MJ, Lobo M, Robinson L, Schow S, Kerner JA, Goldfine ID, Youngren JF. Diarylureas are small-molecule inhibitors of insulin-like growth factor I receptor signaling and breast cancer cell growth. Mol Cancer Ther 2006; 5:1079-86. [PMID: 16648580 DOI: 10.1158/1535-7163.mct-05-0397] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In breast and certain other cancers, receptor tyrosine kinases, including the insulin-like growth factor I receptor (IGF-IR), play an important role in promoting the oncogenic process. The IGF-IR is therefore an important target for developing new anti-breast cancer therapies. An initial screening of a chemical library against the IGF-IR in breast cancer cells identified a diaryl urea compound as a potent inhibitor of IGF-IR signaling. This class of compounds has not been studied as inhibitors of the IGF-IR. We studied the effectiveness of one diaryl urea compound, PQ401, at antagonizing IGF-IR signaling and inhibiting breast cancer cell growth in culture and in vivo. PQ401 inhibited autophosphorylation of the IGF-IR in cultured human MCF-7 cells with an IC50 of 12 micromol/L and autophosphorylation of the isolated kinase domain of the IGF-IR with an IC50 <1 micromol/L. In addition, PQ401 inhibited the growth of cultured breast cancer cells in serum at 10 micromol/L. PQ401 was even more effective at inhibiting IGF-I-stimulated growth of MCF-7 cells (IC50, 6 micromol/L). Treatment of MCF-7 cells with PQ401 was associated with a decrease in IGF-I-mediated signaling through the Akt antiapoptotic pathway. Twenty-four hours of treatment with 15 micromol/L PQ401 induced caspase-mediated apoptosis. In vivo, treatment with PQ401 (i.p. injection thrice a week) reduced the growth rate of MCNeuA cells implanted into mice. These studies indicate that diaryl urea compounds are potential new agents to test in the treatment of breast and other IGF-I-sensitive cancers.
Collapse
Affiliation(s)
- Karissa L Gable
- Diabetes and Endocrine Research University of California, San Francisco/Mt. Zion Medical Center, Box 1616, San Francisco, CA 94143-1616, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Havt A, Schally AV, Halmos G, Varga JL, Toller GL, Horvath JE, Szepeshazi K, Köster F, Kovitz K, Groot K, Zarandi M, Kanashiro CA. The expression of the pituitary growth hormone-releasing hormone receptor and its splice variants in normal and neoplastic human tissues. Proc Natl Acad Sci U S A 2005; 102:17424-9. [PMID: 16299104 PMCID: PMC1297670 DOI: 10.1073/pnas.0506844102] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Various attempts to detect human pituitary growth hormone-releasing hormone receptor (pGHRH-R) in neoplastic extrapituitary tissues have thus far failed. Recently, four splice variants (SVs) of GHRH-R have been described, of which SV1 has the highest structural homology to pGHRH-R and likely plays a role in tumor growth. The aim of this study was to reinvestigate whether human tumors and normal human extrapituitary tissues express the pGHRH-R and to corroborate our previous findings on its SVs. Thus, we developed a real-time PCR method for the detection of the mRNA for the pGHRH-R, its SVs, and the GHRH peptide. Using real-time PCR, Western blotting, and radioligand-binding assays, we detected the mRNA for pGHRH-R and pGHRH-R protein in various human cancer cell lines grown in nude mice and in surgical specimens of human lung cancers. The expression of mRNA for SVs of pGHRH-R and GHRH was likewise found in xenografts of human non-Hodgkin's lymphomas, pancreatic cancer, glioblastoma, small-cell lung carcinomas, and in human nonmalignant prostate, liver, lung, kidney, and pituitary. Western blots showed that these normal and malignant human tissues contain SV1 protein and immunoreactive GHRH. Our results demonstrate that some normal human tissues and tumors express mRNA and protein for the pGHRH-R and its splice variants. These findings confirm and extend the concept that GHRH and its receptors play an important role in the pathophysiology of human cancers.
Collapse
Affiliation(s)
- Alexandre Havt
- Endocrine, Polypeptide, and Cancer Institute, Veterans Affairs Medical Center, New Orleans, LA 70112-1262, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Youngren JF, Gable K, Penaranda C, Maddux BA, Zavodovskaya M, Lobo M, Campbell M, Kerner J, Goldfine ID. Nordihydroguaiaretic Acid (NDGA) Inhibits the IGF-1 and c-erbB2/HER2/neu Receptors and Suppresses Growth in Breast Cancer Cells. Breast Cancer Res Treat 2005; 94:37-46. [PMID: 16142439 DOI: 10.1007/s10549-005-6939-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Nordihydroguaiaretic acid (NDGA) is a phenolic compound isolated from the creosote bush Larrea divaricatta that has anti-cancer activities both in vitro and in vivo. We can now attribute certain of these anti-cancer properties in breast cancer cells to the ability of NDGA to directly inhibit the function of two receptor tyrosine kinases (RTKs), the insulin-like growth factor receptor (IGF-1R) and the c-erbB2/HER2/neu (HER2/neu) receptor. In MCF-7 human breast cancer cells, low micromolar concentrations of NDGA inhibited activation of the IGF-1R, and downstream phosphorylation of both the Akt/PKB serine kinase and the pro-apoptotic protein BAD. In mouse MCNeuA cells, NDGA also inhibited ligand independent phosphorylation of HER2/neu. To study whether this inhibitory effect in cells was due to a direct action on these receptors, we studied the IGF-1-stimulated tyrosine kinase activity of isolated IGF-1R, which was inhibited by NDGA at 10 muM or less. NDGA was also effective at inhibiting autophosphorylation of the isolated HER2/neu receptor at similar concentrations. In addition, NDGA inhibited IGF-1 specific growth of cultured breast cancer cells with an IC50 of approximately 30 muM. NDGA treatment (intraperitoneal injection 3 times per week) also decreased the activity of the IGF-1R and the HER2/neu receptor in MCNeuA cells implanted into mice. This inhibition of RTK activity was associated with decreased growth rates of MCNeuA cells in vivo. These studies indicate that the anti-breast cancer properties of NDGA are related to the inhibition of two important RTKs. Agents of this class may therefore provide new insights into potential therapies for this disease.
