Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Endocr Relat Cancer ; 31(2)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37962553

ABSTRACT

Progesterone receptors (PRs) are biomarkers used as prognostic and predictive factors in breast cancer, but they are still not used as therapeutic targets. We have proposed that the ratio between PR isoforms A and B (PRA and PRB) predicts antiprogestin responsiveness. The MIPRA trial confirmed the benefit of 200 mg mifepristone, administered to patients with tumors with a high PRA/PRB ratio, but dose-ranging has not been conducted. The aim of this study was to establish the plasma mifepristone levels of patients from the MIPRA trial, along with the resultant steroid profiles, and compare these with those observed in mifepristone-treated mice using therapeutic schemes able to induce the regression of experimental mammary carcinomas with high PRA/PRB ratios: 6 mg pellets implanted subcutaneously, or daily doses of 12 mg/kg body weight. The plasma levels of mifepristone and other 19 plasma steroids were measured by liquid chromatography-tandem mass spectometry. In mifepristone-treated mice, plasma levels were lower than those registered in mifepristone-treated patients (i.e. day 7 after treatment initiation, pellet-treated mice: 8.4 ± 3.9 ng/mL; mifepristone-treated patients: 300.3 ± 31.7 ng/mL (mean ± s.d.; P < 0.001)). The increase in corticoid related steroids observed in patients was not observed in mifepristone-treated mice. The increase in progesterone levels was the most significant side effect detected in mifepristone-treated mice after 14 or 21 days of treatment, probably due to an ovarian compensatory effect not observed in postmenopausal patients. We conclude that in future clinical trials using mifepristone, the possibility of lowering the standard daily dose of 200 mg should be considered.


Subject(s)
Breast Neoplasms , Mifepristone , Humans , Mice , Animals , Female , Mifepristone/therapeutic use , Mifepristone/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Receptors, Progesterone , Hormone Antagonists/therapeutic use , Hormone Antagonists/pharmacology , Prognosis
2.
J Proteome Res ; 2023 Jul 27.
Article in English | MEDLINE | ID: mdl-37497607

ABSTRACT

The lipid metabolism adaptations of estrogen and progesterone receptor-positive breast cancer tumors from a mouse syngeneic model are investigated in relation to differences across the transition from hormone-dependent (HD) to hormone-independent (HI) tumor growth and the acquisition of endocrine therapy (ET) resistance (HIR tumors). Results are articulated with reported polar metabolome results to complete a metabolic picture of the above transitions and suggest markers of tumor progression and aggressiveness. Untargeted nuclear magnetic resonance metabolomics was used to analyze tumor and mammary tissue lipid extracts. Tumor progression (HD-HI-HIR) was accompanied by increased nonesterified cholesterol forms and phospholipids (phosphatidylcholine, phosphatidylethanolamine, sphingomyelins, and plasmalogens) and decreased relative contents of triglycerides and fatty acids. Predominating fatty acids became shorter and more saturated on average. These results were consistent with gradually more activated cholesterol synthesis, ß-oxidation, and phospholipid biosynthesis to sustain tumor growth, as well as an increase in cholesterol (possibly oxysterol) forms. Particular compound levels and ratios were identified as potential endocrine tumor HD-HI-HIR progression markers, supporting new hypotheses to explain acquired ET resistance.

