Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Breast Cancer Res ; 13(5): R98, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21999244

ABSTRACT

INTRODUCTION: Breast Cancer Index (BCI) combines two independent biomarkers, HOXB13:IL17BR (H:I) and the 5-gene molecular grade index (MGI), that assess estrogen-mediated signalling and tumor grade, respectively. BCI stratifies early-stage estrogen-receptor positive (ER+), lymph-node negative (LN-) breast cancer patients into three risk groups and provides a continuous assessment of individual risk of distant recurrence. Objectives of the current study were to validate BCI in a clinical case series and to compare the prognostic utility of BCI and Adjuvant!Online (AO). METHODS: Tumor samples from 265 ER+LN- tamoxifen-treated patients were identified from a single academic institution's cancer research registry. The BCI assay was performed and scores were assigned based on a pre-determined risk model. Risk was assessed by BCI and AO and correlated to clinical outcomes in the patient cohort. RESULTS: BCI was a significant predictor of outcome in a cohort of 265 ER+LN- patients (median age: 56-y; median follow-up: 10.3-y), treated with adjuvant tamoxifen alone or tamoxifen with chemotherapy (32%). BCI categorized 55%, 21%, and 24% of patients as low, intermediate and high-risk, respectively. The 10-year rates of distant recurrence were 6.6%, 12.1% and 31.9% and of breast cancer-specific mortality were 3.8%, 3.6% and 22.1% in low, intermediate, and high-risk groups, respectively. In a multivariate analysis including clinicopathological factors, BCI was a significant predictor of distant recurrence (HR for 5-unit increase = 5.32 [CI 2.18-13.01; P = 0.0002]) and breast cancer-specific mortality (HR for a 5-unit increase = 9.60 [CI 3.20-28.80; P < 0.0001]). AO was significantly associated with risk of recurrence. In a separate multivariate analysis, both BCI and AO were significantly predictive of outcome. In a time-dependent (10-y) ROC curve accuracy analysis of recurrence risk, the addition of BCI+AO increased predictive accuracy in all patients from 66% (AO only) to 76% (AO+BCI) and in tamoxifen-only treated patients from 65% to 81%. CONCLUSIONS: This study validates the prognostic performance of BCI in ER+LN- patients. In this characteristically low-risk cohort, BCI classified high versus low-risk groups with ~5-fold difference in 10-year risk of distant recurrence and breast cancer-specific death. BCI and AO are independent predictors with BCI having additive utility beyond standard of care parameters that are encompassed in AO.


Subject(s)
Breast Neoplasms/mortality , Breast Neoplasms/pathology , Online Systems , Adult , Aged , Aged, 80 and over , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cohort Studies , Female , Follow-Up Studies , GTPase-Activating Proteins/genetics , Homeodomain Proteins/genetics , Humans , Middle Aged , NIMA-Related Kinases , Neoplasm Recurrence, Local/drug therapy , Prognosis , Proportional Hazards Models , Protein Serine-Threonine Kinases/genetics , Receptors, Interleukin/genetics , Receptors, Interleukin-17 , Survival Rate , Tamoxifen/therapeutic use
2.
J Mol Diagn ; 13(5): 493-503, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21708287

ABSTRACT

Accurate determination of cancer origin is necessary to guide optimal treatment but remains a diagnostic challenge. Gene expression profiling technologies have aided the classification of tumors and, therefore, could be applied in conjunction with clinicopathologic correlates to improve accuracy. We report an expanded version of the previously described 92-gene assay to classify 30 main tumor types and 54 histological subtypes, with coverage of ≥95% of all solid tumors based on incidence. Increased tissue coverage was achieved through expansion of a reference tumor database containing 2,206 specimens, with a median of 62 samples per main tumor type. The 92-gene classification algorithm demonstrated sensitivities of 87% and 85% for 30 main types and 54 histological subtypes, respectively, in leave-one-out cross validation, and 83% in a test set of 187 tumors representing 28 of the 30 main cancer types. These findings provide further support that broad and diverse tumor classification can be performed using a relatively compact gene set. An additional 300 consecutive cases submitted for clinical testing were profiled to characterize clinical utility in a real-world setting: the 92-gene assay confirmed 78% of samples having a single suspected primary tumor and provided a single molecular prediction in 74% of cases with two or more differential diagnoses. Further development of the 92-gene RT-PCR assay has resulted in a significant expansion in reportable tumor types and histological features with strong performance characteristics and supports the use of molecular classification as an objective standardized adjunct to current methods.