Collapse
Affiliation(s)
- Jack F Youngren
- Division of Diabetes and Endocrine Research, San Francisco/Mt. Zion Medical Center, University of California, San Francisco, CA 94143-1616, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Jaeger LB, Banks WA, Varga JL, Schally AV. Antagonists of growth hormone-releasing hormone cross the blood-brain barrier: a potential applicability to treatment of brain tumors. Proc Natl Acad Sci U S A 2005; 102:12495-500. [PMID: 16118272 PMCID: PMC1194927 DOI: 10.1073/pnas.0504163102] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Hypothalamic growth hormone (GH)-releasing hormone (GHRH) stimulates the synthesis and release of GH from the pituitary gland. GHRH and its mRNA are also found in human cancers of the breast, ovary, prostate, lung, and other tumors, suggesting that GHRH is also a tumor growth factor. Various studies show that GHRH antagonists have antiproliferative effects in many tumor models; however, glioblastomas were examined only recently. Previous studies have demonstrated that s.c. administration of GHRH antagonist (JV-1-36) inhibited growth of s.c. U-87MG human glioblastomas and increased survival of nude mice with orthotopic implants of glioblastomas. Although treatment with JV-1-36 reduced tumorigenicity, it is not known whether peripherally administered GHRH antagonists can cross the blood-brain barrier. Brain endothelial cells joined by tight junctions form the blood-brain barrier, a "barrier" between the general circulation and the CNS. In this study, we administered a GHRH antagonist (JV-1-42) and showed that, after i.v. injection, iodinated JV-1-42 (131I-JV-1-42) enters the brain intact at a rate of 0.8514 mocrol/g per min with a serum half-life of 12.2 min. A one-site binding hyperbolic model indicated that the maximal percent of i.v. dose taken up per gram of brain was 0.41%. Coinjection of unlabeled JV-1-42 indicated that the transport from blood to brain is not saturable; however, transport from brain to blood is saturable and involves P-glycoprotein. Taken together, these results demonstrate that i.v.-administered 131I-JV-1-42 readily crosses the blood-brain barrier and accumulates in the brain. This finding indicates that GHRH antagonists could provide a potential treatment for malignant glioblastomas.
Collapse
Affiliation(s)
- Laura B Jaeger
- Department of Pharmacological and Physiological Sciences, Division of Geriatrics, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | | | | | | |
Collapse
|
25
|
Keller G, Schally AV, Groot K, Toller GL, Havt A, Köster F, Armatis P, Halmos G, Zarandi M, Varga JL, Engel JB. Effective treatment of experimental human non-Hodgkin's lymphomas with antagonists of growth hormone-releasing hormone. Proc Natl Acad Sci U S A 2005; 102:10628-33. [PMID: 16027368 PMCID: PMC1180787 DOI: 10.1073/pnas.0504102102] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Antagonists of growth hormone-releasing hormone (GHRH) were shown to inhibit the growth of various cancers. We investigated the antitumor activity and the mechanism of action of GHRH antagonists in human non-Hodgkin's lymphomas (NHL). Nude mice bearing xenografts of RL and HT human NHL were treated with GHRH antagonists MZ-5-156 and MZ-J-7-138 at a dose of 40 microg twice daily. The concentrations of serum IGF-1 and GHRH, bFGF, and VEGF in tumor tissue were measured by radioimmunoassays. Expression of GHRH and splice variant 1 of the GHRH receptor in both cell lines was examined by RT-PCR. The effects of MZ-5-156, MZ-J-7-138 and GHRH on cell proliferation were evaluated in vitro. Treatment with MZ-5-156 and MZ-J-7-138 significantly (P < 0.05) inhibited the growth of RL and HT tumors by 59.9-73.9%. High-affinity binding sites for GHRH and mRNA for GHRH and splice variant-1 of the GHRH receptors were found on RL and HT tumors. RL and HT cells contained GHRH peptide, and their growth in vitro was significantly inhibited by both antagonists. IGF-I levels in serum of mice were significantly decreased by antagonist MZ-5-156. Therapy with GHRH antagonists also significantly reduced tumoral bFGF, whereas VEGF levels were not suppressed. Our findings suggest that GHRH antagonists inhibit the growth of RL and HT lymphomas by direct effects mediated by tumoral receptors for GHRH. GHRH antagonists could offer a new therapeutic modality for the management of advanced NHL.
Collapse
Affiliation(s)
- Gunhild Keller
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, and Section of Experimental Medicine, Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Rekasi Z, Czompoly T, Schally AV, Boldizsar F, Varga JL, Zarandi M, Berki T, Horvath RA, Nemeth P. Antagonist of growth hormone-releasing hormone induces apoptosis in LNCaP human prostate cancer cells through a Ca2+-dependent pathway. Proc Natl Acad Sci U S A 2005; 102:3435-40. [PMID: 15728367 PMCID: PMC552899 DOI: 10.1073/pnas.0410006102] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Antagonists of growth hormone-releasing hormone (GHRH) exert antiproliferative effects directly on cancer cells, which are mediated by the tumoral GHRH receptors. However, the signal transduction pathways involved in antiproliferative effect of GHRH antagonists have not yet been elucidated. We used flow cytometry to investigate whether GHRH antagonist JV-1-38 can induce changes in the cytosolic free Ca2+ concentration leading to apoptosis in LNCaP human prostate cancer cells. JV-1-38 evoked prompt Ca2+ signal in a dose-dependent way (1-10 microM) and induced early stage of apoptosis in LNCaP human prostate cancer cells at a concentration effective in suppression of cell proliferation (10 microM) peaking after 3 h. Unexpectedly, agonist GHRH(1-29)NH2, which elevates cytosolic free Ca2+ concentration in pituitary somatotrophs at nanomolar concentrations, failed to induce Ca2+ signal or apoptosis even at a 10-fold higher concentration (100 microM). However, agonist GHRH(1-29)NH2 inhibited JV-1-38-induced Ca2+ signals in a dose-dependent way without affecting the antagonist-induced apoptosis. Peptides unrelated to GHRH did not induce Ca2+ signals in LNCaP human prostate cancer cells. EDTA (10 mM) or nifedipine (10 microM) significantly reduced the Ca2+ signal and early stage of apoptosis induced by JV-1-38, supporting the view that the increase in intracellular Ca2+ in response to JV-1-38 occurs primarily through extracellular Ca2+ entry through voltage-operated Ca2+ channels. In conclusion, GHRH antagonists activate tumoral GHRH receptors and are able to induce apoptosis in LNCaP human prostate cancer cells through a Ca2+-dependent pathway. Treatment with GHRH antagonists may offer a new approach to the therapy of prostate and other hormone-sensitive cancers.
Collapse
Affiliation(s)
- Zoltan Rekasi
- Department of Anatomy, University of Pécs, H-7624, Pécs, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Kiaris H, Schally AV, Kalofoutis A. Extrapituitary Effects of the Growth Hormone-Releasing Hormone. VITAMINS AND HORMONES 2005; 70:1-24. [PMID: 15727800 DOI: 10.1016/s0083-6729(05)70001-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
Growth hormone-releasing hormone (GHRH) is a neuropeptide secreted by the hypothalamus that stimulates the synthesis and release of growth hormone (GH) in the pituitary. Accumulating evidence suggests that in addition to GHRH's neuroendocrine action, GHRH is present in several extrahypothalamic tissues and is involved in a variety of cellular processes. Its function is related to the regulation of cell proliferation and differentiation of various nonpituitary cell types. In certain cases, ectopic production of GHRH has also been implicated in carcinogenesis. The mechanisms by which GHRH affects the peripheral extrapituitary tissues remain poorly understood, but it is likely that classic neuroendocrine action as well as paracrine and autocrine pathways are involved. Some headway has been made in the identification of extrapituitary receptors for GHRH and cDNA as splice variants of these GHRH receptors found in various tumors. The fact that the nonpituitary GHRH receptors are not fully identified, however, remains the major obstacle in studying, at a more mechanistic level, the action of local GHRH. This review summarizes the information available regarding the role of GHRH in the extrapituitary tissues with emphasis on its potential therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Hippokratis Kiaris
- Department of Biological Chemistry, Medical School, University of Athens, 115 27 Athens, Greece
| | | | | |
Collapse
|
28
|
Stangelberger A, Schally AV, Varga JL, Zarandi M, Szepeshazi K, Armatis P, Halmos G. Inhibitory Effect of Antagonists of Bombesin and Growth Hormone-Releasing Hormone on Orthotopic and Intraosseous Growth and Invasiveness of PC-3 Human Prostate Cancer in Nude Mice. Clin Cancer Res 2005. [DOI: 10.1158/1078-0432.49.11.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Purpose: To determine whether antagonists of growth hormone-releasing hormone (GHRH) and bombesin/gastrin-releasing peptide (BN/GRP) can inhibit the orthotopic and metastatic growth of PC-3 human androgen-independent prostate cancers.
