Epibatidine

Last updated
Epibatidine
(+)-Epibatidine Structural Formulae V.1.svg
Epibatidine-based-on-xtal-3D-bs-17.png
Identifiers
  • (1R,2R,4S)-(+)-6-(6-Chloro-3-pyridyl)-7-azabicyclo[2.2.1]heptane
CAS Number
PubChem CID
DrugBank
ChemSpider
UNII
KEGG
ChEBI
ChEMBL
ECHA InfoCard 100.162.281 OOjs UI icon edit-ltr-progressive.svg
Chemical and physical data
Formula C11H13ClN2
Molar mass 208.69 g·mol−1
3D model (JSmol)
Density 1.2 ± 0.1 g/cm3
  • ClC1=CC=C([C@@H]2C[C@]3([H])CC[C@@]2([H])N3)C=N1
  • InChI=1S/C11H13ClN2/c12-11-4-1-7(6-13-11)9-5-8-2-3-10(9)14-8/h1,4,6,8-10,14H,2-3,5H2/t8-,9+,10+/m0/s1 Yes check.svgY
  • Key:NLPRAJRHRHZCQQ-IVZWLZJFSA-N Yes check.svgY
 X mark.svgNYes check.svgY  (what is this?)    (verify)

Epibatidine is a chlorinated alkaloid that is secreted by the Ecuadoran frog Epipedobates anthonyi and poison dart frogs from the Ameerega genus. [1] It was discovered by John W. Daly in 1974, but its structure was not fully elucidated until 1992. Whether epibatidine is the first observed example of a chlorinated alkaloid remains controversial, due to challenges in conclusively identifying the compound from the limited samples collected by Daly. By the time that high-resolution spectrometry was used in 1991, there remained less than one milligram of extract from Daly's samples, raising concerns about possible contamination. Samples from other batches of the same species of frog failed to yield epibatidine. [2]

Contents

Epibatidine is toxic. Its toxicity stems from its ability to interact with nicotinic and muscarinic acetylcholine receptors. These receptors are involved in the transmission of painful sensations, and in movement, among other functions. Epibatidine then causes numbness, and, eventually, paralysis. Doses are lethal when the paralysis causes respiratory arrest. Originally, it was thought that epibatidine could be useful as a drug. However, because of its unacceptable therapeutic index, it is no longer being researched for potential therapeutic uses. [3]

History

Epibatidine was discovered by John W. Daly in 1974. It was isolated from the skin of Epipedobates anthonyi frogs collected by Daly and colleague, Charles Myers. Between 1974 and 1979, Daly and Myers collected the skins of nearly 3000 frogs from various sites in Ecuador, after finding that a small injection of a preparation from their skin caused analgesic (painkilling) effects in mice that resembled those of an opioid. [2] Despite its common name - Anthony's Poison Arrow frog - suggesting that it was used by natives when hunting, [4] a paper written by Daly in 2000 claimed that there was no local folklore or folk medicine surrounding the frogs and that they were considered largely unimportant by the locals. [5]

Epipedobates tricolor on a leaf Epipedobates tricolor1.jpg
Epipedobates tricolor on a leaf

The structure of epibatidine was elucidated in 1992, an effort hindered by E. anthonyi gaining IUCN protected status in 1984. [5] Furthermore, these frogs do not produce the toxin when bred and reared in captivity, because they do not synthesize epibatidine themselves. Like other poison dart frogs, they instead obtain it through their diet and then sequester it on their skin. Likely dietary sources are beetles, ants, mites, and flies. [6] Daly and Charles noticed that epibatidine was produced from their diet due to their return trip to Ecuador in 1976 when they found that at one site, none of the frogs present produced alkaloids, such as epibatidine; they discovered that only the frogs at certain sites with the dietary means allowed these frogs to produce epibatidine. [7] Overcoming the difficulties, the structure was eventually determined, and the first synthesis of epibatidine was completed in 1993. Many other synthesis methods have been developed since. [5]

Because of its analgesic effect, there was intense interest in epibatidine's use as a drug, because it was found not to be an opioid. [2] This meant that it could potentially be used without fear of addiction. However, it was soon found that it cannot be used in humans because the dose resulting in toxic symptoms is too low for it to be safe. [8]

Synthesis

Several total synthesis routes have been devised due to the relative scarcity of epibatidine in nature. [9]

