Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 563
Filter
1.
Sci Total Environ ; 806(Pt 1): 150517, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34794910

ABSTRACT

Beta-cypermethrin (ß-CYP), a widely-used pyrethroid pesticide, is considered to have anti-androgenic effects and could impair male reproduction. To ascertain whether MAPK pathways, DNA methyltransferases (DNMTs), and miRNAs played pleiotropic roles in ß-CYP-mediated testicular dysfunction, Sprague-Dawley rats and Leydig cells were employed in this study. Results showed that plasma testosterone levels were declined, testicular histomorphology and ultrastructures were abnormally altered, and Leydig cell functions were damaged after ß-CYP exposure. JNK and p38/MAPK pathways were inactivated, accompanied by the decrease in c-Jun and Sp1 expressions. Specific activators/inhibitors of MAPK pathways and Co-IP demonstrated that DNMT3α was synergistically regulated by JNK/p38 pathways. The activity, mRNA and protein expressions of DNMT3α were all reduced by ß-CYP. ß-CYP induced expressions of intronic miR-140-5p and its host gene Wwp2, and then overexpressed miR-140-5p suppressed steroidogenic StAR, P450scc, and 3ß-HSD by directly targeting SF-1. SF-1 silencing/overexpression, ChIP, and qPCR indicated that SF-1 modulated positively StAR, P450scc, and 3ß-HSD expressions by directly binding to their promoter regions. Intriguingly, 5α-reductase expressions were downregulated after ß-CYP exposure. Collectively, ß-CYP has the anti-androgenic feature and the DNMT3α/miR-140-5p/SF-1 cascade co-regulated by JNK/p38 functions critically in ß-CYP-caused testosterone declines. The downregulation of 5α-reductases may be a potential compensatory mechanism of the organism.


Subject(s)
Insecticides/toxicity , MicroRNAs , Pyrethrins , Testis/drug effects , Testis/pathology , Testosterone/deficiency , Animals , Male , MicroRNAs/genetics , Pyrethrins/toxicity , Rats , Rats, Sprague-Dawley , Testosterone/antagonists & inhibitors
4.
Br J Sports Med ; 55(15): 865-872, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33648944

ABSTRACT

OBJECTIVES: We systemically reviewed the literature to assess how long-term testosterone suppressing gender-affirming hormone therapy influenced lean body mass (LBM), muscular area, muscular strength and haemoglobin (Hgb)/haematocrit (HCT). DESIGN: Systematic review. DATA SOURCES: Four databases (BioMed Central, PubMed, Scopus and Web of Science) were searched in April 2020 for papers from 1999 to 2020. ELIGIBILITY CRITERIA FOR SELECTING STUDIES: Eligible studies were those that measured at least one of the variables of interest, included transwomen and were written in English. RESULTS: Twenty-four studies were identified and reviewed. Transwomen experienced significant decreases in all parameters measured, with different time courses noted. After 4 months of hormone therapy, transwomen have Hgb/HCT levels equivalent to those of cisgender women. After 12 months of hormone therapy, significant decreases in measures of strength, LBM and muscle area are observed. The effects of longer duration therapy (36 months) in eliciting further decrements in these measures are unclear due to paucity of data. Notwithstanding, values for strength, LBM and muscle area in transwomen remain above those of cisgender women, even after 36 months of hormone therapy. CONCLUSION: In transwomen, hormone therapy rapidly reduces Hgb to levels seen in cisgender women. In contrast, hormone therapy decreases strength, LBM and muscle area, yet values remain above that observed in cisgender women, even after 36 months. These findings suggest that strength may be well preserved in transwomen during the first 3 years of hormone therapy.