Experimental Design: The effects of administration of GHRH antagonist MZ-J-7-118, BN/GRP antagonist RC-3940-II, and their combination on the growth and metastatic spread of PC-3 tumors implanted orthotopically into nude mice were evaluated. The efficacy of this treatment on PC-3 tumors implanted intratibially and s.c. was also determined.
Results: Treatment with MZ-J-7-118, RC-3940-II, or their combination significantly inhibited the growth of PC-3 tumors implanted orthotopically, intraosseously, and s.c. The combination of the two antagonists had the greatest effect, inhibiting orthotopic tumor growth by 77%, intratibially implanted tumors by 86%, and s.c. tumors by 86%. The therapy with BN/GRP and GHRH antagonists, especially in combination, also reduced the local tumor spread and distant metastases in animals bearing orthotopic tumors. Combination therapy was likewise the most effective in reducing the incidence and severity of tibial osteolytic lesions and pathologic fractures in intraosseously implanted tumors. High-affinity binding sites for BN/GRP and GHRH were found in s.c. and orthotopic PC-3 tumor samples. MZ-J-7-118, RC-3940-II, and the combination of both compounds inhibited in vitro growth of PC-3 cells.
Conclusions: Our findings show the efficacy of BN/GRP antagonists and GHRH antagonists for the treatment of advanced prostate cancer in preclinical metastatic models. As BN/GRP antagonists are already in clinical trials and GHRH antagonists are effective in androgen-independent prostate cancer models, these analogues could be considered for the management of advanced prostate carcinoma.
Collapse
Affiliation(s)
- Anton Stangelberger
- 1Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center and
- 2Section of Experimental Medicine, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Andrew V. Schally
- 1Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center and
- 2Section of Experimental Medicine, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Jozsef L. Varga
- 1Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center and
- 2Section of Experimental Medicine, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Marta Zarandi
- 1Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center and
- 2Section of Experimental Medicine, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Karoly Szepeshazi
- 1Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center and
- 2Section of Experimental Medicine, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Patricia Armatis
- 1Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center and
- 2Section of Experimental Medicine, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Gabor Halmos
- 1Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center and
- 2Section of Experimental Medicine, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| |
Collapse
|
29
|
Toller GL, Horvath JE, Schally AV, Halmos G, Varga JL, Groot K, Chism D, Zarandi M. Development of a polyclonal antiserum for the detection of the isoforms of the receptors for human growth hormone-releasing hormone on tumors. Proc Natl Acad Sci U S A 2004; 101:15160-5. [PMID: 15469915 PMCID: PMC524040 DOI: 10.1073/pnas.0406348101] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Antagonists of growth hormone-releasing hormone (GHRH) inhibit the growth of various human cancers by multiple mechanisms, which include direct effects on tumor cells through the splice variants (SV) of the GHRH receptor. Our findings suggest that the tumoral protein encoded by SV 1 (SV1) is a likely functional receptor. The aim of this study was to develop a polyclonal antiserum against a polypeptide analog of segment 1-25 of the putative SV1 receptor protein. Rabbits were immunized with [Ala-23]SV1 (1-25)-Tyr-26-Cys-27-NH2 as a hapten, conjugated to BSA or keyhole limpet hemocyanin. The antisera thus generated were evaluated by RIA for binding to the radiolabeled hapten. The specificity and sensitivity of the antisera were studied on xenografts of RL and HT human non-Hodgkin's lymphomas. The sera raised against keyhole limpet hemocyanin-SV1 hapten, showed binding values of 50-75% at a 1:56,000 dilution. In Western blot analyses, the purified polyclonal antibody recognized a specific signal with a molecular mass of approximately 40 kDa in RL and HT lymphomas. This band corresponds to the estimated molecular mass of the GHRH receptor isoform encoded by SV1. RT-PCR and ligand binding studies also revealed the expression of SV1 and the presence of high-affinity binding sites for GHRH on RL and HT tumors. Because the antiserum developed recognizes the tumoral GHRH receptor protein encoded by SV1, it should be of value in various investigations.
Collapse
MESH Headings
- Animals
- Antibodies/isolation & purification
- Cell Line, Tumor
- Female
- Genetic Variation
- Humans
- Lymphoma, Non-Hodgkin/genetics
- Lymphoma, Non-Hodgkin/metabolism
- Mice
- Mice, Nude
- Neoplasms/genetics
- Neoplasms/metabolism
- Protein Isoforms/genetics
- Protein Isoforms/immunology
- Protein Isoforms/metabolism
- Rabbits
- Receptors, Neuropeptide/genetics
- Receptors, Neuropeptide/immunology
- Receptors, Neuropeptide/metabolism
- Receptors, Pituitary Hormone-Regulating Hormone/genetics
- Receptors, Pituitary Hormone-Regulating Hormone/immunology
- Receptors, Pituitary Hormone-Regulating Hormone/metabolism
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Gabor L Toller
- Endocrine, Polypeptide, and Cancer Institute, Veterans Affairs Medical Center, New Orleans, LA 70112-1262, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
The insulin-like growth factors (IGF-I and -II) are potent mitogens and survival factors for both normal and malignant breast cells. These effects are mediated primarily through the IGF-I receptor (IGF-IR), which is significantly overexpressed and highly activated in breast tumors. The IGF-binding proteins are competitive inhibitors of IGF/IGF-IR interaction, limiting cellular proliferation and survival. Higher serum IGF-I levels or an increased ratio of IGF-I to IGF binding protein-3 is associated with an increased risk of developing breast cancer. Hence, interest in the IGF system as a potential target for the development of novel antineoplastic therapies has ensued. Several strategies to interrupt IGF-IR signaling are currently being evaluated for the treatment of breast cancer, including suppression of IGF production, reduction of functional IGF-IR levels, neutralization of IGF action, and inhibition of IGF-IR activation.
