Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 615
Filter
1.
Post Reprod Health ; 29(4): 222-231, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37840298

ABSTRACT

Therapies utilised in breast cancer management have been found to induce or worsen the genitourinary symptoms of menopause (GSM), a group of physical symptoms associated with the systemic loss of estrogen. These symptoms are often undertreated due to concerns surrounding cancer recurrence, especially when considering treatments with possible pro-estrogenic effects. As breast cancer prognosis continues to improve, clinicians are increasingly focussing on managing these symptoms amongst survivors. This systematic review primarily aimed to determine the risk of breast cancer recurrence amongst survivors using vaginal hormones and selective estrogen receptor modulator therapies recommended for use in GSM in the United Kingdom amongst currently published randomised clinical trials (RCTs). The secondary aim was to determine whether these RCTs demonstrated a significant rise in serum estrogen levels following the use of these therapies. A literature search revealed three RCTs suitable for assessment, two evaluating vaginal estrogen and one evaluating vaginal DHEA treatment. Our review determined that amongst published RCTs, no studies have aimed to assess for breast cancer recurrence; however among the studies observing for serious adverse effects of vaginal estrogen preparations, none have reported an increased incidence. Furthermore, these studies did not report a persistent or significant increase in serum estrogen levels following the use of vaginal estrogen products and low concentration (3.25 mg/day) DHEA gel. Larger RCTs studying commonly used vaginal preparations and selective estrogen receptor modulator treatments for GSM over longer follow-up periods will be vital to better assess the risk of breast cancer recurrence in survivors receiving these treatments.


Subject(s)
Breast Neoplasms , Cancer Survivors , Female , Humans , Selective Estrogen Receptor Modulators/adverse effects , Neoplasm Recurrence, Local/complications , Neoplasm Recurrence, Local/drug therapy , Breast Neoplasms/complications , Estrogens/adverse effects , Menopause , Survivors , Dehydroepiandrosterone/therapeutic use
2.
Menopause ; 30(8): 824-830, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37449720

ABSTRACT

OBJECTIVE: To assess the risk of select safety outcomes including endometrial cancer, endometrial hyperplasia, and breast cancer among women using conjugated estrogens/bazedoxifene (CE/BZA) as compared with estrogen/progestin combination hormone therapy (EP). METHODS: We conducted a new-user cohort study in five US healthcare claims databases representing more than 92 million women. We included CE/BZA or EP new users from May 1, 2014, to August 30, 2019. EP users were propensity score (PS) matched to users of CE/BZA. Incidence of endometrial cancer, endometrial hyperplasia, breast cancer, and eight additional cancer and cardiovascular outcomes were ascertained using claims-based algorithms. Rate ratios (RR) and differences pooled across databases were estimated using random-effects models. RESULTS: The study population included 10,596 CE/BZA and 33,818 PS-matched EP new users. Rates of endometrial cancer and endometrial hyperplasia were slightly higher among CE/BZA users (1.6 and 0.4 additional cases per 10,000 person-years), although precision was limited because of small numbers of cases (endometrial cancer: RR, 1.50 [95% confidence interval {CI}, 0.79-2.88]; endometrial hyperplasia: RR, 1.69 [95% CI, 0.51-5.61]). Breast cancer incidence was lower in CE/BZA users (9.1 fewer cases per 10,000 person-years; RR, 0.79; 95% CI, 0.58-1.05). Rates of other outcomes were slightly higher among CE/BZA users, but with confidence intervals compatible with a wider range of possible associations. CONCLUSIONS: CE/BZA users might experience slightly higher rates of endometrial cancer and endometrial hyperplasia, and a lower rate of breast cancer, than EP users in the first years of use.


Subject(s)
Breast Neoplasms , Endometrial Neoplasms , Estrogen Replacement Therapy , Estrogens , Selective Estrogen Receptor Modulators , Estrogens/adverse effects , Estrogens/therapeutic use , Selective Estrogen Receptor Modulators/adverse effects , Selective Estrogen Receptor Modulators/therapeutic use , Estrogen Replacement Therapy/adverse effects , Humans , Female , Breast Neoplasms/chemically induced , Breast Neoplasms/epidemiology , Endometrial Neoplasms/chemically induced , Endometrial Neoplasms/epidemiology , Endometrial Hyperplasia/chemically induced , Endometrial Hyperplasia/epidemiology , Incidence , United States/epidemiology
3.
Biochem Pharmacol ; 214: 115677, 2023 08.
Article in English | MEDLINE | ID: mdl-37419371

ABSTRACT

Breast cancer is the most common cancer in women. Over the past few decades, advances in cancer detection and treatment have significantly improved survival rate of breast cancer patients. However, due to the cardiovascular toxicity of cancer treatments (chemotherapy, anti-HER2 antibodies and radiotherapy), cardiovascular diseases (CVD) have become an increasingly important cause of long-term morbidity and mortality in breast cancer survivors. Endocrine therapies are prescribed to reduce the risk of recurrence and specific death in estrogen receptor-positive (ER +) early breast cancer patients, but their impact on CVD is a matter of debate. Whereas aromatase inhibitors and luteinizing hormone-releasing hormone (LHRH) analogs inhibit estrogen synthesis, tamoxifen acts as a selective estrogen receptor modulator (SERM), opposing estrogen action in the breast but mimicking their actions in other tissues, including arteries. This review aims to summarize the main clinical and experimental studies reporting the effects of tamoxifen on CVD. In addition, we will discuss how recent findings on the mechanisms of action of these therapies may contribute to a better understanding and anticipation of CVD risk in breast cancer patients.


