Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
3.
Br J Cancer ; 113(2): 290-8, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26057454

ABSTRACT

BACKGROUND: Breast cancer clinical outcome is affected by tumor molecular features, and the identification of subtype-specific prognostic biomarkers is relevant for breast cancer translational research. Gene expression signatures proved to be able to complement prognostic information provided by classical clinico-pathological features. Recently, microRNAs (miRNAs) have been causally linked to tumorigenesis and cancer progression and have been associated with patient outcome, also in breast cancer. METHODS: MicroRNAs associated with the development of distant metastasis were identified in a cohort of 92 ESR1+/ERBB2- lymph node-negative breast cancers from patients not receiving adjuvant treatment. Results were confirmed and further investigated in a total of 1246 miRNA and gene expression profiles of the Molecular Taxonomy of Breast Cancer International Consortium data set. Moderated t-test, univariable and multivariable Cox regression models were used for statistical analyses. RESULTS: miR-30e* was identified as independent protective prognostic factor in lymph node-negative untreated patients with ESR1+/ERBB2- tumours and retained a significant association with a good prognosis in treated patients with the same tumor subtype as well as in the ERBB2+ subtype, but not in ESR1-/ERBB2- tumours. CONCLUSIONS: We highlighted a relevant and subtype-specific role in breast cancer for miR-30e* and demonstrated that adding miRNA markers to gene signatures and clinico-pathological features can help for a better prognostication.


Subject(s)
Breast Neoplasms/genetics , MicroRNAs/physiology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Estrogen Receptor alpha/analysis , Female , Humans , Lymphatic Metastasis , MicroRNAs/analysis , Prognosis , Proportional Hazards Models , Receptor, ErbB-2/analysis , Transcriptome
4.
Cell Mol Life Sci ; 60(3): 567-76, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12737316

ABSTRACT

In the last decades, substances with estrogenic activity have been dispersed into the environment. Xenoestrogens act by binding to estrogen receptors, ligand-regulated transcription factors, for which two subtypes have been described, ER-alpha and ER-beta, which are often coexpressed at variable amounts in different tissues. We investigated variations in the expression of ER-alpha and ER-beta mRNAs following treatment with four xenoestrogens (bisphenol A, 4-tert octylphenol, 2-hydroxybiphenyl, 4-hydroxybiphenyl) and with 17beta-estradiol in estrogen-sensitive (T47D) and estrogen-insensitive (BT20) breast cancer cell lines. Although to a variable extent, both estradiol and the tested xenoestrogens increased the expression of ER-beta mRNA, whereas a slight effect on ER-alpha was observed only in T47D cells. Upregulation of ER-beta expression by estradiol and xenoestrogens was observed only in the presence of detectable ER-alpha protein levels. These findings indicate a regulatory role for ER-beta in ER-alpha-mediated transcription and a role for ER-beta in mediating xenoestrogen toxicity.


Subject(s)
Breast Neoplasms/metabolism , Estradiol/pharmacology , Receptors, Estrogen/drug effects , Benzhydryl Compounds , Carcinogens, Environmental/pharmacology , Estrogen Receptor beta , Estrogens, Non-Steroidal/pharmacology , Female , Humans , In Vitro Techniques , Phenols/pharmacology , Tumor Cells, Cultured
5.
Br J Cancer ; 85(6): 795-7, 2001 Sep 14.
Article in English | MEDLINE | ID: mdl-11556826

ABSTRACT

The prognostic contribution of intratumour VEGF, the most important factor in tumour-induced angiogenesis, to NPI was evaluated by using flexible modelling in a series of 226 N-primary breast cancer patients in which steroid receptors and cell proliferation were also accounted for. VEGF provided an additional prognostic contribution to NPI mainly within ER-poor tumours.


