Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Manag Care Spec Pharm ; 29(3): 245-250, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36840953

ABSTRACT

As high-cost and innovative therapies continue to enter the market, health care decision makers (HCDMs) are expressing a need for early information on a product's clinical and economic impacts. Preapproval information exchange (PIE) fulfills these data needs by allowing manufacturers to share drug information with HCDMs prior to US Food and Drug Administration approval. With recent regulatory milestones, such as the Pre-approval Information Exchange Act of 2022, HCDMs look to leverage PIE to forecast budgets and inform reimbursement decisions. However, a lack of stakeholder alignment has challenged the evolving applications of PIE. In addition, manufacturers are still seeking regulatory clarity regarding best practices, and many are developing their own policies for content dissemination. Varying practices have led to heterogeneity of preapproval communications across manufacturers, which may not fully align with HCDM needs and interests. However, recently collected survey data from FormularyDecisions, which focused on HCDM perceptions of both PIE webinars and other formats of PIE (eg, PIE decks and dossiers), indicate that HCDMs have strong preferences regarding the timing and content shared in preapproval engagements. Additionally, product indication and clinical trial information are highly valued, and although desired by HCDMs in other studies, in FormularyDecisions survey data, exact pricing data do not currently appear to be a critical component of PIE. Preapproval communications are expected less than 1 year before anticipated product approval, and PIE webinars, specifically, should prioritize therapeutic areas and products anticipated to have a significant impact on organizational budgets. Although HCDMs prefer nonmanufacturer representatives for PIE webinars and virtual presentations, health outcome liaisons or medical science liaisons are ideal among manufacturer representatives for in-person preapproval engagements. The expectations of HCDMs should be considered as manufacturers establish PIE practices to ensure the exchange of quality and relevant information. DISCLOSURES: This study was funded by Xcenda. Dr Dodda, Dr Bannister, Dr Hydery, Ms Gorey, Ms Dunlap, and Dr Mody report personal fees from Xcenda during the conduct of the study.


Subject(s)
Delivery of Health Care , Prior Authorization , Humans , Budgets , Surveys and Questionnaires , Decision Making
2.
J Trauma Nurs ; 26(4): 199-207, 2019.
Article in English | MEDLINE | ID: mdl-31283749

ABSTRACT

Trauma is a global health problem and a leading cause of mortality. One of the major predictors of trauma mortality is the Injury Severity Score (ISS). Theoretically, as the ISS increases, the probability of survival decreases; ISS = 75 is considered to be not survivable. Studies have shown that some deaths are preventable and some potentially preventable. Hemorrhagic shock is a potentially preventable cause of trauma mortality. A retrospective database review was conducted of the Mississippi Trauma Registry and point-by-serial correlational analyses were conducted to determine the direction of any significant relations between blood product usage, traditional vital signs, and shock index. Pearson correlation, logistic regressions, and odds ratio calculation results revealed that shock index can signal impending hemorrhagic compromise better than traditional vital signs; thus, facilitating early intervention, specifically, as heart rate and shock index increase, the use of blood products increases, and as blood pressure increases, the use of blood products decreases. Independent t tests for shock index and ISS revealed significant differences in the means with relationship to the subgroups "Dead" and "Alive." Higher ISS were found to correlate with higher shock indices. Evaluation of ISS and survivability demonstrates that ISS = 75 is survivable and should not lead one to reflexively assume otherwise. A total mortality finding of only 1.58% (n = 2,010) was unexpected but very encouraging.


Subject(s)
Injury Severity Score , Multiple Trauma/nursing , Adolescent , Adult , Aged , Aged, 80 and over , Cause of Death , Female , Humans , Logistic Models , Male , Middle Aged , Mississippi , Multiple Trauma/mortality , Registries , Young Adult
3.
Cancer Res ; 77(9): 2500-2511, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28373182