Subject(s)
Breast Neoplasms , Cardiovascular Diseases , Female , Humans , Tamoxifen/adverse effects , Antineoplastic Agents, Hormonal/adverse effects , Chemotherapy, Adjuvant , Estrogens , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/prevention & control , Cardiovascular Diseases/drug therapy , Arteries , Selective Estrogen Receptor Modulators/adverse effects
4.
Menopause ; 30(8): 855-866, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37369079

ABSTRACT

IMPORTANCE: Ospemifene is a novel selective estrogen receptor modulator developed for the treatment of moderate to severe postmenopausal vulvovaginal atrophy (VVA). OBJECTIVE: The aim of the study is to perform a systematic literature review (SLR) and network meta-analysis (NMA) to assess the efficacy and safety of ospemifene compared with other therapies used in the treatment of VVA in North America and Europe. EVIDENCE REVIEW: Electronic database searches were conducted in November 2021 in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Randomized or nonrandomized controlled trials targeting postmenopausal women with moderate to severe dyspareunia and/or vaginal dryness and involving ospemifene or at least one VVA local treatment were considered. Efficacy data included changes from baseline in superficial and parabasal cells, vaginal pH, and the most bothersome symptom of vaginal dryness or dyspareunia, as required for regulatory approval. Endometrial outcomes were endometrial thickness and histologic classifications, including endometrial polyp, hyperplasia, and cancer. For efficacy and safety outcomes, a Bayesian NMA was performed. Endometrial outcomes were compared in descriptive analyses. FINDINGS: A total of 44 controlled trials met the eligibility criteria ( N = 12,637 participants). Network meta-analysis results showed that ospemifene was not statistically different from other active therapies in most efficacy and safety results. For all treatments, including ospemifene, the posttreatment endometrial thickness values (up to 52 wk of treatment) were under the recognized clinical threshold value of 4 mm for significant risk of endometrial pathology. Specifically, for women treated with ospemifene, endometrial thickness ranged between 2.1 and 2.3 mm at baseline and 2.5 and 3.2 mm after treatment. No cases of endometrial carcinoma or hyperplasia were observed in ospemifene trials, nor polyps with atypical hyperplasia or cancer after up to 52 weeks of treatment. CONCLUSIONS AND RELEVANCE: Ospemifene is an efficacious, well-tolerated, and safe therapeutic option for postmenopausal women with moderate to severe symptoms of VVA. Efficacy and safety outcomes with ospemifene are similar to other VVA therapies in North America and Europe.


Subject(s)
Dyspareunia , Endometrial Neoplasms , Vaginal Diseases , Female , Humans , Dyspareunia/drug therapy , Dyspareunia/pathology , Vagina/pathology , Hyperplasia/drug therapy , Hyperplasia/pathology , Bayes Theorem , Network Meta-Analysis , Vulva/pathology , Atrophy/drug therapy , Atrophy/pathology , Tamoxifen/adverse effects , Selective Estrogen Receptor Modulators/adverse effects , Vaginal Diseases/drug therapy , Vaginal Diseases/pathology , Endometrial Neoplasms/pathology
5.
Schizophr Bull ; 49(6): 1579-1590, 2023 11 29.
Article in English | MEDLINE | ID: mdl-37116866

ABSTRACT

BACKGROUND AND HYPOTHESIS: Several studies suggest that raloxifene, a selective estrogen receptor modulator, improves symptoms and cognition in post-menopausal women with Schizophrenia-Spectrum Disorders (SSD). We aimed to assess the effects of adjunctive raloxifene in women and men with SSD. STUDY DESIGN: This parallel, randomized, double-blind, placebo-controlled trial included adult SSD patients across the Netherlands and Belgium. Participants were stratified by age, sex, and global functioning and randomly assigned 1:1 to 12-week add-on raloxifene or placebo. Primary outcomes were symptom severity at 6, 12, and 38 weeks and cognition at 12 and 38 weeks, as measured with the Positive and Negative Syndrome Scale and the Brief Assessment of Cognition in Schizophrenia, respectively. Intention-to-treat analyses were performed using linear mixed-effect models. STUDY RESULTS: We assessed 261 patients for eligibility, of which 102 (28% female) were assigned to raloxifene (n = 52) or placebo (n = 48). Although we found no main effect of raloxifene, secondary sex-specific analysis showed that in women, raloxifene had beneficial effects on negative symptoms at week 6 (LSM -2.92; adjusted P = 0.020) and week 12 (LSM -3.12; adjusted P = 0.030), and on working memory at week 38 (LSM 0.73; adjusted P = 0.040), while having negative effects on working memory at week 38 in men (LSM -0.53; adjusted P = 0.026). The number of adverse events was similar between groups. CONCLUSIONS: Our results do not support the use of raloxifene in patients with SSD in general, but suggest female-specific beneficial effects of raloxifene on negative symptoms and working memory. Our findings encourage further research on sex-specific pharmacotherapeutic treatment.