After the discovery of the structure of epibatidine, more than fifty ways to synthesize it in the laboratory have been devised. In the first reported example, a nine-step procedure produces the substance as a racemate (in contrast, the naturally occurring compound is the (+)-enantiomer; the (−)-enantiomer does not occur naturally). It was later determined that the (+) and (-) enantiomers had equivalent analgesic as well as toxic effects. The process has proven to be quite productive, with a yield of about 40%. [10] [11] [12]

An enantioselective synthesis reported by E J Corey starting from chloronicotinaldehyde is outlined below:

The chemical synthesis of epibatidine by Corey Epibatidine Corey.svg
The chemical synthesis of epibatidine by Corey

In addition to Corey's method, other notable methods include those of Broka, [13] Huang and Shen, [14] and Clayton and Regan. [11]

Synthetic analogs

A number of approaches to discovering structural analogs of epibatine that maintain analgesics effects, but without the toxicity, have been attempted. [15] For example, Abbott Laboratories has produced derivatives of epibatidine including ABT-594. [16] ABT-594 retains analgesic properties while avoiding paralysis by still binding to receptors that control pain perception and having a low affinity for muscle-type nicotinic acetylcholine receptors (nAChR) reducing its paralysis effect. [17] Other epibatidine analogs include ABT-418, epiboxidine and their derivatives. [15] [18] [19] [20] [21] A synthesis of epibatidine, utilizing a microbial hydroxylation of an unactivated carbon in a 7-azanorbornane was published in 1999. [22]

Chemical structure

Epibatidine is a piperidine pyridine with a structure similar to that of nicotine. [23] It is a hygroscopic oily substance which is a base.

Biological effects

Mechanism of action

Epibatidine has two mechanisms of action: it can bind to either nicotinic acetylcholine receptors (nAChR) or muscarinic acetylcholine receptors (mAChR). Specifically, the analgesic property of epibatidine is believed to take place by its binding to the α4/β2 subtype of nicotinic receptors. Epibatidine also binds to the α3/β4 subtype and to a much lesser extent α7 receptors (affinity 300-fold less than for α4/β2) [24] The rank order of affinities for the muscle nicotinic receptors is αε > αγ > αδ. [25]

Nicotinic acetylcholine receptors are found in the post-synaptic membranes of nerve cells. These receptors are an example of ion gated channels where binding by a ligand causes a conformational change allowing ions to cross the membrane into the cell. [26] They propagate neurotransmission in the central and peripheral nervous system. When neurotransmitters bind to these receptors, ion channels open, allowing Na+ and Ca2+ ions to move across the membrane. This depolarizes the post-synaptic membrane, inducing an action potential that propagates the signal. This signal will ultimately induce release of dopamine and norepinephrine, resulting in an antinociceptive effect on the organism. The usual neurotransmitter for nAChR is acetylcholine. However, other substances (such as epibatidine and nicotine) are also able to bind to the receptor and induce a similar, if not identical, response. Epibatidine has an extremely high affinity for nAChRs, depending on the receptor subtype, from 0.05 nM at the α4β2 subtype to 22 nM at the α7 subtype. Affinity as well as efficacy (and thus also potency) are much higher than for nicotine. [10]

The paralytic property of epibatidine takes place after its binding to muscle-type nicotinic receptors.

Low doses of epibatidine will only affect the nAChRs, due to a higher affinity to nAChRs than to mAChRs. Higher doses, however, will cause epibatidine to bind to the mAChRs.

Both (+)- and (-)-enantiomers of epibatidine are biologically active, and both have similar binding affinities to nAChRs [10] Only the (+)-enantiomer does not induce tolerance. While this may be a potential therapeutic advantage over morphine, epibatidine has not entered clinical trials because even very small doses are lethal to rodents. [27]

Symptoms

Epibatidine has several toxic consequences. Empirically proven effects include splanchnic sympathetic nerve discharge and increased arterial pressure. [23] The nerve discharge effects can cause antinociception partially mediated by agonism of central nicotinic acetylcholine receptors at low doses of epibatidine; 5 μg/kg. [28] At higher doses, however, epibatidine will cause paralysis and loss of consciousness, coma and eventually death. The median lethal dose (LD50) of epibatidine lies between 1.46 μg/kg and 13.98 μg/kg. [29] This makes epibatidine somewhat more toxic than dioxin (with an average LD50 of 22.8 μg/kg).[ citation needed ] Due to the small difference between its toxic concentration and antinociceptive concentration, its therapeutic uses are very limited.

