Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
J Hematol Oncol ; 14(1): 186, 2021 11 06.
Article in English | MEDLINE | ID: mdl-34742344

ABSTRACT

Poly ADP-ribose polymerase inhibitors (PARPi) have transformed ovarian cancer (OC) treatment, primarily for tumours deficient in homologous recombination repair. Combining VEGF-signalling inhibitors with PARPi has enhanced clinical benefit in OC. To study drivers of efficacy when combining PARP inhibition and VEGF-signalling, a cohort of patient-derived ovarian cancer xenografts (OC-PDXs), representative of the molecular characteristics and drug sensitivity of patient tumours, were treated with the PARPi olaparib and the VEGFR inhibitor cediranib at clinically relevant doses. The combination showed broad anti-tumour activity, reducing growth of all OC-PDXs, regardless of the homologous recombination repair (HRR) mutational status, with greater additive combination benefit in tumours poorly sensitive to platinum and olaparib. In orthotopic models, the combined treatment reduced tumour dissemination in the peritoneal cavity and prolonged survival. Enhanced combination benefit was independent of tumour cell expression of receptor tyrosine kinases targeted by cediranib, and not associated with change in expression of genes associated with DNA repair machinery. However, the combination of cediranib with olaparib was effective in reducing tumour vasculature in all the OC-PDXs. Collectively our data suggest that olaparib and cediranib act through complementary mechanisms affecting tumour cells and tumour microenvironment, respectively. This detailed analysis of the combined effect of VEGF-signalling and PARP inhibitors in OC-PDXs suggest that despite broad activity, there is no dominant common mechanistic inter-dependency driving therapeutic benefit.


Subject(s)
Antineoplastic Agents/therapeutic use , Ovarian Neoplasms/drug therapy , Phthalazines/therapeutic use , Piperazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Cell Line, Tumor , Female , Genes, BRCA1/drug effects , Genes, BRCA2/drug effects , Humans , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Vascular Endothelial Growth Factor A/metabolism
2.
Gynecol Endocrinol ; 37(4): 349-352, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33084436

ABSTRACT

OBJECTIVE: To test whether recombinant anti-Müllerian hormone (rAMH) could exert an inhibitory function on BRCA1/2 expression in human ovarian cortex. METHODS: Pilot study on ovariectomized nude mice xenotransplanted with human vitrified/warmed ovarian cortex and treated with rAMH via infusion pump. Twelve nude mice were ovariectomized and Alzet pumps delivering 1.23 mcg rAMH/day to reach a serum concentration of 17.5 ng/mL, or placebo (controls), were inserted intraabdominally. Previously vitrified/warmed 2x2 mm ovarian cortex fragments were transplanted on day 7 and then harvested on day 14 after pump placement. PCR analyses determined mRNA levels for BRCA1 and BRCA2 in the human ovarian cortex. RESULTS: In mice treated with rAMH, BRCA1 expression was significantly lower (0.196 fg/µg RNA, IQR 0.158, 0.236) than in controls (0.544 fg/µg RNA, IQR 0.458, 0.554; p = .030), while BRCA2 expression remained similar in rAMH mice (5.355 fg/µg RNA, IQR 4.479, 6.230) and in controls (4.011 fg/µg RNA, IQR 3.650, 4.182; p = .327). CONCLUSION: Administration of rAMH in the peri-transplant period caused downregulation of BRCA1, but not of BRCA2 expression, in human ovarian cortex. These results help our understanding of DNA repair mechanism in the ovarian cortex and identify AMH's possible protective effect on ovarian reserve in BRCA1 mutation carriers.