Subject(s)
Body Composition/drug effects , Hemoglobin A/drug effects , Muscle Strength/drug effects , Sports , Testosterone/antagonists & inhibitors , Transgender Persons , Adipose Tissue/drug effects , Androgen Antagonists/pharmacology , Athletic Performance , Body Composition/physiology , Cyproterone Acetate/pharmacology , Estradiol/pharmacology , Female , Hematocrit , Humans , Male , Muscle Strength/physiology , Muscle, Skeletal/drug effects , Sports/physiology , Time Factors , Transsexualism/blood
5.
J Pharmacokinet Pharmacodyn ; 48(4): 465-477, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33538922

ABSTRACT

Intermittent androgen deprivation therapy with gonadotropin-releasing-hormone (GnRH) agonists can prevent or delay disease progression and development of castration resistant prostate cancer for subpopulations of prostate cancer patients. It may also reduce risk and severity of side effects associated with chemical castration in prostate cancer (PCa) patients. One of the earliest comprehensively documented clinical trials on this was reported in a Canadian patient population treated with leuprorelin preceded by a lead-in with cyproterone acetate. A systems-based mixed effect analysis of testosterone response in active and recovery phases allows inference of new information from this patient population. Efficacy of androgen deprivation therapy is presumed to depend on a treshold value for testosterone at the nadir, below which no additional beneficial effects on PSA reponse can be expected, and occurance of testosterone breakthroughs during active therapy. The present analysis results in a mixed effect model, incorporating GnRH receptor activation, testosterone turnover and feedback mechanisms, describing and predicting testosterone inhibition under intermittent androgen deprivation therapy on the individual and population level, during multiple years of therapy. Testosterone levels in these patients decline over time with an estimated first order rate constant of 0.083 year-1(T1/2 = 8.4 y), with a substantial distribution among this patient population, compared to the general population. PCa patients leaving the trial due to unmanageble PSA relapse appear to have slightly higher testosterone levels at the nadir than sustained responders. These findings are expected to contribute to an increased understanding of the role of testosterone in long term disease progression of prostate cancer.


Subject(s)
Androgen Antagonists/therapeutic use , Gonadotropin-Releasing Hormone/agonists , Prostatic Neoplasms/drug therapy , Testosterone/antagonists & inhibitors , Aged , Androgen Antagonists/administration & dosage , Androgen Antagonists/adverse effects , Androgen Antagonists/pharmacology , Cyproterone Acetate/administration & dosage , Cyproterone Acetate/adverse effects , Cyproterone Acetate/therapeutic use , Humans , Leuprolide/administration & dosage , Leuprolide/adverse effects , Leuprolide/therapeutic use , Male , Prostate-Specific Antigen/blood , Testosterone/blood , Treatment Outcome
6.
J Natl Cancer Inst ; 113(2): 192-198, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33527137

ABSTRACT

BACKGROUND: Metformin has been associated with lower breast cancer (BC) risk and improved outcomes in observational studies. Multiple biologic mechanisms have been proposed, including a recent report of altered sex hormones. We evaluated the effect of metformin on sex hormones in MA.32, a phase III trial of nondiabetic BC subjects who were randomly assigned to metformin or placebo. METHODS: We studied the subgroup of postmenopausal hormone receptor-negative BC subjects not receiving endocrine treatment who provided fasting blood at baseline and at 6 months after being randomly assigned. Sex hormone-binding globulin, bioavailable testosterone, and estradiol levels were assayed using electrochemiluminescence immunoassay. Change from baseline to 6 months between study arms was compared using Wilcoxon sum rank tests and regression models. RESULTS: 312 women were eligible (141 metformin vs 171 placebo); the majority of subjects in each arm had T1/2, N0, HER2-negative BC and had received (neo)adjuvant chemotherapy. Mean age was 58.1 (SD=6.9) vs 57.5 (SD=7.9) years, mean body mass index (BMI) was 27.3 (SD=5.5) vs 28.9 (SD=6.4) kg/m2 for metformin vs placebo, respectively. Median estradiol decreased between baseline and 6 months on metformin vs placebo (-5.7 vs 0 pmol/L; P < .001) in univariable analysis and after controlling for baseline BMI and BMI change (P < .001). There was no change in sex hormone-binding globulin or bioavailable testosterone. CONCLUSION: Metformin lowered estradiol levels, independent of BMI. This observation suggests a new metformin effect that has potential relevance to estrogen sensitive cancers.


