Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Steroids ; 65(10-11): 825-30, 2000.
Article in English | MEDLINE | ID: mdl-11108894

ABSTRACT

Progesterone antagonists (PAs) (antiprogestins) or progesterone receptor modulators (PRMs) form an interesting category of new hormonal agents in the treatment of breast cancer. In vitro, antiproliferative effects of different PAs are mainly observed in estrogen-stimulated growth of PR-positive tumor cell lines. Both progestin agonist/antagonist actions on mammary tumor cells are dependent on the type of cell line, culture medium and concentrations of the PAs used, and type of biologic response measured. In various experimental animal tumor models, different PAs showed a greater antitumor activity than tamoxifen or high-dose progestins. Most interestingly, combination treatment of different PAs (mifepristone, ORG 31710, onapristone) or PRMs with different antiestrogens (tamoxifen, droloxifen, ICI 164384) or with an aromatase inhibitor (atemestane) showed greater antitumor efficacy than treatment with each single type of drug alone. These additive antiproliferative effects were demonstrated in various experimental in vitro and in vivo models. In some studies, these effects were accompanied by additive effects on several cell biologic parameters. In pretreated postmenopausal patients with metastatic breast cancer, objective responses have been observed in 10-12%, and stable disease in 42-46% of the patients; in previously untreated patients objective response rates of 11 and 56% have been reported. The clinical development of onapristone was stopped because of liver toxicity. At the present time, apart from development of new pure potent PAs, clinical investigation of combined therapy of PAs with antiestrogens are urgently needed.


Subject(s)
Breast Neoplasms/drug therapy , Hormone Antagonists/therapeutic use , Progesterone/antagonists & inhibitors , Receptors, Progesterone/metabolism , Clinical Trials as Topic , Female , Humans , Receptors, Progesterone/agonists , Receptors, Progesterone/antagonists & inhibitors , Selective Estrogen Receptor Modulators/therapeutic use , Tumor Cells, Cultured
2.
Int J Cancer ; 76(6): 829-35, 1998 Jun 10.
Article in English | MEDLINE | ID: mdl-9626349

ABSTRACT

The urokinase-type plasminogen activator (uPA) may be considered as a key enzyme in the processes of cancer cell invasion and metastasis. Evidence has been presented that, in breast stroma, uPA is expressed predominantly by myofibroblasts located at the invasive areas of the tumor. To examine whether transforming growth factor type-1 (TGF beta(1)) produced by breast-carcinoma cells is a candidate responsible for the induction of uPA-producing myofibroblasts, we studied in vitro the capacity of normal and tumor-derived human breast fibroblasts to express uPA and the myofibroblast marker alpha-smooth-muscle actin in response to TGF beta(1). Next, we compared these influences with those elicited by factor(s) released by epithelial-cancer cells. In all 8 fibroblast strains tested, TGF beta(1) induced a similar concentration-dependent increase in the fraction of alpha-smooth-muscle-actin-positive fibroblasts. While uPA expression was decreased by TGF beta(1) in most of the fibroblast strains, 2 strains were relatively insensitive to TGF beta(1) in this respect. Although factors present in media conditioned by non-uPA-producing epithelial-tumor cells could trigger fibroblasts to become potent producers of uPA, the TGF beta(1) content of the conditioned media were linked to the differential effects of externally added TGF beta(1) with respect to uPA expression. The data demonstrate that, although fibroblasts may utilize TGF beta(1) secreted by tumor cells to differentiate into myofibroblasts, tumor cells secrete factor(s) other than TGF beta(1) ultimately responsible for the generation of powerful uPA-producing fibroblasts.