Subject(s)
Breast Neoplasms/metabolism , Endothelial Growth Factors/metabolism , Lymphokines/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/mortality , Cell Division , Disease-Free Survival , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lymph Nodes , Prognosis , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Survival Rate , United Kingdom , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
6.
J Cell Biochem ; 79(4): 594-600, 2000 Sep 14.
Article in English | MEDLINE | ID: mdl-10996850

ABSTRACT

Genistein, a natural isoflavone phytoestrogen present in soybeans, caused a dose-dependent growth inhibition of the two hormone-sensitive cell lines T47D and ZR75.1 and of the two hormone-independent cell lines MDAMB-231 and BT20. Flow cytometric analysis of cells treated for 4 days with 15 and 30 microM genistein showed a dose-dependent accumulation in the G(2)M phase of the cell cycle. At the highest tested concentration, there was a sevenfold increase in the percentage of cells in G(2)M (63%) with respect to the control (9%) in the case of T47D cells and a 2.4-fold increase in the case of BT20. An intermediate fourfold accumulation was observed in the case of MDAMB-231 and ZR75.1. The G(2)M arrest was coupled with a parallel depletion of the G(0)/G(1) phase. To understand the mechanism of action underlying the block in G(2)M induced by genistein, we investigated the expression and the activity of cyclins and of cyclin-dependent kinases specifically involved in the G(2)-->M transition. As expected, p34(cdc-2) expression, monitored by Western blotting, was unaffected by genistein treatment in all cell lines. With exception of the T47D cell line, we revealed an increase in the tyrosine phosphorylated form of p34, suggesting an inactivation of the p34(cdc-2) catalytic activity consequent to treatment of cells with genistein. In fact, immunoprecipitates from genistein-treated MDAMB-231 and BT20 cells displayed a fourfold decrease in kinase activity evaluated using the histone H1 as substrate. Conversely, no variation in kinase activity was observed between treated and untreated ZR75.1 cells despite the increase in p34 phosphorylation. In cells treated with 30 microM genistein, cyclin B(1) (p62) increased 2.8-,8-and 103-fold, respectively, in BT20, MDAMB-231, and ZR75.1 cells, suggesting an accumulation of the p62, which is instead rapidly degraded in cycling cells. No effects were observed on cyclin expression in T47D cells. We therefore conclude that genistein causes a G(2)M arrest in breast cancer cell lines, but that such growth arrest is not necessarily coupled with deregulation of the p34(cdc-2)/cyclin B(1) complex only in all of the studied cell lines.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Cycle/drug effects , Genistein/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CDC2 Protein Kinase/metabolism , Female , Flow Cytometry , G2 Phase/drug effects , Humans , Maturation-Promoting Factor/chemistry , Maturation-Promoting Factor/metabolism , Mitosis/drug effects , Protein Subunits , Tumor Cells, Cultured
7.
Br J Cancer ; 80(8): 1150-5, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10376965

ABSTRACT

We compared the oestrogenic and anti-oestrogenic properties of the two well-known phyto-oestrogens, genistein and quercetin, on the oestrogen-sensitive breast cancer cell line MCF-7. Genistein exerted a biphasic effect on growth of MCF-7 cells, stimulating at low and inhibiting at high concentrations, whereas quercetin was only growth inhibitory. At doses which did not inhibit cell growth, respectively 5 and 1 microM, genistein and quercetin counteracted oestrogen- and transforming growth factor-alpha-promoted cell growth stimulation. Furthermore, genistein promoted transcription of the oestrogen-regulated genes pS2 and cathepsin-D, whereas quercetin interfered with the oestrogen-induced expression of the proteins. In in vitro binding experiments, genistein competed with oestradiol for binding to the oestrogen receptor (ER), but quercetin did not. Quercetin and genistein down-regulated cytoplasmic ER levels and promoted a tighter nuclear association of the ER, but only genistein was able to up-regulate progesterone receptor protein levels. In gel mobility assays, ER preincubation with oestradiol or with the two phyto-oestrogens led to the appearance of the same retarded band, excluding differences between the various complexes in binding to the consensus sequence. The data allowed us to conclude that quercetin acts like a pure anti-oestrogen, whereas genistein displays mixed agonist/antagonist properties, and to formulate a hypothesis on the possible mechanism of action of such phyto-oestrogens.