Subject(s)
Genes, Neoplasm/genetics , Neoplasms/classification , Neoplasms/genetics , Real-Time Polymerase Chain Reaction/methods , Real-Time Polymerase Chain Reaction/standards , Algorithms , Databases, Genetic , Female , Humans , Male , Middle Aged , Reference Standards , Reproducibility of Results
3.
Expert Opin Biol Ther ; 8(11): 1733-47, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18847308

ABSTRACT

BACKGROUND: The integrated central actions of hormones secreted from pancreatic islets, the gut and adipocytes regulate both energy homeostasis and body weight. Dysregulation in these neurohormonal pathways probably contributes to pathogenesis of obesity and type 2 diabetes. OBJECTIVE: To examine hormone-based therapies targeting these interrelated pathways as potential treatments for obesity and diabetes. METHODS: Preclinical and clinical data on therapies based on hormones secreted from the pancreas (glucagon, insulin, amylin and pancreatic polypeptide), gut (glucagon-like peptide-1, glucose dependent insulinotropic polypeptide, cholecystokinin and peptide YY) and adipose tissue (leptin and adiponectin) as potential treatments for diabetes and obesity are reviewed. RESULTS/CONCLUSIONS: In diabetes, hormone-based treatments have translated into new clinical platforms including insulin analogs, the GLP-1-like peptide receptor agonist exenatide and amylinomimetic pramlintide, which due to their complex interplay and the progressive nature of diabetes, can be utilized in different settings. Various peptide hormones and agonists/antagonists are currently under investigation as new approaches to treatment of obesity and diabetes.


Subject(s)
Body Weight , Energy Metabolism , Hormones/metabolism , Adipocytes/metabolism , Adiponectin/metabolism , Adipose Tissue/metabolism , Amyloid/pharmacology , Animals , Diabetes Mellitus, Type 2/metabolism , Glucagon-Like Peptide 1/chemistry , Homeostasis , Humans , Intestinal Mucosa/metabolism , Islet Amyloid Polypeptide , Leptin/metabolism , Mice , Obesity/metabolism , Pancreas/metabolism
4.
Diabetes Care ; 31(9): 1816-23, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18753666

ABSTRACT

OBJECTIVE: To assess long-term weight loss efficacy and safety of pramlintide used at different dosing regimens and in conjunction with lifestyle intervention (LSI). RESEARCH DESIGN AND METHODS: In a 4-month, double-blind, placebo-controlled, dose-ranging study, 411 obese subjects were randomized to receive pramlintide (six arms: 120, 240, and 360 microg b.i.d. and t.i.d.) or placebo in conjunction with a structured LSI program geared toward weight loss. Of the 4-month evaluable subjects (n = 270), 77% opted to continue preexisting treatment during an 8-month single-blind extension (LSI geared toward weight maintenance). RESULTS: At month 4, mean weight loss from baseline in the pramlintide arms ranged from 3.8 +/- 0.7 to 6.1 +/- 0.8 kg (2.8 +/- 0.8 kg with placebo). By month 12, initial 4-month weight loss was regained in the placebo group but was maintained in all but the 120-microg b.i.d. group. Placebo-corrected weight loss with 120 microg t.i.d. and 360 microg b.i.d. averaged 3.2 +/- 1.2 kg (3.1 +/- 1.1% body wt) and 3.3 +/- 1.1 kg (3.1 +/- 1.0% body wt), respectively, at month 4 (both P < 0.01; 4-month evaluable n = 270) and 6.1 +/- 2.1 kg (5.6 +/- 2.1% body wt) and 7.2 +/- 2.3 kg (6.8 +/- 2.3% body wt), respectively, at month 12 (both P < 0.01; 12-month evaluable n = 146). At month 12, 40 and 43% of subjects treated with 120 microg t.i.d. and 360 microg b.i.d., respectively, achieved >or=10% weight loss (vs. 12% for placebo). Nausea, the most common adverse event with pramlintide in the 4-month study (9-29% pramlintide vs. 2% placebo), was generally mild to moderate and occurred in <10% of subjects during the extension. CONCLUSIONS: When used over 12 months as an adjunct to LSI, pramlintide treatment, with low-dose three-times-daily or higher-dose two-times-daily regimens, helped obese subjects achieve greater initial weight loss and enhanced long-term maintenance of weight loss.