3.
Clin Cancer Res ; 29(5): 866-877, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36269797

ABSTRACT

PURPOSE: Preclinical data suggest that antiprogestins inhibit the growth of luminal breast carcinomas that express higher levels of progesterone receptor isoform A (PRA) than isoform B (PRB). Thus, we designed a presurgical window of opportunity trial to determine the therapeutic effects of mifepristone in patients with breast cancer, based on their high PRA/PRB isoform ratio (MIPRA; NCT02651844). PATIENTS AND METHODS: Twenty patients with luminal breast carcinomas with PRA/PRB > 1.5 (determined by Western blots), and PR ≥ 50%, naïve from previous treatment, were included for mifepristone treatment (200 mg/day orally; 14 days). Core needle biopsies and surgical samples were formalin fixed for IHC studies, while others were snap-frozen to perform RNA sequencing (RNA-seq), proteomics, and/or Western blot studies. Plasma mifepristone levels were determined using mass spectrometry. The primary endpoint was the comparison of Ki67 expression pretreatment and posttreatment. RESULTS: A 49.62% decrease in Ki67 staining was observed in all surgical specimens compared with baseline (P = 0.0003). Using the prespecified response parameter (30% relative reduction), we identified 14 of 20 responders. Mifepristone induced an increase in tumor-infiltrating lymphocytes; a decrease in hormone receptor and pSer118ER expression; and an increase in calregulin, p21, p15, and activated caspase 3 expression. RNA-seq and proteomic studies identified downregulated pathways related to cell proliferation and upregulated pathways related to immune bioprocesses and extracellular matrix remodeling. CONCLUSIONS: Our results support the use of mifepristone in patients with luminal breast cancer with high PRA/PRB ratios. The combined effects of mifepristone and estrogen receptor modulators warrant clinical evaluation to improve endocrine treatment responsiveness in these patients. See related commentary by Ronchi and Brisken, p. 833.


Subject(s)
Breast Neoplasms , Mifepristone , Humans , Female , Mifepristone/pharmacology , Mifepristone/therapeutic use , Receptors, Progesterone/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Proteomics , Ki-67 Antigen , Protein Isoforms/genetics , Protein Isoforms/metabolism
4.
Front Oncol ; 12: 786931, 2022.
Article in English | MEDLINE | ID: mdl-35299741

ABSTRACT

Breast cancer (BC) is the most common type of cancer in women and, in most cases, it is hormone-dependent (HD), thus relying on ovarian hormone activation of intracellular receptors to stimulate tumor growth. Endocrine therapy (ET) aimed at preventing hormone receptor activation is the primary treatment strategy, however, about half of the patients, develop resistance in time. This involves the development of hormone independent tumors that initially are ET-responsive (HI), which may subsequently become resistant (HIR). The mechanisms that promote the conversion of HI to HIR tumors are varied and not completely understood. The aim of this work was to characterize the metabolic adaptations accompanying this conversion through the analysis of the polar metabolomes of tumor tissue and non-compromised mammary gland from mice implanted subcutaneously with HD, HI and HIR tumors from a medroxyprogesterone acetate (MPA)-induced BC mouse model. This was carried out by nuclear magnetic resonance (NMR) spectroscopy of tissue polar extracts and data mining through multivariate and univariate statistical analysis. Initial results unveiled marked changes between global tumor profiles and non-compromised mammary gland tissues, as expected. More importantly, specific metabolic signatures were found to accompany progression from HD, through HI and to HIR tumors, impacting on amino acids, nucleotides, membrane percursors and metabolites related to oxidative stress protection mechanisms. For each transition, sets of polar metabolites are advanced as potential markers of progression, including acquisition of resistance to ET. Putative biochemical interpretation of such signatures are proposed and discussed.

5.
Endocrinology ; 163(2)2022 02 01.
Article in English | MEDLINE | ID: mdl-34977930

ABSTRACT

Luminal breast cancer (BrCa) has a favorable prognosis compared with other tumor subtypes. However, with time, tumors may evolve and lead to disease progression; thus, there is a great interest in unraveling the mechanisms that drive tumor metastasis and endocrine resistance. In this review, we focus on one of the many pathways that have been involved in tumor progression, the fibroblast growth factor/fibroblast growth factor receptor (FGFR) axis. We emphasize in data obtained from in vivo experimental models that we believe that in luminal BrCa, tumor growth relies in a crosstalk with the stromal tissue. We revisited the studies that illustrate the interaction between hormone receptors and FGFR. We also highlight the most frequent alterations found in BrCa cell lines and provide a short review on the trials that use FGFR inhibitors in combination with endocrine therapies. Analysis of these data suggests there are many players involved in this pathway that might be also targeted to decrease FGF signaling, in addition to specific FGFR inhibitors that may be exploited to increase their efficacy.


