Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
2.
Endocr Pract ; 17(6): 922-32, 2011.
Article in English | MEDLINE | ID: mdl-22068254

ABSTRACT

OBJECTIVE: To evaluate the long-term clinical effect of treatment with metreleptin (an analogue of human leptin) on glycemic and lipid abnormalities and markers of hepatic steatosis in patients with inherited or acquired lipodystrophy. METHODS: Fifty-five patients (36 with generalized lipodystrophy and 19 with partial lipodystrophy) with at least 1 of 3 metabolic abnormalities (diabetes mellitus, fasting triglyceride level ≥200 mg/dL, and insulin resistance) and low leptin levels received subcutaneous injections of metreleptin once or twice daily in an ongoing clinical trial at the National Institutes of Health. RESULTS: At baseline, hemoglobin A1c-8.5% ± 2.1% (mean ± standard deviation [SD])-and triglycerides-479 ± 80 mg/dL (geometric mean ± standard error [SE])-were substantially elevated. Robust and sustained reductions in both variables were evident for the observed patient population during a 3-year metreleptin treatment period (-2.1% ± 0.5% [mean ± SE] and -35.4% ± 13.7% [mean ± SE], respectively). Mean alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels were elevated at baseline (100 ± 120 U/L and 71 ± 77 U/L [mean ± SD], respectively) and decreased by -45 ± 19 U/L and -33 ± 14 U/L (mean ± SE), respectively, during the 3-year metreleptin treatment period. Improvements in hemoglobin A1c, triglycerides, ALT, and AST were more pronounced in the subsets of patients having elevated levels at baseline. The most notable adverse events observed in this patient population were likely attributable to underlying metabolic abnormalities or comorbidities. CONCLUSION: Metreleptin treatment substantially reduced glycemic variables, triglycerides, and liver enzymes (ALT and AST) and demonstrated durability of response throughout a 3-year treatment period. These results support metreleptin as a potential treatment for certain metabolic disorders (for example, diabetes mellitus and hypertriglyceridemia) associated with lipodystrophy.


Subject(s)
Hypoglycemic Agents/therapeutic use , Hypolipidemic Agents/therapeutic use , Leptin/analogs & derivatives , Lipodystrophy/drug therapy , Adolescent , Adult , Aged , Biomarkers/blood , Child , Cohort Studies , Fatty Liver/etiology , Fatty Liver/prevention & control , Female , Glycated Hemoglobin/analysis , Humans , Hypertriglyceridemia/etiology , Hypertriglyceridemia/prevention & control , Hypoglycemic Agents/adverse effects , Hypolipidemic Agents/adverse effects , Leptin/adverse effects , Leptin/therapeutic use , Lipodystrophy/blood , Lipodystrophy/physiopathology , Liver/physiopathology , Male , Middle Aged , Time Factors , Young Adult
3.
Obesity (Silver Spring) ; 18(1): 21-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19543217

ABSTRACT

We have previously shown that combined amylin + leptin agonism elicits synergistic weight loss in diet-induced obese (DIO) rats. Here, we assessed the comparative efficacy of amylin, leptin, or amylin + leptin in the maintenance of amylin + leptin-mediated weight loss. DIO rats pretreated with the combination of rat amylin (50 microg/kg/day) and murine leptin (125 microg/kg/day) for 4 weeks were subsequently infused with either vehicle, amylin, leptin, or amylin + leptin for an additional 4 weeks. Food intake, body weight, body composition, plasma parameters, and the expression of key metabolic genes in liver and white adipose tissue (WAT) were assessed. Amylin + leptin treatment (weeks 0-4) reduced body weight to 87.5% of baseline. Rats subsequently maintained on vehicle or leptin regained all weight (to 104.2 and 101.2% of baseline, respectively), those maintained on amylin had partial weight regain (97.0%). By contrast, weight loss was largely maintained with continued amylin + leptin treatment (91.4%), associated with a 10% decrease in adiposity. Cumulative food intake (weeks 5-8) was reduced by amylin and amylin + leptin, but not by leptin alone. Amylin + leptin, but not amylin or leptin alone, reduced plasma triglycerides (by 55%), total cholesterol (by 19%), and insulin (by 57%) compared to vehicle. Amylin + leptin also reduced hepatic stearoyl-CoA desaturase-1 (Scd1) mRNA, and increased WAT mRNA levels of adiponectin, fatty acid synthase (Fasn), and lipoprotein lipase (Lpl). We conclude that, in DIO rats, maintenance of amylin + leptin-mediated weight loss requires continued treatment with both agonists, and is accompanied by sustained improvements in body composition, and indices of lipid metabolism and insulin sensitivity.


