Rimonabant

Last updated
Rimonabant
Rimonabant.svg
Rimonabant ball-and-stick model.png
Clinical data
AHFS/Drugs.com Consumer Drug Information
License data
Pregnancy
category
  • Not assigned, use not recommended
Routes of
administration
Oral
ATC code
Legal status
Legal status
  • Withdrawn from European and Brazilian markets; not approved elsewhere [1] [2]
Pharmacokinetic data
Bioavailability Undetermined
Protein binding Nearly 100%
Metabolism Hepatic, CYP3A4 involved
Elimination half-life Variable:
6 to 9 days with normal BMI
16 days if BMI >30
Excretion Fecal (86%) and renal (3%)
Identifiers
  • 5-(4-Chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-N-(piperidin-1-yl)-1H-pyrazole-3-carboxamide
CAS Number
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
CompTox Dashboard (EPA)
ECHA InfoCard 100.210.978 OOjs UI icon edit-ltr-progressive.svg
Chemical and physical data
Formula C22H21Cl3N4O
Molar mass 463.79 g·mol−1
3D model (JSmol)
  • O=C(NN1CCCCC1)c4nn(c2ccc(Cl)cc2Cl)c(c3ccc(Cl)cc3)c4C
  • InChI=1S/C22H21Cl3N4O/c1-14-20(22(30)27-28-11-3-2-4-12-28)26-29(19-10-9-17(24)13-18(19)25)21(14)15-5-7-16(23)8-6-15/h5-10,13H,2-4,11-12H2,1H3,(H,27,30) Yes check.svgY
  • Key:JZCPYUJPEARBJL-UHFFFAOYSA-N Yes check.svgY
   (verify)

Rimonabant (also known as SR141716; trade names Acomplia, Zimulti) [3] is an anorectic antiobesity drug approved in Europe in 2006 but was withdrawn worldwide in 2008 due to serious psychiatric side effects; it was never approved in the United States. [1] [2] Rimonabant is an inverse agonist for the cannabinoid receptor CB1 and was first-in-class for clinical development. [4] [5]

Contents

History

Rimonabant is a selective CB1 receptor blocker and was discovered and developed by Sanofi-Aventis. [6]

On 21 June 2006, the European Commission approved the sale of rimonabant in the then-25-member European Union as a prescription drug for use in conjunction with diet and exercise for patients with a body mass index (BMI) greater than 30 kg/m2, or patients with a BMI greater than 27 kg/m2 with associated risk factors, such as type 2 diabetes or dyslipidaemia. [7] It was first in its class to be approved anywhere in the world. [5]

Rimonabant was submitted to the Food and Drug Administration (FDA) for approval in the United States in 2005; in 2007, the FDA's Endocrine and Metabolic Drugs Advisory Committee (EMDAC) concluded that Sanofi-Aventis failed to demonstrate the safety of rimonabant and voted against recommending the anti-obesity treatment for approval. [8] The application was deemed not-approvable by FDA, and the company cancelled plans for a re-submission. [9]

The drug was approved in Brazil in April 2007. [2]

In October 2008, the European Medicines Agency recommended the suspension of Acomplia after the Committee for Medicinal Products for Human Use (CHMP) had determined that the risks of Acomplia outweighed its benefits due to the risk of serious psychiatric problems, including suicide. [10] In November 2008 an advisory committee in Brazil recommended suspension as well, and that month Sanofi-Aventis suspended sale of the drug worldwide. [2] The EMA approval was withdrawn in January 2009. [11] [12] In 2009 India prohibited the manufacture and sale of the drug. [13]

Adverse effects

Data from clinical trials submitted to regulatory authorities showed that rimonabant caused depressive disorders or mood alterations in up to 10% of subjects and suicidal ideation in around 1%, and in Europe it was contraindicated for people with any psychiatric disorder, including depressed or suicidal individuals. [7] Data from a large, randomized, clinical trial (CRESCENDO) with > 9000 patients receiving rimonabant treatment demonstrated a rate of psychiatric adverse events (anxiety, depression, depressed mood, or insomnia) of greater than 30%. [14]

Additionally, nausea and upper respiratory tract infections were very common adverse effects (occurring in more than 10% of people); common adverse effects (occurring in between 1% and 10% of people) included gastroenteritis, anxiety, irritability, insomnia and other sleep disorders, hot flushes, diarrhea, vomiting, dry or itchy skin, tendonitis, muscle cramps and spasms, fatigue, flu-like symptoms, and increased risk of falling.

