1932

Abstract

Antiobesity medical management has shown unsatisfactory results to date in terms of efficacy, safety, and long-term maintenance of weight loss. This poor performance could be attributed to the complexity of appetite regulation mechanisms; the serious drug side effects; and, crucially, the lack of profile-matching treatment strategies and individualized, multidisciplinary follow-up. Nevertheless, antiobesity pharmacotherapy remains a challenging, exciting field of intensive scientific interest. According to the latest studies, the future of bariatric medicine lies in developing drugs acting at multiple levels of the brain-gut axis. Currently, research is focused on the generation of combination treatments based on gut hormones in a way that mimics changes underlying surgically induced weight loss, in addition to centrally acting agents; these aim to restore energy balance disruptions and enhance energy expenditure. Collectively, the pharmacological resolution of obesity could potentially be achieved with combination regimens targeting different molecules and levels of the energy homeostasis system, in parallel with matching patients' needs, resulting in a favorable metabolic profile.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-pharmtox-010716-104735
2017-01-06
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/pharmtox/57/1/annurev-pharmtox-010716-104735.html?itemId=/content/journals/10.1146/annurev-pharmtox-010716-104735&mimeType=html&fmt=ahah

Literature Cited

  1. 1. US Dep. Health Hum. Serv., US Food Drug Adm. (FDA), Cent. Drug Eval. Res. (CDER) 2007. Guidance for industry developing products for weight management Silver Spring, MD: FDA http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm071612.pdf
  2. Janssen I, Katzmarzyk PT, Ross R. 2.  2004. Waist circumference and not body mass index explains obesity-related health risk. Am. J. Clin. Nutr. 79:379–84 [Google Scholar]
  3. Pi-Sunyer FX. 3.  2004. The epidemiology of central fat distribution in relation to disease. Nutr. Rev. 62:S120–26 [Google Scholar]
  4. Grube B, Chong PW, Lau KZ, Orzechowski HD. 4.  2013. A natural fiber complex reduces body weight in the overweight and obese: a double-blind, randomized, placebo-controlled study. Obesity 21:58–64 [Google Scholar]
  5. 5. World Health Organ. (WHO) 2016. Obesity and overweight: fact sheet 311. Updated June. http://www.who.int/mediacentre/factsheets/fs311
  6. Hamdy O, Uwaifo GI, Oral EA. 6.  2011. Obesity New York: Medscape Drugs Dis. Accessed Aug. 20, 2011 http://emedicine.medscape.com/article/123702-overview
  7. 7. World Health Organ. (WHO) 2016. Global Health Observatory (GHO) Data Geneva: WHO http://www.who.int/gho/ncd/risk_factors/obesity_text/en/
  8. 8. Local Gov. Assoc., Public Health Engl 2013. Social care and obesity: a discussion paper Disc. Paper, Local Gov. Assoc., London. Accessed Dec. 4, 2013. http://www.local.gov.uk/publications/-/journal_content/56/10180/5610298/PUBLICATION
  9. Finkelstein E, Trogdon JG, Cohen JW, Dietz W. 9.  2009. Annual medical spending attributable to obesity: payer- and service-specific estimates. Health Aff. 28:w822–31 [Google Scholar]
  10. Loring B, Robertson A. 10.  2014. Obesity and Inequities: Guidance for Addressing Inequities in Overweight and Obesity Copenhagen, Den.: World Health Organ http://www.euro.who.int/__data/assets/pdf_file/0003/247638/obesity-090514.pdf
  11. Kopelman P. 11.  2007. Health risks associated with overweight and obesity. Obes. Rev. 8:Suppl. 113–17 [Google Scholar]
  12. De Wit L, Lupino F, van Straten A, Penninx B, Zitman F. 12.  et al. 2010. Depression and obesity: a meta-analysis of community-based studies. Psychiatry Res 178:230–35 [Google Scholar]
  13. Simon GE, Ludman EJ, Linde JA, Operskalski BH, Ichikawa L. 13.  et al. 2008. Association between obesity and depression in middle-aged women. Gen. Hosp. Psychiatry 30:32–39 [Google Scholar]