In research on mice, administration of doses greater than 5 μg/kg of epibatidine caused a dose-dependent paralyzing effect on the organism. With doses over 5 μg/kg, symptoms included hypertension (increased blood pressure), paralysis in the respiratory system, seizures, and, ultimately, death. The symptoms do, however, change drastically when lower doses are given. Mice became resistant to pain and heat with none of the negative effects of higher doses.

Pharmacology

Epibatidine most effectively enters the body through injection. [30] In vitro studies seem to suggest that epibatidine is hardly, if at all, metabolized in the human body. [31]

Also there is currently little information on the path of clearance from the body. Maximum concentration in the brain is reached at about 30 minutes after entering the body. [10]

Potential medical uses

Epibatidine has a high analgesic potency, as stated above. Studies show it has a potency at least 200 times that of morphine. [10] As the compound was not addictive nor did it cause habituation,[ citation needed ], it was initially thought to be very promising to replace morphine as a painkiller. However, the therapeutic concentration is very close to the toxic concentration. This means that even at a therapeutic dose (5 μg/kg [28] ), some epibatidine might bind to the muscarinic acetylcholine receptors and cause adverse effects, such as hypertension, bradycardia and muscular paresis. [23]

Compared to the gold standard in pain management, morphine, epibatidine needed only 2.5 μg/kg (11.98 nmol/kg) to initiate a pain-relieving effect whilst the same effect required approximately 10 mg/kg (35.05 μmol/kg) of morphine (approx. 2,900 times the efficacy.) Currently, only rudimentary research into epibatidine's effects has yet been performed; the drug has been administered only to rodents for analysis at this time. [12]

Antidote

The antidote to epibatidine is mecamylamine, [32] a nicotinic acetylcholine receptor antagonist that is non-selective and non-competitive. [33] Both the (+) and the (-) enantiomers of mecamylamine were seen to be efficient and both have the same affinity for nicotinic acetylcholine receptors. [34]

See also

Related Research Articles

<span class="mw-page-title-main">Nicotinic acetylcholine receptor</span> Acetylcholine receptors named for their selective binding of nicotine

Nicotinic acetylcholine receptors, or nAChRs, are receptor polypeptides that respond to the neurotransmitter acetylcholine. Nicotinic receptors also respond to drugs such as the agonist nicotine. They are found in the central and peripheral nervous system, muscle, and many other tissues of many organisms. At the neuromuscular junction they are the primary receptor in muscle for motor nerve-muscle communication that controls muscle contraction. In the peripheral nervous system: (1) they transmit outgoing signals from the presynaptic to the postsynaptic cells within the sympathetic and parasympathetic nervous system, and (2) they are the receptors found on skeletal muscle that receive acetylcholine released to signal for muscular contraction. In the immune system, nAChRs regulate inflammatory processes and signal through distinct intracellular pathways. In insects, the cholinergic system is limited to the central nervous system.

<span class="mw-page-title-main">Mecamylamine</span> Antihypertensive drug

Mecamylamine is a non-selective, non-competitive antagonist of the nicotinic acetylcholine receptors (nAChRs) that was introduced in the 1950s as an antihypertensive drug. In the United States, it was voluntarily withdrawn from the market in 2009 but was brought to market in 2013 as Vecamyl and eventually was marketed by Turing Pharmaceuticals.

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

Cytisine, also known as baptitoxine, cytisinicline, or sophorine, is an alkaloid that occurs naturally in several plant genera, such as Laburnum and Cytisus of the family Fabaceae. It has been used medically to help with smoking cessation. Although widely used for smoking cessation in Eastern Europe, cytisine remains relatively unknown beyond it. However, it has been found effective in several randomized clinical trials, including some in the United States and a large one in New Zealand, and is being investigated in additional trials in the United States and a non-inferiority trial in Australia in which it is being compared head-to-head with the smoking cessation aid varenicline. It has also been used entheogenically via mescalbeans by some Native American groups, historically in the Rio Grande Valley predating even peyote.

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

Anabasine is a pyridine and piperidine alkaloid found in the Tree Tobacco plant, as well as in the close relative of the common tobacco plant. It is a structural isomer of, and chemically similar to, nicotine. Its principal (historical) industrial use is as an insecticide.

