Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
BMC Med ; 20(1): 154, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35534877

ABSTRACT

BACKGROUND: Thymic epithelial tumors (TETs) are rare malignancies and the treatment options are limited. We aimed to evaluate the efficacy and safety of apatinib, an angiogenesis inhibitor, in advanced TETs. METHODS: This was an open-label, single-arm, phase II trial at three centers in China. Patients with TET who had progressed after failure of at least one line of platinum-based chemotherapy were enrolled. Patients received apatinib 500 mg orally per day. The primary endpoint was objective response rate (ORR). Secondary endpoints were progression-free survival (PFS), overall survival (OS), disease control rate (DCR), and safety. RESULTS: From June 29, 2017, to April 18, 2019, 25 patients were enrolled. At data cut off (September 30, 2021), one patient achieved complete response, nine achieved partial response, and 11 achieved stable disease, with an ORR of 40% (95% CI 21-61%) and DCR of 84% (95% CI 64-95%). The median PFS was 9.0 (95% CI 5.4-12.6) months. The median OS was 24.0 (95% CI 8.2-39.8) months. All patients reported treatment-related adverse events (TRAEs). Grade 3 TRAEs occurred 26 times in 15 patients. No grade 4 or 5 toxicities occurred. CONCLUSIONS: This is the first trial of apatinib for the treatment of TETs. Apatinib showed promising antitumor activity and the toxicities were tolerable and manageable.


Subject(s)
Antineoplastic Agents , Neoplasms, Glandular and Epithelial , Antineoplastic Agents/adverse effects , Humans , Neoplasms, Glandular and Epithelial/chemically induced , Neoplasms, Glandular and Epithelial/drug therapy , Pyridines/adverse effects , Thymus Neoplasms
2.
Invest New Drugs ; 40(3): 586-595, 2022 06.
Article in English | MEDLINE | ID: mdl-35113285

ABSTRACT

PURPOSE: Sym013 contains six humanized monoclonal antibodies that bind to non-overlapping epitopes on three human epidermal growth factor receptors (HER1-3). Preclinical studies suggested Sym013 strongly suppresses growth of multiple epithelial tumors. This is a first-in-human study exploring safety and efficacy of Sym013 in patients with advanced epithelial malignancies. METHODS: Dose escalation used single-patient cohorts until the stopping rule was met, followed by 3 + 3 design. Dose levels planned were: 1, 2, 4, 6, 9, 12, 15, and 18 mg/kg. Treatment cycles were 28 days with imaging every eight weeks. Serum samples were collected at multiple time points for assessment of pharmacokinetics and development of anti-drug antibodies. RESULTS: Thirty-two patients were enrolled with multiple solid tumors, most common being colorectal cancer (CRC; 10/32, 31%). Due to mucositis, rash, and diarrhea at 4 mg/kg once-weekly, dosing was changed to biweekly (Q2W). Mandatory prophylaxis was added due to Grade 3 infusion-related reaction and oral mucositis at 9 mg/kg Q2W. The 15 mg/kg Q2W cohort was enrolling when the study was terminated for business reasons. Most common adverse events were skin (81%) and gastrointestinal (75%) disorders, including dermatitis/rash, stomatitis, and diarrhea. One patient with CRC achieved a partial response; 12 patients with varied malignancies had stable disease. CONCLUSION: During the conduct of the study, management of frequent infusion-related reactions, skin toxicities, and mucosal disorders, which are indicative of HER inhibition, necessitated multiple protocol amendments. The investigators, in concert with the Sponsor, agreed that achieving a tolerated regimen with acceptable target saturation was unlikely. TRIAL REGISTRY: www. CLINICALTRIALS: gov ; NCT02906670 (September 20, 2016).


Subject(s)
Antineoplastic Agents , Exanthema , Neoplasms, Glandular and Epithelial , Neoplasms , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/adverse effects , Diarrhea/chemically induced , Exanthema/chemically induced , Humans , Maximum Tolerated Dose , Neoplasms/metabolism , Neoplasms, Glandular and Epithelial/chemically induced , Neoplasms, Glandular and Epithelial/drug therapy
4.
J Cancer Res Ther ; 16(1): 170-172, 2020.
Article in English | MEDLINE | ID: mdl-32362631