Subject(s)
Amyloid/pharmacology , Body Weight/drug effects , Diet , Leptin/pharmacology , Obesity/drug therapy , Weight Loss/drug effects , Adiponectin/blood , Adipose Tissue, White/metabolism , Amyloid/blood , Analysis of Variance , Animals , Appetite Depressants/pharmacology , Body Composition/drug effects , Drug Interactions , Eating/drug effects , Gene Expression Profiling , Insulin/blood , Islet Amyloid Polypeptide , Leptin/blood , Liver/metabolism , Male , Obesity/metabolism , RNA, Messenger/metabolism , Rats
4.
J Investig Med ; 57(7): 777-83, 2009 Oct.
Article in English | MEDLINE | ID: mdl-20029268

ABSTRACT

The discovery of leptin in 1994 was a seminal event in obesity research. It helped to establish that body weight is tightly regulated by a complex neurohormonal feedback system and that obesity should be viewed as a disorder with a strong biological basis rather than simply the result of poor lifestyle choices and lack of willpower.Leptin, secreted from adipocytes, acts as a prototypic long-term (tonic) adiposity signal. Although nonclinical and clinical studies have provided unequivocal evidence that leptin plays a unique, pivotal role in body weight regulation, efforts to develop recombinant leptin (metreleptin) as a monotherapy for obesity have proven unsuccessful. Amylin, secreted from pancreatic beta-cells, fulfills the criteria for a short-term (episodic) satiety signal. The amylin analog pramlintide elicits sustained reductions in food intake and body weight in obese rodents and humans.A translational research program aimed at elucidating the interaction between different islet-, gut-, and adipocyte-derived hormones led to the discovery that combined amylin/leptin agonism induces marked, synergistic, fat-specific weight loss in leptin-resistant diet-induced obese rodents. In obese humans, combination treatment with pramlintide/metreleptin led to an approximately 13% weight loss after 24 weeks, significantly more than after treatment with pramlintide or metreleptin alone.Collectively, these findings suggest that combined amylin/leptin agonism may have therapeutic utility as part of an integrated, neurohormonal approach to obesity pharmacotherapy.


Subject(s)
Amyloid/agonists , Anti-Obesity Agents/therapeutic use , Leptin/agonists , Obesity/drug therapy , Amyloid/administration & dosage , Animals , Anti-Obesity Agents/administration & dosage , Drug Design , Drug Therapy, Combination , Feedback, Physiological , Humans , Islet Amyloid Polypeptide , Leptin/administration & dosage , Leptin/analogs & derivatives , Neurotransmitter Agents/physiology , Obesity/physiopathology , Satiety Response/drug effects , Satiety Response/physiology , Signal Transduction , Weight Loss/drug effects , Weight Loss/physiology
5.
Obesity (Silver Spring) ; 17(9): 1736-43, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19521351

ABSTRACT

The neurohormonal control of body weight involves a complex interplay between long-term adiposity signals (e.g., leptin), and short-term satiation signals (e.g., amylin). In diet-induced obese (DIO) rodents, amylin/leptin combination treatment led to marked, synergistic, fat-specific weight loss. To evaluate the weight-lowering effect of combined amylin/leptin agonism (with pramlintide/metreleptin) in human obesity, a 24-week, randomized, double-blind, active-drug-controlled, proof-of-concept study was conducted in obese or overweight subjects (N = 177; 63% female; 39 +/- 8 years; BMI 32.0 +/- 2.1 kg/m(2); 93.3 +/- 13.2 kg; mean +/- s.d.). After a 4-week lead-in period with pramlintide (180 microg b.i.d. for 2 weeks, 360 microg b.i.d. thereafter) and diet (40% calorie deficit), subjects achieving 2-8% weight loss were randomized 1:2:2 to 20 weeks of treatment with metreleptin (5 mg b.i.d.), pramlintide (360 microg b.i.d.), or pramlintide/metreleptin (360 microg/5 mg b.i.d.). Combination treatment with pramlintide/metreleptin led to significantly greater weight loss from enrollment to week 20 (-12.7 +/- 0.9%; least squares mean +/- s.e.) than treatment with pramlintide (-8.4 +/- 0.9%; P < 0.001) or metreleptin (-8.2 +/- 1.3%; P < 0.01) alone (evaluable, N = 93). The greater reduction in body weight was significant as early as week 4, and weight loss continued throughout the study, without evidence of a plateau. The most common adverse events with pramlintide/metreleptin were injection site events and nausea, which were mostly mild to moderate and decreased over time. These results support further development of pramlintide/metreleptin as a novel, integrated neurohormonal approach to obesity pharmacotherapy.