Subject(s)
Breast Neoplasms/drug therapy , Fibroblast Growth Factors/physiology , Receptors, Fibroblast Growth Factor/physiology , Receptors, Steroid/physiology , Signal Transduction/physiology , Animals , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/chemistry , Breast Neoplasms/therapy , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Estrogen Receptor alpha/analysis , Female , Fibroblast Growth Factors/genetics , Gene Amplification , Humans , Mice , Mutation , Receptor Cross-Talk/physiology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/genetics
6.
Int J Cancer ; 150(9): 1481-1496, 2022 05 01.
Article in English | MEDLINE | ID: mdl-34935137

ABSTRACT

Progesterone receptors (PRs) ligands are being tested in luminal breast cancer. There are mainly two PR isoforms, PRA and PRB, and their ratio (PRA/PRB) may be predictive of antiprogestin response. Our aim was to investigate: the impact of the PR isoform ratio on metastatic behaviour, the PR isoform ratio in paired primary tumours and lymph node metastases (LNM) and, the effect of antiprogestin/progestins on metastatic growth. Using murine and human metastatic models, we demonstrated that tumours with PRB > PRA (PRB-H) have a higher proliferation index but less metastatic ability than those with PRA > PRB (PRA-H). Antiprogestins and progestins inhibited metastatic burden in PRA-H and PRB-H models, respectively. In breast cancer samples, LNM retained the same PRA/PRB ratio as their matched primary tumours. Moreover, PRA-H LNM expressed higher total PR levels than the primary tumours. The expression of NDRG1, a metastasis suppressor protein, was higher in PRB-H compared to PRA-H tumours and was inversely regulated by antiprogestins/progestins. The binding of the corepressor SMRT at the progesterone responsive elements of the NDRG1 regulatory sequences, together with PRA, impeded its expression in PRA-H cells. Antiprogestins modulate the interplay between SMRT and AIB1 recruitment in PRA-H or PRB-H contexts regulating NDRG1 expression and thus, metastasis. In conclusion, we provide a mechanistic interpretation to explain the differential role of PR isoforms in metastatic growth and highlight the therapeutic benefit of using antiprogestins in PRA-H tumours. The therapeutic effect of progestins in PRB-H tumours is suggested.


Subject(s)
Breast Neoplasms , Cell Cycle Proteins , Intracellular Signaling Peptides and Proteins , Receptors, Progesterone , Animals , Breast Neoplasms/pathology , Cell Cycle Proteins/metabolism , Female , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Neoplasm Metastasis , Progesterone/pharmacology , Progestins/metabolism , Protein Isoforms/metabolism , Receptors, Progesterone/metabolism
7.
Mol Cell Endocrinol ; 538: 111465, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34597725

ABSTRACT

Growth Hormone (GH) plays crucial roles in mammary gland development and growth, and its upregulation has been associated with breast cancer promotion and/or progression. To ascertain how high GH levels could promote mammary tissue oncogenic transformation, morphological characteristics and the expression of receptors involved in mammary growth, development and cancer, and of mitogenic mediators were analyzed in the mammary gland of virgin adult transgenic mice that overexpress GH. Whole mounting and histologic analysis evidenced that transgenic mice exhibit increased epithelial ductal elongation and enlarged ducts along with deficient branching and reduced number of alveolar structures compared to wild type mice. The number of differentiated alveolar structures was diminished in transgenic mice while the amount of terminal end buds (TEBs) did not differ between both groups of mice. GH, insulin-like growth factor 1 (IGF1) and GH receptor mRNA levels were augmented in GH-overexpressing mice breast tissue, as well as IGF1 receptor protein content. However, GH receptor protein levels were decreased in transgenic mice. Fundamental receptors for breast growth and development like progesterone receptor and epidermal growth factor receptor were also increased in mammary tissue from transgenic animals. In turn, the levels of the proliferation marker Ki67, cFOS and Cyclin D1 were increased in GH-overexpressing mice, while cJUN expression was decreased and cMYC did not vary. In conclusion, prolonged exposure to high GH levels induces morphological and molecular alterations in the mammary gland that affects its normal development. While these effects would not be tumorigenic per se, they might predispose to oncogenic transformation.