Subject(s)
Antipsychotic Agents , Schizophrenia , Adult , Male , Female , Humans , Infant, Newborn , Raloxifene Hydrochloride/adverse effects , Schizophrenia/diagnosis , Postmenopause , Selective Estrogen Receptor Modulators/adverse effects , Double-Blind Method , Treatment Outcome
6.
Eur J Pharmacol ; 943: 175558, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36731722

ABSTRACT

PURPOSE: Protective effect of 17ß-estradiol is well-known in pulmonary hypertension. However, estrogen-based therapy may potentially increase the risk of breast cancer, necessitating a search for novel drugs. This study, therefore, investigated the ameliorative effects of a selective estrogen receptor modulator, ormeloxifene, in pulmonary hypertension. METHODS: Cardiomyocytes (H9C2) and human pulmonary arterial smooth muscle cells (HPASMCs) were exposed to hypoxia (1% O2) for 42 and 96 h, respectively, with or without ormeloxifene pre-treatment (1 µM). Also, female (ovary-intact or ovariectomized) and male Sprague-Dawley rats received monocrotaline (60 mg/kg, once, subcutaneously), with or without ormeloxifene treatment (2.5 mg/kg, orally) for four weeks. RESULTS: Hypoxia dysregulated 17ß-hydroxysteroid dehydrogenase (17ßHSD) 1 & 2 expressions, reducing 17ß-estradiol production and estrogen receptors α and ß in HPASMC but increasing estrone, proliferation, inflammation, oxidative stress, and mitochondrial dysfunction. Similarly, monocrotaline decreased plasma 17ß-estradiol and uterine weight in ovary-intact rats. Further, monocrotaline altered 17ßHSD1 & 2 expressions and reduced estrogen receptors α and ß, increasing right ventricular pressure, proliferation, inflammation, oxidative stress, endothelial dysfunction, mitochondrial dysfunction, and vascular remodeling in female and male rats, with worsened conditions in ovariectomized rats. Ormeloxifene was less uterotrophic; however, it attenuated both hypoxia and monocrotaline effects by improving pulmonary 17ß-estradiol synthesis. Furthermore, ormeloxifene decreased cardiac hypertrophy and right ventricular remodeling induced by hypoxia and monocrotaline. CONCLUSION: This study demonstrates that ormeloxifene promoted pulmonary 17ß-estradiol synthesis, alleviated inflammation, improved the NOX4/HO1/Nrf/PPARγ/PGC-1α axis, and attenuated pulmonary hypertension. It is evidently safe at tested concentrations and may be effectively repurposed for pulmonary hypertension treatment.


Subject(s)
Hypertension, Pulmonary , Selective Estrogen Receptor Modulators , Rats , Male , Female , Humans , Animals , Selective Estrogen Receptor Modulators/adverse effects , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/prevention & control , Hypertension, Pulmonary/chemically induced , Rats, Sprague-Dawley , Estrogen Receptor alpha , Monocrotaline/adverse effects , Estradiol/pharmacology , Estradiol/therapeutic use , Pulmonary Artery , Inflammation , Hypoxia
7.
Arch Osteoporos ; 18(1): 18, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36624318

ABSTRACT

This systematic review (SR) assessed the use of denosumab (Prolia®) to treat osteoporosis in cancer patients receiving endocrine therapy. Denosumab was found to prevent vertebral fractures and improve bone mineral density in cancer patients with osteoporosis. This is the first SR to assess treating osteoporotic cancer patients with denosumab. PURPOSE: This study assessed the effectiveness and safety of denosumab (Prolia®) compared to bisphosphonates (alendronate, ibandronate, risedronate, zoledronate), selective estrogen receptor modulators (SERMs) (bazedoxifene, raloxifene) and placebo for the treatment of osteoporosis in hormone-sensitive cancer patients receiving endocrine therapy (men with prostate cancer [MPC] on hormone ablation therapy [HAT], and women with breast cancer [WBC] on adjuvant aromatase inhibitor therapy [AAIT]). METHODS: Systematic literature searches were conducted in three biomedical databases to identify randomized controlled trials (RCTs). Frequentist network meta-analyses and/or pairwise meta-analyses were performed on predetermined outcomes (i.e., vertebral/nonvertebral fractures, bone mineral density [BMD], mortality, treatment-related adverse events [AEs], serious AEs [SAEs], withdrawal due to treatment-related AEs). RESULTS: A total of 14 RCTs (15 publications) were included. Denosumab was found to prevent vertebral fractures in cancer patients receiving endocrine therapy, relative to placebo. Similarly, denosumab, zoledronate, and alendronate improved BMD at the femoral neck (FN) and lumbar spine (LS) in MPC on HAT, relative to placebo. Denosumab, ibandronate and risedronate improved BMD at the LS and total hip (TH) in WBC on AAIT, relative to placebo. Denosumab and risedronate improved trochanteric (TRO) BMD in WBC on AAIT, relative to placebo. Similarly, denosumab improved FN BMD in WBC on AAIT. CONCLUSION: In MPC on HAT, denosumab (relative to placebo) was effective at preventing vertebral fractures and improving BMD at the FN and LS. Moreover, in WBC on AAIT, denosumab (relative to placebo) improved BMD at the FN, LS, TH, and TRO, as well as prevent vertebral fracture.