ABSTRACT

The association between obesity and breast cancer risk and prognosis is well established in estrogen receptor (ER)-positive disease but less clear in HER2-positive disease. Here, we report preclinical evidence suggesting weight maintenance through calorie restriction (CR) may limit risk of HER2-positive breast cancer. In female MMTV-HER2/neu transgenic mice, we found that ERα and ERß expression, mammary tumorigenesis, and survival are energy balance dependent in association with epigenetic reprogramming. Mice were randomized to receive a CR, overweight-inducing, or diet-induced obesity regimen (n = 27/group). Subsets of mice (n = 4/group/time point) were euthanized after 1, 3, and 5 months to characterize diet-dependent metabolic, transcriptional, and epigenetic perturbations. Remaining mice were followed up to 22 months. Relative to the overweight and diet-induced obesity regimens, CR decreased body weight, adiposity, and serum metabolic hormones as expected and also elicited an increase in mammary ERα and ERß expression. Increased DNA methylation accompanied this pattern, particularly at CpG dinucleotides located within binding or flanking regions for the transcriptional regulator CCCTC-binding factor of ESR1 and ESR2, consistent with sustained transcriptional activation of ERα and ERß. Mammary expression of the DNA methylation enzyme DNMT1 was stable in CR mice but increased over time in overweight and diet-induced obesity mice, suggesting CR obviates epigenetic alterations concurrent with chronic excess energy intake. In the survival study, CR elicited a significant suppression in spontaneous mammary tumorigenesis. Overall, our findings suggest a mechanistic rationale to prevent or reverse excess body weight as a strategy to reduce HER2-positive breast cancer risk. Cancer Res; 77(9); 2500-11. ©2017 AACR.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Mammary Neoplasms, Animal/genetics , Obesity/genetics , Animals , Breast Neoplasms/physiopathology , Caloric Restriction , Carcinogenesis/genetics , DNA Methylation/genetics , Energy Metabolism/genetics , Epigenesis, Genetic/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Neoplasms, Animal/etiology , Mammary Neoplasms, Animal/physiopathology , Mice , Mice, Transgenic , Obesity/complications , Obesity/physiopathology , Receptor, ErbB-2/genetics , Risk Factors
4.
Cancer Metab ; 4: 18, 2016.
Article in English | MEDLINE | ID: mdl-27651895

ABSTRACT

BACKGROUND: Calorie restriction (CR) prevents obesity and exerts anticancer effects in many preclinical models. CR is also increasingly being used in cancer patients as a sensitizing strategy prior to chemotherapy regimens. While the beneficial effects of CR are widely accepted, the mechanisms through which CR affects tumor growth are incompletely understood. In many cell types, CR and other nutrient stressors can induce autophagy, which provides energy and metabolic substrates critical for cancer cell survival. We hypothesized that limiting extracellular and intracellular substrate availability by combining CR with autophagy inhibition would reduce tumor growth more effectively than either treatment alone. RESULTS: A 30 % CR diet, relative to control diet, in nude mice resulted in significant decreases in body fat, blood glucose, and serum insulin, insulin-like growth factor-1, and leptin levels concurrent with increased adiponectin levels. In a xenograft model in nude mice involving H-Ras(G12V)-transformed immortal baby mouse kidney epithelial cells with (Atg5 (+/+) ) and without (Atg5 (-/-)) autophagic capacity, the CR diet (relative to control diet) genetically induced autophagy inhibition and their combination, each reduced tumor development and growth. Final tumor volume was greatest for Atg5 (+/+) tumors in control-fed mice, intermediate for Atg5 (+/+) tumors in CR-fed mice and Atg5 (-/-) tumors in control-fed mice, and lowest for Atg5 (-/-) tumors in CR mice. In Atg5 (+/+) tumors, autophagic flux was increased in CR-fed relative to control-fed mice, suggesting that the prosurvival effects of autophagy induction may mitigate the tumor suppressive effects of CR. Metabolomic analyses of CR-fed, relative to control-fed, nude mice showed significant decreases in circulating glucose and amino acids and significant increases in ketones, indicating CR induced negative energy balance. Combining glucose deprivation with autophagy deficiency in Atg5 (-/-) cells resulted in significantly reduced in vitro colony formation relative to glucose deprivation or autophagy deficiency alone. CONCLUSIONS: Combined restriction of extracellular (via CR in vivo or glucose deprivation in vitro) and intracellular (via autophagy inhibition) sources of energy and nutrients suppresses Ras-driven tumor growth more effectively than either CR or autophagy deficiency alone. Interventions targeting both systemic energy balance and tumor-cell intrinsic autophagy may represent a novel and effective anticancer strategy.

5.
Diabetes ; 64(5): 1632-42, 2015 May.
Article in English | MEDLINE | ID: mdl-25576058

ABSTRACT

Metformin treatment is associated with a decreased risk and better prognosis of pancreatic cancer (PC) in patients with type 2 diabetes, but the mechanism of metformin's PC growth inhibition in the context of a prediabetic state is unknown. We used a Panc02 pancreatic tumor cell transplant model in diet-induced obese (DIO) C57BL/6 mice to compare the effects of metformin and the direct mammalian target of rapamycin (mTOR) inhibitor rapamycin on PC growth, glucose regulation, mTOR pathway signaling, and candidate microRNA (miR) expression. In DIO/prediabetic mice, metformin and rapamycin significantly reduced pancreatic tumor growth and mTOR-related signaling. The rapamycin effects centered on decreased mTOR-regulated growth and survival signaling, including increased expression of let-7b and cell cycle-regulating miRs. Metformin (but not rapamycin) reduced glucose and insulin levels and expression of miR-34a and its direct targets Notch, Slug, and Snail. Metformin also reduced the number and size of Panc02 tumor spheres in vitro and inhibited the expression of Notch in spheroids. Our results suggest that metformin and rapamycin can both inhibit pancreatic tumor growth in obese, prediabetic mice through shared and distinct mechanisms. Metformin and direct mTOR inhibitors, alone or possibly in combination, represent promising intervention strategies for breaking the diabetes-PC link.