Subject(s)
Anti-Mullerian Hormone/pharmacology , Gene Expression Regulation/drug effects , Genes, BRCA1/drug effects , Genes, BRCA2/drug effects , Ovary/drug effects , RNA, Messenger/drug effects , Adolescent , Animals , Female , Humans , Mice , Mice, Nude , Ovary/transplantation , Pilot Projects , RNA, Messenger/metabolism
3.
Eur J Obstet Gynecol Reprod Biol ; 230: 212-216, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29631794

ABSTRACT

The endocrine background of breast cancer has raised questions about the increase in risk that might bear the use of hormonal contraceptives. This has been a particular issue in the case of young women, who constitute the population of contraceptive consumers. Observational studies have been the main source of evidence, which has mainly limited to the combined estrogen-progestogen preparations, the popular pill. Studies in the 80's and 90's of the past century found a small, around a 20%, increase in risk. The translation in absolute number of excess cases has been exiguous because the prevalence of the disease is relatively small in premenopausal women. Moreover, the risk slowly seemed to disappear after 5-10 years of use. The more sophisticated analyses provided by new technologies, together with the powerful central registries in some countries, has confirmed increased risk of similar size. Recent preparations, with lower doses of estrogens and new progestogenic molecules, have not substantially modified the risk size. The impact of progestogen only alternatives, either pills or progestogen-loaded intrauterine devices, seems to be similar, but the evidence is still insufficient. Whether there is a preferential effect on histological or molecular subtypes of breast tumours is being debated yet. The data on women at higher risk, either with mutations of the BRCA1/2 genes or with familial weight, have not found specific response patterns, but the experience is still meagre. It is of interest that long-term follow up data on women who enrolled in the initial cohorts, like that of the Royal College of General Practitioners', have shown a considerable protection against cancer of the ovary (relative risk, RR 0.67), endometrium (RR 0.66), or colorectum (RR 0.81).


Subject(s)
Breast Neoplasms/chemically induced , Contraceptives, Oral, Hormonal/adverse effects , Adult , Breast Neoplasms/genetics , Female , Genes, BRCA1/drug effects , Genes, BRCA2/drug effects , Humans , Mutation , Risk Factors , Time Factors , Young Adult
4.
Sci Rep ; 7(1): 17724, 2017 12 18.
Article in English | MEDLINE | ID: mdl-29255221

ABSTRACT

Carboplatin, a second-generation platinum agent, has been used as a cancer therapy for decades and exhibits strong anti-tumor activity. However, the wide application of carboplatin is largely limited due to its side effects, especially myelosuppression. Here, we combined carboplatin with curcumin, a natural product that improves tumor-induced anemia, for the treatment of fibrosarcoma to improve the side effects of carboplatin. We first examined the synergistic and attenuated effects of the two agents in a T241-bearing mouse model. The combination therapy caused no obvious synergistic effect, but curcumin significantly improved the survival rate of carboplatin-treated mice. Histologic analysis of the kidney and bone marrow revealed that curcumin improved carboplatin-induced myelosuppression but did not affect the kidney. To determine the mechanism involved, we introduced a probe derived from curcumin to identify its targets in bone marrow cells and the results provided us a clue that curcumin might affect the DNA repair pathway. Western blot analysis revealed that curcumin up-regulated BRCA1, BRCA2 and ERCC1 expression in bone marrow. In conclusion, curcumin attenuates carboplatin-induced myelosuppression by activating the DNA repair pathway in bone marrow cells.


Subject(s)
Curcumin/pharmacology , DNA Repair/drug effects , Fibrosarcoma/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , Carboplatin/pharmacology , Cell Culture Techniques , Cell Line, Tumor , Cell Survival/drug effects , China , DNA-Binding Proteins/drug effects , Disease Models, Animal , Drug Synergism , Endonucleases/drug effects , Genes, BRCA1/drug effects , Genes, BRCA2/drug effects , Granulocyte Precursor Cells/drug effects , Mice , Mice, Inbred C57BL
5.
Curr Opin Obstet Gynecol ; 26(3): 125-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24752005

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to highlight recent research and insights into the relationship between fertility drug use and ovarian cancer risk. RECENT FINDINGS: Results from two large case-control studies provided further evidence that fertility drug use does not significantly contribute to risk of ovarian cancer among the majority of women when adjusting for known confounding factors. However, questions regarding the effect on certain subgroups, including long-term fertility drug users, women who remain nulligravid after fertility treatment, women with BRCA1 or BRCA2 mutations and borderline ovarian tumours, still remain. In addition, it may currently just be too early to determine whether there is an association between fertility drug use and ovarian cancer risk given that many of the exposed women are only now beginning to reach the ovarian cancer age range. SUMMARY: Whether use of fertility drugs increases the risk of ovarian cancer is an important question that requires further investigation, in particular given the large number of women utilizing fertility treatments. Fortunately, results from recent studies have been mainly reassuring. Large well designed studies with sufficient follow-up time are needed to further evaluate the effects of fertility treatments within subgroups defined by patient and tumour characteristics.


Subject(s)
Clomiphene/adverse effects , Estrogens/adverse effects , Fertility Agents, Female/adverse effects , Gonadotropins/metabolism , Ovarian Neoplasms/epidemiology , Ovulation Induction/adverse effects , Adult , Case-Control Studies , Clomiphene/administration & dosage , Estrogens/administration & dosage , Female , Fertility Agents, Female/administration & dosage , Genes, BRCA1/drug effects , Genes, BRCA2/drug effects , Humans , Infertility, Female/therapy , Mutation , Ovarian Neoplasms/chemically induced , Ovarian Neoplasms/prevention & control , Ovulation Induction/methods , Parity , Pregnancy , Risk Factors , Time Factors
6.
Postepy Hig Med Dosw (Online) ; 66: 311-21, 2012 May 30.
Article in Polish | MEDLINE | ID: mdl-22706117

ABSTRACT

Poly-ADP-ribose polymerases (PARP) are involved in a number of processes that are vital for every living cell. Once activated by the presence of DNA damage they trigger poly-ADP-ribosylation of various proteins which are crucial for DNA repair, preserving of genom integrity, regulation of transcription, proliferation and apoptosis. PARP1, which is the best known enzyme of PARP protein family, plays a role in single-strand breaks (SSB) repair. Decrease of its activity results in accumulation of single strand DNA breaks (SSB) which leads as a consequence to double-strand breaks (DSBs). This disorder is particularly harmful to cells with deficiency of BRCA1/2 protein which is involved in repair of DNA double-strand breaks. This phenomenon is an example of "synthetic lethality" concept and contributes to research on application of PARP inhibitors in treatment of cancers associated with BRCA1/2 protein defect (breast or ovarian cancer). Noticed synergism between PARP inhibitors and genotoxic chemotherapy or radiotherapy determined another direction of research on application of these medicaments. After promising results of phase I and II trials with most commonly investigated PARP inhibitors--iniparib and olaparib--which recruited patients with triple negative breast cancer and ovarian cancer, further studies started. This paper presents theoretical basis of PARP inhibitors action as well as critical review of most important clinical trials of these medicaments.


Subject(s)
Breast Neoplasms/drug therapy , DNA Repair/physiology , Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Antineoplastic Agents/therapeutic use , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Single-Stranded/drug effects , DNA Damage/drug effects , DNA Repair/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Genes, BRCA1/drug effects , Genes, BRCA1/physiology , Genes, BRCA2/drug effects , Genes, BRCA2/physiology , Humans , Mutation , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics
7.
Postepy Hig Med Dosw (Online) ; 66: 372-84, 2012 Jun 15.
Article in Polish | MEDLINE | ID: mdl-22706123

ABSTRACT

 A majority of currently used anticancer drugs belong to a group of chemical agents that damage DNA. The efficiency of the treatment is limited by effective DNA repair systems functioning in cancer cells. Many chemotherapeutic compounds cause strong systemic toxicity. Therefore, there is still a need for new anticancer agents which are less toxic for nontransformed cells and selectively kill cancer cells. One of the most promising molecular targets in cancer therapy is poly(ADP-ribose) polymerases (PARP). PARP play an essential role in repairing DNA strand breaks. Small molecule inhibitors of these enzymes have been developed and have proved to be extremely toxic for cancer cells that lack the functional BRCA1 and BRCA2 proteins that are involved in homologous recombination, a complex repair mechanism of DNA double strand breaks. Mutations in BRCA1/2 genes are associated with genetically inherited breast and ovarian cancers. Therefore PARP inhibitors may prove to be very effective and selective in the treatment of these cancer types. This review is focused on the function of BRCA1/2 proteins and poly(ADP-ribose) polymerases in DNA repair systems, especially in the homologous recombination process. A short history of the studies that led to synthesis of high specificity small molecule PARP inhibitors is also presented, as well as the results of clinical trials concerning the most effective PARP inhibitors in view of their potential application in oncological treatment, particularly breast cancers.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Genes, BRCA1/drug effects , Genes, BRCA2/drug effects , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors , Antineoplastic Agents/therapeutic use , DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , Enzyme Inhibitors/therapeutic use , Female , Humans
8.
Curr Opin Oncol ; 24(3): 316-24, 2012 May.
Article in English | MEDLINE | ID: mdl-22476188

ABSTRACT

PURPOSE OF REVIEW: The success of poly(ADP-ribose) polymerase inhibition in BRCA1 or BRCA2 deficient tumors as an anticancer strategy provided proof-of-concept for a synthetic lethality approach in oncology. There is therefore now active interest in expanding this approach to include other agents targeting the DNA damage response (DDR). We review lessons learnt from the development of inhibitors against DNA damage response mechanisms and envision the future of DNA repair inhibition in oncology. RECENT FINDINGS: Preclinical synthetic lethality screens may potentially identify the best combinations of DNA-damaging drugs with inhibitors of DNA repair and the DDR or two agents acting within the DDR. Efforts are currently being made to establish robust and cost-effective assays that may be implemented within appropriate time-scales in parallel with future clinical studies. Detection of relevant mutations in a high-throughput manner, such as with next-generation sequencing for genes implicated in homologous recombination, including BRCA1, BRCA2, and ataxia telangiectasia mutated is anticipated. Novel approaches targeting the DDR are currently being evaluated and inhibitors of ATM, RAD51 and DNA-dependent protein kinase are now in early drug discovery and development. SUMMARY: There remains great enthusiasm in oncology practice for pursuing the strategy of synthetic lethality. The future development of antitumor agents targeting the DDR should include detailed correlative biomarker work within early phase clinical studies wherever possible, with clear attempts to identify doses at which robust target modulation is observed.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage/drug effects , DNA Repair/drug effects , Neoplasms/drug therapy , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/antagonists & inhibitors , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Binding Proteins/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/pharmacology , Genes, BRCA1/drug effects , Genes, BRCA2/drug effects , Humans , Neoplasms/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rad51 Recombinase/antagonists & inhibitors , Tumor Suppressor Proteins/antagonists & inhibitors
9.
J Cancer Res Clin Oncol ; 138(5): 811-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22274865

ABSTRACT

BACKGROUND: Methotrexate is an inhibitor of folic acid metabolism. Homologous recombination is one of the most important ways to repair double-stranded breaks in DNA and influence the radio- and chemosensitivity of tumor cells. But the relationship between methotrexate and homologous recombination repair has not been elucidated. METHODS: Induction of double-strand breaks by methotrexate in HOS cells is assessed by the neutral comet assay. Inhibition of subnuclear repair foci by methotrexate is measured by immunofluorescence. Western blot and quantitative real-time PCR are conducted to detect whether methotrexate affects the expression level of genes involved in homologous recombination. In addition, we used a pCMV3xnls-I-SceI construct to determine whether methotrexate directly inhibits the process of homologous recombinational repair in cells, and the sensitivity to methotrexate in the Ku80-deficient cells is detected using clonogenic survival assays. RESULTS: The result showed that methotrexate can regulate the repair of DNA double-strand breaks after radiation exposure, and methotrexate inhibition caused the complete inhibition of subnuclear repair foci in response to ionizing radiation. Mechanistic investigation revealed that methotrexate led to a significant reduction in the transcription of RAD51 genes. Treatment with methotrexate resulted in a decreased ability to perform homology-directed repair of I-SceI-induced chromosome breaks. In addition, enhancement of cell death was observed in Ku mutant cells compared to wild-type cells. CONCLUSIONS: These results demonstrate that methotrexate can affect homologous recombination repair of DNA double-strand breaks by controlling the expression of homologous recombination-related genes and suppressing the proper assembly of homologous recombination-directed subnuclear foci.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Homologous Recombination/drug effects , Methotrexate/pharmacology , Neoplasms/genetics , Rad51 Recombinase/genetics , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor , DNA End-Joining Repair/drug effects , DNA End-Joining Repair/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Genes, BRCA2/drug effects , Homologous Recombination/genetics , Humans , Neoplasms/pathology , RNA, Small Interfering/pharmacology , Rad51 Recombinase/antagonists & inhibitors , Rad51 Recombinase/metabolism , Rad52 DNA Repair and Recombination Protein/genetics , Recombinational DNA Repair/drug effects , Recombinational DNA Repair/genetics
10.
J Natl Cancer Inst Monogr ; 2011(43): 79-81, 2011.
Article in English | MEDLINE | ID: mdl-22043047

ABSTRACT

In 2005, 100,514 Australians were diagnosed with cancer, and over 10,000 of these cancers will be due to heritable causes. The impact of familial cancer by definition extends beyond the individual, affecting tens of thousands of parents, siblings, and children. The study of familial cancer causes has arguably made the greatest single contribution to our understanding of cancer biology. This knowledge is used clinically to guide investment in screening and prevention, as well as being translated into new treatments.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Medical Oncology , Translational Research, Biomedical , Australia/epidemiology , Breast Neoplasms/epidemiology , Clinical Trials as Topic/methods , Clinical Trials as Topic/trends , Female , Genes, BRCA1/drug effects , Genes, BRCA2/drug effects , Genetic Predisposition to Disease , Heterozygote , Humans , Medical Oncology/methods , Medical Oncology/organization & administration , Medical Oncology/trends , Mutation , New Zealand/epidemiology , Patient Selection , Translational Research, Biomedical/methods , Translational Research, Biomedical/trends
11.
Rev Med Suisse ; 7(296): 1137-40, 2011 May 25.
Article in French | MEDLINE | ID: mdl-21721203

ABSTRACT

PARP inhibitors are novel drugs under development in oncology, particularly against breast and ovarian cancer. They act on the DNA repair mechanisms in synergy with the loss of BRCA function of the tumor cells, thereby inducing a genetic instability that leads to cell death. The clinical benefit of PARP inhibitors has been demonstrated for breast and ovarian cancer in BRCA germline mutation carriers. Their use in sporadic triple negative breast cancers, that share similarities with BRCA1 mutated tumors, is currently investigated with encouraging preliminary results.


Subject(s)
Breast Neoplasms/drug therapy , Enzyme Inhibitors/therapeutic use , Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Breast Neoplasms/genetics , Evidence-Based Medicine , Female , Genes, BRCA1/drug effects , Genes, BRCA2/drug effects , Humans , Ovarian Neoplasms/genetics , Treatment Outcome
14.
Curr Opin Pharmacol ; 8(4): 363-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18644251

ABSTRACT

Poly (ADP-ribose) Polymerase (PARP) has a well-established role in DNA repair processes, and small molecule inhibitors of PARP have been developed as chemotherapy sensitisers for the treatment of cancer. The subsequent demonstration that PARP inhibition is selective for BRCA1 or BRCA2 deficiency suggests that PARP inhibitors may be particularly useful for the treatment of cancer with BRCA mutations. This would represent one of the first clinically implemented examples of a synthetic lethal approach for cancer treatment. However, there are still unanswered questions surrounding PARP inhibitors, namely the levels of specificity and potency that are required to elicit BRCA selectivity. The recent identification of mechanisms of cellular resistance to PARP inhibitors may provide indications as to how these drugs may be best used in the clinic.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Enzyme Inhibitors/therapeutic use , Genes, BRCA1/drug effects , Genes, BRCA2/drug effects , Humans , Neoplasms/genetics , Neoplasms/prevention & control , Substrate Specificity
16.
Cancer Epidemiol Biomarkers Prev ; 15(10): 1863-70, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17021353

ABSTRACT

BACKGROUND: Understanding the effect of oral contraceptives on risk of breast cancer in BRCA1 or BRCA2 mutation carriers is important because oral contraceptive use is a common, modifiable practice. METHODS: We studied 497 BRCA1 and 307 BRCA2 mutation carriers, of whom 195 and 128, respectively, had been diagnosed with breast cancer. Case-control analyses were conducted using unconditional logistic regression with adjustments for family history and familial relationships and were restricted to subjects with a reference age under 50 years. RESULTS: For BRCA1 mutation carriers, there was no significant association between risk of breast cancer and use of oral contraceptives for at least 1 year [odds ratio (OR), 0.77; 95% confidence interval (95% CI), 0.53-1.12] or duration of oral contraceptive use (P(trend) = 0.62). For BRCA2 mutation carriers, there was no association with use of oral contraceptives for at least 1 year (OR, 1.62; 95% CI, 0.90-2.92); however, there was an association of elevated risk with oral contraceptive use for at least 5 years (OR, 2.06; 95% CI, 1.08-3.94) and with duration of use (OR(trend) per year of use, 1.08; P = 0.008). Similar results were obtained when we considered only use of oral contraceptives that first started in 1975 or later. CONCLUSIONS: We found no evidence overall that use of oral contraceptives for at least 1 year is associated with breast cancer risk for BRCA1 and BRCA2 mutation carriers before age 50. For BRCA2 mutation carriers, use of oral contraceptives may be associated with an increased risk of breast cancer among women who use them for at least 5 years. Further studies reporting results separately for BRCA1 and BRCA2 mutation carriers are needed to resolve this important issue.


Subject(s)
Breast Neoplasms/genetics , Carcinoma in Situ/genetics , Carcinoma, Ductal, Breast/genetics , Contraceptives, Oral/pharmacology , Genes, BRCA1/drug effects , Genes, BRCA2/drug effects , Mutation/drug effects , Adult , Australia/epidemiology , Breast Neoplasms/epidemiology , Canada/epidemiology , Carcinoma in Situ/epidemiology , Carcinoma, Ductal, Breast/epidemiology , Case-Control Studies , Female , Genetic Predisposition to Disease , Heterozygote , Humans , Logistic Models , Middle Aged , Receptors, Estrogen/drug effects , Receptors, Progesterone/drug effects , Risk Factors , Surveys and Questionnaires , Time Factors , United States/epidemiology
17.
Int J Cancer ; 119(5): 1005-13, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16570282

ABSTRACT

Ovarian cancer has a high rate of recurrence and subsequent mortality following chemotherapy despite intense efforts to improve treatment outcomes. Recent trials have suggested that retinoids, especially 4-(N-hydroxyphenyl) retinamide (4-HPR), play an important role as a chemopreventive agent and are currently being used in clinical trials for ovarian cancer chemoprevention as well as treatment. This study examines the mechanism of its activity in premalignant and cancer cells. We investigated the modulation of gene expression by 4-HPR in immortalized ovarian surface epithelial (IOSE) cells and ovarian cancer (OVCA433) cells with DNA microarray. Real time RT-PCR and western blotting were used to confirm the microarray results and metabolic changes were examined with optical fluorescence spectroscopy. 4-HPR resulted in an up-regulation of expression of proapoptotic genes and mitochondrial uncoupling protein in OVCA433 cells and modulation of the RXR receptors in IOSE cells, and down-regulation of mutant BRCA genes in both IOSE and OVCA433 cells. 4-HPR had a larger effect on the redox in the 433 cells compared to IOSE. These findings suggest that 4-HPR acts through different mechanisms in premalignant ovarian surface cells and cancer cells, with a preventive effect in premalignant cells and a treatment effect in cancer cells.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Fenretinide/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Ovarian Neoplasms/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Carrier Proteins/drug effects , Cell Cycle , Cell Line, Tumor , Down-Regulation/drug effects , Female , Genes, BRCA1/drug effects , Genes, BRCA2/drug effects , Humans , Ion Channels , Membrane Proteins/drug effects , Mitochondrial Proteins , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/prevention & control , Retinoid X Receptors/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Fluorescence , Uncoupling Protein 1 , Up-Regulation/drug effects
18.
Br J Nutr ; 95(2): 406-13, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16469160

ABSTRACT

Breast cancer is the most common cancer in women and a significant cause of death. Mutations of the oncosuppressor genes BRCA1 and BRCA2 are associated with a hereditary risk of breast cancer, and dysregulation of their expression has been observed in sporadic cases. Soya isoflavones have been shown to inhibit breast cancer in studies in vitro, but associations between the consumption of isoflavone-containing foods and breast cancer risk have varied in epidemiological studies. Soya is a unique source of the phytoestrogens daidzein (4',7-dihydroxyisoflavone) and genistein (4',5,7-trihydroxyisoflavone), two molecules that are able to inhibit the proliferation of human breast cancer cells in vitro. The aim of the present study was to determine the effects of genistein (5 microg/ml) and daidzein (20 microg/ml) on transcription in three human breast cell lines (one dystrophic, MCF10a, and two malignant, MCF-7 and MDA-MB-231) after 72 h treatment. The different genes involved in the BRCA1 and BRCA2 pathways (GADD45A, BARD1, JUN, BAX, RB1, ERalpha, ERbeta, BAP1, TNFalpha, p53, p21Waf1/Cip1, p300, RAD51, pS2, Ki-67) were quantified by real-time quantitative RT-PCR, using the TaqMan method and an ABI Prism 7700 Sequence Detector (Applied Biosystems). We observed that, in response to treatment, many of these genes were overexpressed in the breast cancer cell lines (MCF-7 and MDA-MB-231) but not in the dystrophic cell line (MCF10a).


Subject(s)
Anticarcinogenic Agents/pharmacology , Breast Neoplasms/genetics , Genes, Tumor Suppressor/drug effects , Genistein/pharmacology , Glycine max/chemistry , Isoflavones/pharmacology , Phytoestrogens/pharmacology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Genes, BRCA1/drug effects , Genes, BRCA1/physiology , Genes, BRCA2/drug effects , Genes, BRCA2/physiology , Genes, Tumor Suppressor/physiology , Humans , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription, Genetic/drug effects
20.
Nutr Cancer ; 45(2): 247-55, 2003.
Article in English | MEDLINE | ID: mdl-12881020

ABSTRACT

A high intake of isoflavones is associated with a reduction of breast cancer among Japanese women. The aim of this study was to quantify BRCA2 tumor suppressor gene expression after treatment of cells with the phytoestrogens daidzein and genistein, the main compounds of soy. The effects of 5 microg/ml genistein and 20 microg/ml daidzein on BRCA2 expression were studied in two human mammary tumor cell lines, MCF7 and MDA-MB-231, and one normal human breast epithelial cell line, MCF10a. BRCA2 mRNA was evaluated by quantitative real time RT-PCR and the amount of BRCA2 protein was measured by affinity chromatography. With Genistein, we observed a 60% increase of BRCA2 mRNA in MDA-MB-231 and MCF10a, which are, respectively, estrogen receptors alpha-/beta+ and alpha-/beta-, and no variation in MCF7, which is ERalpha+/beta+. Dairzein had no effect on BRCA2 mRNA expression. The level of BRCA2 protein with both food components also remained unchanged in all three cell lines. This suggests regulation of BRCA2 between the mRNA and protein levels. Treatment with actinomycin D and cycloheximide demonstrated that the increase in BRCA2 mRNA was not blocked by cycloheximide, indicating that de novo protein synthesis was required in MDA-MB-23, although de novo synthesis was not required in MCF10a for the genistein. With actinomycin D, genistein had a positive effect on the transcriptional level of BRCA2 mRNA in MDA-MB-231 and MCF10a. The use of an anti-estrogen suggested that the action of daidzein and genistein might not be mediated through the ER.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Genes, BRCA2/drug effects , Genistein/pharmacology , Isoflavones/pharmacology , Antibiotics, Antineoplastic/pharmacology , BRCA2 Protein/analysis , BRCA2 Protein/genetics , Breast Neoplasms/metabolism , Cell Division/drug effects , Cell Line , Chromatography, Affinity , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Humans , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...