Subject(s)
Bone Density Conservation Agents , Denosumab , Neoplasms , Female , Humans , Male , Alendronate/adverse effects , Bone Density/drug effects , Bone Density Conservation Agents/adverse effects , Denosumab/adverse effects , Diphosphonates/adverse effects , Hormones , Ibandronic Acid/adverse effects , Neoplasms/drug therapy , Network Meta-Analysis , Osteoporosis/drug therapy , Risedronic Acid/adverse effects , Selective Estrogen Receptor Modulators/adverse effects , Spinal Fractures/prevention & control , Treatment Outcome , Zoledronic Acid/adverse effects , Randomized Controlled Trials as Topic
8.
Best Pract Res Clin Endocrinol Metab ; 36(5): 101686, 2022 09.
Article in English | MEDLINE | ID: mdl-35981955

ABSTRACT

Male hypogonadism is associated with reduced quality of life and the development of co-morbidities including obesity, diabetes mellitus, and dyslipidaemia. The mainstay of treatment for male hypogonadism is testosterone replacement therapy (TRT). However, TRT has recognised side effects including impaired spermatogenesis and there are concerns regarding its use in men with concurrent cardiovascular disease. Thus, there has been an impetus to develop novel androgen therapies for treating male hypogonadism to mitigate the side effects of TRT. This review will discuss the benefits and adverse effects of TRT, and novel therapies including nasal testosterone, aromatase inhibitors, selective oestrogen receptor modulators, and selective androgen receptor modulators.


Subject(s)
Androgen Receptor Antagonists , Androgens , Aromatase Inhibitors , Hormone Replacement Therapy , Hypogonadism , Selective Estrogen Receptor Modulators , Androgen Receptor Antagonists/adverse effects , Androgen Receptor Antagonists/therapeutic use , Androgens/adverse effects , Androgens/therapeutic use , Aromatase Inhibitors/adverse effects , Aromatase Inhibitors/therapeutic use , Humans , Hypogonadism/drug therapy , Male , Quality of Life , Receptors, Androgen , Selective Estrogen Receptor Modulators/adverse effects , Selective Estrogen Receptor Modulators/therapeutic use , Testosterone
9.
Stem Cell Res Ther ; 13(1): 303, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35841004

ABSTRACT

BACKGROUND: The rarity of systemic sclerosis (SSc) has hampered the development of therapies for this intractable autoimmune disease. Induced pluripotent stem cell (iPSC) can be differentiated into the key disease-affected cells in vitro. The generation of patient-derived iPSCs has opened up possibilities for rare disease modeling. Since these cells can recapitulate the disease phenotypes of the cell in question, they are useful high-throughput platforms for screening for drugs that can reverse these abnormal phenotypes. METHODS: SSc iPSC was generated from PBMC by Sendai virus. Human iPSC lines from SSc patients were differentiated into dermal fibroblasts and keratinocytes. The iPSC-derived differentiated cells from the SSc patients were used on high-throughput platforms to screen for FDA-approved drugs that could be effective treatments for SSc. RESULTS: Skin organoids were generated from these cells exhibited fibrosis that resembled SSc skin. Screening of the 770-FDA-approved drug library showed that the anti-osteoporotic drug raloxifene reduced SSc iPSC-derived fibroblast proliferation and extracellular matrix production and skin fibrosis in organoids and bleomycin-induced SSc-model mice. CONCLUSIONS: This study reveals that a disease model of systemic sclerosis generated using iPSCs-derived skin organoid is a novel tool for in vitro and in vivo dermatologic research. Since raloxifene and bazedoxifene are well-tolerated anti-osteoporotic drugs, our findings suggest that selective estrogen receptor modulator (SERM)-class drugs could treat SSc fibrosis.


Subject(s)
Scleroderma, Systemic , Skin Diseases , Animals , Cells, Cultured , Fibroblasts/metabolism , Fibrosis , Humans , Leukocytes, Mononuclear/metabolism , Mice , Raloxifene Hydrochloride/adverse effects , Scleroderma, Systemic/genetics , Selective Estrogen Receptor Modulators/adverse effects , Skin/pathology , Skin Diseases/pathology
10.
Therapie ; 77(6): 683-691, 2022.
Article in English | MEDLINE | ID: mdl-35660259