Subject(s)
Metformin/therapeutic use , MicroRNAs/metabolism , Pancreatic Neoplasms/drug therapy , Prediabetic State/drug therapy , Sirolimus/therapeutic use , Animals , Body Weight , Cell Cycle/drug effects , Cell Cycle/physiology , Diet, Diabetic , Energy Intake , Glucose Intolerance , Hypoglycemic Agents/therapeutic use , Immunosuppressive Agents/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Obese , MicroRNAs/genetics , Neoplasms, Experimental/drug therapy , Random Allocation , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Vimentin/genetics , Vimentin/metabolism
6.
Cancer Metab ; 1(1): 10, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-24280167

ABSTRACT

Calorie restriction (CR) is one of the most potent broadly acting dietary interventions for inducing weight loss and for inhibiting cancer in experimental models. Translation of the mechanistic lessons learned from research on CR to cancer prevention strategies in human beings is important given the high prevalence of excess energy intake, obesity, and metabolic syndrome in many parts of the world and the established links between obesity-associated metabolic perturbations and increased risk or progression of many types of cancer. This review synthesizes findings on the biological mechanisms underlying many of the anticancer effects of CR, with emphasis on the impact of CR on growth factor signaling pathways, inflammation, cellular and systemic energy homeostasis pathways, vascular perturbations, and the tumor microenvironment. These CR-responsive pathways and processes represent targets for translating CR research into effective cancer prevention strategies in human beings.

7.
Endocr Relat Cancer ; 20(6): 797-808, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24025407

ABSTRACT

Despite new therapies, breast cancer continues to be the second leading cause of cancer mortality in women, a consequence of recurrence and metastasis. In recent years, a population of cancer cells has been identified, called cancer stem cells (CSCs) with self-renewal capacity, proposed to underlie tumor recurrence and metastasis. We previously showed that the adipose tissue cytokine LEPTIN, increased in obesity, promotes the survival of CSCs in vivo. Here, we tested the hypothesis that the leptin receptor (LEPR), expressed in mammary cancer cells, is necessary for maintaining CSC-like and metastatic properties. We silenced LEPR via shRNA lentivirus transduction and determined that the expression of stem cell self-renewal transcription factors NANOG, SOX2, and OCT4 (POU5F1) is inhibited. LEPR-NANOG signaling pathway is conserved between species because we can rescue NANOG expression in human LEPR-silenced cells with the mouse LepR. Using a NANOG promoter GFP reporter, we showed that LEPR is enriched in NANOG promoter active (GFP+) cells. In lineage tracing studies, we showed that the GFP+ cells divide in a symmetric and asymmetric manner. LEPR-silenced MDA-MB-231 cells exhibit a mesenchymal to epithelial transition morphologically, increased E-CADHERIN and decreased VIMENTIN expression compared with control cells. Finally, LEPR-silenced cells exhibit reduced cell proliferation, self-renewal in tumor sphere assays, and tumor outgrowth in xenotransplant studies. Given the emergence of NANOG as a pro-carcinogenic protein in multiple cancers, these studies suggest that inhibition of LEPR may be a promising therapeutic approach to inhibit NANOG and thereby neutralize CSC functions.


Subject(s)
Neoplastic Stem Cells/pathology , Receptors, Leptin/metabolism , Triple Negative Breast Neoplasms/pathology , Animals , Blotting, Western , Cell Proliferation , Female , Flow Cytometry , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mesoderm/metabolism , Mesoderm/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nanog Homeobox Protein , Neoplastic Stem Cells/metabolism , Phosphorylation , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptors, Leptin/antagonists & inhibitors , Receptors, Leptin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
8.
PLoS One ; 8(6): e68089, 2013.
Article in English | MEDLINE | ID: mdl-23840816