Subject(s)
Carrier Proteins/genetics , Growth Hormone/genetics , Insulin-Like Growth Factor I/genetics , Mammary Glands, Animal/abnormalities , Animals , Animals, Genetically Modified , Biomarkers/metabolism , Female , Growth Hormone/metabolism , Mammary Glands, Animal/metabolism , Mice , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-myc/metabolism
8.
Essays Biochem ; 65(6): 951-969, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34061163

ABSTRACT

Progesterone receptors (PR) play a pivotal role in many female reproductive tissues such as the uterus, the ovary, and the mammary gland (MG). Moreover, PR play a key role in breast cancer growth and progression. This has led to the development and study of different progestins and antiprogestins, many of which are currently being tested in clinical trials for cancer treatment. Recent reviews have addressed the role of PR in MG development, carcinogenesis, and breast cancer growth. Thus, in this review, in addition to making an overview on PR action in normal and tumor breast, the focus has been put on highlighting the still unresolved topics on hormone treatment involving PR isoforms and breast cancer prognosis.


Subject(s)
Breast Neoplasms , Receptors, Progesterone , Breast/pathology , Breast Neoplasms/drug therapy , Female , Humans , Progesterone/therapeutic use , Progestins/therapeutic use , Receptors, Progesterone/therapeutic use
9.
Cancer Res ; 81(5): 1375-1387, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33268529

ABSTRACT

The role of active antitumor immunity in hormone receptor-positive (HR+) breast cancer has been historically underlooked. The aim of this study was to determine the contribution of the immune system to antiprogestin-induced tumor growth inhibition using a hormone-dependent breast cancer model. BALB/c-GFP+ bone marrow (BM) cells were transplanted into immunodeficient NSG mice to generate an immunocompetent NSG/BM-GFP+ (NSG-R) mouse model. Treatment with the antiprogestin mifepristone (MFP) inhibited growth of 59-2-HI tumors with similar kinetics in both animal models. Interestingly, MFP treatment reshaped the tumor microenvironment, enhancing the production of proinflammatory cytokines and chemokines. Tumors in MFP-treated immunocompetent mice showed increased infiltration of F4/80+ macrophages, natural killer, and CD8 T cells, displaying a central memory phenotype. Mechanistically, MFP induced immunogenic cell death (ICD) in vivo and in vitro, as depicted by the expression and subcellular localization of the alarmins calreticulin and HMGB-1 and the induction of an ICD gene program. Moreover, MFP-treated tumor cells efficiently activated immature dendritic cells, evidenced by enhanced expression of MHC-II and CD86, and induced a memory T-cell response, attenuating tumor onset and growth after re-challenge. Finally, MFP treatment increased the sensitivity of HR+ 59-2-HI tumor to PD-L1 blockade, suggesting that antiprogestins may improve immunotherapy response rates. These results contribute to a better understanding of the mechanisms underlying the antitumor effect of hormonal treatment and the rational design of therapeutic combinations based on endocrine and immunomodulatory agents in HR+ breast cancer. SIGNIFICANCE: Antiprogestin therapy induces immunogenic tumor cell death in PRA-overexpressing tumors, eliciting an adaptive immune memory response that protects mice from future tumor recurrence and increases sensitivity to PD-L1 blockade. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/5/1375/F1.large.jpg.


Subject(s)
Immune Checkpoint Inhibitors/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , Cell Death/drug effects , Cell Death/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Female , Humans , Immunologic Memory/drug effects , Mammary Neoplasms, Experimental/pathology , Mice, Inbred BALB C , Mice, Transgenic , Mifepristone/pharmacology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
12.
Biomolecules ; 10(9)2020 08 27.
Article in English | MEDLINE | ID: mdl-32867141