Subject(s)
Breast Neoplasms/drug therapy , Gonadal Steroid Hormones/antagonists & inhibitors , Metformin/administration & dosage , Body Mass Index , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Estradiol/genetics , Female , Gonadal Steroid Hormones/genetics , Humans , Middle Aged , Receptor, ErbB-2/genetics , Testosterone/antagonists & inhibitors , Testosterone/genetics
8.
Toxicology ; 449: 152663, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33359577

ABSTRACT

Perfluorooctane sulfonate (PFOS), a stable end-product of perfluorinated compounds (PFCs), is associated with male reproductive disorders, but its underlying mechanisms are still unclear. We used in vivo and in vitro models to investigate the effects of PFOS on testosterone biosynthesis and related mechanisms. First, male ICR mice were orally administered PFOS (0-10 mg/kg/bw) for 4 weeks. Bodyweight, sperm count, reproductive hormones, mRNA expression of the genes related to testosterone biosynthesis, and the protein expression of protein kinase A (PKA), p38 mitogen-activated protein kinase (MAPK), cAMP-response element binding protein (CREB), CREB regulated transcription coactivator 2 (CRTC2) and steroidogenic acute regulatory protein (StAR) were evaluated. Furthermore, mouse primary Leydig cells were used to delineate the molecular mechanisms that mediate the effects of PFOS on testosterone biosynthesis. Our results demonstrated that PFOS dose-dependently decreased sperm count, testosterone level, CRTC2/StAR expression, and damaged testicular interstitium morphology, paralleled by increase in phosphorylated PKA, CREB and p38 in testes. Additionally, similar to the in vivo results, PFOS significantly decreased testosterone secretion, CRTC2/StAR expression, interaction between CREB and CRTC2 and binding of CREB/CRTC2 to StAR promoter region, paralleled by increase in phosphorylated-p38, PKA, and CREB expression. Meanwhile, inhibition of p38 by SB203580, or inhibition of PKA by H89 can significantly alleviate the above PFOS-induced effects. As such, the present study highlights a role of the CREB/CRTC2/StAR signaling pathway in PFOS-induced suppression of testosterone biosynthesis, advancing our understanding of molecular mechanisms for PFOS-induced male reproductive disorders.


Subject(s)
Alkanesulfonic Acids/toxicity , Cyclic AMP Response Element-Binding Protein/metabolism , Fluorocarbons/toxicity , Leydig Cells/metabolism , Phosphoproteins/metabolism , Testosterone/biosynthesis , Transcription Factors/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Dose-Response Relationship, Drug , Leydig Cells/drug effects , Male , Mice , Mice, Inbred ICR , Phosphoproteins/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Testosterone/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors
9.
Sex Abuse ; 33(4): 475-500, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32167420

ABSTRACT

For a particular subgroup of individuals with severe paraphilic disorders and a high risk of sexual recidivism, the combination of sex drive-reducing medications and psychotherapy is a promising treatment approach. The present quasi-experimental study aims at comparing differences in clinical characteristics and dynamic risk factors between persons receiving (+TLM, n = 38) versus not receiving (-TLM, n = 22) testosterone-lowering medications (TLMs). Individuals receiving TLM were more frequently diagnosed with paraphilic disorders. Neither the criminal history nor average risk scores differed between the two groups. In the +TLM, Stable-2007 scores showed a stronger decrease after TLM treatment was started. This accounted especially for the general and sexual self-regulation subscales. Individual variations in risk, however, were not predicted by TLM but were significantly related to treatment duration and Psychopathy Checklist-Revised (PCL-R) Factor I. Paraphilic patients with problems in self-regulatory abilities seem to profit most from pharmacological sex drive-reducing treatment. Furthermore, therapists seem to underestimate deviant sexual fantasies in medicated patients.


Subject(s)
Androgen Antagonists/therapeutic use , Criminals/psychology , Paraphilic Disorders/therapy , Psychotherapy , Sex Offenses/psychology , Testosterone/antagonists & inhibitors , Adult , Aged , Duration of Therapy , Germany/epidemiology , Humans , Male , Middle Aged , Recidivism/prevention & control , Risk Assessment/methods , Risk Factors , Self-Control , Treatment Outcome
10.
Front Endocrinol (Lausanne) ; 12: 813067, 2021.
Article in English | MEDLINE | ID: mdl-35002984