The FDA's advisory committee concurred with concerns raised by the review divisions. Based on human and on animal data, it appeared that the therapeutic window with regard to CNS toxicity, and specifically seizures was narrow. [2] [15] [16]

EMA postmarketing surveillance data suggested that the risk of psychiatric disorders in people taking rimonabant was doubled. [2]

Pharmacology

Pharmacodynamics

Rimonabant is an inverse agonist of the cannabinoid CB1 receptor. Originally thought to be selective for the CB1 receptor, rimonabant was subsequently also found to act as an antagonist of the μ-opioid receptor. [17]

Chemistry

The chemical synthesis of rimonabant is described as follows: [18] [ failed verification ]

Rimonabant.png

Research

Along with the clinical trials in obesity that generated the data submitted to regulatory authorities, [19] rimonabant was also studied in clinical trials [2] for diabetes, atherosclerosis, and smoking cessation. [20] [21]

See also

Related Research Articles

<span class="mw-page-title-main">AM-251 (drug)</span> Chemical compound

AM-251 is an inverse agonist at the CB1 cannabinoid receptor. AM-251 is structurally very close to rimonabant; both are biarylpyrazole cannabinoid receptor antagonists. In AM-251, the p-chloro group attached to the phenyl substituent at C-5 of the pyrazole ring is replaced with a p-iodo group. The resulting compound exhibits slightly better binding affinity for the CB1 receptor (with a Ki value of 7.5 nM) than rimonabant, which has a Ki value of 11.5 nM, AM-251 is, however, about two-fold more selective for the CB1 receptor when compared to rimonabant. Like rimonabant, it is additionally a μ-opioid receptor antagonist that attenuates analgesic effects.

<span class="mw-page-title-main">Cannabinoid receptor 1</span> Mammalian protein found in Homo sapiens

Cannabinoid receptor 1 (CB1), is a G protein-coupled cannabinoid receptor that in humans is encoded by the CNR1 gene. The human CB1 receptor is expressed in the peripheral nervous system and central nervous system. It is activated by endogenous cannabinoids called endocannabinoids, a group of retrograde neurotransmitters that include lipids, such as anandamide and 2-arachidonoylglycerol (2-AG); plant phytocannabinoids, such as docosatetraenoylethanolamide found in wild daga, the compound THC which is an active constituent of the psychoactive drug cannabis; and synthetic analogs of THC. CB1 is antagonized by the phytocannabinoid tetrahydrocannabivarin (THCV).

<span class="mw-page-title-main">Taranabant</span> Chemical compound

Taranabant (codenamed MK-0364) is a cannabinoid receptor type 1 (CB1) inverse agonist that was investigated as a potential treatment for obesity due to its anorectic effects. It was discovered by Merck & Co.

<span class="mw-page-title-main">NESS-0327</span> Chemical compound

NESS-0327 is a drug used in scientific research which acts as an extremely potent and selective antagonist of the cannabinoid receptor CB1. It is much more potent an antagonist, and more selective for the CB1 receptor over CB2, than the more commonly used ligand rimonabant, with a Ki at CB1 of 350fM (i.e. 0.00035nM) and a selectivity of over 60,000x for CB1 over CB2. Independently, two other groups have described only modest nanomolar CB1 affinity for this compound (125nM and 18.4nM). Also unlike rimonabant, NESS-0327 does not appear to act as an inverse agonist at higher doses, instead being a purely neutral antagonist which blocks the CB1 receptor but does not produce any physiological effect of its own.

<span class="mw-page-title-main">Surinabant</span> Chemical compound

Surinabant (SR147778) is a cannabinoid receptor type 1 antagonist developed by Sanofi-Aventis. It is being investigated as a potential treatment for nicotine addiction, to assist smoking cessation. It may also be developed as an anorectic drug to assist with weight loss, however there are already several CB1 antagonists or inverse agonists on the market or under development for this application, so surinabant is at present mainly being developed as an anti-smoking drug, with possible application in the treatment of other addictive disorders such as alcoholism. Other potential applications such as treatment of ADHD have also been proposed.

<span class="mw-page-title-main">VCHSR</span> Chemical compound

VCHSR is a drug used in scientific research which acts as a selective antagonist of the cannabinoid receptor CB1. It is derived from the widely used CB1 antagonist rimonabant, and has similar potency and selectivity for the CB1 receptor, but has been modified to remove the hydrogen bonding capability in the C-3 substituent region, which removes the inverse agonist effect that rimonabant produces at high doses, so that VCHSR instead acts as a neutral antagonist, blocking the receptor but producing no physiological effect of its own.