Subject(s)
Breast Neoplasms/metabolism , Breast/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Transforming Growth Factor beta/pharmacology , Urokinase-Type Plasminogen Activator/biosynthesis , Cells, Cultured , Female , Fibroblasts/metabolism , Humans , RNA, Messenger/analysis , Urokinase-Type Plasminogen Activator/genetics
3.
J Nucl Med ; 37(1): 108-17, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8543978

ABSTRACT

UNLABELLED: We evaluated the potential usefulness of a new radiolabeled substance P (SP) analog, [111In-DTPA-Arg1]SP, as a radiopharmaceutical for the in vivo detection of SP receptor-positive (SPR+) immunologic disorders (i.e., inflammatory bowel disease and arthritis) and tumors (i.e., carcinoid). METHODS: Substance P, [DTPA-Arg1]SP and [3-(p-hydroxyphenyl)propionyl-Arg1]SP (Bolton-Hunter-SP, [BH-SP]) were tested as competitors for 125I-BH-SP to SPR in rat brain cortex membranes. An autoradiographic displacement study of the submandibular gland of the rat with the 125I-BH-SP as radioligand and [DTPA-Arg1]SP as competitor was performed. Tissue distribution and ex vivo autoradiography were studied in rats, with and without pretreatment with the selective nonpeptide antagonist CP96,345 to quantify specific binding. In vivo metabolism of [111In-DTPA-Arg1]SP was performed in control rats. Gamma-camera scintigraphic studies were carried out with control rats to visualize the SPR+ salivary glands in rats bearing the SPR+ transplantable pancreatic tumor CA20948 and in rats with SPR+ adjuvant arthritic joints, which was induced after injection of a homogenate of Mycobacterium tuberculosis. RESULTS: Substance P, [DTPA-Arg1]SP and BH-SP dose-dependently inhibited binding of 125I-BH-SP to SPR in rat brain cortex membranes with IC50 values of 0.2, 4 and 2 nM, respectively. In an autoradiographic displacement study of the submandibular gland with 125I-BH-SP as radioligand, an IC50 of 2.7 nM was found for [DTPA-Arg1]SP. In vivo metabolism of the radiopharmaceutical in the rat revealed a renal clearance rate of 50% of the injected radioactive dose in 30 min and a rapid enzymatic degradation of the radiopharmaceutical, resulting in an effective half-life of the intact radiopharmaceutical in blood of approximately 3 min. Tissue distribution and ex vivo autoradiographic studies in rats showed uptake and specific binding of radioactivity in isolated tumors and submandibular and parotid glands. Optimum SPR+ target-to-background ratios were found 24 hr after injection of [111In-DTPA-Arg1]SP. Visualization of normal SPR+ tissues, such as the salivary glands by gamma camera scintigraphy, after administration of [111In-DTPA-Arg1]SP was demonstrated in untreated rats. Pathological SPR+ processes were visualized both in rats bearing the transplantable pancreatic tumor CA20948 and in those with adjuvant mycobacteria tuberculosis-induced arthritic joints. CONCLUSION: [Indium-111-DTPA-Arg1]SP can be used successfully to visualize SPR+ processes in vivo by gamma camera scintigraphy.


Subject(s)
Arthritis, Experimental/diagnostic imaging , Indium Radioisotopes , Pancreatic Neoplasms/diagnostic imaging , Parotid Gland/diagnostic imaging , Pentetic Acid/analogs & derivatives , Receptors, Neurokinin-1/analysis , Submandibular Gland/diagnostic imaging , Substance P/analogs & derivatives , Animals , Biphenyl Compounds/pharmacology , Female , Male , Neurokinin-1 Receptor Antagonists , Parotid Gland/metabolism , Pentetic Acid/pharmacokinetics , Radionuclide Imaging , Rats , Rats, Inbred Lew , Rats, Wistar , Submandibular Gland/metabolism , Substance P/pharmacokinetics , Tissue Distribution
4.
Acta Oncol ; 35 Suppl 5: 30-7, 1996.
Article in English | MEDLINE | ID: mdl-9142962