ABSTRACT

With the discovery of v-Raf murine sarcoma viral oncogene homolog B (BRAF) inhibitors, new treatment possibilities arose against metastatic melanoma. A frequent adverse effect of BRAF inhibitor therapy is the induction of epithelial proliferations such as cutaneous squamous cell carcinoma and verrucous papilloma. Here, we describe a case in which a patient developed extensive anal epithelial proliferations resembling condylomata acuminata, after starting vemurafenib treatment. This adverse effect has rarely been reported in the literature. Interestingly, the lesions in our patient were negative for human papillomavirus, and mutations in BRAF, Neuroblastoma rat sarcoma viral oncogene homolog (NRAS), Kirsten rat sarcoma viral oncogene homolog (KRAS), and Harvey rat sarcoma viral oncogene homolog (HRAS) were not detected. Different pathways can contribute to these epithelial proliferations resembling condylomata acuminata. We show the relevance of a detailed history at the beginning and during treatment, instructions, education, and dermatological follow-up (including the genital area) for patients treated with BRAF inhibitors. Condylomata acuminata can influence the quality of life and are treated, in an early stage, with cryotherapy, coagulation, imiquimod, and/or CO2 laser therapy.


Subject(s)
Anus Neoplasms/diagnosis , Cell Proliferation , Condylomata Acuminata/diagnosis , Melanoma/drug therapy , Neoplasms, Glandular and Epithelial/diagnosis , Skin Neoplasms/drug therapy , Vemurafenib/adverse effects , Antineoplastic Agents/adverse effects , Anus Neoplasms/chemically induced , Anus Neoplasms/genetics , Condylomata Acuminata/chemically induced , Diagnosis, Differential , GTP Phosphohydrolases/genetics , Humans , Male , Melanoma/genetics , Melanoma/pathology , Membrane Proteins/genetics , Middle Aged , Mutation , Neoplasms, Glandular and Epithelial/chemically induced , Neoplasms, Glandular and Epithelial/genetics , Papillomaviridae/isolation & purification , Papillomavirus Infections/virology , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology
5.
Clin Exp Allergy ; 49(10): 1291-1305, 2019 10.
Article in English | MEDLINE | ID: mdl-31295369

ABSTRACT

OBJECTIVE: We investigated whether prolonged treatment with omalizumab influences development or progression of solid epithelial cancer in patients with atopic asthma or chronic idiopathic urticaria. DESIGN: Systematic review and meta-analysis of intervention and observational studies. Randomized controlled trials were assessed for risk of bias using the Cochrane Risk of Bias tool, comparative observational studies were assessed using the Newcastle-Ottawa Scale, and non-comparative observational studies were assessed using the Joanna Briggs Institute Checklist for Prevalence Studies. DATA SOURCES: We searched MEDLINE, EMBASE, Cochrane Library and grey literature for eligible studies to November 2017. All searches were updated in January 2019. ELIGIBILITY CRITERIA FOR INCLUDED STUDIES: Randomized, quasi-randomized, controlled clinical trials and observational studies were included if they involved patients ≥ 12 years with moderate-to-severe persistent asthma or chronic idiopathic urticaria treated with omalizumab for ≥ 40 weeks. Eligible comparators included standard of care, placebo, cromoglycate or no treatment. RESULTS: One hundred and sixty seven unique studies were eligible for inclusion; however, only twelve (7.2%, n = 11 758) reported any outcome of interest, none of which involved patients with urticaria. 195 cancer events were reported. We found no statistically significant increase in the odds of study-emergent solid epithelial cancer in patients randomized to long-term treatment with omalizumab compared to standard of care (Peto OR: 0.65, 95% CI: 0.11, 3.74, I2  = 41%). Less than one per cent of participants of non-comparative observational studies (n = 2350) were diagnosed with a solid epithelial tumour (meta-proportion: 0.86% [95% CI: 0.24, 1.86%, I2  = 56%]). In the only comparative observational study reporting on cancer, the proportion of study-emergent solid epithelial tumour events was nearly identical in both study groups (omalizumab: 2.3%, standard of care: 2.2%). CONCLUSIONS: There is insufficient evidence to determine whether long-term treatment with omalizumab influences development or progression of solid epithelial cancer in these patient populations. PROSPERO registration # CRD 42018082211.