Subject(s)
Amyloid/therapeutic use , Hypoglycemic Agents/therapeutic use , Life Style , Weight Loss/drug effects , Adult , Body Mass Index , Body Size , Double-Blind Method , Female , Follow-Up Studies , Humans , Islet Amyloid Polypeptide , Male , Middle Aged , Placebos , Single-Blind Method , Time Factors
5.
J Clin Endocrinol Metab ; 92(8): 2977-83, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17504894

ABSTRACT

CONTEXT: In previous 1-yr trials, treatment with pramlintide (120 microg), an analog of the beta-cell hormone amylin, induced sustained reductions in A1C and body weight in insulin-using subjects with type 2 diabetes. OBJECTIVE: To assess the potential of pramlintide as an antiobesity agent, we assessed the weight effect, safety, and tolerability of pramlintide in non-insulin-treated obese subjects with and without type 2 diabetes at doses greater than previously studied. DESIGN/SETTING: We performed a randomized, double-blind, placebo-controlled, multicenter study. PATIENTS: A total of 204 obese subjects [80/20% female/male, age 48 +/- 10 yr, and body mass index 37.8 +/- 5.6 kg/m(2) (mean +/- SD)] participated in the study. INTERVENTION: For 16 wk, without concomitant lifestyle intervention, subjects self-administered pramlintide (nonforced dose escalation < or = 240 microg) or placebo via sc injection three times a day before meals. MAIN OUTCOME MEASURES: Weight, waist circumference, tolerability, and safety were the main outcome measures. RESULTS: Pramlintide was generally well tolerated, with 88% of subjects able to escalate to the maximum dose of 240 microg. Withdrawal rates were similar between placebo (25%) and pramlintide-treated subjects (29%). Subjects completing 16 wk of pramlintide treatment experienced placebo-corrected reductions in body weight of 3.7 +/- 0.5% (3.6 +/- 0.6 kg; P < 0.001) and waist circumference (3.6 +/- 1.1 cm; P < 0.01). Approximately 31% of pramlintide-treated subjects achieved > or =5% weight loss (vs. 2% placebo; P < 0.001). More pramlintide than placebo-treated subjects reported improvements in appetite control (72% vs. 31%), weight control (63% vs. 24%), and overall well-being (52% vs. 17%). No unexpected safety signals were observed. The most common adverse event reported was mild, transient nausea. Pramlintide-treated subjects not reporting nausea experienced weight loss similar to those who did (3.6 +/- 0.5% and 3.9 +/- 0.5%, respectively). CONCLUSION: These results support continued evaluation of pramlintide as a potential treatment for obesity.


Subject(s)
Amyloid/therapeutic use , Anti-Obesity Agents/therapeutic use , Body Weight/drug effects , Hypoglycemic Agents/therapeutic use , Obesity/drug therapy , Adult , Amyloid/adverse effects , Anthropometry , Anti-Obesity Agents/adverse effects , Dose-Response Relationship, Drug , Double-Blind Method , Female , Glycated Hemoglobin/metabolism , Humans , Hypoglycemic Agents/adverse effects , Islet Amyloid Polypeptide , Male , Middle Aged , Surveys and Questionnaires , Treatment Outcome
6.
Am J Physiol Endocrinol Metab ; 293(2): E620-7, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17505051

