Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 161
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 24(4)2023 Feb 10.
Article in English | MEDLINE | ID: mdl-36835001

ABSTRACT

Radiation resistance and radiation-related side effects warrant research into alternative strategies in the application of this modality to cancer treatment. Designed in silico to improve the pharmacokinetics and anti-cancer properties of 2-methoxyestradiol, 2-ethyl-3-O-sulfamoyl-estra-1,3,5(10)16-tetraene (ESE-16) disrupts microtubule dynamics and induces apoptosis. Here, we investigated whether pre-exposure of breast cancer cells to low-dose ESE-16 would affect radiation-induced deoxyribonucleic acid (DNA) damage and the consequent repair pathways. MCF-7, MDA-MB-231, and BT-20 cells were exposed to sub-lethal doses of ESE-16 for 24 h before 8 Gy radiation. Flow cytometric quantification of Annexin V, clonogenic studies, micronuclei quantification, assessment of histone H2AX phosphorylation and Ku70 expression were performed to assess cell viability, DNA damage, and repair pathways, in both directly irradiated cells and cells treated with conditioned medium. A small increase in apoptosis was observed as an early consequence, with significant repercussions on long-term cell survival. Overall, a greater degree of DNA damage was detected. Moreover, initiation of the DNA-damage repair response was delayed, with a subsequent sustained elevation. Radiation-induced bystander effects induced similar pathways and were initiated via intercellular signaling. These results justify further investigation of ESE-16 as a radiation-sensitizing agent since pre-exposure appears to augment the response of tumor cells to radiation.


Subject(s)
Breast Neoplasms , DNA Damage , DNA Repair , Estrenes , Female , Humans , 2-Methoxyestradiol/analogs & derivatives , 2-Methoxyestradiol/pharmacology , Apoptosis , Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Cell Line, Tumor , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Estrenes/pharmacology , Estrenes/therapeutic use , Sulfonamides/pharmacology , Sulfonamides/therapeutic use
2.
BMC Vet Res ; 16(1): 284, 2020 Aug 10.
Article in English | MEDLINE | ID: mdl-32778114

ABSTRACT

BACKGROUND: This is the first report about a vaginal leiomyoma concomitant with an ovarian luteoma in a bitch. CASE PRESENTATION: A 11-year-old intact female Labrador retriever was referred because of anuria, constipation and protrusion of a vaginal mass through the vulvar commissure. The bitch had high serum progesterone concentration (4.94 ng/ml). Because of the possibility of progesterone responsiveness causing further increase of the vaginal mass and since the bitch was a poor surgical candidate a 10 mg/kg aglepristone treatment was started SC on referral day 1. A computerized tomography showed a 12.7 × 6.5 × 8.3 cm mass causing urethral and rectal compression, ureteral dilation and hydronephrosis. A vaginal leiomyoma was diagnosed on histology. As serum progesterone concentration kept increasing despite aglepristone treatment, a 0.02 ng/mL twice daily IM alfaprostol treatment was started on day 18. As neither treatment showed remission of clinical signs or luteolysis, ovariohysterectomy was performed on referral day 35. Multiple corpora lutea were found on both ovaries. On histology a luteoma was diagnosed on the left ovary. P4 levels were undetectable 7 days after surgery. Recovery was uneventful and 12 weeks after surgery tomography showed a reduction of 86.7% of the vaginal mass. The bitch has been in good health and able to urinate without any complication ever since. CONCLUSIONS: This case demonstrates the importance of identifying progesterone related conditions as well as the importance of judiciously using a combined medical and surgical approach.


Subject(s)
Dog Diseases/pathology , Leiomyoma/veterinary , Luteoma/veterinary , Progesterone/blood , Animals , Dogs , Estrenes/therapeutic use , Female , Hysterectomy/veterinary , Leiomyoma/drug therapy , Leiomyoma/surgery , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/surgery , Ovarian Neoplasms/veterinary , Ovariectomy/veterinary , Progesterone/antagonists & inhibitors , Prostaglandins F/therapeutic use , Vaginal Neoplasms/drug therapy , Vaginal Neoplasms/surgery , Vaginal Neoplasms/veterinary
3.
Hum Reprod ; 34(4): 623-634, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30865281