Subject(s)
Anticarcinogenic Agents/pharmacology , Breast Neoplasms/physiopathology , Gene Expression Regulation , Genistein/pharmacology , Quercetin/pharmacology , Receptors, Estrogen/drug effects , Amino Acid Sequence , Breast Neoplasms/pathology , Cathepsin D/biosynthesis , Down-Regulation , Female , Humans , Molecular Sequence Data , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/genetics , Tumor Cells, Cultured
8.
Cancer Lett ; 130(1-2): 143-52, 1998 Aug 14.
Article in English | MEDLINE | ID: mdl-9751267

ABSTRACT

Genistein significantly inhibited cell growth (IC50 around 10 microM) of MCF-7, MDAMB-231 and HBL-100 cell lines, but not of skin-derived fibroblasts and counteracted the growth-stimulatory effects exerted by estradiol and growth factors. It abolished the paracrine stimulation observed in MCF-7 cells in co-culture with MDAMB-231 or fibroblasts. Genistein-treated cells accumulated in the S and G2/M phases of the cell cycle and underwent apoptosis. Genistein decreased tyrosine phosphorylation induced upon treatment with transforming growth factor-alpha. Finally, genistein bound the estrogen receptor (ER) (relative affinity constant Kd = 4 nM), induced pS2 and cathepsin-D transcription and increased nuclear ER levels.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms , Genistein/pharmacology , Antineoplastic Agents/metabolism , Apoptosis , Binding, Competitive , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Division/drug effects , Estradiol/metabolism , Estradiol/pharmacology , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Genistein/metabolism , Growth Substances/pharmacology , Humans , Phosphorylation , Receptors, Estrogen/metabolism , Tumor Cells, Cultured/drug effects
9.
Anticancer Res ; 18(3A): 1703-8, 1998.
Article in English | MEDLINE | ID: mdl-9673393

ABSTRACT

BACKGROUND: 1,25-dihydroxycholecalciferol has been previously reported to negatively regulate human breast cancer cell growth. MATERIAL AND METHODS: The antiproliferative effect of 1,25-dihydroxycholecalciferol (Ro 21-5535) and of the two non hypercalcemic analogs (1a,25-dihydroxy-16-ene-23-yne-26,27-hexafluorocholecalciferol+ ++, Ro 24-5531 and 1a,25-dihydroxy-16-ene-23-yne-26,27-hexafluoro-19-nor-cholec alciferol, Ro 25-6760) was studied in MCF-7 and MDAMB-468 human breast cancer cell lines. Cell cycle distribution and apoptosis were evaluated by flow cytometry. Steroid receptor modulation was investigated by radioligand assay. RESULTS: The most effective drug was the Ro 25-6760 which at concentrations ranging between 1-100 nM caused a dose dependent growth inhibition apparently due to accumulation in G0/G1. Vitamin D3 analogs (10 nM) significantly counteracted the growth stimulation induced by TGF-a and IGF-I as well as the paracrine stimulation observed in co-cultures. They antagonized estradiol-promoted growth stimulation and progestrone receptor induction in MCF-7 cells. CONCLUSION: Vitamin D3 analogs represent a class of clinically attractive drugs for treatment of breast cancer due to their ability to counteract estradiol and growth factor-induced growth stimulation.


Subject(s)
Anticarcinogenic Agents/pharmacology , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Cholecalciferol/analogs & derivatives , Cholecalciferol/pharmacology , Apoptosis/drug effects , Breast Neoplasms , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line , ErbB Receptors/analysis , Female , Growth Substances/pharmacology , Humans , Receptor, IGF Type 1/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Structure-Activity Relationship , Tumor Cells, Cultured
10.
Int J Cancer ; 74(6): 620-4, 1997 Dec 19.
Article in English | MEDLINE | ID: mdl-9421359