ABSTRACT

Aim: To explore the chronic effects of metformin on testosterone levels in men with type 2 diabetes mellitus (T2DM). Methods: This is a secondary analysis of a real-world study evaluating the efficacy and safety of premixed insulin treatment in patients with T2DM via 3-month intermittent flash glucose monitoring. Male patients aged 18-60 who were using metformin during the 3-month study period were included as the metformin group. The control group included males without metformin therapy by propensity score matching analysis with age as a covariate. Testosterone levels were measured at baseline and after 3-month treatment. Results: After 3-month treatment, the control group had higher levels of total testosterone, free and bioavailable testosterone than those at baseline (P<0.05). Compared with the control group, the change of total (-0.82 ± 0.59 vs. 0.99 ± 0.59 nmol/L) and bioavailable (-0.13 ± 0.16 vs. 0.36 ± 0.16 nmol/L) testosterone levels in the metformin group significantly decreased (P=0.036 and 0.029, respectively). In Glycated Albumin (GA) improved subgroup, the TT, FT, and Bio-T levels in the control subgroup were higher than their baseline levels (P < 0.05). Compared with the metformin subgroup, TT level in the control subgroup also increased significantly (P=0.044). In GA unimproved subgroup, the change of TT level in the metformin subgroup was significantly lower than that in the control subgroup (P=0.040). Conclusion: In men with T2DM, 3-month metformin therapy can reduce testosterone levels, and counteract the testosterone elevation that accompanied with the improvement of blood glucose. Clinical Trial Registration: https://www.clinicaltrials.gov/ct2/show/NCT04847219?term=04847219&draw=2&rank=1.


Subject(s)
Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Metformin/administration & dosage , Testosterone/blood , Aged , Blood Glucose/drug effects , Blood Glucose/metabolism , Humans , Hypoglycemic Agents/adverse effects , Male , Metformin/adverse effects , Middle Aged , Testosterone/antagonists & inhibitors , Treatment Outcome
11.
Nat Rev Urol ; 18(1): 19-32, 2021 01.
Article in English | MEDLINE | ID: mdl-33214706

ABSTRACT

Marijuana is the most widely consumed recreational drug worldwide, which raises concerns for its potential effects on fertility. Many aspects of human male reproduction can be modulated by cannabis-derived extracts (cannabinoids) and their endogenous counterparts, known as endocannabinoids (eCBs). These latter molecules act as critical signals in a variety of physiological processes through receptors, enzymes and transporters collectively termed the endocannabinoid system (ECS). Increasing evidence suggests a role for eCBs, as well as cannabinoids, in various aspects of male sexual and reproductive health. Although preclinical studies have clearly shown that ECS is involved in negative modulation of testosterone secretion by acting both at central and testicular levels in animal models, the effect of in vivo exposure to cannabinoids on spermatogenesis remains a matter of debate. Furthermore, inconclusive clinical evidence does not seem to support the notion that plant-derived cannabinoids have harmful effects on human sexual and reproductive health. An improved understanding of the complex crosstalk between cannabinoids and eCBs is required before targeting of ECS for modulation of human fertility becomes a reality.


Subject(s)
Cannabinoids/metabolism , Endocannabinoids/metabolism , Genitalia, Male/metabolism , Signal Transduction/physiology , Animals , Cannabinoids/administration & dosage , Endocannabinoids/administration & dosage , Genitalia, Male/drug effects , Humans , Male , Receptors, Cannabinoid/metabolism , Signal Transduction/drug effects , Testosterone/antagonists & inhibitors , Testosterone/metabolism
12.
J Urol ; 205(2): 554-560, 2021 02.
Article in English | MEDLINE | ID: mdl-33090917