ABSTRACT

STUDY QUESTION: Can asoprisnil, a selective progesterone receptor modulator, provide clinically meaningful improvements in heavy menstrual bleeding (HMB) associated with uterine fibroids with an acceptable safety profile? SUMMARY ANSWER: Uninterrupted treatment with asoprisnil for 12 months effectively controlled HMB and reduced fibroid and uterine volume with few adverse events. WHAT IS KNOWN ALREADY: In a 3-month study, asoprisnil (5, 10 and 25 mg) suppressed uterine bleeding, reduced fibroid and uterine volume, and improved hematological parameters in a dose-dependent manner. STUDY DESIGN, SIZE, DURATION: In two Phase 3, double-blind, randomized, placebo-controlled, multicentre studies, women received oral asoprisnil 10 mg, asoprisnil 25 mg or placebo (2:2:1) once daily for up to 12 months. PARTICIPANTS/MATERIALS, SETTING, METHODS: Premenopausal women ≥18 years of age in North America with HMB associated with uterine fibroids were included (N = 907). The primary efficacy endpoint was the percentage of women who met all three predefined criteria at 12 months or the final month for patients who prematurely discontinued: (1) ≥50% reduction in monthly blood loss (MBL) by menstrual pictogram, (2) hemoglobin concentration ≥11 g/dL or an increase of ≥1 g/dL, and (3) no interventional therapy for uterine fibroids. Secondary efficacy endpoints included changes in other menstrual bleeding parameters, volume of the largest fibroids, uterine volume and health-related quality of life (HRQL). MAIN RESULTS AND THE ROLE OF CHANCE: In all, 90% and 93% of women in the asoprisnil 10-mg and 25-mg groups, respectively, and 35% of women in the placebo group met the primary endpoint (P < 0.001). Similar results were observed at month 6 (P < 0.001). The percentage of women who achieved amenorrhea in any specified month ranged from 66-78% in the asoprisnil 10-mg group and 83-93% in the asoprisnil 25-mg group, significantly higher than with placebo (3-12%, P < 0.001). Hemoglobin increased rapidly (by month 2) with asoprisnil treatment and was significantly higher versus placebo throughout treatment. The primary fibroid and uterine volumes were significantly reduced from baseline through month 12 with asoprisnil 10 mg (median changes up to -48% and -28%, respectively) and 25 mg (median changes up to -63% and -39%, respectively) versus placebo (median changes up to +16% and +13%, respectively; all P < 0.001). Dose-dependent, significant improvements in HRQL (Uterine Fibroid Symptom and Quality of Life instrument) were observed with asoprisnil treatment. Asoprisnil was generally well tolerated. Endometrial biopsies indicated dose- and time-dependent decreases in proliferative patterns and increases in quiescent or minimally stimulated endometrium at month 12 of treatment. Although not statistically significantly different at month 6, mean endometrial thickness at month 12 increased by ~2 mm in both asoprisnil groups compared with placebo (P < 0.01). This effect was associated with cystic changes in the endometrium on MRI and ultrasonography, which led to invasive diagnostic and therapeutic procedures in some asoprisnil-treated women. LIMITATIONS, REASONS FOR CAUTION: Most study participants were black; few Asian and Hispanic women participated. The study duration may have been insufficient to fully characterize the endometrial effects. WIDER IMPLICATIONS OF THE FINDINGS: Daily uninterrupted treatment with asoprisnil was highly effective in controlling menstrual bleeding, improving anemia, reducing fibroid and uterine volume, and increasing HRQL in women with HMB associated with uterine fibroids. However, this treatment led to an increase in endometrial thickness and invasive diagnostic and therapeutic procedures, with potential unknown consequences. STUDY FUNDING/COMPETING INTEREST(S): This trial was funded by AbbVie Inc. (prior sponsors: TAP Pharmaceutical Products Inc., Abbott Laboratories). E.A. Stewart was a site investigator in the Phase 2 study of asoprisnil and consulted for TAP during the design and conduct of these studies while at Harvard Medical School and Brigham and Women's Hospital. She received support from National Institutes of Health grants HD063312, HS023418 and HD074711 and research funding, paid to Mayo Clinic for patient care costs related to an NIH-funded trial from InSightec Ltd. She consulted for AbbVie, Allergan, Bayer HealthCare AG, Gynesonics, and Welltwigs. She received royalties from UpToDate and the Med Learning Group. M.P. Diamond received research funding for the conduct of the studies paid to the institution and consulted for AbbVie. He is a stockholder and board and director member of Advanced Reproductive Care. He has also received funding for study conduct paid to the institution from Bayer and ObsEva. A.R.W. Williams consulted for TAP and Repros Therapeutics Inc. He has current consultancies with PregLem SA, Gedeon Richter, HRA Pharma and Bayer. B.R. Carr consulted for and received research funding from AbbVie. E.R. Myers consulted for AbbVie, Allergan and Bayer. R.A. Feldman received compensation for serving as a principal investigator and participating in the conduct of the trial. W. Elger was co-inventor of several patents related to asoprisnil. C. Mattia-Goldberg is a former employee of AbbVie and may own AbbVie stock or stock options. B.M. Schwefel and K. Chwalisz are employees of AbbVie and may own AbbVie stock or stock options. TRIAL REGISTRATION NUMBER: NCT00152269, NCT00160381 (clinicaltrials.gov). TRIAL REGISTRATION DATE: 7 September 2005; 8 September 2005. DATE OF FIRST PATIENT'S ENROLMENT: 12 September 2002; 6 September 2002.


Subject(s)
Estrenes/adverse effects , Estrenes/therapeutic use , Leiomyoma/drug therapy , Menorrhagia/drug therapy , Oximes/adverse effects , Oximes/therapeutic use , Receptors, Progesterone/drug effects , Uterine Neoplasms/drug therapy , Administration, Oral , Adult , Double-Blind Method , Endometrium/drug effects , Estrenes/administration & dosage , Female , Follow-Up Studies , Humans , Leiomyoma/complications , Menorrhagia/complications , Middle Aged , Oximes/administration & dosage , Patient Reported Outcome Measures , Premenopause , Quality of Life , Treatment Outcome , Tumor Burden/drug effects , Uterine Neoplasms/complications
4.
Am J Obstet Gynecol ; 218(6): 563-572.e1, 2018 06.
Article in English | MEDLINE | ID: mdl-29274830

ABSTRACT

Uterine fibroids are common in women of reproductive age and can have a significant impact on quality of life and fertility. Although a number of international obstetrics/gynecology societies have issued evidence-based clinical practice guidelines for the management of symptomatic uterine fibroids, many of these guidelines do not yet reflect the most recent clinical evidence and approved indication for one of the key medical management options: the selective progesterone receptor modulator class. This article aims to share the clinical experience gained with selective progesterone receptor modulators in Europe and Canada by reviewing the historical development of selective progesterone receptor modulators, current best practices for selective progesterone receptor modulator use based on available data, and potential future uses for selective progesterone receptor modulators in uterine fibroids and other gynecologic conditions.


Subject(s)
Contraceptive Agents, Female/therapeutic use , Leiomyoma/drug therapy , Norpregnadienes/therapeutic use , Receptors, Progesterone/agonists , Receptors, Progesterone/antagonists & inhibitors , Uterine Neoplasms/drug therapy , Disease Management , Estrenes/therapeutic use , Female , Forecasting , Humans , Mifepristone/therapeutic use , Oximes/therapeutic use , Population Growth , Steroids/therapeutic use
5.
Cell Mol Biol Lett ; 23: 10, 2018.
Article in English | MEDLINE | ID: mdl-29568313

