Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
JAMA Cardiol ; 2024 Apr 07.
Article in English | MEDLINE | ID: mdl-38583091

ABSTRACT

Importance: High-dose trivalent compared with standard-dose quadrivalent influenza vaccine did not significantly reduce all-cause mortality or cardiopulmonary hospitalizations in patients with high-risk cardiovascular disease in the INVESTED trial. Whether humoral immune response to influenza vaccine is associated with clinical outcomes is unknown. Objective: To examine the antibody response to high-dose trivalent compared with standard-dose quadrivalent inactivated influenza vaccine and its associations with clinical outcomes. Design, Setting, and Participants: This secondary analysis is a prespecified analysis of the immune response substudy of the randomized, double-blind, active-controlled INVESTED trial, which was conducted at 157 sites in the United States and Canada over 3 influenza seasons between September 2016 and January 2019. Antibody titers were determined by hemagglutination inhibition assays at randomization and 4 weeks during the 2017-2018 and 2018-2019 seasons. Eligibility criteria included recent acute myocardial infarction or heart failure hospitalization and at least 1 additional risk factor. Data were analyzed from February 2023 to June 2023. Main Outcomes and Measures: Mean antibody titer change, seroprotection (antibody titer level ≥1:40) and seroconversion (≥4-fold increase in titer) at 4 weeks, and the association between seroconversion status and the risk for adverse clinical outcomes. Interventions: High-dose trivalent or standard-dose quadrivalent inactivated influenza vaccine, with revaccination up to 3 seasons. Results: Antibody data were available for 658 of 5260 randomized participants (12.5%; mean [SD] age, 66.2 [11.4] years; 507 male [77.1%], 151 female [22.9%]; 348 with heart failure [52.9%]). High-dose vaccine was associated with an increased magnitude in antibody titers for A/H1N1, A/H3N2, and B-type antigens compared with standard dose. More than 92% of all participants achieved seroprotection for each of the contained antigens, while seroconversion rates were higher in participants who received high-dose vaccine. Seroconversion for any antigen was not associated with the risk for cardiopulmonary hospitalizations or all-cause mortality (hazard ratio, 1.09; 95% CI, 0.79-1.53; P = .59), irrespective of randomized treatment (P = .38 for interaction). Conclusions and Relevance: High-dose vaccine elicited a more robust humoral response in patients with heart failure or prior myocardial infarction enrolled in the INVESTED trial, with no association between seroconversion status and the risk for cardiopulmonary hospitalizations or all-cause mortality. Vaccination to prevent influenza remains critical in high-risk populations. Trial Registration: ClinicalTrials.gov Identifier: NCT02787044.

2.
Eur J Cancer Prev ; 33(1): 69-72, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37401516

ABSTRACT

BACKGROUND: Here we report clinical risk factors and event rates for the development of new non-melanoma skin cancer (NMSC) in a randomized, double-blind, placebo-controlled trial of the irreversible ornithine decarboxylase (ODC) inhibitor, difluromethylornithine (DFMO), over a 3-5-year follow-up. METHODS: 147 placebo patients (white; mean age 60.2 years; 60% male) were evaluated for event rates and association of initial skin biomarkers and baseline patient characteristics with the development of squamous cell (SCC) and basal cell (BCC) carcinomas. RESULTS: Post-study evaluation (median follow-up 4.4 years) indicates the measures of prior NMSCs ( P  ≤ 0.001), prior BCCs ( P  ≤ 0.001), prior SCCs ( P  = 0.011), prior tumor rate ( P  = 0.002), hemoglobin ( P  = 0.022), and gender ( P  = 0.045) as significant predictors for new NMSC development. Similarly, all measures of prior BCCs and NMSCs ( P  < 0.001), prior tumor rate ( P  = 0.014), and SCCs in the prior 2 years ( P  = 0.047) were statistically significant predictors for new BCC development. Total prior NMSCs and those in the prior 5 years ( P  < 0.001), total prior SCCs and those in the prior 5 years ( P  < 0.001), total prior BCCs and those in the prior 5 years ( P  ≤ 0.001), prior tumor rate ( P  = 0.011) as well as age ( P  = 0.008), hemoglobin ( P  = 0.002), and gender ( P  = 0.003) were statistically significant predictors of new SCC development. TPA-induced ODC activity at baseline showed no statistically significant association with the development of new NMSC ( P  = 0.35), new BCCs ( P  = 0.62), or new SCCs ( P  = 0.25). CONCLUSION: In the studied population, the history of and rate at which prior NMSCs occur are predictive and should be controlled for in future NMSC prevention trials.


Subject(s)
Carcinoma, Basal Cell , Carcinoma, Squamous Cell , Skin Neoplasms , Humans , Male , Middle Aged , Female , Carcinoma, Basal Cell/epidemiology , Carcinoma, Basal Cell/prevention & control , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/epidemiology , Clinical Trials as Topic , Skin Neoplasms/diagnosis , Skin Neoplasms/epidemiology , Skin Neoplasms/pathology , Hemoglobins
3.
J Immunother Cancer ; 10(10)2022 Oct.
Article in English | MEDLINE | ID: mdl-36192087