Collapse
Affiliation(s)
- Lori Jerome
- Department of Oncology, McGill University, Montreal, Quebec, Canada
| | | | | |
Collapse
|
31
|
Kiaris H, Chatzistamou I, Schally AV, Halmos G, Varga JL, Koutselini H, Kalofoutis A. Ligand-dependent and -independent effects of splice variant 1 of growth hormone-releasing hormone receptor. Proc Natl Acad Sci U S A 2003; 100:9512-7. [PMID: 12867592 PMCID: PMC170949 DOI: 10.1073/pnas.1533185100] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Existing evidence indicates that, in addition to its neuroendocrine action, growth hormone-releasing hormone (GHRH) acts directly on several nonpituitary tissues, especially neoplasms, and stimulates cell proliferation. We have recently reported that a splice variant of the receptor (SV1) is expressed in various normal tissues and particularly in tumor tissues, producing mitogenic effects on GHRH binding. By using HEC-1A human endometrial carcinoma cells, which express endogenous SV1, we show that, in addition to its ability to mediate the mitogenic effects of GHRH, SV1 also possesses relatively high intrinsic, ligand-independent activity. By using an antisense RNA-based approach we found that SV1 ablation reduces the efficacy of colony formation and the rate of cell proliferation of HEC-1A cells in the absence of exogenous GHRH, and decreases their sensitivity to GHRH when the neurohormone is added to the culture media. This ligand-independent stimulation of cell proliferation appears to be a characteristic property of the truncated form of the receptor, because the expression of SV1 and not of the full-length GHRH receptor stimulated the proliferation of 3T3 fibroblasts in the absence of exogenous GHRH, whereas both forms mediated the proliferative effects of GHRH. Evaluation of 21 specimens of human primary endometrial carcinoma for expression of SV1 by immunohistochemistry indicated that in contrast to the GHRH receptor, which is absent, SV1 is expressed in approximately 43% of the specimens. These findings indicate that SV1 can operate in a ligand-independent as well as a ligand-dependent manner. The overexpression of this form of GHRH receptor may be associated with carcinogenesis.
Collapse
Affiliation(s)
- Hippokratis Kiaris
- Department of Biological Chemistry,Medical School, University of Athens, 115 27 Athens, Greece.
| | | | | | | | | | | | | |
Collapse
|
32
|
Kiaris H, Koutsilieris M, Kalofoutis A, Schally AV. Growth hormone-releasing hormone and extra-pituitary tumorigenesis: therapeutic and diagnostic applications of growth hormone-releasing hormone antagonists. Expert Opin Investig Drugs 2003; 12:1385-94. [PMID: 12882623 DOI: 10.1517/13543784.12.8.1385] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Growth hormone-releasing hormone (GHRH) regulates growth hormone release from the pituitary. However, in addition to this neuroendocrine action, much evidence implies an additional role for GHRH in carcinogenesis in non-pituitary tissues. This role of GHRH in cancer development appears to be due to the operation of several mechanisms, which involve the regulation of the growth hormone-dependent hepatic insulin-like growth factor I (IGFI) production, tumoural IGF-I and IGF-II secretion and direct action of GHRH on tumour cells by autocrine and/or paracrine pathways. This review summarises the available information regarding the role of GHRH in tumorigenesis with special emphasis on the direct action of GHRH in primary and experimental cancers.
Collapse
Affiliation(s)
- Hippokratis Kiaris
- Department of Biological Chemistry, Medical School, University of Athens, 75 Micras Asias, 115 27 Athens, Greece.
| | | | | | | |
Collapse
|
33
|
Garcia-Fernandez MO, Schally AV, Varga JL, Groot K, Busto R. The expression of growth hormone-releasing hormone (GHRH) and its receptor splice variants in human breast cancer lines; the evaluation of signaling mechanisms in the stimulation of cell proliferation. Breast Cancer Res Treat 2003; 77:15-26. [PMID: 12602901 DOI: 10.1023/a:1021196504944] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Antagonists of growth hormone-releasing hormone (GHRH) inhibit growth of various human cancers including breast cancer, xenografted into nude mice or cultured in vitro. Splice variants (SVs) of receptors for GHRH have been found in several human cancers and cancer cell lines. The antiproliferative actions of GHRH antagonists could be mediated in part through these SVs of GHRH receptors. In this study we examined the expression of mRNA for GHRH and SVs of its receptors in human breast cancer cell lines MCF-7, MCF-7MIII, MDA-MB-231, MDA-MB-435, MDA-MB-468, and T47D. mRNA for GHRH was present in all lines tested. mRNA for SV1 isoform of GHRH receptors was found in MCF-7MIII, MDA-MB-468, and T47D; and for SV2 isoform in MCF-7MIII and T47D cell lines. In proliferation studies in vitro, the growth of T47D cells was stimulated by GHRH and dose-dependently inhibited by GHRH antagonist JV-1-38. H89 (protein kinase A inhibitor), bisindolylmaleimide I (protein kinase C [PKC] inhibitor) and verapamil (voltage-dependent calcium channel blocker) inhibited the GHRH-stimulated proliferation of T47D cells. The GHRH antagonist JV-1-38 suppressed the T47D cell growth in vitro stimulated by PKC activator (phorbol-12-myristate-13-acetate). The stimulation of T47D cells by GHRH was followed by an increase in cAMP production and GHRH antagonist JV-1-38 competitively inhibited this effect. Our results suggest that SVs of GHRH receptors could mediate the responses to GHRH and GHRH antagonists in breast cancer through Ca2+-, cAMP- and PKC-dependent mechanisms. The presence of SV1 of GHRH receptors in human cancers provides a rationale for antitumor therapy based on the blockade of this receptor by specific GHRH antagonists.
Collapse
Affiliation(s)
- M Olga Garcia-Fernandez
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, New Orleans, LA 70112-1262, USA
| | | | | | | | | |
Collapse
|
34
|
Busto R, Schally AV, Braczkowski R, Plonowski A, Krupa M, Groot K, Armatis P, Varga JL. Expression of mRNA for growth hormone-releasing hormone and splice variants of GHRH receptors in human malignant bone tumors. REGULATORY PEPTIDES 2002; 108:47-53. [PMID: 12220726 DOI: 10.1016/s0167-0115(02)00109-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Splice variants (SV) of receptors for growth hormone-releasing hormone (GHRH) have been found in several human cancer cell lines. GHRH antagonists inhibit growth of various human cancers, including osteosarcomas and Ewing's sarcoma, xenografted into nude mice or cultured in vitro and their antiproliferative action could be mediated, in part, through these SV of GHRH receptors. In this study, we found mRNA for the SV(1) isoform of GHRH receptors in human osteosarcoma line MNNG/HOS and SK-ES-1 Ewing's sarcoma line. We also detected mRNA for GHRH, which is apparently translated into the GHRH peptide and secreted by the cells, as shown by the presence of GHRH-like immunoreactivity in the conditioned media of cell cultures. In proliferation studies in vitro, the growth of SK-ES-1 and MNNG/HOS cells was dose-dependently inhibited by GHRH antagonist JV-1-38 and an antiserum against human GHRH. Our study indicates the presence of an autocrine stimulatory loop based on GHRH and SV(1) of GHRH receptors in human sarcomas. The direct antiproliferative effects of GHRH antagonists on malignant bone tumors appear to be exerted through the SV(1) of GHRH receptors on tumoral cells.