ABSTRACT

Endocrine therapy of breast cancer consists of a variety of both medical and surgical ablative treatment modalities, but ablative therapy is increasingly replaced by medical treatment. Most endocrine therapies have more than one endocrine effect, frequently together with direct growth inhibitory actions via receptors. Endocrine therapy can be effective in all phases of the disease, but curative only in early disease while in advanced cancer it can only prolong survival. In the past decade the number of available endocrine agents has been drastically increased. Novel approaches in the endocrine therapy of breast cancer are application of new antiestrogens, antiprogestins, new potent aromatase inhibitors, analogues of luteinizing hormone-releasing hormone (LHRH-A) and somatostatin, inhibitors of prolactin secretion, vitamin A and D analogues, bisphosphonates, growth factor antagonists, tyrosine protein kinase inhibitors, protease inhibitors, inhibitors of angiogenesis, radiolabeled hormones and monoclonal antibodies. New cell biological factors such as oncogenes and suppressorgenes, secretory proteins and membrane receptors can be used not only as prognostic factors but also for prediction of type of response to endocrine and chemotherapy. Thus, these cell biological parameters can be used to select high and low risk patients, type of systemic treatment, and can also be used as targets for new treatment modalities. Future studies on treatment of all stages of disease will increasingly focus on promising combined treatment modalities.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Neoplasms, Hormone-Dependent/drug therapy , Antineoplastic Agents, Hormonal/pharmacology , Aromatase Inhibitors , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Estrogen Antagonists/pharmacology , Estrogen Antagonists/therapeutic use , Gonadotropin-Releasing Hormone/analogs & derivatives , Humans , Insulin-Like Growth Factor I , Neoplasm Metastasis , Neovascularization, Pathologic , Progesterone Congeners/pharmacology , Progesterone Congeners/therapeutic use , Progestins/antagonists & inhibitors , Prolactin/antagonists & inhibitors , Retinoids/pharmacology , Retinoids/therapeutic use , Somatostatin/analogs & derivatives , Transforming Growth Factor beta , Vitamin D/analogs & derivatives
5.
Eur J Nucl Med ; 22(7): 608-16, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7498221

ABSTRACT

The characteristics of terbium-161 diethylene triamine penta-acetic acid (DTPA) labelled octreotide with respect to specific binding to somatostatin (octreotide) receptors on rat brain cortex membranes, biological activity, uptake and excretion by isolated perfused rat livers and metabolism in vivo in normal and tumour-bearing rats were determined and compared to those of indium-111 DTPA-octreotide. The results of the binding studies demonstrate that 161Tb-DTPA-octreotide is a high-affinity radioligand for somatostatin receptors, with an affinity comparable to that of 111In-DTPA-octreotide. Rat growth hormone secretion inhibition experiments showed that 161Tb-DTPA-octreotide has a similar potency to 111In-DTPA-octreotide. 161Tb-DTPA-octreotide appeared to be taken up even less by the isolated perfused rat liver than 111In-DTPA-octreotide, as almost no tracer disappeared from the perfusion medium. Furthermore, hardly any radioactivity was found in the liver, and excretion into the bile was negligible. The biodistribution studies showed that for octreotide receptor-positive organs, such as pancreas and adrenals, uptake of 161Tb-DTPA-octreotide is lower then that of 111In-DTPA-octreotide. However, as the clearance from the blood of the former compound is faster than that of the latter, the tissue/blood ratio is higher in the case of 161Tb-DTPA-octreotide than with 111In-DTPA-octreotide. Furthermore, these studies demonstrated that the uptake of 161Tb-DTPA-octreotide by the renal tubular cells after glomerular filtration can be reduced by administration of lysine or sodium maleate. Increase in urine production before and during the experiment had no effect on the kidney uptake of 161Tb-DTPA-octreotide.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Octreotide/analogs & derivatives , Pentetic Acid/analogs & derivatives , Radioisotopes/therapeutic use , Terbium/therapeutic use , Animals , Brain/metabolism , Kidney/metabolism , Liver/metabolism , Male , Octreotide/pharmacokinetics , Octreotide/therapeutic use , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/radiotherapy , Pentetic Acid/pharmacokinetics , Pentetic Acid/therapeutic use , Rats , Rats, Inbred Lew , Rats, Wistar , Tissue Distribution
6.
Hum Reprod ; 9 Suppl 1: 181-9, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7962463