ABSTRACT

BACKGROUND: The majority of novel chemotherapeutics target the cell cycle, aiming to effect arrest and cause apoptosis. One such agent, 2-methoxyestradiol (2ME), has been shown to possess anticancer properties against numerous cancer types, both in vitro and in vivo. Despite its promise, 2ME has exhibited limitations, including low oral bioavailability and rapid hepatic enzymatic inactivation in vivo. A novel sulphamoylated estrogen analog, 2-ethyl-3-O-sulphamoyl-estra-1,3,5(10)16-tetraene (ESE-16), was in silico-designed in our laboratory to overcome these issues. It was then synthesized by a pharmaceutical company and used in an in vitro antiproliferative effect study on a human cervical carcinoma (HeLa) cell line. RESULTS: Cell proliferation data obtained from the crystal violet assay and real-time cell analysis demonstrated that 0.2 µM of ESE-16 had a significant inhibitory effect on the HeLa cells 24 h post-exposure. Immunofluorescence showed that ESE-16 is a microtubule disruptor that causes cells to undergo a mitotic block. Qualitative morphological studies using polarization-optical transmitted light differential interference contrast (PlasDIC) and light microscopy revealed a decrease in cell density and an increase in the number of cells arrested in metaphase. After ESE-16 exposure, hallmarks of apoptosis were also observed, including membrane blebbing, chromatin condensation and the presence of apoptotic bodies. Flow cytometry provided quantitative results from cell cycle progression analysis, indicating cells undergoing apoptosis and cells in the G2/M phase of the cell cycle, confirming cell cycle arrest in metaphase after ESE-16 treatment. Quantification of the ESE-16-mediated upregulation of cyclin B in HeLa cells and spectrophotometric and flow cytometric confirmation of cell death via apoptosis further confirmed the substance's impact. CONCLUSION: ESE-16 exerts its antiproliferative effects through microtubule disruption, which induces a mitotic block culminating in apoptosis. This research provided information on ESE-16 as a potential antitumor agent and on cellular targets that could aid in the design of prospective microtubule-disrupting compounds. Further in vitro and in vivo investigations of this novel compound are needed.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Estrenes/therapeutic use , Sulfonamides/therapeutic use , Uterine Cervical Neoplasms/drug therapy , Apoptosis , Carcinoma/pathology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Female , HeLa Cells , Humans , Uterine Cervical Neoplasms/pathology
6.
Reprod Domest Anim ; 53 Suppl 3: 85-95, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30474341

ABSTRACT

Since most of dystocia end up in caesarean sections (C-sections), the history of any problem during whelping is a good reason to plan in advance a further C-section. Our aim was to confirm that on a large sample and over an extended period of time, mortality in puppies <2 weeks of age was low, born after a planned C-section using aglepristone as a primer. Seventy-four C-sections on 59 different bitches were included. Bitches were monitored during oestrus to estimate the day of ovulation by progesterone assays; 60, 61 or 62 days after ovulation, foetal viability was checked by ultrasonography and progesterone plasma level was measured. None of the bitches was at term (progesterone plasma level >2 ng/ml). An injection of aglepristone was performed in late afternoon to block the effect of progesterone, mimicking its drop at the end of pregnancy. The C-section was conducted the following morning. Twenty-one breeds were represented most of which were bulldogs (26%, 21/74) and Great Danes (16%, 13/74). Four hundred and thirty-five puppies were born. A total of 43/435 puppies died within the first 2 weeks (9.89%). None of the puppies showed any external signs of prematurity. The average number of deaths per litter relative to the date after ovulation was similar (0.5 pups per litter at day 60, 0.7 at day 61, 0.4 at day 62). This study shows that planned C-section after an accurate determination of ovulation and using aglepristone as a primer is a safe procedure for bitches and their offspring. It may be offered to owners if a pregnant bitch is "at risk" of dystocia.


Subject(s)
Animals, Newborn , Cesarean Section/veterinary , Dogs , Estrenes/therapeutic use , Animals , Estrenes/administration & dosage , Female , Pregnancy , Progesterone/blood , Receptors, Progesterone/antagonists & inhibitors , Retrospective Studies , Ultrasonography, Prenatal/veterinary
7.
Am J Hum Genet ; 94(4): 533-46, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24680886

ABSTRACT

Pelizaeus-Merzbacher disease (PMD) is a severe hypomyelinating disease, characterized by ataxia, intellectual disability, epilepsy, and premature death. In the majority of cases, PMD is caused by duplication of PLP1 that is expressed in myelinating oligodendrocytes. Despite detailed knowledge of PLP1, there is presently no curative therapy for PMD. We used a Plp1 transgenic PMD mouse model to test the therapeutic effect of Lonaprisan, an antagonist of the nuclear progesterone receptor, in lowering Plp1 mRNA overexpression. We applied placebo-controlled Lonaprisan therapy to PMD mice for 10 weeks and performed the grid slip analysis to assess the clinical phenotype. Additionally, mRNA expression and protein accumulation as well as histological analysis of the central nervous system were performed. Although Plp1 mRNA levels are increased 1.8-fold in PMD mice compared to wild-type controls, daily Lonaprisan treatment reduced overexpression at the RNA level to about 1.5-fold, which was sufficient to significantly improve the poor motor phenotype. Electron microscopy confirmed a 25% increase in the number of myelinated axons in the corticospinal tract when compared to untreated PMD mice. Microarray analysis revealed the upregulation of proapoptotic genes in PMD mice that could be partially rescued by Lonaprisan treatment, which also reduced microgliosis, astrogliosis, and lymphocyte infiltration.


Subject(s)
Estrenes/therapeutic use , Hormone Antagonists/therapeutic use , Pelizaeus-Merzbacher Disease/drug therapy , Progesterone/antagonists & inhibitors , Animals , Disease Models, Animal , Estrenes/pharmacokinetics , Estrenes/pharmacology , Gene Expression Regulation/drug effects , Hormone Antagonists/pharmacokinetics , Hormone Antagonists/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Proteolipid Protein/genetics , Phenotype , RNA, Messenger/genetics
8.
Cochrane Database Syst Rev ; 4: CD010770, 2017 Apr 26.
Article in English | MEDLINE | ID: mdl-28444736