Collapse
Affiliation(s)
- R Busto
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Braczkowski R, Schally AV, Plonowski A, Varga JL, Groot K, Krupa M, Armatis P. Inhibition of proliferation in human MNNG/HOS osteosarcoma and SK-ES-1 Ewing sarcoma cell lines in vitro and in vivo by antagonists of growth hormone-releasing hormone: effects on insulin-like growth factor II. Cancer 2002; 95:1735-45. [PMID: 12365022 DOI: 10.1002/cncr.10865] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Antagonists of growth hormone-releasing hormone (GH-RH) can inhibit the proliferation of various tumors either indirectly through the suppression of the pituitary growth hormone/hepatic insulin-like growth factor I (IGF-I) axis and the lowering of serum IGF-I concentration or directly by reducing the levels of IGF-I and IGF-II and their mRNA expression in tumors and blocking the effect of autocrine GH-RH. In this study, the authors investigated the effects of the GH-RH antagonist JV-1-38 on MNNG/HOS human osteosarcoma and SK-ES-1 human Ewing sarcoma cell lines. METHODS Male nude mice bearing subcutaneous xenografts of MNNG/HOS or SK-ES-1 tumors were treated subcutaneously with JV-1-38 at a dose of 20 microg twice daily for 4 weeks. The concentrations of IGF-I and IGF-II in serum and in tumor tissue were measured by radioimmunoassay. Tumor and liver levels of mRNA for IGF-I and IGF-II were determined by reverse transcriptase-polymerase chain reaction analysis. The effects of JV-1-38, IGF-I, and IGF-II on cell proliferation in vitro were evaluated. RESULTS GH-RH antagonist significantly (P < 0.05) inhibited the tumor volume and tumor weight of MNNG/HOS and SK-ES-1 tumors by > 50% after 4 weeks and increased tumor doubling time. JV-1-38 lowered the serum IGF-I level, decreased the expression of mRNA for IGF-I in the liver, and significantly (P < 0.05-0.01) reduced the concentration of IGF-II and mRNA levels for IGF-II in both sarcomas. The concentration of IGF-I was lowered only in SK-ES-1 tumors. In vitro, the proliferation of SK-ES-1 and MNNG/HOS cells was inhibited by JV-1-38 and by antisera to IGF-I and IGF-II. CONCLUSIONS The inhibition of MNNG/HOS osteosarcoma and SK-ES-1 Ewing sarcoma by GH-RH antagonists was linked to a suppression of IGF-II production in tumors. However, in SK-ES-1 tumors, the effects on IGF-I also may be involved.
Collapse
Affiliation(s)
- Ryszard Braczkowski
- Endocrine, Polypeptide, and Cancer Institute, Veterans Affairs Medical Center, New Orleans, Louisiana 70112-1262, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Busto R, Schally AV, Varga JL, Garcia-Fernandez MO, Groot K, Armatis P, Szepeshazi K. The expression of growth hormone-releasing hormone (GHRH) and splice variants of its receptor in human gastroenteropancreatic carcinomas. Proc Natl Acad Sci U S A 2002; 99:11866-71. [PMID: 12186980 PMCID: PMC129360 DOI: 10.1073/pnas.182433099] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2002] [Indexed: 12/28/2022] Open
Abstract
Splice variants (SVs) of receptors for growth hormone-releasing hormone (GHRH) have been found in primary human prostate cancers and diverse human cancer cell lines. GHRH antagonists inhibit growth of various experimental human cancers, including pancreatic and colorectal, xenografted into nude mice or cultured in vitro, and their antiproliferative action could be mediated in part through SVs of GHRH receptors. In this study we examined the expression of mRNA for GHRH and for SVs of its receptors in tumors of human pancreatic, colorectal, and gastric cancer cell lines grown in nude mice. mRNA for both GHRH and SV(1) isoform of GHRH receptors was expressed in tumors of pancreatic (SW1990, PANC-1, MIA PaCa-2, Capan-1, Capan-2, and CFPAC1), colonic (COLO 320DM and HT-29), and gastric (NCI-N87, HS746T, and AGS) cancer cell lines; mRNA for SV(2) was also present in Capan-1, Capan-2, CFPAC1, HT-29, and NCI-N87 tumors. In proliferation studies in vitro, the growth of pancreatic, colonic, and gastric cancer cells was stimulated by GHRH(1-29)NH(2) and inhibited by GHRH antagonist JV-1-38. The stimulation of some gastroenteropancreatic cancer cells by GHRH was followed by an increase in cAMP production, and GHRH antagonist JV-1-38 competitively inhibited this effect. Our study indicates the presence of an autocrine/paracrine stimulatory loop based on GHRH and SV(1) of GHRH receptors in human pancreatic, colorectal, and gastric cancers. The finding of SV(1) receptor in human cancers provides an approach to an antitumor therapy based on the blockade of this receptor by specific GHRH antagonists.
Collapse
Affiliation(s)
- Rebeca Busto
- Endocrine, Polypeptide, and Cancer Institute, Veterans Affairs Medical Center, and Section of Experimental Medicine, Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Swanson SM, Unterman TG. The growth hormone-deficient Spontaneous Dwarf rat is resistant to chemically induced mammary carcinogenesis. Carcinogenesis 2002; 23:977-82. [PMID: 12082019 DOI: 10.1093/carcin/23.6.977] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Recent epidemiologic studies have suggested that the growth hormone (GH)/insulin-like growth factor I axis plays an important role in human breast cancer. The purpose of the present study was to evaluate the function of GH in rat mammary carcinogenesis, a model that closely recapitulates human breast cancer biology. The Spontaneous Dwarf rat (SDR) arose from the Sprague-Dawley rat and harbors a mutation in its GH gene yielding undetectable levels of a severely truncated protein not capable of binding to the GH receptor. When female rats of either strain were exposed to the direct-acting carcinogen N-methyl-N-nitrosourea, all wild-type rats (n = 10) developed multiple mammary cancers (5.3/rat). In contrast, SDR rats (n = 15) developed only three cancers (0.2/rat) and these were very small (<6 mm3). In another experiment, SDRs were backcrossed with wild-type Sprague-Dawley rats and the progeny were exposed to the indirect-acting carcinogen 7,12-dimethylbenz[a]anthracene. Progeny that were either homo- or heterozygous for the wild-type GH gene developed approximately 4 mammary tumors/rat, respectively. In contrast, SDR progeny developed only 0.21 tumors/rat. Mammary glands of SDRs had substantially less alveolar development compared with wild-type, yet ductal branching was similar in the two strains. Infusion of rat GH to SDRs induced mammary epithelial cell proliferation and alveolar development similar to that of wild-type rats. Taken together, these results demonstrate an important role for GH in alveolar development in the virgin rat, and provide the first direct evidence that GH plays a critical role in mammary carcinogenesis.