ABSTRACT

Antiprogestins form a new potential treatment modality for breast cancer and their mode of action has been assessed in vitro on several breast cancer cell lines, in vivo in rats with dimethylbenzanthracene (DMBA)-induced mammary tumours and in vivo in patients with metastatic breast cancer. In vitro in serum-free medium, the progestin Org 2058 and antiprogestins RU486 and Org 31710 caused a dose-dependently stimulated MCF7 cell growth. Both antiprogestins dose-dependently inhibited the oestrogen-stimulated proliferation of progesterone receptor (PgR)-rich T-47D cells in DCC medium. Inhibition by Org 31710 plateaued at 10(-8) M (74% inhibition), compared with RU486 at up to 10(-6) M (53% inhibition). No inhibition was observed at doses of 10(-12)-10(-6) M of both antiprogestins in the absence of oestradiol. The proliferation of the ZR-75.1 and MDA-MB-231 cell lines was not or only marginally affected by either antiprogestin. Rats with DMBA-induced mammary tumours given prophylactic treatment with RU486 displayed a doubled latency period. Antiprogestins were slightly more effective than tamoxifen or progestins in rats with existing tumours. Org 31710 sometimes showed a somewhat more pronounced inhibitory effect than the antiprogestins Org 31806 and RU486. Combined antiprogestational and anti-oestrogenic treatment showed striking additive growth inhibitory effects resulting in clear tumour remissions, in the presence of very strong suppression of oestrogen and PgRs. The growth inhibitory effect of luteinizing hormone-releasing hormone agonists was potentiated by antiprogestins.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Breast Neoplasms/drug therapy , Hormone Antagonists/therapeutic use , Progestins/antagonists & inhibitors , 9,10-Dimethyl-1,2-benzanthracene , Animals , Cell Division/drug effects , Clinical Trials as Topic , Drug Screening Assays, Antitumor , Estradiol/metabolism , Estradiol/pharmacology , Female , Glucocorticoids/antagonists & inhibitors , Gonadotropin-Releasing Hormone/analogs & derivatives , Hormone Antagonists/adverse effects , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/drug therapy , Neoplasms, Hormone-Dependent/drug therapy , Pituitary-Adrenal System/drug effects , Postmenopause , Rats , Salvage Therapy , Tumor Cells, Cultured
7.
Eur J Nucl Med ; 21(4): 328-35, 1994 Apr.
Article in English | MEDLINE | ID: mdl-7911760

ABSTRACT

We have evaluated the potential usefulness of indium-111 labelled [DTPA-D-Phe1]RC-160, derived from the octapeptide somatostatin analogue RC-160, as a radiopharmaceutical for the in vivo detection of somatostatin receptor-positive tumours. For this purpose 111In-and 115In-labelled [DTPA-D-Phe1]RC-160 was tested for its biological activity, and applied for somatostatin receptor scintigraphy in vivo to rats bearing the transplantable rat pancreatic tumour CA20948, which expresses somatostatin receptors. We previously described the development of the 111In-labelled somatostatin analogue [DTPA-D-Phe1]octreotide and its use in the in vivo visualization of somatostatin receptor-positive tumours in rats and in humans. Like [111In-DTPA-D-Phe1]octreotide, [111In-DTPA-D-Phe1]RC-160 showed uptake in and specific binding in vivo to somatostatin receptor-positive organs and tumours, and the tumours were clearly visualized by gamma camera scintigraphy. However, as compared to [111In-DTPA-D-Phe1]octreotide, blood radioactivity (background) was higher, resulting in a lower tumour to blood (background) ratio. Using this animal model we therefore conclude that [111In-DTPA-D-Phe1]RC-160 has no advantage over [111In-DTPA-D-Phe1]octreotide as a radiopharmaceutical in the visualization of somatostatin receptors which bind both analogues. However, recent reports suggest the existence of different somatostatin receptor subtypes on some human cancers, which differentially bind RC-160 and not octreotide. These tumours include cancers of the breast, ovary, exocrine pancreas, prostate and colon. [111In-DTPA-D-Phe1]RC-160 might be of interest for future use in such cancer patients as a radiopharmaceutical for imaging somatostatin receptor-positive tumours, which do not bind octreotide.