ABSTRACT

BACKGROUND: Radiation therapy (RT) has been demonstrated to generate an in situ vaccination (ISV) effect in murine models and in patients with cancer; however, this has not routinely translated into enhanced clinical response to immune checkpoint inhibition (ICI). We investigated whether the commonly used vaccine adjuvant, monophosphoryl lipid A (MPL) could augment the ISV regimen consisting of combination RT and ICI. MATERIALS/METHODS: We used syngeneic murine models of melanoma (B78) and prostate cancer (Myc-CaP). Tumor-bearing mice received either RT (12 Gy, day 1), RT+anti-CTLA-4 (C4, day 3, 6, 9), MPL (20 µg IT injection days 5, 7, 9), RT+C4+MPL, or PBS control. To evaluate the effect of MPL on the irradiated tumor microenvironment, primary tumor with tumor draining lymph nodes were harvested for immune cell infiltration analysis and cytokine profiling, and serum was collected for analysis of antitumor antibody populations. RESULTS: Combination RT+C4+MPL significantly reduced tumor growth, increased survival and complete response rate compared with RT+C4 in both B78 and Myc-CaP models. MPL favorably reprogrammed the irradiated tumor-immune microenvironment toward M1 macrophage and Th1 TBET+CD4+ T cell polarization. Furthermore, MPL significantly increased intratumoral expression of several Th1-associated and M1-associated proinflammatory cytokines. In co-culture models, MPL-stimulated macrophages directly activated CD8 T cells and polarized CD4 cells toward Th1 phenotype. MPL treatment significantly increased production of Th1-associated, IgG2c antitumor antibodies, which were required for and predictive of antitumor response to RT+C4+MPL, and enabled macrophage-mediated antibody-dependent direct tumor cell killing by MPL-stimulated macrophages. Macrophage-mediated tumor cell killing was dependent on FcγR expression. In metastatic models, RT and MPL generated a systemic antitumor immune response that augmented response to ICIs. This was dependent on macrophages and CD4+ but not CD8+T cells. CONCLUSIONS: We report the potential for MPL to augment the ISV effect of combination RT+C4 through FcγR, macrophage, and TBET+CD4+ Th1 cell dependent mechanisms. To our knowledge, this is the first report describing generation of a CD8+ T cell-independent, Th1 polarized, systemic antitumor immune response with subsequent generation of immunologic memory. These findings support the potential for vaccine adjuvants to enhance the efficacy of in situ tumor vaccine approaches.


Subject(s)
Cancer Vaccines , Toll-Like Receptor 4 , Animals , CD8-Positive T-Lymphocytes , Cancer Vaccines/pharmacology , Cytokines , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Male , Mice , Receptors, IgG , Vaccination
4.
Nat Commun ; 13(1): 4948, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35999216

ABSTRACT

Radiation therapy (RT) activates an in situ vaccine effect when combined with immune checkpoint blockade (ICB), yet this effect may be limited because RT does not fully optimize tumor antigen presentation or fully overcome suppressive mechanisms in the tumor-immune microenvironment. To overcome this, we develop a multifunctional nanoparticle composed of polylysine, iron oxide, and CpG (PIC) to increase tumor antigen presentation, increase the ratio of M1:M2 tumor-associated macrophages, and enhance stimulation of a type I interferon response in conjunction with RT. In syngeneic immunologically "cold" murine tumor models, the combination of RT, PIC, and ICB significantly improves tumor response and overall survival resulting in cure of many mice and consistent activation of tumor-specific immune memory. Combining RT with PIC to elicit a robust in situ vaccine effect presents a simple and readily translatable strategy to potentiate adaptive anti-tumor immunity and augment response to ICB or potentially other immunotherapies.


Subject(s)
Multifunctional Nanoparticles , Neoplasms , Animals , Antigens, Neoplasm , Cell Line, Tumor , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Mice , Neoplasms/radiotherapy , Tumor Microenvironment , Vaccination
5.
JAMA ; 325(1): 39-49, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33275134

ABSTRACT

Importance: Influenza is temporally associated with cardiopulmonary morbidity and mortality among those with cardiovascular disease who may mount a less vigorous immune response to vaccination. Higher influenza vaccine dose has been associated with reduced risk of influenza illness. Objective: To evaluate whether high-dose trivalent influenza vaccine compared with standard-dose quadrivalent influenza vaccine would reduce all-cause death or cardiopulmonary hospitalization in high-risk patients with cardiovascular disease. Design, Setting, and Participants: Pragmatic multicenter, double-blind, active comparator randomized clinical trial conducted in 5260 participants vaccinated for up to 3 influenza seasons in 157 sites in the US and Canada between September 21, 2016, and January 31, 2019. Patients with a recent acute myocardial infarction or heart failure hospitalization and at least 1 additional risk factor were eligible. Interventions: Participants were randomly assigned to receive high-dose trivalent (n = 2630) or standard-dose quadrivalent (n = 2630) inactivated influenza vaccine and could be revaccinated for up to 3 seasons. Main Outcomes and Measures: The primary outcome was the time to the composite of all-cause death or cardiopulmonary hospitalization during each enrolling season. The final date of follow-up was July 31, 2019. Vaccine-related adverse events were also assessed. Results: Among 5260 randomized participants (mean [SD] age, 65.5 [12.6] years; 3787 [72%] men; 3289 [63%] with heart failure) over 3 influenza seasons, there were 7154 total vaccinations administered and 5226 (99.4%) participants completed the trial. In the high-dose trivalent vaccine group, there were 975 primary outcome events (883 hospitalizations for cardiovascular or pulmonary causes and 92 deaths from any cause) among 884 participants during 3577 participant-seasons (event rate, 45 per 100 patient-years), whereas in the standard-dose quadrivalent vaccine group, there were 924 primary outcome events (846 hospitalizations for cardiovascular or pulmonary causes and 78 deaths from any cause) among 837 participants during 3577 participant-seasons (event rate, 42 per 100 patient-years) (hazard ratio, 1.06 [95% CI, 0.97-1.17]; P = .21). In the high-dose vs standard-dose groups, vaccine-related adverse reactions occurred in 1449 (40.5%) vs 1229 (34.4%) participants and severe adverse reactions occurred in 55 (2.1%) vs 44 (1.7%) participants. Conclusions and Relevance: In patients with high-risk cardiovascular disease, high-dose trivalent inactivated influenza vaccine, compared with standard-dose quadrivalent inactivated influenza vaccine, did not significantly reduce all-cause mortality or cardiopulmonary hospitalizations. Influenza vaccination remains strongly recommended in this population. Trial Registration: ClinicalTrials.gov Identifier: NCT02787044.