ABSTRACT

BACKGROUND: Uterine fibroids are smooth muscle tumours arising from the uterus. These tumours, although benign, are commonly associated with abnormal uterine bleeding, bulk symptoms and reproductive dysfunction. The importance of progesterone in fibroid pathogenesis supports selective progesterone receptor modulators (SPRMs) as effective treatment. Both biochemical and clinical evidence suggests that SPRMs may reduce fibroid growth and ameliorate symptoms. SPRMs can cause unique histological changes to the endometrium that are not related to cancer, are not precancerous and have been found to be benign and reversible. This review summarises randomised trials conducted to evaluate the effectiveness of SPRMs as a class of medication for treatment of individuals with fibroids. OBJECTIVES: To evaluate the effectiveness and safety of SPRMs for treatment of premenopausal women with uterine fibroids. SEARCH METHODS: We searched the Specialised Register of the Cochrane Gynaecology and Fertility Group, the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, PsycINFO, the Cumulative Index to Nursing and Allied Health Literature (CINAHL) and clinical trials registries from database inception to May 2016. We handsearched the reference lists of relevant articles and contacted experts in the field to request additional data. SELECTION CRITERIA: Included studies were randomised controlled trials (RCTs) of premenopausal women with fibroids who were treated for at least three months with a SPRM. DATA COLLECTION AND ANALYSIS: Two review authors independently reviewed all eligible studies identified by the search. We extracted data and assessed risk of bias independently using standard forms. We analysed data using mean differences (MDs) or standardised mean differences (SMDs) for continuous data and odds ratios (ORs) for dichotomous data. We performed meta-analyses using the random-effects model. Our primary outcome was change in fibroid-related symptoms. MAIN RESULTS: We included in the review 14 RCTs with a total of 1215 study participants. We could not extract complete data from three studies. We included in the meta-analysis 11 studies involving 1021 study participants: 685 received SPRMs and 336 were given a control intervention (placebo or leuprolide). Investigators evaluated three SPRMs: mifepristone (five studies), ulipristal acetate (four studies) and asoprisnil (two studies). The primary outcome was change in fibroid-related symptoms (symptom severity, health-related quality of life, abnormal uterine bleeding, pelvic pain). Adverse event reporting in the included studies was limited to SPRM-associated endometrial changes. More than half (8/14) of these studies were at low risk of bias in all domains. The most common limitation of the other studies was poor reporting of methods. The main limitation for the overall quality of evidence was potential publication bias. SPRM versus placebo SPRM treatment resulted in improvements in fibroid symptom severity (MD -20.04 points, 95% confidence interval (CI) -26.63 to -13.46; four RCTs, 171 women, I2 = 0%; moderate-quality evidence) and health-related quality of life (MD 22.52 points, 95% CI 12.87 to 32.17; four RCTs, 200 women, I2 = 63%; moderate-quality evidence) on the Uterine Fibroid Symptom Quality of Life Scale (UFS-QoL, scale 0 to 100). Women treated with an SPRM showed reduced menstrual blood loss on patient-reported bleeding scales, although this effect was small (SMD -1.11, 95% CI -1.38 to -0.83; three RCTs, 310 women, I2 = 0%; moderate-quality evidence), along with higher rates of amenorrhoea (29 per 1000 in the placebo group vs 237 to 961 per 1000 in the SPRM group; OR 82.50, 95% CI 37.01 to 183.90; seven RCTs, 590 women, I2 = 0%; moderate-quality evidence), compared with those given placebo. We could draw no conclusions regarding changes in pelvic pain owing to variability in the estimates. With respect to adverse effects, SPRM-associated endometrial changes were more common after SPRM therapy than after placebo (OR 15.12, 95% CI 6.45 to 35.47; five RCTs, 405 women, I2 = 0%; low-quality evidence). SPRM versus leuprolide acetate In comparing SPRM versus other treatments, two RCTs evaluated SPRM versus leuprolide acetate. One RCT reported primary outcomes. No evidence suggested a difference between SPRM and leuprolide groups for improvement in quality of life, as measured by UFS-QoL fibroid symptom severity scores (MD -3.70 points, 95% CI -9.85 to 2.45; one RCT, 281 women; moderate-quality evidence) and health-related quality of life scores (MD 1.06 points, 95% CI -5.73 to 7.85; one RCT, 281 women; moderate-quality evidence). It was unclear whether results showed a difference between SPRM and leuprolide groups for reduction in menstrual blood loss based on the pictorial blood loss assessment chart (PBAC), as confidence intervals were wide (MD 6 points, 95% CI -40.95 to 50.95; one RCT, 281 women; low-quality evidence), or for rates of amenorrhoea (804 per 1000 in the placebo group vs 732 to 933 per 1000 in the SPRM group; OR 1.14, 95% CI 0.60 to 2.16; one RCT, 280 women; moderate-quality evidence). No evidence revealed differences between groups in pelvic pain scores based on the McGill Pain Questionnaire (scale 0 to 45) (MD -0.01 points, 95% CI -2.14 to 2.12; 281 women; moderate-quality evidence). With respect to adverse effects, SPRM-associated endometrial changes were more common after SPRM therapy than after leuprolide treatment (OR 10.45, 95% CI 5.38 to 20.33; 301 women; moderate-quality evidence). AUTHORS' CONCLUSIONS: Short-term use of SPRMs resulted in improved quality of life, reduced menstrual bleeding and higher rates of amenorrhoea than were seen with placebo. Thus, SPRMs may provide effective treatment for women with symptomatic fibroids. Evidence derived from one RCT showed no difference between leuprolide acetate and SPRM with respect to improved quality of life and bleeding symptoms. Evidence was insufficient to show whether effectiveness was different between SPRMs and leuprolide. Investigators more frequently observed SPRM-associated endometrial changes in women treated with SPRMs than in those treated with placebo or leuprolide acetate. As noted above, SPRM-associated endometrial changes are benign, are not related to cancer and are not precancerous. Reporting bias may impact the conclusion of this meta-analysis. Well-designed RCTs comparing SPRMs versus other treatments are needed.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Estrenes/therapeutic use , Leiomyoma/drug therapy , Mifepristone/therapeutic use , Norpregnadienes/therapeutic use , Oximes/therapeutic use , Receptors, Progesterone/antagonists & inhibitors , Uterine Neoplasms/drug therapy , Amenorrhea/drug therapy , Female , Humans , Leuprolide/therapeutic use , Menstruation/drug effects , Pelvic Pain/drug therapy , Quality of Life , Randomized Controlled Trials as Topic
9.
Cochrane Database Syst Rev ; 7: CD009881, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28742263