ABSTRACT

Obesity, prevalent in >35% of US women, is an established risk and progression factor for postmenopausal breast cancer, and strategies to break the obesity-breast cancer link are urgently needed. Approximately 30% of breast cancers carry p53 tumor suppressor gene alterations; however, the effects of obesity on breast cancer progression in relation to p53 gene dosage are unclear. Using murine models of postmenopausal breast cancer, we characterized the interactive effects of diet-induced obesity (DIO) and p53 gene dosage on mammary tumor growth and associated p53-related regulatory mechanisms. Ovariectomized C57BL/6 mice were randomly assigned to receive a DIO or control diet, and (at 10 weeks) orthotopic injection of MMTV-Wnt-1 p53(+/-) or MMTV-Wnt-1 p53(+/+) mammary tumor cells (n = 20 mice per diet and genotype group). DIO and control diets produced distinct phenotypes (mean percent body fat at 10 weeks: 57% and 39%, respectively, P<0.001). Regardless of phenotype, time to first palpable tumor was 57% less for Wnt-1 p53(+/-) than Wnt-1 p53(+/+) tumors. Regardless of tumoral p53 genotype, DIO (relative to control) increased tumor burden, tumor cell proliferation (Ki-67), severity of tumor pathology, local tissue invasion, epithelial-to-mesenchymal transition (EMT) programming, and tumoral microRNA-504 (a negative regulator of p53) expression; and suppressed p53, p21, and estrogen receptor-alpha protein expression. These findings in murine models of postmenopausal breast cancer suggest that obesity may augment procancer effects related to p53 gene alterations. Furthermore, microRNA-504, an obesity-responsive negative regulator of p53 and putative EMT regulator, may represent a novel molecular target for breaking the obesity-breast cancer link.


Subject(s)
Gene Dosage/genetics , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , MicroRNAs/genetics , Obesity/complications , Tumor Suppressor Protein p53/genetics , Up-Regulation/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation , Diet , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Estrogen Receptor alpha/genetics , Female , Mice , Mice, Inbred C57BL , Obesity/genetics , Wnt1 Protein/genetics
9.
Cancer Biol Ther ; 14(4): 357-64, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23358472

ABSTRACT

Obesity is associated with increased breast tumor aggressiveness and decreased response to multiple modalities of therapy in postmenopausal women. Delivering cancer chemotherapeutic drugs using nanoparticles has evolved as a promising approach to improve the efficacy of anticancer agents. However, the application of nanoparticles in cancer chemotherapy in the context of obesity has not been studied before. The nucleoside analog gemcitabine is widely used in solid tumor therapy. Previously, we developed a novel stearoyl gemcitabine solid-lipid nanoparticle formulation (GemC18-NPs) and showed that the GemC18-NPs are significantly more effective than gemcitabine in controlling tumor growth in mouse models. In the present study, using ovariectomized diet-induced obese female C57BL/6 mice with orthotopically transplanted MMTV-Wnt-1 mammary tumors as a model of postmenopausal obesity and breast cancer, we discovered that obesity induces tumor cell resistance to gemcitabine. Furthermore, our GemC18-NPs can overcome the obesity-related resistance to gemcitabine chemotherapy. These findings have important clinical implications for cancer chemotherapies involving gemcitabine or other nucleoside analogs in the context of obesity.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/physiopathology , Nanoparticles/administration & dosage , Obesity/physiopathology , Animals , Cell Line, Tumor , Deoxycytidine/pharmacology , Disease Models, Animal , Drug Resistance, Neoplasm , Female , Mice , Mice, Inbred C57BL , Postmenopause , Random Allocation , Gemcitabine
10.
Ann N Y Acad Sci ; 1271: 82-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23050968

ABSTRACT

Obesity is an established risk and progression factor for many cancers. In the United States more than one-third of adults, and nearly one in five children, are currently obese. Thus, a better understanding of the mechanistic links between obesity and cancer is urgently needed to identify intervention targets and strategies to offset the procancer effects of obesity. This review synthesizes the evidence on key biological mechanisms underlying the obesity-cancer association, with particular emphasis on obesity-associated enhancements in growth factor signaling, inflammation, and perturbations in the tumor microenvironment. These interrelated pathways and processes represent mechanistic targets for disrupting the obesity-cancer link.


Subject(s)
Metabolic Diseases/epidemiology , Metabolic Diseases/immunology , Neoplasms/epidemiology , Neoplasms/immunology , Obesity/epidemiology , Obesity/immunology , Tumor Microenvironment , Animals , Comorbidity , Epithelial-Mesenchymal Transition , Humans , Metabolic Diseases/etiology , Metabolic Diseases/metabolism , Neoplasm Proteins/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Obesity/metabolism , Obesity/physiopathology , Risk
11.
Cancer Prev Res (Phila) ; 5(7): 930-42, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22588949