Subject(s)
Amyloid/therapeutic use , Anti-Obesity Agents/therapeutic use , Leptin/analogs & derivatives , Obesity/drug therapy , Overweight/drug therapy , Weight Loss/drug effects , Adiposity/drug effects , Adult , Amyloid/adverse effects , Amyloid/pharmacokinetics , Anti-Obesity Agents/adverse effects , Anti-Obesity Agents/pharmacokinetics , Body Mass Index , Combined Modality Therapy , Double-Blind Method , Drug Combinations , Energy Intake , Feeding Behavior/drug effects , Female , Humans , Islet Amyloid Polypeptide , Leptin/adverse effects , Leptin/pharmacokinetics , Leptin/therapeutic use , Male , Middle Aged , Overweight/diet therapy , Overweight/metabolism , Receptors, Islet Amyloid Polypeptide , Receptors, Leptin/agonists , Receptors, Leptin/metabolism , Receptors, Peptide/agonists , Receptors, Peptide/metabolism , Satiation/drug effects , Signal Transduction/drug effects , Time Factors , Treatment Outcome , United States
6.
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
7.
Endocrinology ; 149(11): 5679-87, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18669592

ABSTRACT

Previously, we reported that combination treatment with rat amylin (100 microg/kg.d) and murine leptin (500 microg/kg.d) elicited greater inhibition of food intake and greater body weight loss in diet-induced obese rats than predicted by the sum of the monotherapy conditions, a finding consistent with amylin-induced restoration of leptin responsiveness. In the present study, a 3 x 4 factorial design was used to formally test for a synergistic interaction, using lower dose ranges of amylin (0, 10, and 50 microg/kg.d) and leptin (0, 5, 25, and 125 microg/kg.d), on food intake and body weight after 4 wk continuous infusion. Response surface methodology analysis revealed significant synergistic anorexigenic (P < 0.05) and body weight-lowering (P < 0.05) effects of amylin/leptin combination treatment, with up to 15% weight loss at doses considerably lower than previously reported. Pair-feeding (PF) experiments demonstrated that reduction of food intake was the predominant mechanism for amylin/leptin-mediated weight loss. However, fat loss was 2-fold greater in amylin/leptin-treated rats than PF controls. Furthermore, amylin/leptin-mediated weight loss was not accompanied by the counterregulatory decrease in energy expenditure and chronic shift toward carbohydrate (rather than fat) utilization observed with PF. Hepatic gene expression analyses revealed that 28 d treatment with amylin/leptin (but not PF) was associated with reduced expression of genes involved in hepatic lipogenesis (Scd1 and Fasn mRNA) and increased expression of genes involved in lipid utilization (Pck1 mRNA). We conclude that amylin/leptin interact synergistically to reduce body weight and adiposity in diet-induced obese rodents through a number of anorexigenic and metabolic effects.


Subject(s)
Amyloid/pharmacology , Drug Resistance/drug effects , Leptin/pharmacology , Obesity/pathology , Signal Transduction/drug effects , Amyloid/administration & dosage , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Body Weight/drug effects , Caloric Restriction , Diet/adverse effects , Drug Evaluation, Preclinical , Drug Synergism , Eating/drug effects , Islet Amyloid Polypeptide , Leptin/administration & dosage , Lipids/blood , Lipogenesis/drug effects , Lipogenesis/genetics , Liver/metabolism , Obesity/drug therapy , Obesity/etiology , Obesity/metabolism , Rats , Rats, Sprague-Dawley
8.
Curr Diabetes Rev ; 4(2): 79-91, 2008 May.
Article in English | MEDLINE | ID: mdl-18473755

ABSTRACT

Contrary to its historical epithet as a lifestyle disorder, obesity is now widely recognized as having a neurobiological basis. This progress is due to our knowledge not only about energy homoeostatic pathways within the central nervous system (CNS), but also about the role of peripheral peptide hormones acting upon the CNS. These hormones include long-term adiposity signals, such as leptin, that inform the CNS primarily of changes in the body's overall fat and energy reserves, and short-term signals such as amylin, peptide YY (PYY) and ghrelin, that primarily reflect changes in the immediate nutritive state (energy intake). The limited weight loss effects achieved with current monotherapy approaches to obesity have been attributed, at least in part, to the redundancies and potent counter-regulatory responses within the neurohormonal feedback loop governing energy balance. Recently, we reported that combinations of amylin, leptin and PYY(3-36) resulted in additive and/or synergistic interactions and caused marked weight loss in the diet-induced obese rat model, which to date has reasonably predicted the clinical effects of several hormones in obese humans. If confirmed in ongoing translational clinical research studies, these findings may provide a physiological rationale for a novel, integrated neurohormonal approach to pharmacotherapy for obesity.