ABSTRACT

Evidence from rodent studies indicates that the beta-cell-derived neurohormone amylin exerts multiple effects on eating behavior, including reductions in meal size, intake of highly palatable foods, and stress-induced sucrose consumption. To assess the effect of amylin agonism on human eating behavior we conducted a randomized, blinded, placebo-controlled, multicenter study investigating the effects of the amylin analog pramlintide on body weight, 24-h caloric intake, portion sizes, "fast food" intake, and perceived control of eating in 88 obese subjects. After a 2-day placebo lead-in, subjects self-administered pramlintide (180 microg) or placebo by subcutaneous injection 15 min before meals for 6 wk without concomitant lifestyle modifications. Compared with placebo, pramlintide treatment elicited significant mean reductions from baseline in body weight on day 44 (-2.1 +/- 0.3 vs. +0.1 +/- 0.4%, P < 0.001), 24-h caloric intake (-990 +/- 94 vs. -243 +/- 126 kcal on day 3, P < 0.0001; -680 +/- 86 vs. -191 +/- 161 kcal on day 43, P < 0.01), portion sizes, and caloric intake at a "fast food challenge" (-385 +/- 61 vs. -109 +/- 88 kcal on day 44, P < 0.05). Pramlintide treatment also improved perceived control of eating, as demonstrated by a 45% placebo-corrected reduction in binge eating scores (P < 0.01). The results of this translational research study confirm in humans various preclinical effects of amylin agonism, demonstrating that pramlintide-mediated weight loss in obese subjects is accompanied by sustained reductions in 24-h food intake, portion sizes, fast food intake, and binge eating tendencies.


Subject(s)
Amyloid/therapeutic use , Energy Intake/drug effects , Feeding Behavior/drug effects , Obesity/drug therapy , Adult , Amyloid/adverse effects , Anti-Obesity Agents/adverse effects , Anti-Obesity Agents/therapeutic use , Appetite Regulation/drug effects , Body Weight/drug effects , Double-Blind Method , Female , Follow-Up Studies , Humans , Hunger/drug effects , Islet Amyloid Polypeptide , Male , Middle Aged , Nausea/chemically induced , Placebos , Satiation/drug effects
7.
J Clin Pharmacol ; 47(7): 798-805, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17463219

ABSTRACT

Pramlintide, an adjunct treatment to mealtime insulin for patients with type 2 and type 1 diabetes, aids glycemic control by suppressing postprandial glucagon secretion, slowing gastric emptying, and enhancing satiety. Because gastric emptying affects oral medication absorption, this placebo-controlled, single-blind, crossover study examined the absorption of 1000 mg of acetaminophen elixir administered -2, -1, 0, +1, and +2 hours relative to pramlintide (120 microg) or 0 hours relative to placebo in 24 patients with type 2 diabetes. When acetaminophen administration occurred 0, +1, or +2 hours relative to pramlintide, the maximum observed plasma concentration of acetaminophen decreased 14% to 29%, and time to maximum observed plasma concentration increased by 0.8 to 1.2 hours compared with administration 0 hours relative to placebo. Pramlintide treatment slowed but did not alter the extent of acetaminophen absorption (area under the concentration-time curve). No serious adverse events or withdrawals were reported. Oral agents should be administered at least 1 hour before or 2 hours after pramlintide injection if rapid onset of action is required for efficacy.


Subject(s)
Acetaminophen/pharmacokinetics , Amyloid/pharmacology , Analgesics, Non-Narcotic/pharmacokinetics , Diabetes Mellitus, Type 2/physiopathology , Hypoglycemic Agents/pharmacology , Acetaminophen/administration & dosage , Acetaminophen/blood , Administration, Oral , Adolescent , Adult , Amyloid/administration & dosage , Amyloid/blood , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/blood , Area Under Curve , Cross-Over Studies , Drug Interactions , Female , Gastric Emptying , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/blood , Injections, Subcutaneous , Islet Amyloid Polypeptide , Male , Middle Aged , Single-Blind Method
8.
Genes Dev ; 19(22): 2645-55, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16291643

ABSTRACT

Extracellular secretion of products is the major mechanism by which Gram-negative pathogens communicate with and intoxicate host cells. Vesicles released from the envelope of growing bacteria serve as secretory vehicles for proteins and lipids of Gram-negative bacteria. Vesicle production occurs in infected tissues and is influenced by environmental factors. Vesicles play roles in establishing a colonization niche, carrying and transmitting virulence factors into host cells, and modulating host defense and response. Vesicle-mediated toxin delivery is a potent virulence mechanism exhibited by diverse Gram-negative pathogens. The biochemical and functional properties of pathogen-derived vesicles reveal their potential to critically impact disease.