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

Methyllycaconitine (MLA) is a diterpenoid alkaloid found in many species of Delphinium (larkspurs). In common with many other diterpenoid alkaloids, it is toxic to animals, although the acute toxicity varies with species. Early research was focused on identifying, and characterizing the properties of methyllycaconitine as one of the principal toxins in larkspurs responsible for livestock poisoning in the mountain rangelands of North America. Methyllycaconitine has been explored as a possible therapeutic agent for the treatment of spastic paralysis, and it has been shown to have insecticidal properties. Most recently, it has become an important molecular probe for studying the pharmacology of the nicotinic acetylcholine receptor.

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

Anatoxin-a, also known as Very Fast Death Factor (VFDF), is a secondary, bicyclic amine alkaloid and cyanotoxin with acute neurotoxicity. It was first discovered in the early 1960s in Canada, and was isolated in 1972. The toxin is produced by multiple genera of cyanobacteria and has been reported in North America, South America, Central America, Europe, Africa, Asia, and Oceania. Symptoms of anatoxin-a toxicity include loss of coordination, muscular fasciculations, convulsions and death by respiratory paralysis. Its mode of action is through the nicotinic acetylcholine receptor (nAchR) where it mimics the binding of the receptor's natural ligand, acetylcholine. As such, anatoxin-a has been used for medicinal purposes to investigate diseases characterized by low acetylcholine levels. Due to its high toxicity and potential presence in drinking water, anatoxin-a poses a threat to animals, including humans. While methods for detection and water treatment exist, scientists have called for more research to improve reliability and efficacy. Anatoxin-a is not to be confused with guanitoxin, another potent cyanotoxin that has a similar mechanism of action to that of anatoxin-a and is produced by many of the same cyanobacteria genera, but is structurally unrelated.

<span class="mw-page-title-main">Anthony's poison arrow frog</span> Species of amphibian

Anthony's poison arrow frog is a species of poison dart frog in the family Dendrobatidae. The species is endemic to Ecuador and Peru.

A nicotinic agonist is a drug that mimics the action of acetylcholine (ACh) at nicotinic acetylcholine receptors (nAChRs). The nAChR is named for its affinity for nicotine.

<span class="mw-page-title-main">Pumiliotoxin 251D</span> Chemical compound

Pumiliotoxin 251D is a toxic organic compound. It is found in the skin of poison frogs from the genera Dendrobates, Epipedobates, Minyobates, and Phyllobates and toads from the genus Melanophryniscus. Its name comes from the pumiliotoxin family (PTXs) and its molecular mass of 251 daltons. When the toxin enters the bloodstream through cuts in the skin or by ingestion, it can cause hyperactivity, convulsions, cardiac arrest and ultimately death. It is especially toxic to arthropods, even at low concentrations.

The alpha-4 beta-2 nicotinic receptor, also known as the α4β2 receptor, is a type of nicotinic acetylcholine receptor implicated in learning, consisting of α4 and β2 subunits. It is located in the brain, where activation yields post- and presynaptic excitation, mainly by increased Na+ and K+ permeability.

<span class="mw-page-title-main">Alpha-7 nicotinic receptor</span>

The alpha-7 nicotinic receptor, also known as the α7 receptor, is a type of nicotinic acetylcholine receptor implicated in long-term memory, consisting entirely of α7 subunits. As with other nicotinic acetylcholine receptors, functional α7 receptors are pentameric [i.e., (α7)5 stoichiometry].

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

Tebanicline is a potent synthetic nicotinic (non-opioid) analgesic drug developed by Abbott. It was developed as a less toxic analog of the potent poison dart frog-derived compound epibatidine, which is about 200 times stronger than morphine as an analgesic, but produces extremely dangerous toxic side effects. Like epibatidine, tebanicline showed potent analgesic activity against neuropathic pain in both animal and human trials, but with far less toxicity than its parent compound. It acts as a partial agonist at neuronal nicotinic acetylcholine receptors, binding to both the α3β4 and the α4β2 subtypes.

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

Epiboxidine is a chemical compound which acts as a partial agonist at neural nicotinic acetylcholine receptors, binding to both the α3β4 and the α4β2 subtypes. It was developed as a less toxic analogue of the potent frog-derived alkaloid epibatidine, which is around 200 times stronger than morphine as an analgesic but produces extremely dangerous toxic nicotinic side effects.