Subject(s)
Adipocytes/metabolism , Adipokines/physiology , Gastrointestinal Hormones/physiology , Intestines/physiology , Islets of Langerhans/metabolism , Pancreatic Hormones/physiology , Body Weight/physiology , Brain/physiology , Energy Intake , Energy Metabolism , Feedback , Homeostasis/physiology , Humans , Leptin/physiology , Models, Biological , Signal Transduction/physiology
9.
Proc Natl Acad Sci U S A ; 105(20): 7257-62, 2008 May 20.
Article in English | MEDLINE | ID: mdl-18458326

ABSTRACT

Body weight is regulated by complex neurohormonal interactions between endocrine signals of long-term adiposity (e.g., leptin, a hypothalamic signal) and short-term satiety (e.g., amylin, a hindbrain signal). We report that concurrent peripheral administration of amylin and leptin elicits synergistic, fat-specific weight loss in leptin-resistant, diet-induced obese rats. Weight loss synergy was specific to amylin treatment, compared with other anorexigenic peptides, and dissociable from amylin's effect on food intake. The addition of leptin after amylin pretreatment elicited further weight loss, compared with either monotherapy condition. In a 24-week randomized, double-blind, clinical proof-of-concept study in overweight/obese subjects, coadministration of recombinant human leptin and the amylin analog pramlintide elicited 12.7% mean weight loss, significantly more than was observed with either treatment alone (P < 0.01). In obese rats, amylin pretreatment partially restored hypothalamic leptin signaling (pSTAT3 immunoreactivity) within the ventromedial, but not the arcuate nucleus and up-regulated basal and leptin-stimulated signaling in the hindbrain area postrema. These findings provide both nonclinical and clinical evidence that amylin agonism restored leptin responsiveness in diet-induced obesity, suggesting that integrated neurohormonal approaches to obesity pharmacotherapy may facilitate greater weight loss by harnessing naturally occurring synergies.


Subject(s)
Amyloid/agonists , Amyloid/chemistry , Leptin/metabolism , Adipose Tissue/metabolism , Amyloid/metabolism , Amyloid/pharmacology , Animals , Body Weight , Caloric Restriction , Disease Models, Animal , Hormones/metabolism , Hypothalamus/metabolism , Islet Amyloid Polypeptide , Leptin/analogs & derivatives , Leptin/pharmacology , Models, Biological , Obesity/genetics , Obesity/therapy , Oxygen Consumption , Rats
10.
Am J Clin Nutr ; 87(2): 295-302, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18258617

ABSTRACT

BACKGROUND: Enlargement of adipocytes from subcutaneous abdominal adipose tissue (SAT), increased intrahepatic lipid content (IHL), intramyocellular lipid content (IMCL), and low circulating adiponectin concentrations are associated with insulin resistance. OBJECTIVE: Because adiponectin increases fat oxidation in skeletal muscle and liver, and the expression of the adiponectin gene in SAT is inversely associated with adipocyte size, we hypothesized that hypoadiponectinemia links hypertrophic obesity with insulin resistance via increased IMCL and IHL. DESIGN: Fifty-three obese Pima Indians with a mean (+/-SD) age of 27 +/- 8 y, body fat of 35 +/- 5%, and normal glucose regulation (normal fasting and 2-h glucose concentration per WHO 1999 criteria) underwent euglycemic-hyperinsulinemic clamp, biopsies of SAT and vastus lateralis muscle, and magnetic resonance imaging of the abdomen. RESULTS: Adipocyte diameter (AD) correlated positively with body fat (P < 0.0001) and IHL (estimated from magnetic resonance imaging intensity of liver; P = 0.047). No association was found between AD and plasma adiponectin or IMCL. Plasma adiponectin negatively correlated with type II IMCL (IIA, P = 0.004; IIX, P = 0.009) or IHL (P = 0.02). In a multivariate analysis, plasma adiponectin, AD, and visceral adipose tissue (VAT) independently predicted IHL. Low insulin-mediated glucose disposal was associated with low plasma adiponectin (P = 0.02) and high IHL (P = 0.0003), SAT (P = 0.02), and VAT (P = 0.04). High IHL was the only predictor of reduced insulin-mediated suppression of hepatic glucose production (P = 0.02) and the only independent predictor of insulin-mediated glucose disposal in a multivariate analysis. CONCLUSIONS: Increased lipid content in the liver may independently link hypoadiponectinemia, hypertrophic obesity, and increased visceral adiposity with peripheral and hepatic insulin resistance.