ABSTRACT

INTRODUCTION: Some users of anabolic androgenic steroids (AAS) secretly consume aromatase inhibitors (AI) and selective oestrogen receptor modulators (SERM). Cyber-forums can be potential sources of information. Our aim was to determine the cycles used, and to identify the adverse drug reactions (ADRs) experienced, reported in a bodybuilding forum. METHOD: We collected discussions on a French forum for AAS users (MESO-Rx®), from January 2013 to 2019 on concerning clomiphene, tamoxifen, anastrozole, exemestane and letrozole were collected. Characteristics of the users, duration of cures, treatments, dosages, point of purchase and occurrence of ADRs were analysed. RESULTS: Among the 1792 posts published on the forum, 845 concerned SERM and 571 concerned AI, i.e. 2180 drugs used (several cycles included concomitant consumption). Our population was exclusively male, with an average age of 28.2±6.3years, and had been practising weight training for 6.7±5.6years. The SERMs were mainly used to "revive the hypothalamohypophyseal axis" and the AIs to "fight against androgen aromatisation". The median treatment duration was 22 days for SERM, 70days for anastrozole, 84days for exemestane and 30days for letrozole, with a mean dose of 69mg/d for clomiphene, 22mg/d for tamoxifen, 0.4mg/d for anastrozole, 10mg/d for exemestane, 2mg/d for letrozole. The main way of obtaining these drugs was through the internet. 157 ADRs were identified: 95 for SERMs and 62 for AI. The most represented were acne, musculoskeletal, mood and reproductive disorders. Impaired quality of life (according to the patient) was described in 54% of the SERM posts and 26% of the AI posts. CONCLUSIONS: Patient narratives posted on forums can be a useful tool in the context of doping, to better understand practices, motivations and possibly to bring up pharmacovigilance signals.


Subject(s)
Breast Neoplasms , Drug-Related Side Effects and Adverse Reactions , Male , Humans , Young Adult , Adult , Aromatase Inhibitors/adverse effects , Anastrozole , Letrozole/adverse effects , Selective Estrogen Receptor Modulators/adverse effects , Estrogen Receptor Modulators , Quality of Life , Tamoxifen/therapeutic use , Clomiphene , Drug-Related Side Effects and Adverse Reactions/epidemiology , Breast Neoplasms/drug therapy
11.
Osteoporos Int ; 33(7): 1579-1589, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35138412

ABSTRACT

The aim of this study was to evaluate the risk of acute myocardial infarction in patients taking osteoporosis medication. Patients were taken from the SIDIAP or CPRD database and were matched using propensity scores. Patients with diabetes and chronic kidney disease taking SERMs were at an increased risk. The results favour the cardiovascular safety of alendronate as a first-line choice for osteoporosis treatment. INTRODUCTION: This study aims to evaluate the comparative safety of anti-osteoporosis drugs based on the observed risk of acute myocardial infarction while on treatment in a primary care setting. METHODS: This is a propensity-matched cohort study and meta-analysis. This study was conducted in two primary care record databases covering UK NHS (CPRD) and Catalan healthcare (SIDIAP) patients during 1995-2014 and 2006-2014, respectively. The outcome was acute myocardial infarction while on treatment. Users of alendronate (reference group) were compared to those of (1) other oral bisphosphonates (OBP), (2) strontium ranelate (SR), and (3) selective oestrogen receptor modulator (SERM), after matching on baseline characteristics (socio-demographics, fracture risk factors, comorbidities, and concomitant drug use) using propensity scores. Multiple imputation was used to handle missing data on confounders and competing risk modelling for the calculation of relative risk (sub-distribution hazard ratios (SHR)) according to therapy. Country-specific data were analysed individually and meta-analysed. RESULTS: A 10% increased risk of acute myocardial infarction was found in users of other bisphosphonates as compared to alendronate users within CPRD. The meta-analysis of CPRD and SIDIAP results showed a 9% increased risk in users of other bisphosphonate as compared to alendronate users. Sensitivity analysis showed SERMS users with diabetes and chronic kidney disease were at an elevated risk. CONCLUSIONS: This study provides additional data on the risk of acute myocardial infarction in patients receiving osteoporosis treatment. The results favour the cardiovascular safety of alendronate as a first-line choice for osteoporosis treatment.


Subject(s)
Bone Density Conservation Agents , Myocardial Infarction , Osteoporosis , Alendronate/adverse effects , Bone Density Conservation Agents/adverse effects , Cohort Studies , Databases, Factual , Diabetes Mellitus/epidemiology , Diphosphonates/adverse effects , Humans , Myocardial Infarction/chemically induced , Myocardial Infarction/epidemiology , Osteoporosis/drug therapy , Primary Health Care , Renal Insufficiency, Chronic/epidemiology , Risk Assessment , Selective Estrogen Receptor Modulators/adverse effects , Thiophenes/adverse effects , United Kingdom/epidemiology
12.
Expert Opin Investig Drugs ; 31(6): 515-529, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34694932

ABSTRACT

INTRODUCTION: The selective estrogen receptor degrader (SERD) and full receptor antagonist provides an important therapeutic option for hormone receptor (HR)-positive breast cancer. Endocrine therapies include tamoxifen, a selective estrogen receptor modulator (SERM), that exhibits receptor agonist and antagonist activity, and aromatase inhibitors that block estrogen biosynthesis but which demonstrate acquired resistance. Fulvestrant, the only currently approved SERD, is limited by poor drug-like properties. A key focus for improving disease management has been development of oral SERDs with optimized target occupancy and potency and superior clinical efficacy. AREAS COVERED: Using PubMed, clinicaltrials.gov, and congress websites, this review explored the preclinical development and clinical pharmacokinetics from early phase clinical studies (2015 or later) of novel oral SERDs, including giredestrant, amcenestrant, camizestrant, elacestrant, and rintodestrant. EXPERT OPINION: Numerous oral SERDs are in clinical development, aiming to form the core endocrine therapy for HR-positive breast cancer. Through property- and structure-based drug design of estrogen receptor-binding, antagonism, degradation, anti-proliferation, and pharmacokinetic properties, these SERDs have distinct profiles which impact clinical dosing, efficacy, and safety. Assuming preliminary safety and activity data are confirmed in phase 3 trials, these promising agents could further improve the management, outcomes, and quality of life in HR-positive breast cancer.