ABSTRACT

PURPOSE: Nonadherence to dosing schedules for androgen deprivation therapy increases the risk of testosterone escape for patients with prostate cancer. Two approved formulations of leuprolide acetate, the most commonly prescribed androgen deprivation therapy in the United States, use different extended release delivery technologies: an in situ gel and microspheres. We evaluated the prevalence and impact of late dosing on testosterone suppression for gel and microsphere formulations of leuprolide acetate. MATERIALS AND METHODS: We retrospectively analyzed records of patients with prostate cancer treated with gel or microsphere delivery of leuprolide acetate. Analyses used 2 definitions of "month," "28-day" (late dosing after day 28, 84, 112 or 168) and "extended" (late dosing after day 32, 97, 128 and 194). Frequencies of late dosing and associated testosterone values were calculated. RESULTS: A total of 2,038 patients received gel and 8,360 received microsphere formulations of leuprolide acetate. More than 80% and 27% of injections were late for 28-day and extended month, respectively. For 28-day month late injections 10% (gel delivery) and 14% (microsphere delivery) of testosterone values were above 50 ng/dl, and 25% (gel) vs 33% (microsphere) were above 20 ng/dl. For extended month 18% (gel) vs 25% (microsphere) were above 50 ng/dl, and 34% (gel) vs 44% (microsphere) were above 20 ng/dl. Microsphere leuprolide acetate was 1.5 times more likely to have testosterone above 50/20 ng/dl vs gel. Least square mean testosterone was 34 ng/dl (gel) vs 46 ng/dl (microsphere) for 28-day month, and 48 ng/dl (gel) vs 76 ng/dl (microsphere) for extended month. CONCLUSIONS: Leuprolide acetate therapies were frequently administered late. Gel formulation demonstrated higher rates of testosterone 50 ng/dl or less and 20 ng/dl or less than microsphere formulation. Optimal testosterone suppression can impact prostate cancer progression and patient survival, and differences in extended release technology for androgen deprivation therapy appear relevant.


Subject(s)
Androgen Antagonists/administration & dosage , Leuprolide/administration & dosage , Prostatic Neoplasms/drug therapy , Testosterone/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Gels , Humans , Male , Microspheres , Middle Aged , Retrospective Studies , Time Factors , United States , Young Adult
14.
Physiol Res ; 69(Suppl 2): S187-S192, 2020 09 30.
Article in English | MEDLINE | ID: mdl-33094617

ABSTRACT

The adrenal glands produce significant amounts of steroid hormones and their metabolites, with various levels of androgenic activities. Until recently, the androgenic potency of these adrenal-derived compounds were not well known, but some recent studies have shown that the production of 11-oxo- and 11beta-hydroxy-derived testosterone and dihydrotestosterone evidently have high androgenic activity. This fact has clinical importance, for instance, in various types of congenital adrenal hyperplasia with androgenization or polycystic ovarian syndrome, and laboratory determinations of these substances could help to better evaluate the total androgen pressure in patients with these disorders. Another area of concern is the treatment of prostate cancer with androgen deprivation, which loses effectiveness after a certain time. The concurrent blocking of the secretion of adrenal C(19)-steroids, whether using corticoids or adrenostatics, could increase the effectiveness of androgen-deprivation therapy.


Subject(s)
Adrenal Hyperplasia, Congenital/drug therapy , Androgen Antagonists/therapeutic use , Androgens/chemistry , Androgens/metabolism , Prostatic Neoplasms/drug therapy , Testosterone/analogs & derivatives , Adrenal Hyperplasia, Congenital/metabolism , Adrenal Hyperplasia, Congenital/pathology , Animals , Humans , Male , Molecular Targeted Therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Testosterone/antagonists & inhibitors , Testosterone/metabolism
15.
Eur J Endocrinol ; 183(6): 561-569, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33055297

ABSTRACT

OBJECTIVE: The impact of different combinations of long-term gender-affirming hormone therapy (GAHT) in transwomen (TW) is largely unknown. To assess the effects of 5-year administration of cyproterone acetate (CPA) or leuprolide acetate (Leu) plus transdermal or oral estradiol (E). DESIGN: Cohort study based on prospectively collected data. Fifty TW received 50 mg CPA daily orally (n = 25; CPA+E group) or 3.75 mg Leu i.m. monthly (n = 25; Leu+E group) with 1 or 2 mg E daily for 5 years. Reproductive hormones, biochemical and anthropometric parameters, body composition and bone mineral density (BMD) were assessed. RESULTS: LH, FSH and total testosterone levels were similarly and significantly suppressed in both groups. Prolactin increased only in the CPA+E group (P = 0.002). Fasting insulin resistance and glucose progressively increased in the CPA+E group only (treatment × time effect P = 0.002 and P = 0.043, respectively). Total cholesterol increased more in the Leu+E group than in the CPA+E group and HDL-cholesterol decreased in the CPA+E group (time × treatment interaction effect, P = 0.007). Lumbar and total body BMD increased in both groups after 3 years. No serious adverse events were recorded. CONCLUSIONS: Both regimens were effective in suppression of T production. CPA+E worsened the metabolic profile with a slight increase in PRL levels. All subjects presented an increase in BMD regardless of treatment. These preliminary data could have clinical implications in the choice of GAHT, in particular for those TW not requiring gender-affirming surgery.