ABSTRACT

The role of int-2 oncogene amplification on the prognosis of breast cancer patients was investigated in 128 patients with node-negative primary breast cancers given first-line local-regional treatments until relapse and with a median follow-up of 65 months. Tumours had been previously characterised for oestrogen (ER) and progesterone receptor (PgR) status and proliferative activity (3H-thymidine labelling index). Amplification of the int-2 oncogene occurred in 18% of cases and was significantly related to the presence of hormone receptors and to menopausal status or age, but not to proliferative status. Patients with tumours exhibiting int-2 amplification had a lower probability of disease-free survival than patients with non-amplified tumours and frequently developed local-regional recurrence. Disease-free survival analysis, adjusted for the prognostic contribution provided by tumour size, steroid receptors and proliferative rate, indicated that the association between int-2 amplification and risk of relapse was maintained and remained constant even in the presence of the other co-variates. Interestingly, int-2 amplification was a further prognostic discriminant within subsets of patients with a putatively good (i.e., tumour size <20 mm, ER+ and PgR+) or poor prognosis (i.e., high labelling index). Our exploratory study suggests that within node-negative patients, int-2 amplification could be a valuable and independent prognosticator, useful to identify patients at high risk of local-regional recurrence.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Fibroblast Growth Factors/genetics , Gene Amplification , Proto-Oncogene Proteins/genetics , Analysis of Variance , Breast Neoplasms/ultrastructure , DNA, Neoplasm/genetics , Disease-Free Survival , Female , Fibroblast Growth Factor 3 , Humans , Lymph Nodes/pathology , Middle Aged , Prognosis , Proportional Hazards Models , Receptors, Estrogen/analysis
11.
Tumour Biol ; 17(5): 290-8, 1996.
Article in English | MEDLINE | ID: mdl-8792855

ABSTRACT

Cathepsin-D and pS2 are two estrogen-regulated proteins in human breast cancer cell lines. They have been considered possible prognostic factors in breast cancer, but results have been contradictory. To better understand the regulation of these proteins, we investigated the role of estradiol (E2), serum, and growth factors in hormone-dependent (MCF-7, ZR75.1) and hormone-independent (MDAMB-231, BT20) breast cancer cell lines. E2 treatment in serum-free conditions increased intracellular and secreted levels of pS2 in ZR75.1 and in MCF-7, secreted levels only of cathepsin-D in MCF-7, and both levels of cathepsin-D in ZR75.1. Insulin-like growth factor I (IGF-I) and progesterone receptors were also stimulated by E2, whereas the estrogen receptor was down-regulated. Following treatment with epidermal growth factor (EGF), secreted pS2 levels doubled only in MCF-7 cells. IGF-I did not modify cathepsin-D or pS2 levels in either cell line, but caused an increase in its own receptor. Cathepsin-D and pS2 doubled in MCF-7 cells grown in medium supplemented with denaturated serum, but estrogen regulation of these proteins was still maintained. Cathepsin-D was expressed in MDAMB-231 and BT20, but its levels were modified by neither E2 nor growth factor treatment. Conversely, neither cell line expressed detectable levels of pS2 before or after treatment. In conclusion, our results show that in different types of breast cancer cells, some estrogen-regulated proteins (e.g. pS2) are also regulated by growth factors-such as EGF and other unknown serum factors. This may account for the contradictory results obtained regarding the prognostic relevance of cathepsin-D and pS2.


Subject(s)
Breast Neoplasms/chemistry , Cathepsin D/analysis , Neoplasm Proteins/analysis , Proteins , Cathepsin D/physiology , Estradiol/pharmacology , Female , Humans , Neoplasm Proteins/physiology , Receptor, IGF Type 1/analysis , Trefoil Factor-1 , Tumor Cells, Cultured , Tumor Suppressor Proteins
12.
Anticancer Res ; 15(6B): 2551-5, 1995.
Article in English | MEDLINE | ID: mdl-8669822