A cannabinoid receptor antagonist, also known simply as a cannabinoid antagonist or as an anticannabinoid, is a type of cannabinoidergic drug that binds to cannabinoid receptors (CBR) and prevents their activation by endocannabinoids. They include antagonists, inverse agonists, and antibodies of CBRs. The discovery of the endocannabinoid system led to the development of CB1 receptor antagonists. The first CBR inverse agonist, rimonabant, was described in 1994. Rimonabant blocks the CB1 receptor selectively and has been shown to decrease food intake and regulate body-weight gain. The prevalence of obesity worldwide is increasing dramatically and has a great impact on public health. The lack of efficient and well-tolerated drugs to cure obesity has led to an increased interest in research and development of CBR antagonists. Cannabidiol (CBD), a naturally occurring cannabinoid and a non-competitive CB1/CB2 receptor antagonist, as well as Δ9-tetrahydrocannabivarin (THCV), a naturally occurring cannabinoid, modulate the effects of THC via direct blockade of cannabinoid CB1 receptors, thus behaving like first-generation CB1 receptor inverse agonists, such as rimonabant. CBD is a very low-affinity CB1 ligand, that can nevertheless affect CB1 receptor activity in vivo in an indirect manner, while THCV is a high-affinity CB1 receptor ligand and potent antagonist in vitro and yet only occasionally produces effects in vivo resulting from CB1 receptor antagonism. THCV has also high affinity for CB2 receptors and signals as a partial agonist, differing from both CBD and rimonabant.

Pitolisant, sold under the brand name Wakix among others, is a medication used for the treatment of excessive daytime sleepiness in adults with narcolepsy. It is a histamine 3 (H3) receptor antagonist/inverse agonist (an antihistamine drug specific to that kind of receptors). It represents the first commercially available medication in its class, so that the US Food and Drug Administration (FDA) declares it a first-in-class medication. Pitolisant enhances the activity of histaminergic neurons in the brain that function to improve a person's wakefulness.

<span class="mw-page-title-main">Ibipinabant</span> Chemical compound

Ibipinabant (SLV319, BMS-646,256) is a drug used in scientific research which acts as a potent and highly selective CB1 antagonist. It has potent anorectic effects in animals, and was researched for the treatment of obesity, although CB1 antagonists as a class have now fallen out of favour as potential anorectics following the problems seen with rimonabant, and so ibipinabant is now only used for laboratory research, especially structure-activity relationship studies into novel CB1 antagonists. SLV330, which is a structural analogue of Ibipinabant, was reported active in animal models related to the regulation of memory, cognition, as well as in addictive behavior. An atom-efficient synthesis of ibipinabant has been reported.

<span class="mw-page-title-main">Pimavanserin</span> Atypical antipsychotic medication

Pimavanserin, sold under the brand name Nuplazid, is an atypical antipsychotic which is approved for the treatment of Parkinson's disease psychosis and is also being studied for the treatment of Alzheimer's disease psychosis, schizophrenia, agitation, and major depressive disorder. Unlike other antipsychotics, pimavanserin is not a dopamine receptor antagonist.

<span class="mw-page-title-main">Org 27569</span> Chemical compound

Org 27569 is a drug which acts as a potent and selective negative allosteric modulator of the cannabinoid CB1 receptor. Studies in vitro suggest that it binds to a regulatory site on the CB1 receptor target, causing a conformational change that increases the binding affinity of CB1 agonists such as CP 55,940, while decreasing the binding affinity of CB1 antagonists or inverse agonists such as rimonabant. However while Org 27569 increases the ability of CB1 agonists to bind to the receptor, it decreases their efficacy at stimulating second messenger signalling once bound, and so in practice behaves as an insurmountable antagonist of CB1 receptor function.

<span class="mw-page-title-main">TM-38837</span> Chemical compound

TM-38837 is a small molecule inverse agonist/antagonist of the CB1 cannabinoid receptor, with peripheral selectivity. It is being developed for the treatment of obesity and metabolic disorders by 7TM Pharma. The company has announced phase I clinical trials.