Subject(s)
Adipocytes/pathology , Adiponectin/blood , Fatty Liver/metabolism , Insulin Resistance , Intra-Abdominal Fat/metabolism , Obesity/metabolism , Subcutaneous Fat, Abdominal/metabolism , Adipocytes/metabolism , Adult , Fatty Liver/pathology , Female , Glucose Clamp Technique , Humans , Indians, North American , Intra-Abdominal Fat/pathology , Magnetic Resonance Imaging , Male , Multivariate Analysis , Obesity/pathology , Subcutaneous Fat, Abdominal/pathology
11.
Endocrinology ; 148(12): 6054-61, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17761760

ABSTRACT

Circulating levels of the pancreatic beta-cell peptide hormone amylin and the gut peptide PYY[3-36] increase after nutrient ingestion. Both have been implicated as short-term signals of meal termination with anorexigenic and weight-reducing effects. However, their combined effects are unknown. We report that the combination of amylin and PYY[3-36] elicited greater anorexigenic and weight-reducing effects than either peptide alone. In high-fat-fed rats, a single ip injection of amylin (10 microg/kg) plus PYY[3-36] (1000 microg/kg) reduced food intake for 24 h (P < 0.05 vs. vehicle), whereas the anorexigenic effects of either PYY[3-36] or amylin alone began to diminish 6 h after injection. These anorexigenic effects were dissociable from changes in locomotor activity. Subcutaneous infusion of amylin plus PYY[3-36] for 14 d suppressed food intake and body weight to a greater extent than either agent alone in both rat and mouse diet-induced obesity (DIO) models (P < 0.05). In DIO-prone rats, 24-h metabolic rate was maintained despite weight loss, and amylin plus PYY[3-36] (but not monotherapy) increased 24-h fat oxidation (P < 0.05 vs. vehicle). Finally, a 4 x 3 factorial design was used to formally describe the interaction between amylin and PYY[3-36]. DIO-prone rats were treated with amylin (0, 4, 20, and 100 microg/kg.d) and PYY[3-36] (0, 200, 400 microg/kg.d) alone and in combination for 14 d. Statistical analyses revealed that food intake suppression with amylin plus PYY[3-36] treatment was synergistic, whereas body weight reduction was additive. Collectively, these observations highlight the importance of studying peptide hormones in combination and suggest that integrated neurohormonal approaches may hold promise as treatments for obesity.


Subject(s)
Amyloid/pharmacology , Obesity/drug therapy , Peptide YY/pharmacology , Weight Loss/drug effects , Amyloid/administration & dosage , Amyloid/therapeutic use , Animals , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Dietary Fats , Drug Synergism , Drug Therapy, Combination , Eating/drug effects , Humans , Islet Amyloid Polypeptide , Male , Mice , Obesity/chemically induced , Peptide Fragments , Peptide YY/administration & dosage , Peptide YY/therapeutic use , Rats
12.
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
13.
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
14.
Obesity (Silver Spring) ; 15(5): 1179-86, 2007 May.
Article in English | MEDLINE | ID: mdl-17495194

ABSTRACT

OBJECTIVE: We previously reported that a single preprandial injection (120 microg) of pramlintide, an analog of the beta-cell hormone amylin, reduced ad libitum food intake in obese subjects. To further characterize the meal-related effects of amylin signaling in humans, we studied a lower pramlintide dose (30 microg) in normal-weight subjects. RESEARCH METHODS AND PROCEDURES: In a randomized, double-blind, placebo-controlled, cross-over study, 15 healthy men (age, 24 +/- 7 years; BMI, 22.2 +/- 1.8 kg/m(2)) underwent a standardized buffet meal test on two occasions. After an overnight fast, subjects received a single subcutaneous injection of pramlintide (30 microg) or placebo, followed immediately by a standardized pre-load meal. After 1 hour, subjects were offered an ad libitum buffet meal, and total caloric intake and meal duration were measured. RESULTS: Compared with placebo, pramlintide reduced total caloric intake (1411 +/- 94 vs. 1190 +/- 117 kcal; Delta, -221 +/- 101 kcal; -14 +/- 9%; p = 0.05) and meal duration (36 +/- 2 vs. 31 +/- 3 minutes; Delta, -5.1 +/- 1.4 minutes; p < 0.005). Visual analog scale profiles of hunger trended lower and fullness higher during the first hour after pramlintide administration. In response to the buffet, hunger and fullness changed to a similar degree after pramlintide and placebo, despite subjects on pramlintide consuming 14% fewer kilocalories. Visual analog scale nausea ratings remained near baseline, without differences between treatments. Plasma peptide YY, cholecystokinin, and ghrelin concentrations did not differ with treatment, whereas glucagon-like peptide-1 concentrations after meals were lower in response to pramlintide than to placebo. DISCUSSION: These observations add support to the concept that amylin agonism may have a role in human appetite control.