ABSTRACT

BACKGROUND: Reports about the effects of progestins on cell proliferation are contradictory. We investigated the effect of progesterone, medroxyprogesterone acetate, megestrol acetate, ORG 2058 and the antiprogestin RU 486 on two hormone-dependent cell lines, T47D and MCF-7 (characterized by a different content of PgR). The aim of the study was to understand the eventual ability of progestins to interfere with cell proliferation stimulated by estradiol and various growth factors (TGF-a, IGF-I, IGF-II). MATERIAL AND METHODS: MCF-7 and T47D cells were maintained in DMEM/F12 medium supplemented with 2% FCS while experiments were carried out in the same culture medium using DCC-stripped FCS. RESULTS: In the absence of estradiol, all tested progestins generally tended to stimulate cell growth in the T47D cell line, but in the MCF-7 cell line only the highest concentrations (10(-6) M and 10(-7) M) were found to be stimulatory. In contrast, in the presence of 10(-8) M estradiol, progestins tended to inhibit cell growth stimulation in MCF-7 and T47D cell lines. The antiprogestin RU 486 exerted a stimulatory effect similar to that promoted by estradiol itself in MCF-7 cells. Instead, in T47D cells, RU 486 did not modify cell growth in the absence of estradiol, but tended to counteract the estradiol-promoted cell proliferation. In MCF-7 cells, medroxyprogesterone acetate and megestrol acetate were also able to effectively counteract the cell growth induced by TGF-alpha. However, none of these progestins was able to abolish cell proliferation promoted by IGF-I or IGF-II. CONCLUSION: We therefore concluded that failure to respond to progestin treatment may be due to the very heterogeneous nature of human breast tumors and to the inability of these molecules to interfere with the IGF-R pathway.


Subject(s)
Adenocarcinoma/pathology , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Estradiol/pharmacology , Estrogens , Growth Substances/pharmacology , Neoplasms, Hormone-Dependent/pathology , Progesterone , Progestins/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Cell Division/drug effects , Drug Interactions , Female , Humans , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/pharmacology , Medroxyprogesterone Acetate/pharmacology , Megestrol/analogs & derivatives , Megestrol/pharmacology , Megestrol Acetate , Mifepristone/pharmacology , Neoplasm Proteins/analysis , Pregnenediones/pharmacology , Progesterone/pharmacology , Receptors, Progesterone/analysis , Transforming Growth Factor alpha/pharmacology , Tumor Cells, Cultured/drug effects
13.
Breast Cancer Res Treat ; 26(3): 275-81, 1993.
Article in English | MEDLINE | ID: mdl-8251652

ABSTRACT

A permeable solid support (Transwell Coll.) was used to develop serum-free co-cultures allowing paracrine interactions between hormone-dependent (MCF-7, ZR75.1) and hormone-independent (MDAMB-231, BT20) breast cancer cell lines. Both hormone-independent cell lines were able to stimulate the growth of the hormone-dependent lines, whereas the opposite was observed only in the case of BT20 co-culture with ZR75.1 cells. The cell growth stimulation observed in co-cultures could be abolished by the addition to the culture medium of an excess of transforming growth factor alpha (TGF alpha) or insulin-like growth factor I (IGF-I). Similarly, treatment with a neutralizing anti TGF alpha antibody impaired the growth stimulation exerted by hormone-independent cells on hormone-dependent cells. These results confirm the important role of paracrine interactions in control of the growth of human heterogeneous breast tumors and suggest that the main growth factors involved in such interactions are TGF alpha and probably some growth factors from the insulin-like growth factor family rather than IGF-I itself.


Subject(s)
Breast Neoplasms/pathology , Cell Communication/physiology , Neoplasms, Hormone-Dependent/pathology , Breast Neoplasms/ultrastructure , Cell Division/physiology , Culture Media , Growth Substances/physiology , Humans , Neoplasms, Hormone-Dependent/ultrastructure , Receptor, IGF Type 1/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Tumor Cells, Cultured
14.
Anticancer Res ; 10(6): 1699-705, 1990.
Article in English | MEDLINE | ID: mdl-2285244