Collapse
Affiliation(s)
- Steven M Swanson
- Program for Collaborative Research in the Pharmaceutical Sciences and the Center for Pharmaceutical Biotechnology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | | |
Collapse
|
38
|
Kiaris H, Schally AV, Busto R, Halmos G, Artavanis-Tsakonas S, Varga JL. Expression of a splice variant of the receptor for GHRH in 3T3 fibroblasts activates cell proliferation responses to GHRH analogs. Proc Natl Acad Sci U S A 2002; 99:196-200. [PMID: 11773624 PMCID: PMC117538 DOI: 10.1073/pnas.012590999] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2001] [Indexed: 01/02/2023] Open
Abstract
The stimulatory effects of growth hormone-releasing hormone (GHRH) and the antiproliferative action of GHRH antagonists have been demonstrated in various cancers, but the receptors that mediate these responses are not clearly identified. Recently, we reported that human cancer cell lines express splice variants (SVs) of the receptors for GHRH. SV1 exhibits the greatest similarity to the pituitary GHRH receptor and is most likely to be functional. To ascertain whether SV1 mediates mitogenic effects on nonpituitary tissues, we expressed SV1 in 3T3 mouse fibroblasts and studied the properties of the transfected cells. Radioligand binding assays with (125)I-labeled GHRH antagonist JV-1-42 detected high affinity (K(d) = 0.58 +/- 0.17 nM) binding sites for GHRH with a maximal binding capacity (B(max)) of 103 +/- 17.4 fmol/mg of membrane protein in 3T3 cells transfected with pcDNA3-SV1, whereas the control cells transfected with the empty vector did not show any GHRH binding. Cell proliferation studies showed that cells expressing SV1 are much more sensitive to GHRH analogs than the pcDNA3 controls. Thus, the expression of SV1 augments the stimulatory responses to GHRH(1-29)NH(2) or GHRH agonist JI-38 and inhibitory responses to GHRH antagonist JV-1-38 as compared with pcDNA3 controls. The stimulation of SV1-expressing cells by GHRH or JI-38 is followed by an increase in cAMP production, but no GH release occurs. Vasoactive intestinal peptide had no effect, and its antagonist JV-1-53 did not inhibit the proliferation of SV1-expressing cells stimulated by GHRH. Our results suggest that SV1 could mediate responses of nonpituitary cells and various tumors to GHRH and GHRH antagonists. The presence of SV1 in several human cancer cell lines provides a rationale for antitumor therapy based on the blockade of this receptor by specific GHRH antagonists.
Collapse
Affiliation(s)
- Hippokratis Kiaris
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | | | | | | | | | | |
Collapse
|
39
|
Schally AV, Comaru-Schally AM, Nagy A, Kovacs M, Szepeshazi K, Plonowski A, Varga JL, Halmos G. Hypothalamic hormones and cancer. Front Neuroendocrinol 2001; 22:248-91. [PMID: 11587553 DOI: 10.1006/frne.2001.0217] [Citation(s) in RCA: 203] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The use of peptide analogs for the therapy of various cancers is reviewed. Inhibition of the pituitary-gonadal axis forms the basis for oncological applications of luteinizing hormone-releasing hormone (LH-RH) agonists and antagonists, but direct effects on tumors may also play a role. Analogs of somatostatin are likewise used for treatment of various tumors. Radiolabeled somatostatin analogs have been successfully applied for the localization of tumors expressing somatostatin receptors. Studies on the role of tumoral LH-RH, growth hormone-releasing hormone (GH-RH), and bombesin/GRP and their receptors in the proliferation of various tumors are summarized, but the complete elucidation of all the mechanisms involved will require much additional work. Human tumors producing hypothalamic hormones are also discussed. Treatment of many cancers remains a major challenge, but new therapeutic modalities are being developed based on antagonists of GH-RH and bombesin, which inhibit growth factors or their receptors. Other approaches consist of the use of cytotoxic analogs of LH-RH, bombesin, and somatostatin, which can be targeted to receptors for these peptides in various cancers and their metastases. These new classes of peptide analogs should lead to a more effective treatment for various cancers.
Collapse
Affiliation(s)
- A V Schally
- Endocrine, Polypeptide, and Cancer Institute, Veterans Affairs Medical Center, 1601 Perdido Str., New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Chatzistamou I, Schally AV, Varga JL, Groot K, Busto R, Armatis P, Halmos G. Inhibition of growth and metastases of MDA-MB-435 human estrogen-independent breast cancers by an antagonist of growth hormone-releasing hormone. Anticancer Drugs 2001; 12:761-8. [PMID: 11593058 DOI: 10.1097/00001813-200110000-00008] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Antagonists of growth hormone-releasing hormone (GH-RH) inhibit the growth of various cancers by mechanism(s) that include the suppression of the insulin-like growth factors (IGF)-I and/or -II. In this study, nude mice bearing orthotopic implants of MDA-MB-435 human estrogen-independent breast carcinoma received 39 days of therapy with GH-RH antagonist JV-1-36 (20 microg/day). The treatment significantly inhibited tumor growth by 71.1% (p<0.01) and nullified the metastatic potential of MDA-MB-435 cells. Four of eight control mice (50%) developed metastases in the lymph nodes and one (12.5%) in the lung, but none of the animals receiving JV-1-36 showed metastatic spread. GH-RH antagonist JV-1-36 inhibited the growth of MDA-MB-435 cells in vitro, while IGF-I stimulated it. However, mRNA for IGF-I or -II was not detected in MDA-MB-435 cells, indicating that the suppression of autocrine IGFs may not be involved in the antiproliferative mechanism. Using ligand competition assays with (125)I-labeled GH-RH antagonist JV-1-42, specific high-affinity binding sites for GH-RH were found on tumor membranes. Reverse transcription-polymerase chain reaction revealed the expression of mRNA for GH-RH receptor splice variant-1 in MDA-MB-435 tumors. Our results suggest that the antitumorigenic action of GH-RH antagonists on MDA-MB-435 breast cancer could be direct and mediated by tumoral GH-RH receptors.
Collapse
Affiliation(s)
- I Chatzistamou
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, 1601 Perdido Street, New Orleans, LA 70112-1262, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Rekasi Z, Varga JL, Schally AV, Plonowski A, Halmos G, Csernus B, Armatis P, Groot K. Antiproliferative actions of growth hormone-releasing hormone antagonists on MiaPaCa-2 human pancreatic cancer cells involve cAMP independent pathways. Peptides 2001; 22:879-86. [PMID: 11390017 DOI: 10.1016/s0196-9781(01)00413-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We evaluated the effects of GHRH antagonists on the proliferation of MiaPaCa-2 human pancreatic cancer cells and cAMP signaling in vitro. GHRH antagonists inhibited the proliferation of MiaPaCa-2 cells in vitro in a dose-dependent way and caused a significant elevation in cAMP production. In a superfusion system, short-term exposure of the cells to GHRH antagonists evoked an acute, dose-dependent release of cAMP into the medium. Native GHRH, which stimulates cAMP efflux from pituitary at nanomolar doses, did not influence cAMP release from cultured or superfused MiaPaCa-2 cells even at 10-30 microM. VIP, PACAP, secretin and glucagon also did not influence cell proliferation or cAMP production. Adenylate cyclase activator forskolin (FSK) caused a greater cAMP response, but a smaller antiproliferative effect than GHRH antagonists. Combined treatment with FSK and GHRH antagonist JV-1-38 potentiated the cAMP-inducing effect of FSK, but did not produce a greater inhibition of cell proliferation than JV-1-38 alone. A selective accumulation of radiolabeled GHRH antagonist [(125)I]JV-1-42 in vivo in MiaPaCa-2 carcinoma xenografted into nude mice was also observed. In conclusion, second messengers other than cAMP participate in the signal transduction pathways of GHRH analogs mediated by tumoral GHRH receptors.
