Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 245
Filter
1.
Med ; 5(4): 311-320.e3, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38471508

ABSTRACT

BACKGROUND: Intravenous immune checkpoint blockade (ICB) has shown poor response rates in recurrent gynecologic malignancies. Intraperitoneal (i.p.) ICB may result in enhanced T cell activation and anti-tumor immunity. METHODS: In this phase 1b study, registered at Clinical. TRIALS: gov (NCT03508570), initial cohorts received i.p. nivolumab monotherapy, and subsequent cohorts received combination i.p. nivolumab every 2 weeks and i.p. ipilimumab every 6 weeks, guided by a Bayesian design. The primary objective was determination of the recommended phase 2 dose (RP2D) of the combination. Secondary outcomes included toxicity, objective response rate (ORR), progression-free survival (PFS), and overall survival (OS). FINDINGS: The trial enrolled 23 patients: 18 with ovarian cancer, 2 with uterine cancer, and 3 with cervical cancer. Study evaluable patients (n = 16) received a median of 2 prior lines of therapy (range: 1-8). Partial response was observed in 2 patients (12.5%; 1 ovarian, 1 uterine), and complete response was observed in 1 patient (6.3%) with cervical cancer, for an ORR of 18.8% (95% confidence interval: 4.0%-45.6%). The median duration of response was 14.8 months (range: 4.1-20.8), with one complete response ongoing. Median PFS and OS were 2.7 months and not reached, respectively. Grade 3 or higher immune-related adverse events occurred in 2 (8.7%) patients. CONCLUSIONS: i.p. administration of dual ICB is safe and demonstrated durable responses in a subset of patients with advanced gynecologic malignancy. The RP2D is 3 mg/kg i.p. nivolumab every 2 weeks plus 1 mg/kg ipilimumab every 6 weeks. FUNDING: This work was funded by Bristol Myers Squibb (CA209-9C7), an MD Anderson Cancer Center Support Grant (CA016672), the Ovarian Cancer Moon Shots Program, the Emerson Collective Fund, and a T32 training grant (CA101642).


Subject(s)
Genital Neoplasms, Female , Ovarian Neoplasms , Peritoneal Neoplasms , Uterine Cervical Neoplasms , Humans , Female , Nivolumab/adverse effects , Ipilimumab/adverse effects , Genital Neoplasms, Female/chemically induced , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/chemically induced , Uterine Cervical Neoplasms/chemically induced , Bayes Theorem , Neoplasm Recurrence, Local/drug therapy , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/chemically induced
2.
Anticancer Drugs ; 35(5): 450-458, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38452059

ABSTRACT

The purpose of this study is to establish the recommended phase 2 dose for regorafenib in combination with sildenafil for patients with advanced solid tumors. Secondary outcomes included identification of antitumor effects of regorafenib and sildenafil, toxicity of the combination, determination of PDE5 expression in tumor samples, and the impact of sildenafil on the pharmacokinetics of regorafenib. This study was a phase 1, open-label single-arm dose-escalation trial using a 3 + 3 design. Additional patients were enrolled at the maximum tolerated dose (MTD) until a total of 12 patients were treated at the MTD. A total of 29 patients were treated in this study. The median duration of treatment was 8 weeks. The recommended phase 2 doses determined in this study are regorafenib 160 mg daily with sildenafil 100 mg daily. The most common toxicities included palmar-plantar erythrodysesthesia syndrome (20 patients, 69%) and hypophosphatemia (18 patients, 62%). Two patients (7%) experienced grade 4 lipase increase. Objective responses were not observed; however, 14 patients (48%) had a period of stable disease during the study. Stable disease for up to 12 months was observed in patients with ovarian cancer as well as up to 20 months for a patient with cervical cancer. The combination of regorafenib and sildenafil at the recommended phase 2 dose is safe and generally well tolerated. Disease control in patients with gynecologic malignancies was especially encouraging. Further evaluation of the combination of regorafenib and sildenafil in gynecologic malignancies is warranted. Clinical Trial Registration Number: NCT02466802.