ABSTRACT

Laboratory data from Milan and Houston were evaluated to determine the extent to which the distribution of estrogen receptor (ER) and progesterone receptor (PgR) has changed with time. Results from over 11,500 ER and over 8,200 PgR determinations (6,194 ER and 3,127 PgR from Milan) were analyzed. All assays in Milan were performed by a dextran-coated charcoal method and in Houston by a sucrose density-gradient method. The data demonstrate a time-dependent, upward drift in the amount of ER and PgR detected, with the effect most pronounced at the lower end of the distribution curves. We attribute this change to optimization of all facets of the receptor assay procedures (tissue harvesting and storage) as well as to a change in breast cancer biology. These results suggest that studies correlating certain biological parameters with receptor status (whether using qualitative or quantitative scales) need to be re-examined. For example, a population of 349 node-negative patients who did not receive any adjuvant treatment was studied in Milan to determine any association between disease-free survival (DFS) and receptor status. If the "historical" threshold values (10 fmol/mg protein) were used to determine receptor status, no significant difference in DFS at 5 years was detected. Even the combination of ER and PgR did not improve the predictive power of receptor status. In the premenopausal subgroup, ER status did predict the 5-year DFS. However, if the threshold value for PgR was adjusted to 25 fmol/mg protein, patients with ER-positive, PgR-positive tumors had significantly better 5-year DFS than patients with ER-negative, PgR-negative tumors. In addition, PgR status alone was associated with significantly improved 3-year DFS if the subgroups of PgR less than 5 fmol/mg protein and PgR greater than 100 fmol/mg protein were compared. We conclude from these data that: 1) historical threshold values for receptor positivity should be re-examined in all laboratories; 2) studies involving receptor results determined over an extended period of time should attempt to "normalize" these results; and 3) the quantitative assessment of receptor status should be used whenever possible.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Breast Neoplasms/surgery , Female , Follow-Up Studies , Humans , Italy , Mastectomy , Prognosis , Texas
15.
Anticancer Res ; 8(6): 1297-301, 1988.
Article in English | MEDLINE | ID: mdl-3218961

ABSTRACT

Steroid receptors were measured in a series of 30 operable canine mammary gland tumors; both cytoplasmic estrogen (ERc) and progesterone (PgRc) receptor mean concentrations were very low with respect to the mean levels found in humans. Therefore a study was designed to modulate receptor levels by administration of Tamoxifen and Etretinate, either alone or in combination. Forty dogs with resectable, histologically documented mammary gland tumors were subdivided into the following treatment groups: a. Etretinate (1 mg/kg/d) p.o. for 7 days followed by Tamoxifen (0.7 mg/kg/d) p.o. for 7 days; b. Tamoxifen (0.7 mg/kg/d) p.o. for 14 days; c. Etretinate (1 mg/kg/d) p.o. for 14 days; d. 14 days placebo, and cytoplasmic ERc and PgRc and nuclear ER (ERn) were measured before and after the treatment. An increase of ERc and ERn was observed after administration of Tamoxifen, while an increase of ERc only was seen after treatment with Etretinate. We conclude that canine mammary tumors are indeed hormone sensitive despite their very low receptor concentrations and a suitable treatment can in fact modulate receptor levels. However, further studies are needed better to define the optimal treatment regimen in order to achieve maximal steroid receptor induction.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dog Diseases/drug therapy , Etretinate/therapeutic use , Neoplasms/veterinary , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Tamoxifen/therapeutic use , Animals , Cytosol/metabolism , Dog Diseases/metabolism , Dog Diseases/pathology , Dogs , Etretinate/administration & dosage , Female , Mammary Glands, Animal/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Receptors, Estrogen/drug effects , Receptors, Progesterone/drug effects , Tamoxifen/administration & dosage
16.
Tumour Biol ; 9(4): 200-11, 1988.
Article in English | MEDLINE | ID: mdl-3420376