  14. Pories WJ, Swanson MS, McDonald KG, Long SB, Morris PG. 14.  et al. 1995. Who would have thought it? An operation proves to be the most effective therapy for adult onset diabetes mellitus. Ann. Surg. 222:339–50 [Google Scholar]
  15. Wright SM, Aronne LJ. 15.  2011. Obesity in 2010: the future of obesity medicine: Where do we go from here?. Nat. Rev. Endocrinol. 7:69–70 [Google Scholar]
  16. Jensen MD, Ryan DH, Apovian CM, Ard JD, Comuzzie AG. 16.  et al. 2014. 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults: a report of the American College of Cardiology/American Heart Association/Task Force on Practice Guidelines and The Obesity Society. Circulation 129:25, Suppl. 2102–38 [Google Scholar]
  17. Ioannides-Demos LL, Proietto J, Tokin AM, McNeil JJ. 17.  2006. Safety of drug therapies used for weight loss and treatment of obesity. Drug Saf 29:277–302 [Google Scholar]
  18. 18. US Food Drug Admin. (FDA) 2010. Meridia (sibutramine): market withdrawal due to risk of serious cardiovascular events News Release, Aug. 10. http://www.fda.gov/safety/medwatch/safetyinformation/safetyalertsforhumanmedicalproducts/ucm228830.htm
  19. Christensen R, Kristensen PC, Bartels EM, Biddal H, Astrup A. 19.  2007. Efficacy and safety of the weight-loss drug rimonabant: a meta-analysis of randomized trials. Lancet 370:1706–13 [Google Scholar]
  20. 20. Eur. Med. Agency 2013. Withdrawal of the marketing authorisation application for Belviq (lorcaserin). News Release, May 30. Accessed Dec. 31, 2013. http://www.ema.europa.eu/docs/en_GB/document_library/Medicine_QA/2013/05/WC500143811.pdf
  21. 21. Vivus, Inc. 2013. Vivus receives decision regarding Qsiva appeal News Release, Feb. 21. http://ir.vivus.com/releasedetail.cfm?ReleaseID=742340
  22. 22. Natl. Heart Lung Blood Inst. (NHLBI), Natl. Inst. Diabetes Dig. Kidney Dis. (NIDDK) 1998. Clinical Guidelines on the Identification, Evaluation, and Treatment of Overweight and Obesity in Adults: The Evidence Report Bethesda, MD: Natl. Inst. Health
  23. Ikramuddin S, Korner J, Lee WJ, Connett JE, Inabnet WB. 23.  et al. 2013. Roux-en-Y gastric bypass versus intensive medical management for the control of type 2 diabetes, hypertension and hyperlipidemia: the Diabetes Surgery Study randomized controlled trial. JAMA 309:2240–49 [Google Scholar]
  24. Camastra S, Muscelli E, Gastaldelli A, Holst JJ, Astiarraga B. 24.  et al. 2013. Long-term effects of bariatric surgery on meal disposal and β-cell function in diabetic and nondiabetic patients. Diabetes 62:3709–17 [Google Scholar]
  25. Kashyap SR, Bhatt DL, Wolski K, Watanabe RM, Abdul-Ghani M. 25.  et al. 2013. Metabolic effects of bariatric surgery in patients with moderate obesity and type 2 diabetes: analysis of a randomized control trial comparing surgery with intensive medical treatment. Diabetes Care 36:2175–82 [Google Scholar]
  26. Woods SC, D'Alessio DA. 26.  2008. Central control of body weight and appetite. J. Clin. Endocrinol. Metab. 93:S37–50 [Google Scholar]
  27. Reece AS. 27.  2011. Hypothalamic opioid-melanocortin appetitive balance and addictive craving. Med. Hypotheses 76:132–37 [Google Scholar]
  28. Greenway FL, Whitehouse MJ, Guttadauria M, Anderson JW, Atkinson RL. 28.  et al. 2009. Rational design of a combination medication for the treatment of obesity. Obesity 17:30–39 [Google Scholar]
  29. van Bloemendaal L, ten Kulve JS, la Fleur SE, Ijzerman RG, Diamant M. 29.  2014. Effects of glucagon-like peptide 1 on appetite and body weight: focus on the CNS. J. Endocrinol. 221:T1–16 [Google Scholar]
  30. Le Roux CW, Aylwin SJ, Batterham RL, Borg CM, Coyle F. 30.  et al. 2006. Gut hormone profiles following bariatric surgery favour an anorectic state, facilitate weight loss and improve metabolic parameters. Ann. Surg. 243:108–14 [Google Scholar]