Subject(s)
Cyproterone Acetate/administration & dosage , Estradiol/administration & dosage , Leuprolide/administration & dosage , Testosterone/blood , Transsexualism/blood , Transsexualism/drug therapy , Adult , Androgen Antagonists/administration & dosage , Cohort Studies , Drug Administration Schedule , Drug Therapy, Combination , Estrogens/administration & dosage , Female , Fertility Agents, Female/administration & dosage , Humans , Male , Middle Aged , Prospective Studies , Testosterone/antagonists & inhibitors , Transgender Persons
16.
Br J Cancer ; 123(12): 1715-1719, 2020 12.
Article in English | MEDLINE | ID: mdl-32989230

ABSTRACT

Androgen receptor (AR) signalling drives neoplastic growth and therapy resistance in prostate cancer. Recent clinical data show that docetaxel combined with androgen deprivation therapy improves outcome in hormone-sensitive disease. We studied whether testosterone and AR signalling interferes with docetaxel treatment efficacy in castration-resistant prostate cancer (CRPC). We found that testosterone supplementation significantly impaired docetaxel tumour accumulation in a CRPC model, resulting in decreased tubulin stabilisation and antitumour activity. Furthermore, testosterone competed with docetaxel for uptake by the drug transporter OATP1B3. Irrespective of docetaxel-induced tubulin stabilisation, AR signalling by testosterone counteracted docetaxel efficacy. AR-pathway activation could also reverse long-term tumour regression by docetaxel treatment in vivo. These results indicate that to optimise docetaxel efficacy, androgen levels and AR signalling need to be suppressed. This study lends evidence for continued maximum suppression of AR signalling by combining targeted therapeutics with docetaxel in CRPC.


Subject(s)
Androgen Antagonists/therapeutic use , Antineoplastic Agents/therapeutic use , Docetaxel/therapeutic use , Drug Resistance, Neoplasm/physiology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism , Testosterone/pharmacology , Acetylation , Androgen Antagonists/pharmacokinetics , Androgen Receptor Antagonists/therapeutic use , Animals , Antineoplastic Agents/pharmacokinetics , Cell Death , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival , Disease Progression , Docetaxel/pharmacokinetics , Drug Interactions , Humans , In Situ Nick-End Labeling , Male , Mice , Mice, Nude , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Prostate-Specific Antigen/biosynthesis , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/drug effects , Signal Transduction/drug effects , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Testosterone/administration & dosage , Testosterone/antagonists & inhibitors , Testosterone/metabolism , Tubulin/drug effects , Tubulin/metabolism
17.
Eur Urol Oncol ; 3(6): 717-724, 2020 12.
Article in English | MEDLINE | ID: mdl-32807727