Subject(s)
Indium Radioisotopes , Octreotide/analogs & derivatives , Pancreatic Neoplasms/diagnostic imaging , Pentetic Acid/analogs & derivatives , Somatostatin/analogs & derivatives , Animals , Male , Pancreatic Neoplasms/chemistry , Radionuclide Imaging , Rats , Rats, Inbred Lew , Receptors, Somatostatin/analysis , Tissue Distribution
8.
Eur J Nucl Med ; 20(11): 1089-94, 1993 Nov.
Article in English | MEDLINE | ID: mdl-7904561

ABSTRACT

We have evaluated the potential usefulness of the radioiodinated octapeptide RC-160, a somatostatin analogue, which might serve as a radiopharmaceutical for the in vivo detection of somatostatin receptor-positive tumours. For this purpose, iodine-123 and iodine-125 labelled RC-160 was tested for biological activity and applied in vivo in rats bearing the transplantable rat pancreatic tumour CA20948, which expresses somatostatin receptors. Our group has recently described the in vivo visualization of such tumours in rats and in humans with the radioiodinated somatostatin analogue [Tyr3]octreotide. Like [123I-Tyr3]octreotide, 123I-RC-160 showed uptake in and specific binding in vivo to somatostatin receptor-positive organs and tumors. However, blood radioactivity (background) was higher, resulting in a lower tumour to blood (background) ratio. We therefore conclude that in this animal model 123I-RC-160 has no advantage over [123I-Tyr3]octreotide as a radiopharmaceutical for the in vivo use as a somatostatin receptor imager, although, like [123I-Tyr3]octreotide, 123I-RC-160 shows specific binding to different somatostatin receptor-positive organs. Recently differences were reported in affinity between somatostatin and its analogues for somatostatin receptors expressed in different human cancers, like those of the breast, ovary, exocrine pancreas, prostate and colon. Therefore 123I-RC-160 might be of interest for future use in humans as a radiopharmaceutical for imaging octreotide receptor-negative tumours.


Subject(s)
Antineoplastic Agents , Pancreatic Neoplasms/diagnostic imaging , Somatostatin/analogs & derivatives , Animals , Iodine Radioisotopes , Male , Octreotide/analogs & derivatives , Radionuclide Imaging , Rats , Rats, Inbred Lew , Receptors, Somatostatin
9.
Life Sci ; 49(22): 1593-601, 1991.
Article in English | MEDLINE | ID: mdl-1658516

ABSTRACT

Radioiodinated somatostatin analogues are useful ligands for the in vitro and in vivo detection of somatostatin receptors. [111In-DTPA-D-Phe1]-octreotide, a somatostatin analogue labeled with a different radionuclide, also binds specifically to somatostatin receptors in vitro. In this study we investigated its in vivo application in the visualization of somatostatin receptor-positive tumors in rats. The distribution of the radiopharmaceutical was investigated after intravenous injection in normal rats and in rats bearing the somatostatin receptor-positive rat pancreatic carcinoma CA 20948. After injection the radiopharmaceutical was rapidly cleared (50% decrease in maximal blood radioactivity in 4 min), predominantly by the kidneys. Excreted radioactivity was mainly in the form of the intact radiopharmaceutical. Ex vivo autoradiographic studies showed that specific accumulation of radioactivity occurred in somatostatin receptor-containing tissue (anterior pituitary gland). However, in contrast to the adrenals and pituitary, the tracer accumulation in the kidneys was not mediated by somatostatin receptors. Increasing radioactivity over the somatostatin receptor-positive tumors was measured rapidly after injection and the tumors were clearly visualized by gamma camera scintigraphy. In rats pretreated with 1 mg octreotide accumulation of [111In-DTPA-D-Phe1]-octreotide in the tumors was prevented. Because of its relatively long effective half-life, [111In-DTPA-D-Phe1]-octreotide is a radionuclide-coupled somatostatin analogue which can be used to visualize somatostatin receptor-bearing tumors efficiently after 24 hr, when interfering background radioactivity is minimized by renal clearance. This is an advantage over the previously used [123I-Tyr3]-octreotide which has a shorter effective half-life and shows high abdominal interference due to its hepato-biliary clearance. Therefore, [111In-DTPA-D-Phe1]-octreotide seems a better alternative for scintigraphic imaging of somatostatin receptor-bearing tumors.