ABSTRACT

Using novel murine models of claudin-low and basal-like breast cancer, we tested the hypothesis that diet-induced obesity (DIO) and calorie restriction (CR) differentially modulate progression of these aggressive breast cancer subtypes. For model development, we characterized two cell lines, "mesenchymal (M)-Wnt" and "epithelial (E)-Wnt," derived from MMTV-Wnt-1 transgenic mouse mammary tumors. M-Wnt, relative to E-Wnt, cells were tumor-initiating cell (TIC)-enriched (62% vs. 2.4% CD44(high)/CD24(low)) and displayed enhanced ALDEFLUOR positivity, epithelial-to-mesenchymal transition (EMT) marker expression, mammosphere-forming ability, migration, invasion, and tumorigenicity (P < 0.001; each parameter). M-Wnt and E-Wnt cells clustered with claudin-low and basal-like breast tumors, respectively, in gene expression profiles and recapitulated these tumors when orthotopically transplanted into ovariectomized C57BL/6 mice. To assess the effects of energy balance interventions on tumor progression and EMT, mice were administered DIO, control, or CR diets for 8 weeks before orthotopic transplantation of M-Wnt or E-Wnt cells (for each cell line, n = 20 mice per diet) and continued on their diets for 6 weeks while tumor growth was monitored. Relative to control, DIO enhanced M-Wnt (P = 0.01), but not E-Wnt, tumor progression; upregulated EMT- and TIC-associated markers including N-cadherin,fibronectin, TGFß, Snail, FOXC2, and Oct4 (P < 0.05, each); and increased intratumoral adipocytes. Conversely, CR suppressed M-Wnt and E-Wnt tumor progression (P < 0.02, each) and inhibited EMT and intratumoral adipocyte accumulation. Thus, dietary energy balance interventions differentially modulate EMT and progression of claudin-low and basal-like tumors. EMT pathway components may represent targets for breaking the obesity-breast cancer link, particularly for preventing and/or controlling TIC-enriched subtypes such as claudin-low breast cancer.


Subject(s)
Carcinoma, Basal Cell/pathology , Claudins/deficiency , Diet , Energy Metabolism , Epithelial-Mesenchymal Transition , Mammary Neoplasms, Animal/pathology , Neoplastic Stem Cells/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Basal Cell/etiology , Carcinoma, Basal Cell/metabolism , Cell Adhesion , Cell Differentiation , Cell Movement , Cell Proliferation , Disease Progression , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Mammary Neoplasms, Animal/etiology , Mammary Neoplasms, Animal/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Ovariectomy , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Wnt Proteins/genetics , Wnt Proteins/metabolism
12.
Proc Natl Acad Sci U S A ; 109(9): 3475-80, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22345562

ABSTRACT

Insulin-like growth factor-binding protein 2 (IGFBP2) is increasingly recognized as a glioma oncogene, emerging as a target for therapeutic intervention. In this study, we used an integrative approach to characterizing the IGFBP2 network, combining transcriptional profiling of human glioma with validation in glial cells and the replication-competent ASLV long terminal repeat with a splice acceptor/tv-a glioma mouse system. We demonstrated that IGFBP2 expression is closely linked to genes in the integrin and integrin-linked kinase (ILK) pathways and that these genes are associated with prognosis. We further showed that IGFBP2 activates integrin ß1 and downstream invasion pathways, requires ILK to induce cell motility, and activates NF-κB. Most significantly, the IGFBP2/integrin/ILK/NF-κB network functions as a physiologically active signaling pathway in vivo by driving glioma progression; interfering with any point in the pathway markedly inhibits progression. The results of this study reveal a signaling pathway that is both targetable and highly relevant to improving the survival of glioma patients.


Subject(s)
Brain Neoplasms/pathology , Genetic Therapy , Genetic Vectors/therapeutic use , Glioblastoma/pathology , Insulin-Like Growth Factor Binding Protein 2/physiology , Integrin beta1/physiology , NF-kappa B/physiology , Neoplasm Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Astrocytoma/genetics , Astrocytoma/metabolism , Avian Proteins/genetics , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic , Genes, Synthetic , Genes, sis , Genetic Vectors/administration & dosage , Glioblastoma/genetics , Glioblastoma/therapy , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/toxicity , Insulin-Like Growth Factor Binding Protein 2/biosynthesis , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 2/toxicity , Intermediate Filament Proteins/genetics , Kaplan-Meier Estimate , Mice , Mice, Transgenic , NF-KappaB Inhibitor alpha , Neoplasm Invasiveness , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Nestin , Oligodendroglioma/genetics , Oligodendroglioma/metabolism , Prognosis , Protein Serine-Threonine Kinases/toxicity , Receptors, Virus/genetics , Retroviridae , Signal Transduction/physiology
13.
Alcohol Clin Exp Res ; 36(4): 577-87, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21981381