Subject(s)
Asthma , Chronic Urticaria , Neoplasms, Glandular and Epithelial , Neoplasms, Second Primary , Omalizumab , Asthma/drug therapy , Asthma/epidemiology , Chronic Urticaria/drug therapy , Chronic Urticaria/epidemiology , Female , Humans , Male , Neoplasms, Glandular and Epithelial/chemically induced , Neoplasms, Glandular and Epithelial/epidemiology , Neoplasms, Second Primary/chemically induced , Neoplasms, Second Primary/epidemiology , Omalizumab/adverse effects , Omalizumab/therapeutic use , Randomized Controlled Trials as Topic , Time Factors
6.
Drug Chem Toxicol ; 41(3): 330-337, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29281929

ABSTRACT

The present study assessed the protective effect of aspirin against carcinogenicity induced by mitomycin C (MMC) by the test for detection of warts/epithelial tumor clones in Drosophila melanogaster. Larvae were treated with different concentrations of aspirin alone (10, 20 or 40 mg/mL) or aspirin in association with MMC. MMC and ultrapure water were employed as the positive and negative control, respectively. Antioxidant activity was determined using the DPPH method. For performing cytotoxicity assay on HeLa cells, the aspirin concentrations used ranged from 200 mmol/L to 3,125 mmol/L. For assessment of apoptosis and necrosis, cells were incubated for 24 h with complete medium in the absence (control group) or presence of aspirin (12.5 mmol/L and 25 mmol/L). The results obtained in the assessment of the possible carcinogenic effects of aspirin at the three concentrations tested indicate no statistically significant increase in tumor frequency compared to the negative control. The anticarcinogenic activity assessment, where the larvae of D. melanogaster were previously induced to tumor formation by MMC and later treated with aspirin, showed a statistically significant reduction in the number of tumors compared to the positive control. Antioxidant activity across the three aspirin concentrations (10, 20 or 40 mg/mL) ranged from 20.81% to 26.5%. It was observed that aspirin reduced growth viability of HeLa cells in a concentration-dependent manner in comparison with the control. These results indicate that aspirin did not induce tumors in Drosophila and reduced MMC-induced carcinogenicity. The antioxidant activity and apoptosis induction appear to be the main mechanisms involved in reducing the frequency of tumors.


Subject(s)
Aspirin/pharmacology , Mitomycin/toxicity , Neoplasms, Glandular and Epithelial/prevention & control , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Drosophila melanogaster , HeLa Cells , Humans , Neoplasms, Glandular and Epithelial/chemically induced
7.
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
8.
J Ovarian Res ; 10(1): 65, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-28950890

ABSTRACT

BACKGROUND: To investigate dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) for assessing histopathological and molecular biological features in induced rat epithelial ovarian carcinomas (EOCs). METHODS: 7,12-dimethylbenz[A]anthracene (DMBA) was applied to induce EOCs in situ in 46 SD rats. Conventional MRI and DCE-MRI were performed to evaluate the morphology and perfusion features of the tumors, including the time-signal intensity curve (TIC), volume transfer constant (Ktrans), rate constant (Kep), extravascular extracellular space volume ratio (Ve) and initial area under the curve (IAUC). DCE-MRI parameters were correlated with histological grade, microvascular density (MVD), vascular endothelial growth factor (VEGF) and fraction of Ki67-positive cells and the serum level of cancer antigen 125 (CA125). RESULTS: Thirty-five of the 46 rats developed EOCs. DCE-MRI showed type III TIC more frequently than type II (29/35 vs. 6/35, p < 0.001) in EOCs. The two types of TIC of tumors had significant differences in the histological grade, MVD, expression of VEGF and Ki67, and the serum level of CA125 (all p < 0.01). Ktrans, Kep and IAUC values showed significant differences in different histological grades in overall and pairwise comparisons except for IAUC in grade 2 vs. grade 3 (all p < 0.01). There was no significant difference in Ve values among the three grade groups (p > 0.05). Ktrans, Kep and IAUC values were positively correlated with MVD, VEGF and Ki67 expression (all p < 0.01). Ve was not significantly correlated with MVD, VEGF expression, Ki67 expression and the CA125 level (all p > 0.05). CONCLUSIONS: TIC types and perfusion parameters of DCE-MRI can reflect tumor grade, angiogenesis and cell proliferation to some extent, thereby helping treatment planning and predicting prognosis.