  31. Karamanakos SM, Vagenas K, Kalfarentzos F, Alexandrides TK. 31.  2008. Weight loss appetite suppression and changes in fasting and postprandial ghrelin and peptide-YY levels after Roux-en-Y gastric bypass and sleeve gastrectomy: a prospective, double-blind study. Ann. Surg. 247:401–7 [Google Scholar]
  32. Burton PR, Brown WA. 32.  2011. The mechanisms of weight loss with laparoscopic adjustable gastric banding: induction of satiety not restriction. Int. J. Obes. 35:S26–30 [Google Scholar]
  33. Dar MS, Chapman WH III, Pender JR, Drake AJ III, O'Brien K. 33.  et al. 2012. GLP-1 response to a mixed meal: What happens 10 years after Roux-en-Y gastric bypass (RYGB). Obes. Surg. 22:1077–83 [Google Scholar]
  34. Chandarana K, Gelegen C, Irvine EE, Choudhury AI, Amouyal C. 34.  et al. 2013. Peripheral activation of the Y2-receptor promotes secretion of GLP-1 and improves glucose tolerance. Mol. Metab. 2:142–52 [Google Scholar]
  35. Ahima RS, Flier JS. 35.  2000. Leptin. Annu. Rev. Physiol. 62:413–37 [Google Scholar]
  36. Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H. 36.  et al. 1997. Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 387:903–8 [Google Scholar]
  37. Rosenbaum M, Goldsmith R, Bloomfield D, Magnano A, Weimer L. 37.  et al. 2005. Low-dose leptin reverses skeletal muscle, autonomic and neuroendocrine adaptations to maintenance of reduced weight. J. Clin. Investig. 115:3579–86 [Google Scholar]
  38. Rosenbaum M, Murphy EM, Heymsfield SB, Matthews DE, Leibel RL. 38.  2002. Low dose leptin administration reverses effects of sustained weight-reduction on energy expenditure and circulating concentrations of thyroid hormones. J. Clin. Endocrinol. Metab. 87:2391–94 [Google Scholar]
  39. Kissileff HR, Thornton JC, Torres M, Pavlovich K, Mayer LS. 39.  et al. 2012. Leptin reverses declines in satiation in weight-reduced obese humans. Am. J. Clin. Nutr. 95:309–17 [Google Scholar]
  40. Heymsfield SB, Greenberg AS, Fujioka K, Dixon RM, Kushner R. 40.  et al. 1999. Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, dose-escalation trial. JAMA 282:1568–75 [Google Scholar]
  41. Le Roux CW, Welbourn R, Werling M, Osborne A, Kokkinos A. 41.  et al. 2007. Gut hormones as mediators of appetite and weight loss after Roux-en-Y gastric bypass. Ann. Surg. 246:780–85 [Google Scholar]
  42. Roth JD, Roland BL, Cole RL, Trevaskis JL, Weyer C. 42.  et al. 2008. Leptin responsiveness restored by amylin agonism in diet-induced obesity: evidence from nonclinical and clinical studies. PNAS 105:7257–62 [Google Scholar]
  43. 43. Takeda Pharm 2010. Amylin and Takeda announce decision to advance development of pramlintide/metreleptin combination treatment for obesity News Release, Feb. 23. http://www.takeda.com/press/article_35851.html
  44. 44. Takeda Pharm 2011. Amylin and Takeda discontinue development of pramlintide/metreleptin combination treatment for obesity following commercial reassessment of the program News Release, Aug. 4. http://www.takeda.com/news/2011/20110805_3889.html
  45. Launay JM, Hervé P, Tournois C, Callebert J, Nebigil CG. 45.  et al. 2002. Function of the serotonin 5-hydroxytryptamine 2B receptor in pulmonary hypertension. Nat. Med. 8:1129–35 [Google Scholar]
  46. Fitzgerald LW, Burn TC, Brown BS, Patterson JP, Corjay MH. 46.  et al. 2000. Possible role of valvular serotonin 5-HT2B receptors in the cardiopathy associated with fenfluramine. Mol. Pharmacol. 57:75–81 [Google Scholar]
  47. Thomsen WJ, Grottick AJ, Menzaghi F, Reyes-Saldana H, Espitia S. 47.  2008. Lorcaserin, a novel selective human 5-hydroxytryptamine2C agonist: in vitro and in vivo pharmacological characterization. J. Pharmacol. Exp. Ther. 325:577–87 [Google Scholar]
  48. 48. Arena Pharm 2005. Arena Pharmaceuticals announces positive Phase 2b clinical trial results of novel anti-obesity compound News Release, Dec. 13. http://invest.arenapharm.com/releasedetail.cfm?ReleaseID=320421
  49. 49. Arena Pharm 2010. FDA issues complete response letter for lorcaserin new drug application News Release, Oct. 23. http://www.drugs.com/nda/lorcaserin_101025.html