Subject(s)
Amyloid/pharmacology , Eating/physiology , Energy Intake/drug effects , Adult , Aged , Body Mass Index , Body Weight , Cross-Over Studies , Double-Blind Method , Humans , Hypoglycemic Agents/pharmacology , Islet Amyloid Polypeptide , Male , Middle Aged , Placebos , Postprandial Period , Reference Values , Time Factors
15.
Am J Health Syst Pharm ; 62(8): 816-22, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15821274

ABSTRACT

PURPOSE: The pharmacokinetics, pharmacodynamics, and safety of pramlintide and various insulin formulations in patients with type 1 diabetes mellitus (DM) when given as separate injections or mixed in the same syringe before injection were studied. METHODS: In two randomized, open-label, placebo-controlled, five-period-crossover studies, patients with type 1 DM received preprandial injections of pramlintide, short-acting insulin, and long-acting insulin administered either by separate injections or after mixing in various combinations. Serum free insulin and plasma glucose concentrations were measured for 10 hours and plasma pramlintide concentrations for 5 hours after injection. RESULTS: Blood samples were collected from a total of 51 patients. All treatments involving mixtures were comparable to separate injections with respect to the area under the concentration-versus-time curve (AUC) and the maximum concentration (Cmax) of serum free insulin. There were some minor differences in the AUC and Cmax of pramlintide. No injection-site reactions or other unexpected adverse events were observed. CONCLUSION: Mixing pramlintide with short- or long-acting insulin in the same syringe before subcutaneous injection did not affect the pharmacodynamics of glucose or the pharmacokinetics of insulin or pramlintide in a clinically significant manner.


Subject(s)
Amyloid/administration & dosage , Diabetes Mellitus, Type 1/drug therapy , Insulin/administration & dosage , Adolescent , Adult , Aged , Amyloid/adverse effects , Amyloid/pharmacokinetics , Area Under Curve , Cross-Over Studies , Drug Combinations , Female , Humans , Insulin/adverse effects , Insulin/pharmacokinetics , Islet Amyloid Polypeptide , Male , Middle Aged , Placebos , United States
16.
Diabetes Care ; 28(3): 632-7, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15735200

ABSTRACT

OBJECTIVE: Oxidative stress has been shown to be increased in the postprandial period in patients with diabetes and has been implicated in the pathogenesis of micro- and macrovascular complications. The aim of this post hoc analysis was to assess the effects of pramlintide, an amylin analog shown to reduce postprandial glucose excursions in patients with diabetes, on markers of oxidative stress in the postprandial period. RESEARCH DESIGN AND METHODS: In a randomized, single-blind, placebo-controlled, crossover study, 18 evaluable subjects with type 1 diabetes underwent two standardized breakfast meal tests and received pramlintide or placebo in addition to their preprandial insulin. The plasma concentrations of glucose and markers of oxidative stress (nitrotyrosine, oxidized LDL [ox-LDL], and total radical-trapping antioxidant parameter [TRAP]) were measured at baseline and during the 4-h postprandial period. RESULTS: Compared with placebo, pramlintide treatment significantly reduced postprandial excursions of glucose, nitrotyrosine, and ox-LDL and prevented a decline in TRAP (P < 0.03 for all comparisons). Correlation analyses adjusted for treatment revealed a significant association between postprandial mean incremental area under the curve from 0 to 4 h (AUC(0-4 h)) for glucose and postprandial mean incremental AUC(0-4 h) for each measure of oxidative stress (r = 0.75, 0.54, and -0.63 for nitrotyrosine, ox-LDL, and TRAP, respectively; P < 0.001 for all correlations). CONCLUSIONS: These findings indicate that the postprandial glucose-lowering effect of pramlintide in type 1 diabetes is associated with a significant reduction in postprandial oxidative stress.


Subject(s)
Amyloid/therapeutic use , Blood Glucose/metabolism , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/therapeutic use , Lipoproteins, LDL/blood , Oxidative Stress/physiology , Tyrosine/analogs & derivatives , Tyrosine/blood , Adult , Blood Glucose/drug effects , Cross-Over Studies , Diabetes Mellitus, Type 1/blood , Enzyme-Linked Immunosorbent Assay , Glycated Hemoglobin/metabolism , Humans , Islet Amyloid Polypeptide , Male , Middle Aged , Oxidative Stress/drug effects , Placebos , Postprandial Period , Safety , Single-Blind Method
17.
Diabetes ; 53(12): 3091-6, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15561938