ABSTRACT

BACKGROUND: Alcohol consumption is an established risk factor for breast cancer. Yet, the mechanism by which alcohol affects breast cancer development remains unresolved. The transition from the premenopausal to the postmenopausal phase is associated with a drastic reduction in systemic estrogen levels. It is not clear whether the risk of breast cancer attributable to alcohol consumption is modified by the different levels of estrogen found in pre- and postmenopausal women. The objective of this study is to determine whether the effects of alcohol on mammary tumor development are dependent on the presence of ovarian estrogen. METHODS: As a model of breast cancer, we used mouse mammary tumor virus (MMTV)-neu transgenic mice that overexpress the human epidermal growth factor receptor 2 (HER2/neu) in the mammary epithelium, resulting in the development of estrogen receptor alpha (ERα)-negative mammary tumors. The mammary tumorigenesis process in these mice is similar to that of patients with HER2 breast cancer. Nonovariectomized (NOVX) and ovariectomized (OVX) MMTV-neu mice were exposed to 0, 5, and 20% ethanol in the drinking water. Breast cancer development and progression were determined alongside the effects of alcohol on estrogen availability and signaling. RESULTS: Our data show that 20% alcohol consumption promoted tumor development in MMTV-neu mice only in the presence of ovarian hormones. Tumor promotion was associated with increased systemic estrogen levels, increased expression of aromatase (the rate-limiting enzyme in estrogen synthesis), and increased expression of ERα in the tumors of 20% alcohol-consuming MMTV-neu mice. Additionally, we show that ovariectomy (removal of the ovaries and ovarian hormone production) blocked the effects of 20% alcohol on tumor development. CONCLUSIONS: Our results support the notion that alcohol consumption promotes HER2 breast cancer development via the estrogen signaling pathway. Additionally, they suggest that the effects of alcohol on breast cancer may be prevented by blocking estrogen signaling.


Subject(s)
Central Nervous System Depressants/toxicity , Estrogen Receptor alpha/genetics , Estrogens/physiology , Ethanol/toxicity , Mammary Neoplasms, Experimental/pathology , Receptor, ErbB-2/genetics , Signal Transduction/drug effects , Adiposity/drug effects , Animals , Aromatase/metabolism , Blotting, Western , Body Composition/physiology , Body Weight/drug effects , Central Nervous System Depressants/blood , Ethanol/blood , Female , Immunohistochemistry , Mammary Tumor Virus, Mouse , Mice , Ovariectomy , Receptor, IGF Type 1/biosynthesis , Receptor, IGF Type 1/genetics , Retroviridae Infections/pathology , Tumor Virus Infections/pathology
14.
Endocr Relat Cancer ; 19(1): 57-68, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22143497

ABSTRACT

Obesity is an established risk and progression factor for postmenopausal breast cancer. Interventions to decrease caloric intake and/or increase energy expenditure beneficially impact tumor progression in normoweight humans and animal models. However, despite the increasingly high global prevalence of obesity, the effects and underlying mechanisms of these energy balance modulating interventions are poorly characterized in obese individuals. The goal of this study was to better characterize the mechanism(s) responsible for the link between energy balance and breast cancer progression in the postmenopausal obesity context. We compared the effects of calorie restriction (CR), treadmill exercise (EX), and mammalian target of rapamycin (mTOR inhibitor) treatment on body composition, serum biomarkers, cellular signaling, and mammary tumor growth in obese mice. Ovariectomized C57BL/6 mice were administered a diet-induced obesity regimen for 8 weeks, then randomized into three treatment groups: control (semipurified diet fed ad libitum, maintained the obese state); 30% CR (isonutrient relative to control except 30% reduction in carbohydrate calories); and EX (control diet fed ad libitum plus treadmill exercise). Mice were implanted with syngeneic MMTV-Wnt-1 mammary tumor cells at week 12. Rapamycin treatment (5  mg/kg every 48  h) started at week 14. Tumors were excised at week 18. CR and rapamycin (but not EX) significantly reduced final tumor weight compared to control. In follow-up analysis, constitutive activation of mTOR ablated the inhibitory effects of CR on Wnt-1 mammary tumor growth. We conclude that mTOR inhibition may be a pharmacologic strategy to mimic the anticancer effects of CR and break the obesity-breast cancer progression link.