Subject(s)
Indium Radioisotopes , Pancreatic Neoplasms/diagnostic imaging , Receptors, Neurotransmitter/analysis , Animals , Autoradiography , Half-Life , Kidney/metabolism , Male , Pancreatic Neoplasms/metabolism , Radionuclide Imaging , Rats , Rats, Inbred Lew , Receptors, Somatostatin
10.
J Steroid Biochem Mol Biol ; 37(6): 789-94, 1990 Dec 20.
Article in English | MEDLINE | ID: mdl-2285592

ABSTRACT

UNLABELLED: Treatment with antiprogestins is a new treatment modality for breast cancer. Previously, in rats with DMBA-induced mammary tumors we observed significant growth inhibitory effects of chronic treatment with the antiprogestin mifepristone (RU486). In addition, in 11 postmenopausal breast cancer patients, we observed one objective response, six instances of short-term stable disease, and four instances of progressive disease. Side-effects appeared mainly due to antiglucocorticoid properties of the drug. Increased plasma estradiol levels were observed which probably resulted from ovarian (rat) and adrenal (patients) steroidogenesis. Combined treatment with an antiestrogen in the rat model caused additive growth inhibitory effects. Tumor inhibition after single treatment with mifepristone or tamoxifen was 90 and 75%, respectively. In contrast, when combined, tumor remission similar to that caused by LHRH-agonist treatment (50%) was observed. Even higher tumor remission was found after combined treatment with mifepristone plus LHRH-agonist (75%). In first studies in the rat model we observed significant tumor growth inhibitory effects with two new antiprogestins of seemingly greater potency which cause less unfavorable endocrine side-effects. IN CONCLUSION: combined treatment (antiprogestin plus antiestrogen or LHRH-agonist) may be of value in endocrine therapy of breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Mifepristone/pharmacology , Tamoxifen/pharmacology , Adult , Aged , Animals , Disease Models, Animal , Drug Therapy, Combination , Gonadotropin-Releasing Hormone/pharmacology , Hormones/blood , Humans , Middle Aged , Organ Size , Rats , Receptors, Steroid/metabolism
11.
J Steroid Biochem Mol Biol ; 37(6): 1089-95, 1990 Dec 20.
Article in English | MEDLINE | ID: mdl-1981013

ABSTRACT

UNLABELLED: Interference in growth factor mediated pathways is a new strategy in the treatment of cancer. Somatostatin analogs can inhibit hormone and growth factor secretion, while suramin can block the binding of several growth factors to their receptors. In addition, somatostatin analogs can cause direct growth inhibitory effects after binding to tumoral somatostatin receptors. We tested the efficacy and endocrine effects of chronic treatment with three somatostatin analogs (Sandostatin, R RC-160 and CGP 15-425) or suramin in several tumor models and in patients with various types of cancer. Treatment with somatostatin analogs caused growth inhibition of breast cancer cells (MCF-7) in vitro, and of rat transplantable pancreatic (50-70% inhibition) and prostatic Dunning tumors (12% inhibition). No tumor growth inhibition was observed with respect to DMBA-induced rat mammary tumors, a transplantable colon tumor and a rhabdomyosarcoma in rats. In 34 patients with metastatic pancreatic or gastrointestinal adenocarcinomas chronic Sandostatin treatment caused stable disease in 27% of the patients, but no objective remissions. Somatostatin receptors were found in the responding MCF-7 mammary tumor cells, rat pancreatic tumors and in 20-45% of human breast cancer specimens [J. Steroid Biochem. Molec. Biol. 37 (1990) 1073-1077], but not in rat DMBA-mammary tumors or in 10 human pancreatic adenocarcinomas. Suramin caused significant dose-dependent growth inhibition of human breast cancer cells in vitro and of rat pancreatic tumors in vivo in the presence of plasma levels up to 150 micrograms/ml. In a preliminary clinical study concerning 11 patients with various tumor types we observed significant hematological, biochemical, endocrine and clinical side effects, but no objective remissions in spite of relevant peak plasma suramin concentrations of 270-330 micrograms/ml. IN CONCLUSION: somatostatin analogs and suramin can cause growth inhibition of various experimental tumors in vitro and in vivo, but the clinical value has to be established for several types of cancer, especially with respect to suramin and suramin-like compounds.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Somatostatin/therapeutic use , Suramin/therapeutic use , Clinical Trials as Topic , Humans , Somatostatin/analogs & derivatives
12.
Breast Cancer Res Treat ; 17(1): 23-32, 1990 Nov.
Article in English | MEDLINE | ID: mdl-1965705