Subject(s)
Neoplasms, Glandular and Epithelial/diagnostic imaging , Ovarian Neoplasms/diagnostic imaging , 9,10-Dimethyl-1,2-benzanthracene , Animals , Carcinogens , Carcinoma, Ovarian Epithelial , Contrast Media , Disease Models, Animal , Female , Ki-67 Antigen/metabolism , Magnetic Resonance Imaging , Neoplasms, Glandular and Epithelial/chemically induced , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/chemically induced , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Proteins/metabolism , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/metabolism
9.
Environ Res ; 156: 388-393, 2017 07.
Article in English | MEDLINE | ID: mdl-28395243

ABSTRACT

Ovarian cancer survival varies geographically throughout California. The objective of this study is to determine the impact of living in disadvantaged communities on spatial patterns of survival disparities. Including a bivariate spatial smooth of geographic location within the Cox proportional hazard models is an effective approach for spatial analyses of cancer survival. Women diagnosed with advanced Stage IIIC/IV epithelial ovarian cancer (1996-2006) were identified from the California Cancer Registry. The impact of living in disadvantaged communities, as measured by the California Office of Environmental Health Hazard Assessment cumulative CalEnviroScreen 2.0 score, on geographic disparities in survival was assessed while controlling for age, tumor characteristics, quality of care, and race. Community-level air quality indicators and socioeconomic status (SES) were also independently examined in secondary analyses. The Cox proportional hazard spatial methods are available in the MapGAM package implemented in R. An increase in the community disadvantage from the 5th (less disadvantage) to the 95th percentile (more disadvantage) was significantly associated with poorer ovarian cancer survival (hazard ratio [HR], 1.16; 95% confidence interval [CI], 1.07-1.26). Ozone levels and SES were the most influential indicators on geographic disparities that warrant further investigation. The use of a bivariate smoother of location within the survival model allows for more advanced spatial analyses for exploring potential air quality-related predictors of geographic disparities.


Subject(s)
Air Pollutants/toxicity , Neoplasms, Glandular and Epithelial/mortality , Ovarian Neoplasms/mortality , Ozone/toxicity , Particulate Matter/toxicity , Vehicle Emissions/toxicity , Aged , California/epidemiology , Carcinoma, Ovarian Epithelial , Female , Gasoline/toxicity , Humans , Middle Aged , Neoplasms, Glandular and Epithelial/chemically induced , Ovarian Neoplasms/chemically induced , Proportional Hazards Models , Residence Characteristics , Social Class
10.
Article in English | MEDLINE | ID: mdl-27925846

ABSTRACT

The primary objective of this study is to understand the profiles, sources and cytotoxic effects of atmospheric polycyclic aromatic hydrocarbons (PAHs), which are closely related to urban air contamination and public health, in urban roadside environments. On-road sampling campaigns were conducted from 2014 to 2015 at three urban road sites in Hangzhou, China. Sixteen gaseous and particulate matter (PM) 2.5-bound PAHs were identified and quantified using gas chromatography-mass spectrometry (GC-MS). The total PAH concentrations at the three sites ranged from 750 to1142 ng/m3 and 1050 to 1483 ng/m3 in summer and winter, respectively. Low molecular weight PAHs were the most abundant compounds (77-86%) and primarily existed in gas phase. The concentrations and phase distributions of high molecular weight PAHs were varied at three sites due to the differences in traffic volume, vehicle composition, engine loading, and nearby artificial activity. Diagnostic ratios of the principal mass (m/z,178, 202, 228 and 276) parent PAHs were statistically described to determine the PAH sources to urban roadsides; principal component analysis (PCA) was applied to apportion the sources. The results indicated that high- and low-temperature fuel processes, as well as residential and industrial emissions, were major contributors to roadside PAHs. The cytotoxic potential of the roadside PAHs was evaluated using a human epithelial lung cell line (A549). Cell viability was measured after a direct exposure to PAH extract. The results reflected the profiles of roadside PAHs at the three sites. The cytotoxicity of reference PAHs was evaluated to provide further insights into the cytotoxic potential of PAHs. We found that low molecular weight PAHs, which are less cytotoxic compounds, synergistically promoted the lethal effect of cytotoxic compounds, posing a potential threat to public health.