ABSTRACT

BACKGROUND: Endometriosis is defined as the presence of endometrial tissue (glands and stroma) outside the uterine cavity. This condition is oestrogen-dependent and thus is seen primarily during the reproductive years. Owing to their antiproliferative effects in the endometrium, progesterone receptor modulators (PRMs) have been advocated for treatment of endometriosis. OBJECTIVES: To assess the effectiveness and safety of PRMs primarily in terms of pain relief as compared with other treatments or placebo or no treatment in women of reproductive age with endometriosis. SEARCH METHODS: We searched the following electronic databases, trial registers, and websites: the Cochrane Gynaecology and Fertility Group (CGFG) Specialised Register of Controlled Trials, the Central Register of Studies Online (CRSO), MEDLINE, Embase, PsycINFO, clinicaltrials.gov, and the World Health Organization (WHO) platform, from inception to 28 November 2016. We handsearched reference lists of articles retrieved by the search. SELECTION CRITERIA: We included randomised controlled trials (RCTs) published in all languages that examined effects of PRMs for treatment of symptomatic endometriosis. DATA COLLECTION AND ANALYSIS: We used standard methodological procedures as expected by the Cochrane Collaboration. Primary outcomes included measures of pain and side effects. MAIN RESULTS: We included 10 randomised controlled trials (RCTs) with 960 women. Two RCTs compared mifepristone versus placebo or versus a different dose of mifepristone, one RCT compared asoprisnil versus placebo, one compared ulipristal versus leuprolide acetate, and four compared gestrinone versus danazol, gonadotropin-releasing hormone (GnRH) analogues, or a different dose of gestrinone. The quality of evidence ranged from high to very low. The main limitations were serious risk of bias (associated with poor reporting of methods and high or unclear rates of attrition in most studies), very serious imprecision (associated with low event rates and wide confidence intervals), and indirectness (outcome assessed in a select subgroup of participants). Mifepristone versus placebo One study made this comparison and reported rates of painful symptoms among women who reported symptoms at baseline.At three months, the mifepristone group had lower rates of dysmenorrhoea (odds ratio (OR) 0.08, 95% confidence interval (CI) 0.04 to 0.17; one RCT, n =352; moderate-quality evidence), suggesting that if 40% of women taking placebo experience dysmenorrhoea, then between 3% and 10% of women taking mifepristone will do so. The mifepristone group also had lower rates of dyspareunia (OR 0.23, 95% CI 0.11 to 0.51; one RCT, n = 223; low-quality evidence). However, the mifepristone group had higher rates of side effects: Nearly 90% had amenorrhoea and 24% had hot flushes, although the placebo group reported only one event of each (1%) (high-quality evidence). Evidence was insufficient to show differences in rates of nausea, vomiting, or fatigue, if present. Mifepristone dose comparisons Two studies compared doses of mifepristone and found insufficient evidence to show differences between different doses in terms of effectiveness or safety, if present. However, subgroup analysis of comparisons between mifepristone and placebo suggest that the 2.5 mg dose may be less effective than 5 mg or 10 mg for treating dysmenorrhoea or dyspareunia. Gestrinone comparisons Ons study compared gestrinone with danazol, and another study compared gestrinone with leuprolin.Evidence was insufficient to show differences, if present, between gestrinone and danazol in rate of pain relief (those reporting no or mild pelvic pain) (OR 0.71, 95% CI 0.33 to 1.56; two RCTs, n = 230; very low-quality evidence), dysmenorrhoea (OR 0.72, 95% CI 0.39 to 1.33; two RCTs, n = 214; very low-quality evidence), or dyspareunia (OR 0.83, 95% CI 0.37 to 1.86; two RCTs, n = 222; very low-quality evidence). The gestrinone group had a higher rate of hirsutism (OR 2.63, 95% CI 1.60 to 4.32; two RCTs, n = 302; very low-quality evidence) and a lower rate of decreased breast size (OR 0.62, 95% CI 0.38 to 0.98; two RCTs, n = 302; low-quality evidence). Evidence was insufficient to show differences between groups, if present, in rate of hot flushes (OR 0.79, 95% CI 0.50 to 1.26; two RCTs, n = 302; very low-quality evidence) or acne (OR 1.45, 95% CI 0.90 to 2.33; two RCTs, n = 302; low-quality evidence).When researchers compared gestrinone versus leuprolin through measurements on the 1 to 3 verbal rating scale (lower score denotes benefit), the mean dysmenorrhoea score was higher in the gestrinone group (MD 0.35 points, 95% CI 0.12 to 0.58; one RCT, n = 55; low-quality evidence), but the mean dyspareunia score was lower in this group (MD 0.33 points, 95% CI 0.62 to 0.04; low-quality evidence). The gestrinone group had lower rates of amenorrhoea (OR 0.04, 95% CI 0.01 to 0.38; one RCT, n = 49; low-quality evidence) and hot flushes (OR 0.20, 95% CI 0.06 to 0.63; one study, n = 55; low quality evidence) but higher rates of spotting or bleeding (OR 22.92, 95% CI 2.64 to 198.66; one RCT, n = 49; low-quality evidence).Evidence was insufficient to show differences in effectiveness or safety between different doses of gestrinone, if present. Asoprisnil versus placebo One study (n = 130) made this comparison but did not report data suitable for analysis. Ulipristal versus leuprolide acetate One study (n = 38) made this comparison but did not report data suitable for analysis. AUTHORS' CONCLUSIONS: Among women with endometriosis, moderate-quality evidence shows that mifepristone relieves dysmenorrhoea, and low-quality evidence suggests that this agent relieves dyspareunia, although amenorrhoea and hot flushes are common side effects. Data on dosage were inconclusive, although they suggest that the 2.5 mg dose of mifepristone may be less effective than higher doses. We found insufficient evidence to permit firm conclusions about the safety and effectiveness of other progesterone receptor modulators.


Subject(s)
Endometriosis/drug therapy , Hormone Antagonists/therapeutic use , Mifepristone/therapeutic use , Receptors, Progesterone/antagonists & inhibitors , Danazol/therapeutic use , Dysmenorrhea/drug therapy , Dysmenorrhea/epidemiology , Dyspareunia/drug therapy , Dyspareunia/epidemiology , Estrenes/therapeutic use , Female , Gestrinone/adverse effects , Gestrinone/therapeutic use , Gonadotropin-Releasing Hormone/analogs & derivatives , Hormone Antagonists/administration & dosage , Hormone Antagonists/adverse effects , Humans , Leuprolide/adverse effects , Leuprolide/therapeutic use , Mifepristone/administration & dosage , Mifepristone/adverse effects , Norpregnadienes/therapeutic use , Oximes/therapeutic use , Prevalence , Randomized Controlled Trials as Topic
10.
Clin Obstet Gynecol ; 59(1): 30-52, 2016 03.
Article in English | MEDLINE | ID: mdl-26756261

ABSTRACT

Fibroids are the most common tumor of the female reproductive tract, but approved medical treatments are limited. Patients demand uterine-sparing treatments which preserve fertility and avoid surgery. We systematically reviewed PubMed and Cochrane databases from January 1985 to November 2015 for evidence-based medical therapies for fibroids in the context of disease prevention, treatment of early disease, treatment of symptomatic disease, and preoperative management. We identified 2182 studies, of which 52 studies met inclusion and exclusion criteria. Published data affirm the efficacy of multiple agents, which are promising avenues for the development of medical alternatives to surgery.