Subject(s)
Breast Neoplasms , Receptors, Estrogen , Breast Neoplasms/drug therapy , Estrogen Antagonists/pharmacology , Estrogen Antagonists/therapeutic use , Female , Fulvestrant/pharmacokinetics , Fulvestrant/therapeutic use , Humans , Quality of Life , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/adverse effects
13.
Obes Rev ; 23(2): e13358, 2022 02.
Article in English | MEDLINE | ID: mdl-34559450

ABSTRACT

The incidence of obesity, a recognized risk factor for various metabolic and chronic diseases, including numerous types of cancers, has risen dramatically over the recent decades worldwide. To date, convincing research in this area has painted a complex picture about the adverse impact of high body adiposity on breast cancer onset and progression. However, an emerging but overlooked issue of clinical significance is the limited efficacy of the conventional endocrine therapies with selective estrogen receptor modulators (SERMs) or degraders (SERDs) and aromatase inhibitors (AIs) in patients affected by breast cancer and obesity. The mechanisms behind the interplay between obesity and endocrine therapy resistance are likely to be multifactorial. Therefore, what have we actually learned during these years and which are the main challenges in the field? In this review, we will critically discuss the epidemiological evidence linking obesity to endocrine therapeutic responses and we will outline the molecular players involved in this harmful connection. Given the escalating global epidemic of obesity, advances in understanding this critical node will offer new precision medicine-based therapeutic interventions and more appropriate dosing schedule for treating patients affected by obesity and with breast tumors resistant to endocrine therapies.


Subject(s)
Breast Neoplasms , Aromatase Inhibitors/adverse effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Humans , Obesity/complications , Obesity/drug therapy , Receptors, Estrogen , Selective Estrogen Receptor Modulators/adverse effects
14.
J Korean Med Sci ; 36(27): e186, 2021 Jul 12.
Article in English | MEDLINE | ID: mdl-34254473

ABSTRACT

BACKGROUND: Selective estrogen receptor modulators (SERMs) were associated with an increased risk of venous thromboembolism (VTE) due to the estrogen effect. In this study, we investigated the effect of SERMs on VTE compared to bisphosphonates (BPs) using the Korean National Health Insurance claims database. METHODS: This was a retrospective cohort study. Women over 50 years old who were first prescribed BPs or SERMs for osteoporosis treatment in 2012 were included. The difference in VTE incidence between the SERMs and BP groups was compared. Both groups were followed up for VTE or PE occurrence, death, or until December 2016. The study population was analyzed by 3:1 matching according to age using a multivariate Cox model. RESULTS: The hazard ratio (HR) for VTE was 0.72 (95% confidence interval [CI], 0.40-1.28) in the SERMs group compared to BP group. Older age (60-69 vs. 50-59 years: HR, 3.77; 95% CI, 2.07-6.86 and 70-79 vs. 50-59 years: HR, 5.88; 95% CI, 3.14-11.02), major osteoporotic fracture (HR, 1.77; 95% CI, 1.16- 2.70), atrial fibrillation (HR, 3.31; 95% CI, 1.35-8.11), and estrogen replacement (HR, 3.40; 95% CI, 2.01-5.73) all increased VTE risk. In subgroup analysis of the SERMs group, past hospitalization (HR, 2.24; 95% CI, 1.02-4.92), estrogen replacement (HR, 5.75; 95% CI, 2.29-14.39), and glucocorticoid replacement (HR, 2.71; 95% CI, 1.05-7.0) increased VTE risk. CONCLUSION: SERMs did not increase the risk of VTE compared to BPs in Koreans with osteoporosis. However, old age and estrogen replacement both increased VTE risk.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Diphosphonates/therapeutic use , Osteoporosis/drug therapy , Selective Estrogen Receptor Modulators/therapeutic use , Venous Thromboembolism/epidemiology , Aged , Aged, 80 and over , Bone Density Conservation Agents/adverse effects , Diphosphonates/adverse effects , Humans , Incidence , Middle Aged , Osteoporosis/epidemiology , Republic of Korea/epidemiology , Selective Estrogen Receptor Modulators/adverse effects
15.
Drugs ; 81(7): 771-823, 2021 May.
Article in English | MEDLINE | ID: mdl-33788182

ABSTRACT

The proliferation of targeted anticancer agents over the last two decades has revolutionized cancer treatment and improved survival in many previously refractory malignancies. However, many agents are associated with characteristic ophthalmic adverse effects. It is important that ophthalmologists recognize and maintain a high index of suspicion for these side effects in patients on targeted therapy. Most ophthalmic adverse effects can be treated with specific ocular therapy without discontinuation of cancer treatment, although it is important to be aware of the life-threatening and vision-threatening circumstances that would require therapy cessation in conjunction with the patient's oncologist. This review aims to summarize the ophthalmic adverse effects of targeted and hormonal anticancer agents and briefly describe their management.