ABSTRACT

The metabolic characteristics of metastatic and non-metastatic breast carcinomas remain poorly studied. In this work, untargeted Nuclear Magnetic Resonance (NMR) metabolomics was used to compare two medroxyprogesterone acetate (MPA)-induced mammary carcinomas lines with different metastatic abilities. Different metabolic signatures distinguished the non-metastatic (59-2-HI) and the metastatic (C7-2-HI) lines, with glucose, amino acid metabolism, nucleotide metabolism and lipid metabolism as the major affected pathways. Non-metastatic tumours appeared to be characterised by: (a) reduced glycolysis and tricarboxylic acid cycle (TCA) activities, possibly resulting in slower NADH biosynthesis and reduced mitochondrial transport chain activity and ATP synthesis; (b) glutamate accumulation possibly related to reduced glutathione activity and reduced mTORC1 activity; and (c) a clear shift to lower phosphoscholine/glycerophosphocholine ratios and sphingomyelin levels. Within each tumour line, metabolic profiles also differed significantly between tumours (i.e., mice). Metastatic tumours exhibited marked inter-tumour changes in polar compounds, some suggesting different glycolytic capacities. Such tumours also showed larger intra-tumour variations in metabolites involved in nucleotide and cholesterol/fatty acid metabolism, in tandem with less changes in TCA and phospholipid metabolism, compared to non-metastatic tumours. This study shows the valuable contribution of untargeted NMR metabolomics to characterise tumour metabolism, thus opening enticing opportunities to find metabolic markers related to metastatic ability in endocrine breast cancer.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Adenocarcinoma/chemically induced , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Citric Acid Cycle , Female , Glucose/metabolism , Glycolysis , Humans , Lipid Metabolism , Magnetic Resonance Spectroscopy , Mammary Neoplasms, Experimental/chemically induced , Medroxyprogesterone Acetate , Metabolome , Mice , Mice, Inbred BALB C , Neoplasm Metastasis/pathology , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology
13.
Sci Rep ; 10(1): 15062, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32934281

ABSTRACT

Anti-Müllerian hormone (AMH) is secreted by Sertoli cells of the testes from early fetal life until puberty, when it is downregulated by androgens. In conditions like complete androgen insensitivity syndrome (CAIS), AMH downregulation does not occur and AMH increases at puberty, due in part to follicle-stimulating hormone (FSH) effect. However, other conditions like Peutz-Jeghers syndrome (PJS), characterised by low FSH, also have increased AMH. Because both CAIS and PJS may present as hyperoestrogenic states, we tested the hypothesis that oestradiol (E2) upregulates AMH expression in peripubertal Sertoli cells and explored the molecular mechanisms potentially involved. The results showed that E2 is capable of inducing an upregulation of endogenous AMH and of the AMH promoter activity in the prepubertal Sertoli cell line SMAT1, signalling through ERα binding to a specific ERE sequence present on the hAMH promoter. A modest action was also mediated through the membrane oestrogen receptor GPER. Additionally, the existence of ERα expression in Sertoli cells in patients with CAIS was confirmed by immunohistochemistry. The evidence presented here provides biological plausibility to the hypothesis that testicular AMH production increases in clinical conditions in response to elevated oestrogen levels.


Subject(s)
Androgen-Insensitivity Syndrome/metabolism , Anti-Mullerian Hormone/metabolism , Estrogen Receptor alpha/biosynthesis , Neoplasm Proteins/biosynthesis , Peutz-Jeghers Syndrome/metabolism , Response Elements , Sertoli Cells/metabolism , Androgen-Insensitivity Syndrome/pathology , Animals , Cell Line , Child , Child, Preschool , Estradiol/metabolism , Female , Humans , Male , Mice , Peutz-Jeghers Syndrome/pathology , Sertoli Cells/pathology
14.
Article in English | MEDLINE | ID: mdl-32349923

ABSTRACT

Progesterone (Pg) is a pregnancy-related hormone that prepares the endometrium for the implantation of the fertilized zygote and suppresses myometrial contractility for the maintenance of pregnancy. At high concentrations, it acts as a natural immunosuppressant avoiding the rejection of a half allogeneic foetus. It is the precursor of many other related steroid hormones, but what is its role in the human breast? In this chapter, we will discuss some aspects related to Pg and its role in breast development and in the neoplastic disease. Understanding the mechanisms related to Pg-induced effects in the normal and neoplastic mammary gland will light the way to exploit this hormone signalling pathway therapeutically. We will introduce some aspects of the effects of progestins in normal breast development, breast cancer risk and in neoplastic growth, and we will describe ongoing clinical trials in breast cancer using progestins or antiprogestins.


Subject(s)
Breast Neoplasms , Progesterone , Breast , Breast Neoplasms/drug therapy , Female , Hormones , Humans , Pregnancy , Progestins , Receptors, Progesterone
15.
Breast Cancer Res Treat ; 180(1): 257-263, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31950383

ABSTRACT

PURPOSE: Expression of estrogen receptor alpha (ER) and/or progesterone receptor (PR) defines luminal breast cancer. Even though androgen (AR) and glucocorticoid receptors (GR) are highly expressed in luminal breast cancers, prognostic value remains uncertain and concomitant expression of these four hormone receptors is still unexplored. METHODS: Here, we evaluated ER, PR, AR, and GR expression, using immunohistochemistry, in a cohort of 169 breast cancer patients and correlated these findings with clinical and pathological parameters. RESULTS: We found that AR is more frequently expressed and at higher levels in the ER+PR- subset compared to ER+PR+ tumors. There were no significant differences in GR expression between tumor subsets. Moreover, most luminal tumors also expressed either AR or GR and most basal tumors were also negative for AR and GR. CONCLUSION: These data suggest that targeting AR in ER+PR- tumors may represent a promising therapeutic alternative in hormonal refractory tumors.


Subject(s)
Breast Neoplasms/genetics , Gene Expression , Receptors, Androgen/genetics , Adult , Aged , Biomarkers, Tumor , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Cluster Analysis , Female , Gene Frequency , Humans , Immunohistochemistry , Middle Aged , Neoplasm Grading , Neoplasm Staging , Receptors, Androgen/metabolism , Receptors, Estrogen , Receptors, Progesterone
16.
Medicina (B Aires) ; 79(Spec 6/1): 540-545, 2019.
Article in English | MEDLINE | ID: mdl-31864223

ABSTRACT

Seventy per cent of breast cancers are luminal carcinomas that express estrogen receptor alpha (ER). For several decades, its expression has been used as a therapeutic target in patients with breast cancer. These therapies are aimed at blocking ER or inhibiting ligand synthesis. The expression of progesterone receptors (PR) is evaluated as a prognostic factor together with ER. It has been shown that there are two predominant PR isoforms with different molecular weight, isoform A and isoform B, which are not distinguished by immunohistochemical techniques. The available evidence indicates that the PR isoform ratio may have both a prognostic and predictive value of the response to antiprogestin treatment. In luminal mammary carcinomas, androgen receptors (AR) are expressed in a high percentage and the AR/ER or AR/PR ratio could be a prognostic factor. In ER negative (-) tumors, AR expression is an indicator of poor prognosis and it is proposed that they may be susceptible to antiandrogen treatment. Finally, the expression of glucocorticoid receptors (GR) would be an indicator of good or bad prognosis in luminal or ER- tumors, respectively. In ER- tumors, metastases express higher levels of nuclear GR than primary tumors and therapies that block GR could improve the efficacy of chemotherapy. Given the crosstalk of pathways triggered by different hormone receptors, it is possible that in the future, a therapeutic scheme can be administered that contemplates the expression of ER, PR isoforms, AR and GR.


El 70 por ciento de los carcinomas mamarios son luminales y expresan receptores de estrógenos alfa (RE). Desde hace varias décadas, su expresión se utiliza como blanco terapéutico en pacientes con cáncer de mama. Estas terapias están dirigidas a bloquear el RE o a inhibir la síntesis del ligando. La expresión de receptores de progesterona (RP) se evalúa como factor pronóstico junto con los RE. Se ha comprobado que existen dos isoformas predominantes de RP de distinto peso molecular, isoforma A e isoforma B, que no se distinguen por técnicas de inmunohistoquímica. Las evidencias indican que la proporción de isoformas de RP podría tener tanto un valor pronóstico como predictivo de la respuesta a un tratamiento con antiprogestágenos. En tumores mamarios luminales, los receptores de andrógenos (RA) se expresan en un alto porcentaje y la proporción de RA/RE o RA/RP podría ser un factor pronóstico. En tumores RE-, la expresión de RA es indicador de mal pronóstico y se propone que serían susceptibles a un tratamiento con antiandrógenos. Por último, la expresión de receptores de glucocorticoides (RG) sería un indicador de buen o mal pronóstico en tumores luminales o RE-, respectivamente. En tumores RE-, las metástasis expresan mayores niveles de RG nuclear que los tumores primarios y las terapias que bloquean los RG podrían mejorar la eficacia de la quimioterapia. Dado los entrecruzamientos de vías gatilladas por distintos receptores hormonales es posible que en un futuro se pueda administrar un esquema terapéutico que contemple la expresión de RE, isoformas de RP, RA y RG.


Subject(s)
Breast Neoplasms/metabolism , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/metabolism , Biomarkers/metabolism , Breast Neoplasms/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Prognosis , Receptors, Androgen/genetics , Receptors, Estrogen/genetics , Receptors, Glucocorticoid/genetics , Receptors, Progesterone/genetics
17.
Endocr Relat Cancer ; 2018 Jul 10.
Article in English | MEDLINE | ID: mdl-29991638

ABSTRACT

There is a consensus that progestins and thus their cognate receptor molecules, the progesterone receptors (PR), are essential in the development of the adult mammary gland and regulators of proliferation and lactation. However, a role for natural progestins in breast carcinogenesis remains poorly understood. A hint to that possible role came from studies in which the synthetic progestin medroxyprogesterone acetate was associated with an increased breast cancer risk in women under hormone replacement therapy. However, progestins have been also used for breast cancer treatment and to inhibit the growth of several experimental breast cancer models. More recently, PR have been shown to be regulators of estrogen receptor signaling. With all this information, the question is how can we target PR, and if so, which patients may benefit from such an approach? PR are not single unique molecules. Two main PR isoforms have been characterized, PRA and PRB, that exert different functions and the relative abundance of one isoform respect to the other determines the response of PR agonists and antagonists. Immunohistochemistry with standard antibodies against PR do not discriminate between isoforms. In this review, we summarize the current knowledge on the expression of both PR isoforms in mammary glands, in experimental models of breast cancer and in breast cancer patients, to better understand how the PRA/PRB ratio can be exploited therapeutically to design personalized therapeutic strategies.

18.
Horm Cancer ; 9(5): 338-348, 2018 10.
Article in English | MEDLINE | ID: mdl-29956066

ABSTRACT

Endocrine resistance may develop as a consequence of enhanced growth factor signaling. Fibroblast growth factor 2 (FGF2) consists of a low and several high molecular weight forms (HMW-FGF2). We previously demonstrated that antiprogestin-resistant mammary carcinomas display lower levels of progesterone receptor A isoforms (PRA) than B isoforms (PRB). Our aim was to evaluate the role of FGF2 isoforms in breast cancer progression. We evaluated FGF2 expression, cell proliferation, and pathway activation in models with different PRA/PRB ratios. We performed lentiviral infections of different FGF2 isoforms using the human hormone-responsive T47D-YA cells, engineered to only express PRA, and evaluated tumor growth, metastatic dissemination, and endocrine responsiveness. We assessed FGF2 expression and localization in 81 human breast cancer samples. Antiprogestin-resistant experimental mammary carcinomas with low PRA/PRB ratios and T47D-YB cells, which only express PRB, displayed higher levels of HMW-FGF2 than responsive variants. HMW-FGF2 overexpression in T47D-YA cells induced increased tumor growth, lung metastasis, and antiprogestin resistance compared to control tumors. In human breast carcinomas categorized by their PRA/PRB ratio, we found nuclear FGF2 expression in 55.6% of tumor cells. No differences were found between nuclear FGF2 expression and Ki67 proliferation index, tumor stage, or tumor grade. In low-grade tumor samples, moderate to high nuclear FGF2 levels were associated to carcinomas with low PRA/PRB ratio. In conclusion, we show that HMW-FGF2 isoforms are PRB targets which confer endocrine resistance and are localized in the nuclei of breast cancer samples. Hence, targeting intracellular FGF2 may contribute to overcome tumor progression.