Subject(s)
Androgens/therapeutic use , Aromatase Inhibitors/therapeutic use , Contraceptive Agents, Female/therapeutic use , Gonadotropin-Releasing Hormone/agonists , Hormone Antagonists/therapeutic use , Leiomyoma/drug therapy , Plant Extracts/therapeutic use , Uterine Neoplasms/drug therapy , Contraceptives, Oral, Combined/therapeutic use , Curcumin , Delayed-Action Preparations , Drugs, Chinese Herbal/therapeutic use , Estradiol/analogs & derivatives , Estradiol/therapeutic use , Estrenes/therapeutic use , Estrogen Receptor Antagonists/therapeutic use , Evidence-Based Medicine , Female , Fulvestrant , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Humans , Intrauterine Devices, Medicated , Leiomyoma/prevention & control , Levonorgestrel/therapeutic use , Medroxyprogesterone Acetate/therapeutic use , Mifepristone/therapeutic use , Neoadjuvant Therapy , Norpregnadienes/therapeutic use , Oximes/therapeutic use , Receptors, Progesterone/antagonists & inhibitors , Selective Estrogen Receptor Modulators/therapeutic use , Tea , Uterine Myomectomy , Uterine Neoplasms/prevention & control , Vitamin D/therapeutic use , Vitamins/therapeutic use
11.
J Immunol ; 191(5): 2226-35, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23913972

ABSTRACT

Uterine NK cells (uNK) play a role in the regulation of placentation, but their functions in nonpregnant endometrium are not understood. We have previously reported suppression of endometrial bleeding and alteration of spiral artery morphology in women exposed to asoprisnil, a progesterone receptor modulator. We now compare global endometrial gene expression in asoprisnil-treated versus control women, and we demonstrate a statistically significant reduction of genes in the IL-15 pathway, known to play a key role in uNK development and function. Suppression of IL-15 by asoprisnil was also observed at mRNA level (p < 0.05), and immunostaining for NK cell marker CD56 revealed a striking reduction of uNK in asoprisnil-treated endometrium (p < 0.001). IL-15 levels in normal endometrium are progesterone-responsive. Progesterone receptor (PR) positive stromal cells transcribe both IL-15 and IL-15RA. Thus, the response of stromal cells to progesterone will be to increase IL-15 trans-presentation to uNK, supporting their expansion and differentiation. In asoprisnil-treated endometrium, there is a marked downregulation of stromal PR expression and virtual absence of uNK. These novel findings indicate that the IL-15 pathway provides a missing link in the complex interplay among endometrial stromal cells, uNK, and spiral arteries affecting physiologic and pathologic endometrial bleeding.


Subject(s)
Estrenes/therapeutic use , Killer Cells, Natural/metabolism , Leiomyoma/drug therapy , Oximes/therapeutic use , Uterine Neoplasms/drug therapy , Double-Blind Method , Endometrium/drug effects , Endometrium/immunology , Endometrium/metabolism , Female , Humans , Immunohistochemistry , Interleukin-15 , Killer Cells, Natural/immunology , Leiomyoma/complications , Leiomyoma/immunology , Lymphocyte Activation/drug effects , Oligonucleotide Array Sequence Analysis , Receptors, Progesterone/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome , Uterine Neoplasms/complications , Uterine Neoplasms/immunology , Uterus
12.
BMC Vet Res ; 10: 296, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25515784

ABSTRACT

BACKGROUND: Progesterone receptors play a key role in the development of canine mammary tumours, and recent research has focussed on their possible value as therapeutic targets using antiprogestins. Cloning and sequencing of the progesterone receptor gene has shown that the receptor has two isoforms, A and B, transcribed from a single gene. Experimental studies in human breast cancer suggest that the differential expression of progesterone receptor isoforms has implications for hormone therapy responsiveness. This study examined the effects of the antiprogestin aglepristone on cell proliferation and mRNA expression of progesterone receptor isoforms A and B in mammary carcinomas in dogs treated with 20 mg/Kg of aglepristone (n = 22) or vehicle (n = 5) twice before surgery. RESULTS: Formalin-fixed, paraffin-embedded tissue samples taken before and after treatment were used to analyse total progesterone receptor and both isoforms by RT-qPCR and Ki67 antigen labelling. Both total progesterone receptor and isoform A mRNA expression levels decreased after treatment with aglepristone. Furthermore, a significant decrease in the proliferation index (percentage of Ki67-labelled cells) was observed in progesterone-receptor positive and isoform-A positive tumours in aglepristone-treated dogs. CONCLUSIONS: These findings suggest that the antiproliferative effects of aglepristone in canine mammary carcinomas are mediated by progesterone receptor isoform A.


Subject(s)
Dog Diseases/drug therapy , Estrenes/therapeutic use , Mammary Neoplasms, Animal/drug therapy , Neoadjuvant Therapy/veterinary , Receptors, Progesterone/physiology , Animals , Cell Proliferation/drug effects , Dog Diseases/physiopathology , Dogs , Female , Mammary Neoplasms, Animal/physiopathology , Neoadjuvant Therapy/methods , Polymerase Chain Reaction/veterinary , Receptors, Progesterone/metabolism
13.
Reprod Domest Anim ; 49 Suppl 2: 28-32, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24947858

ABSTRACT

Pyometra is a reproductive disorder very common in bitches over 8 years of age in which physiological effects of progesterone on the uterus play a major role. The traditional therapy for pyometra is ovariohysterectomy. The main advantage of ovariohysterectomy over medical management is that it is both curative and preventive for recurrence of pyometra. However, surgery is associated with the risk of anaesthesia and renders the bitch sterile. During the last 10 years, numerous medical treatments have been proposed to treat both open and closed cervix pyometra. The most effective medical treatment with minor side effects seems to be the repeated administration of aglepristone with or without the additional treatment with low doses of prostaglandins.


Subject(s)
Dog Diseases/drug therapy , Estrenes/therapeutic use , Pyometra/veterinary , Receptors, Progesterone/antagonists & inhibitors , Animals , Dogs , Estrenes/administration & dosage , Female , Prostaglandins/administration & dosage , Prostaglandins/therapeutic use , Pyometra/drug therapy
14.
Vet Med Sci ; 10(6): e70060, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39365140

ABSTRACT

Feline mammary fibroadenomatous hyperplasia (FMFH) is an extensive proliferation of feline mammary tissues in response to endogenous or exogenous progestogens. The treatment of choice for FMFH is anti-progestins or ovariohysterectomy (OVH). OVH if there is no intention of breeding the queen. This report describes a case of recurrent FMFH in an 11-month-old female Maine Coon cat. The condition was successfully treated with aglepristone during its first occurrence but recurred during the cat's subsequent gestation. OVH was performed along with aglepristone administration, resulting in complete regression of FMFH 21 days after aglepristone injection. The recurrence of FMFH successful treatment is well described; however, this case illustrates the necessity for further discussion regarding the utilisation of cats previously diagnosed with FMFH for breeding purposes.