Subject(s)
Cardiovascular Diseases/epidemiology , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Mortality , Aged , Cardiovascular Diseases/mortality , Double-Blind Method , Female , Heart Failure/complications , Hospitalization/statistics & numerical data , Humans , Influenza Vaccines/adverse effects , Influenza, Human/mortality , Male , Middle Aged , Myocardial Infarction/complications , Risk Factors , Survival Analysis , Vaccines, Inactivated/administration & dosage
6.
AJR Am J Roentgenol ; 213(6): 1259-1266, 2019 12.
Article in English | MEDLINE | ID: mdl-31386573

ABSTRACT

OBJECTIVE. The purpose of this study was to evaluate the utility of CT texture analysis (CTTA) in differentiating low-attenuation renal cell carcinoma (RCC) from renal cysts on unenhanced CT. MATERIALS AND METHODS. Ninety-four patients with low-attenuation RCC on unenhanced CT were compared with a cohort of 192 patients with benign renal cysts. CT characteristics (size and minimum, maximum, and mean attenuation) and CTTA features were recorded using an ROI approximately two-thirds the size of the mass. Masses were subjectively assessed by two expert genitourinary readers and two novice readers using a 5-point Likert scale (1 = definite cyst, 5 = definite renal cell carcinoma). Results of first-order CTTA and subjective evaluation were compared using ROC analysis. RESULTS. The group of 94 patients with low-attenuation RCC included 62 men and 32 women (mean age, 58.0 years). On unenhanced CT, the RCC were larger than 10 mm and of a median size of 50 mm with less than or equal to 20 HU (mean attenuation, 16 ± 4 HU). Of the RCC cohort, 83 were clear cell subtype. The cohort of 192 patients included 134 men and 58 women (mean age, 64.7 years) with benign renal cysts greater than 10 mm and a median size of 27 mm and less than or equal to 20 HU (mean attenuation, 9 ± 6 HU). The mean follow-up time was 6.2 years. Mean entropy in the low-attenuation RCC group (4.1 ± 0.7) was significantly higher than in the cyst group (2.8 ± 1.3, p < 0.0001). Entropy showed an ROC AUC of 0.89, with sensitivity of 84% and specificity of 80% at threshold 3.9. The AUC was better than subjective evaluation by novice readers (AUC, 0.77) and comparable to subjective evaluation by two expert readers (AUC, 0.90). A model combining the three best texture features (unfiltered mean gray-level attenuation, coarse entropy, and kurtosis) showed an improved AUC of 0.92. CONCLUSION. High entropy revealed with CTTA may be used to differentiate low-attenuation RCC from cysts at unenhanced CT; this technique performs as well as expert readers.


Subject(s)
Carcinoma, Renal Cell/diagnostic imaging , Kidney Diseases, Cystic/diagnostic imaging , Kidney Neoplasms/diagnostic imaging , Tomography, X-Ray Computed/methods , Aged , Diagnosis, Differential , Female , Humans , Male , Middle Aged , Radiographic Image Interpretation, Computer-Assisted , Retrospective Studies , Sensitivity and Specificity
7.
Abdom Radiol (NY) ; 44(6): 1999-2008, 2019 06.
Article in English | MEDLINE | ID: mdl-29804215

ABSTRACT

PURPOSE: To assess CT texture features of small renal cell carcinomas (≤ 4cm) for association with key pathologic features including protein biomarkers. METHODS: Quantitative CT texture analysis (CTTA) of small renal cancers (≤ 4cm) was performed on non-contrast and portal venous phase abdominal MDCT scans with an ROI drawn at the largest cross-sectional diameter of the tumor using commercially available software. Texture parameters including mean pixel attenuation, the standard deviation (SD) of the pixel distribution histogram, entropy, the mean of positive pixels, the skewness (i.e., asymmetry) of the pixel histogram, kurtosis (i.e., peakness) of the pixel histogram, and the percentage of positive pixels were correlated with pathologic data from surgical resection, including histology and nuclear grade, as well as microarray analysis in a subset (n = 40) including Ki67 index, CRP, and neovascularization (CD105/CD31). RESULTS: Portal venous phase images were available in 249 patients (105 women, 144 men; mean age, 56.7 years) with tumors ≤ 4cm (mean, median, range, ± SD; 2.66, 2.60, 0.3-4.0 ± 0.85 cm). CT texture features of standard deviation, mean of the positive pixels, and entropy of the pixel histogram were significantly associated with histologic cell type (clear vs. non-clear; p < 0.001). Entropy and mean of the positive pixels also showed an association with nuclear grade, although not statistically significant. In the microarray analysis subset, kurtosis of the pixel histogram was associated with CD105/CD31 (p = 0.05). SD also showed some association with CD 105 positivity (p = 0.02) and CAIX expression (p = 0.01). Non-contrast CT images were available in 174 patients (72 women, 102 men; mean age, 57.5 years). Although the association with histology was not as strong as on the portal venous phase, in the subset of patients with microarray data, SD was found to correlate with CRP (p = 0.08), kurtosis with CRP (p = 0.004), CD105/CD31 (p = 0.002), and with Ki 67 index (p < 0.001). CONCLUSION: CT texture features were significantly associated with important histopathologic features in small renal cancers. These non-invasive measures can be performed retrospectively and may provide useful information when determining follow-up and treatment of small renal cancers.