ABSTRACT

The effects of treatment with a somatostatin analog (Sandostatin, SMS201-995) were investigated in female rats with dimethylbenzanthracene (DMBA)-induced rat mammary tumors. A 3-week treatment was performed using sandostatin, the LHRH-agonist buserelin alone, or buserelin in combination with sandostatin. Twice daily sandostatin treatment was performed with dosages of 0.05 microgram, 0.2 microgram, 1 microgram, 5 micrograms, and 20 micrograms. Buserelin was used in a 2 x 5 micrograms/day dosage. The combined results from six different experiments show that the various dosages of sandostatin caused no tumor growth inhibition. Somatostatin receptors could not be demonstrated in these mammary tumors. Sandostatin treatment by daily injections did not suppress levels of growth hormone, prolactin, or epidermal growth factor-like activities. Estrogen (ER) and progesterone (PgR) receptor contents of the mammary tumors were not changed. In contrast, buserelin treatment caused highly significant tumor remission. The combined treatment with sandostatin and buserelin did not alter the treatment results obtained after treatment with buserelin alone. In conclusion, sandostatin treatment in this tumor model had no direct growth inhibitory effect and did not cause an endocrine inhibition of mammary tumor growth. However, these results do not exclude antitumor effects in human breast cancer in view of the presence of somatostatin receptors in approximately 20-45% of human tumors, besides possible different endocrine effects.


Subject(s)
Mammary Neoplasms, Experimental/drug therapy , Octreotide/therapeutic use , 9,10-Dimethyl-1,2-benzanthracene , Animals , Buserelin/administration & dosage , Buserelin/therapeutic use , Drug Screening Assays, Antitumor , Female , Mammary Neoplasms, Experimental/chemically induced , Octreotide/administration & dosage , Pituitary Hormones, Anterior/blood , Rats , Rats, Inbred Strains , Receptors, Neurotransmitter/analysis , Receptors, Somatostatin , Receptors, Steroid/analysis
13.
Endocrinology ; 125(3): 1593-8, 1989 Sep.
Article in English | MEDLINE | ID: mdl-2527151

ABSTRACT

Rats bearing mammary tumors induced with dimethylbenzanthracene were treated with the antiprogestin mifepristone (RU486; 10 mg/kg.day, sc), the antiestrogen tamoxifen (400 micrograms/kg.day, sc), LHRH agonists administered by either sc injections (buserelin; 40 micrograms/kg.day) or implant (buserelin or zoladex), or combinations of mifepristone and tamoxifen or LHRH agonists. Single treatment with mifepristone or tamoxifen caused a significant inhibition of tumor growth (90% and 75%, respectively), but no tumor remission. In contrast, single treatment with LHRH agonists caused remission of mammary tumor growth by 50% (injection) or 70% (implant), respectively. Combined treatment with mifepristone and tamoxifen caused additive tumor growth inhibitory effects resulting in the same extent of tumor remission as that observed after treatment with LHRH agonist injections alone. Combination of mifepristone with either manner of LHRH agonist administration resulted in the highest tumor remission (approximately 75%). Significant reductions in cytosolic steroid (estrogen and progesterone) receptor contents of mammary tumors were noted after various treatment modalities. The most pronounced decrements were observed after combined treatment with mifepristone and tamoxifen (residual estrogen receptor; 10%; residual progesterone receptor, 0%). On the other hand, suppression of pituitary-ovarian function was most pronounced after treatment with LHRH agonist implants alone or in combination with mifepristone. It is concluded that combination treatment with an antiprogestin and an antiestrogen or an LHRH agonist may be of great value in the endocrine therapy of breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Buserelin/analogs & derivatives , Buserelin/therapeutic use , Estrenes/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Progestins/antagonists & inhibitors , Tamoxifen/therapeutic use , 9,10-Dimethyl-1,2-benzanthracene , Animals , Buserelin/administration & dosage , Cytosol/analysis , Drug Implants , Estrenes/administration & dosage , Female , Follicle Stimulating Hormone/blood , Goserelin , Injections, Subcutaneous , Luteinizing Hormone/blood , Mammary Neoplasms, Experimental/analysis , Mammary Neoplasms, Experimental/blood , Mifepristone , Rats , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Tamoxifen/administration & dosage
14.
Cancer Treat Rep ; 71(11): 1021-7, 1987 Nov.
Article in English | MEDLINE | ID: mdl-3119199