ABSTRACT

Human breast cancer cell lines, as well as human breast cancer biopsies, possess specific high-affinity epidermal growth factor receptors (EGF-r). However, reports on the presence of EGF-r in human breast cancer biopsies are still controversial, especially concerning the relationship between EGF-r and other biological variables whose prognostic relevance is well known, such as the estrogen (ER) and progesterone (PgR) receptors. In the present study, the EGF-r content was estimated in a series of 136 unselected breast cancer primaries along with cytoplasmic (ERc) and nuclear (ERn) ER and cytoplasmic PgR. EGF-binding activity consisted of a single class of high-affinity binding sites (Kd = 0.55 nM) and ranged from 0 to 275 fmol/mg protein. We observed a strong inverse association between EGF-r and ERc (p less than 0.001); in fact, about two thirds of the tumors were ERc-positive/EGF-r-negative or ERc-negative/EGF-r-positive. The same type of association was found between EGF-r and either ERn or PgR. Kendall's rank correlation test confirmed that the EGF-r concentrations were correlated with the levels of ERc (tau = -0.291, p less than 0.0001), ERn (tau = -0.27, p less than 0.0005) and PgR (tau = -0.162, p less than 0.01). The EGF-r content was significantly higher (p less than 0.0001) in the ERc-negative tumors (72.6 +/- 54.4 fmol/mg protein) as compared to the ERc-positive ones (33.0 +/- 37.4 fmol/mg protein). Similarly, the subset of PgR-positive tumors was characterized by lower EGF-r mean concentrations when compared to PgR-negative cases (35.4 +/- 54.4 vs. 63.8 +/- 54.4 fmol/mg protein). These results confirm the previously described inverse relationship between EGF-r and steroid receptors. Moreover, they suggest the existence of an interaction between steroid hormones and EGF and support the need for further studies to better understand their respective roles in modulating breast cancer growth.


Subject(s)
Breast Neoplasms/analysis , ErbB Receptors/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Breast Neoplasms/mortality , Female , Humans , Prognosis
17.
Cancer Detect Prev ; 10(5-6): 327-33, 1987.
Article in English | MEDLINE | ID: mdl-3032437

ABSTRACT

The presence of cellular retinoic acid binding protein (cRABP) was analyzed in 13 consecutive patients with large bowel cancer (one right colon and 12 rectum-sigmoid, classified as three Duke B, eight C, and two D). Specimens of neoplastic tissue and of adjacent mucosa were obtained at surgery, and cRABP was determined by an assay based on incubation of partially purified cytosol with labeled retinoic acid and ultracentrifugation in sucrose density gradients. Sixty-one percent of tumors contained detectable levels of cRABP, whereas 58.3% of normal mucosal specimens were positive for cRABP. Among the positive tumors 62.5% contained cRABP also in the corresponding mucosa; in the group of cRABP-negative tumors, 40% showed cRABP in the adjacent mucosa. No correlation could be established with the grading or the stage of the tumors; however, interestingly, 100% (three cases) of gelatinous carcinomas were cRABP positive. Since cRABP seems to be a character of neoplastic cells contrary to normal ones, it would be interesting to investigate the conditions that influence the presence of this protein in normal appearing mucosa adjacent to carcinoma.


Subject(s)
Carrier Proteins/analysis , Colonic Neoplasms/analysis , Neoplasm Proteins/analysis , Rectal Neoplasms/analysis , Humans , In Vitro Techniques , Intestinal Mucosa/analysis , Receptors, Retinoic Acid
18.
Cancer Res ; 46(8 Suppl): 4278s-4281s, 1986 Aug.
Article in English | MEDLINE | ID: mdl-3524809

ABSTRACT

We have compared a new enzyme immunoassay (EIA) for estrogen receptors (ER) with our conventional radioligand binding assays (multipoint dextran-coated charcoal assay for cytoplasmic ER and hydroxylapatite exchange assay for nuclear ER). Cytoplasmic ERs were measured in 76 human breast cancer specimens by EIA and by five-point Scatchard analysis. The correlation between the two assays yielded a straight line with a slope of 0.92 (r = 0.95; P less than 0.001); conversely, in 31 nuclear salt extracts, linear regression analysis of hydroxylapatite exchange assay data with EIA showed a clear correlation (r = 0.93; P less than 0.001) but a slope of 1.7, demonstrating that EIA detects more ER sites. The binding of the antibody to the cytoplasmic ER molecules was investigated by sucrose density gradient analysis, which showed that EIA recognizes both cytoplasmic forms (9 and 3S), but does not distinguish between them. Advantages and drawbacks of this method are discussed with respect to its application for routine receptor determination for clinical management of breast cancer patients.