ABSTRACT

BACKGROUND: ECOG3805 is a randomized trial of testosterone suppression with or without docetaxel for metastatic hormone-sensitive prostate cancer (mHSPC). Deeper prostate-specific antigen (PSA) suppression is prognostic for outcome. However, the concordance of PSA rise and radiographic progression has not been examined previously in mHSPC, whereas this has been reported in metastatic castration-resistant prostate cancer. OBJECTIVE: To determine the patterns of progression by PSA and radiographic parameters in patients in ECOG3805. DESIGN, SETTING, AND PARTICIPANTS: We conducted a retrospective analysis of all patients in ECOG3805. Patients were classified according to the PSA level at progression (whether PSA level was below 2.0 ng/mL or not) and the type of progression event in the study (either PSA progression as defined by the study with or without clinical progression, or clinical progression alone). Baseline demographics, clinical outcomes, and patterns of progression were compared between the groups. RESULTS AND LIMITATIONS: One in eight patients had clinical progression below a PSA level of 2 ng/mL, and approximately 25% developed clinical progression in the absence of confirmed PSA progression. Overall survival from randomization was shorter in patients with clinical progression without confirmed PSA progression than in patients with PSA progression alone as the first progression. Patient demographics at study entry were not predictive of the pattern of progression. Study limitations include its retrospective and post hoc nature. CONCLUSIONS: Clinical progression prior to PSA rise or at low PSA levels is a relatively frequent phenomenon in mHSPC and is associated with poorer overall survival. Further biological and clinical studies of these patients are warranted. PATIENT SUMMARY: Reliance on prostate-specific antigen (PSA) alone is an inadequate strategy to monitor patients undergoing treatment for metastatic hormone-sensitive prostate cancer. Prostate cancer can get worse on scans even with low PSA and/or no or small changes in PSA. Imaging should be added to PSA testing to monitor patients with metastatic prostate cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Kallikreins/blood , Prostate-Specific Antigen/blood , Prostate/diagnostic imaging , Prostatic Neoplasms/drug therapy , Testosterone/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Disease Progression , Docetaxel/pharmacology , Docetaxel/therapeutic use , Gonadotropin-Releasing Hormone/pharmacology , Gonadotropin-Releasing Hormone/therapeutic use , Humans , Male , Middle Aged , Neoplasm Grading , Prognosis , Progression-Free Survival , Prospective Studies , Prostate/drug effects , Prostate/pathology , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Retrospective Studies , Tumor Burden/drug effects
18.
Biol Trace Elem Res ; 195(1): 125-134, 2020 May.
Article in English | MEDLINE | ID: mdl-31313246

ABSTRACT

Exposure to arsenic (AS) causes abnormalities in the reproductive system; however, the precise cellular pathway of AS toxicity on steroidogenesis in developing F1-male mice has not been clearly defined. In this study, paternal mice were treated with arsenic trioxide (As2O3; 0, 0.2, 2, and 20 ppm in drinking water) from 5 weeks before mating until weaning and continued for male offspring from weaning until maturity (in vivo). Additionally, Leydig cells (LCs) were isolated from the testes of sacrificed F1-intact mature male mice and incubated with As2O3 (0, 1, 10, and 100 µM) for 48 h (in vitro). Biomarkers of mitochondrial impairment, oxidative stress, and several steroidogenic genes, including the steroidogenic acute regulatory (StAR) protein, cytochrome P450 side-chain cleaving enzyme (P450scc; Cyp11a), 3ß-hydroxysteroid dehydrogenase (3ß-HSD), and 17ß-hydroxysteroid dehydrogenase (17ß-HSD), were evaluated. High doses of As2O3 interrupted testosterone (T) biosynthesis and T-related gene expression in these experimental models. Altogether, overconsumption of As2O3 can cause testicular and LC toxicity through mitochondrial-related pathways and oxidative stress indices as well as downregulation of androgenic-related genes in mice and isolated LCs. These results could lead to the development of preventive/therapeutic procedures against As2O3-induced reproductive toxicity. Graphical Abstract Mohammad Mehdi Ommati and Reza Heidari contributed equally to this study.


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Arsenic Trioxide/pharmacology , Cholesterol Side-Chain Cleavage Enzyme/antagonists & inhibitors , Down-Regulation/drug effects , Mitochondria/drug effects , Testosterone/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , Administration, Oral , Animals , Arsenic Trioxide/administration & dosage , Cell Survival/drug effects , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Leydig Cells/drug effects , Leydig Cells/metabolism , Male , Mice , Mice, Inbred BALB C , Mitochondria/metabolism , Oxidative Stress/drug effects , Testosterone/metabolism
19.
J Urol ; 203(4): 743-750, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31580749