Collapse
Affiliation(s)
- Z Rekasi
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Chatzistamou I, Schally AV, Varga JL, Groot K, Armatis P, Busto R, Halmos G. Antagonists of growth hormone-releasing hormone and somatostatin analog RC-160 inhibit the growth of the OV-1063 human epithelial ovarian cancer cell line xenografted into nude mice. J Clin Endocrinol Metab 2001; 86:2144-52. [PMID: 11344219 DOI: 10.1210/jcem.86.5.7487] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The effects of antagonists of GHRH and the somatostatin analog RC-160 on the growth of OV-1063 human epithelial ovarian cancer cells xenografted into nude mice were investigated. Treatment with 20 microg/day of the GHRH antagonist JV-1-36 or MZ-5-156 and 60 microg/day of the somatostatin analog RC-160 for 25 days decreased tumor volume by 70.9% (P < 0.01), 58.3% (P < 0.05), and 60.6% (P < 0.01), respectively, vs. the control value. The levels of GH in serum were decreased in all of the treated groups, but only RC-160 significantly reduced serum insulin-like growth factor I (IGF-I). The levels of messenger ribonucleic acid (mRNA) for IGF-I and -II and for their receptors in OV-1063 tumors were investigated by multiplex RT-PCR. No expression of mRNA for IGF-I was detected, but treatment with JV-1-136 caused a 51.8% decrease (P < 0.05) in the level of mRNA for IGF-II in tumors. Exposure of OV-1063 cells cultured in vitro to GHRH, IGF-I, or IGF-II significantly (P < 0.05) stimulated cell growth, but 10(-5) mol/L JV-1-36 nearly completely inhibited (P < 0.001) OV-1063 cell proliferation. OV-1063 tumors expressed mRNA for GHRH receptors and showed the presence of binding sites for GHRH. Our results indicate that antagonistic analogs of GHRH and the somatostatin analog RC-160 inhibit the growth of epithelial ovarian cancers. The effects of RC-160 seem to be exerted more on the pituitary GH-hepatic IGF-I axis, whereas GHRH antagonists appear to reduce IGF-II production and interfere with the autocrine regulatory pathway. The antitumorigenic action of GHRH antagonists appears to be mediated by GHRH receptors found in OV-1063 tumors.
Collapse
Affiliation(s)
- I Chatzistamou
- Endocrine, Polypeptide, and Cancer Institute, Veterans Affairs Medical Center, Section of Experimental Medicine, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Kiaris H, Schally AV, Armatis P. Direct action of growth hormone-releasing hormone agonist JI-38 on normal human fibroblasts: evidence from studies on cell proliferation and c-myc proto-oncogene expression. REGULATORY PEPTIDES 2001; 96:119-24. [PMID: 11111017 DOI: 10.1016/s0167-0115(00)00166-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Growth hormone-releasing hormone (GHRH) is secreted by the hypothalamus and stimulates the release of growth hormone from the pituitary. Recent studies also indicate that in addition to its neuroendocrine function, GHRH may play a direct role in the proliferation of cancer cells, acting as growth factor for various human tumors. In the present study we investigated the effects of JI-38, an agonistic analog of GHRH, on the rate of proliferation of normal human diploid dermal fibroblasts (NHF) cultured in vitro. The effects of JI-38 on the levels of mRNA for c-myc proto-oncogene were also tested. Exposure to 10(-7) M JI-38 stimulated the rate of proliferation of early passage NHF by about 100%. Exposure of NHF cells to 10(-8)-5x10(-6) M JI-38 for 24 h resulted in about 0.5-3.5 fold increase in the levels of mRNA for c-myc proto-oncogene. The ability of JI-38 to stimulate the proliferation of NHF cells was abolished in cells cultured at late passage. Continuous exposure to 10(-7) M JI-38, over 6-7 passages (15-20 population doublings), progressively reduced the rate of proliferation of NHF compared with cells exposed to medium alone, indicating that GHRH agonist acted as a growth inhibitor. Our results suggest that at certain developmental stages, GHRH may act on various tissues, stimulating cell proliferation.
Collapse
Affiliation(s)
- H Kiaris
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center and Section of Experimental Medicine, Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112-1262, USA
| | | | | |
Collapse
|
44
|
Halmos G, Schally AV, Varga JL, Plonowski A, Rekasi Z, Czompoly T. Human renal cell carcinoma expresses distinct binding sites for growth hormone-releasing hormone. Proc Natl Acad Sci U S A 2000; 97:10555-60. [PMID: 10962030 PMCID: PMC27063 DOI: 10.1073/pnas.180313097] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2000] [Indexed: 12/28/2022] Open
Abstract
Antagonists of growth hormone-releasing hormone (GHRH) inhibit the proliferation of various human cancers in vitro and in vivo by mechanisms that include apparent direct effects through specific binding sites expressed on tumors and that differ from pituitary human GHRH (hGHRH) receptors. In this study, GHRH antagonist JV-1-38 (20 microgram/day per animal s.c.) inhibited the growth of orthotopic CAKI-1 human renal cell carcinoma (RCC) by 83% and inhibited the development of metastases to lung and lymph nodes. Using ligand competition assays with (125)I-labeled GHRH antagonist JV-1-42, we demonstrated the presence of specific high-affinity (K(d) = 0.25 +/- 0.03 nM) binding sites for GHRH with a maximal binding capacity (B(max)) of 70.2 +/- 4.1 fmol/mg of membrane protein in CAKI-1 tumors. These receptors bind GHRH antagonists preferentially and display a lower affinity for hGHRH. The binding of (125)I-JV-1-42 is not inhibited by vasoactive intestinal peptide (VIP)-related peptides sharing structural homology with hGHRH. The receptors for GHRH antagonists on CAKI-1 tumors are distinct from binding sites detected with (125)I-VIP (K(d) = 0.89 +/- 0.14 nM; B(max) = 183.5 +/- 2.6 fmol/mg of protein) and also have different characteristics from GHRH receptors on rat pituitary as documented by the insignificant binding of [His(1),(125)I-Tyr(10), Nle(27)]hGHRH(1-32)NH(2). Reverse transcription-PCR revealed the expression of splice variants of hGHRH receptor in CAKI-1 RCC. Biodistribution studies demonstrate an in vivo uptake of (125)I-JV-1-42 by the RCC tumor tissue. The presence of specific receptor proteins that bind GHRH antagonists in CAKI-1 RCC supports the view that distinct binding sites that mediate the inhibitory effect of GHRH antagonists are present on various human cancers.