Subject(s)
Breast Neoplasms/genetics , Fibroblast Growth Factor 2/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Molecular Weight
19.
Curr Cancer Drug Targets ; 18(10): 979-987, 2018.
Article in English | MEDLINE | ID: mdl-29237381

ABSTRACT

BACKGROUND: Endocrine resistance and metastatic dissemination comprise major clinical challenges for breast cancer treatment. The fibroblast growth factor receptor family (FGFR) consists of four tyrosine kinase transmembrane receptors, involved in key biological processes. Genomic alterations in FGFR have been identified in advanced breast cancer and thus, FGFR are an attractive therapeutic target. However, the efficacy of FGFR inhibitors on in vivo tumor growth is still controversial. OBJECTIVE: The purpose of this study was to evaluate the role of FGFR in tumor growth and breast cancer progression. METHODS: Cell proliferation was assessed by 3H-thymidine uptake and cell counting in primary cultures of endocrine resistant mammary carcinomas and a human cell line, respectively. Tumor transplants and cell injections were used to determine in vivo growth and spontaneous metastasis. FGFR1-3 and αSMA expression were evaluated on primary tumors by immunohistochemistry. RESULTS: Antiprogestin resistant murine transplants and a human xenograft express high levels of total FGFR1-3. In vitro treatment with the FGFR inhibitor, BGJ398, impaired cell proliferation of resistant variants versus vehicle. In vivo, versus control, BGJ398 treatment decreased one out of four resistant tumors, however all tumors showed a decreased epithelial/stromal ratio. Finally, in a model of hormone resistant mammary cancer that spontaneously metastasizes to the lung, BGJ398 decreased the number of mice with lung metastasis. CONCLUSION: FGFR inhibitors are promising tools that require further investigation to identify sensitive tumors. These studies suggest that targeting FGFR combined with other targeted therapies will be useful to impair breast cancer progression.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Phenylurea Compounds/metabolism , Phenylurea Compounds/therapeutic use , Pyrimidines/metabolism , Pyrimidines/therapeutic use , Receptors, Fibroblast Growth Factor/metabolism , Analysis of Variance , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chi-Square Distribution , Disease Progression , Drug Resistance, Neoplasm , Female , Humans , Mice , Neoplasm Metastasis , Phenylurea Compounds/pharmacology , Pyrimidines/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects
20.
PLoS One ; 8(5): e64049, 2013.
Article in English | MEDLINE | ID: mdl-23691144

ABSTRACT

The basic component of Silastic® glue (Dow Corning) used to prepare Silastic® pellets is polydimethylsiloxane. This compound is also present in other commercial adhesives such as FASTIX® (Akapol SA) that are available in any store for that category. In the present study we developed low cost, easy to prepare handmade steroid pellets (HMSP) by mixing 17ß-estradiol, progesterone or other synthetic steroids with FASTIX® adhesive. We assessed serum levels of 17ß-estradiol, progesterone, prolactin and luteinizing hormone in ovariectomized mice treated for 24 and 48 h or 7, 14 and 28 days with 20 µg or 5 mg of 17ß-estradiol or 5 mg progesterone HMSP. We found a time dependent and significant increase in the levels of both natural hormones, and a downregulation of serum luteinizing hormone levels, while both 17ß-estradiol doses increased serum prolactin. Uterine weights at sacrifice and histological examination of the uteri and the mammary glands correlated with estrogen or progestin action. Finally, we evaluated the biological effects of HMSP compared to commercial pellets or daily injections in the stimulation or inhibition of hormone dependent mammary tumor growth, and found that HMSP were as effective as the other methods of hormone administration. These data show that HMSP represent a useful, low cost, easily accessible method for administering steroids to mice.


Subject(s)
Chemistry, Pharmaceutical , Drug Costs , Estradiol/administration & dosage , Progesterone/administration & dosage , Animals , Body Weight/drug effects , Cell Division/drug effects , Estradiol/pharmacology , Female , Gonadal Steroid Hormones/blood , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Organ Size/drug effects , Progesterone/pharmacology , Radioimmunoassay
SELECTION OF CITATIONS
SEARCH DETAIL
...