Subscriber access provided by Stockholm University Library
Review
DARK classics in chemical neuroscience: atropine, scopolamine and other anticholinergic deliriant hallucinogens Anton Lakstygal, Tatyana Kolesnikova, Sergey Khatsko, Konstantin Zabegalov, Andrey Volgin, Konstantin Demin, Vadim Shevyrin, Edina Wappler-Guzzetta, and Allan Kalueff ACS Chem. Neurosci., Just Accepted Manuscript • DOI: 10.1021/acschemneuro.8b00615 • Publication Date (Web): 19 Dec 2018 Downloaded from http://pubs.acs.org on December 20, 2018
Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.
is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.
Page 1 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
DARK classics in chemical neuroscience: atropine, scopolamine and other anticholinergic deliriant hallucinogens
Anton M. Lakstygal1, Tatiana O. Kolesnikova2, Sergey L. Khatsko2, Konstantin N. Zabegalov2, Andrey D. Volgin3, Konstantin A. Demin3,4, Vadim A. Shevyrin2, Edina A. Wappler-Guzzetta5 and Allan V. Kalueff6,7,8,9,10,11*
1Graduate 2Ural
School of Biology, St. Petersburg State University, St. Petersburg, Russia
Federal University, Ekaterinburg 620002, Russia
3Institute
of Experimental Medicine, Almazov National Medical Research Centre, Ministry of
Healthcare of Russian Federation, St. Petersburg, Russia 4Institute
of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg
199034, Russia 5
ZENEREI Research Center, Slidell LA 70458, USA
6School
of Pharmacy, Southwest University, Chongqing, 400700, China
7Anatomy
and Physiology Laboratory, Ural Federal University, Ekaterinburg, Russia
8Laboratory
of Biological Psychiatry, ITBM, St Petersburg State University, St Petersburg 199034,
Russia 9Scientific 10Granov
Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
Russian Scientific Center of Radiology and Surgical Technologies, Ministry of Healthcare
of Russian Federation, St. Petersburg, Russia 11Almazov
National Medical Research Centre, Ministry of Healthcare of Russian Federation, St.
Petersburg, Russia *Corresponding author: Allan V. Kalueff, PhD School of Pharmacy, Southwest University, Chongqing, 400700, China Tel/Fax: +1-240-899-9571 Email:
[email protected] Funding source: The research was supported by the Russian Foundation for Basic Research (RFBR) grant 16-04-00851 to АVK. KAD is supported by RFBR grant 18-34-00996 and Special Rector’s Fellowship for SPSU PhD Students.
Conflict of interest: Authors declare no conflicts of interest. ACS Paragon Plus Environment
1
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 2 of 38
TOC Graphic
ACS Paragon Plus Environment
2
Page 3 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
Abstract:
Anticholinergic drugs based on tropane alkaloids, including atropine, scopolamine and hyoscyamine, have been used for various medicinal and toxic purposes for millennia. These drugs are competitive antagonists of acetylcholine muscarinic (M-) receptors that potently modulate the central nervous system (CNS). Currently used clinically to treat vomiting, nausea and bradycardia, as well as alongside other anesthetics to avoid vagal inhibition, these drugs also evoke potent psychotropic effects, including characteristic delirium-like states with hallucinations, altered mood and cognitive deficits. Given the growing clinical importance of anti-M deliriant hallucinogens, here we discuss their use and abuse, clinical importance, and the growing value in preclinical (experimental) animal models relevant to modeling CNS functions and dysfunctions.
Keywords: the cholinergic system, deliriant drugs, atropine, scopolamine, hallucinogens, antimuscarinic effects
Abbreviations: AS – anticholinergic syndrome, BBB – the blood-brain barrier, MDD – major depressive disorder, BDD – bipolar depressive disorder, CNS – central nervous system, DAT – dopamine transporter, DFSA - drug facilitating sexual assault, GABA – gamma-aminobutyric acid, M – muscarinic (receptors), PNS – peripheral nervous system, TCA – tricyclic antidepressant, MDMA – 3,4-methylenedioxymethamphetamine
ACS Paragon Plus Environment
3
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 4 of 38
1. Introduction: chemistry and synthesis of deliriant anticholinergic agents Atropine, hyoscyamine and scopolamine (hyoscine, Fig. 1) are antagonists of the muscarinic (M) acetylcholine receptors that play an important role in modulating the central nervous system (CNS, Table 1). Chemically, they belong to tropane alkaloids, a group of over 200 analogs sharing a common tropane (N-methyl-8-azabicyclo[3.2.1]octane) fragment1-3. Structurally similar to acetylcholine, hyoscyamine and atropine are esters of tropine and optically active tropic acid. Pure biologically active hyoscyamine is a levorotatory optic isomer (S)-(–)-hyoscyamine and atropine is optically inactive (±)-racemate of hyoscyamine3, 4. Scopolamine is an optically and biologically active, racemation-stable (S)-(–)-isomer5 whose chemical structure, unlike atropine and hyoscyamine, includes the epoxy group in positions 6 and 7 of the tropane ring (Fig. 1). Atropine, scopolamine and hyoscyamine are most studied tropane alkaloids (Fig. 1, inset) naturally occurring in plants of the Solanaceae family (Anisodus, Anthocercis, Mandragora, Brugmansia, Duboisia, Hyoscyamus, Datura, Atropa, and Scopolia)1,
5-7.
Their alkaloid content
varies between 0.01–3%, depending on the species1, 5. Chemical names of these alkaloids originate from the respective plants from which they were extracted in the past. Atropine was extracted from Atropa belladonna (the deadly nightshade, belladonna) as the principally active compound by Mein and then – as a purified substance - by Runge3, 6, 8. The name ‘tropane’ itself originated from the Latin name for this plant2, 3, 8, also called ‘Atropos’ by the ancient Greeks9. Hyoscyamine was extracted from Hyoscyamus niger (the black henbane), and scopolamine from Scopolia carniolica (the henbane bell)2, 5, 10. Common street drug names of major tropane alkaloids are given in Table 2. The biosynthesis of tropane alkaloids occurs in roots, and then they are transported within the xylem1, 5. Since their chemical synthesis is complex and less feasible, plants remain the main source of tropane alkaloids5. Various Solanaceae plants accumulate pharmacologically active hyoscyamine and scopolamine as their major alkaloids, and are widely used for industrial production7, 11 as salts (after acidic/water treatment) or bases (following treatment with organic solvents like methanol, ethanol or chloroform)1. To isolate specific alkaloids, chromatography can also be used12. Furthermore, while plant cultivation is limited by ecological and climatic factors, it can be enhanced
ACS Paragon Plus Environment
4
Page 5 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
by hydroponic culturing and genetic engineering of alkaloid-producing plants and/or hairy root cultures1, 5, 7, 13-15. Enzymatic biosynthesis of tropane alkaloids involves amino acids L-ornithine, L- arginine (to form tropane ring) and L-phenylalanine (to produce tropic acid), as precursors1, 5, 7, 16-18 (Fig. 2). Ornithine undergoes enzymatic decarboxylation to produce putrescine, whereas arginine metabolism requires an additional step to form аgmatine and N-carbamoylputrescine. Putrescine is next methylated into N-methylputrescine, oxidatively desaminated into 4-(methylamino)butanal, which undergoes spontaneous cyclization into a reactive precursor of tropane ring, 1-methyl-3,4-dihydro2H-pyrrolium cation. Its reaction with acetoacetic acid generates 4-(1-methylpyrrolidin-2-yl)-3oxobutanoic acid, whose cyclization with decarboxylation produces tropinone (the first intermediate with the tropane ring). Reduction of its carbonyl group generates tropine which, in turn, reacts with phenyllactic acid (produced from phenylalanine) to generate littorine – a key precursor of hyoscyamine and other tropane alkaloids, whose 2-step reaction (via 6β-hydroxyhyoscyamine) produces scopolamine (Fig. 2). The first attempt to establish the structure of atropine was made by Kraut in 186319, detailing its degradation to tropine and tropic acid3. The full synthesis of atropine was reported by Willstätter in 190120 as a process that starts from cycloheptanone which undergoes a 8-step conversion into cycloheptatriene (Fig. 3). Sequential bromination of cycloheptatriene, replacing bromine with dimethylamine and reducing one of the two double links, results in 1-dimethylaminocyclohept-4-en3, 6, 18, 20,
followed by bromination of the remaining double link, creating a ring in 2-bromotropane salt
that produces tropidine upon reaction with bases. Tropane can then be produced in a 4-step reaction from tropidine3, 18, 20. Overall, albeit generally complex and low-yield, this atropine synthesis has been critical for proving its chemical structure. In 1917, a new efficient method was proposed by Robinson3,
6, 21,
based on 3-component condensation of succinaldehyde, methylamine and
acetonedicarboxylic acid (Fig. 3). Subsequent reduction of tropinone generates tropine, and its fusion with tropic acid in the presence of hydrogen chloride produces atropine3. This method of synthesis, with some modifications, remains widely used today3, 6, although alternative methods have also been proposed22-24, including continuous-flow synthesis25,
26.