ABSTRACT

Pancreatic polypeptide (PP) is released from the pancreas in response to a meal. In humans, low-circulating PP levels have been observed in obesity, and administration of pharmacological doses of PP has been shown to decrease food intake. The aim of the present study was to investigate whether low circulating PP is associated with weight gain in Pima Indians. Plasma PP concentrations were measured after an overnight fast and 30 min after a standardized mixed meal in 33 nondiabetic male subjects who had a follow-up visit 4.9 +/- 2.5 years later. Cross-sectionally, fasting and postprandial PP levels were negatively associated with body size and adiposity. Prospectively, the change in PP response to the meal was negatively associated with the change in body weight (r = -0.53, P = 0.002). In contrast, a high fasting PP level was positively associated with change in body weight (r = 0.45, P = 0.009). In conclusion, our results provide evidence that, even within the physiological range, PP contributes to the regulation of energy balance in humans. However this contribution appears to be more complex than anticipated because of the opposite effect of fasting and postprandial PP on the risk of future weight gain.


Subject(s)
Body Weight/physiology , Indians, North American , Pancreatic Polypeptide/physiology , Adult , Arizona , Body Mass Index , Eating , Energy Metabolism/physiology , Fasting , Homeostasis , Humans , Male , Pancreatic Polypeptide/blood , Postprandial Period
18.
Metabolism ; 53(9): 1227-32, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15334389

ABSTRACT

Pramlintide is an analog of the human glucoregulatory hormone amylin. Previous studies have shown no clear evidence that pramlintide modifies the response to insulin-induced hypoglycemia; however, a detailed assessment of responses at hypoglycemic thresholds has not been conducted. To further test the effect of pramlintide on symptom, catecholamine, and glucagon responses, a 3-step hypoglycemic clamp was investigated in healthy volunteers. In a randomized, double-blind, placebo-controlled, crossover study, 18 healthy subjects without diabetes received subcutaneous premeal injections of either placebo or 60 microg pramlintide 3 times daily for 5 consecutive days. On day 6, subjects received study drug with breakfast and, after a 7-hour fast, were connected to a Biostator for a 3-step, 3-hour clamp experiment (insulin infusion rate: 1.0 mU/kg/min; blood glucose targets: 70, 55, and 45 mg/dL). An intravenous (IV) infusion of pramlintide (16 microg/h) or placebo was initiated at t = 60 minutes. At the end of each 60-minute clamp step, autonomic (sweating, palpitations, hunger, etc) and neuroglycopenic (confusion, headache, odd behavior, etc) symptoms were assessed using a validated visual analog scale questionnaire. Blood samples were collected at 30-minute intervals for measurement of plasma glucose, insulin, pramlintide, catecholamine, and glucagon concentrations. Intraindividual and group mean responses showed that autonomic symptoms and plasma catecholamine and glucagon concentrations increased progressively during the clamp, with no discernible differences between pramlintide and placebo treatments. Group means for catecholamines at 60 minutes were: epinephrine 233 +/- 42, 892 +/- 85, 2,340 +/- 302 and 202 +/- 25, 774 +/- 114, 2,751 +/- 404 pg/mL and norepinephrine 1,138 +/- 86, 1,236 +/- 77, 1,721 +/- 158 and 1,278 +/- 108, 1,259 +/- 109, 1,580 +/-136 pg/mL (+/- SEM) for placebo- and pramlintide-treated groups at 70, 55, and 45 mg/dL glucose, respectively. Group means for glucagon were 72 +/- 6.3, 98 +/- 11.1, 130 +/- 14.7 and 63 +/- 3.6, 92 +/- 9.4, 120 +/- 16.0 pmol/L (+/- SEM) for placebo- and pramlintide-treated groups at 70, 55, and 45 mg/dL glucose, respectively. These results showed that pramlintide did not impair the symptom, catecholamine, and glucagon responses to insulin-induced hypoglycemia in healthy subjects.


Subject(s)
Amyloid/pharmacology , Catecholamines/blood , Glucagon/blood , Hypoglycemia/blood , Hypoglycemic Agents/pharmacology , Adolescent , Adult , Amyloid/adverse effects , Amyloid/blood , Autonomic Nervous System/drug effects , Blood Glucose/metabolism , Body Mass Index , Cross-Over Studies , Double-Blind Method , Female , Glucose Clamp Technique , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/blood , Islet Amyloid Polypeptide , Kinetics , Male
19.
Metabolism ; 53(7): 904-11, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15254885