Collapse
Affiliation(s)
- G Halmos
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112-2699, USA
| | | | | | | | | | | |
Collapse
|
45
|
Rekasi Z, Czompoly T, Schally AV, Halmos G. Isolation and sequencing of cDNAs for splice variants of growth hormone-releasing hormone receptors from human cancers. Proc Natl Acad Sci U S A 2000; 97:10561-6. [PMID: 10962031 PMCID: PMC27064 DOI: 10.1073/pnas.180313297] [Citation(s) in RCA: 125] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2000] [Indexed: 01/15/2023] Open
Abstract
The proliferation of various tumors is inhibited by the antagonists of growth hormone-releasing hormone (GHRH) in vitro and in vivo, but the receptors mediating the effects of GHRH antagonists have not been identified so far. Using an approach based on PCR, we detected two major splice variants (SVs) of mRNA for human GHRH receptor (GHRH-R) in human cancer cell lines, including LNCaP prostatic, MiaPaCa-2 pancreatic, MDA-MB-468 breast, OV-1063 ovarian, and H-69 small-cell lung carcinomas. In addition, high-affinity, low-capacity binding sites for GHRH antagonists were found on the membranes of cancer cell lines such as MiaPaCa-2 that are negative for the vasoactive intestinal peptide/pituitary adenylate cyclase-activating polypeptide receptor (VPAC-R) or lines such as LNCaP that are positive for VPAC-R. Sequence analysis of cDNAs revealed that the first three exons in SV(1) and SV(2) are replaced by a fragment of retained intron 3 having a new putative in-frame start codon. The rest of the coding region of SV(1) is identical to that of human pituitary GHRH-R, whereas in SV(2) exon 7 is spliced out, resulting in a 1-nt upstream frameshift, which leads to a premature stop codon in exon 8. The intronic sequence may encode a distinct 25-aa fragment of the N-terminal extracellular domain, which could serve as a proposed signal peptide. The continuation of the deduced protein sequence coded by exons 4-13 in SV(1) is identical to that of pituitary GHRH-R. SV(2) may encode a GHRH-R isoform truncated after the second transmembrane domain. Thus SVs of GHRH-Rs have now been identified in human extrapituitary cells. The findings support the view that distinct receptors are expressed on human cancer cells, which may mediate the antiproliferative effect of GHRH antagonists.
Collapse
Affiliation(s)
- Z Rekasi
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | | | | |
Collapse
|
46
|
Kiaris H, Schally AV, Varga JL. Antagonists of growth hormone-releasing hormone inhibit the growth of U-87MG human glioblastoma in nude mice. Neoplasia 2000; 2:242-50. [PMID: 10935510 PMCID: PMC1507563 DOI: 10.1038/sj.neo.7900074] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Antagonists of growth hormone-releasing hormone(GH-RH)inhibit the growth of various cancers by mechanisms that involve the suppression of the insulin-like growth factor (IGF)-I and/or IGF-II. In view of the importance of the IGF system in glioma tumorigenesis, the effects of GH-RH antagonists MZ-5-156 and JV-1-36 were investigated in nude mice bearing subcutaneous and orthotopic xenografts of U-87MG human glioblastomas. After 4 weeks of therapy with MZ-5-156 or JV-1 -36 at the dose of 20 microg/day per animal, the final volume of subcutaneous U-87MG tumors was significantly (P < .01) decreased by 84% and 76%, respectively, as compared with controls. Treatment with GH-RH antagonists also reduced tumor weight and the levels of mRNA for IGF receptor type I (IGFR-I). A reduction in the mRNA levels for IGF-II was found in tumors of mice treated with MZ-5-156. Treatment with MZ-5-156 or JV-1 -36 also extended the survival of nude mice implanted orthotopically with U-87MG glioblastomas by 81% (P < .005) and 18%, respectively, as compared with the controls. Exposure in vitro to GH-RH antagonists MZ-5-156 or JV-1 -36 at 1 microM concentration for 24 hours decreased the tumorigenicity of U-87MG cells in nude mice by 10% to 30% and extended the latency period for the development of subcutaneous palpable tumors by 31% to 56%, as compared with the controls. Exposure of U-87MG cells to GH-RH antagonists in vitro also resulted in a time-dependent increase in the mRNA levels of IGFR-II or a decrease in the mRNA levels of IGFR-I. mRNA for GH-RH was detected in U-87MG cells and xenografts implying that GH-RH may play a role in the pathogenesis of this tumor. Our results suggest that GH-RH antagonists MZ-5-156 and JV-1-36 inhibit the growth of U-87MG human glioblastoma by mechanisms that involve the suppression of IGF system. Antagonistic analogs of GH-RH merit further development for the treatment of malignant glioblastoma.
Collapse
Affiliation(s)
- H Kiaris
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112-1262, USA
| | | | | |
Collapse
|
47
|
Kiaris H, Schally AV, Varga JL, Groot K, Armatis P. Growth hormone-releasing hormone: an autocrine growth factor for small cell lung carcinoma. Proc Natl Acad Sci U S A 1999; 96:14894-8. [PMID: 10611309 PMCID: PMC24744 DOI: 10.1073/pnas.96.26.14894] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/1999] [Indexed: 11/18/2022] Open
Abstract
Antagonists of growth hormone-releasing hormone (GHRH) inhibit the growth of various cancers in vivo. This effect is thought to be exerted through suppression of the pituitary growth hormone-hepatic insulin-like growth factor I (IGF-I) axis and direct inhibition of autocrine/paracrine production of IGF-I and -II in tumors. However, other evidence points to a direct effect of GHRH antagonists on tumor growth that may not implicate IGFs, although an involvement of GHRH in the proliferation of cancer cells has not yet been established. In the present study we investigated whether GHRH can function as an autocrine/paracrine growth factor in small cell lung carcinoma (SCLC). H-69 and H-510A SCLC lines cultured in vitro express mRNA for GHRH, which apparently is translated into peptide GHRH and then secreted by the cells, as shown by the detection of GHRH-like immunoreactivity in conditioned media from the cells cultured in vitro. In addition, the levels of GHRH-like immunoreactivity in serum from nude mice bearing H-69 xenografts were higher than in tumor-free mice. GHRH(1-29)NH(2) stimulated the proliferation of H-69 and H-510A SCLCs in vitro, and GHRH antagonist JV-1-36 inhibited it. JV-1-36 administered s.c. into nude mice bearing xenografts of H-69 SCLC reduced significantly (P < 0.05) tumor volume and weight, after 31 days of therapy, as compared with controls. Collectively, our results suggest that GHRH can function as an autocrine growth factor in SCLCs. Treatment with antagonistic analogs of GHRH may offer a new approach to the treatment of SCLC and other cancers.
Collapse
Affiliation(s)
- H Kiaris
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, New Orleans, LA 70112, USA
| | | | | | | | | |
Collapse
|