ABSTRACT

PURPOSE: We evaluated the timeliness of androgen deprivation therapy dosing, the impact of dosing nonadherence on testosterone, and the frequency of testosterone and prostate specific antigen testing in patients with prostate cancer. MATERIALS AND METHODS: We retrospectively analyzed the records of 22,860 patients with prostate cancer treated with luteinizing hormone-releasing hormone agonists. Analyses were done using 2 definitions of month, including a 28-day month (late dosing after day 28, 84, 112 or 168) and an extended month (late after day 32, 97, 128 or 194) for 1, 3, 4 and 6-month formulations, respectively. The prevalence of late dosing, associated testosterone values, and the frequency of testosterone and prostate specific antigen testing were assessed. Statistical significance was assessed with the unpaired t-test. RESULTS: Of the injections 84% and 27% were late for the 28-day and extended month analyses, respectively. For the 28-day month 60% and 29% of injections were late by more than 1 and more than 2 weeks, respectively. Of testosterone values 4% were greater than 50 ng/dl for early/on time injections using both definitions, and 15% and 27% were greater than 50 ng/dl when late, and for the 28-day month and the extended month, respectively. For early/on time vs late injections 22% vs 31% of testosterone values were greater than 20 ng/dl for the 28-day month and 21% vs 43% for the extended month. Mean testosterone was higher when late (49 ng/dl for 28-day month, 79 ng/dl for extended month) vs early/on time (both 21 ng/dl). Of the injections prostate specific antigen measurements were performed in 83% and testosterone assessment was done in only 13%. CONCLUSIONS: Luteinizing hormone-releasing hormone agonists were frequently (84%) administered later than the schedules used in pivotal trials. Nearly half of the late testosterone values for the extended month were greater than 20 ng/dl and mean testosterone was almost double the castration level. Elevated testosterone remained unidentified with infrequent testing.


Subject(s)
Antineoplastic Agents, Hormonal/administration & dosage , Gonadotropin-Releasing Hormone/agonists , Medication Adherence/statistics & numerical data , Prostatic Neoplasms/drug therapy , Testosterone/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Dose-Response Relationship, Drug , Drug Administration Schedule , Humans , Kallikreins/blood , Male , Middle Aged , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Retrospective Studies , Testosterone/blood , Time Factors , United States , Young Adult
20.
Niger J Physiol Sci ; 34(1): 69-75, 2019 Jun 30.
Article in English | MEDLINE | ID: mdl-31449274

ABSTRACT

Anti-androgenic substances, mainly prostate 5α-reductase inhibitors, used in the treatment of benign prostatic hyperplasia (BPH) have been associated with side effects in man and animals. To reduce these side effects as well as suppress BPH development, the management of the condition has come to include dietary interventions. This study investigated the effect of some cooking oils on testosterone-induced hyperplasia of the prostate in rats. Male Sprague-dawley rats were distributed into eighteen groups (n=6) as A-R. A negative control group was injected subcutaneously with soya oil; while prostatic hyperplasia was induced subcutaneously in groups B-R with 3mg/kg testosterone daily for 14days. Group B was the positive control (BPH group) while groups C-R were also administered orally 800mg/kg of coconut, castor, canola, cottonseed, pomegranate, blackseed, sheabutter, olive oil, codliver, sardine, palm, repeatedly heated palm (RHPO), vegetable, repeatedly-heated vegetable (RHVO), sesame, and groundnut oils respectively, daily, for 14 days. Blood sample was drawn via retro-orbital sinus for the estimation of serum testosterone(T) and dihydrotestosterone (DHT) level and rats were thereafter euthanized to obtain the prostates for T and DHT determination as well as tissue weights. Data are mean ± SEM, compared by ANOVA. The oils significantly reduced the increase in prostate weight (PW) to body weight (BW) ratio induced by testosterone. Apart from the fact that all the oils reduced the PW:BW ratio, the blackseed, sheabutter, sardine, vegetable and groundnut oils suppressed the DHT level in the serum, while pomegranate, olive, RHPO reduced DHT level in the prostate compared to the BPH rats. This study suggests that blackseed, sheabutter, sardine, vegetable, groundnut, pomegranate, olive, and RHPO oils could inhibit testosterone-induced hyperplasia of the prostate and therefore may be beneficial in the management of BPH.


Subject(s)
Disease Models, Animal , Fatty Acids/administration & dosage , Plant Oils/administration & dosage , Prostatic Hyperplasia/blood , Prostatic Hyperplasia/drug therapy , Testosterone/blood , Administration, Oral , Animals , Male , Prostatic Hyperplasia/pathology , Rats , Rats, Sprague-Dawley , Testosterone/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...