Subject(s)
Cell Wall/physiology , Gram-Negative Bacteria/cytology , Gram-Negative Bacteria/pathogenicity , Gram-Negative Bacterial Infections/metabolism , Gram-Negative Bacterial Infections/microbiology , Animals , Cell Wall/ultrastructure , Gram-Negative Bacteria/physiology , Humans
9.
EMBO J ; 23(23): 4538-49, 2004 Nov 24.
Article in English | MEDLINE | ID: mdl-15549136

ABSTRACT

Enterotoxigenic Escherichia coli (ETEC) is a prevalent cause of traveler's diarrhea and infant mortality in third-world countries. Heat-labile enterotoxin (LT) is secreted from ETEC via vesicles composed of outer membrane and periplasm. We investigated the role of ETEC vesicles in pathogenesis by analyzing vesicle association and entry into eukaryotic cells. Fluorescently labeled vesicles from LT-producing and LT-nonproducing strains were compared in their ability to bind adrenal and intestinal epithelial cells. ETEC-derived vesicles, but not control nonpathogen-derived vesicles, associated with cells in a time-, temperature-, and receptor-dependent manner. Vesicles were visualized on the cell surface at 4 degrees C and detected intracellularly at 37 degrees C. ETEC vesicle endocytosis depended on cholesterol-rich lipid rafts. Entering vesicles partially colocalized with caveolin, and the internalized vesicles accumulated in a nonacidified compartment. We conclude that ETEC vesicles serve as specifically targeted transport vehicles that mediate entry of active enterotoxin and other bacterial envelope components into host cells. These data demonstrate a role in virulence for ETEC vesicles.


Subject(s)
Bacterial Toxins/metabolism , Cell Membrane/metabolism , Enterotoxins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , Transport Vesicles/physiology , Virulence Factors/metabolism , Animals , Caveolin 1 , Caveolins/metabolism , Cell Line , Endocytosis , Escherichia coli/pathogenicity , Fluorescent Antibody Technique, Indirect , HT29 Cells , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Membrane Microdomains/metabolism , Mice , Microscopy, Electron, Transmission , Periplasm/metabolism , Transport Vesicles/ultrastructure , Virulence
10.
J Biol Chem ; 279(3): 2069-76, 2004 Jan 16.
Article in English | MEDLINE | ID: mdl-14578354

ABSTRACT

Gram-negative bacteria shed outer membrane vesicles composed of outer membrane and periplasmic components. Since vesicles from pathogenic bacteria contain virulence factors and have been shown to interact with eukaryotic cells, it has been proposed that vesicles behave as delivery vehicles. We wanted to determine whether heterologously expressed proteins would be incorporated into the membrane and lumen of vesicles and whether these altered vesicles would associate with host cells. Ail, an outer membrane adhesin/invasin from Yersinia enterocolitica, was detected in purified outer membrane and in vesicles from Escherichia coli strains DH5alpha, HB101, and MC4100 transformed with plasmid-encoded Ail. In vesicle-host cell co-incubation assays we found that vesicles containing Ail were internalized by eukaryotic cells, unlike vesicles without Ail. To determine whether lumenal vesicle contents could be modified and delivered to host cells, we used periplasmically expressed green fluorescent protein (GFP). GFP fused with the Tat signal sequence was secreted into the periplasm via the twin arginine transporter (Tat) in both the laboratory E. coli strain DH5alpha and the pathogenic enterotoxigenic E. coli ATCC strain 43886. Pronase-resistant fluorescence was detectable in vesicles from Tat-GFP-transformed strains, demonstrating that GFP was inside intact vesicles. Inclusion of GFP cargo increased vesicle density but did not result in morphological changes in vesicles. These studies are the first to demonstrate the incorporation of heterologously expressed outer membrane and periplasmic proteins into bacterial vesicles.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Cell Membrane/metabolism , Escherichia coli/metabolism , Periplasm/chemistry , Green Fluorescent Proteins , Luminescent Proteins/metabolism , Protein Sorting Signals , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...