Subject(s)
Caloric Restriction , Disease Models, Animal , Mammary Neoplasms, Animal/prevention & control , Mammary Tumor Virus, Mouse/genetics , Obesity/prevention & control , Postmenopause , Sirolimus/therapeutic use , Wnt1 Protein/genetics , Animals , Antibiotics, Antineoplastic/therapeutic use , Blotting, Western , Cell Cycle , Cell Proliferation , Energy Intake , Female , Mammary Neoplasms, Animal/etiology , Mammary Neoplasms, Animal/mortality , Mice , Mice, Inbred C57BL , Obesity/complications , Obesity/mortality , Ovariectomy , Physical Conditioning, Animal , Survival Rate , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
15.
Endocr Relat Cancer ; 18(4): 491-503, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21636700

ABSTRACT

Obesity increases both the risk and mortality associated with many types of cancer including that of the breast. In mice, obesity increases both incidence of spontaneous tumors and burden of transplanted tumors. Our findings identify leptin, an adipose secreted cytokine, in promoting increased mammary tumor burden in obese mice and provide a link between this adipokine and cancer. Using a transplantable tumor that develops spontaneously in the murine mammary tumor virus-Wnt-1 transgenic mice, we show that tumors transplanted into obese leptin receptor (LepRb)-deficient (db/db) mice grow to eight times the volume of tumors transplanted into lean wild-type (WT) mice. However, tumor outgrowth and overall tumor burden is reduced in obese, leptin-deficient (ob/ob) mice. The residual tumors in ob/ob mice contain fewer undifferentiated tumor cells (keratin 6 immunopositive) compared with WT or db/db mice. Furthermore, tumors in ob/ob mice contain fewer cells expressing phosphorylated Akt, a growth promoting kinase activated by the LepRb, compared with WT and db/db mice. In vivo limiting dilution analysis of residual tumors from ob/ob mice indicated reduced tumor initiating activity suggesting fewer cancer stem cells (CSCs). The tumor cell populations reduced by leptin deficiency were identified by fluorescence-activated cell sorting and found to express LepRb. Finally, LepRb expressing tumor cells exhibit stem cell characteristics based on the ability to form tumorspheres in vitro and leptin promotes their survival. These studies provide critical new insight on the role of leptin in tumor growth and implicate LepRb as a CSC target.


Subject(s)
Carcinoma/pathology , Cell Proliferation , Leptin/deficiency , Mammary Neoplasms, Animal/pathology , Mammary Tumor Virus, Mouse/genetics , Neoplastic Stem Cells/physiology , Wnt1 Protein/genetics , Animals , Carcinoma/complications , Carcinoma/genetics , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Down-Regulation/physiology , Female , Leptin/genetics , Leptin/physiology , Mammary Neoplasms, Animal/complications , Mammary Neoplasms, Animal/genetics , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Obesity/complications , Obesity/genetics , Receptors, Leptin/genetics , Receptors, Leptin/physiology
16.
Proc Natl Acad Sci U S A ; 104(28): 11736-41, 2007 Jul 10.
Article in English | MEDLINE | ID: mdl-17606927

ABSTRACT

Overexpression of insulin-like growth factor binding protein 2 (IGFBP2) is associated with progression in many types of human cancer. In this study we used a glial-specific transgenic mouse model to examine the active role of IGFBP2 in tumorigenesis and progression. Our studies show that IGFBP2 coexpression results in progression to a higher-grade glioma in platelet-derived growth factor beta (PDGFB)-driven tumors. These anaplastic oligodendrogliomas are characterized by increased cellularity, vascular proliferation, small regions of necrosis, increased mitotic activity, and increased activation of the Akt pathway. Combined expression of IGFBP2 or Akt with K-Ras was required to form astrocytomas, indicating that activation of two separate pathways is necessary for gliomagenesis. In ex vivo experiments, blockade of Akt by an inhibitor led to decreased viability of cells coexpressing IGFBP2 versus PDGFB expression alone. Thus, this study provides definitive evidence that IGFBP2 plays a key role in activation of the Akt pathway and collaborates with K-Ras or PDGFB in the development and progression of two major types of glioma.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Glioma/metabolism , Glioma/pathology , Insulin-Like Growth Factor Binding Protein 2/physiology , Animals , Animals, Newborn , Astrocytoma/genetics , Astrocytoma/pathology , Cell Line, Transformed , Cell Survival/physiology , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Chickens , Disease Progression , Glioma/genetics , Humans , Insulin-Like Growth Factor Binding Protein 2/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , ras Proteins/physiology
17.
Cancer Biol Ther ; 5(3): 300-4, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16410724

ABSTRACT

Allelic loss of the short arm of chromosome 1 has been observed frequently in oligodendroglioma (60-80%). We evaluated 177 oligodendroglial tumor samples and defined a consensus region of deletion of approximately 630 kb. This region contains a single gene, SHREW1, which encodes a novel transmembrane protein in adherens junctions. Whereas a mutation was not detected in the coding region of the SHREW1 gene in oligodendrogliomas, restoration of SHREW1 expression resulted in suppression of cell adhesion and migration. Thus, SHREW1 inactivation may play a role in the development of oligodendroglial tumors.