Subject(s)
Air Pollutants/toxicity , Environmental Monitoring/methods , Neoplasms, Glandular and Epithelial/chemically induced , Particulate Matter/toxicity , Polycyclic Aromatic Hydrocarbons/toxicity , Vehicle Emissions/toxicity , Air Pollutants/analysis , Cell Line, Tumor/drug effects , Cell Survival/drug effects , China , Cities , Gas Chromatography-Mass Spectrometry , Humans , Neoplasms, Glandular and Epithelial/physiopathology , Particulate Matter/analysis , Polycyclic Aromatic Hydrocarbons/analysis , Seasons , Vehicle Emissions/analysis
11.
Cancer Epidemiol Biomarkers Prev ; 25(10): 1411-1417, 2016 10.
Article in English | MEDLINE | ID: mdl-27197282

ABSTRACT

BACKGROUND: Epidemiologic studies indicate increased ovarian cancer risk among women who use genital powder, but this has not been thoroughly investigated in African American (AA) women, a group with a high prevalence of use. We evaluate the relationship between use of genital powder and nongenital powder in invasive epithelial ovarian cancer (EOC). METHODS: Subjects are 584 cases and 745 controls enrolled in the African American Cancer Epidemiology Study (AACES), an ongoing, population-based case-control study of EOC in AA women in 11 geographic locations in the United States. AA controls were frequency matched to cases on residence and age. Logistic regression was used to calculate ORs and 95% confidence intervals (CI) for associations between genital and nongenital powder exposure and EOC risk, controlling for potential confounders. RESULTS: Powder use was common (62.8% of cases and 52.9% of controls). Genital powder was associated with an increased risk of EOC (OR = 1.44; 95% CI, 1.11-1.86) and a dose-response relationship was found for duration of use and number of lifetime applications (P < 0.05). Nongenital use was also associated with EOC risk, particularly among nonserous EOC cases (OR = 2.28; 95% CI, 1.39-3.74). An association between powder use and upper respiratory conditions suggests an enhanced inflammatory response may explain the association between body powder and EOC. CONCLUSIONS: In a study of AA women, body powder use was significantly associated with EOC risk. IMPACT: The results support that body powder is a modifiable risk factor for EOC among AA women. Cancer Epidemiol Biomarkers Prev; 25(10); 1411-7. ©2016 AACRSee related commentary by Trabert, p. 1369.


Subject(s)
Neoplasms, Glandular and Epithelial/chemically induced , Neoplasms, Glandular and Epithelial/epidemiology , Ovarian Neoplasms/chemically induced , Ovarian Neoplasms/epidemiology , Powders/adverse effects , Adult , Black or African American , Aged , Carcinoma, Ovarian Epithelial , Case-Control Studies , Female , Humans , Middle Aged , Odds Ratio , Young Adult
12.
Anticancer Res ; 36(1): 111-20, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26722034

ABSTRACT

A single intraperitoneal injection of 50 or 75 mg/kg N-methyl-N-nitrosourea in male Sprague-Dawley rats at 4 weeks of age, dose-dependently resulted in cutaneous epithelial cysts and tumors of pilosebaceous origin. Cysts were composed of epidermal cysts or mixed epidermal and inner root sheath hybrid cysts. The majority of induced tumors were keratoacanthomas. A few tumors were trichofolliculomas, trichoblastomas, pilomatricomas, or sebaceous adenomas. All tumors were benign pilosebaceous tumors. Keratoacanthomas were crater-shaped tumors with thick infoldings of epithelium containing keratohyalin granules (epidermal lip) that abruptly changed to epithelium containing trichohyalin granules. The morphological similarity and resemblance of keratin 1, 10, and 14 profiles, and p63 and ß-catenin expression between mixed epidermal and inner root sheath hybrid cysts and keratoacanthomas suggests that hybrid cysts progressed to keratoacanthomas, and the cells from infundibular cells to inner root sheath cells of the pilar segment seem to be the origin of rat keratoacanthomas. Immunohistochemical localization of keratins 1, 10 and 14, p63, and ß-catenin in trichofolliculoma, trichoblastoma, and pilomatricoma, as well as keratoacanthoma, may indicate tumor histogenesis.


Subject(s)
Epidermal Cyst/chemically induced , Keratoacanthoma/chemically induced , Methylnitrosourea , Neoplasms, Glandular and Epithelial/chemically induced , Skin Neoplasms/chemically induced , Skin/drug effects , Animals , Biomarkers, Tumor/metabolism , Disease Models, Animal , Epidermal Cyst/metabolism , Epidermal Cyst/pathology , Humans , Immunohistochemistry , Keratoacanthoma/metabolism , Keratoacanthoma/pathology , Male , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Rats, Sprague-Dawley , Skin/metabolism , Skin/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
14.
Int J Cancer ; 137(1): 173-82, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25430487