Subject(s)
Antineoplastic Agents, Hormonal/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Eye/drug effects , Toxic Optic Neuropathy/epidemiology , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Aromatase Inhibitors/adverse effects , Clinical Trials as Topic , ErbB Receptors/antagonists & inhibitors , Fusion Proteins, bcr-abl/antagonists & inhibitors , Humans , Immune Checkpoint Inhibitors/adverse effects , Janus Kinase 2/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, ErbB-2/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Selective Estrogen Receptor Modulators/adverse effects , Severity of Illness Index , fms-Like Tyrosine Kinase 3/antagonists & inhibitors
16.
Lancet Oncol ; 22(1): 74-84, 2021 01.
Article in English | MEDLINE | ID: mdl-33387497

ABSTRACT

BACKGROUND: Oral fluoropyrimidines, such as S-1, have been shown to have a role in controlling disease progression in metastatic breast cancer. We examined adjuvant treatment with S-1 in patients with oestrogen receptor (ER)-positive and HER2-negative primary breast cancer. METHODS: We did a multicentre, open-label, randomised, controlled, phase 3 trial in 139 sites (137 hospitals and two clinics). Eligible patients were women aged 20-75 years with histologically diagnosed stage I to IIIB invasive breast cancer (intermediate to high risk of recurrence). Patients were temporarily registered at participating institutions and biopsy or surgical samples were collected and sent for central pathological assessment. Patients received 5 years of standard adjuvant endocrine therapy (selective oestrogen receptor modulators with or without ovarian suppression and aromatase inhibitors) with or without 1 year of S-1. Oral S-1 80-120 mg/day was administered twice a day for 14 days with 7 days off. Randomisation (1:1) using the minimisation method was done with six stratification factors (age, axillary lymph node metastasis at surgery or sentinel lymph node biopsy, preoperative or postoperative (neoadjuvant or adjuvant) chemotherapy, preoperative endocrine therapy, proportion of ER-positive cells, and study site). The primary endpoint was invasive disease-free survival, in the full analysis set (all randomly assigned patients, excluding those with significant protocol deviations). The safety analysis set consisted of all patients who received at least one dose of study treatment. Here, we report the results from the interim analysis at the data cutoff date Jan 31, 2019. This trial is registered with Japan Registry of Clinical Trials, jRCTs051180057, and the University hospital Medical Information Network, UMIN000003969. FINDINGS: Between Feb 1, 2012, and Feb 1, 2016, 1930 patients were enrolled in the full analysis set, 957 (50%) received endocrine therapy plus S-1 and 973 (50%) received endocrine therapy alone. Median follow-up was 52·2 months (IQR 42·1-58·9). 155 (16%) patients in the endocrine therapy alone group and in 101 (11%) patients in the endocrine therapy plus S-1 group had invasive disease-free survival events (hazard ratio 0·63, 95% CI 0·49-0·81, p=0·0003). As the primary endpoint was met at interim analysis, the trial was terminated early. The most common grade 3 or worse adverse events were decreased neutrophil count (72 [8%] of 954 patients in the endocrine therapy plus S-1 group vs seven [1%] of 970 patients in the endocrine therapy alone group), diarrhoea (18 [2%] vs none), decreased white blood cells (15 [2%] vs two [<1%]), and fatigue (six [<1%] vs none). Serious adverse events were reported in nine (1%) of 970 patients in the endocrine therapy alone group and 25 (3%) of 954 patients in the endocrine therapy plus S-1 group. There was one (<1%) possible treatment-related death in the endocrine therapy plus S-1 group due to suspected pulmonary artery thrombosis. INTERPRETATION: These data suggest that this combination of S-1 with endocrine therapy could be a potential treatment option for this intermediate and high-risk group of patients with ER-positive, HER2-negative primary breast cancer. FUNDING: Public Health Research Foundation (Japan), Taiho Pharmaceutical.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aromatase Inhibitors/administration & dosage , Biomarkers, Tumor/analysis , Breast Neoplasms/drug therapy , Oxonic Acid/administration & dosage , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Selective Estrogen Receptor Modulators/administration & dosage , Tegafur/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Aromatase Inhibitors/adverse effects , Breast Neoplasms/chemistry , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Disease-Free Survival , Drug Combinations , Female , Humans , Middle Aged , Neoadjuvant Therapy , Oxonic Acid/adverse effects , Selective Estrogen Receptor Modulators/adverse effects , Tegafur/adverse effects , Time Factors , Young Adult
17.
Eur J Cancer ; 145: 19-28, 2021 03.
Article in English | MEDLINE | ID: mdl-33418233