<span class="mw-page-title-main">ABT-418</span> Chemical compound

ABT-418 is a drug developed by Abbott, that has nootropic, neuroprotective and anxiolytic effects, and has been researched for treatment of both Alzheimer's disease and ADHD. It acts as an agonist at neural nicotinic acetylcholine receptors, subtype-selective binding with high affinity to the α4β2, α7/5-HT3, and α2β2 nicotinic acetylcholine receptors but not α3β4 receptors ABT-418 was reasonably effective for both applications and fairly well tolerated, but produced some side effects, principally nausea, and it is unclear whether ABT-418 itself will proceed to clinical development or if another similar drug will be used instead.

<span class="mw-page-title-main">Histrionicotoxins</span>

Histrionicotoxins are a group of related toxins found in the skin of poison frogs from the family Dendrobatidae, notably Oophaga histrionica, which are native to Colombia. It is likely that, as with other poison frog alkaloids, histrionicotoxins are not manufactured by the amphibians, but absorbed from insects in their diet and stored in glands in their skin. They are notably less toxic than other alkaloids found in poison frogs, yet their distinct structure acts as a neurotoxin by non-competitive inhibition of nicotinic acetylcholine receptors.

<span class="mw-page-title-main">A-84,543</span> Chemical compound

A-84543 is a drug developed by Abbott, which acts as an agonist at neural nicotinic acetylcholine receptors with high selectivity for the α4β2 subtype. It is widely used in scientific research into the structure and function of this receptor subtype and has been the lead compound for the development of a large family of related derivatives.

The alpha-3 beta-4 nicotinic receptor, also known as the α3β4 receptor and the ganglion-type nicotinic receptor, is a type of nicotinic acetylcholine receptor, consisting of α3 and β4 subunits. It is located in the autonomic ganglia and adrenal medulla, where activation yields post- and/or presynaptic excitation, mainly by increased Na+ and K+ permeability.

<span class="mw-page-title-main">RJR-2429</span> Chemical compound

RJR-2429 is a drug that acts as an agonist at neural nicotinic acetylcholine receptors, binding to both the α3β4 and the α4β2 subtypes. RJR-2429 is stronger than nicotine but weaker than epibatidine in most assays, and with high affinity for both α3β4 and α4β2 subtypes, as well as the less studied α1βγδ subtype.

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

Phantasmidine is a toxic substance derived from the Ecuadorian poisonous frog Anthony's poison arrow frog, more commonly known as the “phantasmal poison frog”. It is a nicotinic agonist, meaning it binds to nicotinic receptors in the body and mimics the effects of the neurotransmitter acetylcholine. This causes the stimulation of the body's parasympathetic nervous system, which induces many inhibitory behaviors in the body such as decreased heart rate.

<span class="mw-page-title-main">3-Bromocytisine</span> Chemical compound

3-Bromocytisine is a derivative of the toxic alkaloid cytisine that acts as a highly potent agonist at neural nicotinic acetylcholine receptors, binding primarily to the α4β2 and α7 subtypes. 3-Bromocytisine is a full agonist at the α7 subtype while it is only a partial agonist at α4β2, but has an extremely strong binding affinity at α4β2 with 200-fold selectivity for α4β2 over α7. In animal studies 3-bromocytisine stimulates the release of dopamine and noradrenaline and increases locomotor activity.