Subject(s)
Breast Neoplasms/analysis , Receptors, Estrogen/analysis , Cell Nucleus/analysis , Cytoplasm/analysis , Female , Humans , Immunoenzyme Techniques , Radioligand Assay , Receptors, Estrogen/immunology
19.
Tumori ; 72(3): 251-7, 1986 Jun 30.
Article in English | MEDLINE | ID: mdl-3739005

ABSTRACT

Estrogen (ER) and progesterone receptors (PgR) appear to be a prerequisite to elicit a biologic response by a hormone-target organ. Current methodologies for analysis of these proteins (e.g., dextran-coated charcoal, DCC) in single-label assay (SLA) require relatively large amounts of tissue material, time and laboriousness. Therefore, we have developed for breast cancer tissue an improved dual-label assay (DLA) for simultaneous titration (by DCC) and/or characterization (by sedimentation properties) of ER and PgR on the same sample, using 125I-E2 and 3H-Org 2058 as tracers. The interaction of 125I-E2 with ER and plasma proteins in comparison to 3H-E2 was studied in terms of specificity, time course, affinity binding and sedimentation pattern. 125I-E2 bound the same molecular forms displayed by 3H-E2 (9 and 3S) but with lower titers (about 1.3-fold), irrespective of the technique used, and did not bind to sex hormone-binding globulin. Simultaneous detection of 125I and 3H was achieved by use of a gamma counter plus a beta counter sequentially. ER and PgR titrations with DCC in DLA were in good agreement with those obtained with SLA, in terms of titers and Ka values. An analogous result was obtained with sucrose density gradient (SDG) analysis. Both the DLA methods were highly reproducible (CV less than 8.0%). Between the rotors available for SDG, the vertical one was preferable because of the larger number of samples processed and of less perturbation of sedimenting receptor molecules. Furthermore, a biochemical application of the method is described. In conclusion, the DLA procedure, by simplifying ER and PgR estimation, makes it possible to study, even on small tumor biopsies, the molecular properties of these proteins in relation to the clinical response of the disease.


Subject(s)
Breast Neoplasms/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Breast Neoplasms/secondary , Centrifugation, Density Gradient , Cytosol/analysis , Estradiol/analysis , Female , Humans , Iodine Radioisotopes , Methods , Pregnenediones/analysis , Scintillation Counting , Tritium
20.
J Urol ; 135(1): 18-21, 1986 Jan.
Article in English | MEDLINE | ID: mdl-2934557

ABSTRACT

From July 1, 1979 to June 30, 1983, 136 consecutive patients with category M0 renal cell cancer who had undergone transperitoneal radical nephrectomy at 5 centers entered a prospective randomized trial to compare 500 mg. adjuvant medroxyprogesterone 3 times a week for 1 year to no treatment. Sex steroid hormone receptors also were studied in the renal tumor and in the surrounding healthy parenchyma with the dextran-coated charcoal technique. After a median followup period of 3 years (range 13 to 60 months) 30 of 121 evaluable patients (24.8 per cent) experienced relapse, usually in the lung or bones. Relapses occurred in 15 of 58 evaluable patients in the adjuvant treatment group (25.8 per cent) and 15 of 63 evaluable controls (23.8 per cent). The disease recurred more frequently (35.1 per cent) in the 57 patients with no receptors in the tumor than in the 45 with at least 1 receptor (17.8 per cent). These results were independent of adjuvant therapy. After a median 3-year followup, adjuvant medroxyprogesterone acetate was of no therapeutic benefit in patients who had undergone radical nephrectomy and the side effects of the therapy were evident in more than 50 per cent of the patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/therapy , Kidney Neoplasms/therapy , Medroxyprogesterone/analogs & derivatives , Receptors, Steroid/analysis , Carcinoma, Renal Cell/analysis , Clinical Trials as Topic , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Kidney/analysis , Kidney Neoplasms/analysis , Male , Medroxyprogesterone/therapeutic use , Medroxyprogesterone Acetate , Nephrectomy , Prospective Studies , Random Allocation , Tamoxifen/therapeutic use , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...