ABSTRACT

Nitrate and nitrite are precursors in the endogenous formation of N-nitroso compounds (NOC), potential human carcinogens. We evaluated the association of nitrate and nitrite ingestion with postmenopausal ovarian cancer risk in the Iowa Women's Health Study. Among 28,555 postmenopausal women, we identified 315 incident epithelial ovarian cancers from 1986 to 2010. Dietary nitrate and nitrite intakes were assessed at baseline using food frequency questionnaire data. Drinking water source at home was obtained in a 1989 follow-up survey. Nitrate-nitrogen (NO3 -N) and total trihalomethane (TTHM) levels for Iowa public water utilities were linked to residences and average levels were computed based on each woman's duration at the residence. We computed multivariable-adjusted hazard ratios (HR) and 95% confidence intervals (CI) using Cox proportional hazards regression. We tested interactions of nitrate with TTHMs and dietary factors known to influence NOC formation. Ovarian cancer risk was 2.03 times higher (CI = 1.22-3.38, ptrend = 0.003) in the highest quartile (≥2.98 mg/L) compared with the lowest quartile (≤0.47 mg/L; reference) of NO3 -N in public water, regardless of TTHM levels. Risk among private well users was also elevated (HR = 1.53, CI = 0.93-2.54) compared with the same reference group. Associations were stronger when vitamin C intake was

Subject(s)
Neoplasms, Glandular and Epithelial/chemically induced , Nitrates/adverse effects , Nitrites/toxicity , Ovarian Neoplasms/chemically induced , Water Pollutants, Chemical/toxicity , Aged , Carcinoma, Ovarian Epithelial , Diet , Female , Humans , Iowa/epidemiology , Middle Aged , Neoplasms, Glandular and Epithelial/epidemiology , Ovarian Neoplasms/epidemiology , Postmenopause , Risk Factors , Trihalomethanes/adverse effects
15.
Maturitas ; 76(1): 81-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23871271

ABSTRACT

OBJECTIVE: To evaluate the clinical safety of bazedoxifene (BZA) on the reproductive tract in postmenopausal women with osteoporosis over 7 years. STUDY DESIGN: This was a second, blinded, 2-year extension of a 3-year, randomized, double-blind, placebo (PBO)- and active-controlled phase 3 trial. In the core study, subjects were randomized to receive BZA 20 or 40mg, raloxifene 60mg, or PBO. During years 4-5, the raloxifene arm was discontinued and subjects receiving BZA 40mg were transitioned to BZA 20mg. Subjects continued to receive BZA 20mg or PBO during years 6-7. MAIN OUTCOME MEASURES: The primary endpoint was the incidence of new vertebral fractures at 7 years (reported separately). Reproductive tract safety findings at 7 years are reported here. Endometrial thickness was assessed by transvaginal ultrasonography for subjects in the endometrial safety substudy. Adverse events (AEs) were recorded throughout the study. RESULTS: At 7 years, the adjusted mean (±standard error) change in endometrial thickness was similar with BZA and PBO (-0.11 ± 0.21 and 0.07 ± 0.32 mm, respectively). The incidence of endometrial hyperplasia was low (0.1% for both groups). BZA showed significantly lower rates than PBO of endometrial carcinoma (0.1% vs. 0.4%; P=0.020) and vaginitis (6.1% vs. 7.6%; P=0.035). There were more cases of ovarian carcinoma with BZA (n=4 [0.1%]) than PBO (n=0); the difference was not statistically significant. Rates of breast-related and other gynecologic AEs were similar among groups. CONCLUSIONS: BZA was associated with a favorable reproductive safety profile in postmenopausal women with osteoporosis over 7 years.


Subject(s)
Bone Density Conservation Agents/adverse effects , Endometrium/drug effects , Indoles/adverse effects , Osteoporosis, Postmenopausal/drug therapy , Selective Estrogen Receptor Modulators/adverse effects , Aged , Bone Density Conservation Agents/therapeutic use , Carcinoma, Ovarian Epithelial , Double-Blind Method , Endometrial Hyperplasia/epidemiology , Endometrial Neoplasms/epidemiology , Endometrium/diagnostic imaging , Female , Humans , Incidence , Indoles/therapeutic use , Middle Aged , Neoplasms, Glandular and Epithelial/chemically induced , Neoplasms, Glandular and Epithelial/epidemiology , Ovarian Neoplasms/chemically induced , Ovarian Neoplasms/epidemiology , Postmenopause , Selective Estrogen Receptor Modulators/therapeutic use , Spinal Fractures/prevention & control , Ultrasonography , Vaginitis/epidemiology
16.
Curr Drug Metab ; 14(6): 695-705, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23701164