Subject(s)
Carcinoma, Renal Cell/diagnostic imaging , Kidney Neoplasms/diagnostic imaging , Tomography, X-Ray Computed/methods , Biomarkers, Tumor/analysis , Carcinoma, Renal Cell/pathology , Female , Humans , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Grading , Proteins/analysis , Radiographic Image Interpretation, Computer-Assisted
8.
FASEB J ; 33(3): 3198-3211, 2019 03.
Article in English | MEDLINE | ID: mdl-30379590

ABSTRACT

Growth factor receptor-binding protein 10 (GRB10) is a well-known adaptor protein and a recently identified substrate of the mammalian target of rapamycin (mTOR). Depletion of GRB10 increases insulin sensitivity and overexpression suppresses PI3K/Akt signaling. Because the major reason for the limited efficacy of PI3K/Akt-targeted therapies in prostate cancer (PCa) is loss of mTOR-regulated feedback suppression, it is therefore important to assess the functional importance and regulation of GRB10 under these conditions. On the basis of these background observations, we explored the status and functional impact of GRB10 in PCa and found maximum expression in phosphatase and tensin homolog (PTEN)-deficient PCa. In human PCa samples, GRB10 inversely correlated with PTEN and positively correlated with pAKT levels. Knockdown of GRB10 in nontumorigenic PTEN null mouse embryonic fibroblasts and tumorigenic PCa cell lines reduced Akt phosphorylation and selectively activated a panel of receptor tyrosine kinases. Similarly, overexpression of GRB10 in PTEN wild-type PCa cell lines accelerated tumorigenesis and induced Akt phosphorylation. In PTEN wild-type PCa, GRB10 overexpression promoted mediated PTEN interaction and degradation. PI3K (but not mTOR) inhibitors reduced GRB10 expression, suggesting primarily PI3K-driven regulation of GRB10. In summary, our results suggest that GRB10 acts as a major downstream effector of PI3K and has tumor-promoting effects in prostate cancer.-Khan, M. I., Al Johani, A., Hamid, A., Ateeq, B., Manzar, N., Adhami, V. M., Lall, R. K., Rath, S., Sechi, M., Siddiqui, I. A., Choudhry, H., Zamzami, M. A., Havighurst, T. C., Huang, W., Ntambi, J. M., Mukhtar, H. Proproliferatve function of adaptor protein GRB10 in prostate carcinoma.


Subject(s)
GRB10 Adaptor Protein/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Carcinogens/antagonists & inhibitors , Carcinogens/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , GRB10 Adaptor Protein/antagonists & inhibitors , GRB10 Adaptor Protein/genetics , Gene Knockdown Techniques , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Models, Biological , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/genetics , RNA, Messenger , Signal Transduction
9.
Cancer Prev Res (Phila) ; 10(5): 298-307, 2017 May.
Article in English | MEDLINE | ID: mdl-28325826

ABSTRACT

We performed a phase II pharmacodynamic prevention trial of Polyphenon E [a green tea polyphenol formulation primarily consisting of epigallocatechin gallate (EGCG)] in patients prior to bladder cancer surgery. Patients with a bladder tumor were randomized to receive Polyphenon E containing either 800 or 1,200 mg of EGCG or placebo for 14 to 28 days prior to transurethral resection of bladder tumor or cystectomy. The primary objective was to compare the postintervention EGCG tissue levels in patients receiving Polyphenon E as compared with placebo. Secondary objectives included assessments of tissue expression of PCNA, MMP2, clusterin, VEGF, p27, IGF-1, IGFBP-3; correlation of tissue, plasma, and urine levels of EGCG; and EGCG metabolism by catechol-O-methyltransferase and UDP-glucuronosyltransferase pharmacogenomic mutations. Thirty-one patients (male:female, 26:5; mean age, 67.2 years) were randomized and 29 (94%) completed the study. There was not an observed significant difference (P = 0.12) in EGCG tissue levels between two Polyphenon E dosage groups combined versus placebo. However, a dose-response relationship for EGCG levels was observed in both normal (P = 0.046) and malignant bladder tissue (P = 0.005) across the three study arms. In addition, EGCG levels in plasma (P < 0.001) and urine (P < 0.001) increased and PCNA (P = 0.016) and clusterin (P = 0.008) were downregulated in a dose-dependent fashion. No pharmacogenomic relationship was observed. EGCG levels in plasma, urine, and bladder tissue followed a dose-response relationship, as did modulation of tissue biomarkers of proliferation and apoptosis. Despite the limitations of this pilot study, the observed pharmacodynamics and desirable biologic activity warrant further clinical studies of this agent in bladder cancer prevention. Cancer Prev Res; 10(5); 298-307. ©2017 AACR.