Subject(s)
Cell Adhesion , Cell Movement , Genes, Tumor Suppressor , Membrane Proteins/genetics , Oligodendroglioma/genetics , Oligodendroglioma/pathology , Cell Adhesion Molecules , Gene Deletion , Humans , Membrane Proteins/physiology , Microsatellite Repeats , Mutation , Polymerase Chain Reaction , Transfection
18.
Cancer ; 106(3): 566-75, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16369983

ABSTRACT

BACKGROUND: Cyclooxygenase-2 (COX-2) plays a major role in the development of cancer through numerous mechanisms. COX-2 is expressed in the majority of renal cell carcinoma (RCC) tumors and correlates with stage, grade, and microvessel density. Based on potential additive or synergistic antitumor effects, interferon-alpha (IFNalpha) and celecoxib, an oral COX-2 inhibitor, were given to metastatic RCC patients in a Phase II trial. METHODS: Patients with untreated, metastatic RCC received IFNalpha 3 million units (MU) daily and celecoxib 400 mg orally (p.o.) twice daily continuously until disease progression or unacceptable toxicity. Pretreatment, paraffin-embedded RCC tumor samples were immunohistochemically stained for COX-2 expression and plasma basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) levels were assayed to determine predictive or prognostic potential. RESULTS: There were three partial responses among 25 patients treated (objective response rate, 12%; 95% confidence interval [CI], 3-31%). The observed median time to disease progression (TTP) for the entire cohort was 3.3 months. A significant association between maximal COX-2 staining and clinical response was observed: all patients who experienced an objective response demonstrated 3+ COX-2 tumor immunostaining (trend test: P=0.03). Therapy was well tolerated without cardiac or other notable toxicity. CONCLUSIONS: The addition of celecoxib to IFNalpha did not increase the objective response rate or TTP of this unselected cohort. Maximal COX-2 tumor immunostaining may identify RCC patents more likely to achieve clinical benefit with COX-2 inhibition in combination with IFNalpha. Further investigation of this combination in 3+ COX-2-overexpressing RCC tumors is warranted.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Cyclooxygenase 2/biosynthesis , Cyclooxygenase Inhibitors/therapeutic use , Interferon-alpha/therapeutic use , Kidney Neoplasms/drug therapy , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , Administration, Oral , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Celecoxib , Cyclooxygenase 2/analysis , Disease Progression , Female , Fibroblast Growth Factor 2/analysis , Humans , Immunohistochemistry , Kidney Neoplasms/enzymology , Kidney Neoplasms/pathology , Male , Middle Aged , Predictive Value of Tests , Prognosis , Treatment Outcome , Vascular Endothelial Growth Factor A/analysis
19.
J Am Acad Child Adolesc Psychiatry ; 44(3): 265-73, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15725971

ABSTRACT

OBJECTIVE: The current study examined how self-reported maternal stress and distress are associated with child disruptive behaviors. METHOD: Mother and teacher ratings of child disruptive behavior problems (attention problems, aggression, and delinquency) were collected for 215 male participants, ranging in age from 9 to 12 years. Participating mothers also provided self-report data on socioeconomic status (SES), parenting stress, and distress (depression and anxiety/somatization). RESULTS: Low SES was significantly associated with both mother- and teacher-reported child disruptive behavior problems. Regression analyses indicated a relation between parenting stress and mother-reported child disruptive behavior problems, even when controlling for SES. Results also indicated a significant relation between maternal distress and mother-reported child disruptive behavior problems (particularly attention problems), even when controlling for SES and parenting stress. Maternal stress and distress were not significantly related to teacher-reported child disruptive behavior problems. CONCLUSIONS: Although the lack of an association between teacher-reported behavior problems and maternal stress and distress could be interpreted as a rater bias by these mothers, it may be that the mothers' symptoms are associated with a stressful home environment, thus exacerbating child disruptive behavior problems and eventually leading to a reciprocal relation between symptomatology in mothers and children.


Subject(s)
Attention Deficit and Disruptive Behavior Disorders/psychology , Depressive Disorder, Major/epidemiology , Depressive Disorder, Major/psychology , Mothers/psychology , Stress, Psychological/epidemiology , Stress, Psychological/psychology , Adult , Attention Deficit and Disruptive Behavior Disorders/diagnosis , Attention Deficit and Disruptive Behavior Disorders/epidemiology , Child , Cognition Disorders/diagnosis , Cognition Disorders/epidemiology , Depressive Disorder, Major/diagnosis , Female , Humans , Male , Observer Variation , Severity of Illness Index , Stress, Psychological/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL
...