Subject(s)
Genital Neoplasms, Female , Neoplasms , Adult , Female , Humans , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Genital Neoplasms, Female/chemically induced , Genital Neoplasms, Female/drug therapy , Maximum Tolerated Dose , Neoplasms/drug therapy , Neoplasms/pathology , Phenylurea Compounds/adverse effects , Pyridines/therapeutic use , Sildenafil Citrate/adverse effects
3.
J Formos Med Assoc ; 123(4): 487-495, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37852875

ABSTRACT

OBJECTIVE: The approved standard dose of pembrolizumab (200 mg administrated every 3 weeks) for cancer treatment imposes a significant financial burden on patients. However, no study has analyzed the clinical outcomes of low-dose pembrolizumab among individuals diagnosed with gynecologic cancer. The primary objective of this study was to assess the effectiveness and safety of a low-dose pembrolizumab regimen in real-world clinical practice. METHODS: We retrospectively assessed the efficacy and safety data of patients with gynecologic malignancies who received pembrolizumab between 2017 and 2022 at Kaohsiung Chang Gung Memorial Hospital. Furthermore, we conducted a comparative analysis of the objective response rate (ORR) and progression-free survival (PFS) between patients with deficient mismatch repair (dMMR) and proficient MMR (pMMR). RESULTS: A total of thirty-nine patients were included and received pembrolizumab at fixed dosages of 50 mg (5.1%), 100 mg (84.6%) and 200 mg (10.3%) per cycle. Compared to the pMMR group, the dMMR group exhibited a tendency toward improved ORR (45.5% vs. 13.0%, p = 0.074), and notably, the median duration of response remained unreached. There was no significant difference in PFS between the dMMR and pMMR groups; however, the patients with dMMR in tumor tissue had a trend of better survival (p = 0.079). Incidence of immune-related adverse events (irAEs) of any grade was observed in 13 patients (33.3%), with 3 individuals (7.7%) experiencing grade 3 or 4 events. CONCLUSION: Low-dose pembrolizumab may be a cost-effective and safe treatment option without compromising clinical outcomes in patients with refractory gynecologic cancers.


Subject(s)
Genital Neoplasms, Female , Humans , Female , Genital Neoplasms, Female/drug therapy , Genital Neoplasms, Female/genetics , Genital Neoplasms, Female/chemically induced , Retrospective Studies , Antibodies, Monoclonal, Humanized/adverse effects , Progression-Free Survival
4.
Clin Appl Thromb Hemost ; 29: 10760296231178300, 2023.
Article in English | MEDLINE | ID: mdl-37231620

ABSTRACT

The aim of this study was to determine which type of prophylaxis was effective for postoperative symptomatic venous thromboembolism (VTE) in patients with gynecological malignancies. A total of 1756 consecutive patients undergoing laparotomy as first-line treatment were included. In Period 1 (2004-2009), low-molecular weight heparin (LMWH) was not available for postoperative VTE prophylaxis, but available in after Period 2 (2009-2013). In Period 3 (2013-2020), patients with pretreatment VTE could switch from LMWH to direct oral anticoagulant (DOAC) as of 2015. Preoperative VTE was screened by measuring D-dimer, followed by venous ultrasound imaging, and computed tomography and/or perfusion lung scintigraphy. Postoperative symptomatic VTE occurred with an incidence of 2.8% by the measures without prophylactic LMWH administration in Period 1. The incidence of postoperative symptomatic VTE was 0.6% in Period 2 and 0.3% in Period 3, being significantly reduced compared with Period 1 (P < .01 and < .0001). The incidences were not significantly different between Periods 2 and 3, but no patient switching to DOAC in Period 3 (n = 79) developed symptomatic VTE. Our preoperative VTE screening and postoperative selective LMWH administration were significantly preventive against postoperative symptomatic VTE.


Subject(s)
Genital Neoplasms, Female , Venous Thromboembolism , Female , Humans , Heparin, Low-Molecular-Weight , Venous Thromboembolism/etiology , Venous Thromboembolism/prevention & control , Venous Thromboembolism/epidemiology , Genital Neoplasms, Female/surgery , Genital Neoplasms, Female/chemically induced , Genital Neoplasms, Female/complications , Anticoagulants
5.
Anticancer Drugs ; 34(7): 837-843, 2023 08 01.
Article in English | MEDLINE | ID: mdl-36730297

ABSTRACT

Nab-PTX is a special dosage form of antitumor drug that is different from other injections. In order to explore the efficacy and safety of albumin-bound paclitaxel, we developed an analytical method with UPLC-MS/MS to quantify the total and free paclitaxel in plasma, and prospectively evaluate the impact of unbound fraction fu (%) on the prognosis and adverse reactions of patients with gynecological tumors. From 2020.10 to 2021.10, a total of 116 patients with gynecological tumors were included, application of albumin-bound paclitaxel combined with platinum chemotherapy drugs, the blood collection time is 18-30 h after nab-PTX intravenous infusion. The collection time and the start (end) time of intravenous drip are recorded correctly, and a high-precision and sensitive UPLC-MS/MS method for the simultaneous determination of total and free paclitaxel was established. With fu (%) = Cunbound/Ctotal as the evaluation index, the concentration of total paclitaxel and free paclitaxel were determined by UPLC-MS/MS. The value of fu (%) was closely related to clinical adverse reactions, neutropenia, thrombocytopenia, leukopenia and bone marrow suppression. Neurotoxicity was statistically remarkable ( P up0.001), and fu (%) has a significant correlation with clinical efficacy ( P up0.001). We have developed a highly precise, highly sensitive and specific UPLC-MS/MS method for the simultaneous determination of binding and free albumin-bound paclitaxel concentrations in patients' serum. In addition, we found that fu (%) could be used as the detection index. The higher the fu (%) was, the more taxol could be free, the more adverse reactions related to toxic events occurred in patients.