Subject(s)
Catechin/analogs & derivatives , Urinary Bladder Neoplasms , Urinary Bladder/drug effects , Adult , Aged , Biomarkers, Tumor/analysis , Catechin/administration & dosage , Catechin/pharmacokinetics , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Male , Middle Aged , Pilot Projects , Tissue Distribution
10.
Biomaterials ; 101: 20-31, 2016 09.
Article in English | MEDLINE | ID: mdl-27267625

ABSTRACT

Triple negative breast cancer (TNBC) is an aggressive subtype of breast cancer for which there is no available targeted therapy. TNBC cases contribute disproportionately to breast cancer-related mortality, thus the need for novel and effective therapeutic methods is urgent. We have previously shown that a National Cancer Institute (NCI) investigational drug aminoflavone (AF) exhibits strong growth inhibitory effects in TNBC cells. However, in vivo pulmonary toxicity resulted in withdrawal or termination of several human clinical trials for AF. Herein we report the in vivo efficacy of a nanoformulation of AF that enhances the therapeutic index of AF in TNBC. We engineered a unique unimolecular micelle nanoparticle (NP) loaded with AF and conjugated with GE11, a 12 amino acid peptide targeting epidermal growth factor receptor (EGFR), since EGFR amplification is frequently observed in TNBC tumors. These unimolecular micelles possessed excellent stability and preferentially released drug payload at endosomal pH levels rather than blood pH levels. Use of the GE11 targeting peptide resulted in enhanced cellular uptake and strong growth inhibitory effects in TNBC cells. Further, AF-loaded, GE11-conjugated (targeted) unimolecular micelle NPs significantly inhibit orthotopic TNBC tumor growth in a xenograft model, compared to treatment with AF-loaded, GE11-lacking (non-targeted) unimolecular micelle NPs or free AF. Interestingly, the animals treated with AF-loaded, targeted NPs had the highest plasma and tumor level of AF among different treatment groups yet exhibited no increase in plasma aspartate aminotransferase (AST) activity level or observable tissue damage at the time of sacrifice. Together, these results highlight AF-loaded, EGFR-targeted unimolecular micelle NPs as an effective therapeutic option for EGFR-overexpressing TNBC.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Carriers/chemistry , ErbB Receptors/metabolism , Flavonoids/administration & dosage , Nanoparticles/chemistry , Peptides/chemistry , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Breast/drug effects , Breast/metabolism , Breast/pathology , Cell Line, Tumor , Drug Carriers/metabolism , Drug Delivery Systems , Female , Flavonoids/pharmacokinetics , Flavonoids/therapeutic use , Humans , Micelles , Nanoparticles/metabolism , Nanoparticles/ultrastructure , Peptides/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
11.
Carcinogenesis ; 37(1): 72-80, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26586792

ABSTRACT

Protein kinase C epsilon (PKCε), a Ca(2+)-independent phospholipid-dependent serine/threonine kinase, is among the six PKC isoforms (α, δ, ε, η, µ, ζ) expressed in both mouse and human skin. Epidermal PKCε level dictates the susceptibility of PKCε transgenic (TG) mice to the development of cutaneous squamous cell carcinomas (SCC) elicited either by repeated exposure to ultraviolet radiation (UVR) or by using the DMBA initiation-TPA (12-O-tetradecanoylphorbol-13-acetate) tumor promotion protocol (Wheeler,D.L. et al. (2004) Protein kinase C epsilon is an endogenous photosensitizer that enhances ultraviolet radiation-induced cutaneous damage and development of squamous cell carcinomas. Cancer Res., 64, 7756-7765). Histologically, SCC in TG mice, like human SCC, is poorly differentiated and metastatic. Our earlier studies to elucidate mechanisms of PKCε-mediated development of SCC, using either DMBA-TPA or UVR, indicated elevated release of cytokine TNFα. To determine whether TNFα is essential for the development of SCC in TG mice, we generated PKCε transgenic mice/TNFα-knockout (TG/TNFαKO) by crossbreeding TNFαKO with TG mice. We now present that deletion of TNFα in TG mice inhibited the development of SCC either by repeated UVR exposures or by the DMBA-TPA protocol. TG mice deficient in TNFα elicited both increase in SCC latency and decrease in SCC incidence. Inhibition of UVR-induced SCC development in TG/TNFαKO was accompanied by inhibition of (i) the expression levels of TNFα receptors TNFRI and TNFRII and cell proliferation marker ornithine decarboxylase and metastatic markers MMP7 and MMP9, (ii) the activation of transcription factors Stat3 and NF-kB and (iii) proliferation of hair follicle stem cells and epidermal hyperplasia. The results presented here provide the first genetic evidence that TNFα is linked to PKCε-mediated sensitivity to DMBA-TPA or UVR-induced development of cutaneous SCC.


Subject(s)
Carcinoma, Squamous Cell/prevention & control , Protein Kinase C-epsilon/genetics , Skin Neoplasms/prevention & control , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics , 9,10-Dimethyl-1,2-benzanthracene , Animals , Carcinogenesis/chemically induced , Carcinogenesis/genetics , Carcinogenesis/radiation effects , Carcinogens , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/radiation effects , Female , Male , Mice , Mice, Knockout , Mice, Transgenic , Protein Kinase C-epsilon/biosynthesis , Skin Neoplasms/etiology , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Tetradecanoylphorbol Acetate , Ultraviolet Rays
12.
Eur J Cancer Prev ; 25(4): 312-20, 2016 07.
Article in English | MEDLINE | ID: mdl-26313229