ABSTRACT

Individuals with low birth weight (LBW) are at increased risk of developing type 2 diabetes in later life. Whether impairments in endogenous glucose production (EGP), insulin action, insulin secretion, or a combination thereof account for this association is unclear. We, therefore, examined these parameters in Pima Indians with normal glucose tolerance. Body composition, glucose and insulin responses during a 75-g oral glucose tolerance test (OGTT), EGP, insulin-stimulated glucose disposal during low- and high-dose insulin infusion (M-low and M-high, hyperinsulinemic glucose clamp), and acute insulin response (AIR) to a 25-g intravenous glucose challenge were measured in 230 Pima Indians (147 men and 83 women, aged 25 +/- 0.4 years [mean +/- SE; range, 18 to 44]) with normal glucose tolerance. A subgroup of 63 subjects additionally underwent biopsies of subcutaneous adipose tissue for determination of adipocyte cell size and lipolysis. Subjects in the lowest quartile of birth weight (birth weight: 2,891 +/- 33 g, LBW, n = 58) were compared to those whose birth weight was in the upper 3 quartiles (birth weight: 3,657 +/- 28 g, NBW, n = 172). Age- and sex-adjusted body mass index (BMI), percent body fat, and waist-to-thigh ratio (WTR) were similar in LBW and NBW subjects. Suppression of EGP during the clamp was less in LBW than in NBW subjects before (P = .002) and after adjustment for age, sex, percent body fat, and M-low (P = .02). M-low and M-high were less in LBW than in NBW subjects before (P = .05 and P = .01) and after adjustment for age, sex, percent body fat, and WTR (P = .04 and P = .05). AIR was not different in LBW compared to NBW subjects before adjustments (P = .06), but it was lower in LBW than in NBW subjects after adjustment for age, sex, percent body fat, and M-low (P = .02), suggesting that AIR did not increase appropriately for the decrease in insulin-stimulated glucose disposal (M). In addition, average adipocyte cell size (P = .08) and basal lipolysis (P = .02) were higher in the LBW than in the NBW group. These results show that Pima Indians with LBW manifest a variety of impairments in metabolism in adulthood. Among these, a lesser insulin-stimulated suppression of EGP and a lesser insulin secretory capacity are the predominant ones. We conclude that interaction of multiple defects may contribute to increased susceptibility to type 2 diabetes among individuals with LBW.


Subject(s)
Glucose Intolerance/epidemiology , Glucose/biosynthesis , Indians, North American , Infant, Low Birth Weight/blood , Insulin Resistance/physiology , Insulin/metabolism , Adipocytes/physiology , Adipocytes/ultrastructure , Adult , Body Composition/physiology , Body Mass Index , Diabetes Mellitus/blood , Diabetes Mellitus/epidemiology , Diabetes Mellitus/genetics , Female , Glucose Clamp Technique , Glucose Intolerance/blood , Humans , Infant, Newborn , Lipolysis/physiology , Male , Risk Factors , United States/epidemiology
20.
Obes Res ; 12(4): 661-8, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15090634

ABSTRACT

OBJECTIVE: Several randomized, placebo-controlled, double-blind trials in insulin-treated patients with type 2 diabetes have shown that adjunctive therapy with pramlintide reduces hemoglobin (Hb)A1c with concomitant weight loss. This analysis further characterizes the weight-lowering effect of pramlintide in this patient population. RESEARCH METHODS AND PROCEDURES: This pooled post hoc analysis of two long-term trials included all patients who were overweight/obese at baseline (BMI > 25 kg/m2), and who were treated with either 120 microg pramlintide BID (n = 254; HbA1c 9.2%; weight, 96.1 kg) or placebo (n = 244; HbA1c 9.4%; weight, 95.0 kg). Statistical endpoints included changes from baseline to week 26 in HbA1c, body weight, and insulin use. RESULTS: Pramlintide treatment resulted in significant reductions from baseline to week 26, compared with placebo, in HbA1c and body weight (both, p < 0.0001), for placebo-corrected reductions of -0.41% and -1.8 kg, respectively. Approximately three times the number of patients using pramlintide experienced a > or = 5% reduction of body weight than with placebo (9% vs. 3%, p = 0.0005). Patients using pramlintide also experienced a proportionate decrease in total daily insulin use (r = 0.39, p < 0.0001). The greatest placebo-corrected reductions in weight at week 26 were observed in pramlintide-treated patients with a BMI >40 kg/m2 and in those concomitantly treated with metformin (both, p < 0.001), for placebo-corrected reductions of -3.2 kg and -2.5 kg, respectively. DISCUSSION: These findings support further evaluation of the weight-lowering potential of pramlintide in obese patients with type 2 diabetes.


Subject(s)
Amyloid/therapeutic use , Body Weight/drug effects , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Obesity , Aged , Amyloid/adverse effects , Body Mass Index , Double-Blind Method , Drug Therapy, Combination , Female , Glycated Hemoglobin/analysis , Humans , Insulin/administration & dosage , Islet Amyloid Polypeptide , Male , Metformin/administration & dosage , Middle Aged , Nausea/chemically induced , Placebos , Weight Loss
SELECTION OF CITATIONS
SEARCH DETAIL
...