ABSTRACT

BACKGROUND: Four randomised controlled trials (RCTs) in postmenopausal women with advanced breast cancer (ABC) comparing aromatase inhibitors (AIs) versus the selective estrogen receptor modulator tamoxifen, each individually reported significantly longer progression-free survival (PFS) but none showed a significant difference in overall survival (OS). In these trials between 6.8% and 55% of tumours were hormone receptor (HR) status unknown or negative. This meta-analysis restricted the comparison to HR-positive (HR+) tumours. METHODS: Anonymised individual patient data were obtained from three RCTs, EORTC (exemestane versus tamoxifen), Study 0027 and Study 0030 (both anastrozole versus tamoxifen). For the remaining RCT (Femara Study PO25; letrozole versus tamoxifen), odds ratio (OR) or hazard ratio (HzR), with confidence intervals were obtained from the clinical study report, for patients with HR+ tumours, in addition to published data. In total, data were obtained from 2296 patients; 1560 (68%) had HR+ ABC. FINDINGS: The OR for clinical benefit rate was 1.56, in favour of AIs (p < 0.001). The duration of clinical benefit was not significantly increased by AIs (HzR 0·88; p = 0.08). For PFS the HzR (0.82) was in favour of AIs (p = 0·007). However, for OS the HzR (1.05) was not significantly different between AIs and tamoxifen (p = 0.42). INTERPRETATION: Although third generation AIs put significantly more patients into 'clinical benefit', their tumours were not controlled for significantly longer. Overall, while this resulted in a significantly greater PFS in favour of the AIs, this did not translate into improvement in OS.


Subject(s)
Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Postmenopause , Receptors, Estrogen/analysis , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/therapeutic use , Aged , Aromatase Inhibitors/adverse effects , Breast Neoplasms/chemistry , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Clinical Trials, Phase III as Topic , Female , Humans , Middle Aged , Progression-Free Survival , Randomized Controlled Trials as Topic , Selective Estrogen Receptor Modulators/adverse effects , Tamoxifen/adverse effects , Time Factors
18.
J Orthop Res ; 39(7): 1572-1580, 2021 07.
Article in English | MEDLINE | ID: mdl-32485026

ABSTRACT

The use of tamoxifen-inducible models of Cre recombinase in the tendon field is rapidly expanding, resulting in an enhanced understanding of tendon homeostasis and healing. However, the effects of tamoxifen on the tendon are not well-defined, which is particularly problematic given that tamoxifen can have both profibrotic and antifibrotic effects in a tissue-specific manner. Therefore, in the present study, we examined the effects of tamoxifen on tendon homeostasis and healing in male and female C57Bl/6J mice. Tamoxifen-treated mice were compared to corn oil (vehicle)-treated mice. In the "washout" treatment regimen, mice were treated with tamoxifen or corn oil for 3 days beginning 1 week prior to undergoing complete transection and surgical repair of the flexor digitorum longus tendon. In the second regimen, mice were treated with tamoxifen or corn oil beginning on the day of surgery, daily through day 2 postsurgery, and every 48 hours thereafter (D0-2q48) until harvest. All repaired tendons and uninjured contralateral control tendons were harvested at day 14 postsurgery. Tamoxifen treatment had no effect on tendon healing in male mice, regardless of the treatment regimen, while Max load was significantly decreased in female repairs in the Tamoxifen washout group, relative to corn oil. In contrast, D0-2q48 corn oil treatment in female mice led to substantial disruptions in tendon homeostasis, relative to washout corn oil treatment. Collectively, these data clearly define the functional effects of tamoxifen and corn oil treatment in the tendon and inform future use of tamoxifen-inducible genetic models.


Subject(s)
Selective Estrogen Receptor Modulators/adverse effects , Tamoxifen/adverse effects , Tendon Injuries , Tendons/drug effects , Wound Healing/drug effects , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Homeostasis/drug effects , Male , Mice, Inbred C57BL , Mice, Transgenic
20.
Curr Protoc Chem Biol ; 12(4): e90, 2020 12.
Article in English | MEDLINE | ID: mdl-33315311

ABSTRACT

Drug-induced liver injury is an important cause of non-approval in drug development and the withdrawal of already approved drugs from the market. Screening human hepatic cell lines for toxicity has been used extensively to predict drug-induced liver injury in preclinical drug development. Assessing hepatic-cell health with more diverse markers will increase the value of in vitro assays and help predict the mechanism of toxicity. We describe three live cell-based assays using HepG2 cells to measure cell health parameters indicative of hepatotoxicity. The first assay measures cellular ATP levels using luciferase. The second and third assays are multiparametric high-content screens covering a panel of cell health markers including cell count, mitochondrial membrane potential and structure, nuclear morphology, vacuolar density, and reactive oxygen species and glutathione levels. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Measurement of cellular ATP content Basic Protocol 2: High-content analysis assay to assess cell count, mitochondrial membrane potential and structure, and reactive oxygen species Basic Protocol 3: High-content analysis assay to assess nuclear morphology, vacuoles, and glutathione content Support Protocol 1: Subculturing and maintaining HepG2 cells Support Protocol 2: Plating HepG2 cell line Support Protocol 3: Transferring compounds by pin tool Support Protocol 4: Generating dose-response curves.


Subject(s)
Adenosine Triphosphate/analysis , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , High-Throughput Screening Assays , Models, Biological , Oxidative Stress/drug effects , Adenosine Triphosphate/antagonists & inhibitors , Biomarkers/analysis , Chemical and Drug Induced Liver Injury/diagnosis , Hep G2 Cells , Humans , Raloxifene Hydrochloride/adverse effects , Selective Estrogen Receptor Modulators/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...