Subject(s)
Albumin-Bound Paclitaxel , Genital Neoplasms, Female , Female , Humans , Albumin-Bound Paclitaxel/adverse effects , East Asian People , Chromatography, Liquid , Genital Neoplasms, Female/drug therapy , Genital Neoplasms, Female/chemically induced , Tandem Mass Spectrometry , Paclitaxel , Albumins/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects
6.
Anesth Analg ; 137(3): 525-533, 2023 09 01.
Article in English | MEDLINE | ID: mdl-36727850

ABSTRACT

BACKGROUND: Surgery for gynecologic malignancy via midline-laparotomy leads to severe postoperative pain. Adequate pain control while sparing opioid consumption does offer benefits in postoperative complications and recovery. Intrathecal morphine (ITM) provides simple and effective analgesia. In this randomized trial, we compared postoperative opioid consumption in patients who received either ITM or a sham procedure. METHODS: We enrolled 68 adult patients undergoing open gynecologic oncology surgery from June 2021 to November 2021. They were randomly allocated to the ITM group (ITM; 200 µg injection) or sham group (sham procedure) to achieve a final 1:1 ratio between groups. We compared opioid consumption and pain severity during 72 hours after surgery. The variables regarding postoperative recovery and patient-centered outcomes were collected. The primary outcome is cumulative intravenous (IV) opioid consumption 24 hours after surgery. RESULTS: The median (interquartile range) cumulative IV opioid consumption during 24 hours after surgery was 18 mg (12-29) in the ITM group and 36 mg (27-42) in the sham group (median difference, 13; 95% confidence interval, 7.2-20.7; P < .001). Patient satisfaction regarding pain control was statistically significantly higher in the ITM group than in the sham group at postoperative 24 and 48 hours ( P < .001 and P = .005, respectively). There were no significant differences in the variables associated with postoperative recovery and frequency of complications requiring treatment. CONCLUSIONS: ITM is a safe and effective analgesic method after curative intent laparotomy for gynecologic malignancy. ITM provides better pain relief, reduces opioid consumption, and improves patient satisfaction without additional evident adverse events.


Subject(s)
Analgesics, Opioid , Genital Neoplasms, Female , Adult , Humans , Female , Morphine , Genital Neoplasms, Female/surgery , Genital Neoplasms, Female/chemically induced , Genital Neoplasms, Female/drug therapy , Injections, Spinal , Pain, Postoperative/diagnosis , Pain, Postoperative/drug therapy , Pain, Postoperative/etiology
7.
Asian Pac J Allergy Immunol ; 41(4): 340-346, 2023 Dec.
Article in English | MEDLINE | ID: mdl-33068367

ABSTRACT

BACKGROUND: Immediate hypersensitivity reactions (IHRs) are commonly found in patients receiving paclitaxel. Effects of paclitaxel vary because of variable co-therapy or re-challenge with paclitaxel. OBJECTIVE: Our objective was to investigate the incidence, patterns, and risk factors for paclitaxel-related IHRs and management of IHRs in gynecologic malignancy patients. METHODS: This retrospective study was performed in gynecologic cancer patients receiving paclitaxel-based regimens at Siriraj hospital from January 2012 to December 2017. RESULTS: 416 subjects were included and received ranitidine 50 mg, dexamethasone 20 mg, ondansetron 16 mg intravenously and diphenhydramine 50 mg orally 30 minutes before starting chemotherapy. The incidence of IHRs was 17.79%. IHRs occurring on first exposure to paclitaxel was 81.1% and occurred within 30 minutes after starting paclitaxel. The most commonly found presentation of IHRs were skin reactions (86.5%). In multivariate analysis, age < 54.5 years, stage of cancer < 2, and leukocyte cell count < 7.735 × 109/L were significantly associated with IHRs. Seventy-two out of 74 patients that recovered from IHRs were reintroduced paclitaxel. Forty-seven patients (97.92%) of 48 patients with mild reactions were successfully reintroduced to paclitaxel after treatment with chlorpheniramine or other interventions. CONCLUSIONS: The incidence of paclitaxel-related IHRs was about one in five. Skin reactions were the most commonly occurring reactions. Younger age, stage of cancer < 2, and leukocytes < 7.735 × 109/L were significant risk factors for IHRs. Patients with IHRs recovered without the use of dexamethasone and antihistamines before the reintroduction of paclitaxel.