ABSTRACT

Epidemiologic, preclinical, and early phase I studies of the cruciferous vegetable bioactive metabolite, 3,3'-diindolylmethane (DIM), support its potential prostate cancer chemopreventive ability. We performed a multicenter, double-blind, placebo-controlled trial of DIM in patients diagnosed with prostate cancer and scheduled for radical prostatectomy. A total of 45 patients with organ-confined prostate cancer were randomized to 21-28 days of an absorption-enhanced formulation of DIM (BR-DIM) at doses of 100 or 200 mg per os twice daily or to placebo twice daily. Prostate tissue levels of DIM were the primary endpoint, with selected secondary biomarker endpoints including blood levels of DIM, total prostate-specific antigen, testosterone, and the insulin-like growth factor-1: insulin-like growth factor binding protein-3 ratio and the urinary 2-hydroxyestrone/16-hydroxyestrone ratio, obtained at baseline, at day 15, and before surgery, as well as tissue expression of androgen receptor, prostate-specific antigen, Ki67, caspase 3 with cytochrome p450 mRNA expression and genotyping (polymorphisms). DIM was well tolerated with excellent study compliance and relatively rapid accrual of all 45 patients within 1 year. DIM levels were detected in only seven of 28 prostate tissue specimens. There was a statistically significant difference in the change in the urinary 2-hydroxyestrone/16-hydroxyestrone ratio from baseline until before surgery between the placebo and 400 mg DIM groups, with otherwise statistically nonsignificant changes in plasma biomarker expression. The administration of BR-DIM to prostate cancer patients before prostatectomy yields detectable plasma levels but without consistent or significant tissue accumulation or biomarker modulation. This study demonstrates the feasibility of biologic evaluation of relatively nontoxic preventive agents in the preprostatectomy setting with the potential for rapid accrual.


Subject(s)
Adenocarcinoma/metabolism , Anticarcinogenic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Indoles/therapeutic use , Prostatectomy , Prostatic Neoplasms/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Double-Blind Method , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery
13.
Cancer Prev Res (Phila) ; 8(12): 1146-55, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26391916

ABSTRACT

The purpose of this study was to assess the feasibility of using the selective estrogen receptor modulator (SERM) acolbifene as a breast cancer prevention agent in premenopausal women. To do so, we assessed change in proliferation in benign breast tissue sampled by random periareolar fine-needle aspiration (RPFNA) as a primary endpoint, along with changes in other risk biomarkers and objective and subjective side effects as secondary endpoints. Twenty-five women with cytologic hyperplasia ± atypia and ≥2% of breast epithelial cells staining positive for Ki-67, received 20 mg acolbifene daily for 6-8 months, and then had benign breast tissue and blood risk biomarkers reassessed. Ki-67 decreased from a median of 4.6% [interquartile range (IQR), 3.1%-8.5%] at baseline to 1.4% (IQR, 0.6%-3.5%) after acolbifene (P < 0.001; Wilcoxon signed-rank test), despite increases in bioavailable estradiol. There were also significant decreases in expression (RT-qPCR) of estrogen-inducible genes that code for pS2, ERα, and progesterone receptor (P ≤ 0.026). There was no significant change in serum IGF1, IGFBP3, IGF1:IGFBP3 ratio, or mammographic breast density. Subjective side effects were minimal with no significant increase in hot flashes, muscle cramps, arthralgias, or fatigue. Objective measures showed a clinically insignificant decrease in lumbar spine bone density (DEXA) and an increase in ovarian cysts but no change in endometrial thickness (sonography). In summary, acolbifene was associated with favorable changes in benign breast epithelial cell proliferation and estrogen-inducible gene expression but minimal side effects, suggesting a phase IIB placebo-controlled trial evaluating it further for breast cancer prevention.


Subject(s)
Breast Neoplasms/prevention & control , Breast/drug effects , Cell Proliferation/drug effects , Piperidines/therapeutic use , Premenopause , Selective Estrogen Receptor Modulators/therapeutic use , Adult , Biopsy, Fine-Needle , Bone Density/drug effects , Epithelial Cells/drug effects , Female , Humans , Ki-67 Antigen/analysis , Ki-67 Antigen/biosynthesis , Middle Aged , Ovarian Cysts/epidemiology , Pilot Projects , Real-Time Polymerase Chain Reaction , Risk Factors , Transcriptome/drug effects
14.
J Aging Phys Act ; 22(4): 571-7, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24306390

ABSTRACT

This study examined the intensity of activity contributing to physical activity energy expenditure in older adults. In 57 men and women aged ≥ 65, total energy expenditure (TEE) was measured using doubly labeled water and resting metabolic rate was measured using indirect calorimetry to calculate a physical activity index (PAI). Sedentary time and physical activity of light and moderate to vigorous (mod/vig) intensity was measured using an accelerometer. The subjects were 75 ± 7 yrs (mean ± SD) of age and 79% female. Subjects spent 66 ± 8, 25 ± 5, and 9 ± 4% of monitor wear time in sedentary, light, and mod/vig activity per day, respectively. In a mixture regression model, both light (ß = 29.6 [15.6-43.6, 95% CI]), p < .001) and mod/vig intensity activity (ß = 28.7 [7.4-50.0, 95% CI]), p = .01) were strongly associated with PAI, suggesting that both light and mod/vig intensity activities are major determinants of their physical activity energy expenditure.