<span class="mw-page-title-main">SR-144,528</span> Chemical compound

SR144528 is a drug that acts as a potent and highly selective CB2 receptor inverse agonist, with a Ki of 0.6 nM at CB2 and 400 nM at the related CB1 receptor. It is used in scientific research for investigating the function of the CB2 receptor, as well as for studying the effects of CB1 receptors in isolation, as few CB1 agonists that do not also show significant activity as CB2 agonists are available. It has also been found to be an inhibitor of sterol O-acyltransferase, an effect that appears to be independent from its action on CB2 receptors.

<span class="mw-page-title-main">O-1269</span> Chemical compound

O-1269 is a drug that is a diarylpyrazole derivative, related to potent cannabinoid antagonist drugs such as rimonabant and surinabant. However O-1269 and several related drugs were unexpectedly found to act as full or partial agonists at the cannabinoid receptors rather than antagonists, and so produce the usual effects expected of cannabinoid agonists in animal tests, such as sedation and analgesic effects. The N-heptyl homolog O-1270 and the N-propyl homolog O-1399 also act as cannabinoid agonists with similar potency in vivo, despite weaker binding affinity at cannabinoid receptors compared to the pentyl homolog O-1269. Agonist-like and atypical cannabinoid activity has also been observed with a number of related compounds.

<span class="mw-page-title-main">O-2050</span> Chemical compound

O-2050 is a drug that is a classical cannabinoid derivative, which acts as an antagonist for the CB1 receptor. This gives it an advantage in research over many commonly used cannabinoid antagonists, such as rimonabant, which at higher doses act as inverse agonists at CB1 as well as showing off-target effects. However, while O-2050 acts as a silent antagonist in vitro, some tests in vivo have suggested it may show agonist activity under certain circumstances.

<span class="mw-page-title-main">AM-6545</span> Chemical compound

AM-6545 is a drug which acts as a peripherally selective silent antagonist for the CB1 receptor, and was developed for the treatment of obesity. Other cannabinoid antagonists such as rimonabant have been marketed for this application, but have subsequently been withdrawn from sale because of centrally mediated side effects such as depression and nausea. Because AM-6545 does not cross the blood–brain barrier to any significant extent, it does not produce these kinds of side effects, but has still been shown to effectively reduce appetite and food consumption in animal studies.

<span class="mw-page-title-main">Rosonabant</span> Chemical compound

Rosonabant (INN; E-6776) is a drug acting as a CB1 receptor antagonist/inverse agonist that was under investigation by Esteve as an appetite suppressant for the treatment of obesity. Development of the drug for clinical use was apparently halted shortly after the related CB1 antagonist rimonabant was discontinued in November 2008, due to the reports of severe psychiatric adverse effects such as anxiety, depression, and suicidal ideation associated with it and with similarly acting agents.

<span class="mw-page-title-main">Drinabant</span> Chemical compound

Drinabant (INN; AVE-1625) is a drug that acts as a selective CB1 receptor antagonist, which was under investigation varyingly by Sanofi-Aventis as a treatment for obesity, schizophrenia, Alzheimer's disease, Parkinson's disease, and nicotine dependence. Though initially studied as a potential treatment for a variety of different medical conditions, Sanofi-Aventis eventually narrowed down the therapeutic indications of the compound to just appetite suppression. Drinabant reached phase IIb clinical trials for this purpose in the treatment of obesity but was shortly thereafter discontinued, likely due to the observation of severe psychiatric side effects including anxiety, depression, and thoughts of suicide in patients treated with the now-withdrawn rimonabant, another CB1 antagonist that was also under development by Sanofi-Aventis.

<span class="mw-page-title-main">Samidorphan</span> Opioid antagonist

Samidorphan is an opioid antagonist that in the form of olanzapine/samidorphan is used in the treatment of schizophrenia and bipolar disorder. Samidorphan reduces the weight gain associated with olanzapine. Samidorphan is taken by mouth.

<span class="mw-page-title-main">JD5037</span> Chemical compound

JD5037 is an antiobesity drug candidate which acts as a peripherally-restricted cannabinoid inverse agonist at CB1 receptors. It is very selective for the CB1 subtype, with a Ki of 0.35nM, >700-fold higher affinity than it has for CB2 receptors.