References

  1. Fitch RW, Spande TF, Garraffo HM, Yeh HJ, Daly JW (March 2010). "Phantasmidine: an epibatidine congener from the ecuadorian poison frog Epipedobates anthonyi". Journal of Natural Products. 73 (3): 331–337. doi:10.1021/np900727e. PMC   2866194 . PMID   20337496.
  2. 1 2 3 Garraffo HM, Spande TF, Williams M (April 2009). "Epibatidine: from frog alkaloid to analgesic clinical candidates: a testimonial to" true grit"!" (PDF). Heterocycles. 79 (1): 207–217. doi:10.3987/REV-08-SR(D)5 (inactive 2024-03-07).{{cite journal}}: CS1 maint: DOI inactive as of March 2024 (link)
  3. Schwarcz J (2012). The Right Chemistry. Random House.
  4. Maxon J, Whittaker K (March 2009). "Epipedobates anthonyi". Berkeley, CA, USA: University of California. Retrieved 2015-05-06.
  5. 1 2 3 Daly JW, Garraffo HM, Spande TF, Decker MW, Sullivan JP, Williams M (April 2000). "Alkaloids from frog skin: the discovery of epibatidine and the potential for developing novel non-opioid analgesics". Natural Product Reports. 17 (2): 131–135. doi:10.1039/a900728h. PMID   10821107.
  6. Lasley EN (December 1999). "Having Their Toxins and Eating Them Too Study of the natural sources of many animals' chemical defenses is providing new insights into nature's medicine chest". BioScience. 45 (12). Oxford Journals: 945–950. doi: 10.1525/bisi.1999.49.12.945 .
  7. Daly JW, Garraffo HM, Spande TF, Decker MW, Sullivan JP, Williams M (April 2000). "Alkaloids from frog skin: the discovery of epibatidine and the potential for developing novel non-opioid analgesics". Natural Product Reports. 17 (2): 131–135. doi:10.1039/A900728H. PMID   10821107.
  8. Donnelly-Roberts DL, Puttfarcken PS, Kuntzweiler TA, Briggs CA, Anderson DJ, Campbell JE, et al. (May 1998). "ABT-594 [(R)-5-(2-azetidinylmethoxy)-2-chloropyridine]: a novel, orally effective analgesic acting via neuronal nicotinic acetylcholine receptors: I. In vitro characterization". The Journal of Pharmacology and Experimental Therapeutics. 285 (2): 777–786. PMID   9580626.
  9. Olivo HF, Hemenway MS (2002). "Recent syntheses of epibatidine. A review". Organic Preparations and Procedures International. 34 (1): 1–26. doi:10.1080/00304940209355744. S2CID   98696766.
  10. 1 2 3 4 5 Traynor JR (July 1998). "Epibatidine and pain". British Journal of Anaesthesia. 81 (1): 69–76. doi: 10.1093/bja/81.1.69 . PMID   9771274.
  11. 1 2 Clayton SC, Regan AC (1993). "A total synthesis of (±)-epibatidine". Tetrahedron Letters. 34 (46): 7493–7496. doi:10.1016/S0040-4039(00)60162-4.
  12. 1 2 Broka CA (1994). "Synthetic approaches to epibatidine". Medicinal Chemistry Research. 4 (7): 449–460.
  13. Broka CA (1993). "Total synthesis of epibatidine". Tetrahedron Lett. 34 (20): 3251–3254. doi:10.1016/s0040-4039(00)73674-4.
  14. Huang DF, Shen TY (1993). "A versatile total synthesis of epibatidine and analogs". Tetrahedron Lett. 34 (28): 4477–4480. doi:10.1016/0040-4039(93)88063-o.
  15. 1 2 Dowd MJ. "Epibatidine". Department of Medicinal Chemistry. Virginia Commonwealth University. Archived from the original on December 5, 2010.
  16. "Deriving a non-opiate painkiller [ABT-594] from Epipedobates tricolor". Mongabay.com. Retrieved 2014-03-12.
  17. "Epibatidine". School of Chemistry. University of Bristol. Retrieved 2021-11-12.
  18. Rizzi L, Dallanoce C, Matera C, Magrone P, Pucci L, Gotti C, et al. (August 2008). "Epiboxidine and novel-related analogues: a convenient synthetic approach and estimation of their affinity at neuronal nicotinic acetylcholine receptor subtypes". Bioorganic & Medicinal Chemistry Letters. 18 (16): 4651–4654. doi:10.1016/j.bmcl.2008.07.016. hdl: 2434/59291 . PMID   18644719.
  19. Dallanoce C, Matera C, De Amici M, Rizzi L, Pucci L, Gotti C, et al. (July 2012). "The enantiomers of epiboxidine and of two related analogs: synthesis and estimation of their binding affinity at α4β2 and α7 neuronal nicotinic acetylcholine receptors". Chirality. 24 (7): 543–551. doi:10.1002/chir.22052. PMID   22566097.
  20. Dallanoce C, Matera C, Pucci L, Gotti C, Clementi F, Amici MD, Micheli CD (January 2012). "Synthesis and binding affinity at α4β2 and α7 nicotinic acetylcholine receptors of new analogs of epibatidine and epiboxidine containing the 7-azabicyclo[2.2.1]hept-2-ene ring system". Bioorganic & Medicinal Chemistry Letters. 22 (2): 829–832. doi:10.1016/j.bmcl.2011.12.052. PMID   22222032.
  21. Dallanoce C, Magrone P, Matera C, Lo Presti L, De Amici M, Riganti L, et al. (December 2010). "Synthesis of novel chiral Δ2-isoxazoline derivatives related to ABT-418 and estimation of their affinity at neuronal nicotinic acetylcholine receptor subtypes". European Journal of Medicinal Chemistry. 45 (12): 5594–5601. doi:10.1016/j.ejmech.2010.09.009. PMID   20932609.
  22. Olivo HF, Hemenway MS (November 1999). "Total Synthesis of (+/-)-Epibatidine Using a Biocatalytic Approach". The Journal of Organic Chemistry. 64 (24): 8968–8969. doi:10.1021/jo991141q. PMID   11674810.
  23. 1 2 3 Fisher M, Huangfu D, Shen TY, Guyenet PG (August 1994). "Epibatidine, an alkaloid from the poison frog Epipedobates tricolor, is a powerful ganglionic depolarizing agent". The Journal of Pharmacology and Experimental Therapeutics. 270 (2): 702–707. PMID   8071862.
  24. Traynor JR (July 1998). "Epibatidine and pain". British Journal of Anaesthesia. 81 (1): 69–76. doi: 10.1093/bja/81.1.69 . PMID   9771274.
  25. Prince RJ, Sine SM (April 1998). "Epibatidine binds with unique site and state selectivity to muscle nicotinic acetylcholine receptors". The Journal of Biological Chemistry. 273 (14): 7843–7849. doi: 10.1074/jbc.273.14.7843 . PMID   9525877.
  26. Hogg RC, Raggenbass M, Bertrand D (2003). "Nicotinic acetylcholine receptors: from structure to brain function". Reviews of Physiology, Biochemistry and Pharmacology. 147: 1–46. doi:10.1007/s10254-003-0005-1. ISBN   978-3-540-01365-5. PMID   12783266.
  27. Olena A (September 21, 2017). "How Poison Frogs Avoid Poisoning Themselves". The Scientist.
  28. 1 2 Badio B, Daly JW (April 1994). "Epibatidine, a potent analgetic and nicotinic agonist". Molecular Pharmacology. 45 (4): 563–9. PMID   8183234.
  29. Sihver W, Långström B, Nordberg A (2002). "Ligands for in vivo imaging of nicotinic receptor subtypes in Alzheimer brain". Acta Neurologica Scandinavica. Supplementum. 176: 27–33. doi: 10.1034/j.1600-0404.2000.00304.x . PMID   11261802. S2CID   23541883.
  30. Sullivan JP, Bannon AW (1996). "Epibatidine: Pharmacological Properties of a Novel Nicotinic Acetylcholine Receptor Agonist and Analgesic Agent". CNS Drug Reviews. 2 (1): 21–39. doi:10.1111/j.1527-3458.1996.tb00288.x.
  31. Watt AP, Hitzel L, Morrison D, Locker KL (October 2000). "Determination of the in vitro metabolism of (+)- and (-)-epibatidine". Journal of Chromatography A. 896 (1–2): 229–38. doi:10.1016/s0021-9673(00)00597-5. PMID   11093658.
  32. Damaj MI, Creasy KR, Grove AD, Rosecrans JA, Martin BR (November 1994). "Pharmacological effects of epibatidine optical enantiomers". Brain Research. 664 (1–2): 34–40. doi:10.1016/0006-8993(94)91950-x. PMID   7895043. S2CID   46489298.
  33. Bacher I, Wu B, Shytle DR, George TP (November 2009). "Mecamylamine - a nicotinic acetylcholine receptor antagonist with potential for the treatment of neuropsychiatric disorders". Expert Opinion on Pharmacotherapy. 10 (16): 2709–2721. doi:10.1517/14656560903329102. PMID   19874251. S2CID   25690407.
  34. Nickell JR, Grinevich VP, Siripurapu KB, Smith AM, Dwoskin LP (July 2013). "Potential therapeutic uses of mecamylamine and its stereoisomers". Pharmacology, Biochemistry, and Behavior. 108: 28–43. doi:10.1016/j.pbb.2013.04.005. PMC   3690754 . PMID   23603417.