Subject(s)
Aging , Energy Metabolism/physiology , Motor Activity/physiology , Accelerometry/methods , Aged , Aged, 80 and over , Aging/physiology , Aging/psychology , Calorimetry, Indirect/methods , Female , Humans , Male , Sedentary Behavior
15.
Clin Med Res ; 11(3): 107-12, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23580787

ABSTRACT

OBJECTIVE: We conducted a pilot survey to evaluate breast cancer patients' willingness to participate in a preoperative chemoprevention (ie, window-of-opportunity) study. Design A 27-question written survey was developed and administered to participants. Setting A breast cancer specialty clinic at the University of Wisconsin Hospital and Clinics. Participants 30 adult patients with newly diagnosed operable breast cancer participated after signing informed consent. METHODS: A convenience sample of 30 participants was recruited from July 2005 through January 2006. Participants were administered the survey in clinic. Univariate ordinal logistic regression models were used to identify predictors of willingness to participate in window-of-opportunity trials. RESULTS: Overall, 26.7% of respondents were willing to participate in a research trial between the time of breast cancer diagnosis and surgery. Univariate ordinal logistic regression models identified that women with a prior history of breast cancer (P=0.060), prior research participation (P=0.006), more education (P=0.034), and self-reported breast cancer knowledge (P=0.043) were more willing to participate. On average, women preferred to have surgery 7 days (range 1-14) after their diagnosis, but the actual average wait time between diagnostic biopsy and surgery was 37.5 days (standard deviation = 23.4 days). CONCLUSION: There is ample time before breast surgery to conduct preoperative window-of-opportunity trials. Interventions aimed at expanding patients' breast cancer knowledge may improve accrual to window-of-opportunity studies.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/surgery , Patient Education as Topic , Patient Participation , Aged , Aged, 80 and over , Female , Humans , Logistic Models , Middle Aged , Retrospective Studies , Surveys and Questionnaires
16.
Med Sci Sports Exerc ; 43(5): 867-76, 2011 May.
Article in English | MEDLINE | ID: mdl-20881882

ABSTRACT

PURPOSE: To compare the validity of various physical activity measures with doubly labeled water (DLW)-measured physical activity energy expenditure (PAEE) in free-living older adults. METHODS: Fifty-six adults aged ≥65 yr wore three activity monitors (New Lifestyles pedometer, ActiGraph accelerometer, and a SenseWear (SW) armband) during a 10-d free-living period and completed three different surveys (Yale Physical Activity Survey (YPAS), Community Health Activities Model Program for Seniors (CHAMPS), and a modified Physical Activity Scale for the Elderly (modPASE)). Total energy expenditure was measured using DLW, resting metabolic rate was measured with indirect calorimetry, the thermic effect of food was estimated, and from these, estimates of PAEE were calculated. The degree of linear association between the various measures and PAEE was assessed, as were differences in group PAEE, when estimable by a given measure. RESULTS: All three monitors were significantly correlated with PAEE (r=0.48-0.60, P<0.001). Of the questionnaires, only CHAMPS was significantly correlated with PAEE (r=0.28, P=0.04). Statistical comparison of the correlations suggested that the monitors were superior to YPAS and modPASE. Mean squared errors for all correlations were high, and the median PAEE from the different tools was significantly different from DLW for all but the YPAS and regression-estimated PAEE from the ActiGraph. CONCLUSIONS: Objective devices more appropriately rank PAEE than self-reported instruments in older adults, but absolute estimates of PAEE are not accurate. Given the cost differential and ease of use, pedometers seem most useful in this population when ranking by physical activity level is adequate.


Subject(s)
Actigraphy/instrumentation , Motor Activity/physiology , Surveys and Questionnaires/standards , Aged , Energy Metabolism/physiology , Female , Humans , Male , Models, Statistical
17.
Am J Pathol ; 176(6): 2986-96, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20382709

ABSTRACT

TMPRSS2, a type II transmembrane serine protease, is highly expressed by the epithelium of the human prostate gland. To explore the regulation and function of TMPRSS2 in the prostate, a panel of monoclonal antibodies with high sensitivity and specificity were generated. Immunodetection showed TMPRSS2 on the apical plasma membrane of the prostate luminal cells and demonstrated its release into semen as a component of prostasomes, organelle-like vesicles that may facilitate sperm function and enhance male reproduction. In prostate cancer cells, TMPRSS2 expression was increased and the protein mislocalized over the entire tumor cell membrane. In both LNCaP prostate cancer cells and human semen, TMPRSS2 protein was detected predominantly as inactive zymogen forms as part of an array of multiple noncovalent and disulfide-linked complexes, suggesting that TMPRSS2 activity may be regulated by unconventional mechanisms. Our data suggested that TMPRSS2, an apical surface serine protease, may have a normal role in male reproduction as a component of prostasomes. The aberrant cellular localization, and increased expression of the protease seen in cancer, may contribute to prostate tumorigenesis by providing access of the enzyme to nonphysiological substrates and binding-proteins.


Subject(s)
Epithelial Cells , Prostate/cytology , Prostate/enzymology , Prostatic Neoplasms/enzymology , Semen/enzymology , Serine Endopeptidases/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Monoclonal/metabolism , Cell Line, Tumor , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Male , Mice , Microarray Analysis , Middle Aged , Prostate/pathology , Prostatic Neoplasms/pathology , Serine Endopeptidases/genetics
18.
Cancer Prev Res (Phila) ; 3(1): 35-47, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20051371

ABSTRACT

Preclinical studies have shown that the inhibition of ornithine decarboxylase (ODC) by alpha-difluoromethylornithine (DFMO) and resultant decreases in tissue concentrations of polyamines (putrescine and spermidine) prevents neoplastic developments in many tissue types. Clinical studies of oral DFMO at 500 mg/m(2)/day revealed it to be safe and tolerable and resulted in significant inhibition of phorbol ester-induced skin ODC activity. Two hundred and ninety-one participants (mean age, 61 years; 60% male) with a history of prior nonmelanoma skin cancer (NMSC; mean, 4.5 skin cancers) were randomized to oral DFMO (500 mg/m(2)/day) or placebo for 4 to 5 years. There was a trend toward a history of more prior skin cancers in subjects randomized to placebo, but all other characteristics including sunscreen and nonsteroidal anti-inflammatory drug use were evenly distributed. Evaluation of 1,200 person-years of follow-up revealed a new NMSC rate of 0.5 events/person/year. The primary end point, new NMSCs, was not significantly different between subjects taking DFMO and placebo (260 versus 363 cancers, P = 0.069, two-sample t test). Evaluation of basal cell (BCC) and squamous cell cancers separately revealed very little difference in squamous cell cancer between treatment groups but a significant difference in new BCC (DFMO, 163 cancers; placebo, 243 cancers; expressed as event rate of 0.28 BCC/person/year versus 0.40 BCC/person/year, P = 0.03). Compliance with DFMO was >90% and it seemed to be well tolerated with evidence of mild ototoxicity as measured by serial audiometric examination when compared with placebo subjects. The analysis of normal skin biopsies revealed a significant (P < 0.05) decrease in 12-0-tetradecanoylphorbol-13-acetate-induced ODC activity (month 24, 36, and 48) and putrescine concentration (month 24 and 36 only) in DFMO subjects. Subjects with a history of skin cancer taking daily DFMO had an insignificant reduction (P = 0.069) in new NMSC that was predominantly due to a marked reduction in new BCC. Based on these data, the potential of DFMO, alone or in combination, to prevent skin cancers should be explored further.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Basal Cell/prevention & control , Carcinoma, Squamous Cell/prevention & control , Eflornithine/therapeutic use , Skin Neoplasms/prevention & control , Audiometry , Carcinoma, Basal Cell/mortality , Carcinoma, Squamous Cell/mortality , Double-Blind Method , Female , Hearing/drug effects , Humans , Kaplan-Meier Estimate , Male , Medication Adherence , Middle Aged , Ornithine Decarboxylase Inhibitors , Skin Neoplasms/mortality
19.
J Invest Dermatol ; 130(1): 270-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19626035

ABSTRACT

Chronic exposure to UVR is the major etiologic factor in the development of human skin cancers including squamous-cell carcinoma (SCC). We have previously shown that protein Kinase C epsilon (PKCepsilon) transgenic mice on FVB/N background, which overexpress PKCepsilon protein approximately eightfold over endogenous levels in epidermis, exhibit about threefold more sensitivity than wild-type littermates to UVR-induced development of SCC. To determine whether it is PKCepsilon and not the mouse genetic background that determines susceptibility to UVR carcinogenesis, we cross-bred PKCepsilon FVB/N transgenic mice with SKH-1 hairless mice to generate PKCepsilon-overexpressing SKH-1 hairless mice. To evaluate the susceptibility of PKCepsilon SKH-1 hairless transgenic mice to UVR carcinogenesis, the mice were exposed to UVR (1-2 KJ m(-2)) three times weekly from a bank of six kodacel-filtered FS40 sunlamps. As compared with the wild-type hairless mice, PKCepsilon overexpression in SKH-1 hairless mice decreased the latency (12 weeks), whereas it increased the incidence (twofold) and multiplicity (fourfold) of SCC. The SKH hairless transgenic mice were observed to be as sensitive as FVB/N transgenic mice to UVR-induced development of SCC and expression of proliferative markers (proliferating cell nuclear antigen, signal transducers and activators of transcription 3, and extracellular signal-regulated kinase 1/2). The results indicate that PKCepsilon level dictates susceptibility, irrespective of genetic background, to UVR carcinogenesis.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Protein Kinase C-epsilon/genetics , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Ultraviolet Rays/adverse effects , Animals , Cell Nucleus/metabolism , Dose-Response Relationship, Radiation , Epidermis/physiology , Epidermis/radiation effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Male , Mice , Mice, Hairless , Mice, Transgenic , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/radiation effects , Proliferating Cell Nuclear Antigen/metabolism , Protein Kinase C-epsilon/metabolism , STAT3 Transcription Factor/metabolism , Species Specificity
20.
Anticancer Res ; 29(10): 3769-75, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19846907

ABSTRACT

BACKGROUND: Celecoxib and other non-steroidal anti-inflammatory drugs (NSAIDs) are being evaluated in the prevention of bladder and other cancers. Here we investigate molecular effects of celecoxib independent of cyclooxygenase (COX)-2 expression levels in urothelial carcinoma of the bladder. MATERIALS AND METHODS: Low-grade RT-4 and high-grade UM-UC-3 bladder cancer cells were treated with 0-50 muM celecoxib. Growth, cell cycle and apoptosis were measured by crystal violet elution and flow cytometry. Western analysis was performed for COX-2, Rb, cyclin B1/D1, and phospho-cyclin B1/D1. COX-2 induction was achieved with phorbol ester. RESULTS: Celecoxib inhibited growth of RT-4 and UM-UC-3, with G(1) cell cycle arrest and altered cyclin B1/D1 expression in RT-4, whereas Rb up-regulation occurred in UM-UC-3. Apoptosis occurred in both cell lines. CONCLUSION: Celecoxib induces G(1) cell cycle arrest in low- and high-grade bladder cancer by different pathways. This heterogeneous molecular response supports combination approaches to prevention and treatment.


Subject(s)
Cyclin B1/biosynthesis , Cyclin D1/biosynthesis , G1 Phase/drug effects , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Celecoxib , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Cyclooxygenase 2/biosynthesis , Humans , Retinoblastoma Protein/biosynthesis , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...