  50. 50. US Food Drug Admin. (FDA) 2012. FDA approves Belviq to treat some overweight or obese adults News Release, June 27. http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm309993.htm
  51. Padwal R. 51.  2009. Contrave, a bupropion and naltrexone combination therapy for the potential treatment of obesity. Curr. Opin. Investig. Drugs 10:1117–25 [Google Scholar]
  52. Apovian CM, Aronne L, Rubino D, Still C, Wyatt H. 52.  et al. 2013. A randomized, phase 3 trial of naltrexone SR/bupropion SR on weight and obesity-related risk factors (COR-II). Obesity 21:935–43 [Google Scholar]
  53. 53. US Food Drug Admin. (FDA) 2014. FDA approves weight-management drug Contrave News Release, Sept. 10. http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm413896.htm
  54. 54. Med. Econ 2000. Topiramate (Topamax tablets). Physicians' Desk2590–95 Montvale, NJ: Med. Econ. [Google Scholar]
  55. Rosenfeld WE. 55.  1997. Topiramate: a review of preclinical, pharmacokinetic and clinical data. Clin. Ther. 19:1294–308 [Google Scholar]
  56. Bray GA, Hollander P, Klein S, Kushner R, Levy B. 56.  et al. 2003. A 6-month randomized, placebo-controlled dose-ranging trial of topiramate for weight loss in obesity. Obes. Res. 11:722–33 [Google Scholar]
  57. Wilding J, Van Gaal L, Rissanen A, Vercruysse F, Fitchet M. 57.  et al. 2004. A randomized double-blind placebo-controlled study of the long-term efficacy and safety of topiramate in the treatment of obese subjects. Int. J. Obes. 28:1399–410 [Google Scholar]
  58. McElroy SL, Arnold LM, Shapira NA, Keck PE Jr., Rosenthal NR. 58.  et al. 2003. Topiramate in the treatment of binge eating disorder associated with obesity: a randomized, placebo-controlled trial. Am. J. Psychiatry 160:255–61 [Google Scholar]
  59. Hoopes SP, Reimherr FW, Hedges DW, Rosenthal NR, Kamin M. 59.  et al. 2003. Treatment of bulimia nervosa with topiramate in a randomized, double-blind, placebo-controlled trial, part 1: improvement in binge and purge measures. J. Clin. Psychiatry 64:1335–41 [Google Scholar]
  60. Van Issum C, Mavrakanas N, Schutz JS, Shaarawy T. 60.  2011. Topiramate-induced acute bilateral angle closure and myopia: pathophysiology and treatment controversies. Eur. J. Ophthalmol. 21:404–9 [Google Scholar]
  61. Hunt S, Russell A, Smithson WH, Parsons L, Robertson I. 61.  et al. 2008. Topiramate in pregnancy: preliminary experience from the UK Epilepsy and Pregnancy Register. Neurology 71:272–76 [Google Scholar]
  62. 62. Food Drug Adm. (FDA) 2011. FDA Drug Safety Communication: risk of oral clefts in children born to mothers taking Topamax (topiramate) News Release, Mar. 4. Accessed Apr. 3, 2011. http://www.fda.gov/Drugs/DrugSafety/ucm245085.htm
  63. Gadde KM, Allison DB, Ryan DH, Peterson CA, Troupin B. 63.  et al. 2011. Effects of low-dose, controlled-release, phentermine plus topiramate combination on weight and associated comorbidities in overweight and obese adults (CONQUER): a randomized, placebo-controlled, phase 3 trial. Lancet 377:1341–52 [Google Scholar]
  64. 64. Vivus, Inc 2008. Qnexa meets primary endpoint by demonstrating superior weight loss over components and placebo in the 28-week equate study (OB-3010). News Release, Dec. 11. http://ir.vivus.com/releasedetail.cfm?ReleaseID=353965
  65. Kelly EM, Tungol AA, Wesolowicz LA. 65.  2013. Formulary management of 2 new agents: lorcaserin and phentermine/topiramate for weight loss. J. Manag. Care Pharm. 19:642–54 [Google Scholar]
  66. 66. Vivus Inc 2010. FDA issues complete response letter to Vivus regarding new drug application for Qnexa® News Release, Oct. 28. http://ir.vivus.com/releasedetail.cfm?ReleaseID=524576
  67. Broberger C, Landry M, Wong H, Walsh JN, Hokfelt T. 67.  1997. Subtypes Y1 and Y2 of the neuropeptide Y receptor are respectively expressed in pro-opiomelanocortin and neuropeptide Y containing neurons of the rat hypothalamic arcuate nucleus. Neuroendocrinology 66:393–408 [Google Scholar]
  68. Erondu N, Gantz I, Musser B, Suryawanshi S, Mallick M. 68.  et al. 2006. Neuropeptide Y5 receptor antagonism does not induce clinically meaningful weight loss in overweight and obese adults. Cell Metab. 4:275–82 [Google Scholar]
  69. Sargent BJ, Moore NA. 69.  2009. New central targets for the treatment of obesity. Br. J. Clin. Pharmacol. 68:852–60 [Google Scholar]
  70. Salem V, Bloom SR. 70.  2010. Approaches to the pharmacological treatment of obesity. Expert Rev. Clin. Pharmacol. 3:73–88 [Google Scholar]
  71. Kim GW, Lin JL, Valentino MA, Colon-Gonzalez F, Waldman SA. 71.  2011. Regulation of appetite to treat obesity. Expert Rev. Clin. Pharmacol. 4:243–59 [Google Scholar]
  72. Lee JW, Ye J, Gao Z, Youn HS, Lee WH. 72.  et al. 2003. Reciprocal modulation of Toll-like receptor-4 signalling pathways involving MyD88 and phosphatidylinositol 3-kinase/AKT by saturated and polyunsaturated fatty acids. J. Biol. Chem. 278:37041–51 [Google Scholar]
  73. Verdich C, Flint A, Gutzwiller JP, Naslünd E, Beglinger C. 73.  et al. 2001. A meta-analysis of the effect of glucagon-like peptide-1 (7–36) amide on ad libitum energy intake in humans. J. Clin. Endocrinol. Metab. 86:4382–89 [Google Scholar]
  74. Gutzwiller JP, Goke B, Drewe J, Hildenbrand P, Ketterer S. 74.  et al. 1999. Glucagon-like peptide 1: a potent regulator of food intake in humans. Gut 44:81–86 [Google Scholar]
  75. Naslünd E, Barkeling B, King N, Gutniak M, Blundell JE. 75.  et al. 1999. Energy intake and appetite are suppressed by glucagon-like peptide-1 (GLP-1) in obese men. Int. J. Obes. 23:304–11 [Google Scholar]
  76. Toft-Nielsen MB, Madsbad S, Holst JJ. 76.  1999. Continuous subcutaneous infusion of glucagon-like peptide 1 lowers plasma glucose and reduces appetite in type 2 diabetic patients. Diabetes Care 22:1137–43 [Google Scholar]
  77. Zander M, Madsbad S, Madsen JL, Holst JJ. 77.  2002. Effect of 6-week course of glucagon-like peptide 1 on glycaemic control, insulin sensitivity and β-cell function in type 2 diabetes: a parallel-group study. Lancet 359:824–30 [Google Scholar]
  78. Riddle MC, Henry RR, Poon TH, Zhang B, Mac SM. 78.  et al. 2006. Exenatide elicits sustained glycaemic control and progressive reduction of body weight in patients with type 2 diabetes inadequately controlled by sulphonylureas with or without metformin. Diabetes Metab. Res. Rev. 22:483–91 [Google Scholar]
  79. Abbot CR, Monteiro M, Small CJ, Sajedi A, Smith KL. 79.  et al. 2005. The inhibitory effects of peripheral administration of peptide YY3–36 and glucagon-like peptide-1 on food intake are attenuated by ablation of the vagal–brainstem–hypothalamic pathway. Brain Res 1044:127–31 [Google Scholar]
  80. Talsania T, Anini Y, Siu S, Drucker DJ, Brubaker PL. 80.  2005. Peripheral exendin-4 and peptide YY3–36 synergistically reduce food intake through different mechanisms in mice. Endocrinology 146:3748–56 [Google Scholar]
  81. Baggio LL, Huang Q, Brown TJ, Drucker DJ. 81.  2004. Oxyntomodulin and glucagon-like peptide-1 differentially regulate murine food intake and energy expenditure. Gastroenterology 127:546–58 [Google Scholar]
  82. Drucker DJ. 82.  2006. The biology of incretin hormones. Cell Metab. 3:153–65 [Google Scholar]
  83. Astrup A, Rössner S, Van Gaal L, Rissanen A, Niskanen L. 83.  et al. 2009. Effects of liraglutide in the treatment of obesity: a randomised, double-blind, placebo-controlled study. Lancet 374:1606–16 [Google Scholar]
  84. Wadden TA, Hollander P, Klein S, Niswender K, Woo V. 84.  et al. 2013. Weight maintenance and additional weight loss with liraglutide after low-calorie-diet-induced weight loss: the SCALE Maintenance randomized study. Int. J. Obes. 37:1443–51 [Google Scholar]
  85. Knudsen LB, Madsen LW, Andersen S, Almholt K, de Boer AS. 85.  et al. 2010. Glucagon-like peptide-1 receptor agonists activate rodent thyroid C-cells causing calcitonin release and C-cell proliferation. Endocrinol 151:1473–86 [Google Scholar]
  86. Lean ME, Carraro R, Finer N, Hartviq H, Lindegaard ML. 86.  et al. 2013. Tolerability of nausea and vomiting and associations with weight loss in a randomized trial of liraglutide in obese, non-diabetic adults. Int. J. Obes. 38:689–97 [Google Scholar]
  87. Tzefos M, Harris K, Brackett A. 87.  2012. Clinical efficacy and safety of once-weekly glucagon-like peptide-1 agonists in development for treatment of type 2 diabetes mellitus in adults. Ann. Pharmacother. 46:68–78 [Google Scholar]
  88. Barnett AH. 88.  2011. Lixisenatide: evidence for its potential use in the treatment of type 2 diabetes. Core Evid. 6:67–79 [Google Scholar]
  89. Pinelli NR, Hurren KM. 89.  2011. Efficacy and safety of long-acting glucagon-like peptide-1 receptor agonists compared with exenatide twice daily and sitagliptin in type 2 diabetes mellitus: a systematic review and meta-analysis. Ann. Pharmacother. 45:850–60 [Google Scholar]
  90. Madsbad S, Kielgast U, Asmar M, Deacon CF, Torekov SS. 90.  et al. 2011. An overview of once-weekly glucagon-like peptide-1 receptor agonists—available efficacy and safety data and perspectives for the future. Diabetes Obes. Metab. 13:394–407 [Google Scholar]
  91. Steinert RE, Poller B, Castelli MC, Drewe J, Beglinger C. 91.  2010. Oral administration of glucagon-like peptide 1 or peptide YY 3–36 affects food intake in healthy male subjects. Am. J. Clin. Nutr. 92:810–17 [Google Scholar]
  92. Beglinger C, Poller B, Arbit E, Ganzoni C, Gass S. 92.  et al. 2008. Pharmacokinetics and pharmacodynamics effects of oral GLP-1 and PYY3–36: a proof-of-concept study in healthy subjects. Clin. Pharmacol. Ther. 84:468–74 [Google Scholar]
  93. Steinert RE, Poller B, Castelli MC, Friedman K, Huber AR. 93.  et al. 2009. Orally administered glucagon-like peptide-1 affects glucose homeostasis following a glucose tolerance test in healthy male subjects. Clin. Pharmacol. Ther. 86:644–50 [Google Scholar]
  94. Pironi L, Stanghellini V, Miglioli V, Corinaldesi R, De Giorgio R. 94.  et al. 1993. Fat-induced ileal brake in humans: a dose-dependent phenomenon correlated to the plasma levels of peptide YY. Gastroenterology 105:733–39 [Google Scholar]
  95. Batterham RL, Cohen MA, Ellis SM, Le Roux CW, Withers DJ. 95.  et al. 2003. Inhibition of food intake in obese subjects by peptide YY3–36. N. Engl. J. Med. 349:941–48 [Google Scholar]
  96. Degen L, Oesch S, Casanova M, Graf S, Ketterer S. 96.  et al. 2005. Effect of peptide YY3–36 on food intake in humans. Gastroenterology 129:1430–36 [Google Scholar]
  97. Field BCT, Wren AM, Peters V, Baynes KCR, Martin NM. 97.  et al. 2010. PYY3–36 and oxyntomodulin can be additive in their effect on food intake in overweight and obese humans. Diabetes 59:1635–39 [Google Scholar]
  98. Gantz I, Erondu N, Mallick M, Musser B, Krishna R. 98.  et al. 2007. Efficacy and safety of intranasal peptide YY3–36 for weight reduction in obese adults. J. Clin. Endocrinol. Metab. 92:1754–57 [Google Scholar]
  99. Scott V, Kimura N, Stark JA, Luckman SM. 99.  2005. Intravenous peptide YY3–36 and Y2 receptor antagonism in the rat: effects on feeding behaviour. J. Neuroendocrinol. 17:452–57 [Google Scholar]
  100. Halatchev IG, Ellacott KLJ, Fan W, Cone RD. 100.  2004. Peptide YY3–36 inhibits food intake in mice through a melanocortin-4 receptor-independent mechanism. Endocrinology 145:2585–90 [Google Scholar]
  101. Adams SH, Won WB, Schonhoff SE, Leiter AB, Paterniti JR. 101.  2004. Effects of peptide YY[3–36] on short-term food intake in mice are not affected by prevailing plasma ghrelin levels. Endocrinology 145:4967–75 [Google Scholar]
  102. Challis BG, Coll AP, Yeo GSH, Pinnock SB, Dickson SL. 102.  et al. 2004. Mice lacking pro-opiomelanocortin are sensitive to high-fat feeding but respond normally to the acute anorectic effects of peptide-YY3–36. PNAS 101:4695–700 [Google Scholar]
  103. Pittner RA, Moore CX, Bhavsar SP, Gedulin BR, Smith PA. 103.  et al. 2004. Effects of PYY[3–36] in rodent models of diabetes and obesity. Int. J. Obes. 28:963–71 [Google Scholar]
  104. Chelikani PK, Haver AC, Reeve JR, Keire DA, Reidelburger RD. 104.  2006. Daily, intermittent intravenous infusion of peptide YY(3–36) reduces daily food intake and adiposity in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 290:R298–305 [Google Scholar]
  105. Moran TH, Smedh U, Kinzig KP, Scott KA, Knipp S. 105.  et al. 2005. Peptide YY(3–36) inhibits gastric emptying and produces acute reductions in food intake in rhesus monkeys. Am. J. Physiol. Regul. Integr. Comp. Physiol. 288:R384–88 [Google Scholar]
  106. Batterham RL, Cowley MA, Small CJ, Herzog H, Cohen MA. 106.  et al. 2002. Gut hormone PY3–36 physiologically inhibits food intake. Nature 418:650–54 [Google Scholar]
  107. Dakin CL, Gunn I, Small CJ, Edwards CM, Hay DL. 107.  et al. 2001. Oxyntomodulin inhibits food intake in the rat. Endocrinology 142:4244–50 [Google Scholar]
  108. Dakin CL, Small CJ, Batterham RL, Neary NM, Cohen MA. 108.  et al. 2004. Peripheral oxyntomodulin reduces food intake and body weight gain in rats. Endocrinology 145:2687–95 [Google Scholar]
  109. Yamamoto H, Lee CE, Marcus JN, Williams TD, Overton JM. 109.  et al. 2002. Glucagon-like peptide-1 receptor stimulation increases blood pressure and heart rate and activates autonomic regulatory neurons. J. Clin. Investig. 110:43–52 [Google Scholar]
  110. Cohen MA, Ellis SM, Le Roux CW, Batterham RL, Park A. 110.  et al. 2003. Oxyntomodulin suppresses appetite and reduces food intake in humans. J. Clin. Endocrinol. Metab. 88:4696–701 [Google Scholar]
  111. Wynne K, Park AJ, Small CJ, Patterson M, Ellis SM. 111.  et al. 2005. Subcutaneous oxyntomodulin reduces body weight in overweight and obese subjects: a double-blind, randomized, controlled trial. Diabetes 54:2390–95 [Google Scholar]
  112. Wynne K, Park AJ, Small CJ, Meeran K, Ghatei MA. 112.  et al. 2006. Oxyntomodulin increases energy expenditure in addition to decreasing energy intake in overweight and obese humans: a randomised controlled trial. Int. J. Obes. 30:1729–36 [Google Scholar]
  113. Santoprete A, Capito E, Carrington PE, Pocai A, Finotto M. 113.  et al. 2011. DPP-IV-resistant, long-acting oxyntomodulin derivatives. J. Pept. Sci. 17:270–80 [Google Scholar]
  114. Kerr BD, Flatt PR, Gault VA. 114.  2010. (d-Ser2)Oxm[mPEG-PAL]: a novel chemically modified analogue of oxyntomodulin with anti-hyperglycaemic, insulinotropic and anorexigenic actions. Biochem. Pharmacol. 80:1727–35 [Google Scholar]
  115. Cummings DE, Foster-Schubert KE, Overduin J. 115.  2005. Ghrelin and energy balance: focus on current controversies. Curr. Drug Targets 6:153–69 [Google Scholar]
  116. Wren AM, Seal LJ, Cohen AJ, Brynes AE, Frost GS. 116.  et al. 2001. Ghrelin enhances appetite and increases food intake in humans. J. Clin. Endocrinol. Metab. 86:5992–95 [Google Scholar]
  117. Zorrilla EP, Iwasaki S, Moss JA, Chang J, Otsuji J. 117.  et al. 2006. Vaccination against weight gain. PNAS 103:13226–31 [Google Scholar]
  118. Xin Z, Serby MD, Zhao H, Kosogof C, Szczepankiewicz BG. 118.  et al. 2006. Discovery and pharmacological evaluation of growth hormone secretagogue receptor antagonists. J. Med. Chem. 49:4459–69 [Google Scholar]
  119. Yang J, Zhao TJ, Goldstein JL, Brown MS. 119.  2008. Inhibition of ghrelin O-acyltransferase (GOAT) by octanoylated penta-peptides. PNAS 105:10750–55 [Google Scholar]
  120. Katsuura G, Asakawa A, Inui A. 120.  2002. Roles of pancreatic polypeptide in regulation of food intake. Peptides 23:323–29 [Google Scholar]
  121. Asakawa A, Inui A, Ueno N, Fujimiya M, Fujino MA. 121.  et al. 1999. Mouse pancreatic polypeptide modulates food intake, while not influencing anxiety in mice. Peptides 20:1445–48 [Google Scholar]
  122. Batterham RL, Le Roux CW, Cohen MA, Park AJ, Ellis SM. 122.  et al. 2003. Pancreatic polypeptide reduces appetite and food intake in humans. J. Clin. Endocrinol. Metab. 88:3989–92 [Google Scholar]
  123. Jesudason DR, Monteiro MP, McGowan BM, Neary NM, Park AJ. 123.  et al. 2007. Low-dose pancreatic polypeptide inhibits food intake in man. Br. J. Nutr. 97:426–29 [Google Scholar]
  124. Neary NM, McGowan BM, Monteiro MP, Jesudason DR, Ghatei MA. 124.  et al. 2008. No evidence of an additive inhibitory feeding effect following PP and PYY3–36 administration. Int. J. Obes. 32:1438–40 [Google Scholar]
  125. 125. 7TM Pharma 2007. 7TM Pharma obtains patent protection for the use of both obinepitide and TM 30339 in the treatment of obesity News Release, Apr. 12. Accessed Apr. 12, 2012
  126. Lutz TA, Althaus J, Rossi R, Scharrer E. 126.  1998. Anorectic effect of amylin is not transmitted by capsaicin-sensitive nerve fibers. Am. J. Physiol. 274:R1777–82 [Google Scholar]
  127. Roth JD, Anderson C. 127.  2006. Anti-obesity effects of the β-cell hormone amylin in diet-induced obese rats: effects on food intake, body weight, composition, energy expenditure and gene expression. Endocrinology 147:5855–64 [Google Scholar]
  128. Rushing PA, Hagan MM, Seeley RJ, Lutz TA, Woods SC. 128.  2000. Amylin: a novel action in the brain to reduce body weight. Endocrinology 141:850–53 [Google Scholar]
  129. Lutz TA, Geary N, Szabady MM, Del Prete E, Scharrer E. 129.  1995. Amylin decreases meal size in rats. Physiol. Behav. 58:1197–202 [Google Scholar]
  130. Young AA. 130.  1997. Amylin physiology and its role in diabetes. Curr. Opin. Endocrinol. Diabetes 4:282–90 [Google Scholar]
  131. Young AA, Vine W, Gedulin BR, Pittner R, Janes S. 131.  et al. 1996. Preclinical pharmacology of pramlintide in the rat: comparisons with human and rat amylin. Drug Dev. Res. 37:231–48 [Google Scholar]
  132. Hollander P, Maggs DG, Ruggles JA, Fineman M, Shen L. 132.  et al. 2004. Effect of pramlintide on weight in overweight and obese insulin-treated type 2 diabetes patients. Obes. Res. 12:661–68 [Google Scholar]
  133. Chapman I, Parker B, Doran S, Feinle-Bisset C, Wishart J. 133.  et al. 2005. Effect of pramlintide on satiety and food intake in obese subjects and subjects with type 2 diabetes. Diabetologia 48:838–48 [Google Scholar]
  134. Hollander PA, Levy P, Fineman MS, Maggs DG, Shen LZ. 134.  et al. 2003. Pramlintide as an adjunct to insulin therapy improves long-term glycemic and weight control in patients with type 2 diabetes: a 1-year randomized controlled trial. Diabetes Care 26:784–90 [Google Scholar]
  135. Aronne L, Fujioka K, Aroda V, Chen K, Halseth A. 135.  et al. 2007. Progressive reduction in body weight after treatment with the amylin analogue pramlintide in obese subjects: a phase 2 randomized, placebo-controlled, dose-escalation study. J. Clin. Endocrinol. Metab. 92:2977–83 [Google Scholar]
  136. Roth JD, Coffey T, Jodka CM, Maier H, Athanacio JR. 136.  et al. 2007. Combination therapy with amylin and peptide YY[3–36] in obese rodents: anorexigenic synergy and weight loss additivity. Endocrinology 148:6054–61 [Google Scholar]
  137. Aronne LJ, Halseth AE, Burns CM, Miller S, Shen LZ. 137.  2010. Enhanced weight loss following co-administration of pramlintide with sibutramine or phentermine in a multicentre trial. Obesity 18:1739–46 [Google Scholar]
  138. Wang T, Jia W, Hu C. 138.  2015. Advancement in genetic variants conferring obesity susceptibility from genome-wide association studies. Front. Med. 9:146–61 [Google Scholar]
  139. Basile KJ, Johnson ME, Xia Q, Grant SF. 139.  2014. Genetic susceptibility to type 2 diabetes and obesity: follow-up findings from genome-wide association studies. Int. J. Endocrinol. 2014:769671 [Google Scholar]
/content/journals/10.1146/annurev-pharmtox-010716-104735
Loading
/content/journals/10.1146/annurev-pharmtox-010716-104735
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error