References

  1. 1 2 Sam AH, Salem V, Ghatei MA (2011). "Rimonabant: From RIO to Ban". Journal of Obesity. 2011: 432607. doi: 10.1155/2011/432607 . PMC   3136184 . PMID   21773005.
  2. 1 2 3 4 5 6 7 Moreira FA, Crippa JA (June 2009). "The psychiatric side-effects of rimonabant". Revista Brasileira de Psiquiatria. 31 (2): 145–153. doi: 10.1590/s1516-44462009000200012 . PMID   19578688.
  3. "Rimonabant". AdisInsight. Retrieved 21 February 2017.
  4. Fong TM, Heymsfield SB (September 2009). "Cannabinoid-1 receptor inverse agonists: current understanding of mechanism of action and unanswered questions". International Journal of Obesity. 33 (9): 947–955. doi: 10.1038/ijo.2009.132 . PMID   19597516.
  5. 1 2 "European Approval Comes Early for Sanofi-Aventis' Acomplia". IHS. June 23, 2006.
  6. Barth F, Rinaldi-Carmona M (August 1999). "The development of cannabinoid antagonists". Current Medicinal Chemistry. 6 (8): 745–755. doi:10.2174/0929867306666220401143808. PMID   10469889. S2CID   247893317.
  7. 1 2 "Acomplia EPAR" (PDF). EMA. January 30, 2009. Archived from the original (PDF) on February 22, 2017. Retrieved February 21, 2017. From EMA index page Archived 2017-02-22 at the Wayback Machine
  8. Saul S (14 June 2007). "F.D.A. Panel Rejects Drug for Obesity". The New York Times.
  9. "Sanofi-Aventis Drops Application for Drug". The New York Times. 30 June 2007.
  10. "The European Medicines Agency recommends suspension of the marketing authorisation of Acomplia". European Medicines Agency. 23 October 2008. Retrieved 18 January 2016.
  11. "Anti-obesity drug use suspended". BBC News. 23 October 2008. Retrieved 4 March 2010.
  12. "Public Statement on Acomplia (rimonabant) Withdrawal of the Marketing Authorisation in the European Union" (PDF). European Medicines Agency. 30 January 2009. Retrieved 18 January 2016.
  13. "Drugs banned in India". Central Drugs Standard Control Organization, Dte.GHS, Ministry of Health and Family Welfare, Government of India. Archived from the original on 2017-02-22. Retrieved 2013-09-17.
  14. Topol EJ, Bousser MG, Fox KA, Creager MA, Despres JP, Easton JD, et al. (August 2010). "Rimonabant for prevention of cardiovascular events (CRESCENDO): a randomised, multicentre, placebo-controlled trial". Lancet. 376 (9740): 517–523. doi:10.1016/S0140-6736(10)60935-X. PMID   20709233. S2CID   36404292.
  15. "FDA Briefing Document NDA 21-888 Zimulti (rimonabant) Tablets, 20" (PDF). FDA. June 13, 2007.
  16. Davis-Bruno K (June 13, 2007). "Nonclinical Overview: CNS Toxicity with Rimonabant". FDA, Division of Metabolism & Endocrinology Products.
  17. Seely KA, Brents LK, Franks LN, Rajasekaran M, Zimmerman SM, Fantegrossi WE, Prather PL (October 2012). "AM-251 and rimonabant act as direct antagonists at mu-opioid receptors: implications for opioid/cannabinoid interaction studies". Neuropharmacology. 63 (5): 905–915. doi:10.1016/j.neuropharm.2012.06.046. PMC   3408547 . PMID   22771770.
  18. Yoshioka T, Fujita T, Kanai T, Aizawa Y, Kurumada T, Hasegawa K, Horikoshi H (February 1989). "Studies on hindered phenols and analogues. 1. Hypolipidemic and hypoglycemic agents with ability to inhibit lipid peroxidation". Journal of Medicinal Chemistry. 32 (2): 421–428. doi:10.1021/jm00122a022. PMID   2913302.
  19. Pi-Sunyer FX, Aronne LJ, Heshmati HM, Devin J, Rosenstock J (February 2006). "Effect of rimonabant, a cannabinoid-1 receptor blocker, on weight and cardiometabolic risk factors in overweight or obese patients: RIO-North America: a randomized controlled trial". JAMA. 295 (7): 761–775. doi: 10.1001/jama.295.7.761 . PMID   16478899.
  20. Cahill K, Ussher MH (March 2011). "Cannabinoid type 1 receptor antagonists for smoking cessation". The Cochrane Database of Systematic Reviews. 2011 (3): CD005353. doi:10.1002/14651858.CD005353.pub4. PMC   6486173 . PMID   21412887.
  21. Maldonado R, Valverde O, Berrendero F (April 2006). "Involvement of the endocannabinoid system in drug addiction". Trends in Neurosciences. 29 (4): 225–232. doi:10.1016/j.tins.2006.01.008. PMID   16483675. S2CID   16125335.