ABSTRACT

The effects of the synthetic antiprogestin mifepristone (RU486) on growth of dimethylbenzanthracene (DMBA)-induced mammary tumors in female rats were investigated. Prophylactic treatment with mifepristone (10 mg/kg/day) for 3 weeks starting from the day of first DMBA injection resulted in a doubling of the average tumor latency period (81 +/- 16 days, n = 17 treated, versus 39 +/- 5 days, n = 75 controls; P less than 0.005) and was accompanied by a significant growth retardation as shown by lower body weight increments. A 3-week therapeutic treatment of rats bearing mammary tumors was performed by administration of different dosages of mifepristone (2.5, 10, or 40 mg/kg/day) or megestrol acetate (2.5 or 10 mg/kg/day), with the luteinizing hormone-releasing hormone agonist buserelin (40 micrograms/kg/day), buserelin plus mifepristone (10 mg/kg/day), or by ovariectomy. The effects of treatment on tumor load, pituitary, adrenal and reproductive organ weights, steroid receptor contents of mammary tumors, and blood plasma hormone concentrations were investigated. Mifepristone and megestrol acetate treatment gave rise to inhibition of mammary tumor growth with all dosages studied, in which mifepristone was more potent than megestrol acetate (80%-90% vs 40% inhibition, P less than 0.01). In contrast, buserelin treatment and ovariectomy resulted not only in inhibition, but in tumor remission by about 50%. Combined treatment with buserelin and mifepristone gave the same tumor remission as resulted from ovariectomy or single treatment with buserelin. Estradiol-stimulated growth of the human mammary cancer MCF-7 cells in culture was fully abolished by mifepristone (3.6 X 10(-8) M) or tamoxifen (4 X 10(-8) M), whereas growth of MCF-7 cells under control incubation was not affected by either agent. Therefore, a direct inhibition of the growth of rat mammary tumor cells by mifepristone appears likely. Based on the effects of mifepristone on plasma hormone levels (increased: luteinizing hormone, estradiol, progesterone; unchanged: follicle-stimulating hormone, adrenocorticotropic hormone, corticosterone), organ weights (increased: pituitary, ovaries, uterus; unchanged: adrenals) and steroid receptor contents of mammary tumors (decreased: estrogen receptor and progesterone receptor contents), the main mechanism of action is probably a direct antiprogestational effect at the level of the mammary tumor cells through occupancy of the progesterone receptor.


Subject(s)
Buserelin/therapeutic use , Estrenes/therapeutic use , Mammary Neoplasms, Experimental/therapy , Megestrol/analogs & derivatives , Ovariectomy , Animals , Body Weight/drug effects , Combined Modality Therapy , Female , Follicle Stimulating Hormone/blood , Luteinizing Hormone/blood , Megestrol/therapeutic use , Megestrol Acetate , Mifepristone , Rats , Rats, Inbred Strains , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis
15.
Cancer Res ; 47(6): 1566-70, 1987 Mar 15.
Article in English | MEDLINE | ID: mdl-2880660

ABSTRACT

Various hormones and growth factors are involved in the growth regulation of breast (tumor) cells. In this report we show for the first time that an analogue of the neuropeptide somatostatin (Sandostatin) can also influence the proliferation of human breast cancer cells (MCF-7), namely, in an inhibitory fashion. With respect to dose-response relationship a bell-shaped curve was observed with the maximal inhibition of tumor cell growth at a sharply defined amount of Sandostatin (10 nM). The same effects were found with the natural hormone somatostatin-14 and another analogue (CGP 15-425). These results, together with the observation that high affinity binding sites for an iodinated derivative of Sandostatin are present in MCF-7 cells, support the conclusion that somatostatin and analogues act directly on breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , Somatostatin/pharmacology , Cell Division/drug effects , Cell Line , Dose-Response Relationship, Drug , Female , Humans , Receptors, Neurotransmitter/analysis , Receptors, Somatostatin , Somatostatin/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL
...