Subject(s)
Cat Diseases , Hyperplasia , Animals , Cats , Female , Cat Diseases/pathology , Cat Diseases/drug therapy , Cat Diseases/surgery , Hyperplasia/veterinary , Pregnancy , Estrenes/therapeutic use , Mammary Neoplasms, Animal/pathology , Recurrence , Hysterectomy/veterinary
15.
Ann Oncol ; 24(10): 2543-2548, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23788750

ABSTRACT

BACKGROUND: The progesterone-receptor (PR) antagonists onapristone (type I) and mifepristone (type II) showed modest activity in hormone-receptor-positive breast cancer; however, onapristone in particular was associated with hepatotoxicity. Lonaprisan is a novel, type III PR antagonist that was well tolerated in phase I studies. PATIENTS AND METHODS: This randomized, open-label, phase II study evaluated the efficacy and tolerability of lonaprisan as second-line endocrine therapy in postmenopausal women with stage IV, PR-positive, HER2-negative, metastatic breast cancer. RESULTS: Patients received once-daily lonaprisan 25 mg (n = 34) or 100 mg (n = 34). The primary objective was not met (≥ 35% clinical benefit rate: complete/partial responses at any time until month 6 or stable disease [SD] for ≥ 6 months from start of treatment). There were no complete/partial responses. In the 25 mg and 100 mg groups, 6 of 29 patients (21%) and 2 of 29 patients (7%), respectively, had SD ≥ 6 months. Overall, 61 of 68 patients (90%) had ≥ 1 adverse event (AE), the most frequent (≥ 10% overall) being fatigue, hot flush, dyspnoea, nausea, asthenia, headache, constipation, vomiting, and decreased appetite; 33 patients had serious AEs. CONCLUSION: Lonaprisan showed limited efficacy as second-line endocrine therapy in postmenopausal women with PR-positive metastatic breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Estrenes/therapeutic use , Receptors, Progesterone/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Hormonal/adverse effects , Estrenes/adverse effects , Female , Humans , Middle Aged , Receptors, Progesterone/metabolism , Treatment Outcome
16.
Vet Med Sci ; 9(3): 1297-1303, 2023 05.
Article in English | MEDLINE | ID: mdl-36634253

ABSTRACT

BACKGROUND: Induction of parturition in guinea pigs appears to be essential because these animals have a higher rate of reproductive problems than rabbits and small rodents. OBJECTIVES: Since aglepristone (AGL) is a competitive progesterone antagonist acting through binding to progesterone receptors while oxytocin (OT) is a powerful constituent of uterine smooth muscle, the aim of this study was to evaluate the clinical and ultrasonographic impacts of AGL and OT on guinea pig parturition induction. METHODS: In this study, guinea pigs were allocated into five groups; each included five animals on the 61st day of pregnancy. In the aglepristone group (Agle), AGL was administrated subcutaneously (SC) once daily on 2 consecutive days (Days 61 and 62 post mating). Oxytocin (OT) was administered subcutaneously once and twice at 4-h intervals on Day 62 post mating in oxytocin 1 (Oxy1) and oxytocin 2 (Oxy2) groups, respectively. The animals in the aglepristone-oxytocin group (Agle-Oxy) received AGL subcutaneously once daily on 2 consecutive days (Days 61 and 62 post mating) and OT on Day 62 post mating. The remaining sows received saline solution (0.9% NaCl) in the control group. RESULTS: According to the results, fetal heart rate, temperature, neonatal and maternal survival rates were not significantly different between the treatment and control groups (p > 0.05). Biparietal diameter of head and body weight of neonates in the Agle, Oxy2 and Agle-Oxy groups showed a significant decline, compared to the control group (p < 0.05). The time interval between injection and delivery and the duration of pregnancy was significantly reduced in Agle, Oxy2, Agl-Oxy groups, compared to the control and Oxy1 groups. CONCLUSIONS: In conclusion, it seems that treatment Oxy2 can induce parturition in guinea pigs without side effects and lower pain during induction of parturition.


Subject(s)
Oxytocin , Parturition , Pregnancy , Animals , Guinea Pigs , Female , Swine , Rabbits , Oxytocin/pharmacology , Estrenes/pharmacology , Estrenes/therapeutic use , Uterus
17.
Invest New Drugs ; 29(2): 340-6, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20084425

ABSTRACT

BACKGROUND: 2-methoxyestradiol (2ME2) is an estradiol-17ß metabolite with antiproliferative and antiangiogenic activities. ENMD-1198 is an analog of 2ME2 which was developed to decrease the metabolism and increase both the bioavailability and antitumor activities of the parent molecule. This first-in-human phase I study evaluated the tolerability, pharmacokinetics and preliminary evidence of activity of ENMD-1198 in advanced cancer patients. METHODS: Eligible patients received ENMD-1198 orally once daily in Part A (standard 3 + 3 dose escalation design), or in Part B (accelerated dose escalation design). Cycle 1 consisted of 28 days daily dosing followed by a 14-(Part A) or 7-(Part B) day observation period, then continuously in 28 day cycles thereafter. RESULTS: A total of 29 patients were enrolled in 12 dose cohorts (5 to 550 mg/m²)/d). The most common drug-related toxicities were Grade 1/2 fatigue (55%), nausea and vomiting (37%), and constipation (34%). Two DLTs (Grade 4 neutropenia) occurred at 550 mg/m²/day, and 425 mg/m²/d was declared the maximum tolerated dose. ENMD-1198 was absorbed rapidly with a T(max) of 1-2 h. Exposure to ENMD-1198 (C(max) and AUC0₋24 hr increased linearly with dose. The mean terminal half-life was 15 h. A 3-fold accumulation was found after multiple doses. Five patients achieved stabilization of disease for at least 2 cycles, three of whom (with neuroendocrine carcinoma of pancreas, prostate cancer and ovarian cancer) demonstrated prolonged stabilization ranging from 8-24.5 cycles. CONCLUSION: ENMD-1198 is well-tolerated with a pharmacokinetic exposure profile compatible with once daily dosing. The recommended phase II dose of ENMD-1198 is 425 mg/m²/d. Early evidence of prolonged disease stabilization in pre-treated patients suggests ENMD-1198 is worthy of additional investigation.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Estradiol/analogs & derivatives , Estrenes/pharmacokinetics , Estrenes/therapeutic use , Neoplasms/drug therapy , 2-Methoxyestradiol , Administration, Oral , Adult , Aged , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Demography , Dogs , Dose-Response Relationship, Drug , Estradiol/administration & dosage , Estradiol/adverse effects , Estradiol/pharmacokinetics , Estradiol/therapeutic use , Estrenes/administration & dosage , Estrenes/adverse effects , Female , Humans , Male , Middle Aged , Neoplasms/blood , Rats , Species Specificity
18.
Proc Natl Acad Sci U S A ; 105(50): 20004-8, 2008 Dec 16.
Article in English | MEDLINE | ID: mdl-19066214