Subject(s)
Drug Hypersensitivity , Genital Neoplasms, Female , Hypersensitivity, Immediate , Humans , Female , Middle Aged , Paclitaxel/adverse effects , Dexamethasone/adverse effects , Genital Neoplasms, Female/drug therapy , Genital Neoplasms, Female/chemically induced , Genital Neoplasms, Female/complications , Retrospective Studies , Drug Hypersensitivity/diagnosis , Drug Hypersensitivity/epidemiology , Drug Hypersensitivity/etiology , Hypersensitivity, Immediate/complications
8.
Eur J Obstet Gynecol Reprod Biol ; 268: 74-81, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34875557

ABSTRACT

BACKGROUND: Recent evidence has demonstrated that the salutary effect of statins on the prevention and prognosis of cancers, including gynecologic cancers. However, due to the heterogeneity of tumors, the results from related studies regarding the association between statin therapy and gynecologic cancers are conflicting. Thus, we conducted this meta-analysis to better understand the relationship between statins use and gynecologic cancers. METHODS: We searched for articles published before July 2021 in the databases: PubMed, Web of Science, Medline, EMBASE and Google Scholar. We computed odds ratio (OR)/relative risk (RR) or hazard ratio (HR) and 95% confidence intervals (CI) regarding the association between statin use and the risk or prognosis of gynecologic cancers by using STATA 12.0 software. RESULTS: The present meta-analysis showed that statin use was associated with a lower risk of gynecologic cancer (OR/RR = 0.89, 95% CI 0.83 to 0.96, I2 = 60.6%, p < 0.001). Statin use was associated with lower risks of endometrial cancer and ovarian cancer (endometrial cancer: OR/RR = 0.81, 95% CI 0.70 to 0.94, I2 = 62.3%, p = 0.001; ovarian cancer: OR/RR = 0.92, 95% CI 0.85 to 1.00, I2 = 42.1%, p = 0.077). The present meta-analysis showed that statin use was associated with a lower mortality of gynecologic cancer (HR = 0.73, 95% CI 0.67 to 0.80, I2 = 39.0%, p = 0.03). Statin use was associated with lower mortalities of endometrial cancer and ovarian cancer (endometrial cancer: HR = 0.71, 95% CI 0.64 to 0.80, I2 = 31.9%, p = 0.144; ovarian cancer: HR = 0.78, 95% CI 0.73 to 0.83, I2 = 43.9%, p = 0.051). CONCLUSION: Statins use was inversely associated with the risk and mortality of gynecologic cancers. Meanwhile, we need more well-designed and high-quality studies with strong evidence for definite conclusions that determine clinical practice.


Subject(s)
Endometrial Neoplasms , Genital Neoplasms, Female , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Ovarian Neoplasms , Female , Genital Neoplasms, Female/chemically induced , Genital Neoplasms, Female/epidemiology , Genital Neoplasms, Female/prevention & control , Humans , Ovarian Neoplasms/epidemiology , Ovarian Neoplasms/prevention & control , Prognosis
9.
BMB Rep ; 51(11): 557-562, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29921411

ABSTRACT

Reactive oxygen species (ROS) are major sources of cellular oxidative stress. Specifically, cancer cells harbor genetic alterations that promote a continuous and elevated production of ROS. While such oxidative stress conditions could be harmful to normal cells, they facilitate cancer cell growth in multiple ways by causing DNA damage and genomic instability, and ultimately by reprogramming cancer cell metabolism. This review provides up to date findings regarding the roles of ROS generation induced by diverse biological molecules and chemicals in representative women's cancer. Specifically, we describe the cellular signaling pathways that regulate direct or indirect interactions between ROS homeostasis and metabolism within female genital cancer cells. [BMB Reports 2018; 51(11): 557-562].


Subject(s)
Carcinogenesis/chemically induced , Carcinogens/pharmacology , Genital Neoplasms, Female/chemically induced , Reactive Oxygen Species/pharmacology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinogens/metabolism , Female , Genital Neoplasms, Female/metabolism , Genital Neoplasms, Female/pathology , Genitalia, Female/drug effects , Genitalia, Female/metabolism , Humans , Oxidation-Reduction/drug effects , Oxidative Stress/physiology , Pregnancy , Reactive Oxygen Species/metabolism
10.
J Steroid Biochem Mol Biol ; 176: 38-48, 2018 02.
Article in English | MEDLINE | ID: mdl-28595943

ABSTRACT

Mechanisms of carcinogenesis by estrogen center on its mitogenic and genotoxic potential on tumor target cells. These models suggest that estrogen receptor (ER) signaling promotes expansion of the transformed population and that subsequent accumulation of somatic mutations that drive cancer progression occur via metabolic activation of cathecol estrogens or by epigenetic mechanisms. Recent findings that GPER is linked to obesity, vascular pathology and immunosuppression, key events in the development of metabolic syndrome and intra-tissular estrogen synthesis, provides an alternate view of estrogen-induced carcinogenesis. Consistent with this concept, GPER is directly associated with clinicopathological indices that predict cancer progression and poor survival in breast and gynecological cancers. Moreover, GPER manifests cell biological responses and a microenvironment conducive for tumor development and cancer progression, regulating cellular responses associated with glandular homeostasis and survival, invading surrounding tissue and attracting a vascular supply. Thus, the cellular actions attributed to GPER fit well with the known molecular mechanisms of G-protein coupled receptors, GPCRs, namely, their ability to transactivate integrins and EGF receptors and alter the interaction between glandular epithelia and their extracellular environment, affecting epithelial-to-mesenchymal transition (EMT) and allowing for tumor cell survival and dissemination. This perspective reviews the molecular and cellular responses manifested by GPER and evaluates its contribution to female reproductive cancers as diseases that progress as a result of dysregulated glandular homeostasis resulting in chronic inflammation and metastasis. This review is organized in sections as follows: I) a brief synopsis of the current state of knowledge regarding estrogen-induced carcinogenesis, II) a review of evidence from clinical and animal-based studies that support a role for GPER in cancer progression, and III) a mechanistic framework describing how GPER-mediated estrogen action may influence the tumor and its microenvironment.


Subject(s)
Carcinogenesis/pathology , Estrogens/toxicity , Genital Neoplasms, Female/pathology , Neoplasms, Glandular and Epithelial/secondary , Receptors, G-Protein-Coupled/metabolism , Animals , Carcinogenesis/chemically induced , Carcinogenesis/metabolism , Female , Genital Neoplasms, Female/chemically induced , Genital Neoplasms, Female/metabolism , Genital Neoplasms, Female/mortality , Humans , Neoplasms, Glandular and Epithelial/chemically induced , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/mortality
11.
J Dermatol Sci ; 87(2): 116-122, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28416366

ABSTRACT

BACKGROUND: Crisaborole is a novel, topical nonsteroidal, anti-inflammatory, phosphodiesterase 4 (PDE4) inhibitor for the treatment of mild to moderate atopic dermatitis. OBJECTIVE: As part of a nonclinical safety testing program, these 2-year studies tested the carcinogenic potential of crisaborole. METHODS: Crisaborole ointment, 2%, 5%, or 7%, was applied once daily topically to mice, and crisaborole was administered orally to rats at doses of 30, 100, or 300mg/kg/day for up to 104 weeks. Systemic exposure to crisaborole and its metabolites, moribundity/death, clinical signs, and tumor formation were assessed in each study. RESULTS: Crisaborole treatment was not tumorigenic in mice at any of the doses administered and did not increase the incidence of neoplastic or nonneoplastic microscopic lesions compared with controls. Oral administration of crisaborole at the high dose (300mg/kg/day) to female rats increased the incidence of treatment-related benign granular cell tumors in the distal reproductive tract (uterus with cervix and vagina) but did not cause moribundity/death. CONCLUSION: Crisaborole was well tolerated and not tumorigenic in mice. It was not tumorigenic in male rats at 300mg/kg/day at exposures that were 3× the human area under the concentration-time curve (AUC24) and was nontumorigenic in female rats at 100mg/kg/day at exposures that were 1× the human AUC24.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Boron Compounds/adverse effects , Bridged Bicyclo Compounds, Heterocyclic/adverse effects , Carcinogenesis/chemically induced , Genital Neoplasms, Female/epidemiology , Phosphodiesterase 4 Inhibitors/adverse effects , Administration, Cutaneous , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Carcinogenicity Tests , Dermatitis, Atopic/drug therapy , Drug Evaluation, Preclinical , Female , Genital Neoplasms, Female/chemically induced , Humans , Incidence , Male , Mice , Ointments/adverse effects , Phosphodiesterase 4 Inhibitors/administration & dosage , Rats
12.
Fertil Steril ; 106(4): 795-819, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27568524

ABSTRACT

This systematic review identified 45 original published research articles related to oil and gas extraction activities and human reproductive endpoints. Reproductive outcomes were categorized as [1] birth outcomes associated with maternal exposure, [2] semen quality, fertility, and birth outcomes associated with adult paternal exposure, [3] reproductive cancers, and [4] disruption of human sex steroid hormone receptors. The results indicate there is moderate evidence for an increased risk of preterm birth, miscarriage, birth defects, decreased semen quality, and prostate cancer. The quality of the evidence is low and/or inadequate for stillbirth, sex ratio, and birth outcomes associated with paternal exposure, and testicular cancer, female reproductive tract cancers, and breast cancer, and the evidence is inconsistent for an increased risk of low birth weight; therefore, no conclusions can be drawn for these health effects. There is ample evidence for disruption of the estrogen, androgen, and progesterone receptors by oil and gas chemicals, which provides a mechanistic rationale for how exposure to oil and gas activities may increase the health risks we have outlined. The results from this systematic review suggest there is a negative impact on human reproduction from exposure to oil and gas activities. Many of the 45 studies reviewed identified potential human health effects. Most of these studies focused on conventional oil and gas activities. Few studies have been conducted to evaluate the impact of unconventional oil and gas operations on human health. The impact of unconventional oil and gas activities may be greater than that of conventional activity, given that unconventional activities employ many of the same approaches and use dozens of known endocrine-disrupting chemicals in hydraulic fracturing.


Subject(s)
Endocrine Disruptors/adverse effects , Environmental Exposure/adverse effects , Natural Gas/adverse effects , Oil and Gas Fields , Oil and Gas Industry , Petroleum/adverse effects , Reproduction/drug effects , Abnormalities, Drug-Induced/etiology , Cell Line, Tumor , Female , Fertility/drug effects , Genital Neoplasms, Female/chemically induced , Genital Neoplasms, Male/chemically induced , Humans , Hydraulic Fracking , Infertility/chemically induced , Infertility/physiopathology , Male , Maternal Exposure/adverse effects , Paternal Exposure/adverse effects , Pregnancy , Pregnancy Complications/chemically induced , Prenatal Exposure Delayed Effects , Receptors, Steroid/drug effects , Receptors, Steroid/metabolism , Risk Assessment , Risk Factors , Spermatozoa/drug effects , Spermatozoa/pathology
13.
Med Hypotheses ; 92: 84-7, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27241264

ABSTRACT

The effects of the natural and synthetic estrogens have been studied for a long time but the data regarding estrogen related chemicals (endocrine disrupting chemicals, EDCs) and their effects on reproductive system are scarce. EDCs are hormone like agents that are readily present in the environment, which may alter the endocrine system of humans and animals. Approximately 800 chemicals are known or suspected to have the potential to function as EDC. Potential role of EDCs on reproductive disease has gained attention in medical literature in recent years. We hypothesize that exposure to low doses of EDCs in a chronic manner could cause hormone dependent genital cancers including ovarian and endometrial cancer. Long term exposure to low concentrations of EDCs may exert potentiation effect with each other and even with endogenous estrogens and could inhibit enzymes responsible for estrogen metabolism. Exposure time to these EDCs is essential as we have seen from Diethylstilbestrol experience. Dose-response curves of EDCs are also unpredictable. Hence mode of action of EDCs are more complex than previously thought. In the light of these controversies lower doses of EDCs in long term exposure is not harmless. Possibility of this relationship and this hypothesis merit further investigation especially through in vivo studies that could better show the realistic environmental exposure. With the confirmation of our hypothesis, possible EDCs could be identified and eliminated from general use as a public health measure.


Subject(s)
Endocrine Disruptors/chemistry , Endocrine System/drug effects , Environmental Pollutants/adverse effects , Genital Neoplasms, Female/chemically induced , Animals , Carcinoma/chemically induced , Diethylstilbestrol/chemistry , Endometrial Neoplasms/chemically induced , Environmental Exposure , Environmental Pollutants/chemistry , Estrogens/chemistry , Female , Humans , Ligands , Models, Theoretical , Ovarian Neoplasms/chemically induced
14.
Z Rheumatol ; 75(1): 63-7, 2016 Feb.
Article in German | MEDLINE | ID: mdl-26813689

ABSTRACT

The risk of gynecological cancers in patients with inflammatory rheumatic diseases only seems to be elevated with respect to cervical cancer and mainly in patients with systemic lupus erythematosus. There is increasing evidence for an influence of the immune system on tumor control of gynecological malignancies; however, an adverse influence of immunosuppressive treatment in rheumatic patients was indicated only for the risk of cervical cancer. In contrast, biologics could not be shown to cause an increased risk of cervical cancer but data on this topic are limited. General screening recommendations exist for breast cancer and cervical cancer. Recommendations for follow-up after oncological treatment are presented. Because of limited evidence immunosuppressive and biological treatment should be applied with great restraint at least within the first 5 years after curative oncological treatment also for gynecological tumors. As far as breast cancer is concerned an even longer interval is under discussion.


Subject(s)
Antirheumatic Agents/administration & dosage , Antirheumatic Agents/adverse effects , Genital Neoplasms, Female/chemically induced , Genital Neoplasms, Female/prevention & control , Lupus Erythematosus, Systemic/drug therapy , Practice Guidelines as Topic , Dose-Response Relationship, Drug , Drug Administration Schedule , Europe , Evidence-Based Medicine , Female , Germany , Humans , Lupus Erythematosus, Systemic/complications , Treatment Outcome
15.
Reprod Toxicol ; 59: 167-82, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26493093

ABSTRACT

The estrogenic properties of bisphenol A (BPA), a ubiquitous synthetic monomer that can leach into the food and water supply, have prompted considerable research into exposure-associated health risks in humans. Endocrine-disrupting properties of BPA suggest it may impact developmental plasticity during early life, predisposing individuals to disease at doses below the oral reference dose (RfD) established by the Environmental Protection Agency in 1982. Herein, we review the current in vivo literature evaluating the carcinogenic properties of BPA. We conclude that there is substantial evidence from rodent studies indicating that early-life BPA exposures below the RfD lead to increased susceptibility to mammary and prostate cancer. Based on the definitions of "carcinogen" put forth by the International Agency for Research on Cancer and the National Toxicology Program, we propose that BPA may be reasonably anticipated to be a human carcinogen in the breast and prostate due to its tumor promoting properties.


Subject(s)
Benzhydryl Compounds/toxicity , Breast Neoplasms/chemically induced , Cell Transformation, Neoplastic/chemically induced , Endocrine Disruptors/toxicity , Environmental Pollutants/toxicity , Genital Neoplasms, Female/chemically induced , Genital Neoplasms, Male/chemically induced , Phenols/toxicity , Animals , Breast Neoplasms/epidemiology , Female , Genital Neoplasms, Female/epidemiology , Genital Neoplasms, Male/epidemiology , Humans , Male , Risk Assessment , Risk Factors
16.
Eur J Public Health ; 26(1): 76-83, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26330492

ABSTRACT

BACKGROUND: Worrying trends regarding human reproductive endpoints (e.g. semen quality, reproductive cancers) have been reported and there is growing circumstantial evidence for a possible causal link between these trends and exposure to endocrine disrupting chemicals (EDCs). However, there is a striking lack of human data to fill the current knowledge gaps. To answer the crucial questions raised on human reproductive health, there is an urgent need for a reproductive surveillance system to be shared across countries. METHODS: A multidisciplinary network named HUman Reproductive health and Global ENvironment Network (HURGENT) was created aiming at designing a European monitoring system for reproductive health indicators. Collaborative work allowed setting up the available knowledge to design such a system. Furthermore we conducted an overview of 23 potential indicators, based upon a weight of evidence (WoE) approach according to their potential relation with EDC exposure. RESULTS: The framework and purposes of the surveillance system are settled as well as the approach to select suitable reproductive indicators. The indicators found with the highest scores according to the WoE approach are prostate and breast cancer incidence, sex ratio, endometriosis and uterine fibroid incidence, indicators related to the testicular dysgenesis syndrome, precocious puberty incidence and reproductive hormone levels. CONCLUSION: Not only sentinel health endpoints, but also diseases with high burdens in public health are highlighted as prior indicators in the context of EDC exposure. Our work can serve as a basis to construct, as soon as possible, the first multi-country reproductive monitoring system.


Subject(s)
Endocrine Disruptors/toxicity , Environmental Exposure/adverse effects , Environmental Pollutants/toxicity , Public Health Surveillance/methods , Reproductive Health/statistics & numerical data , Breast Neoplasms/chemically induced , Female , Genital Neoplasms, Female/chemically induced , Gonadal Disorders/chemically induced , Humans , Incidence , Male , Prostatic Neoplasms/chemically induced
17.
J Cancer Res Clin Oncol ; 142(1): 287-93, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26337160

ABSTRACT

OBJECTIVE: To investigate whether an association exists between a history of fertility treatments and future risk of female malignancies. STUDY DESIGN: A population-based study compared the incidence of long-term female malignancies in a cohort of women with and without a history of fertility treatments including in vitro fertilization (IVF) and ovulation induction (OI). Deliveries occurred between the years 1988-2013, with a mean follow-up duration of 12 years. Excluded from the study were women with known genetic predisposition for malignancies or known malignancies prior to the index pregnancy. Female malignancies were divided into specific types including ovarian, uterine, breast and cervix. Kaplan-Meier survival curve was used to estimate cumulative incidence of malignancies. Cox proportional hazard models were used to estimate the adjusted hazard ratios (HRs) for female malignancy. RESULTS: During the study period, 106,031 women met the inclusion criteria; 4.1 % (n = 4363) occurred in patients following fertility treatments. During the follow-up period, patients with a history of IVF treatments had a significantly increased risk of being diagnosed with ovarian and uterine cancer as compared to patients after OI and patients with no history of fertility treatments. Cox proportional hazard models were constructed for ovarian and uterine cancer separately, controlling for confounders such as maternal age and obesity. A history of IVF treatment remained independently associated with ovarian and uterine cancer (adjusted HR 3.9; 95 % CI 1.2-12.6; P = 0.022 and adjusted HR 4.6; 95 % CI 1.4-14.9; P = 0.011; respectively). CONCLUSION: IVF treatments pose a significant risk of subsequent long-term ovarian and uterine cancer.


Subject(s)
Breast Neoplasms/chemically induced , Fertility Agents, Female/adverse effects , Fertility/drug effects , Fertilization in Vitro/adverse effects , Genital Neoplasms, Female/chemically induced , Hospitalization/statistics & numerical data , Ovulation Induction/adverse effects , Adult , Breast Neoplasms/diagnosis , Breast Neoplasms/epidemiology , Cohort Studies , Female , Follow-Up Studies , Genital Neoplasms, Female/diagnosis , Genital Neoplasms, Female/epidemiology , Humans , Israel/epidemiology , Neoplasm Staging , Pregnancy , Prevalence , Prognosis , Risk Factors , Time Factors
19.
Rev Endocr Metab Disord ; 16(4): 359-64, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26831296

ABSTRACT

Breast and uterine cancer are the most frequent female gender related neoplasms whose growth is mostly estrogen dependent. Therefore, any EDC exhibiting estrogenic effects may increase the risk of these two malignancies. This review focuses on the potential role of EDCs with estrogenic potential on the risk of breast and uterine neoplasms but also points to the possible role of the exposure to EDCs in the pathogenesis of ovarian and cervical cancer. It also underlines the necessity of informing the public about the presence of EDCs in common consumer products, their detrimental health effects and methods of reducing the exposure risk.


Subject(s)
Breast Neoplasms/chemically induced , Endocrine Disruptors/toxicity , Environmental Pollutants/toxicity , Estrogens/toxicity , Genital Neoplasms, Female/chemically induced , Ovarian Neoplasms/chemically induced , Female , Humans
20.
Cancer Epidemiol ; 37(6): 870-5, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24189467

ABSTRACT

Spironolactone, an aldosterone-antagonist, is associated with gynecomastia. Digoxin, which can also cause gynecomastia, has been associated with increased incidence of breast and uterus cancers. We therefore postulated that spironolactone use might also increase these cancer risks. Using a nationwide prescription drug registry between 1995 and 2010, we identified use of spironolactone in a cohort of Danish women (≥20 years old). In users and non-users, incidence rate ratios adjusted by age group and calendar-year examined risk of breast and uterus cancers, both estrogen-sensitive, and ovary and cervix cancers, both relatively estrogen-insensitive. As an added control exposure, risk ratios in women who used another diuretic, furosemide, were examined by the same approach. Among 2.3 million women (28.5 million person-years), risks of breast, uterus, ovary, and cervix cancers were generally increased about 10-30% in both spironolactone and furosemide users. In the first year of drug exposure, incidences were increased, especially for ovary cancers. However, incidence increases in the first year of use were not specific for estrogen-sensitive cancers, occurred with both spironolactone and furosemide, and were driven by exposures immediately prior to diagnosis. For drug exposure ≥1 years before cancer diagnosis, incidences of these cancers were not significantly increased. We conclude that associations observed with first use in the year immediately before cancer diagnosis were driven by reverse causality, i.e., because of treatment for symptoms related to the incipient cancer. With respect to breast, uterus, ovarian and cervical cancer, there is no evidence of increased risk with spironolactone or furosemide use.


Subject(s)
Breast Neoplasms/epidemiology , Genital Neoplasms, Female/epidemiology , Mineralocorticoid Receptor Antagonists/therapeutic use , Spironolactone/therapeutic use , Adult , Aged , Aged, 80 and over , Breast Neoplasms/chemically induced , Breast Neoplasms/diagnosis , Cohort Studies , Denmark/epidemiology , Drug Prescriptions/statistics & numerical data , Female , Follow-Up Studies , Genital Neoplasms, Female/chemically induced , Genital Neoplasms, Female/diagnosis , Humans , Middle Aged , Mineralocorticoid Receptor Antagonists/adverse effects , Prognosis , Risk Factors , Spironolactone/adverse effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...