ABSTRACT

Aristolochic acid (AA), a plant nephrotoxin and carcinogen, causes aristolochic acid nephropathy (AAN) and its associated urothelial malignancy, and is hypothesized to be responsible for Balkan endemic nephropathy (BEN). The major component of AA, aristolochic acid I (AAI), is the predominant compound responsible for these diseases. The reductive activation of AAI leads to the formation of covalent DNA adducts. The most abundant DNA adduct, 7-(deoxyadenosin-N6-yl)aristolactam I, causes characteristic AT→TA transversions found in the TP53 tumor suppressor gene in tumors from AAN and BEN patients. Understanding which human enzymes are involved in AAI activation to species forming DNA adducts and/or detoxication to the AAI O-demethylated metabolite, aristolochic acid Ia (AAIa), is important in the assessment of the susceptibility to this carcinogen. This review summarizes the latest data on identifying human and rodent enzymes participating in AAI metabolism. NAD(P)H:quinone oxidoreductase (NQO1) is the most efficient cytosolic nitroreductase activating AAI in vitro and in vivo. In human hepatic microsomes, AAI is activated by cytochrome P450 1A2 (CYP1A2) and, to a lesser extent, by CYP1A1; NADPH:CYP oxidoreductase also plays a minor role. Human and rodent CYP1A1 and 1A2 are also the principal enzymes involved in oxidative detoxication of AAI to AAIa in vitro and in vivo. The orientation of AAI in the active sites of human CYP1A1/2 and NQO1 was predicted from molecular modeling and is consistent with the efficient reduction of AAI by them observed experimentally. Molecular modeling also shows why CYP1A2 plays an important role in the oxidation of AAI to AAIa.


Subject(s)
Aristolochic Acids/pharmacokinetics , Kidney Diseases/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Oxidoreductases/metabolism , Animals , Aristolochic Acids/toxicity , Humans , Kidney Diseases/chemically induced , Neoplasms, Glandular and Epithelial/chemically induced , Urothelium/metabolism
17.
Cancer Epidemiol Biomarkers Prev ; 22(4): 653-60, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23417989

ABSTRACT

BACKGROUND: Acrylamide is a probable human carcinogen formed during cooking of starchy foods. Two large prospective cohort studies of dietary acrylamide intake and ovarian cancer risk observed a positive association, although two other studies reported no association. METHODS: We measured acrylamide exposure using red blood cell acrylamide and glycidamide hemoglobin adducts among women in two large prospective cohorts: the Nurses' Health Study and Nurses' Health Study II. Between blood collection and 2010, we identified 263 incident cases of epithelial ovarian cancer, matching two controls per case. We used logistic regression models to examine the association between acrylamide exposure and ovarian cancer risk, adjusting for matching factors, family history of ovarian cancer, tubal ligation, oral contraceptive use, body mass index, parity, alcohol intake, smoking, physical activity, and caffeine intake. RESULTS: The multivariate-adjusted relative risk (RR) of ovarian cancer comparing the highest versus lowest tertile of total acrylamide adducts was 0.79 (95% CI, 0.50-1.24, P trend = 0.08). The comparable RR of ovarian cancer among non-smokers at blood draw was 0.85 (95% CI, 0.57-1.27, P trend = 0.14). The association did not differ by tumor histology (serous invasive versus not), P for heterogeneity = 0.86. Individual adduct types (acrylamide or glycidamide) were not associated with risk. CONCLUSIONS: We observed no evidence that acrylamide exposure as measured by adducts to hemoglobin is associated with an increased risk of ovarian cancer. IMPACT: Our finding indicates that acrylamide intake may not increase risk of ovarian cancer.


Subject(s)
Acrylamide/adverse effects , Hemoglobins/metabolism , Neoplasms, Glandular and Epithelial/chemically induced , Ovarian Neoplasms/chemically induced , Adult , Carcinoma, Ovarian Epithelial , Case-Control Studies , Epoxy Compounds/adverse effects , Female , Humans , Middle Aged , Neoplasms, Glandular and Epithelial/metabolism , Nurses , Ovarian Neoplasms/metabolism , Ovary/metabolism , Prognosis , Prospective Studies , Risk Factors
19.
Gynecol Oncol ; 127(2): 316-20, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22835717

ABSTRACT

OBJECTIVE: To compare clinico-pathologic variables and protein expression of potential regulatory components in patients who develop type II endometrial cancer with and without antecedent tamoxifen. METHODS: Clinico-pathologic variables were compared for all surgically staged patients (2000-2008) with grade 3 endometrioid, papillary serous, clear cell, and carcinosarcoma of the uterus based on tamoxifen exposure [Tam (+) vs. Tam (-)]. Overall survival was analyzed using a multivariable Cox regression model and Kaplan-Meier estimates. Protein expression of ERα, PR, mTOR, p-mTOR, IGF-1R, EGFR, VEGF and HER-2/neu was compared by immunohistochemistry using a semiquantitative scoring system. RESULTS: Of 115 patients with high grade endometrial cancers, 15 received tamoxifen. These patients were older at diagnosis than Tam (-) patients. A higher percentage of Tam (+) patients had carcinosarcoma compared to Tam (-) patients (60% vs. 30%, P=0.038). Overall survival for Tam (+) patients was shorter than Tam (-) patients (16.6 vs. 32.2 months, P=0.004). The hazard ratio for death for Tam (+) patients was 2.53 (P=0.014), controlling for age and stage. Intensity and extent of staining were similar for ERα, PR, VEGF, EGFR, p-mTOR and HER-2/neu. The average expression score for IGF-1R and mTOR in the Tam (+) group was significantly higher than the Tam (-) group: 10.3 vs 7.0, P=0.001 and 6.0 vs 3.1, P=0.029, respectively. CONCLUSION: There are differences in the biology of type II endometrial cancers that develop in women with prior tamoxifen exposure. Tamoxifen associated cancers show higher expression of IGF-1R and mTOR, which should be further investigated.


Subject(s)
Antineoplastic Agents, Hormonal/adverse effects , Biomarkers, Tumor/metabolism , Carcinosarcoma/chemically induced , Endometrial Neoplasms/chemically induced , Neoplasms, Glandular and Epithelial/chemically induced , Tamoxifen/adverse effects , Adenocarcinoma, Clear Cell/chemically induced , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/mortality , Adenocarcinoma, Clear Cell/pathology , Aged , Carcinoma, Endometrioid/chemically induced , Carcinoma, Endometrioid/metabolism , Carcinoma, Endometrioid/mortality , Carcinoma, Endometrioid/pathology , Carcinosarcoma/metabolism , Carcinosarcoma/mortality , Carcinosarcoma/pathology , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/mortality , Endometrial Neoplasms/pathology , Female , Follow-Up Studies , Humans , Immunohistochemistry , Neoplasm Grading , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/mortality , Neoplasms, Glandular and Epithelial/pathology , Prognosis , Receptor, IGF Type 1/metabolism , Survival Analysis , TOR Serine-Threonine Kinases/metabolism , Tissue Array Analysis
20.
Genet Mol Res ; 10(4): 3236-45, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22194187

ABSTRACT

The search for new and effective antitumor agents with fewer cytotoxic side effects on normal tissue has increasingly become important. Lapachol, a natural organic compound isolated from the lapacho tree (Tabebuia avellandedae), is chemically identified as belonging to the naphthoquinone group and is known for its anti-inflammatory, analgesic and antibiotic properties, although there are questions about its effectiveness for treating neoplasic cells. We evaluated the antitumoral effects of lapachol by testing for clones of epithelial tumors in Drosophila melanogaster. Seventy-two-hour old larvae bred from wts/TM3, Sb(1) females and mwh/mwh males, were treated with different concentrations of lapachol (20, 40 and 60 µg/mL). Lapachol alone did not significantly increase the number of epithelial tumors. However, lapachol did significantly reduce the number of tumors provoked by doxorubicin.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drosophila Proteins/genetics , Drosophila melanogaster/drug effects , Naphthoquinones/pharmacology , Neoplasms, Experimental/drug therapy , Neoplasms, Glandular and Epithelial/drug therapy , Protein Kinases/genetics , Tabebuia/chemistry , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Crosses, Genetic , Dose-Response Relationship, Drug , Doxorubicin , Drosophila melanogaster/genetics , Female , Heterozygote , Humans , Larva/drug effects , Larva/genetics , Male , Naphthoquinones/therapeutic use , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Neoplasms, Glandular and Epithelial/chemically induced , Neoplasms, Glandular and Epithelial/genetics , Plant Extracts/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...