ABSTRACT

Treatment of neuropathic pain is a major clinical problem. This study shows expression of phospholipase ss3 (PLCss3) in mouse and human DRG neurons, mainly in small ones and mostly with a nonpeptidergic phenotype. After spared nerve injury, the pain threshold was strongly reduced, and systemic treatment of such animals with the unselective PLC inhibitor U73122 caused a rapid and long-lasting (48-h) increase in pain threshold. Thus, inhibition of PLC may provide a way to treat neuropathic pain.


Subject(s)
Estrenes/therapeutic use , Hyperalgesia/drug therapy , Pain/drug therapy , Phosphodiesterase Inhibitors/therapeutic use , Phospholipase C beta/antagonists & inhibitors , Pyrrolidinones/therapeutic use , Spinal Cord Injuries/enzymology , Animals , Disease Models, Animal , Ganglia, Spinal/drug effects , Ganglia, Spinal/enzymology , Ganglia, Spinal/injuries , Humans , Hyperalgesia/enzymology , Hyperalgesia/etiology , Mice , Mice, Mutant Strains , Pain/enzymology , Pain/etiology , Pain Threshold/drug effects , Phospholipase C beta/genetics , Spinal Cord/drug effects , Spinal Cord/enzymology , Spinal Cord Injuries/complications
19.
FEBS Open Bio ; 11(2): 435-445, 2021 02.
Article in English | MEDLINE | ID: mdl-33326693

ABSTRACT

Osteoarthritis (OA) is an age-related, chronic degenerative disease. With the increasing median age of the population, this disease has become an important public health problem. New, disease-modifying therapies are needed. A potential novel molecular target is phospholipase Cγ1 (PLCγ1), a critical enzyme with important functions including calcium signaling regulation and cell proliferation. In rat chondrocytes treated with IL-1ß (20 ng·mL-1 for 36 h), inhibition of PLCγ1 with U73122 (2 µm for 12 h) increased levels and expression of the cartilage matrix components Collagen2 and Aggrecan. This beneficial effect of PLCγ1 inhibition was counteracted by increased chondrocyte apoptosis and necroptosis, increased cell death, and increase levels of ROS, all potentially negative for OA. Combined treatment of IL-1ß + U73122-treated chondrocytes with inhibitors of apoptosis (Z-VAD, 10 µm) and necroptosis (Nec-1, 30 µm) enhanced the increases in levels and expression of Collagen2 and Aggrecan, and prevented the increases in cell death and ROS levels. These results suggest that PLCγ1 inhibition may be a viable approach for an OA therapy, if combined with targeted inhibition of chondrocyte apoptosis and necroptosis.


Subject(s)
Interleukin-1beta/immunology , Osteoarthritis/drug therapy , Phospholipase C gamma/antagonists & inhibitors , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/immunology , Cartilage, Articular/cytology , Cartilage, Articular/drug effects , Cartilage, Articular/immunology , Cartilage, Articular/pathology , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/immunology , Chondrocytes/pathology , Disease Models, Animal , Drug Therapy, Combination/methods , Estrenes/pharmacology , Estrenes/therapeutic use , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Interleukin-1beta/metabolism , Necroptosis/drug effects , Necroptosis/immunology , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Osteoarthritis/immunology , Osteoarthritis/pathology , Phospholipase C gamma/metabolism , Primary Cell Culture , Pyrrolidinones/pharmacology , Pyrrolidinones/therapeutic use , Rats , Recombinant Proteins/immunology , Recombinant Proteins/metabolism
20.
Cell Biochem Funct ; 28(5): 412-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20589734

ABSTRACT

A priority in recent anti-cancer drug development has been attaining better side-effect profiles for potential compounds. To produce highly specific cancer therapies it is necessary to understand both the effects of the proposed compound on cancer and on normal cells comprising the rest of the human body. Thus in vitro evaluation of these compounds against non-carcinogenic cell lines is of critical importance. One of the most recent developments in experimental anti-cancer agents is 2-methoxyestradiol-bis-sulphamate (2ME-BM), a sulphamoylated derivative of 2-methoxyestradiol. The aim of this study was to evaluate the in vitro effects of 2ME-BM on cell proliferation, morphology and mechanisms of cell death in the non-carcinogenic MCF-12A breast epithelial cell line. The study revealed changes in proliferative capacity, morphology and cell death induction in response to 2ME-BM exposure (24 h at 0.4 microM). Microscopy showed decreased cell density and cell death-associated morphology (increased apoptotic characteristics), a slight increase in acidic intracellular vesicles and insignificant ultra-structural aberrations. Mitotic indices revealed a G(2)M-phase cell cycle block. This was confirmed by flow cytometry, where an increased fraction of abnormal cells and a decrease in cyclin B1 levels were observed. These results evidently demonstrate that the non-carcinogenic MCF-12A cell line is less susceptible when compared to 2ME-BM-exposed cancer cell lines previously tested. Further in vitro research into the mechanism of this potentially useful compound is warranted.


Subject(s)
Antineoplastic Agents/therapeutic use , Estrenes/therapeutic use , Antineoplastic Agents/chemistry , Apoptosis , Breast Neoplasms/drug therapy , Breast Neoplasms/ultrastructure , Cell Division , Cell Line, Tumor , Cyclin B1/metabolism , Estrenes/chemistry , Female , Flow Cytometry , G2 Phase , Humans , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL