Abstract
The rising tide of obesity and its related disorders is one of the most pressing health concerns worldwide, yet existing medicines to combat the problem are disappointingly limited in number and effectiveness. Recent advances in mechanistic insights into the neuroendocrine regulation of body weight have revealed an expanding list of molecular targets for novel, rationally designed antiobesity pharmaceutical agents. Antiobesity drugs act via any of four mechanisms: 1) decreasing energy intake, 2) increasing energy expenditure or modulating lipid metabolism, 3) modulating fat stores or adipocyte differentiation, and 4) mimicking caloric restriction. Various novel drug candidates and targets directed against obesity are currently being explored. A few of them are also in the later phases of clinical trials. This review discusses the development of novel antiobesity drugs based on current understanding of energy homeostasis
Keywords: Obesity, Pharmacotherapy, Drug targets.
INTRODUCTION
Obesity has progressed to epidemic proportions globally, with more than 1.6 billion adults overweight and at least 400 million of them clinically obese [1]. It is a leading contributor to the global burden of chronic diseases like type 2 diabetes, cardiovascular disease, hypertension, stroke, and certain forms of cancer. The health outcomes range from increased risk of premature death to serious chronic conditions that reduce the overall quality of life [1]. Each year, an estimated 300,000 adults in the United States die of causes related to obesity [2]
According to the guidelines of the National Institute of Health (NIH) regarding the treatment of obesity, “weight loss drugs approved by the FDA [US Food and Drug Administration] may only be used as part of a comprehensive weight loss program, including dietary therapy and physical activity, for patients with a BMI [body mass index] of ≥30 with no concomitant obesity-related risk factors or diseases, and for patients with a BMI of ≥27 with concomitant obesity-related risk factors or diseases”[3].The risk factors and diseases considered important enough to warrant pharmacotherapy at a BMI of 27--29.9 are hypertension, dyslipidemia, coronary heart disease (CHD), type 2 diabetes, and sleep apnea [3]. In 2007--2008, the prevalence of American adults having a BMI greater than 30 was 32.2% among men and 35.5% among women [4]. In light of the NIH guidelines, this startling preponderance of obesity indicates pharmacotherapy is warranted for more than one-third of all American adults. Yet, existing medicines to combat the problem are disappointingly few in number and efficacy. In the past few years, understanding of the mechanisms involved in regulating feeding behavior and energy expenditure has evolved considerably, and targeting these mechanisms for drug development has garnered greater attention as well. This review discusses the neuroendocrine control of energy homeostasis, drug targets, and the possible future range of anti-obesity agents.
PATHOPHYSIOLOGY OF OBESITY
The etiology of obesity is complex. Genetic, environmental, and psychological factors are implicated in its causation. Obesity is a disorder of energy balance. When food-derived energy chronically exceeds energy expenditure, the excess calories are stored as triglycerides in adipose tissue. Notwithstanding the marked fluctuations in daily food intake, body weight remains remarkably stable in most humans. In response to alterations in body adiposity, the brain triggers compensatory physiological adaptations that resist weight change. The two sides of the energy equation, i.e., consumption and expenditure, are finely regulated by neural and humoral mechanisms [5,6]. These neural and humoral mechanisms work in concert as the neurohumoral system to maintain body energy status. The neurohumoral system is comprised of the afferent pathway, the central processing unit, and the efferent pathway [5] (Fig. 1).
The afferent system generates humoral signals from adipose tissue (leptin), pancreas (insulin), and stomach (ghrelin). Acting in the brain, leptin and, to a lesser extent, insulin decrease food intake and increase energy expenditure, promoting weight loss. They are consequently termed catabolic adiposity signals. Impinging on the same neuronal targets, ghrelin exerts opposite effects and is thus an anabolic hormone. The amount of leptin is in direct proportion to adipose stores. Weight gain evokes proportionate increases in catabolic hormones and decreases ghrelin, whereas weight loss causes the opposite [5]. Long-term regulation of energy stores depends on adiposity signals like leptin and insulin, while ghrelin communicates short-term changes in nutritional status. Apart from ghrelin, many other gastrointestinal signals communicate short-term alterations in nutritional status to the brain via humoral and neural stimuli. A number of satiety signals that are released from the intestine include cholecystokinin (CCK), pancreatic polypeptide (PP), peptide-YY (PYY), oxyntomodulin, enterostatin, and glucagon -like peptide-1 (GLP-1) [5-10]. Another satiety signal, amylin, is cosecreted with insulin from the pancreas. These short-acting satiety and hunger signals are transmitted via afferent fibers of the vagus nerve to the caudal brainstem in nucleus tractus solitarius, from where they also influence the hypothalamus. The sensitivity of brainstem responses to afferent gastrointestinal signals is modulated by long-acting catabolic adiposity signals [5] (Fig. 2).
The central processing unit is located primarily in the hypothalamus, especially in the arcuate nucleus, which integrates the afferent signals from leptin, insulin, and ghrelin. There are two major types of neurons in this locale that bear leptin receptors; these are anorexigenic neurons and orexigenic neurons. Pro-opiomelanocortin (POMC) and cocaine- and amphetamine-related transcript (CART) neurons are first-order anorexigenic neurons. These are activated by leptin and secrete α-melanocyte stimulating hormone (α-MSH), which acts on second-order neurons present in the paraventricular area that express melanocortin 4 receptor(MC4R). The signaling through these pathways induces expression of corticotrophic-releasing hormone (CRH) and thyrotrophin-releasing hormone (TRH) [5]. Neuropeptide Y (NPY)/ Agouti-related peptide (AgRP) neurons are the first-order orexigenic neurons. They produce NPY and AgRP, which are the initial targets of leptin action. NPY acts on second-order neurons in the lateral hypothalamic area and the perifornical area. The second-order neurons express orexigenic peptides that are melanin-concentrating hormone (MCH) and orexins A and B. The signaling through these neurons is increased by ghrelin and attenuated by leptin. AgRP exerts its anabolic effect by inhibiting the anorexigenic or melanocortin arm [5].
The efferent system carries out orders from the hypothalamic nuclei in the form of feeding behavior and energy expenditure [5]. Anorexigenic signaling through TRH and CRH induces anorexia and thermogenesis. On the other hand, the orexigenic signaling via orexigenic peptides (MCH and orexin A and B neuropeptides) results in increased food intake and decreased energy expenditure.
Several neurotransmitters (serotonin, noradrenaline, dopamine, and histamine), peptides (neuromedin U, urocortin, bombesin, amylin, galanin), hormones (thyroid hormone, growth hormone) and cytokines (ciliary neurotrophic factor) also play a role in modulating feeding behavior and energy expenditure [5-10].
LEPTIN AND LEPTIN RESISTANCE
No discovery has had a greater impact on obesity research than that of leptin in 1994 [11]. Leptin is the product of OB gene, a 146 amino acid protein, secreted from white adipose tissue [12]. The gene encoding for leptin is mutated in genetically obese (ob/ob) mice. The gene encoding for the leptin receptor, a class I cytokine receptor, is mutated in fatty (fa/fa) rats and in diabetic (db/db) mice [12]. Leptin binds to its receptor and activates Janus kinase and STAT (signal transducers and activators of transcription) signal transduction pathway. STAT3 appears to be an important transcription factor recruited during signaling. It translocates to the nucleus and induces the production of anorectic neuropeptide POMC, while repressing the production of orexigenic neuropeptide AgRP. It also induces the expression of suppressor of cytokine signaling-3, which is a negative regulator of leptin signaling [13].
Leptin is an important hormone that maintains body weight by acting through a negative feedback mechanism of energy consumption and utilization. The finding that leptin deficiency in animals results in morbid obesity generated hope that exogenous administration of leptin would ameliorate obesity [11]. However, as common obesity is a state of leptin resistance, the exogenous administration of even exceedingly high doses of leptin has proven relatively ineffective at reducing body weight [14-16].
In the past few years, insight into the mechanisms of leptin resistance has been elucidated. Leptin resistance results from impairments in leptin action at multiple levels. Leptin is usually transported across the blood-brain barrier by a specialized leptin-transporter that is impaired in obesity [17]. In mice, a high-fat diet has also been shown to impair leptin signaling by attenuating STAT3 activation, thus contributing to leptin resistance [18].Leptin receptor activation engages intracellular proteins, such as protein tyrosine phosphatase1 B (PTP1B) and suppressor of cytokine signaling-3, that terminate receptor signaling. Increased activity of these autoinhibitory factors attenuates leptin signaling and is also implicated in leptin resistance [17]. All of these factors are targeted in drug development in order to increase leptin sensitivity in obese individuals (Fig. 3).
PHARMACOTHERAPY OF OBESITY
Obesity is a state of sustained disequilibrium between energy intake and energy expenditure. The pharmacotherapies for obesity are directed at the following: 1) decreasing energy intake (e.g. appetite suppressants and lipase inhibitors), 2) increasing energy expenditure, 3) modulating fat storage or adipocyte differentiation, or 4) mimicking caloric restriction [8,19] (Fig. 4).
The history of the pharmacotherapy of obesity dates back to the 1930s. Many drugs used to treat obesity in the past have been discontinued because of their potential to be abused and their toxicities [20-27] (Table 1). Rimonabant is a cannabinoid receptor 1 antagonist that became available for long-term treatment of obesity in the United Kingdom and other countries beginning in 2006 [27]. In the United States, the FDA did not approve the agent because of its association with a risk of psychiatric disorders including depression and suicidal tendencies [27]. In 2009, the EMEA (European Medicine Agency) withdrew market authorisation for rimonabant in all countries of the European Union due to safety concerns [21]. Sibutramine, a popular antiobesity drug used worldwide since 1997, was withdrawn from the European and US markets in 2010 because of safety concerns [27,28]. In the Sibutramine Cardiovascular Outcomes (SCOUT) trial, the drug was shown to be associated with increased risk of major adverse cardiovascular events (a composite of non-fatal heart attack, non-fatal stroke, resuscitation after cardiac arrest, and cardiovascular death) in obese patients, as compared to placebo [29].
Table 1.
Drugs Used in Past (Year of Introduction) | Year of Discontinuation | Cause of Discontinuation |
---|---|---|
Thyroid hormone | Off label use till 1980s despite toxicity | Hyperthyroidism, cardiac arrhythmias, and sudden death |
Dinitrophenol (introduction in 1930) | Banned by the FDA in 1938 | Dermatitis, neuropathy, agranulocytosis, visual impairment, death |
Amphetamine and its derivatives (1936) | Became a schedule II drug under the Controlled Substances Act in 1971 | Addiction, hypertension, myocardial toxicity |
Aminorex (1965 in Europe) | 1968 | Chronic pulmonary hypertension resulting in high mortality |
Fenfluramine + phentermine (1992) | 1997 | Valvular heart disease |
Phenylpropanolamine(available since 1970) | 2000 | Hemorrhagic stroke |
Rimonabant(available in UK and other European countries since 2006 for long term treatment of obesity) | EMEA withdrew marketing authorization in 2009 Failed to get US-FDA approval due to safety factors | Psychiatric disorders, depression and suicidal ideation |
Sibutramine (popular antiobesity drug used since 1997) | 2010 | Increase in risk of major adverse cardiovascular events (a composite of non-fatal heart attack, non-fatal stroke, resuscitation after cardiac arrest and cardiovascular death) |
(Abbreviation = FDA: Food and Drug Administration; EMEA= European Medicine Agency)
The medications currently approved by the FDA for use in the United States include phendimetrazine, benzphetamine, diethylpropion, phentermine, and orlistat. The US Drug Enforcement Administration classifies phendimetrazine and benzphetamine as schedule 3 drugs and diethylpropion and phentermine as schedule 4 drugs [20,30]. These drugs are recommended for the short-term (12 weeks or less) treatment of obesity [20,30].
Orlistat is the only drug currently approved by the FDA for long-term management of obesity [21,31]. It acts by reversibly inhibiting pancreatic lipase enzyme, thus preventing the hydrolyzation and absorption of dietary fat by approximately 30%, and thereby decreasing the caloric intake of obese patients [32]. A meta-analysis of 29 studies of orlistat for weight loss in adult patients reported a weight loss of 2.59 kg and 2.89 kg at 6 months and 12 months, respectively [33,34]. In comparison to patients on diet and placebo only, the patients treated with orlistat demonstrated a significant improvement in blood glucose concentrations, insulin resistance, waist circumference, total and low-density lipoprotein cholesterol (LDL-C), and blood pressure [35-37]. Owing to the unabsorbed fat in the intestine, orlistat causes some side effects, among which the most common are diarrhea, flatulence, bloating, abdominal pain, and dyspepsia [38].
The only currently approved long-term therapy for obesity in the United States is not only having limited efficacy, but is also associated with number of side effects. Thus, a dire need exists for the development of novel antiobesity drugs with high efficacy and long-term safety. In recent years, there has been an explosion in knowledge of the mechanisms involved in the regulation of feeding behavior and the cellular mechanisms involved in energy expenditure. This has led to a rich harvest of new potential targets and drug candidates that may offer hope of safe and effective treatments for obesity.
NOVEL TARGETS AND DRUG CANDIDATES
Drugs that Decrease Energy Intake
These include appetite suppressants and lipase inhibitors. Among the appetite suppressants, the targets are classified according to the energy pathways they modulate. These can either be stimulators of anorexigenic pathways or inhibitors of orexigenic pathways (Fig. 4). The lipase inhibitors inhibit dietary fat absorption and hence decrease energy intake.
Appetite Suppressants: Stimulators of anorexigenic pathway
Leptin
Leptin treatment has proved to be ineffective for attaining weight reduction in obese individuals. However, the use of low doses of leptin to maintain weight loss attained by other anorectic medicines and lifestyle modifications is emerging as a pharmacotherapeutic option [39,40]. In human beings, the maintenance of reduced body weight is achieved via coordination of metabolic, neuroendocrine, and autonomic responses that normally favor weight regain following a loss [40]. Weight loss decreases energy expenditure, reduces the tone of the sympathetic nervous system, and decreases the circulating levels of leptin and thyroid hormones [40]. The weight-reduced state is considered to be a state of relative leptin deficiency [41-43]. These counter-regulatory changes induce hyperphagia and a hypometabolic state, which result in tachyphylaxis to anorectic medications and favor weight regain. Administration of low doses of leptin in humans after weight loss has been shown to reverse these counter-regulatory neuroendocrine responses and increase energy expenditure, thus maintaining the weight reduction [40,44].
Leptin mimetics
To overcome leptin resistance (Fig. 3), drugs that can bypass the leptin transporter are being explored. These include oral leptin mimetics and intranasal leptin, both of which are in preclinical stages of drug development [15,45,46].
Stimulating leptin signaling
Recent preclinical evidence has shown that coadministration of amylin with leptin improves leptin-mediated STAT3 signaling in the ventromedial hypothalamus of rats with diet-induced obesity [47]. This observation suggests that combination therapy with leptin and amylin has potential for the treatment of obesity. One such combination therapy is currently being evaluated in clinical trials and is discussed later in this article.
Another strategy to improve sensitivity to leptin is to inhibit the autoinhibitory factors involved in leptin signaling, i.e., suppressor of cytokine signaling-3 and PTP1B. This strategy is particularly compelling as the activity of these autoinhibitory factors is increased in obesity, hinting at an etiological role for them in leptin resistance [15, 48,49]. Suppressor of cytokine signaling-3 haploinsufficiency in mice enhances the weight-reducing effects of leptin and confers resistance to diet-induced obesity [50]. PTP1B is a negative regulator of the leptin and insulin signaling pathways, so it has emerged as an attractive novel target for the treatment of both type 2 diabetes and obesity [51]. The development of PTP1B inhibitors for the treatment of obesity is in preclinical stages. One PTP1B inhibitor, trodusquemine, has been shown to suppress appetite, reduce body weight, and improve plasma insulin and leptin levels in a murine model of diet-induced obesity [52].
Another logical strategy to overcome leptin resistance is to manipulate leptin-regulated pathways distal to the first-order neuron, which is the melanocortin pathway.
Melanocortin pathway stimulation
In the leptin-melanocortin pathway, POMC is the first key intermediary downstream of leptin-receptor signaling. Pharmacological mechanisms to increase POMC expression are not evident, due to the involvement of POMC-derived peptides in adrenal physiology [53]. Hence, the downstream pathways are targeted for drug development.
Melanocortin receptor agonists
Cleavage of POMC produces, among other peptides, α-MSH, which activates melanocortin 3 and 4 receptors (Mc3r, Mc4r) to exert catabolic effects [54]. These receptors are highly promising targets for obesity treatment because of their vital functions and relative specificity in energy homeostasis [55]. Ample genetic evidence proves Mc4r signaling to be a critical component of the body-weight regulation system. Null mutations in Mc4r cause pronounced, dominantly inherited, monogenic obesity in rodents and humans, and are associated with increased food intake, decreased energy expenditure, and increased lean body and fat mass [56,57]. These observations have led to the development of Mc4 receptor agonists; however, their development has proceeded less successfully than expected due to the melanocortin pathway’s role in increasing heart rate and blood pressure as well as induction of penile erection [58,59]. These effects cannot be dissociated from that of appetite. There is also ample evidence to demonstrate that stimulation of the melanocortin pathway by inflammatory cytokines results in cachexia. This is another issue of concern with long-term use of melanocortin agonists [60,61]. Some Mc4 receptor agonists have been tested in preclinical and clinical studies. A melanocortin 4 receptor agonist, 3 RY764, reduced food intake and augmented erectile activity in rodents [62]. MK-0493, another potent and selective agonist of Mc4 receptor, was associated with statistically insignificant weight reductions in a clinical trial[63]. (Table 3) Given these discouraging results, Mc4 receptor agonists are currently not being developed for the treatment of obesity. However, they are being explored for other conditions such as sexual dysfunction [64]. Melanocortin antagonists are also being evaluated for the treatment of cachexia [65,66].
Table 3.
Stage of Development in which the Drug Failed or was Not Pursued Further | Drug | Pharmacological Approach | Pharmaceutical Company | Comment | Reference |
---|---|---|---|---|---|
NDA rejected by FDA | Lorcaserin | Selective serotonin receptor agonist | Arena Pharmaceuticals Inc | High doses of the drug associated with tumor formation in rats | Smith et al. (2010) [83], Fidler et al. (2011) [84], Pollack A (2010) [85] |
Qnexa | Low doses of phentermine and topiramate | Vivus Inc | Rejected due to safety concerns including increased heart rate, teratogenic potential and psychiatric problems | Gadde et al. (2011) [246], Pollack A (2010) [247] | |
Contrave | Fixed dose combination of bupropion and naltrexone | Orexigen Therapeutics | Deferred approval till cardiovascular safety concerns are addressed | Greenway et al. (2010) [240], Wadden et al. (2011) [242], Pollack A (2011) [243] | |
Phase 3 | Axokine | Recombinant CNTF | Regeneron pharmaceuticals Inc | Failed due to development of antibodies against CNTF | Ettinger et al. (2003) [74], Pollack A (2003) [75] |
Phase 2 | PYY3-36 (nasal) | Neuropeptide presynaptic Y2 receptor agonist | Nastech Pharmaceutical Company | Did not meet primary efficacy endpoint | Gantz et al. (2007) [131] |
GI181771X | CCK1 receptor agonists | Glaxo Smithkline | Failed to demonstrate efficacy | Jordan et al. (2008) [116] | |
AOD 9604 | Growth hormone analogue | Metabolic Pharmaceuticals | Drug failed to demonstrate efficacy in phase 2b studies | Valentino et al. (2010) [192] | |
MK0557 | Neuropeptide Y 5 receptor antagonist | Merck & Co., Inc. | Failed to demonstrate efficacy | Erondu et al. (2006) [155] | |
SCH497079 | Histamine 3 receptor antagonist | Schering plough | Current status not known | ClinicalTrial.gov [92] | |
MK-0493 | Melanocortin 4 receptor agonist | Merck & Co., Inc. | Lack of efficacy | Krishna et al. (2009) [63] | |
Pramlinitide and metreleptin | Combination of long-term adiposity signal eg. leptin (metreleptin, analog of human leptin) and short-term satiety signals e.g. amylin (pramlintide, analog of amylin) | Amylin Pharmaceuticals Inc, Takeda Pharmaceuticals Limited | Discountinued the project after commercial reassessment | Ravussin et al. (2009) [248], Tam et al. (2011) [249] | |
Hoodia P57 extract | Appetite suppression | Phytopharm, Pfizer Inc,Unilever | Difficulty in synthesizing the extract in drug form, Inconclusive data on efficacy and safety | Vermaak et al. (2011) [94], Bray et al. (2007) [95] | |
Famoxin | Proteolytic cleavage product of adipocyte complement-related protein (Acrp30) | Genset Corporation (acquired by Serono Co. in 2003) | Current status not known | Cheetham et al. (2004) [220] | |
N-5984 | β3 adrenoceptor agonist | Nisshin Kyorin Pharmaceuticals Co. | Current status not known | Farrigan et al. (2002) [202] |
(Abbreviations: NDA: new drug application; FDA: Food and Drug Administration; CCK= cholecystokinin; PYY3-36: peptide YY3-36; CNTF: ciliary neurotrophic factor)
Melanocortin receptor signaling
Recent evidence indicates the single-minded homologue 1 (SIM1) transcription factor acts as a proximal mediator for the anorectic, but not thermogenic, effects of melanocortins [67]. In rodents and in humans genetic evidence demonstrates that the loss of SIM1 causes hyperphagic obesity in addition to causing resistance to the anorectic effects of melanocortins [68,69]. Conversely, SIM1 overexpression reduces food ingestion and body weight in mice fed a high-fat diet, acting downstream of melanocortin receptors [70]. These observations identify SIM1 stimulation as a potential antiobesity strategy.
Ciliary neurotrophic factor
Ciliary neurotrophic factor (CNTF) is a glial cell-produced neuroprotective cytokine. It has been explored for the treatment of neurodegenerative diseases. Unexpectedly, subjects receiving CNTF in clinical trials for this indication experienced weight losses of 10--15%, prompting researchers to consider using CNTF to treat obesity [71]. CNTF either cross-reacts with leptin receptors or directly activates its own receptors present on the hypothalamus, initiating a transduction pathway analogous to that of leptin [72]. In hypothalamic feeding centers, CNTF stimulates the proliferation of neurons that contain leptin-responsive elements [73]. Based on these promising findings, axokine, a recombinant human variant of CNTF, was used for testing in humans. Modestly successful results were observed in phase 1 and 2 clinical trials [74]. However, in one year-long phase 3 trial involving 2,000 severely obese patients, disappointing results were seen in the axokine-treated group [75]. In this trial, axokine resulted in an average weight loss of 2.9 kg, as compared to an average weight loss of 1.1 kg with placebo. While the difference was considered statistically significant, it fell short of the goal set by the FDA for the approval of antiobesity drugs, which is a 5% weight loss beyond that achieved with placebo. (Table 3) This limited efficacy was due to the development of anti-CNTF antibodies in these patients [75]. CNTF congeners that do not elicit an immune response would be rational antiobesity drug candidates in the future [15].
Reuptake inhibitor of serotonin, noradrenaline, and dopamine
Tesofensine increases monoaminergic transmission by inhibiting the neuronal uptake of serotonin, dopamine, and noradrenaline, thus causing appetite suppression. In phase 2b clinical trials, this drug achieved degrees of weight loss that were significantly greater than those achieved with any other currently available antiobesity drug [76]. Over a period of 6 months, patients lost an average of 12.8 kg, 11.3 kg, and 6.7 kg on the 1 mg, 0.5 mg, and 0.25 mg doses, respectively, as compared to a 2.2 kg weight loss in the placebo group [76]. The most common adverse effects in the tesofensine group were dry mouth, nausea, constipation, hard stools, diarrhea, and insomnia. Tesofensine also increased blood pressure, heart rate, and frequency of mood changes. Tesofensine at doses of 0.5 mg and 1.0 mg increased heart rate by 7.4 and 8.1 beats per min, respectively [76]. This effect on heart rate is an important safety issue that needs special attention in future trials since obese individuals are at increased cardiovascular risk. Another issue of concern that must be comprehensively explored in future trials is the association between tesofensine and increased frequency of agitation and mood changes. Its efficacy and tolerability is currently being evaluated in a phase 3 trial [77] (Table 2).
Table 2.
Current Development Status | Drug | Pharmacological Approach | Pharmaceutical Company | References |
---|---|---|---|---|
Phase 3 | Cetilistat | Pancreatic lipase inhibitor | Amylin Pharmaceuticals Inc. /Takeda Pharmaceutical Co. Ltd | Kopelman et al. (2007) [185], Kopelman et al. (2010) [186], Padwal R (2008) [187] |
Tesofensine | Reuptake inhibitor of noradrenaline, dopamine and serotonin | NeuroSearch A/S | Astrup et al. (2008) [76], Bello et al. (2009) [77] | |
Liraglutide | GLP1 agonist | Novo Nordisk A/S | Astrup et al. (2009) [127], Neary et al. (2009) [128] | |
Phase 2 | Obinepitide | Analogue of PYY3-36 and PP. Agonist of neuropeptide Y2 and Y4 receptor | 7TM Pharma | Neary et al. (2009) [128] |
Empatic | Combination of bupropion and zonisamide | Orexigen Therapeutics Inc | Valentino et al. (2010) [192] | |
Phase 1 | OAP 189 (TKS-1225) | Oxyntomodulin analogue | Pfizer Inc (previously developed by Thiakis/ Wyeth) | Wyne et al. (2006) [135], Bloomgarden ZT (2009) [136], McGavigan et al. (2012) [137] |
(Abbreviations : PYY3-36: peptide YY3-36; PP= pancreatic polypeptide; GLP1: glucagon- like peptide1)
Serotonin (5- hydroxytryptamine or 5HT) receptor modulation
The nonselective serotonergic antiobesity drugs fenfluramine and dexfenfluramine have been associated with severe side effects like valvular heart disease and pulmonary hypertension; as a result, these drugs were discontinued [78,79]. A detailed analysis of the antiobesity action of fenfluramine disclosed that it directly activates hypothalamic POMC neurons through 5-hydroxytryptamine 2C (5-HT2C) receptors that are expressed on a majority of these cells [80]. This finding led to selective targeting of 5-HT2C receptors in the brain. Lorcaserin is a selective agonist of the 5-HT2C serotonin receptor in the hypothalamus, which regulates satiety and the metabolic rate [81]. In a phase 2 clinical study, patients who were administered 20 mg/day lorcaserin attained an average weight loss of 3.6 kg, which was significantly greater than the average weight loss of 0.3 kg in the placebo group [82].
Two phase 3 trials BLOOM (Behavioral modification and Lorcaserin for Overweight and Obesity Management) and BLOSSOM (Behavioral modification and LOrcaserin Second Study for Obesity Management) evaluated obese and overweight patients to demonstrate the efficacy of lorcaserin (10 mg twice daily) over placebo [83.84]. In the BLOOM trial, patients treated with lorcaserin achieved highly significant and absolute weight loss in 1 year, and continuous treatment with lorcaserin over 2 years helped two-thirds of the patients maintain a weight loss of 5% or greater [83]. The average amount of weight lost on lorcaserin treatment was 5.8 ± 0.2 kg , compared with 2.2 ± 0.1 kg in the placebo group (P<0.001). Lorcaserin was very well tolerated; it did not result in increased risk of depression or the development of cardiac valvular insufficiency. The BLOSSOM trial confirmed the results reported by the BLOOM trial [84]. In this 52-week trial, 47.2% of patients in the lorcaserin group lost at least 5% of their body weight, compared to 25% of patients in the placebo group [84]. In the lorcaserin group, 22.6% of patients lost at least 10% of their body weight, as compared to 9.7% of patients in the placebo group [84]. Lorcaserin had successfully completed phase 3 trials and its new drug application was reviewed by the FDA<B87></C. The drug failed to get FDA approval because of safety concerns Related to the finding that high doses of the drug were associated with tumor formation in rats [85] (Table 3).
5-HT6 receptors have also been targeted for the treatment of obesity. Central application of antisense oligonucleotide against 5-HT6 receptors decreased food intake in rats [86]. Selective 5-HT6 receptor antagonists have also been shown to reduce body weight in mice with diet-induced obesity [87]. However, recent evidence suggests the distribution of 5-HT6 receptors in the mouse brain is very different from that in rats or humans [88]. As a result, the potential role of the 5-HT6 receptor in regulating human energy homeostasis requires additional evaluation.
Histamine H3 receptor antagonists
Histamine-synthesizing neurons in the tuberomammillary nucleus project to the paraventricular nucleus and the ventromedial hypothalamus, two hypothalamic regions recognized to regulate food intake [89]. In 2001, Masaki et al. reported that central infusion of histamine in a leptin-resistant mouse model of obesity reduced fat accumulation, decreased leptin, and improved insulin sensitivity [90]. These findings initiated the development of H3 receptor antagonists, which increase histamine levels in the hypothalamus and decrease food intake. Novo Nordisk extensively investigated the antiobesity action of the potent and selective non-imidazole H3 receptor antagonist NNC38-1202. However, NNC38-1202 did not progress as an antiobesity pharmaceutical option due to possible drug interactions [91]. Schering plough was evaluating another H3 receptor antagonist, SCH497079, as an antiobesity drug candidate in phase 2 trial. The data of its clinical testing are not available [92].(Table 3) A-331440, a potent and selective non-imidazole H3 receptor antagonist developed by Abbott Laboratories, reduced weight in a dose-dependent manner in an animal model using mice with diet-induced obesity and it prevented weight gain in genetically obese (ob/ob) mice [93]. The weight loss of the mice with diet-induced obesity was associated with reductions in both food consumption and plasma leptin levels.
Hoodia gordonii extract
The Hoodia gordonii cactus plant was traditionally used by the Bushmen of the Kalahari Desert to suppress hunger. The South Africa’s Council for Scientific and Industrial Research (CSIR) investigated the appetite suppressant activity of Hoodia extracts and made a breakthrough with structure elucidation of an active ingredient, p57 [94]. In 1998, CSIR licensed an agreement with Phytopharm (a British pharmaceutical company specializing in Phytomedicines) to further develop p57 [94]. Phytopharm collaborated with Pfizer for development and commercialization of the extract In an early clinical trial of 19 obese subjects, either P57 or placebo was administered for 15 days. A statistically significant reduction was observed in the average daily calorie intake in the P57 group compared with the placebo group, and no serious adverse effects were experienced by any of the subjects [94,95]. However, in 2003, Pfizer ended its collaboration with Phytopharm on the further development of p57 citing difficulties in synthesizing the extract into drug form [94,96] (Table 3). In 2004, Unilever entered into an agreement with Phytopharm to start marketing Hoodia gordonii commercially in the form of a functional food for obesity management [94]. However, in 2008, Unilever severed its agreement with Phytopharm as the extract could not live to their expectations of safety and efficacy [94]. (Table 3) Much work remains to be done before the scientific evidence supports the use of Hoodia plant extract as an appetite suppressant.
Nesfatin
Nesfatin-1/nucleobindin 2 (NuCB2) is a naturally occurring satiety molecule produced in the brains of mammals [97]. Chronic intracerebroventricular injection of nesfatin-1 in rats has been shown to reduce body weight, whereas that of antisense oligonucleotide against the NUCB2-encoding gene increased body weight [98]. These findings suggest nesfatin and its analogues have potential as possible antiobesity drug candidates.
Corticotrophin releasing factor (CRF)/urocortin
The CRF-like neuropeptide (urocortin), acting via CRF2 receptors in the brain, is a potent suppressor of appetite [99]. Administration of urocortin-3 in the ventromedial hypothalamus suppresses feeding and elevates blood glucose levels in rats, suggesting the activation of CRF2 receptor in the brain promotes stress-like responses [100].It was recently shown that transgenic mice overexpressing urocortin-3 were protected against the untoward metabolic consequences of an obesogenic high-fat diet challenge [101].These observations indicate urocortin may be another antiobesity drug target with promise.
Neuromedin U
Neuromedin U (or NmU) is a neuropeptide found in the brains of humans and other mammals. This peptide is involved in diverse physiological functions, including appetite suppression [102]. Intracerebroventricular administration of neuromedin U in rats is associated with marked reductions in body weight [103]. On the other hand, injection of an antibody to neuromedin U increased food ingestion [103]. In mice with diet-induced obesity, intracerebroventricular infusion of NmU for 7 days decreased body weight and total energy intake as compared with the mice given intracerebroventricular infusion of vehicle [104]. The role of neuromedin U as an appetite suppressant acting through NmU receptor 2 (Nmur2) has recently been identified from the studies using Nmur2 -/- mice (carrying 2 copies of the mutant Nmur2 allele) [104,105]. These mice were resistant to central NmU induced weight loss when fed a high fat diet as compared to wild type mice [105].
Fatty acid synthase (FAS) inhibitor
FAS enzyme is involved in the conversion of malonyl CoA to palmitate. Inhibition of FAS by a specific FAS inhibitor, C75, has been shown to reduce food intake and body weight in mice [106]. This anorectic effect is presumably mediated via interference with the expression of orexigenic neuropeptides in the brain. FAS inhibitors also stimulate carnitine palmitoyl transferase 1 (CPT-1), the rate-limiting enzyme for mitochondrial fatty acid oxidation [107].Thus, FAS inhibitors also show potential as antiobesity agents.
Bombesin and bombesin-like peptides
Bombesin (and its family of bombesin-like peptides) modulates various biological and behavioral functions in humans. Intensive psychopharmacological studies in animals and humans have shown it to be an appetite suppressant [108]. Lieverse et al. demostarted that intravenous infusion of bombesin reduced the intake of a carbohydrate- rich meal in human subjects [109-111]. However, the satiety inducing effects of bombesin were observed in lean subjects, and not in obese individuals [111].
BRS-3 (bombesin receptor subtype 3) ligands comprise a new class of potential antiobesity agents that may strongly suppress feeding [112]. In animal studies, the selective antagonist of BRS3 increased food intake and body weight, whereas the selective BRS3 agonists increased metabolic rate and reduced food intake and body weight. Prolonged high levels of receptor occupancy by BRS3 agonists continued to cause weight loss in animal studies, suggesting a lack of tachyphylaxis [113].
Gastrointestinal peptides
In response to ingested nutrients, a number of peptides are released from the gastrointestinal tract. These include cholecystokinin (CKK), glucagon-like peptide 1(GLP-1), peptide YY (PYY), pancreatic polypeptide (PP), oxyntomodulin, amylin, and enterostatin. These peptides act in concert with other postprandial gastrointestinal signals (e.g., gastric distention) to cause satiation and promote meal termination [7-10]. Thus, stimulation of each is being researched to determine their potential as antiobesity targets.
Cholecystokinin receptor agonists
CCK decreases food intake in numerous species including humans. It has been proposed to act as a satiety signal via CCK1 receptor activation [114]. A few non-peptide, selective CCK1 receptor agonists (GI-181771X [GlaxoSmithKline], CE326597 [Pfizer]) had been evaluated in clinical trials for obesity [115]. However, none of these compounds are currently undergoing clinical development. GI181771X, when given to obese patients for 24 weeks, failed to produce any reduction in body weight or waist circumference or to alter cardiometabolic risk markers [116]. (Table 3) A possible explanation for the failure of CCK1 agonist monotherapy might be the development of tachyphylaxis upon chronic stimulation of the receptor [117]. Though CCK monotherapy has not proven successful, some studies have reported an enhanced effect of leptin on weight reduction when it is co-administrated with CCK in rats [118]. Future studies should focus on co-administration of CCK1 receptor agonists along with leptin or other hormones for the treatment of human obesity [9]
Glucagon-like peptide 1
The protease-resistant GLP-1 (glucagon-like peptide 1) congener, exenatide, is already marketed for the treatment of diabetes. In clinical trials, exenatide has been shown to reduce hemoglobin A1C, while also inducing modest but progressive weight loss that persists for at least 2 years [119]. Exenatide given subcutaneously twice daily at doses of 5 or 10 µg elicited mean weight reductions of up to 2.8 kg at 30 weeks and 5.3 kg at 3.5 years [120-122]. Exenatide administered at doses of 0.8 mg or 2 mg once weekly resulted in mean weight reductions of up to 3.8 kg at 15 weeks and 3.7 kg at 30 weeks [123,124]. Liraglutide is a novel GLP1 agonist that was approved by the FDA in January 2010 for the treatment of type 2 diabetes [125]. It has a long duration of action, is suitable for once-daily dosing and is more homologous to native GLP1 than exenatide [126]. Liraglutide has also shown promising results in clinical trials for the treatment of obesity in individuals without diabetes, resulting in weight losses of 4.8--7.2 kg with different doses ranging from 1.2 to 3 mg [127]. This was significantly higher than that seen with placebo (P = 0.003 for 1.2 mg liraglutide and P < 0.0001 for 1.8--3 mg liraglutide) and orlistat (P = 0.003 for 2.4 mg liraglutide and P < 0.0001 for 3 mg liraglutide). More than 50% of the patients on liraglutide therapy achieved a weight reduction in the range of 5--10% [127]. Nausea and vomiting were the most common side effects. These were transient in nature and rarely led to the discontinuation of therapy. Liraglutide has progressed to phase 3 trials for the treatment of obesity [128] (Table 2).
Peptide YY3-36 and pancreatic polypeptide
Peripheral administration of peptide YY 3-36 (PYY3-36) inhibits food ingestion and reduces body weight in rodents, apparently by activating autoinhibitory Y2 receptors on orexigenic NPY/Agrp neurons in the hypothalamus, thereby derepressing adjacent anorexigenic POMC neurons [129]. In obese individuals, PYY3-36 intravenous infusion has been reported to ease hunger, decrease single-meal food intake by 30%, and significantly reduce the cumulative 24-hour caloric intake [130]. This treatment was well tolerated, with no complaints of altered food palatability or nausea. Based on these promising results, an intranasal formulation of PYY3-36 was evaluated in clinical trials [131]. However, this formulation demonstrated disappointing results; the amounts of weight lost at low doses were similar to those lost with placebo. Weight loss at high doses of intranasal PYY could not be assessed because 60% of patients dropped out due to nausea and vomiting. This high incidence of nausea with intranasal administration might be related to PYY’s C-max (peak serum concentration of drug), since concentration spikes with the nasal route of delivery [132]. Novel formulations providing sustained levels of PYY might be well tolerated and are desirable targets for future research.
PP is another gut-derived peptide with similar action to PYY. Obinepitide, a synthetic analogue of both PYY3-36 and PP and an agonist of Y2 and Y4 receptors, is under clinical development [128]. (Table 2) In obese human subjects, once-daily subcutaneous dosing of obinepitide significantly inhibited food intake up to 9 hours after dosing [128].
Oxyntomodulin
Oxyntomodulin is a product of the proglucagon gene from which GLP-1 is cleaved. In humans, intravenous infusions of oxyntomodulin have been shown to acutely lessen hunger and single-meal food intake without reducing food palatability [133]. Repeated subcutaneous injections of oxyntomodulin for 4 weeks reduced body weight by up to 2.3 ± 0.4 kg as compared with 0.5 ± 0.5 kg in the control group (P = 0.0106) [134]. Importantly, oxyntomodulin reduced single-meal intake by 25% at the start of this study and by up to 35% at the end, indicating there was no tachyphylaxis related to its anorectic effects [134]. In another study, subcutaneous injection of oxyntomodulin for 4 days reduced energy intake by 17.3 ± 5.5%, with no change in meal palatability [135]. Oxyntomodulin did not alter resting energy expenditure, but it increased activity-related energy expenditure by 26.2 ± 9.9% and total energy expenditure by 9.4 ± 4.8%. Reductions in body weight of 0.5 ± 0.2% were also observed during the period of oxyntomodulin administration. These modest and favorable results led to the development of the potent and synthetic long-acting oxyntomodulin analogue, OAP189 (TKS1225), which is currently being investigated in a phase 1 clinical trial [136,137] (Table 2).
Amylin
Amylin is a peptide that is postprandially cosecreted with insulin from pancreatic cells. Food intake causes a rapid increase in plasma amylin that is directly proportional to the meal size [138,139]. Amylin functions as a satiety signal by acting on area prostema in the hindbrain [140]. Administration of exogenous amylin prior to a meal dose-dependently decreases meal size in rodents thereby reduces food intake [141,142]. Amylin also suppresses gastric emptying, gastric acid output, and glucagon secretion. Pramlintide, a synthetic amylin analog, is currently marketed for the treatment of diabetes, but it has also been found to induce mild progressive weight loss in humans for at least 16 weeks [143]. In one study, subjects completing treatment with pramlintide experienced placebo-corrected reductions in body weight of 3.7 ± 0.5% [144]. Approximately 31% of the pramlintide-treated subjects achieved a weight loss of 5% or greater, as compared with 2% in the placebo group [144]. To date, much of the research with amylin has focused on its potential to increase leptin sensitivity. Infusions of an ineffective dose of leptin (500 µg/kg/day had no significant effect on the body weight of rodents with diet-induced obesity) in combination with a moderately effective dose of amylin (100 g/kg/day achieved weight losses of up to 6.5%) have been shown to elicit marked, synergistic weight loss (up to 12%) in rats with diet-induced obesity [47]. These findings provide a rationale for using pramlintide in combination with leptin for the treatment of obesity. This combination is currently being evaluated in clinical trials and is discussed again later in this article.
Enterostatin
Enterostatins are pentapeptides generated in the small intestine as a result of N-terminal proteolytic processing of pancreatic procolipase. Enterostatin is absorbed from the gut and acts as a powerful anorectic peptide, particularly in regards to reducing the intake of fatty foods [145]. In a study performed in rats, chronic intracerebroventricular injections of enterostatin decreased intake of a high-fat diet and reduced body weight [146]. Previous experiments explored the hypothesis that an association exists between human obesity and enterostatin dysregulation [147,148]. However, in clinical trials, intravenous enterostatin failed to show any effect on feelings of hunger, satiety, or food preference [149]. Present knowledge suggests the molecular identification of enterostatin receptors is required in order for enterostatin to be developed further as antiobesity drug target.
Appetite Suppressants: Inhibitors of Orexigenic Pathway
Neuropeptide Y receptor antagonists
Neuropeptide Y (NPY) is a 36 amino-acid peptide that was found to potently stimulate food intake in a variety of species [10]. This led to a surge of interest in developing NPY receptor antagonists as antiobesity drugs. The orexigenic effects of NPY are chiefly mediated by the Y1 and Y5 receptors. Thus, antagonists for these receptors were viewed as potential antiobesity drugs. Neuropeptide Y1 antagonists demonstrated promising weight-reducing results in animal studies, whereas neuropeptide Y5 antagonists showed equivocal effects [150-154]. Few of these antagonists progressed to clinical trials, and since they did not live up to expectations, none are being currently pursued. NGD 95/1, a NPY1 antagonist, was evaluated in a phase 1b clinical trial conducted by Neurogen and Pfizer, but it was not developed further because of elevated liver enzymes [10]. MK-0557, an orally active NPY5 receptor antagonist, was evaluated in a year-long clinical trial, but since it did not produce clinically meaningful weight loss, further development was abandoned [155] (Table 3).
In addition to the disappointing clinical experience with NPY antagonists, the critical role of NPY in energy regulation was also disputed by the finding that NPY deficiency did not result in leanness in mice [15,156]. However, NPY deficiency in ob/ob (leptin deficient) mice was found to attenuate the obesity phenotype by reducing food intake and increasing energy expenditure [157]. This observation implies that NPY is required for a full response of leptin deficiency, even though it might not be required to maintain body weight. Thus, its blockade might be useful for preventing the regain of weight lost by other means [15]. More recently, interest has focused on the stimulation of presynaptic NPY2 receptors in the arcuate nucleus as a mechanism to reduce NPY release and, thus, food intake [131]. As discussed above, peptide YY (PYY), a gut hormone peptide with selectivity for NPY2 receptors, has been explored in clinical trials.
Melanin concentrating hormone 1 receptor antagonist
Melanin-concentrating hormone (MCH) is a 19-amino acid, cyclic neuropeptide that is present in the hypothalamus. MCH administration or its transgenic overexpression in mice increases body weight by stimulating food ingestion and adipogenesis, while decreasing energy expenditure [158,159]. This suggests that MCH1 receptor antagonists might promote weight loss, but there are several issues of concern with the development of these compounds. MCH1 receptor knockout mice demonstrate weight reduction, hyperactivity, and increased energy expenditure, but they are also hyperphagic [160]. This situation of chronically elevated energy intake due to marked hyperactivity and energy expenditure might be hazardous, as ample evidence suggests that long-term caloric restriction increases lifespan, and chronic increased calorie intake in the face of increased energy expenditure can theoretically shorten life span [161-163]. MCH antagonism also poses a risk of cardiac arrhythmias, and MCH is known to modulate many functions beyond feeding, such as locomotor activity, anxiety, aggression, sensory processing, learning, and sleep cycle [159,164,165]. Thus, it might be challenging to design anti-MCH agents that selectively regulate energy homeostasis without exerting adverse side effects. Early clinical testing of the MCH1 receptor antagonist NGD-4715 failed after recipients complained of vivid dreams and awakenings [166].
Orexin antagonists
Orexin A and orexin B (or hypocretin-1 and -2) are a pair of excitatory neuropeptides present in the brain. These peptides have diverse physiological functions like the regulation of feeding, the sleep-wake cycle, and the reward pathway [167,168]. It has been reported that intracerebroventricular injection of orexin increases food intake in rats [10,167,168]. The antiobesity activity of a prototype selective orexin-1 receptor antagonist, SB-334867-A, has been explored in preclinical studies [169]. This drug reduced cumulative food intake and body weight in genetically obese mice when administered over a period of 14 days [169]. Stimulation of thermogenesis also occurred, as evidenced by an increase in uncoupling protein-1 mRNA expression in intrascapular brown adipose tissue. A single dose of SB-334867-A was found to increase metabolic rate over a period of 4 h in ob/ob mice [170].
Galanin antagonists
Galanin, a 29-30 amino acid neuropeptide, is widely distributed in both the central and peripheral nervous systems. Pharmacological studies indicate galanin plays a role in regulating feeding and nutritional balance [10,171].Injection of galanin into the central nucleus of the amygdala and the paraventricular nucleus of the hypothalamus in rats significantly increases food intake [172]. Central injection of galanin stimulates the intake of fatty foods in particular [173].Similarly, galanin gene knock-out mice also demonstrate significantly lower intake of fat in comparison with wild-type mice [174]. However, repeated galanin infusions in the third ventricle in rats failed to induce obesity or hyperphagia, suggesting tachyphylaxis may have developed [175]. Thus, further evaluation of the role of galanin in modulating feeding behavior is required. To date, three galanin receptors have been cloned and attempts are underway to synthesize specific galanin antagonists for the treatment of obesity [10,176].
Ghrelin antagonist
Ghrelin is an orexigenic hormone secreted from the stomach and upper intestine. Its plasma levels rise before the start of a meal (coincident with meal times of a fixed feeding schedule) and fall again rapidly after the meal is initiated [177]. This pattern of secretion implies that ghrelin plays a role in mealtime hunger and meal initiation and has led to the development of ghrelin antagonists for the treatment of obesity. However, the role of ghrelin antagonism in this aspect has been challenged by studies performed with ghrelin-deficient mice. Mice with congenital deletions of the gene encoding ghrelin or its receptor exhibited minimal body weight reductions on standard chow but they were resistant to highfat-diet-induced obesity [178,179]. In spite of these observations, a number of other studies demonstrated that ghrelin antagonism in adult animals reduces body weight [180,181]. Ghrelin receptor antagonists have been found to decrease energy intake in lean mice, obese (ob/ob) mice, and mice with diet-induced obesity [182]. They also reduce the rate of gastric emptying. Repeated administration of ghrelin receptor antagonist has been shown to decrease body weight gain and improve glycemic control in ob/ob obese mice [183]. The possible efficacy of ghrelin inhibitors as weight-loss agents is consequently being reevaluated. Evidence is also emerging that suggests there are chronic effects of ghrelin on fat accumulation by modulation of hypothalamic genes coding for the peptides and receptors involved in energy balance regulation, independent of food intake [184].
Lipase Inhibitors
Lipase inhibitors inhibit gastric and pancreatic lipases in the lumen of the alimentary tract to decrease systemic absorption of dietary fat, without affecting food intake. The lipase inhibitor presently in clinical development for the treatment of obesity is cetilistat [185] (Table 2) The clinical data currently available suggest that cetilistat has comparable efficacy to orlistat (Phase 2b clinical trial showed 3 kg weight loss over placebo in 3 months). Also, as compared to orlistat, this drug has a favorable side-effect profile (90% fewer severe gastrointestinal side effects) [186,187].
DRUGS THAT INCREASE ENERGY EXPENDITURE AND INCREASE METABOLIC ACTIVITY, THERMOGENESIS, AND LIPOLYSIS PERIPHERALLY
Thyroid Hormone Receptor Agonist
Thyroid hormones are known to stimulate weight loss by increasing the metabolic rate. However, their use has been associated with cardiac stimulation and protein loss. Recently, several subtypes of the thyroid receptors have been discovered. The alpha subtype is located in the heart and mediates the cardiostimulatory effect of the thyroid hormones. The beta subtype is chiefly located in the liver and is involved in hepatic cholesterol metabolism. A beta subtype-specific agonist, KB-141, has been developed [188]. In one study, KB-141 was administered orally to obese zucker fa/fa rats for 21 days and ob/ob mice for 7 days, after which body weight, adiposity, and lipid levels were examined [189]. In fa/fa rats, KB-141 reduced body weight by 6--8% and adiposity by 5--6%, without causing tachycardia. In ob/ob mice, KB-141 lowered serum cholesterol, triacylglycerols, and both serum and hepatic levels of free fatty acids, without causing tachycardia. Treatment of ob/ob mice with KB-141 for 2 weeks improved glucose tolerance and insulin sensitivity in a dose-dependent manner with no effect on heart rate [189]. Another thyroid beta subtype receptor agonist, GC1, when administered to cynomolgus monkeys for 7 days, caused significant reductions in cholesterol and lipoprotein (a), and an average weight loss of 4%, without causing cardiac stimulation [190].
Human Growth Hormone Analogue
Growth hormone has profound lipolytic and antilipogenic activity and is known to reduce body fat and cause weight loss. AOD-9604 is an orally active analogue of peptide fragment of human growth hormone (hGH 177--191) that selectively activates lipolysis in adipose tissue. In one study, AOD-9604 reduced body weight in genetically obese zucker rats and ob/ob mice, without inducing the untoward effect on glycemic control normally observed with growth hormone [191]. In a 12-week randomized clinical trial, subjects receiving AOD-9604 (1 mg/d) lost an average of 2.6 kg, compared to 0.8 kg in the placebo group [192]. Development of this drug was terminated in 2007 as the drug failed to induce significant weight loss in a 24-week trial of 536 subjects [192] (Table 3).
β3-adrenoceptor Agonists
β3-adrenoceptor agonists were first described as an antiobesity target in 1983, and their seven-transmembrane G-protein-coupled β3-adrenoceptor was cloned 6 years later [193,194]. To date, however, β3-adrenoceptor agonists have not been marketed for the treatment of obesity. The first generation β3-agonists were selected on the basis of their thermogenic and antiobesity actions observed in rats and mice. In obese rodents, they produced good weight loss results by increasing metabolic rate. Unfortunately, clinical experience with these compounds has been less encouraging. The side effects of first-generation compounds in humans were tachycardia and tremors, indicating lack of β3 receptor selectivity. They were also not effective at reducing weight by increasing thermogenesis, since their agonistic potential for human β3 receptors was not as strong as for rodent β3-adrenoceptors [10,195,196]. In rodents, the β3-receptors are present in both white adipose tissue and brown adipose tissue (BAT), but in humans, β3 receptors are predominantly present in BAT. Newborn infants have relatively large amounts of BAT, which plays a thermoregulatory role in affording protection from cold exposure. However, with increasing age, the amount of BAT decreases. Thus, it has been argued that the expression of β3 receptors might be insufficient in human adults for β3-adrenoceptor agonists to elicit a response [10,195]. Treatment of obese patients with the first-generation β3 agonists BRL26830 and BRL35135 had no effect on thermogenesis or fuel metabolism. These drugs also caused undesirable tremors and tachycardia [195, 197, 198].
The presence of BAT and its physiological role in adults has been a subject of great debate. Recently, the presence of BAT in adult humans was demonstrated with radiological evidence [199]. In addition, body mass index and the amount of BAT have been shown to be inversely proportional, especially in older patients; this suggests BAT may play a protective role against obesity in adults [199]. A few studies have also shown that chronic stimulation with selective agonists causes upregulation of β3-adrenoceptors and induces BAT hypertrophy in rodents, dogs, and monkeys, a finding that suggests recruitment of BAT could also occur in humans. These recent developments have led to a resurgence of research into drugs targeting β3 receptors. Novel generation of highly selective β3 receptor agonists have been tried in clinical trials [10, 19, 200, 201]. CL 316243 increased insulin-mediated glucose disposal and fat oxidation in human volunteers, without causing tremors and cardiac stimulation [200]. However, the poor oral bioavailability of CL316243 is a major drawback. Prodrugs with improved bioavailability have also been developed [201]. The oral bioavailability of a single dose of LY377604 was greater than 20%, and in obese sbjects it was shown to increase metabolic rate by 17.5% [10]. Another β3 agonist N5984, developed by Nisshin Kyorin Pharmaceuticals was in early phase of clinical testing. However, the clinical study data for N5984 is not available [202] (Table 3).
Uncoupling Proteins
Uncoupling protein 1 (UCP1) or thermogenin, a member of the mitochondrial transporter superfamily, is involved in energy expenditure and brown fat thermogenesis. UCP1 protein dissociates the mitochondrial fatty acid oxidation from production of ATP and the energy is dissipated as heat (Fig. 5) [10, 203]. This is a key process for non- shivering thermogenesis and body weight regulation in mammals [203]. Although UCP1 is very well characterized, it has not been pursued as a target for increasing energy expenditure because the protein is uniquely present in BAT, which is barely detected in adult humans. In the past few years, a few proteins with high sequence homology to UCP1 have been discovered that are also expressed in non-BAT. These include UCP2, which is widely distributed in the body, and UCP3, which is expressed mainly in skeletal muscle [204]. Owing to the ubiquitous localization of UCP2, the likelihood of unwanted side effects makes this protein a less appealing target for obesity treatment. In contrast, stimulation of the skeletal-muscle-specific UCP3 activity could provide a more dependable mechanism by which whole-body thermogenesis in humans could be increased. Mutations in the gene encoding UCP3 have been discovered in some individuals suffering from severe obesity and non-insulin-dependent diabetes mellitus [205]. Transgenic mice with muscle-directed overexpression of UCP3 were hyperphagic and lean when fed a palatable diet, and their glucose tolerance was improved [206]. Thus, the pharmacological stimulation of UCP3 activity could result in beneficial effects against obesity and type 2 diabetes mellitus [206].
Diacylglycerol Acyltransferase Inhibitors
Diacylglycerol acyltransferase (DGAT) enzyme catalyzes the final reaction of triacylgycerol synthesis. Two isozymes of DGAT, DGAT1 and DGAT2, have been described. DGAT1 plays an important role in the synthesis of very-low-density lipoprotein (VLDL). Since increased plasma VLDL concentrations might promote obesity, the inhibition of DGAT1enzyme is considered a possible therapeutic target for obesity treatment [207]. DGAT gene knockout mice are resistant to diet-induced obesity and appear to compensate for the reduced ability to store fat by increasing energy expenditure [208]. A potent and highly selective DGAT1 inhibitor, compound 4a, reportedly reduced weight and liver triglycerides when dosed chronically in diet-induced obese mice [209]. It also depleted serum triglycerides in a dose-dependent manner following a lipid challenge in a murine model of obesity, thus reproducing major phenotypical characteristics of DGAT1 knockout mice [209].
Stearoyl-CoA Desaturase1 Inhibitors
Stearoyl-CoA desaturase (SCD) is the rate-limiting enzyme involved in monounsaturated fatty acid synthesis. It has recently been reported to be a vital control point regulating hepatic lipogenesis and lipid oxidation. SCD1-deficient mice exhibit increased energy expenditure, reduced body adiposity, and increased insulin sensitivity. These mice are resistant to diet-induced obesity [210]. Much evidence suggests that the direct antiobesity effect of SCD1 deficiency results from increased fatty acid oxidation and decreased lipid synthesis. All of these findings suggest that the pharmacological manipulation of SCD activity might be of benefit in the treatment of obesity, diabetes, and metabolic syndrome [211].
Acetyl-CoA Carboxylase Inhibitors
The acetyl-coenzyme A carboxylase (ACC) enzyme plays an important role in fatty acid metabolism in most living organisms. ACC is a biotin-dependent enzyme that catalyzes the carboxylation of acetyl-CoA to produce malonyl-CoA. Mice lacking ACC2 have continuous fatty acid oxidation and reduced body fat and body weight. This validates efforts to develop an anti-obesity drug based on antagonism of this enzyme [212].
Adipocyte-complement-related Protein or Adiponectin
Adiponectin (adipocyte-complement-related protein of 30 kDa) is a hormone produced by fat cells that is associated with fatty acid oxidation, increased insulin sensitivity, and antiatherogenic properties [213]. Adiponectin levels are inversely correlated with obesity and obesity-associated complications such as type 2 diabetes mellitus, cardiovascular disease, and hepatic dysfunction [213-216]. The association of low adiponectin levels with obesity underscores its important role in obesity-related comorbidities. It has been recognized as a key molecule in obesity as well as in the metabolic syndrome, and is a potentially promising target for the prevention and treatment of the metabolic syndrome [213, 217,218]. Famoxin is a proteolytic cleavage product of this protein. Administration of famoxin to mice has been shown to increase fatty-acid oxidation and induce weight-loss without affecting food ingestion [219]. The Genset Corporation was preparing famoxin for phase 1 clinical studies; however, further clinical development of this compound is not known when Genset was acquired by Serono Company in 2003 [220] (Table 3). A few drugs have been reported to stimulate the secretion or to induce the expression of adiponectin, including pioglitazone, endocannabinoid (CB1) receptor antagonists, and inhibitors of the renin-angiotensin pathway [221-223].
11 Beta Hydroxysteroid Dehydrogenase Type 1 Inhibitors
There is strong evidence from rodent obesity models indicating that the glucocorticoid action underlies the pathology of metabolic syndrome. In these models, it was observed that removing glucocorticoids reverses obesity and its metabolic abnormalities [224,225]. 11 beta-hydroxysteroid dehydrogenase type 1 (11β-HSD1) is an enzyme that regenerates the active glucocorticoid cortisol from its inactive metabolite cortisone. Animals with 11β-HSD1 knockout have normal plasma glucocorticoid levels, but they cannot regenerate glucocorticoid inside liver and adipose tissue. As a result, they are protected from insulin resistance, hyperglycemia, and weight gain induced by high-fat feeding [225]. Conversely, mice with selective overexpression of 11β-HSD1 in adipose tissue have increased intra-adipose glucocorticoid concentrations, despite no change in plasma levels. These animals display a dramatic phenotype of central obesity, insulin resistance, and hyperglycemia [225]. In idiopathic obesity in humans, 11β-HSD1 activity in adipose tissue is selectively increased to a similar degree as that seen in transgenic 11β-HSD1 overexpressing mice [226]. Pharmacological inhibition of 11β-HSD1 to lower intracellular cortisol concentrations in liver and adipose tissue, without altering circulating cortisol concentrations or responses to stress, is an exciting potential therapy for obesity and diabetes. A few 11β-HSD1 inhibitors have been tested in preclinical studies. BVT.2733, an11β-HSD1inhibitor, has been reported to reduce food intake and weight as well as improve glucose tolerance in a mouse model of obesity and diabetes [227].
MODULATION OF FAT STORAGE OR ALTERATION OF ADIPOCYTE DIFFERENTIATION
Peroxisome proliferator-activated receptor gamma (PPAR-γ) ligands have been reported to stimulate UCP1 expression in BAT but not in white adipocytes. This finding led to the identification of BAT-specific transcription cofactor, PPAR-γ coactivator-1 (PGC-1)[8, 228, 229]. PGC-1 is a family of transcription coactivators that functionally interact with nuclear receptor PPAR-γ [230]. It plays a cardinal role in the transcriptional programme of adaptive thermogenesis occurring in BAT as mitochondrial biogenesis, respiration, and thermogenesis [230]. Ectopic expression of PGC-1 in white adipocytes converts them to a more BAT-like phenotype [8]. The treatment of obese mice with rosiglitazone has been shown to boost PGC-1 transcription in white adipose tissue, concomitant with increased mitochondrial function and insulin sensitivity [231]. Increasing PGC-1 levels in skeletal muscle and white adipose tissue could, therefore, be potentially beneficial for the treatment of obesity and type 2 diabetes.
MIMICKING CALORIC RESTRICTION
Caloric restriction triggers an adaptive drop in metabolic rate in rodents, nonhuman primates, and humans. This adaptation plays a central role in the rebound of body weight after weight loss observed in most patients<B19></C>; it also evokes beneficial effects in lowering the incidence of metabolic disorders related with aging, overweight or obesity, insulin resistance, and cardiovascular disease [19]. Caloric restriction has been demonstrated to increase lifespan in organisms, ranging from yeast to rodents, as well as in nonhuman primates [161-163,232,233]. Recently, a class of proteins known as SIRTS or sirtuins (also known as silent information regulator 2 [Sir2]-related enzymes) has been implicated in an effect of CR on lifespan extension and the prevention of comorbidities associated with obesity [233, 234]. Sirtuins are the family of enzymes with nicotinamide adenine dinucleotide-(NAD) dependent histone deacetylase or ADP-ribosyl transferase activity [235, 236]. The mammalian sirtuins (Sirt1--Sirt7) participate in a number of cellular and physiological functions such as gene silencing, apoptosis, mitochondrial function, energy homeostasis, and longevity [237].Transgenic mice with overexpression of the sirt1 gene demonstrate decreased lipid-induced inflammation, improved glucose tolerance, and protection from hepatic steatosis when fed a high-fat diet [238].
COMBINATION THERAPIES
Various fixed-dose combination antiobesity drugs are in different phases of clinical development. These include the following:
Bupropion and Naltrexone (Contrave)
Contrave is a fixed dose combination of bupropion and naltrexone in a single tablet. Bupropion, a nonselective dopamine- and norepinephrine-reuptake inhibitor, is considered to reduce weight by stimulating hypothalamic POMC neurons. The stimulation of hypothalamic POMC neurons is inhibited by β-endorphin as an opioid reward process is involved in the short-term control of eating [239]. In combination with bupropion, naltrexone, a µ-opioid receptor antagonist, acts in a synergistic manner to stimulate weight loss by blocking the β-endorphin-mediated inhibition of the POMC neuron [192]. In phase 3 trials, after 56 weeks of treatment, 62% of patients receiving contrave 32 (bupropion SR 360 mg/naltrexone SR 32 mg) lost at least 5% of their body weight as compared to 23% of patients in the placebo group [240]. Approximately 34% patients lost 10% or more of their body weight and 17% patients lost at least 15% of their mean body weight in the contrave group. Patients receiving contrave32 had a mean weight loss of 8 kg, as compared to 1.8 kg in the placebo group, after 56 weeks of treatment [240]. Obese patients on contrave also demonstrated significant improvements in important markers of cardiometabolic risk, including waist circumference, high-density lipoprotein cholesterol, insulin resistance and triglycerides. Contrave had completed phase 3 trials successfully [240-242]. Its NDA has recently been reviewed by the FDA. The FDA panel has deferred the approval of the drug, pending the conduction of a long-term study demonstrating cardiovascular safety [243] (Table 3).
Bupropion and Zonisamide (Empatic)
Empatic is a fixed-dose combination drug containing bupropion and zonisamide. Zonisamide is an antiepileptic drug that has serotonergic and dopaminergic activity in addition to its ability to act as a blockade of sodium and calcium channels. Weight loss was an adverse effect associated with zonisamide treatment in clinical trials for epilepsy. Although the exact mechanism for this weight-reducing effect is unknown, the proposed mechanism includes alterated perception of taste due to carbonic anhydrase activity and modulation of dopamine and serotonin levels in the brain [241, 244, 245].Empatic has shown promising results in phase 2 clinical trials [241] (Table 2). Phase 2 trials demonstrated that patients completing 24 weeks of empatic360 (bupropion SR 360 mg/zonisamide SR 360 mg) and empatic-120 (bupropion 360 mg/zonisamide 120 mg) therapy attained a significant weight loss as compared to placebo (9.9%, 7.7% vs. 1.7%, respectively (p<0.001)). In the empatic-360 group, 82.6% of patients lost at least 5% of their baseline body weight and 47.7% lost at least 10% of their baseline body weight as compared to 18.9% and 5.7% patients in the placebo group, respectively [192].
Phenteramine and Topiramate (Qnexa)
Qnexa is a combination drug that consists of low doses of phentermine and topiramate. Phentermine is a popular appetite suppressant used for weight loss. Topiramate is approved as an anti-epileptic and antimigraine drug having multifactorial effects on the central nervous system. It is thought to act as a γ-aminobutyric acid agonist that increases satiety, although the precise mechanism of action is unclear. Qnexa has shown promising clinical data in a phase 3 trial [246]. An average weight loss of -8.1 kg and -10.2 kg was attained at the end of 56 weeks with phentermine 7·5 mg plus topiramate 46·0 mg, and phentermine 15·0 mg plus topiramate 92·0 mg, respectively. Also, there were significant improvements in cardiovascular and metabolic risk factors among patients treated with Qnexa [246]. An FDA panel recently voted against the NDA of Qnexa because of safety concerns (Table 3). The potential benefits of the drug were overridden by its adverse effect profile, which includes its teratogenic potential, ability to increase heart rate, and propensity to cause psychiatric problems [247].
Pramlintide and Metreleptin
This combination targets the long-term adiposity signals (e.g., leptin) and short-term satiety signals (e.g., amylin). This is a novel integrated neurohormonal strategy recently employed for obesity drug development. Various preclinical and clinical studies have demonstrated a synergistic effect of amylin and leptin combination in reducing food intake and body weight [47]. Amylin has also been implicated in enhancing leptin signaling [47]. The combination of the amylin analogue pramlintide and the recombinant human leptin metreleptin caused significant weight reductions (-11.5±kg) in comparison with monotherapy in a 24-week phase 2 clinical trial [248]. The pramlintide/ metreleptin combination was well tolerated. The most common side effects were mild-to-moderate adverse events at the injection site and nausea. This combination therapy had completed phase 2 trials successfully. However, the further clinical development of pramlintide/metreleptin combination therapy has been discontinued by the manufacturing companies because of safety concerns [249](Table 3).
CONCLUSION
While the development of antiobesity drugs continues to look challenging, researchers retain the hope that effective and safe medications might be on the horizon. A number of drug candidates and targets are currently being explored. Liraglutide, cetilistat, and tesofensine are already in late phases of clinical development. Thus, notwithstanding the existing skepticism about the future development of these drugs, substantial optimism persists that new antiobesity drugs will be available in the foreseeable future.
ACKNOWLEDGEMENTS
Declared none.
CONFLICT OF INTEREST
The author confirms that this article content has no conflicts of interest.
REFERENCES
- 1.World Health Organization. Unhealthy diets and physical inactivity. The problem. Available from:http://www.who.int/nmh/publications/ fact sheet_diet_en.pdf . [Accessed on September 9, 2011].
- 2.Allison DB, Fontaine KR, Manson JE, Stevens J, VanItallie TB. Annual deaths attributable to obesity in the United States. JAMA. 1999;82(16 ):1530–8. doi: 10.1001/jama.282.16.1530. [DOI] [PubMed] [Google Scholar]
- 3.National Institutes of Health, National Heart, Lung, and Blood Institute. Clinical guidelines on the identification, evaluation, and treatment of overweight and obesity in adults: the evidence report. Obes Res. 1998;6(suppl 2):S5–S210. [PubMed] [Google Scholar]
- 4.Flegal KM, Carroll MD, Ogden CL, Curtin LR. Prevalence and trends in obesity among US adults, 1999-2008. JAMA. 2010;303(3 ):235–41. doi: 10.1001/jama.2009.2014. [DOI] [PubMed] [Google Scholar]
- 5.Morton GJ, Cummings DE, Baskin DG, Barsh GS, Schwartz MW. Central nervous system control of food intake and body weight. Nature. 2006;443(7109 ):289–95. doi: 10.1038/nature05026. [DOI] [PubMed] [Google Scholar]
- 6.Flier JS. Obesity wars molecular progress confronts an expanding epidemic. Cell. 2004;116:337–350. doi: 10.1016/s0092-8674(03)01081-x. [DOI] [PubMed] [Google Scholar]
- 7.Cumming DE, Overduin J. Gastrointestinal regulation of food intake. J Clin Invest. 2007;17(1 ): 13–23. doi: 10.1172/JCI30227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Chiesi M, Huppertz C, Hofbauer KG. Pharmacotherapy of obesity: targets and perspectives. Trends Pharmacol Sci. 2001;2(5 ):247–54. doi: 10.1016/s0165-6147(00)01664-3. [DOI] [PubMed] [Google Scholar]
- 9.Field BC, Chaudhri OB, Bloom SR. Obesity treatment novel peripheral targets. Br J Clin Pharmacol . 009 ;8(6 ):830–43. doi: 10.1111/j.1365-2125.2009.03522.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Clapham JC, Arch JR, Tadayyon M. Anti-obesity drugs a critical review of current therapies and future opportunities. Pharmacol Ther. 2001;89(1 ):81–121. doi: 10.1016/s0163-7258(00)00105-4. [DOI] [PubMed] [Google Scholar]
- 11.Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372(6505 ):425–32. doi: 10.1038/372425a0. [DOI] [PubMed] [Google Scholar]
- 12.Dallongeville J, Fruchart JC, Auwerx J. Leptin, a pleiotropic hormone physiology, pharmacology, and strategies for discovery of leptin modulators. J Med Chem . 998 ;41(27 ):5337–52. doi: 10.1021/jm9802867. [DOI] [PubMed] [Google Scholar]
- 13.Frühbeck G. Intracellular signalling pathways activated by leptin. Biochem J . 006 ;393(Pt 1 ):7–20. doi: 10.1042/BJ20051578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Heymsfield SB, Greenberg AS, Fujioka K, et al. Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, dose-escalation trial. JAMA. 1999;282(16 ):1568–75. doi: 10.1001/jama.282.16.1568. [DOI] [PubMed] [Google Scholar]
- 15.Foster-Schubert KE, Cummings DE. Emerging therapeutic strategies for obesity. Endocr Rev. 2006;27(7 ):779–93. doi: 10.1210/er.2006-0041. [DOI] [PubMed] [Google Scholar]
- 16.Veniant MM, LeBel CP. Leptin from animals to humans. Curr Pharm Des. 2003;9(10 ):811–8. doi: 10.2174/1381612033455369. [DOI] [PubMed] [Google Scholar]
- 17.Morris DL, Rui L. Recent advances in understanding leptin signaling and leptin resistance. Am J Physio Endocrinol Metab. 2009;297(6 ):E1247–59. doi: 10.1152/ajpendo.00274.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.El-Haschimi K, Pierroz DD, Hileman SM, Bjørbaek C, Flier JS. Two defects contribute to hypothalamic leptin resistance in mice with diet-induced obesity. J Clin Invest. 2000;105(12 ):1827–32. doi: 10.1172/JCI9842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hofbauer KG, Nicholson JR, Boss O. The obesity epidemic current and future pharmacological treatments. Annu Rev Pharmacol Toxicol. 2007;47:565–92. doi: 10.1146/annurev.pharmtox.47.120505.105256. [DOI] [PubMed] [Google Scholar]
- 20.Weigle DS. Pharmacological therapy of obesity past, present, and future. Clin Endocrinol Metab. 2003;88(6 ): 2462–9. doi: 10.1210/jc.2003-030151. [DOI] [PubMed] [Google Scholar]
- 21.Ioannides-Demos LL, Piccenna L, McNeil JJ. Pharmacotherapies for obesity: past, current, and future therapies. J Obes. 2011:1–18. doi: 10.1155/2011/179674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Bhasin S, Wallace W, Lawrence JB, Lesch M. Sudden death associated with thyroid hormone abuse. Am J Med. 1981;71(5 ): 887–90. doi: 10.1016/0002-9343(81)90392-2. [DOI] [PubMed] [Google Scholar]
- 23.Gurtner HP. Aminorex and pulmonary hypertension. A review. Cor Vasa. 1985;27(2-3 ): 160–71. [PubMed] [Google Scholar]
- 24.Holden C. Amphetamines: tighter controls on the horizon. Science. 1976;194(4269 ): 1027–8. doi: 10.1126/science.194.4269.1027. [DOI] [PubMed] [Google Scholar]
- 25.Pi-Sunyer FX. Heart valve lesions in patients treated with appetite suppressants. Obes Res. 1999;7(4 ):414–6. doi: 10.1002/j.1550-8528.1999.tb00426.x. [DOI] [PubMed] [Google Scholar]
- 26.Kernan WN, Viscoli CM, Brass LM. Phenylpropanolamine and the risk of hemorrhagic stroke. N Engl J Med. 2000;343(25 ): 1826–32. doi: 10.1056/NEJM200012213432501. [DOI] [PubMed] [Google Scholar]
- 27.Li MF, Cheung BM. Rise and fall of anti-obesity drugs. World J Diabetes. 2011;2(2 ): 19–23. doi: 10.4239/wjd.v2.i2.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.McNeely W, Goa KL. Sibutramine. A review of its contribution to the management of obesity. Drugs. 1998;56(6 ): 1093–124. doi: 10.2165/00003495-199856060-00019. [DOI] [PubMed] [Google Scholar]
- 29.James WP, Caterson ID, Coutinho W. Effect of sibutramine on cardiovascular outcomes in overweight and obese subjects. N Engl J Med. 2010;363(10 ):905–17. doi: 10.1056/NEJMoa1003114. [DOI] [PubMed] [Google Scholar]
- 30.Yanovski SZ, Yanovski JA. Obesity. N Engl J Med. 2002;46(8 ):591–602. doi: 10.1056/NEJMra012586. [DOI] [PubMed] [Google Scholar]
- 31.Padwal RS, Majumdar SR. Drug treatments for obesity orlistat, sibutramine, and rimonabant. Lancet. 2007;369(9555 ):71–7. doi: 10.1016/S0140-6736(07)60033-6. [DOI] [PubMed] [Google Scholar]
- 32.Guerciolini R. Mode of action of orlistat. Int J Obes Relat Metab Disord. 1997;21(Suppl 3 ):S12–23. [PubMed] [Google Scholar]
- 33.Li M, Cheung BM. Pharmacotherapy for obesity. Br J Clin Pharmacol. 2009 ;68(6 ):804–10. doi: 10.1111/j.1365-2125.2009.03453.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Li Z, Maglione M, Tu W. Meta-analysis pharmacologic treatment of obesity. Ann Intern Med. 2005;42(7 ):532–46. doi: 10.7326/0003-4819-142-7-200504050-00012. [DOI] [PubMed] [Google Scholar]
- 35.Torgerson JS, Hauptman J, Boldrin MN, Sjöström L. XENical in the prevention of diabetes in obese subjects (XENDOS) study a randomized study of orlistat as an adjunct to lifestyle changes for the prevention of type 2 diabetes in obese patients. Diabetes Care. 2004;27(1 ): 155–61. doi: 10.2337/diacare.27.1.155. [DOI] [PubMed] [Google Scholar]
- 36.Schneider R, Golzman B, Turkot S, Kogan J, Oren S. Effect of weight loss on blood pressure, arterial compliance, and insulin resistance in normotensive obese subjects. Am J Med Sci. 2005;330(4 ):157–60. doi: 10.1097/00000441-200510000-00001. [DOI] [PubMed] [Google Scholar]
- 37.Beck-da-Silva L, Higginson L, Fraser M, Williams K, Haddad H. Effect of Orlistat in obese patients with heart failure a pilot study. Congest Heart Fail. 2005;11(3 ): 118–23. doi: 10.1111/j.1527-5299.2005.03827.x. [DOI] [PubMed] [Google Scholar]
- 38.Snow V, Barry P, Fitterman N, Qaseem A, Weiss K. Efficacy Assessment Subcommittee of the American College of Physicians. Pharmacologic and surgical management of obesity in primary care: a clinical practice guideline from the American College of Physicians. Ann Intern Med. 2005;142(7 ): 525–31. doi: 10.7326/0003-4819-142-7-200504050-00011. [DOI] [PubMed] [Google Scholar]
- 39.Boozer CN, Leibel RL, Love RJ, Cha MC, Aronne LJ. Synergy of sibutramine and low-dose leptin in treatment of diet-induced obesity in rats. Metabolism. 2001;50(8 ): 889–93. doi: 10.1053/meta.2001.24917. [DOI] [PubMed] [Google Scholar]
- 40.Rosenbaum M, Goldsmith R, Bloomfield D. Low-dose leptin reverses skeletal muscle, autonomic, and neuroendocrine adaptations to maintenance of reduced weight. J Clin Invest. 2005;115(12 ): 3579–86. doi: 10.1172/JCI25977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.MacLean PS, Higgins JA, Johnson GC. Enhanced metabolic efficiency contributes to weight regain after weight loss in obesity-prone rats. Am J Physiol Regul Integr Comp Physiol. 2004;287(6 ):R1306–15. doi: 10.1152/ajpregu.00463.2004. [DOI] [PubMed] [Google Scholar]
- 42.Breslow MJ, Min-Lee K, Brown DR, Chacko VP, Palmer D, Berkowitz DE. Effect of leptin deficiency on metabolic rate in ob/ob mice. Am J Physiol. 1999;276(3 Pt 1 ):E443–9. doi: 10.1152/ajpendo.1999.276.3.E443. [DOI] [PubMed] [Google Scholar]
- 43.Farooqi IS, Matarese G, Lord GM, et al. Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J Clin Invest. 2002;110(8 ): 1093–103. doi: 10.1172/JCI15693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Rosenbaum M, Murphy EM, Heymsfield SB, Matthews DE, Leibel RL. Low dose leptin administration reverses effects of sustained weight-reduction on energy expenditure and circulating concentrations of thyroid hormones. J Clin Endocrinol Metab. 2002;87(5 ): 2391–4. doi: 10.1210/jcem.87.5.8628. [DOI] [PubMed] [Google Scholar]
- 45.Novakovic ZM, Leinung MC, Lee DW, Grasso P. Intranasal administration of mouse [D-Leu-4]OB3, a synthetic peptide amide with leptin-like activity, enhances total uptake and bioavailability in Swiss Webster mice when compared to intraperitoneal, subcutaneous, and intramuscular delivery systems. Regul Pept. 2009;154(1-3 ): 107–11. doi: 10.1016/j.regpep.2009.01.002. [DOI] [PubMed] [Google Scholar]
- 46.Otvos L Jr, Terrasi M, Cascio S, et al. Development of a pharmacologically improved peptide agonist of the leptin receptor. Biochem Biophys Acta. 2008;1783(10 ): 1745–54. doi: 10.1016/j.bbamcr.2008.05.007. [DOI] [PubMed] [Google Scholar]
- 47.Roth JD, Roland BL, Cole RL, et al. Leptin responsiveness restored by amylin agonism in diet-induced obesity evidence from nonclinical and clinical studies. Proc Natl AcadSci USA. 2008;105(20 ): 7257–62. doi: 10.1073/pnas.0706473105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Talbert ME, Langefeld CD, Ziegler J, et al. Polymorphisms near SOCS3 are associated with obesity and glucose homeostasis traits in Hispanic Americans from the Insulin Resistance Atherosclerosis Family Study. Hum Genet. 2009;125(2 ):153–62. doi: 10.1007/s00439-008-0608-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Cheyssac C, Lecoeur C, Dechaume A, et al. Analysis of common PTPN1 gene variants in type 2 diabetes, obesity and associated phenotypes in the French population. BMC Med Genet. 2006;7:44–54. doi: 10.1186/1471-2350-7-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Howard JK, Cave BJ, Oksanen LJ, Tzameli I, Bjørbaek C, Flier JS. Enhanced leptin sensitivity and attenuation of diet-induced obesity in mice with haploinsufficiency of SOCS3. Nat Med. 2004;10(7 ): 734–8. doi: 10.1038/nm1072. [DOI] [PubMed] [Google Scholar]
- 51.Zhang ZY, Lee SY. PTP1B inhibitors as potential therapeutics in the treatment of type 2 diabetes and obesity. Expert Opin Investig Drugs. 2003;12(2 ): 223–33. doi: 10.1517/13543784.12.2.223. [DOI] [PubMed] [Google Scholar]
- 52.Lantz KA, Hart SG, Planey SL. Inhibition of PTP1B by Trodusquemine (MSI-1436) Causes Fat-specific Weight Loss in Diet-induced Obese Mice. Obesity (Silver Spring) 2010;18(8 ):1516–23. doi: 10.1038/oby.2009.444. [DOI] [PubMed] [Google Scholar]
- 53.Coll AP, Loraine Tung YC. Pro-opiomelanocortin (POMC)-derived peptides and the regulation of energy homeostasis. Mol Cell Endocrinol. 2009;300(1-2 ): 147–51. doi: 10.1016/j.mce.2008.09.007. [DOI] [PubMed] [Google Scholar]
- 54.Barb CR, Robertson AS, Barrett JB, Kraeling RR, Houseknecht KL. The role of melanocortin-3 and -4 receptor in regulating appetite, energy homeostasis and neuroendocrine function in the pig. J Endocrinol. 2004;181(1 ): 39–52. doi: 10.1677/joe.0.1810039. [DOI] [PubMed] [Google Scholar]
- 55.Masuzaki H, Tanaka T, Ebihara K, Hosoda K. Energy homeostasis regulated by an adipocyte-derived hormone-- mechanism of leptin resistance and role of hypothalamic melanocortin signaling. Nippon Rinsho. 2009;67(2 ): 287–96. [PubMed] [Google Scholar]
- 56.Huszar D, Lynch CA, Fairchild-Huntress V. Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell. 1997;88(1 ): 131–41. doi: 10.1016/s0092-8674(00)81865-6. [DOI] [PubMed] [Google Scholar]
- 57.Vaisse C, Clement K, Durand E, Hercberg S, Guy-Grand B, Froguel P. Melanocortin-4 receptor mutations are a frequent and heterogeneous cause of morbid obesity. J Clin Invest. 2000;106(2 ): 253–62. doi: 10.1172/JCI9238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ni XP, Butler AA, Cone RD, Humphreys MH. Central receptors mediating the cardiovascular actions of melanocyte stimulating hormones. J Hypertens. 2006;24(11 ): 2239–46. doi: 10.1097/01.hjh.0000249702.49854.fa. [DOI] [PubMed] [Google Scholar]
- 59.Giuliano F. Control of penile erection by the melanocortinergic system experimental evidences and therapeutic perspectives. J Androl. 2004;25(5 ): 683–91. doi: 10.1002/j.1939-4640.2004.tb02842.x. [DOI] [PubMed] [Google Scholar]
- 60.Deboer MD, Marks DL. Cachexia lessons from melanocortin antagonism. Trends EndocrinolMetab. 2006;17(5 ): 199–204. doi: 10.1016/j.tem.2006.05.005. [DOI] [PubMed] [Google Scholar]
- 61.Lee M, Wardlaw SL. The central melanocortin system and the regulation of energy balance. Front Biosci. 2007;12:3994–4010. doi: 10.2741/2366. [DOI] [PubMed] [Google Scholar]
- 62.Ye Z, Guo L, Barakat KJ, et al. Discovery and activity of (1R,4S,6R)-N-[(1R)-2-[4-cyclohexyl-4-[[(1,1- dimethylethyl)amino]carbonyl]-1-piperidinyl]-1-[(4- fluorophenyl)methyl]-2-oxoethyl]-2-methyl-2- azabicyclo[2.2.2]octane-6-carboxamide (3, RY764), a potent and selective melanocortin subtype-4 receptor agonist. Bioorg Med Chem Lett. 2005;15(15 ):3501–5. doi: 10.1016/j.bmcl.2005.05.109. [DOI] [PubMed] [Google Scholar]
- 63.Krishna R, Gumbiner B, Stevens C, et al. Potent and selective agonism of the melanocortin receptor 4 with MK-0493 does not induce weight loss in obese human subjects: energy intake predicts lack of weight loss efficacy. Clin Pharmacol Ther. 2009;86(6 ): 659–66. doi: 10.1038/clpt.2009.167. [DOI] [PubMed] [Google Scholar]
- 64.Diamond LE, Earle DC, Rosen RC, Willett MS, Molinoff PB. Double-blind, placebo-controlled evaluation of the safety, pharmacokinetic properties and pharmacodynamic effects of intranasal PT-141, a melanocortin receptor agonist, in healthy males and patients with mild-to-moderate erectile dysfunction. Int J Impot Res. 2004;16(1 ): 51–9. doi: 10.1038/sj.ijir.3901139. [DOI] [PubMed] [Google Scholar]
- 65.Wisse BE, Frayo RS, Schwartz MW, Cummings DE. Reversal of cancer anorexia by blockade of central melanocortin receptors in rats. Endocrinology. 2001;142(8 ): 3292–301. doi: 10.1210/endo.142.8.8324. [DOI] [PubMed] [Google Scholar]
- 66.Vos TJ, Caracoti A, Che JL, et al. Identification of 2-[2-[2-(5-bromo-2- methoxyphenyl)-ethyl]-3-fluorophenyl]-4,5-dihydro-1H-imidazole (ML00253764), a small molecule melanocortin 4 receptor antagonist that effectively reduces tumor-induced weight loss in a mouse model. J Med Chem. 2004;47(7 ):1602–4. doi: 10.1021/jm034244g. [DOI] [PubMed] [Google Scholar]
- 67.Baskin DG. Single-minded view of melanocortin signaling in energy homeostasis. Endocrinology. 2006;147(10 ): 4539–41. doi: 10.1210/en.2006-0807. [DOI] [PubMed] [Google Scholar]
- 68.Kublaoui BM, Holder JL Jr, Gemelli T, Zinn AR. SIM1 haploin-sufficiency impairs melanocortin-mediated anorexia and activation of paraventricular nucleus neurons. Mol Endocrinol. 2006;20(10 ): 2483–92. doi: 10.1210/me.2005-0483. [DOI] [PubMed] [Google Scholar]
- 69.Michaud JL, Boucher F, Melnyk A, et al. SIM1 haploinsufficiency causes hyperphagia, obesity and reduction of the paraventricular nucleus of the hypothalamus. Hum Mol Genet. 2001;10:1465–73. doi: 10.1093/hmg/10.14.1465. [DOI] [PubMed] [Google Scholar]
- 70.Kublaoui BM, Holder JL , Jr, Tolson KP, Gemelli T, Zinn AR. SIM1 overexpression partially rescues agouti yellow and diet-induced obesity by normalizing food intake. Endocrinology. 2006;147(10 ): 4542–9. doi: 10.1210/en.2006-0453. [DOI] [PubMed] [Google Scholar]
- 71.ALS CNTF Treatment Study Group. A double-blind placebo-controlled clinical trial of subcutaneous recombinant human ciliary neurotrophic factor (rHCNTF) in amyotrophic lateral sclerosis. Neurology. 1996;46(5 ): 1244–9. doi: 10.1212/wnl.46.5.1244. [DOI] [PubMed] [Google Scholar]
- 72.Lambert PD, Anderson KD, Sleeman MW, et al. Ciliary neurotrophic factor activates leptin-like pathways and reduces body fat, without cachexia or rebound weight gain, even in leptin-resistant obesity. Proc Natl Acad Sci USA. 2001;98(8 ): 4652–7. doi: 10.1073/pnas.061034298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Kokoeva MV, Yin H, Flier JS. Neurogenesis in the hypothalamus of adult mice: potential role in energy balance. Science. 2005;310(5748 ): 679–83. doi: 10.1126/science.1115360. [DOI] [PubMed] [Google Scholar]
- 74.Ettinger MP, Littlejohn TW, Schwartz SL, et al. Recombinant variant of ciliary neurotrophic factor for weight loss in obese adults a randomized, dose-ranging study. JAMA. 2003;289(14 ): 1826–32. doi: 10.1001/jama.289.14.1826. [DOI] [PubMed] [Google Scholar]
- 75.Pollack A. Obesity Drug Trial Produces Disappointing Weight Loss (axokine).The New York Times. Published on: April 1, 2003. Available from: http://www.nytimes.com/2003/04/01/business/obesity-drug-trialproduces- disappointing-weight-loss.html . [Accessed on September 9, 2011].
- 76.Astrup A, Madsbad S, Breum L, Jensen TJ, Kroustrup JP, Larsen TM. Effect of tesofensine on bodyweight loss, body composition, and quality of life in obese patients a randomised, double-blind, placebo-controlled trial. Lancet. 2008;372(9653 ): 1906–13. doi: 10.1016/S0140-6736(08)61525-1. [DOI] [PubMed] [Google Scholar]
- 77.Bello NT, Zahner MR. Tesofensine, a monoamine reuptake inhibitor for the treatment of obesity. Curr Opin Investig Drugs. 2009;10(10 ): 1105–16. [PubMed] [Google Scholar]
- 78.Rothman RB, Baumann MH. Serotonergic drugs and valvular heart disease. Expert Opin Drug Saf. 2009;8(3 ): 317–29. doi: 10.1517/14740330902931524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Blaise G, Langleben D, Hubert B. Pulmonary arterial hypertension pathophysiology and anesthetic approach. Anesthesiology. 2003;99(6 ): 1415–32. doi: 10.1097/00000542-200312000-00027. [DOI] [PubMed] [Google Scholar]
- 80.Heisler LK, Cowley MA, Kishi T, et al. Central serotonin and melanocortin pathways regulating energy homeostasis. Ann N Y Acad Sci. 2003;994:169–74. doi: 10.1111/j.1749-6632.2003.tb03177.x. [DOI] [PubMed] [Google Scholar]
- 81.Smith BM, Smith JM, Tsai JH. Discovery and structure-activity relationship of (1R)-8-chloro-2,3,4,5-tetrahydro-1-methyl-1H-3-benzazepine (Lorcaserin), a selective serotonin 5-HT2C receptor agonist for the treatment of obesity. J Med Chem. 2008;51(2 ): 305–13. doi: 10.1021/jm0709034. [DOI] [PubMed] [Google Scholar]
- 82.Smith SR, Prosser WA, Donahue DJ, Morgan ME, Anderson CM, Shanahan WR. Lorcaserin (APD356), a selective 5-HT(2C) agonist, reduces body weight in obese men and women. Obesity (Silver Spring) 2009;17(3 ): 494–503. doi: 10.1038/oby.2008.537. [DOI] [PubMed] [Google Scholar]
- 83.Smith SR, Weissman NJ, Anderson CM, et al. Multicenter, placebo-controlled trial of lorcaserin for weight management. N Engl J Med. 2010;363(3 ): 245–56. doi: 10.1056/NEJMoa0909809. [DOI] [PubMed] [Google Scholar]
- 84.Fidler MC, Sanchez M, Raether B. One-Year Randomized Trial of Lorcaserin for Weight Loss in Obese and Overweight Adults: The BLOSSOM Trial. J Clin Endocrinol Metab. 2011;96(10 ): 3067–77. doi: 10.1210/jc.2011-1256. [DOI] [PubMed] [Google Scholar]
- 85.Pollack A. No FDA approval for new diet pill. The New York Times. Published on: October 23, 2010. Available from: http://www.nytimes.com/2010/10/24/business/24obesity.html . [Accessed on September 9, 2011].
- 86.Woolley ML, Bentley JC, Sleight AJ, Marsden CA, Fone KC. A role for 5-HT6 receptors in retention of spatial learning in the Morris water maze. Neuropharmacology. 2001;41(2 ): 210–9. doi: 10.1016/s0028-3908(01)00056-9. [DOI] [PubMed] [Google Scholar]
- 87.Heal DJ, Smith SL, Fisas A, Codony X, Buschmann H. Selective 5-HT6 receptor ligands progress in the development of a novel pharmacological approach to the treatment of obesity and related metabolic disorders. Pharmacol Ther. 2008;117(2 ): 207–31. doi: 10.1016/j.pharmthera.2007.08.006. [DOI] [PubMed] [Google Scholar]
- 88.Hirst WD, Abrahamsen B, Blaney FE, et al. Differences in the central nervous system distribution and pharmacology of the mouse 5-hydroxytryptamine-6 receptor compared with rat and human receptors investigated by radioligand binding, site-directed mutagenesis, and molecular modeling. Mol Pharmacol. 2003;64(6 ): 1295–308. doi: 10.1124/mol.64.6.1295. [DOI] [PubMed] [Google Scholar]
- 89.Schwartz JC, Arrang JM, Garbarg M, Pollard H, Ruat M. Histaminergic transmission in the mammalian brain. Physiol Rev. 1991;71(1 ): 1–51. doi: 10.1152/physrev.1991.71.1.1. [DOI] [PubMed] [Google Scholar]
- 90.Masaki T, Yoshimatsu H, Chiba S, Watanabe T, Sakata T. Central infusion of histamine reduces fat accumulation and upregulates UCP family in leptin-resistant obese mice. Diabetes. 2001;50(2 ): 376–84. doi: 10.2337/diabetes.50.2.376. [DOI] [PubMed] [Google Scholar]
- 91.Sander K, Kottke , Stark H. Histamine H3 Receptor Antagonists Go to Clinics. Biol Pharm Bull. 2008;31(12 ):2163–81. doi: 10.1248/bpb.31.2163. [DOI] [PubMed] [Google Scholar]
- 92.ClinicalTrial.go . Efficacy and Safety Trial to Evaluate the Effect of SCH 497079 on Weight in Obese and Overweight Subjects (Study P05483)( Last Updated on September 30, 2009) Available from: http://clinicaltrials.gov/ct2/show/NCT00642993 . [Accessed on September 9, 2011].
- 93.Hancock AA, Bennani YL, Bush EN, et al. Antiobesity effects of A-331440, a novel non-imidazole histamine H3 receptor antagonist. Eur J Pharmacol. 2004;487(1-3 ): 183–97. doi: 10.1016/j.ejphar.2004.01.015. [DOI] [PubMed] [Google Scholar]
- 94.Vermaak I, Hamman JH, Viljoen AM. Hoodia gordonii: an up-to-date review of a commercially important anti-obesity plant. Planta Med. 2011;77(11 ): 1149–60. doi: 10.1055/s-0030-1250643. [DOI] [PubMed] [Google Scholar]
- 95.Bray GA, Greenway FL. Pharmacological treatment of the over-weight patient. Pharmacol Rev. 2007;59(2 ): 151–84. doi: 10.1124/pr.59.2.2. [DOI] [PubMed] [Google Scholar]
- 96.Duenwald M. An Appetite Killer for a Killer Appetite? Not Yet. The New York Times. Published on: April 19, 2005. Available at: http://www.nytimes.com/2005/04/19/health/nutrition/19cons.html . [Accessed on September 9, 2011].
- 97.Oh-I S, Shimizu H, Satoh T, et al. Identification of nesfatin-1 as a satiety molecule in the hypothalamus. Nature. 2006;443(7112 ): 709–12. doi: 10.1038/nature05162. [DOI] [PubMed] [Google Scholar]
- 98.Shimizu H, Oh-I S, Okada S, Mori M. Nesfatin-1: an overview and future clinical application. Endocr J. 2009;56(4 ): 537–43. doi: 10.1507/endocrj.k09e-117. [DOI] [PubMed] [Google Scholar]
- 99.Inoue K, Valdez GR, Reyes TM, et al. Human urocortin II, a selective agonist for the type 2 corticotropin-releasing factor receptor, decreases feeding and drinking in the rat. J Pharmacol Exp Ther. 2003;305(1 ): 385–93. doi: 10.1124/jpet.102.047712. [DOI] [PubMed] [Google Scholar]
- 100.Chen P, Vaughan J, Donaldson C, Vale W, Li C. Injection of Urocortin 3 into the ventromedial hypothalamus modulates feeding, blood glucose levels and hypothalamic POMC gene expression but not the HPA axis. Am J Physiol Endocrinol Metab. 2010;298(2 ): E337–45. doi: 10.1152/ajpendo.00402.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Jamieson PM, Cleasby ME, Kuperman Y, et al. Urocortin 3 transgenic mice exhibit a metabolically favourable phenotype resisting obesity and insulin resistance on a high fat diet. Diabetologia. 2011;54(9 ): 2392–403. doi: 10.1007/s00125-011-2205-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Budhiraja S, Chugh A. Neuromedin U physiology, pharmacology and therapeutic potential. Fundam Clin Pharmacol. 2009;23(2 ): 149–57. doi: 10.1111/j.1472-8206.2009.00667.x. [DOI] [PubMed] [Google Scholar]
- 103.Doggrell SA. Neuromedin U- a new target in obesity. Expert OpinTher Targets. 2005;9(4 ): 875–7. doi: 10.1517/14728222.9.4.875. [DOI] [PubMed] [Google Scholar]
- 104.Egecioglu E, Ploj K, Xu X, et al. Central NMU signaling in body weight and energy balance regulation: evidence from NMUR2 deletion and chronic central NMU treatment in mice. m J Physiol Endocrinol Metab. 2009;297(3 ): E708–16. doi: 10.1152/ajpendo.91022.2008. [DOI] [PubMed] [Google Scholar]
- 105.Peier a, Kosinski j, Cox-york k, et al. The antiobesity effects of centrally administered neuromedin u and neuromedin s are mediated predominantly by the neuromedin u receptor 2 (nmur2) Endocrinology. 2009;150(7 ): 3101–9. doi: 10.1210/en.2008-1772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Ronnett GV, Kleman AM, Kim EK, Landree LE, Tu Y. Fatty acid metabolism, the central nervous system and feeding. Obesity (Silver Spring) 2006;14(Suppl 5 ): 201S–7S. doi: 10.1038/oby.2006.309. [DOI] [PubMed] [Google Scholar]
- 107.Kuhajda FP, Landree LE, Ronnett GV. The connections between C75 and obesity drug-target pathways. Trends Pharmacol Sci. 2005;26(11 ): 541–4. doi: 10.1016/j.tips.2005.09.002. [DOI] [PubMed] [Google Scholar]
- 108.Yamada K, Wada E, Santo-Yamada Y, Wada K. Bombesin and its family of peptides prospects for the treatment of obesity. Eur J Pharmacol. 2002;440(2-3 ): 281–90. doi: 10.1016/s0014-2999(02)01435-8. [DOI] [PubMed] [Google Scholar]
- 109.Lieverse RJ, Jansen JB, van de Zwan A, et al. Bombesin reduces food intake in lean man by a cholecystokinin-independent mechanism. J Clin Endocrinol Metab. 1993;76(6 ): 1495–8. doi: 10.1210/jcem.76.6.8501156. [DOI] [PubMed] [Google Scholar]
- 110.Lieverse RJ, Jansen JB, Masclee AA, Lamers CB. Bombesin reduces food intake after a preload in man by a cholecystokinin-independent mechanism. ClinSci (Lond) 1993;85(3 ): 277–80. doi: 10.1042/cs0850277. [DOI] [PubMed] [Google Scholar]
- 111.Lieverse RJ, Jansen JB, Masclee AA, Lamers CB. Significant satiety effect of bombesin in lean but not in obese subjects. Int J Obes Relat Metab Disord. 1994;18(8 ): 579–83. [PubMed] [Google Scholar]
- 112.Ohki-Hamazaki H, Watase K, Yamamoto K, et al. Mice lacking bombesin receptor subtype-3 develop metabolic defects and obesity. Nature. 1997;390(6656 ): 165–9. doi: 10.1038/36568. [DOI] [PubMed] [Google Scholar]
- 113.Guan XM, Chen H, Dobbelaar PH, et al. Regulation of energy homeostasis by bombesin receptor subtype-3 selective receptor agonists for the treatment of obesity. Cell Metab. 2010;11(2 ): 101–12. doi: 10.1016/j.cmet.2009.12.008. [DOI] [PubMed] [Google Scholar]
- 114.Steigerwalt RW, Williams JA. Binding specificity of the mouse cerebral cortex receptor for small cholecystokinin peptides. Regul Pept. 1984;8(1 ): 51–9. doi: 10.1016/0167-0115(84)90028-4. [DOI] [PubMed] [Google Scholar]
- 115.Szewczyk JR, Laudeman C. CCK1R agonists: a promising target for the pharmacological treatment of obesity. Curr Top Med Chem. 2003;3(8 ): 837–54. doi: 10.2174/1568026033452258. [DOI] [PubMed] [Google Scholar]
- 116.Jordan J, Greenway FL, Leiter LA. Stimulation of cholecystokinin-A receptors with GI181771X does not cause weight loss in overweight or obese patients. Clin Pharmacol Ther. 2008;83(2 ): 281–7. doi: 10.1038/sj.clpt.6100272. [DOI] [PubMed] [Google Scholar]
- 117.Crawley JN, Beinfeld MC. Rapid development of tolerance to the behavioral actions of cholecystokinin. Nature. 1983;302(5910 ): 703–6. doi: 10.1038/302703a0. [DOI] [PubMed] [Google Scholar]
- 118.Matson CA, Reid DF, Ritter RC. Daily CCK injection enhances reduction of body weight by chronic intracerebroventricular leptin infusion. Am J Physiol Regul Integr Comp Physiol. 2002;282(5 ): R1368–73. doi: 10.1152/ajpregu.00080.2001. [DOI] [PubMed] [Google Scholar]
- 119.FernándezLandó L, Casellini CM. Exenatide trials for the treatment of type 2 diabetes. Medicina (B Aires) 2009;69(4 ): 447–57. [PubMed] [Google Scholar]
- 120.DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care. 2005;28(5 ): 1092–100. doi: 10.2337/diacare.28.5.1092. [DOI] [PubMed] [Google Scholar]
- 121.Kendall DM, Riddle MC, Rosenstock J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea. Diabetes Care. 2005;28(5 ): 1083–91. doi: 10.2337/diacare.28.5.1083. [DOI] [PubMed] [Google Scholar]
- 122.Klonoff DC, Buse JB, Nielsen LL, et al. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin. 2008;24(1 ): 275–86. doi: 10.1185/030079908x253870. [DOI] [PubMed] [Google Scholar]
- 123.Kim D, MacConell L, Zhuang D, et al. Effects of once-weekly dosing of a long-acting release formulation of exenatide on glucose control and body weight in subjects with type 2 diabetes. Diabetes Care. 2007;30(6 ): 1487–93. doi: 10.2337/dc06-2375. [DOI] [PubMed] [Google Scholar]
- 124.Drucker DJ, Buse JB, Taylor K, et al. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes a randomised, open-label, non-inferiority study. Lancet. 2008;372(9645 ): 1240–50. doi: 10.1016/S0140-6736(08)61206-4. [DOI] [PubMed] [Google Scholar]
- 125.Parks M, Rosebraugh C. Weighing risks and benefits of liraglutide--the FDA's review of a new antidiabetic therapy. N Engl J Med. 2010;362(9 ): 774–7. doi: 10.1056/NEJMp1001578. [DOI] [PubMed] [Google Scholar]
- 126.Neumiller JJ. Differential chemistry (structure), mechanism of action, and pharmacology of GLP-1 receptor agonists and DPP-4 inhibitors. J Am Pharm Assoc. 2009;49(Suppl 1 ): S16–29. doi: 10.1331/JAPhA.2009.09078. [DOI] [PubMed] [Google Scholar]
- 127.Astrup A, Rössner S, Van Gaal L, et al. NN8022-1807 Study Group. Effects of liraglutide in the treatment of obesity: a randomised, double-blind, placebo-controlled study. Lancet. 2009;374(9701 ): 1606–16. doi: 10.1016/S0140-6736(09)61375-1. [DOI] [PubMed] [Google Scholar]
- 128.Neary MT, Batterham RL. Gut hormones implications for the treatment of obesity. Pharmacol Ther. 2009;124(1 ): 44–56. doi: 10.1016/j.pharmthera.2009.06.005. [DOI] [PubMed] [Google Scholar]
- 129.Batterham RL, Cowley MA, Small CJ, et al. Gut hormone PYY(3-36) physiologically inhibits food intake. Nature. 2002;418(6898 ): 650–4. doi: 10.1038/nature00887. [DOI] [PubMed] [Google Scholar]
- 130.Batterham RL, Cohen MA, Ellis SM, et al. Inhibition of food intake in obese subjects by peptide YY3-36. N Engl J Med. 2003;349(10 ): 941–8. doi: 10.1056/NEJMoa030204. [DOI] [PubMed] [Google Scholar]
- 131.Gantz I, Erondu N, Mallick M. Efficacy and safety of intranasal peptide YY3-36 for weight reduction in obese adults. J Clin Endocrinol Metab. 2007;92(5 ): 1754–7. doi: 10.1210/jc.2006-1806. [DOI] [PubMed] [Google Scholar]
- 132.Chen SC, Eiting K, Cui K, et al. Therapeutic Utility of a Novel Tight Junction Modulating Peptide for Enhancing Intranasal Drug Delivery. J Pharm Sci. 2006;95(6 ): 1364–71. doi: 10.1002/jps.20510. [DOI] [PubMed] [Google Scholar]
- 133.Cohen MA, Ellis SM, Le Roux CW, et al. Oxyntomodulin suppresses appetite and reduces food intake in humans. J Clin Endocrinol Metab. 2003;88(10 ): 4696–701. doi: 10.1210/jc.2003-030421. [DOI] [PubMed] [Google Scholar]
- 134.Wynne K, Park AJ, Small CJ, et al. Subcutaneous oxyntomodulin reduces body weight in overweight and obese subjects a double-blind, randomized, controlled trial. Diabetes. 2005;54(8 ): 2390–5. doi: 10.2337/diabetes.54.8.2390. [DOI] [PubMed] [Google Scholar]
- 135.Wynne K, Park AJ, Small CJ, et al. Oxyntomodulin increases energy expenditure in addition to decreasing energy intake in overweight and obese humans a randomised controlled trial. Int J Obes (Lond) 2006;30(12 ): 1729–36. doi: 10.1038/sj.ijo.0803344. [DOI] [PubMed] [Google Scholar]
- 136.Bloomgarden ZT. Obesity mediators and treatment approaches. Diabetes Care. 2009;32(5 ): e48–52. doi: 10.2337/dc09-zb05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Mcgavigan ak, Murphy Kg. Gut hormones the future of obesity treatment? Br j clin pharmacol. 2012 Mar 27; doi: 10.1111/j.1365-2125.2012.04278.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Woods SC, Lutz TA, Geary N, Langhans W. Pancreatic signals controlling food intake; insulin, glucagon and amylin . Philos Trans R Soc Lond B Biol Sci. 2006;361(1471 ): 1219–35. doi: 10.1098/rstb.2006.1858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Butler PC, Chou J, Carter WB, et al. Effects of meal ingestion on plasma amylin concentration in NIDDM and nondiabetic humans. Diabetes. 1990;39(6 ): 752–6. doi: 10.2337/diab.39.6.752. [DOI] [PubMed] [Google Scholar]
- 140.Lutz ta. Pancreatic amylin as a centrally acting satiating hormone. Curr drug targets. 2005;6(2 ): 181–9. doi: 10.2174/1389450053174596. [DOI] [PubMed] [Google Scholar]
- 141.lutz ta, geary n, szabady mm, del prete e, scharrer e. Amylin decreases meal size in rats. Physiol behav. 1995;58(6 ): 1197–202. doi: 10.1016/0031-9384(95)02067-5. [DOI] [PubMed] [Google Scholar]
- 142. Reidelberger Rd, Haver ac, arnelo u, smith dd, schaffert cs, permert j. Amylin receptor blockade stimulates food intake in rats. Am j physiol regul integr comp physiol. 2004;287(3 ): r568–7. doi: 10.1152/ajpregu.00213.2004. [DOI] [PubMed] [Google Scholar]
- 143.Ryan G, Briscoe TA, Jobe L. Review of pramlintide as adjunctive therapy in treatment of type 1 and type 2 diabetes. Drug Des Devel Ther. 2009;2:203–14. doi: 10.2147/dddt.s3225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Aronne L, Fujioka K, Aroda V, et al. Progressive reduction in body weight after treatment with the amylin analog pramlintide in obese subjects a phase 2, randomized, placebo-controlled, dose-escalation study. J Clin Endocrinol Metab. 2007;92(8 ): 2977–83. doi: 10.1210/jc.2006-2003. [DOI] [PubMed] [Google Scholar]
- 145.Okada S, York DA, Bray GA, Erlanson-Albertsson C. Enterostatin (Val-Pro-Asp-Pro-Arg), the activation peptide of procolipase, selectively reduces fat intake. Physiol Behav. 1991;49(6 ): 1185–9. doi: 10.1016/0031-9384(91)90349-s. [DOI] [PubMed] [Google Scholar]
- 146.Lin L, Chen J, York DA. Chronic ICV enterostatin preferentially reduced fat intake and lowered body weight. Peptides. 1997;18(5 ): 657–61. doi: 10.1016/s0196-9781(97)00128-9. [DOI] [PubMed] [Google Scholar]
- 147.Bowyer RC, Rowston WM, Jehanli AM, Lacey JH, Hermon-Taylor J. Effect of a satiating meal on the concentrations of procolipase-propeptide in the serum and urine of normal and morbidly obese subjects. Gut. 1993;34(11 ): 1520–5. doi: 10.1136/gut.34.11.1520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Prasad C, Imamura M, Debata C, Svec F, Sumar N, Hermon-Taylor J. Hyperenterostatinemia in premenopausal obese women. J Clin Endocrinol Metab. 1999;84(3 ): 937–41. doi: 10.1210/jcem.84.3.5562. [DOI] [PubMed] [Google Scholar]
- 149.Rössner S, Barkeling B, Erlanson-Albertsson C, Larsson P, Wåhlin-Boll E. Intravenous enterostatin does not affect single meal food intake in man. Appetite. 1995;24(1 ): 37–42. doi: 10.1016/s0195-6663(95)80004-2. [DOI] [PubMed] [Google Scholar]
- 150.Kanatani A, Hata M, Mashiko S, et al. A typical Y1 receptor regulates feeding behaviors: effects of a potent and selective Y1 antagonist, J-115814. Mol Pharmacol. 2001;59(3 ): 501–5. doi: 10.1124/mol.59.3.501. [DOI] [PubMed] [Google Scholar]
- 151.Antal-Zimanyi I, Bruce MA, Leboulluec KL, et al. Pharmacological characterization and appetite suppressive properties of BMS-193885, a novel and selective neuropeptide Y(1) receptor antagonist. Eur J Pharmacol. 2008;590(1-3 ): 224–32. doi: 10.1016/j.ejphar.2008.06.032. [DOI] [PubMed] [Google Scholar]
- 152.Daniels AJ, Grizzle MK, Wiard RP, Matthews JE, Heyer D. Food intake inhibition and reduction in body weight gain in lean and obese rodents treated with GW438014A, a potent and selective NPY-Y5 receptor antagonist. Regul Pept. 2002;106(1-3 ): 47–54. doi: 10.1016/s0167-0115(02)00034-4. [DOI] [PubMed] [Google Scholar]
- 153.Turnbull AV, Ellershaw L, Masters DJ, et al. Selective antagonism of the NPY Y5 receptor does not have a major effect on feeding in rats. Diabetes. 2002;51(8 ): 2441–9. doi: 10.2337/diabetes.51.8.2441. [DOI] [PubMed] [Google Scholar]
- 154.Kamiji MM, Inui A. Neuropeptide Y receptor selective ligands in the treatment of obesity. Endocr Rev. 2007;28(6 ): 664–84. doi: 10.1210/er.2007-0003. [DOI] [PubMed] [Google Scholar]
- 155.Erondu N, Gantz I, Musser B, et al. Neuropeptide Y5 receptor antagonism does not induce clinically meaningful weight loss in overweight and obese adults. Cell Metab. 2006;4(4 ):275–82. doi: 10.1016/j.cmet.2006.08.002. [DOI] [PubMed] [Google Scholar]
- 156.Erickson JC, Clegg KE, Palmiter RD. Sensitivity to leptin and susceptibility to seizures of mice lacking neuropeptide Y. Nature. 1996;381(6581 ): 415–21. doi: 10.1038/381415a0. [DOI] [PubMed] [Google Scholar]
- 157.Erickson JC, Hollopeter G, Palmiter RD. Attenuation of the obesity syndrome of ob/ob mice by the loss of neuropeptideY. Science. 1996;274(5293 ): 1704–7. doi: 10.1126/science.274.5293.1704. [DOI] [PubMed] [Google Scholar]
- 158.Ludwig DS, Tritos NA, Mastaitis JW, et al. Melanin-concentrating hormone overexpression in transgenic mice leads to obesity and insulin resistance. J Clin Invest. 2001;107(3 ): 379–86. doi: 10.1172/JCI10660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Pissios P, Bradley RL, Maratos-Flier E. Expanding the scales: The multiple roles of MCH in regulating energy balance and other biological functions. Endocr Rev. 2006;27(6 ): 606–20. doi: 10.1210/er.2006-0021. [DOI] [PubMed] [Google Scholar]
- 160.Marsh DJ, Weingarth DT, Novi DE, et al. Melanin-concentrating hormone 1 receptor-deficient mice are lean, hyperactive, and hyperphagic and have altered metabolism. Proc Natl Acad Sci USA. 2002;99(5 ): 3240–5. doi: 10.1073/pnas.052706899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Lin SJ, Ford E, Haigis M, Liszt G, Guarente L. Calorie restriction extends yeast life span by lowering the level of NADH. Genes Dev. 2004;18(1 ): 12–6. doi: 10.1101/gad.1164804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Anderson RM, Shanmuganayagam D, Weindruch R. Caloric restriction and aging studies in mice and monkeys. Toxicol Pathol. 2009;37(1 ): 47–51. doi: 10.1177/0192623308329476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Heilbronn LK, de Jonge L, Frisard MI, et al. Effect of 6-month calorie restriction on biomarkers of longevity, metabolic adaptation, and oxidative stress in overweight individuals: a randomized controlled trial. JAMA. 2006;295(13 ): 1539–48. doi: 10.1001/jama.295.13.1539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Méndez-Andino JL, Wos JA. MCH-R1 antagonists what is keeping most research programs away from the clinic? Drug Discov Today. 2007;12(21-22 ): 972–9. doi: 10.1016/j.drudis.2007.08.010. [DOI] [PubMed] [Google Scholar]
- 165.Willie JT, Sinton CM, Maratos-Flier E, Yanagisawa M. Abnormal response of melanin-concentrating hormone deficient mice to fasting hyperactivity and rapid eye movement sleep suppression. Neuroscience. 2008;156(4 ): 819–29. doi: 10.1016/j.neuroscience.2008.08.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Haberman AB. Obesity drug pipeline: developing therapies for a complex disease. Obesity drug pipeline report overview. Insight Pharma reports. (Published in: September 2008) Available at: http://www.insightpharmareports.com/reports_report.aspx?r=653& id=81176 . [Accessed on: September 9, 2011].
- 167.Sakurai T, Amemiya A, Ishii M, et al. Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell. 1998;92(4 ): 573–85. doi: 10.1016/s0092-8674(00)80949-6. [DOI] [PubMed] [Google Scholar]
- 168.Piper DC, Upton N, Smith MI, Hunter AJ. The novel brain neuropeptide, orexin-A, modulates the sleep-wake cycle of rats. Eur J Neurosci. 2000;12(2 ): 726–30. doi: 10.1046/j.1460-9568.2000.00919.x. [DOI] [PubMed] [Google Scholar]
- 169.Smart D, Sabido-David C, Brough SJ, et al. SB-334867-A the first selective orexin-1 receptor antagonist. Br J Pharmacol. 2001;132(6 ): 1179–82. doi: 10.1038/sj.bjp.0703953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Haynes AC, Chapman H, Taylor C, et al. Anorectic thermogenic and anti-obesity activity of a selective orexin-1 receptor antagonist in ob/ob mice. Regul Pept. 2002;104(1-3 ): 153–9. doi: 10.1016/s0167-0115(01)00358-5. [DOI] [PubMed] [Google Scholar]
- 171.Kyrkouli SE, Stanley BG, Leibowitz SF. Galanin stimulation of feeding induced by medial hypothalamic injection of this novel peptide. Eur J Pharmacol. 1986;122(1 ): 159–60. doi: 10.1016/0014-2999(86)90175-5. [DOI] [PubMed] [Google Scholar]
- 172.Corwin RL, Robinson JK, Crawley JN. Galanin antagonists block galanin-induced feeding in the hypothalamus and amygdala of the rat. Eur J Neurosci. 1993;5(11 ): 1528–33. doi: 10.1111/j.1460-9568.1993.tb00221.x. [DOI] [PubMed] [Google Scholar]
- 173.Leibowitz SF. Differential functions of hypothalamic galanin cell grow in the regulation of eating and body weight. Ann N Y Acad Sci. 1998;863:206–20. doi: 10.1111/j.1749-6632.1998.tb10696.x. [DOI] [PubMed] [Google Scholar]
- 174.Adams AC, Clapham JC, Wynick D, Speakman JR. Feeding behavior in galanin knockout mice supports a role of galanin in fat intake and preference. J Neuroendocrinol. 2008;20(2 ): 199–206. doi: 10.1111/j.1365-2826.2007.01638.x. [DOI] [PubMed] [Google Scholar]
- 175.Smith BK, York DA, Bray GA. Chronic cerebroventricular galanin does not induce sustained hyperphagia or obesity. Peptides. 1994;15(7 ): 1267–72. doi: 10.1016/0196-9781(94)90152-x. [DOI] [PubMed] [Google Scholar]
- 176.Gundlach AL. Galanin/GALP and galanin receptors role in central control of feeding, body weight/obesity and reproduction? Eur J Pharmacol. 2002;440(2-3 ): 255–68. doi: 10.1016/s0014-2999(02)01433-4. [DOI] [PubMed] [Google Scholar]
- 177.Cummings DE, Purnell JQ, Frayo RS, Schmidova K, Wisse BE, Weigle DS. A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes. 2001;50(8 ): 1714–9. doi: 10.2337/diabetes.50.8.1714. [DOI] [PubMed] [Google Scholar]
- 178.Sun Y, Ahmed S, Smith RG. Deletion of ghrelin impairs neither growth nor appetite. Mol Cell Biol. 2003;23(22 ): 7973–81. doi: 10.1128/MCB.23.22.7973-7981.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Wortley KE, Anderson KD, Garcia K, et al. Genetic deletion of ghrelin does not decrease food intake but influences metabolic fuel preference. Proc Natl Acad Sci USA. 2004;101(21 ): 8227–32. doi: 10.1073/pnas.0402763101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Wortley KE, del Rincon JP, Murray JD, et al. Absence of ghrelin protects against early-onset obesity. J Clin Invest. 2005;115(12 ): 3573–8. doi: 10.1172/JCI26003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Asakawa A, Inui A, Kaga T, et al. Antagonism of ghrelin receptor reduces food intake and body weight gain in mice. Gut. 2003;52(7 ): 947–52. doi: 10.1136/gut.52.7.947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Shuto Y, Shibasaki T, Otagiri A, et al. Hypothalamic growth hormone secretagogue receptor regulates growth hormone secretion, feeding, and adiposity. J Clin Invest. 2002;109(11 ): 1429–36. doi: 10.1172/JCI13300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Esler WP, Rudolph J, Claus TH, et al. Small-molecule ghrelin receptor antagonists improve glucose tolerance, suppress appetite, and promote weight loss. Endocrinology. 2007;148(11 ): 5175–85. doi: 10.1210/en.2007-0239. [DOI] [PubMed] [Google Scholar]
- 184.Salomé N, Hansson C, Taube M, et al. On the central mechanism underlying ghrelin's chronic pro-obesity effects in rats new insights from studies exploiting a potent ghrelin receptor antagonist. J Neuroendocrinol. 2009;21(9 ): 777–85. doi: 10.1111/j.1365-2826.2009.01895.x. [DOI] [PubMed] [Google Scholar]
- 185.Kopelman P, Bryson A, Hickling R, et al. Cetilistat (ATL-962), a novel lipase inhibitor a 12-week randomized, placebo-controlled study of weight reduction in obese patients. Int J Obes (Lond) 2007;31(3 ): 494–9. doi: 10.1038/sj.ijo.0803446. [DOI] [PubMed] [Google Scholar]
- 186.Kopelman P, Groot Gde H, Rissanen A, et al. Weight loss, HbA1c reduction, and tolerability of cetilistat in a randomized, placebo-controlled phase 2 trial in obese diabetics comparison with orlistat (Xenical) Obesity (Silver Spring) 2010;18(1 ): 108–15. doi: 10.1038/oby.2009.155. [DOI] [PubMed] [Google Scholar]
- 187.Padwal R. Cetilistat, a new lipase inhibitor for the treatment of obesity. Curr Opin Investig Drugs. 2008;9(4 ): 414–21. [PubMed] [Google Scholar]
- 188.Grover GJ, Mellstrom K, Malm J. Development of the thyroid hormone receptor beta-subtype agonist KB-141 a strategy for body weight reduction and lipid lowering with minimal cardiac side effects. Cardiovasc Drug Rev. 2005;23(2 ): 133–48. doi: 10.1111/j.1527-3466.2005.tb00161.x. [DOI] [PubMed] [Google Scholar]
- 189.Bryzgalova G, Effendic S, Khan A, et al. Anti-obesity, anti-diabetic, and lipid lowering effects of the thyroid receptor beta subtype selective agonist KB-141. J Steroid Biochem Mol Biol. 2008;111(3-5 ): 262–7. doi: 10.1016/j.jsbmb.2008.06.010. [DOI] [PubMed] [Google Scholar]
- 190.Grover GJ, Egan DM, Sleph PG, et al. Effects of the thyroid hormone receptor agonist GC-1 on metabolic rate and cholesterol in rats and primates selective actions relative to 3,5,3'-triiodo-L-thyronine. Endocrinology. 2004;145(4 ): 1656–61. doi: 10.1210/en.2003-0973. [DOI] [PubMed] [Google Scholar]
- 191.Ng FM, Sun J, Sharma L, Libinaka R, Jiang WJ, Gianello R. Metabolic studies of a synthetic lipolytic domain (AOD9604) of human growth hormone. Horm Res. 2000;53(6 ): 274–8. doi: 10.1159/000053183. [DOI] [PubMed] [Google Scholar]
- 192.Valentino MA, Lin JE, Waldman SA. Central and peripheral molecular targets for antiobesity pharmacotherapy. Clin Pharmacol Ther. 2010;87(6 ): 652–62. doi: 10.1038/clpt.2010.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Arch JR, Ainsworth AT, Cawthorne MA, et al. Atypical beta-adrenoceptor on brown adipocytes as target for anti-obesity drugs. Nature. 1984;309(5964 ): 163–5. doi: 10.1038/309163a0. [DOI] [PubMed] [Google Scholar]
- 194.Emorine L, Blin N, Strosberg AD. The human beta 3-adrenoceptor: the search for a physiological function. Trends Pharmacol Sci. 1994;15(1 ): 3–7. doi: 10.1016/0165-6147(94)90118-x. [DOI] [PubMed] [Google Scholar]
- 195.Weyer C, Gautier JF, Danforth E ., Jr Development of beta 3-adrenoceptor agonists for the treatment of obesity and diabetes--an update. Diabetes Metab. 1999;25(1 ): 11–21. [PubMed] [Google Scholar]
- 196.Ito M, Grujic D, Abel ED, et al. Mice expressing human but not murine beta3-adrenergic receptors under the control of human gene regulatory elements. Diabetes. 1998;47(9 ): 1464–71. doi: 10.2337/diabetes.47.9.1464. [DOI] [PubMed] [Google Scholar]
- 197.Connacher AA, Bennet WM, Jung RT. Clinical studies with the beta-adrenoceptor agonist BRL 26830A. Am J Clin Nutr. 1992;55(Suppl1 ):258S–261S. doi: 10.1093/ajcn/55.1.258s. [DOI] [PubMed] [Google Scholar]
- 198.Wheeldon NM, McDevitt DG, Lipworth BJ. Cardiac effects of the beta 3-adrenoceptor agonist BRL35135 in man. Br J Clin Pharmacol. 1994;37(4 ): 363–9. doi: 10.1111/j.1365-2125.1994.tb04290.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Cypess AM, Lehman S, Williams G, et al. Identification and importance of brown adipose tissue in adult humans. N Engl J Med. 2009;360(15 ): 1509–17. doi: 10.1056/NEJMoa0810780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Weyer C, Tataranni PA, Snitker S, Danforth E, Jr, Ravussin E. Increase in insulin action and fat oxidation after treatment with CL 316,243, a highly selective beta3-adrenoceptor agonist in humans. Diabetes. 1998;47(10 ): 1555–61. doi: 10.2337/diabetes.47.10.1555. [DOI] [PubMed] [Google Scholar]
- 201.Sum FW, Gilbert A, Venkatesan AM, et al. Prodrugs of CL316243: a selective beta3-adrenergic receptor agonist for treating obesity and diabetes. Bioorg Med Chem Lett. 1999;9(14 ): 1921–6. doi: 10.1016/s0960-894x(99)00316-9. [DOI] [PubMed] [Google Scholar]
- 202. Farrigan c, Pang k. Obesity market overview. Nat Rev Drug Discov. 2002;1(4 ): 257–8. doi: 10.1038/nrd781. [DOI] [PubMed] [Google Scholar]
- 203.Klingenberg M, Huang SG. Structure and function of the uncoupling protein from brown adipose tissue. Biochim Biophys Acta. 1999;1415(2 ): 271–96. doi: 10.1016/s0005-2736(98)00232-6. [DOI] [PubMed] [Google Scholar]
- 204.Boss O, Hagen T, Lowell BB. Uncoupling proteins 2 and 3 potential regulators of mitochondrial energy metabolism. Diabetes. 2000;49(2 ): 143–56. doi: 10.2337/diabetes.49.2.143. [DOI] [PubMed] [Google Scholar]
- 205.Argyropoulos G, Brown AM, Willi SM, et al. Effects of mutations in the human uncoupling protein 3 gene on the respiratory quotient and fat oxidation in severe obesity and type 2 diabetes. J Clin Invest. 1998;102(7 ): 1345–51. doi: 10.1172/JCI4115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Clapham JC, Arch JR, Chapman H, et al. Mice overexpressing human uncoupling protein-3 in skeletal muscle are hyperphagic and lean. Nature. 2000;406(6794 ): 415–8. doi: 10.1038/35019082. [DOI] [PubMed] [Google Scholar]
- 207.Matsuda D, Tomoda H. DGAT inhibitors for obesity. Curr Opin Investig Drugs. 2007;8(10 ): 836–41. [PubMed] [Google Scholar]
- 208.Chen HC, Farese RV., Jr DGAT and triglyceride synthesis a new target for obesity treatment? Trends Cardiovasc Med. 2000;10(5 ): 188–92. doi: 10.1016/s1050-1738(00)00066-9. [DOI] [PubMed] [Google Scholar]
- 209.Zhao G, Souers AJ, Voorbach M, et al. Validation of diacylglycerolacyltransferase I as a novel target for the treatment of obesity and dyslipidemia using a potent and selective small molecule inhibitor. J Med Chem. 2008;51(3 ): 380–3. doi: 10.1021/jm7013887. [DOI] [PubMed] [Google Scholar]
- 210.Dobrzyn A, Ntambi JM. Stearoyl-CoA desaturase as a new drug target for obesity treatment. Obes Rev. 2005;6(2 ): 169–74. doi: 10.1111/j.1467-789X.2005.00177.x. [DOI] [PubMed] [Google Scholar]
- 211.Dobrzyn A, Ntambi JM. The role of stearoyl-CoA desaturase in the control of metabolism. Prostaglandins Leukot Essent Fatty Acids. 2005;73(1 ): 35–41. doi: 10.1016/j.plefa.2005.04.011. [DOI] [PubMed] [Google Scholar]
- 212.Tong L. Acetyl-coenzyme A carboxylase crucial metabolic enzyme and attractive target for drug discovery. Cell Mol Life Sci. 2005;62(16 ): 1784–803. doi: 10.1007/s00018-005-5121-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Shetty S, Kusminski CM, Scherer PE. Adiponectin in health and disease evaluation of adiponectin-targeted drug development strategies. Trends Pharmacol Sci. 2009;30(5 ): 234–9. doi: 10.1016/j.tips.2009.02.004. [DOI] [PubMed] [Google Scholar]
- 214.Bredella MA, Torriani M, Ghomi RH, et al. Adiponectin Is Inversely Associated With Intramyocellular and Intrahepatic Lipids in Obese Premenopausal Women. Obesity (Silver Spring) 2011;19(5 ): 911–6. doi: 10.1038/oby.2010.296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Liu YL, Liang HR, Liu HT, et al. Association of serum adiponectin levels with artherosclerosis and the metabolic syndrome in obese children. J Pediatr Endocrinol Metab. 2010;23(8 ): 743–51. doi: 10.1515/jpem.2010.122. [DOI] [PubMed] [Google Scholar]
- 216.Hotta K, Funahashi T, Arita Y, et al. Plasma concentrations of a novel, adipose-specific protein, adiponectin, in type 2 diabetic patients. Arterioscler Thromb Vasc Biol. 2000;20(6 ): 1595–9. doi: 10.1161/01.atv.20.6.1595. [DOI] [PubMed] [Google Scholar]
- 217.Yamauchi T, Kamon J, Waki H, et al. The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat Med. 2000;7(8 ): 941–6. doi: 10.1038/90984. [DOI] [PubMed] [Google Scholar]
- 218.Yamauchi T, Hara K, Kubota N, et al. Dual roles of adiponectin/Acrp30 in vivo as an anti-diabetic and anti-atherogenic adipokine. Curr Drug Targets Immune Endocr Metabol Disord. 2003;3(4 ): 243–54. doi: 10.2174/1568008033340090. [DOI] [PubMed] [Google Scholar]
- 219.Fruebis J, Tsao TS, Javorschi S, et al. Proteolytic cleavage product of 30-kDa adipocyte complement-related protein increases fatty acid oxidation in muscle and causes weight loss in mice. Proc Natl Acad Sci USA. 2001;98(4 ): 2005–10. doi: 10.1073/pnas.041591798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Cheetham SC, Jackson HC, Vickers SP, Dickinson K, Jones RB, Heal DJ. Novel targets for the treatment of obesity: a review of progress. Drug Discovery Today: Therapeutic Strategies. 2004;1(2 ): 227–35. [Google Scholar]
- 221.Bodles AM, Banga A, Rasouli N, Ono F, Kern PA, Owens RJ. Pioglitazone increases secretion of high-molecular-weight adiponectin from adipocytes. Am J Physiol Endocrinol Metab. 2006;291(5 ): E1100–5. doi: 10.1152/ajpendo.00187.2006. [DOI] [PubMed] [Google Scholar]
- 222.Bensaid M, Gary-Bobo M, Esclangon A, et al. The cannabinoid CB1 receptor antagonist SR141716 increases Acrp30 mRNA expression in adipose tissue of obese fa/fa rats and in cultured adipocyte cells. Mol Pharmacol. 2003;63(4 ): 908–14. doi: 10.1124/mol.63.4.908. [DOI] [PubMed] [Google Scholar]
- 223.Yamada S. Pleiotropic Effects of ARB in Metabolic Syndrome. Curr Vasc Pharmacol. 2011;9(2 ): 158–61. doi: 10.2174/157016111794519318. [DOI] [PubMed] [Google Scholar]
- 224.Sukhija R, Kakar P, Mehta V, Mehta JL. Enhanced 11beta-hydroxysteroid dehydrogenase activity, the metabolic syndrome, and systemic hypertension. Am J Cardiol. 2006;98(4 ): 544–8. doi: 10.1016/j.amjcard.2006.03.028. [DOI] [PubMed] [Google Scholar]
- 225.Morton NM, Seckl JR. 11beta-hydroxysteroid dehydrogenase type 1 and obesity. Front Horm Res. 2008;36:146–64. doi: 10.1159/000115363. [DOI] [PubMed] [Google Scholar]
- 226.Muñoz R, Carvajal C, Escalona A, et al. 11beta-hydroxysteroid dehydrogenase type 1 is overexpressed in subcutaneous adipose tissue of morbidly obese patients. Obes Surg. 2009;19(6 ): 764–70. doi: 10.1007/s11695-008-9616-6. [DOI] [PubMed] [Google Scholar]
- 227.Wang SJ, Birtles S, de Schoolmeester J, et al. Inhibition of 11beta-hydroxysteroid dehydrogenase type 1 reduces food intake and weight gain but maintains energy expenditure in diet-induced obese mice. Diabetologia. 2006;49(6 ): 1333–7. doi: 10.1007/s00125-006-0239-y. [DOI] [PubMed] [Google Scholar]
- 228.Tontonoz P, Hu E, Spiegelman BM. Stimulation of adipogenesis in fibroblasts by PPAR gamma 2, a lipid-activated transcription factor. Cell. 1994;79(7 ): 1147–56. doi: 10.1016/0092-8674(94)90006-x. [DOI] [PubMed] [Google Scholar]
- 229.Puigserver P, Wu Z, Park CW, Graves R, Wright M, Spiegelman BM. A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell. 1998;92(6 ): 829–39. doi: 10.1016/s0092-8674(00)81410-5. [DOI] [PubMed] [Google Scholar]
- 230.Handschin C, Spiegelman BM. Peroxisome proliferator-activated receptor gamma coactivator 1 coactivators, energy homeostasis, and metabolism. Endocr Rev. 2006;27(7 ): 728–35. doi: 10.1210/er.2006-0037. [DOI] [PubMed] [Google Scholar]
- 231.Wilson-Fritch L, Nicoloro S, Chouinard M. Mitochondrial remodeling in adipose tissue associated with obesity and treatment with rosiglitazone. J Clin Invest. 2004;114(9 ): 1281–9. doi: 10.1172/JCI21752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Weindruch R, Walford RL, Fligiel S, Guthrie D. The retardation of aging in mice by dietary restriction: longevity, cancer, immunity and lifetime energy intake. J Nutr. 1986;116(4 ): 641–54. doi: 10.1093/jn/116.4.641. [DOI] [PubMed] [Google Scholar]
- 233.Chaudhary N, Pfluger PT. Metabolic benefits from Sirt1 and Sirt1 activators. Curr Opin Clin Nutr Metab Care. 2009;12(4 ): 431–7. doi: 10.1097/MCO.0b013e32832cdaae. [DOI] [PubMed] [Google Scholar]
- 234.Elliott PJ, Jirousek M. Sirtuins novel targets for metabolic disease. Curr Opin Investig Drugs. 2008;9(4 ): 371–8. [PubMed] [Google Scholar]
- 235.Imai s, Armstrong cm, Kaeberlein m, Guarente l. Transcriptional silencing and longevity protein sir2 is an nad-dependent histone deacetylase. Nature. 2000;403(6771 ): 795–800. doi: 10.1038/35001622. [DOI] [PubMed] [Google Scholar]
- 236.Van Meter M, Mao Z, Gorbunova V, Seluanov A. Repairing split ends: SIRT6, mono-ADP ribosylation and DNA repair. Aging (Albany NY) 2011;3(9 ): 829–35. doi: 10.18632/aging.100389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Yamamoto H, Schoonjans K, Auwerx J. Sirtuin functions in health and disease. Mol Endocrinol. 2007;21(8 ): 1745–55. doi: 10.1210/me.2007-0079. [DOI] [PubMed] [Google Scholar]
- 238.Pfluger PT, Herranz D, Velasco-Miguel S, Serrano M, Tschöp MH. Sirt1 protects against high-fat diet-induced metabolic damage. Proc Natl Acad Sci USA. 2008;105(28 ): 9793–8. doi: 10.1073/pnas.0802917105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Yeomans MR, Gray RW. Opioid peptides and control of human ingestivebehaviour. Neurosci Biobehav Rev. 2002;26(6 ): 713–2. doi: 10.1016/s0149-7634(02)00041-6. [DOI] [PubMed] [Google Scholar]
- 240.Greenway FL, Fujioka K, Plodkowski RA, et al. COR-I Study Group. Effect of naltrexone plus bupropion on weight loss in overweight and obese adults (COR-I): a multicentre, randomised, double- blind, placebo-controlled, phase 3 trial. Lancet . 2010;376(9741 ): 595–605. doi: 10.1016/S0140-6736(10)60888-4. [DOI] [PubMed] [Google Scholar]
- 241.Jones D. Novel pharmacotherapies for obesity poised to enter market. Nat Rev Drug Discov. 2009;8(11 ): 833–4. doi: 10.1038/nrd3044. [DOI] [PubMed] [Google Scholar]
- 242.Wadden TA, Foreyt JP, Foster GD, et al. Weight loss with naltrexone SR/bupropion SR combination therapy as an adjunct to behavior modification the COR-BMOD trial. Obesity (Silver Spring) 2011;19(1 ): 110–20. doi: 10.1038/oby.2010.147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Pollack A. FDA Declines to Approve Diet Drug. The New York Times. Published on: 1 February, 2011. Available from: http://www.nytimes.com/2011/02/02/business/02drug.html . [Accessed on: February 26, 2012].
- 244.Gadde KM, Franciscy DM, Wagner HR, 2nd, Krishnan KR. Zonisamide for weight loss in obese adults: a randomized controlled trial. JAMA. 2003;289(14 ): 1820–5. doi: 10.1001/jama.289.14.1820. [DOI] [PubMed] [Google Scholar]
- 245.Supuran CT, Di Fiore A, De Simone G. Carbonic anhydrase inhibitors as emerging drugs for the treatment of obesity. Expert Opin Emerg Drugs. 2008;13(2 ): 383–92. doi: 10.1517/14728214.13.2.383. [DOI] [PubMed] [Google Scholar]
- 246.Gadde KM, Allison DB, Ryan DH, et al. Effects of low-dose, controlled-release, phentermine plus topiramate combination on weight and associated comorbidities in overweight and obese adults (CONQUER): a randomised, placebo-controlled, phase 3 trial. Lancet. 2011;377(9774 ): 1341–52. doi: 10.1016/S0140-6736(11)60205-5. [DOI] [PubMed] [Google Scholar]
- 247.Pollack A. FDA Rejects Qnexa, a Third Weight-Loss Drug. The New York Times. Published on: 28 October, 2010. Available from: http://www.nytimes.com/2010/10/29/health/policy/29drug.html . [Accessed on: September 9, 2011].
- 248.Ravussin E, Smith SR, Mitchell JA, et al. Enhanced weight loss with pramlintide/metreleptin: an integrated neurohormonal approach to obesity pharmacotherapy. Obesity (Silver Spring) 2009;17(9 ): 1736–43. doi: 10.1038/oby.2009.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Tam CS, Lecoultre V, Ravussin E. Novel strategy for the use of leptin for obesity therapy. Expert Opin Biol Ther. 2011;11(12 ): 1677–85. doi: 10.1517/14712598.2011.619974. [DOI] [PMC free article] [PubMed] [Google Scholar]