Since scopolamine is a more expensive
ACS Paragon Plus Environment
5
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 6 of 38
product than atropine6, chemical epoxidation of tropane ring may be considered. The first total synthesis of scopolamine from (–)-tropane-3α,6β-diol was described in 19597, and while alternative methods of scopolamine synthesis exist23, its chemical synthesis remains less than extraction from plants6. Benzatropine, a tropane ring-containing anti-Parkinson agent (Fig. 1), was synthesized via the diphenyldiazomethane reaction with tropine by Merck & Co, patented in USA in 1952, and appeared on the marked as a mesylate salt in 195427, 28 (Table 2; diphenyldiazomethane was later replaced with a more safer agent, diphenylhalomethane29). Laboratory combinatorial synthesis of benzatropine and its analogs via azidoether has also been described30. Biperiden (Fig. 1) is another structurally related agent synthesized in Germany in 195331, which was patented in USA in 195732 and appeared in the market in 195928 as a therapy for Parkinson’s disease (Table 2). Biperiden can be synthesized by Mannich's reaction from acetophenone, formaldehyde and piperidine hydrochloride, followed by treatment with the Grignard reagent prepared in diethyl ether by 5-chloro-2-norbornene reaction with magnesium shavings31, 32. A modern method of industrial synthesis of biperiden utilizes 5-ethylene2-norbornene, whose selective oxidation produces 5-acetyl norbornene, which enters into a Mannich's reaction with formaldehyde and piperidine hydrochloride, followed by reaction with phenylmagnesium bromide33. Containing a fragment of tropic acid (Fig. 1), tropicamide was approved in USA in 1959 as an ophthalmological drug28, originally developed in 1952 by Hoffmann-La Roche, Inc. (Switzerland) and patented in USA34, 35. Tropicamide was synthesized from N-(pyridin-4-ylmethyl)ethanamine, obtained from 4-(chloromethyl)pyridine and ethylamine reaction with chloroanhydride of 3-acetoxy2-phenylpropanoic acid, and subsequent acidic hydrolysis of the O-acetyl group34, 35 (also note a similar patented reaction using modern technological approach and reagents36). The same method was used for synthesis of enantiomers of tropicamide37. Two other anti-Parkinson drugs, trihexyphenidyl and procyclidine, appeared in the USA market in the 1950s28 (Table 2), both sharing a common 1-cyclohexyl-1-phenylpropanol moiety but different (piperidine or pyrrolidine) rings (Fig. 1). Trihexyphenidyl has been originally developed as a spasmolytic agent, and its synthesis has been described in research and patent literature in early ACS Paragon Plus Environment
6
Page 7 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
1950s38-42
via
Grignard
reaction
of
cyclohexylmagnesium
bromide
with
3-(1-
piperidino)propiophenone, which is in turn obtained through Mannich's reaction of acetophenone, formaldehyde and piperidine hydrochloride28, 39, 40. Using phenylmagnesium bromide as a Grignard reagent, and ethyl cyclohexyl ketone for Mannich’s reaction can also be possible40. Moreover, partial catalytic reduction of diphenyl alkanolamines39, 42, albeit complicated by purification steps43, can be used. To increase yield, trihexyphenidyl can be synthesized from 3-chloropropanoyl chloride through sequential Grignard reaction with cyclohexylmagnesium and phenylmagnesium bromides, and treating the tertiary alcohol with piperidine43 (see synthesis of trihexyphenidyl enantiomers in 44, 45). Early reports on procyclidine appeared in the 1950s39,
46,
and its synthesis resembles that of
trihexyphenidyl, using pyrrolidine as an amine instead of piperidine38-46. 2. History of use and abuse Tropane alkaloids have a long history of use and abuse, as their spiritual, medicinal and toxic properties have been known since antiquity47 (Fig. 4). For example, fertility-promoting effects of mandrake, a plant of the Mandragora genus, are mentioned in the Old Testament (Genesis 30:1422). Ancient warriors and assassins have often soaked their weapons in the extract of the deadly nightshade9. The Solanaceae plants have been widely used as a poison for political assassinations, including killing of Macbeth, Duncan and Roman Emperors Claudius and Augustus9. In Shakespeare’s “Hamlet”, his father was poisoned by an ear-drop prepared from henbane48. Folk medicine has been using tropane alkaloids for centuries as analgesic agents, hallucinogens and poisons, representing one of the most ancient plant-derived compounds known to man49. Apart from religious rituals, pre-Columbian American civilizations medicated asthma, rheumatism and anxiety with plants of the Datura genus, whereas in medieval Spain extracts from of the Solanaceae family plants have been used as both recreational drugs and poisons50. In some Middle Eastern countries, people continue to use Datura plants to treat musculoskeletal problems, such as limbs swelling51. In medieval time, Solanaceae plants have commonly been used by witches, who would often prepare Pharmaka diabolics - the ointment that caused mental alterations (e.g., feeling of flying) and stimulated sexual desires52. By the 16-17th centuries, the Solanaceae plants have also begun to interest herbalists and apothecaries for their therapeutic effects53. In the 18th century, extracts from ACS Paragon Plus Environment
7
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 8 of 38
belladonna and other plants became a famous cosmetic ingredient used to enlarge pupils - a standard of female beauty at the time54 (hence is the plant name from Italian ‘bella donna’, a beautiful lady). More recently, tropane alkaloids have been widely utilized in physiology and pharmacology research. For instance, Schmiedeberg demonstrated that heart slowed by electrical stimulation returns to its normal rate after atropine exposure55. Later, the need to block sea sickness has drawn attention to clinical effects of tropane alkaloids – the line of research actively supported by the UK military and the Medical Research Council56. In the first half of the 20th century, atropine and scopolamine have been used as anesthetics, therapies for bradycardia, and to treat nausea and vomiting during seasickness57 or post-operative sickness58-60. Furthermore, atropine and other anti-M tropane alkaloids exert potent central nervous system (CNS) effects, including delirium-like and hallucinogenic effects (Table 1). This shared action has placed these drugs into a special group of deliriants hallucinogens61, 62
that evoke characteristic agitation, confusion, hallucinations and cognitive deficits63, 64 (Table 1,
Fig. 5). Atropine and scopolamine are the two best-studied anti-M deliriant agents (Table 3, Fig. 1). As they cause both central and peripheral M-mediated anticholinergic effects (Tables 1 and 4), their common action includes restlessness, excitement, hallucinations, euphoria, disorientation, stupor, coma or even death (typically, from respiratory depression). Scopolamine also produces rapid and robust antidepressant effects in unipolar and bipolar depression patients65, 66. At the same time, atropine, scopolamine and other deliriants can trigger anticholinergic syndrome (AS, Table 4), a critical diagnosis accounting for nearly 15000 toxic cases each year67. Even at therapeutic doses, patients on scopolamine are at risk for AS toxicity, especially when combined with other medications with anticholinergic effects. For example, beyond atropine and scopolamine, there are hundreds of unique pharmacological and herbal compounds that can cause anti-M effects68, such as antihistamines, tricyclic antidepressants (TCAs) and sleeping pills (e.g., doxylamine)69. Thus, both plant and synthetic anticholinergics have multiple therapeutic and side effects, and their clinical importance continues to grow (Fig. 1). Today, atropine is a controlled substance, currently listed in Schedule V in the US. Drugs from this schedule are the least regulated controlled substances, as they have a relatively low potential ACS Paragon Plus Environment
8
Page 9 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
for abuse and a currently accepted medicinal use. Scopolamine and hyoscyamine are not listed as controlled substances in the US. However, access to these three deliriant drugs is administratively limited, and prescriptions are needed for them in many countries worldwide. US researchers must have an approved state license to be able to explore atropine effects in vivo or in vitro, due to its current scheduling as a controlled substance. 3. Clinical and preclinical pharmacology Scopolamine is an M-receptor antagonist with peripheral and central anticholinergic, as well as sedative and amnestic properties (Table 1). In humans, scopolamine patches may evoke transient psychosis even without AS-like toxicity67. Fast-acting antidepressant effects of scopolamine have also been described70, with somewhat stronger action in women71. Scopolamine administration in randomized placebo–controlled study in patients with major depressive or bipolar disorders evokes antidepressant effects72, and scopolamine-containing drugs are efficient in panic attack, nervousness and anxiety73. Preclinical effects of scopolamine, recently discussed in-depth74 (Table 5), include cognitive impairments in passive avoidance task and Morris water maze75-77, poorer fear-context conditioning78 and increased anxiety-like behavior79. Atropine is another typical anti-M drug80. As a parasympathetic blocker, it reduces salivary and bronchial secretions, raises the heart rate and relaxes the smooth muscle of the urinary bladder, gastro-intestinal tract and bronchi81. Since atropine easily crosses the blood-brain barrier (BBB), it quickly causes CNS effects - restlessness, mental excitement/agitation and hallucinations (Table 1). While nerve gases used in warfare (e.g., Sarin) inactivate acetylcholinesterase82 and cause lethality, atropine acts as a potent antidote83 commonly used by the military84. Atropine also prevents accidental poisoning by organophosphate insecticides (which also inhibit acetylcholinesterase activity)85. In rodents, similarly to scopolamine (Table 4), atropine impairs newly formed fear memories86 and evokes anxiety74, 79. Atropine also acts as competitive antagonist of serotonin 5HT3 receptors87 and may modulate various neurotransmitters involved in emotionality (e.g., cholinergic, dopaminergic, glutamatergic and GABA-ergic)88 and behavior87. Benzatropine is an atypical anti-M tropane alkaloid (Table 3) which also inhibits the dopamine transporter (DAT)89, is used clinically to treat Parkinson’s symptoms90, and evokes ACS Paragon Plus Environment
9
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 10 of 38
antihistaminic effects91. Several benzatropine analogs also inhibit histamine H1 receptors91 and DAT, predictably inducing reward-like effects (e.g., in primate models)92, 93. Procyclidine exerts strong antidotal anti-M, antiglutamatergic and antiepileptic effects94. It also reduces anxiety in depressed patients treated jointly with chlorpromazine and, to a lesser extent, with procyclidine alone95. However, it impairs learning and memory in both rodent96 and clinical studies97, and (similarly to other anticholinergic drugs) evokes strong delirium and visual hallucinations that are suppressed by anticholinesterase drugs98. Tropicamide is an anticholinergic tropic acid derivative with an anti-M activity, often causing mydriasis and cycloplegia99. While topical abuse of ophthalmic solutions has occurred previously, intravenous abuse of tropicamide has emerged as a recent medical problem100. Notably, some case reports suggest that tropicamide may improve subjective effects of heroin intake, causing a more powerful relief and relaxation (compared to heroin alone), but also AS-like blurred vision, dizziness, illusions and hallucinations, as well as memory- and appetite loss101. Furthermore, tropicamide also induces palpitations, dissociation, anxiety, and sweating in humans101, and suppresses some Parkinson-like phenotypes (e.g., stereotypic jaw movements) in rodents102. Biperiden is a predominantly M1-antagonist103 (Table 3) which impairs cognitive performance (verbal memory, visuospatial processes, and motor learning) clinically104 and disrupts short-term memory, but not sensorimotor responding, food motivation or attention, in rodents105. The most common central side-effects of biperiden are drowsiness, headache, vertigo and dizziness106 (Table 1). Its peripheral AS-like side effects include blurred vision, mydriasis, dry mouth, abdominal discomfort and constipation107, 108. Finally, trihexyphenidyl is an anti-M drug acting both centrally and peripherally109, and currently used to treat tremors, spasms, stiffness and muscle weakness in patients with Parkinson’s disease110 as well as in similar conditions evoked by some CNS drugs (e.g., fluphenazine, haloperidol and chlorpromazine)111. Like biperiden, trihexyphenidyl impairs memory in rats103 and worsens cognitive performance clinically112. 4. Metabolism and pharmacokinetics Atropine, scopolamine and hyoscyamine are usually taken orally, intravenously, intramuscularly or in the form of suppository, aerosols or transdermal patches68, ACS Paragon Plus Environment
113-115.
Low 10
Page 11 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
molecular weights of scopolamine (303 g/mol) and atropine (289 g/mol) and their lipophilic character enable an easy passing the gastric mucosa of humans and mammals after intake, ensuring their fast absorption and action116. The absorption of atropine and other tropane alkaloids through the skin is moderate115. The first signs of clinical atropine poisoning appear 15-30 min after ingestion; its halflife is 4 h, and 90% of the drug is excreted with urine within 24 h117. The peak plasma levels of atropine depend on the route of administration, ranging from 13 min for intramuscular to 1 h for oral and 1.5-4 h for aerosol administration118. Most of the drug effects disappear within 24-48 h117, except for mydriasis that can persist for days119. Scopolamine has a short half-life in plasma, peaking 30 min after ingestion120, but causing overt physiological effects lasting for hours. Bioavailability of hyoscyamine is approximately 43%. Its pharmacological effects and their duration depend on the form of the drug, appearing within minutes for tablets, solutions or elixirs (peaking at 15-60 min and lasting for 4 h) and within 20-30 min for capsules (peaking at 40-90 min and lasting for 12 h)121. The half-life of hyoscyamine is 3.5 h in serum and 7 h in plasma121. Hyoscyamine is distributed through the whole body, readily passing through organ barriers and in small quantities penetrating into breast milk121. Tropane alkaloids are metabolized in the liver and excreted with urine, with 13-50% atropine excreted unchanged, and ~65% scopolamine excreted within 24 h as glucuronide and sulfide conjugates122. Hyoscyamine is also metabolized in liver121, and is excreted with urine within 12 h, mostly unchanged123. Post-mortem concentrations of hyoscyamine can reach 0.2 mg/L in the blood and 14.2 mg/L in the urine124. Atropine (8-15 pg/mg) and scopolamine (1–1.3 pg/mg) can also be detected in hair125. The lethal concentration in the peripheral blood for atropine is 0.3 mg/L and for scopolamine 0.005 mg/L126. Procyclidine, trihexylphenyl, biperidine are typically administered as hydrochloride salts and are rapidly absorbed in the gastrointestinal tract, whereas benzatropine is used as a mesylate salt with a slower absorption. Plasma concentrations of these drugs peak within 2.5 h (7 h for benzatropine)127. Depending on the dose, maximal plasma concentrations after oral intake are 1.5 ng/ml for benzotropine128, 10-50 ng/ml for trihexyphenyl129, 130, 4 ng/mL for biperidine131 and 113 ng/ml for procyclidine132. Biperidine quickly penetrates BBB in rats, and its concentration in the brain peaks ACS Paragon Plus Environment
11
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 12 of 38
within 3-10 min after an intravenous injection133 due to high lipophilicity of this drug. Fasting and change in the volume of adipose tissue in rats reduce biperidine accumulation134. In humans, biperedine plasma levels after an oral intake are lower than after intravenous injection, peaking at 1.5-2 h with the half-life of ~21 h. The bioavailability of biperidine is 33%, and it is excreted by kidneys and the liver131. Tropicamide is usually administered intravenously, and its physiological effect lasts for 0.5-6 h135. Data are currently limited on its metabolism and pharmacokinetics. Following an eye instillation, it absorbs rapidly, within 5 min reaching the peak plasma concentration, and disappears from the system within 2 h136. Following an oral administration in rats, the maximal concentration of the drug is detected in the stomach within 1-6 h, with much lower levels also found in the brain, lungs and spleen137. 5. Current social and cultural impact Plants containing anti-M deliriant tropane alkaloids have long been used in different cultures for the unique bodily and mental/spiritual experiences they evoke (Table 1, Fig. 4). While the use of tropane alkaloids for political poisoning or witchcraft is long gone, their societal impact remains high due to active medicinal use, severe toxicity, some abuse potential, robust deliriant/hallucinogenic properties and the availability of synthetic analogs (Fig. 1) with potent CNS action. For example, since antiquity, people have used belladonna and other plants for special beverages that bring people ‘closer to gods’ through ecstasy and dreaming52. The link between tropane alkaloid use and other drugs has also been long recognized. In Ancient Egypt, mandrake plant has been added to beer to potentiate alcohol action52. In Europe, torn apples and henbane were commonly utilized for this purpose138,
139.
Similar synergism occurs in pharmacotherapy, since some anticholinergic drugs
potentiate the effect of other agents (e.g., procyclidine increases antidepressant effects of chlorpromazine140), which can be used therapeutically but may also worsen side-effects. Although a generally unpleasant subjective experience with deliriant hallucinations and multiple AS-like side-effects (Tables 5 and 6) limit the appeal of these drugs, their abuse is possible. In modern history, the use and abuse of deliriants as psychoactive substances has been popular among some young people. For example, in the late 1960s, deliriant poisoning became widespread among USA students using asthma medicine ‘Asthmador’ (containing an extract of D. stramonium) to ACS Paragon Plus Environment
12
Page 13 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
experience delirium and hallucinations141. Anticholinergic drugs are abused in low- and middleincome countries (e.g., Saudi Arabia, Jordan, Iraq and Brazil), especially by young unemployed and marginalized people with polysubstance abuse and a family history of mental disorders142. A recent rise of abuse of anticholinergics reported in Europe involves poor African immigrants142. Such targeting of people on low income and welfare by anti-M deliriant drugs can be due to their low cost (vs. other psychoactive substances - cannabis, ‘ecstasy’/MDMA or cocaine)143, and easy access to them (due to weak control of anticholinergic drugs)142. Presently, the most commonly abused anticholinergic drug is trihexyphenidyl because of its strong euphorigenic effect144. Biperiden abuse also occurs among people who have previously abused other substances145. Albeit a less potent anticholinergic then trihexyphenidyl, it has a strong potential to abuse, causing various mood disorders and (upon withdrawal) dysphoria, fatigue, lack of motivation and insomnia142. Recently, scopolamine abuse has been noted in USA prisoners with prior history of abuse (mainly cocaine and heroin), using crushed scopolamine tablets for smoking146. Overall, the abuse of deliriants tends to occur in groups with high risk of social vulnerability, as only populations that do not have access to other drugs would become abusers of drugs that are toxic and induce unpleasant subjective effects. The high prevalence of abuse of such substances may therefore be more indictive of socioeconomic status of the individuals, rather than reflect the abuse potential of deliriant drugs per se. Delirium-like hallucinogenic effects of tropane alkaloids is another key biomedical concern. For example, tropane alkaloids, especially scopolamine from Brugmansia arborea, have been used for centuries by Peruvian sorcerers to interact with spiritual forces147. Delirium-inducing properties of Solanaceae plants have also been used for banquets and entertainment, when intoxicated people felt and behaved ‘like animals’139. In the first half of 20th century, scopolamine was suggested to evoke “twilight sleep”, during which exposed individual honestly answered the questions without remembering it after recovering from the drug. While this led to several cases of interrogating criminals by police using scopolamine as a ‘truth serum’, this practice was deemed highly controversial and is discontinued, according to CIA report7. Nevertheless, toxic, amnestic and sedative effects of scopolamine have been adopted by Columbian criminals to render and kidnap their victims7, 148, thus becoming a societal problem in South America in recent years. ACS Paragon Plus Environment
13
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 14 of 38
Another aspect to consider is the effects of anti-M deliriants on sexual behavior, since they have historically been used to stimulate fertility (Genesis 30:14-22) and sexual desires52. Representing naturally occurring aphrodisiacs, tropane alkaloids often stimulate libido but also cause toxicity in their users149, 150. Presently, both atropine and scopolamine are included in some, but not all, studies of drugs facilitating sexual assault (DFSA)151. Scopolamine may draw more attention in this regard due to a stronger sedation (than atropine) and overt amnestic effects151, which can promote sexual assault or violence152, 153. Some cases of hypersexuality are described following intake of other drugs, such as clomipramine, with overt anticholinergic side-effects154. Rodent literature supports the role of deliriants in the regulation of sexual behavior. For example, central microinjections of scopolamine in male hamsters increases intromission frequency and ejaculation latency, also causes continuous strings without the usual separation by dismounts155. Rodent and primate data also support sex differences in responses to scopolamine, since it inhibits sexual behaviors in females, but not in males156-158. However, such sexual inhibition following scopolamine may also be due to sedation, commonly observed in rodents159 and therefore meriting further scrutiny. A typical delirium experience usually involves confusion with hallucinations and complex visual imagery160. Neuropsychiatric effects of anticholinergic deliriants are dose-dependent, and include various positive and negative symptoms (Fig. 5), ranging from hallucinations to drowsiness, sedation, ataxia, amnesia, paranoia, picking behaviors and coma161,
162.
Psychotogenic effects of
deliriants is another problem, especially among abusers of anti-M drugs163 (also see similar animal data164). In addition to atropine-165, 166 and scopolamine-evoked psychoses167-169, self-injury and death during hallucinations170,
171
also occur, making deliriants a common cause of death due to self-
damage or toxicity171. For instance, only in the US, over 100000 cases of plant poisoning are reported annually, with a 16-% risk of death for some species172, further supporting clinical and societal importance of deliriant drugs. 6. Concluding remarks In summary, anticholinergic deliriant hallucinogens are an important class of drugs relevant to multiple CNS conditions in both humans and animal models (Tables 1 and 3). Some new CNS properties of tropane alkaloids include rapid (3-4 days) antidepressant properties of scopolamine173ACS Paragon Plus Environment
14
Page 15 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience 175,
particularly important since depression is often resistant to traditional antidepressants176,
177.
However, scopolamine can also be pro-depressant in some models: for instance, in a rat model of Alzheimer’s disease, as it increases immobility time in the forced swim ‘despair’ test178,
179.
Furthermore, other drugs from this group may not show antidepressant activity, as, for example, atropine coma therapy (previously used in psychiatric practice) is not recommended for depressed patients180. Thus, further studies are needed to understand rapid antidepressant action of scopolamine and the role of anti-M effects in such responses. A core feature of anti-M drugs is their ability to induce delirium – an important mental state clinically overlapping with schizophrenia, unipolar and bipolar depression, anxiety and addiction181 (Fig. 5), with hyper- and hypoactivity182, 183 that quickly and unpredictably switch184. While similar alternating states can be observed in animal models after biperiden exposure185, the exact nature of this switch remains unclear. Likewise, while delirium can be triggered by many agents beyond tropane alkaloids, it is unclear how deliriant effects of the later are neurobiologically related to antiM delirium186, 187. As clinical delirium is a highly heterogenic condition188 (Fig. 4), the potential of deliriant drugs to evoke such heterogenic multifactorial state may indeed be promising. Recent findings implicate the immune system and microglia in the regulation of brain functioning and behavior, including psychiatric disorders189,
190.
Since delirium is associated with excessive
production of inflammatory mediators by microglia191, it may be interesting to examine the impact of deliriant hallucinogens on neuroimmune mechanisms and microglia activity following their acute and chronic administration. Moreover, since deliriant agents discussed here have several medicinal properties, including overt anti-Parkinson effects (Table 1), their clinical applications merit further scrutiny, and may eventually be expanded to target a wider range of CNS conditions. Overall, despite being used for millennia as psychoactive compounds, anticholinergic deliriants remain an interesting but under-studied group potently affecting multiple behavioral domains (e.g., cognition, sleep, memory, hallucinations, motor activity, anxiety and reward) relevant to many psychiatric conditions (Table 1). The study of deliriants can also improve our understanding of delirium per se (Fig. 2). While such cross-endophenotypic anticholinergics effects and the heterogeneity of delirium may complicate these studies, they may be beneficial in a long-run due to ACS Paragon Plus Environment
15
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 16 of 38
unique potential to unravel connections between these endpoints, and the role of specific (anti-M) mechanisms in their modulation (Table 3). Collectively, this necessitates further studies of deliriant, cognitive, abuse-related and affective activity of anticholinergic hallucinogenic compounds.
Funding source: The research was supported by the Russian Foundation for Basic Research (RFBR) grant 16-04-00851 to А.V.K. K.A.D. is supported by RFBR grant 18-34-00996 and Special Rector’s Fellowship for SPSU PhD Students.
Acknowledgement: The authors thank Lyudmyla E. Kalueva for providing an artwork for the TOC Graphic of this paper.
Conflict of interest: Authors declare no conflicts of interest.
Author Contributions: All authors have extensively contributed to this manuscript. A.V.K. conceived and coordinated the project, with conceptual input from V.A.Sh. All authors have participated in data collection, analysis and interpretation. A.M.L., K.A.D, T.O.K., S.L.Kh., K.N.Z. and V.A.Sh drafted the manuscript. A.M.L., K.A.D., A.V.K. and V.A.Sh. participated in critical review and further revision of the manuscript. All authors contributed to critical discussions and finalizing the manuscript before submission, and have approved its final form.
ACS Paragon Plus Environment
16
Page 17 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
Figure 1. Chemical structure of major tropane alkaloids (top row) and some synthetic analogs (bottom row) of tropane alkaloids. Inset - representation of various anticholinergic deliriant hallucinogens in scientific literature, based on Pubmed (www.ncbi.nlm.nih.gov/pubmed/) queries. Line diagram (top) shows the yearly cumulative numbers of papers published in 1955-2018 and mentioning the respective drugs in case reports, clinical studies, clinical trials (I-IV), evaluation studies, meta-analyses, observational studies and randomized controlled trials (the Y-axis is logarithmic, for better visuality). Bottom panel (pie-charts) presents relative number of articles published on various deliriant drugs (expressed in % of total) filtered for human studies (left) and animal studies (right), respectively.
ACS Paragon Plus Environment
17
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 18 of 38
N N O
N OH
O
OH
OH N
OH N
N Benztropine
Biperiden
Tropicamide
Trihexyphenidyl
Procyclidine
Number of articles in Pubmed
% of total (clinical)
% of total (pre-clinical)
ACS Paragon Plus Environment
18
Page 19 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
Figure 2. Biosynthetic pathway of major tropane alkaloids
ACS Paragon Plus Environment
19
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 20 of 38
Figure 3. Chemical synthesis of atropine (inset)
ACS Paragon Plus Environment
20
Page 21 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
Figure 4. Biological effects of anti-muscarinic hallucinogenic deliriants. The most known deliriants, atropine and scopolamine, are extracted from Atropa belladonna, used for ages for various purposes. In ancient times, it has been used as an efficient poison, and for witchcraft in medieval times. In the renaissance era, they became popular among women for expanding pupils. In the 20th century, atropine and scopolamine were actively used for research and also were introduced into clinical practice, to treat vomiting and nausea. Currently, their putative ‘fast’ antidepressant properties are also being studied.
Poison used by assassins and military structures
Treatment for postoperative nausea and vomiting
Atropa belladonna (AB)
Extract of AB has been widely used among renaissance era to extend eye-pupils (wide pupilsbeauty standard for women of this era)
Nowadays anti-cholinergic deliriants are considered as a potential treatment for depression (e.g. scopolamine can modulate activity of serotoninergic system
ACS Paragon Plus Environment
21
ACS Chemical Neuroscience
Figure 5. Summary of clinical delirium-like states common for clinical delirium or evoked by antimuscarinic deliriant hallucinogens. Note that hyper- and hypoactive states often fluctuate and switch. Most of the states shown in this diagram are easy to model in animal experimental models, and are commonly seen in clinical patients following intake of anticholinergic deliriants (see Table 1 for details).
Hyperactivity
Hypoactivity
Confusion, disturbed attention Hallucinations Hyperlocomotion Agitation Illusions and delusions Restlessness Nervousness/Anxiety Hypersexuality Sleep problems Aggression
Hypolocomotion Reduced alertness Drowsiness Lethargy Disordered thought Executive dysfunction
Mixed type
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 22 of 38
Cognitive deficits
ACS Paragon Plus Environment
22
Page 23 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
Table 1. Summary of clinical effects of antimuscarinic deliriants (also see Table 2) Drug
Therapeutic effects
Side effects
Atropine
Reduced motion sickness, ophthalmic effect
Delirium-related: Confusion, hallucinations, excitement, disorientation Cognitive: Memory impairment Other behavioral: Anxiety, coma and convulsions Body-related: Blurred vision, change in color vision, difficulty seeing at night, eye pain or stinging, fast or irregular heartbeat or pulse, high photosensitivity, hyperthermia
Scopolamine
Reduced motion sickness
Delirium-related: Confusion, hallucinations, mood or mental changes, restlessness Cognitive: Impairment of selection/evaluation of environmental information and memory Other behavioral: Dizziness, faintness, tiredness, anxiety/fear Body-related: Blurred vision, chest pain/discomfort, difficulty urinating, dilation of pupils, lightheadedness when getting up, eye pain, skin redness, muscle weakness, nausea or vomiting, rash, shortness of breath slow or irregular heartbeat, sweating, unusually warm skin
Hyoscyamine
Mydriatic, cardiovascular, antispasmodic effects
Delirium-related: Confusion, hallucinations, disorientation, depersonalization Cognitive: Memory impairment Other behavioral: Auditory changes, ataxia Body-related: Muscular incoordination, blurred vision, hyperthermia, reduced salivation
Benztropine
Anti-Parkinson effects
Delirium-related: Altered consciousness, false fact-less beliefs, unusual excitement, nervousness or restlessness Cognitive: Memory impairment Other behavioral: Dizziness, faintness, tiredness, loss of consciousness, shakiness and unsteady walk, motor incoordination Body-related: Poor/blurred vision, cold skin, lightheadedness, muscle weakness, nausea or vomiting, breathing problems, nosebleeds, numbness or tingling in the face, arms or legs, sweating, tearing, wheezing
Biperiden
Anti-Parkinson effects
Delirium-related: Anxiety, confusion (about identity, place and time), false or unusual sense of well-being, irritability, nervousness, restlessness, trouble sleeping, unusual tiredness Cognitive: Visual memory impairment, short-term memory impairment Other behavioral: Disturbed behavior, dizziness, faintness, twisting, uncontrolled movement of the face, hands, arms or legs, shaking Body-related: Chest pain, chills, cold sweats, reduced or painful urination, lightheadedness, dry mouth, hyperventilation, slow or irregular heartbeats, shortness of breath
Tropicamide
Ophthalmic effect
Delirium-related: Confusion, hallucinations Cognitive: Memory impairment Other behavioral: Clumsiness or unsteadiness, slurred speech, drowsiness, tiredness and general weakness, unusual/atypical behaviors, especially in children Body-related: Fast heartbeat, flushing or redness of face, increased thirst, dry mouth, skin rash, swollen stomach
Trihexypheni dyl
Anti-Parkinson effects
Delirium-related: Nervousness, disorientation (especially in children) Cognitive: Memory impairment Other behavioral: Delusions of persecution, mistrust, suspiciousness and combativeness Body-related: Dizziness, mild nausea
Procyclidine
Anti-Parkinson effects
Delirium-related: Agitation, confusion, overactive behavior, sexual hyperactivity, irritability Cognitive: Memory impairment Body-related: Dry mouth, blurred vision, nausea, vomiting, epigastric distress, constipation, lightheadedness, muscular weakness
ACS Paragon Plus Environment
23
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 24 of 38
Table 2. Chemical, market and street names of common anticholinergic deliriants (see Table 1 for summary of clinical effects) Drugs
Selected marketed names
Atropine*,**
Atropine Care, Isopto Atropine
Scopolamine (hyoscine, scopine tropate)**
Scopace, Maldemar
Hyoscyamine (daturine)*,**
Anaspaz, Cystospaz, Hyosyne, Levbid, Levsin, Levsinex, NuLev, Symax, Oscimine
Benztropine***
Cogentin, Cogentinol, Cogentine, Akitan, Bensylate, Cobrentin
Biperiden****
Akineton HCl, Akinophyl, Akineton, Dekinet, Ipsatol, Paraden, Tasmolin
Tropicamide (benzeneacetamide)
Mydryacyl, Mydriaticum, Mydrin, Mydrum, Tropimil, Tryptar, Visumidriatic
Trihexyphenidyl****
Atrane, Pipanol, Tremin, Antitrem, Anti-Spas, Aparkan, Broflex, Aparkane, Novohexidyl, Paralest, Pargitan, Parkinane, Parkopan, Partane, Peragit, Pyramistin, Rodenal, Sedrena, Trihexane, Trihexy, Triphedinon
Procyclidine****
Kemadrin, Kemadrine, Arpicolin, Kemadren, Osnervan, Procyclid
*Commonly used as a sulfate salt **These drugs share the street names, originating from Datura stramonium: Jamestown weed, Jimson weed, datura, stinkweed, devil’s snare, devil’s seed, angel/devil’s trumpet, Korean morning glory, crazy tea, moonflower, thorn apple, malpitte, ***Commonly used as a mesylate salt ****Commonly used as a hydrochloride salt
ACS Paragon Plus Environment
24
Page 25 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
Table 3. A brief summary of muscarinic acetylcholine receptor subtypes (M1-M5) and deliriant binding profiles in mammals (+ potent activity, 0 no effects, ? unclear effects or no data) Drugs
M1
M2
M3
M4
M5
References
Atropine
+++
++
++
+
+
192, 193
Scopolamine
+++
+++
+++
+++
+
194
Hyoscyamine
0
+++
0
0
0
195
Benztropine
+
0
+
0
0
196
Biperiden
+++
+
+++
+++
+++
197, 198
Tropicamide
+++
0
0
+++
0
199, 200
Trihexyphenidyl
+++
+
+
+++
+
201
Procyclidine
+++
+++
?
+++
?
202
ACS Paragon Plus Environment
25
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 26 of 38
Table 4. A brief summary of symptoms recognized as anticholinergic syndrome (AS) commonly evoked by anticholinergic deliriant hallucinogens (also see Table 1 for details of drug-evoked mental effects)
AS symptoms
Drugs most commonly evoking the symptoms
Tachycardia
Atropine, scopolamine203
High blood pressure
Atropine, scopolamine203
Fever
Atropine, scopolamine203
Agitation, anxiety
Atropine, scopolamine203, tropicamine204
Memory impairment
Atropine, scopolamine203
Sweating
Atropine, scopolamine203, tropicamine204
Palpitations
Tropicamide204
Blurred vision, mydriasis
Atropine, scopolamine203, tropicamine204, biperiden205
Dry mouth
Atropine, scopolamine203, tropicamine204, biperiden205
Abdominal discomfort
Biperiden205
ACS Paragon Plus Environment
26
Page 27 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
Table 5. A brief summary of preclinical effects of tropane anti-cholinergic tropane alkaloids (see 74 for review and Tables 1 and 6 for a summary of similar clinical effects) Drug
Effects in rodents
Atropine
Delirium-like: Impaired stimulus sensitivity, sensory discrimination, vision and behavioral patterning Cognitive: Reduced passive avoidance, impaired memory Other: Evoked sleep-like slow wave cortical activity
Scopolamine
Delirium-like: Increased spontaneous locomotion, longer latency between stimuli in the choice reaction task Cognitive: Impaired working and social memory, fear conditioning Other: Lower food motivation, antidepressant-like effects
Hyoscyamine
Delirium-like: Atropine-like stimulatory effects on central and motor neurons Other: Nociceptive effect
Benztropine
Delirium-like: Increased locomotion and rearing Cognitive: Cognitive deficits
Biperiden
Delirium-like: Alternating hyper- and hypoactive states Cognitive: Memory impairments (passive avoidance test) Other: Antidepressant-like effects
Tropicamide
Delirium-like: Increased anxiety-like behavior in rats Cognitive: Cognitive deficits Other: Suppressed stereotypic jaw movements in a model of Parkinson’s disease
Trihexyphenidyl
Delirium-like: Sleep deficits, enhanced cocaine effects, hyperactivity and increased male sexual behavior Cognitive: Cognitive deficits
Procyclidine
Delirium-like: Agitation, confusion Cognitive: Cognitive deficits
ACS Paragon Plus Environment
27
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 28 of 38
Table 6. Selected personal experiences with various deliriant hallucinogens described in the Erowid database (www.erowid.org, accessed November 2018) and/or published case reports. Note a strikingly overlapping clinical picture of drug-evoked delirium and hallucinogenic states. Drugs Atropine
Dose 32 mg i.v. 2 eye-drops
Scopola-mine
1.6 mg i.v. 7 mg orally
Patch 6 mg orally
Hyoscyamine
Benztropine
Biperiden
3 mg orally 7 mg orally 250 mg sublingually 2 mg i.m. 2 mg orally 4 mg orally 50 mg orally 4 mg orally 6 mg orally
Tropicamide
Not specified, i.v.
Trihexyphenidyl
Not specified, orally
Procyclidine
300 mg orally 15 mg orally 8 tablets
Neurobehavioral effects Disorientation, delayed recall, inattention, delusions of persecution, following only simple commands, inability to cooperate during a formal mental examination Psychomotor excitation, disorientation to time and place, delusions and visual hallucinations (seeing mice) Visual hallucinations for 11 days, severely impaired vision for 3 days Blurred vision, ataxia and slurred speech after 30 min, overt delirium, low muscle tone after 1 h, severe myorelaxation, impaired hearing, colorful bright visual hallucinations, poor vision, impaired depth perception (walls ‘breathing’, objects swirling and taking on living forms), robust frightening hallucinations, total disorientation after 2 h. Confusion, poor concentration and memory, blurry vision, dilated pupils, mild paranoia after 16 h. General perception: highly negative Sleep-like walking in a dreamy state within several hours Perceptual changes, blurred vision, poor concentration, severe spatial distortion, incoordination after 40 min; muscle weakness, lucid dreaming sleep, poor perception, visual and auditory hallucinations, communication with imaginary persons, talking to himself, memory loss, fatigue, sleepiness after 2-4 h, poorer concentration 24 h later. General perception: negative Confusion, hallucinations, memory loss, Delirium Impaired perception (moving furniture, volumetric vision of flat picture) Feeling of rush after injection Delirium, hallucinations, restlessness, disorientation Confusion, agitation, aimless wandering Feeling of relaxation and increased sociability, somnolence and disorientation to time/place, incoherent speech, blurry vision, resting tremor, poor appetite, hypotonia, flaccidity in the facial muscles and slow gait Improved mood, reduced agitation and anxiety, normalized sleep-wake schedule Auditory hallucinations and bizarre behaviors (talking to himself, picking up imaginary objects), altered mental status, disorientation, restlessness, insomnia Feeling of fun, joy, but also horrific, scary and dangerous experience Dissociation, hallucinations, anxiety and blurred vision Powerful relief and relaxation (in combination with heroin), but hallucinations and dissociation after tropicamide alone Euphoria (feeling of ‘loving everybody in the world’) and increased sociability Animated and pleasant hallucinations Auditory and visual hallucinations (imaginary objects around) Agitation and hallucinations Disorientation to time/place, agitation, emotional lability and frequent fears Delirium, disorientation and motor incoordination
ACS Paragon Plus Environment
References or Erowid record ID 206 207 208
16448
36749 27661
58661 39749 21232 209 210 211 212
213 214 215 101 101 209 209 100 216 217 218
28
Page 29 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
References 1. Georgiev, V., Marchev, A., Berkov, S., and Pavlov, A. (2013) Plant In Vitro Systems as Sources of Tropane Alkaloids. In Natural Products: Phytochemistry, Botany and Metabolism of Alkaloids, Phenolics and Terpenes (Ramawat, K. G., and Mérillon, J.-M., Eds.), pp 173-211, Springer Berlin Heidelberg, Berlin, Heidelberg. 2. Philipov, S., and Doncheva, T. (2013) Alkaloids Derived from Ornithine: Tropane Alkaloids. In Natural Products: Phytochemistry, Botany and Metabolism of Alkaloids, Phenolics and Terpenes (Ramawat, K. G., and Mérillon, J.-M., Eds.), pp 343-358, Springer Berlin Heidelberg, Berlin, Heidelberg. 3. Schulte, M. L., and Lindsley, C. W. (2012) Tropane-Based Alkaloids as Muscarinic Antagonists for the Treatment of Asthma, Obstructive Pulmonary Disease, and Motion Sickness, Bioactive Heterocyclic Compound Classes: Pharmaceuticals, 21-36. 4. Gualtieri, F., Conti, G., Dei, S., Giovannoni, M. P., Nannucci, F., Romanelli, M. N., Scapecchi, S., Teodori, E., Fanfani, L., and Ghelardini, C. (1994) Presynaptic cholinergic modulators as potent cognition enhancers and analgesic drugs. 1. Tropic and 2-phenylpropionic acid esters, Journal of medicinal chemistry 37, 1704-1711. 5. Oksman-Caldentey, K., and Arroo, R. (2000) Regulation of tropane alkaloid metabolism in plants and plant cell cultures. In Metabolic engineering of plant secondary metabolism pp 253-281, Springer. 6. Grynkiewicz, G., and Gadzikowska, M. (2008) -Tropane alkaloids as medicinally useful natural products and their synthetic derivatives as new drugs, Pharmacological Reports 60, 439. 7. Ullrich, S. F., Hagels, H., and Kayser, O. (2017) Scopolamine: a journey from the field to clinics, Phytochem Rev 16, 333-353. 8. Mein, K. (1833) Ueber die Darstellung des Atropins in weißen Krystallen, Ann. Pharm 6, 67-72. 9. Smulyan, H. (2018) The Beat Goes On: The Story of 5 Ageless Cardiac Drugs, Am J Med Sci. 10. Cieri, U. R. (2005) Identification and estimation of the levo isomer in raw materials and finished products containing atropine and/or hyoscyamine, J AOAC Int 88, 1-4. 11. Griffin, W. J., and Lin, G. D. (2000) Chemotaxonomy and geographical distribution of tropane alkaloids, Phytochemistry 53, 623-637. 12. He, Y., Luo, J., and Kong, L. (2011) Preparative separation of atropine and scopolamine from Daturae metelis Flos using pH-zone-refining counter-current chromatography with counter-rotation and dual-mode elution procedure, Journal of separation science 34, 806-811. 13. Cardillo, A. B., Perassolo, M., Sartuqui, M., Talou, J. R., and Giulietti, A. M. (2017) Production of tropane alkaloids by biotransformation using recombinant Escherichia coli whole cells, Biochemical Engineering Journal 125, 180-189. 14. Wang, J., Li, J.-l., Li, J., Li, J.-x., Liu, S.-j., Huang, L.-q., and Gao, W.-y. (2017) Production of Active Compounds in Medicinal Plants: From Plant Tissue Culture to Biosynthesis, Chinese Herbal Medicines 9, 115125. 15. Xia, K., Liu, X., Zhang, Q., Qiang, W., Guo, J., Lan, X., Chen, M., and Liao, Z. (2016) Promoting scopolamine biosynthesis in transgenic Atropa belladonna plants with pmt and h6h overexpression under field conditions, Plant Physiology and Biochemistry 106, 46-53. 16. Cordell, G. A. (2013) Fifty years of alkaloid biosynthesis in Phytochemistry, Phytochemistry 91, 29-51. 17. Philipov, S., and Doncheva, T. (2013) Alkaloids derived from ornithine: tropane alkaloids. In Natural Products pp 343-358, Springer. 18. Talapatra, S. K., and Talapatra, B. (2015) Chemistry of plant natural products, Springer. 19. Kraut, K. (1863) Ueber das Atropin, Justus Liebigs Annalen der Chemie 128, 280-285. 20. Willstätter, R. (1901) Synthesen in der Tropingruppe. I. Synthese des Tropilidens, Justus Liebigs Annalen der Chemie 317, 204-265. 21. Robinson, R. (1917) LXIII.—A synthesis of tropinone, Journal of the Chemical Society, Transactions 111, 762-768. 22. Iida, H., Watanabe, Y., and Kibayashi, C. (1985) A new synthetic route to tropane alkaloids based on [4+ 2] nitroso cycloaddition to 1, 3-cycloheptadienes, The Journal of Organic Chemistry 50, 1818-1825. 23. Nocquet, P. A., and Opatz, T. (2016) Total Synthesis of (±)-Scopolamine: Challenges of the Tropane Ring, European Journal of Organic Chemistry 2016, 1156-1164. 24. Pollini, G. P., Benetti, S., De Risi, C., and Zanirato, V. (2006) Synthetic approaches to enantiomerically pure 8-azabicyclo [3.2. 1] octane derivatives, Chemical reviews 106, 2434-2454. 25. Bédard, A.-C., Longstreet, A. R., Britton, J., Wang, Y., Moriguchi, H., Hicklin, R. W., Green, W. H., and Jamison, T. F. (2017) Minimizing E-factor in the continuous-flow synthesis of diazepam and atropine, Bioorganic & medicinal chemistry 25, 6233-6241. ACS Paragon Plus Environment
29
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 30 of 38
26. Dai, C., Snead, D. R., Zhang, P., and Jamison, T. F. (2015) Continuous-flow synthesis and purification of atropine with sequential in-line separations of structurally similar impurities, Journal of Flow Chemistry 5, 133-138. 27. Phillips, R. (1952) Benzhydryl ethers of tropines and their production, US Pat 2. 28. Sittig, M. (1988) Pharmaceutical manufacturing encyclopedia, Noyes Publications. 29. John, W. (1955) Benzhydryl ethers of tropine and processes of preparation, Google Patents. 30. Pedersen, H., Sinning, S., Bülow, A., Wiborg, O., Falborg, L., and Bols, M. (2004) Combinatorial synthesis of benztropine libraries and their evaluation as monoamine transporter inhibitors, Organic & biomolecular chemistry 2, 2861-2869. 31. Stockdale, T. P., and Williams, C. M. (2015) Pharmaceuticals that contain polycyclic hydrocarbon scaffolds, Chemical Society Reviews 44, 7737-7763. 32. Wilfrid, K. (1957) Amino alcohols substituted by bicycloalkyl residues and a process of making same, Google Patents. 33. WANG., K. X. L. L. Y. (2016) Novel biperiden hydrochloride preparation method, China. 34. Spiegelberg, G. R.-B. a. H. (1955) SUBSTITUTED TROPIC ACID N-(γ-PICOLYL)-AMIDES AND PROCESS FOR THE MANUFACTURE THEREOF, US. 35. Gerald Rey-Bellet, H. S. (1953) Verfahren zur Herstellung von substituierten Tropasäure-N-(γ-picolyl)amiden, Deutschland. 36. WEIYAN, X. Y. Z. Q. Z. H. X. X. L. G. Z. (2018) Synthetic method of tropicamide, China. 37. Dei, S., Bellucci, C., Ghelardini, C., Romanelli, M. N., and Spampinato, S. (1996) Synthesis, characterization and pharmacological profile of tropicamide enantiomers, Life sciences 58, 2147-2153. 38. Wayne, R. A. (1954) Substituted tertiary-aminoalkyl carbinols, Google Patents. 39. Wallace, A. D., and Samuel, W. (1954) Catalytic reduction of diphenyl alkanolamines and resulting products, Google Patents. 40. Ruddy, A. W., and Buckley Jr, J. S. (1950) Antispasmodics. N-(3-Phenylpropyl)-amines and 3-Amino-1phenyl-1-propanols1, Journal of the American Chemical Society 72, 718-721. 41. Joseph, D. J. (1955) Basic tertiary piperidino alcohols, Google Patents. 42. Adamson, D., and Billinghurst, J. (1950) 212. Aminoalkyl tertiary carbinols and derived products. Part III. 3-Tertiary-amino-1-aryl-1-(2-pyridyl)-propan-1-ols and-prop-1-enes, Journal of the Chemical Society (Resumed), 1039-1045. 43. Pfanz, H., and Jassmann, E. (1958) Beiträge zur Synthese spasmolytisch wirksamer Verbindungen. II. Mitteilung: Über neue Darstellungsverfahren für tertiäre Aminoalkohole, Archiv der Pharmazie 291, 36-44. 44. Ukaji, Y., Yamamoto, K., Fukui, M., and Fujisawa, T. (1991) Stereodivergent synthesis of optically active tertiary alcohols via addition reaction of chiral 2-acyl oxazolidine with organometallics, Tetrahedron letters 32, 2919-2922. 45. Schjelderup, L., Harbitz, O., Groth, P., and Aasen, A. J. (1987) Syntheses of (S)-(+)-trihexyphenidyl hydrochloride and (S)-(+)-procyclidine hydrochloride, two anticholinergics, using (S)-(-)-3-cyclohexyl-3hydroxy-3-phenylpropanoic acid as chiral synthon, Acta Chem. Scand. 41, 356. 46. Wallace, A. D. (1959) Method of treating parkinsonism, Google Patents. 47. Pearn, J., and Thearle, J. (1982) The history of hyoscine, Hist Sci Med 17, 257-261. 48. Huizinga, E. (1971) Murder through the ear, ORL 33, 361-365. 49. Kukula-Koch, W. A., and Widelski, J. (2017) Alkaloids, Pharmacognosy: Fundamentals, Applications and Strategies, 163-198. 50. Benítez, G., March-Salas, M., Villa-Kamel, A., Cháves-Jiménez, U., Hernández, J., Montes-Osuna, N., Moreno-Chocano, J., and Cariñanos, P. (2018) The genus Datura L. (Solanaceae) in Mexico and Spain – Ethnobotanical perspective at the interface of medical and illicit uses, Journal of Ethnopharmacology 219, 133-151. 51. Malik, K., Ahmad, M., Zhang, G. L., Rashid, N., Zafar, M., Sultana, S., and Shah, S. N. (2018) Traditional plant based medicines used to treat musculoskeletal disorders in Northern Pakistan, Eur J Integr Med 19, 1764. 52. Müller, J. L. (1998) Love potions and the ointment of witches: historical aspects of the nightshade alkaloids, Journal of Toxicology: Clinical Toxicology 36, 617-627. 53. Kwakye, G. F., Jimenez, J., Jimenez, J. A., and Aschner, M. (2018) Atropa belladonna neurotoxicity: Implications to neurological disorders, Food Chem Toxicol 116, 346-353. 54. Forbes, T. R. (1977) Why is it called 'beautiful lady'? A note on belladonna, Bull N Y Acad Med 53, 403406. ACS Paragon Plus Environment
30
Page 31 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
55. Lee, M., Dukan, E., and Milne, I. (2018) History & Humanities, JR Coll Physicians Edinb 48, 85-91. 56. Sanger, G. J., and Andrews, P. L. (2018) A History of Drug Discovery for Treatment of Nausea and Vomiting and the Implications for Future Research, Frontiers in pharmacology 9. 57. Welliver, M. (2015) Consideration of Acetylcholine Antagonists for Nausea and Vomiting, Gastroenterol Nurs 38, 319-321. 58. Baciarello, M., Cornini, A., Zasa, M., Pedrona, P., Scrofani, G., Venuti, F. S., and Fanelli, G. (2011) Intrathecal atropine to prevent postoperative nausea and vomiting after Cesarean section: a randomized, controlled trial, Minerva Anestesiol 77, 781-788. 59. Boeke, A. J., de Lange, J. J., van Druenen, B., and Langemeijer, J. J. (1994) Effect of antagonizing residual neuromuscular block by neostigmine and atropine on postoperative vomiting, Br J Anaesth 72, 654-656. 60. Isaacs, B. (1956) The influence of hyoscine and atropine on apomorphine-induced vomiting in man, Clin Sci 15, 177-182. 61. Volgin, A. D., Yakovlev, O. V., Demin, K. A., Alekseeva, P. A., Kyzar, E., Collins, C., Nichols, D. E., and Kalueff, A. (2018) Understanding CNS effects of deliriant hallucinogenic drugs through experimental animal models, ACS Chem Neurosci. 62. Ali, S., Patel, M., Jabeen, S., Bailey, R. K., Patel, T., Shahid, M., Riley, W. J., and Arain, A. (2011) Insight into delirium, Innov Clin Neurosci 8, 25-34. 63. Alizadeh, A., Moshiri, M., Alizadeh, J., and Balali-Mood, M. (2014) Black henbane and its toxicity - a descriptive review, Avicenna J Phytomed 4, 297-311. 64. Janowsky, D. S., and Overstreet, D. H. (1990) Cholinergic dysfunction in depression, Pharmacol Toxicol 66 Suppl 3, 100-111. 65. Furey, M. L., and Drevets, W. C. (2006) Antidepressant efficacy of the antimuscarinic drug scopolamine: a randomized, placebo-controlled clinical trial, Arch Gen Psychiatry 63, 1121-1129. 66. Drevets, W. C., and Furey, M. L. (2010) Replication of scopolamine's antidepressant efficacy in major depressive disorder: a randomized, placebo-controlled clinical trial, Biol Psychiatry 67, 432-438. 67. Zhang, X. C., Farrell, N., Haronian, T., and Hack, J. (2017) Postoperative Anticholinergic Poisoning: Concealed Complications of a Commonly Used Medication, J Emerg Med 53, 520-523. 68. Zhang, X. C., Farrell, N., Haronian, T., and Hack, J. (2017) Postoperative Anticholinergic Poisoning: Concealed Complications of a Commonly Used Medication, The Journal of emergency medicine 53, 520-523. 69. Duran, C. E., Azermai, M., and Vander Stichele, R. H. (2013) Systematic review of anticholinergic risk scales in older adults, Eur J Clin Pharmacol 69, 1485-1496. 70. Gillin, J. C., Sutton, L., Ruiz, C., Darko, D., Golshan, S., Risch, S. C., and Janowsky, D. (1991) The effects of scopolamine on sleep and mood in depressed patients with a history of alcoholism and a normal comparison group, Biol Psychiatry 30, 157-169. 71. Furey, M. L., Khanna, A., Hoffman, E. M., and Drevets, W. C. (2010) Scopolamine produces larger antidepressant and antianxiety effects in women than in men, Neuropsychopharmacology 35, 2479-2488. 72. Furey, M. L., and Drevets, W. C. (2006) Antidepressant efficacy of the antimuscarinic drug scopolamine: a randomized, placebo-controlled clinical trial, Archives of general psychiatry 63, 1121-1129. 73. Thomas, T., Benjamin, A., and Dooley, T. (2017) Treatment of Acute Anxiety Episodes in Patients using a Fast-Acting Beta Blocker–Scopolamine Combination Drug, Annals of Depression and Anxiety 4, 1088. 74. Volgin, A. D., Yakovlev, O. A., Demin, K. A., Alekseeva, P. A., and Kalueff, A. V. (2018) Acute behavioral effects of deliriant hallucinogens atropine and scopolamine in adult zebrafish, Behavioural Brain Research. 75. Budzynska, B., Boguszewska-Czubara, A., Kruk-Slomka, M., Skalicka-Wozniak, K., Michalak, A., Musik, I., and Biala, G. (2015) Effects of imperatorin on scopolamine-induced cognitive impairment and oxidative stress in mice, Psychopharmacology (Berl) 232, 931-942. 76. Klinkenberg, I., and Blokland, A. (2010) The validity of scopolamine as a pharmacological model for cognitive impairment: a review of animal behavioral studies, Neurosci Biobehav Rev 34, 1307-1350. 77. Araujo, J. A., Chan, A. D., Winka, L. L., Seymour, P. A., and Milgram, N. W. (2004) Dose-specific effects of scopolamine on canine cognition: impairment of visuospatial memory, but not visuospatial discrimination, Psychopharmacology (Berl) 175, 92-98. 78. Luyten, L., Nuyts, S., and Beckers, T. (2017) Low-dose systemic scopolamine disrupts context conditioning in rats, Journal of Psychopharmacology 31, 667-673. 79. Smythe, J. W., Murphy, D., Bhatnagar, S., Timothy, C., and Costall, B. (1996) Muscarinic antagonists are anxiogenic in rats tested in the black-white box, Pharmacology Biochemistry and Behavior 54, 57-63.
ACS Paragon Plus Environment
31
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 32 of 38
80. Ten Berge, R. E., Roffel, A. F., and Zaagsma, J. (1993) The interaction of selective and non-selective antagonists with pre-and postjunctional muscarinic receptor subtypes in the guinea pig trachea, European journal of pharmacology 233, 279-284. 81. Wood, M. J., and Thompson, J. P. (2011) Drugs affecting the autonomic nervous system, Anaesthesia & Intensive Care Medicine 12, 73-77. 82. Kovacic, P. (2003) Mechanism of organophosphates (nerve gases and pesticides) and antidotes: electron transfer and oxidative stress, Current medicinal chemistry 10, 2705-2709. 83. Bardin, P. G., van Eeden, S. F., Moolman, J. A., Foden, A. P., and Joubert, J. R. (1994) Organophosphate and carbamate poisoning, Archives of internal medicine 154, 1433-1441. 84. Grob, D., and Harvey, A. (1953) The effects and treatment of nerve gas poisoning, The American journal of medicine 14, 52-63. 85. Sungur, M., and Güven, M. (2001) Intensive care management of organophosphate insecticide poisoning, Critical care 5, 211. 86. Rafiq, S., Ahmad, S., Ahmed, F., Batool, Z., Ahmed, S. B., Saleem, S., Naqvi, F., Liaquat, L., Afzal, A., and Haider, S. (2018) Anticholinergic drug atropine diminishes newly formed fear memory in male rats, Pakistan journal of pharmaceutical sciences 31. 87. Lochner, M., and Thompson, A. J. (2016) The muscarinic antagonists scopolamine and atropine are competitive antagonists at 5-HT3 receptors, Neuropharmacology 108, 220-228. 88. Seyedabadi, M., Fakhfouri, G., Ramezani, V., Mehr, S. E., and Rahimian, R. (2014) The role of serotonin in memory: interactions with neurotransmitters and downstream signaling, Experimental brain research 232, 723-738. 89. Ukairo, O. T., Bondi, C. D., Newman, A. H., Kulkarni, S. S., Kozikowski, A. P., Pan, S., and Surratt, C. K. (2005) Recognition of benztropine by the dopamine transporter (DAT) differs from that of the classical dopamine uptake inhibitors cocaine, methylphenidate, and mazindol as a function of a DAT transmembrane 1 aspartic acid residue, Journal of Pharmacology and Experimental Therapeutics 314, 575-583. 90. Hai, N. T., Kim, J., Park, E.-S., and Chi, S.-C. (2008) Formulation and biopharmaceutical evaluation of transdermal patch containing benztropine, International journal of pharmaceutics 357, 55-60. 91. Kulkarni, S. S., Kopajtic, T. A., Katz, J. L., and Newman, A. H. (2006) Comparative structure-activity relationships of benztropine analogues at the dopamine transporter and histamine H(1) receptors, Bioorg Med Chem 14, 3625-3634. 92. Woolverton, W. L., Hecht, G. S., Agoston, G. E., Katz, J. L., and Newman, A. H. (2001) Further studies of the reinforcing effects of benztropine analogs in rhesus monkeys, Psychopharmacology 154, 375-382. 93. Newman, A. H., and Kulkarni, S. (2002) Probes for the dopamine transporter: New leads toward a cocaine-abuse therapeutic—A focus on analogues of benztropine and rimcazole, Medicinal research reviews 22, 429-464. 94. Myhrer, T., Enger, S., and Aas, P. (2008) Anticonvulsant efficacy of drugs with cholinergic and/or glutamatergic antagonism microinfused into area tempestas of rats exposed to soman, Neurochemical research 33, 348-354. 95. Donald, F. K. (1966) Chlorpromazine-procyclidine combination, imipramine and placebo in depressive disorders, Canadian Psychiatric Association Journal 11, 146-149. 96. Cho, S., Park, W., Cho, Y., Lee, S., Kim, H., Kim, Y., Seong, Y., Hur, G., Cheon, K., and Kim, W. (2004) Effect of Physostigmine and Procyclidine, as a Combinational Prophylactic Regimen for Organophosphate Poisoning on Learning and Memory Process in Rats, The Korean Journal of Laboratory Animal Science. 97. Frith, C. D. (1984) Schizophrenia, memory, and anticholinergic drugs, Journal of Abnormal Psychology 93, 339. 98. Ashton, H. (2002) Delirium and hallucinations, ADVANCES IN CONSCIOUSNESS RESEARCH 36, 181-204. 99. Smolen, V. F., and Schoenwald, R. D. (1971) Drug-absorption analysis from pharmacological data I: Method and confirmation exemplified for the mydriatic drug tropicamide, Journal of pharmaceutical sciences 60, 96-103. 100. Spagnolo, P. A., Badiani, A., and Nencini, P. (2013) Polydrug abuse by intravenous use of heroin and tropicamide-containing eyedrops, Clinical neuropharmacology 36, 100-101. 101. Bozkurt, M., Karabulut, V., Evren, C., Seker, M., and Kan, H. (2015) Intravenous abuse of tropicamide in opioid use disorder: presentation of 2 cases, Substance abuse 36, 170-173. 102. Betz, A. J., McLaughlin, P. J., Burgos, M., Weber, S. M., and Salamone, J. D. (2007) The muscarinic receptor antagonist tropicamide suppresses tremulous jaw movements in a rodent model of parkinsonian tremor: possible role of M4 receptors, Psychopharmacology 194, 347-359. ACS Paragon Plus Environment
32
Page 33 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
103. Roldán, G., Bolaños-Badillo, E., González-Sánchez, H., Quirarte, G. L., and Prado-Alcalá, R. A. (1997) Selective M1 muscarinic receptor antagonists disrupt memory consolidation of inhibitory avoidance in rats, Neuroscience letters 230, 93-96. 104. Wezenberg, E., Verkes, R. J., Sabbe, B., Ruigt, G., and Hulstijn, W. (2005) Modulation of memory and visuospatial processes by biperiden and rivastigmine in elderly healthy subjects, Psychopharmacology 181, 582-594. 105. Klinkenberg, I., and Blokland, A. (2011) A comparison of scopolamine and biperiden as a rodent model for cholinergic cognitive impairment, Psychopharmacology 215, 549-566. 106. Sambeth, A., Riedel, W. J., Klinkenberg, I., Kähkönen, S., and Blokland, A. (2015) Biperiden selectively induces memory impairment in healthy volunteers: no interaction with citalopram, Psychopharmacology 232, 1887-1897. 107. Vingerhoets, C., Bakker, G., van Dijk, J., Bloemen, O. J., Wang, Y., Chan, R. C., Booij, J., and van Amelsvoort, T. A. (2017) The effect of the muscarinic M1 receptor antagonist biperiden on cognition in medication free subjects with psychosis, European Neuropsychopharmacology 27, 854-864. 108. Ogino, S., Miyamoto, S., Tenjin, T., Kitajima, R., Ojima, K., Miyake, N., Funamoto, Y., Arai, J., Tsukahara, S., and Ito, Y. (2011) Effects of discontinuation of long-term biperiden use on cognitive function and quality of life in schizophrenia, Progress in Neuro-Psychopharmacology and Biological Psychiatry 35, 78-83. 109. Guthrie, S. K., Manzey, L., Scott, D., Giordani, B., and Tandon, R. (2000) Comparison of central and peripheral pharmacologic effects of biperiden and trihexyphenidyl in human volunteers, Journal of Clinical Psychopharmacology 20, 77-83. 110. Jilani, T. N., and Sharma, S. (2018) Trihexyphenidyl. In StatPearls [Internet] StatPearls Publishing. 111. Azmi, A. N., and Desrini, S. (2016) A cross sectional study of trihexyphenidyl utilization on patient receiving antipsychotic therapy, JKKI: Jurnal Kedokteran dan Kesehatan Indonesia 7, 102-108. 112. Koller, W. C. (1984) Disturbance of recent memory function in parkinsonian patients on anticholinergic therapy, Cortex 20, 307-311. 113. Muhtadi, F. J., Analytical Profiles of Drug Substances and Excipients vol. 23: Hyoscyamine, 1994, Academic Press. 114. Lin, Y.-G., Chen, P.-H., Chang, F.-Y., Wu, L.-T., Liao, K.-Y., and Wu, T.-C. (2011) Delirium due to scopolamine patch in a 4-year-old boy, Journal of the Formosan Medical Association 110, 208-211. 115. Kwakye, G. F., Jiménez, J., Jiménez, J. A., and Aschner, M. (2018) Atropa belladonna neurotoxicity: implications to neurological disorders, Food and Chemical Toxicology. 116. John, H., Rychlik, M., Thiermann, H., and Schmidt, C. (2018) Simultaneous quantification of atropine and scopolamine in infusions of herbal tea and Solanaceae plant material by matrix-assisted laser desorption/ionization time-of-flight (tandem) mass spectrometry, Rapid Communications in Mass Spectrometry. 117. Brown JH, T. P. (2003) Agonists and antagonists of mucarinic receptors. In: Hardman JG, Limbird LE, Goodman, Gilman A,editors. Pharmacological basis of therapeutic, Pharmacological basis of therapeuticб 10 ed., New York: McGraw-Hill, 162–181. 118. Pillay, P. (2005) Modern Medical Toxicology, Jaypee Brothers Medical Publishers. 119. Faivre, A., Mounier, C., Gaillard, T., Alla, P., and Goutorbe, P. (2012) Intoxication grave à l’atropine mimant une urgence neurovasculaire, Revue Neurologique 168, 450-453. 120. Putcha, L., Tietze, K. J., Bourne, D. W., Parise, C. M., Hunter, R. P., and Cintrón, N. M. (1996) Bioavailability of intranasal scopolamine in normal subjects, Journal of pharmaceutical sciences 85, 899-902. 121. Muhtadi, F. J. (1994) Hyoscyamine. In Analytical Profiles of Drug Substances and Excipients pp 153-228, Elsevier. 122. Bosman, I., Douma, W., Ensing, K., and De Zeeuw, R. (1997) A semi-automated solid-phase extraction and radioreceptor assay for the analysis of scopolamine in urine and plasma, European journal of pharmaceutical sciences 5, 315-325. 123. Verma, P. S. (1986) Sensitive radioimmunoassay using antibody to L-hyoscyamine, Google Patents. 124. Boumba, V. A., Mitselou, A., and Vougiouklakis, T. (2004) Fatal poisoning from ingestion of Datura stramonium seeds, Veterinary and human toxicology 46, 81-82. 125. Ricard, F., Abe, E., Duverneuil-Mayer, C., Charlier, P., de la Grandmaison , G., & Alvarez,. (2012) Datura stramonium chronic exposure. Forensic science international, Forensic science international, 223 (221-223), 256-260. 126. Lusthof, K., Bosman, I., Kubat, B., and Vincenten-van Maanen, M. (2017) Toxicological results in a fatal and two non-fatal cases of scopolamine-facilitated robberies, Forensic science international 274, 79-82. ACS Paragon Plus Environment
33
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 34 of 38
127. Brocks, D. R. (1999) Anticholinergic drugs used in Parkinson’s disease: an overlooked class of drugs from a pharmacokinetic perspective, J Pharm Pharm Sci 2, 39-46. 128. He, H., McKay, G., and Midha, K. K. (1993) Development of a sensitive and specific radioimmunoassay for benztropine, Journal of pharmaceutical sciences 82, 1027-1032. 129. Owen, J. A., Sribney, M., Lawson, J., Delva, N., and Letemendia, F. (1989) Capillary gas chromatography of trihexyphenidyl, procyclidine and cycrimine in biological fluids, Journal of Chromatography B: Biomedical Sciences and Applications 494, 135-142. 130. Desage, M., Rousseau-Tsangaris, M., Lecompte, D., and Brazier, J. (1991) Quantitation of trihexyphenidyl from plasma using a mass-selective detector and electron-impact ionization, Journal of Chromatography B: Biomedical Sciences and Applications 571, 250-256. 131. Grimaldi, R., Perucca, E., Ruberto, G., Gelmi, C., Trimarchi, F., Hollmann, M., and Crema, A. (1986) Pharmacokinetic and pharmacodynamic studies following the intravenous and oral administration of the antiparkinsonian drug biperiden to normal subjects, European journal of clinical pharmacology 29, 735-737. 132. Whiteman, P., Fowle, A., Hamilton, M., Peck, A., Bye, A., Dean, K., and Webster, A. (1985) Pharmacokinetics and pharmacodynamics of procyclidine in man, European journal of clinical pharmacology 28, 73-78. 133. Yokagawa, K., Nakashima, E., Ishizaki, J., Hasegawa, M., Kido, H., and Ichimura, F. (1992) Brain regional pharmacokinetics of biperiden in rats, Biopharmaceutics & drug disposition 13, 131-140. 134. Nakashima, E., Yokogawa, K., Ichimura, F., Hashimoto, T., Yamana, T., and Tsuji, A. (1987) Effects of fasting on biperiden pharmacokinetics in the rat, Journal of pharmaceutical sciences 76, 10-13. 135. Bersani, F. S., Corazza, O., Simonato, P., Mylokosta, A., Levari, E., Lovaste, R., and Schifano, F. (2013) Drops of madness? Recreational misuse of tropicamide collyrium; early warning alerts from Russia and Italy, General Hospital Psychiatry 35, 571-573. 136. VUORI, M.-L., KAILA, T., IISALO, E., and SAARI, K. M. (1994) Systemic absorption and anticholinergic activity of topically applied tropicamide, Journal of Ocular Pharmacology and Therapeutics 10, 431-437. 137. Shormanov, V., Pravdyuk, M., Cheked, M., and Barannikova, V. (2016) The distribution of tropicamide in the body of warm-blooded animals after its intragastric administration, Sudebno-meditsinskaia ekspertiza 59, 27-34. 138. Schultes, R. E., and Hofmann, A. (1980) Pflanzen der Götter die magischen Kräfte der Rausch-und Giftgewächse;[mit Pflanzenlexikon und Bildatlas], Hallwag. 139. FUHNER, H. (1925) Solanazeen als Berauschungsmittel. Eine historisch-ethnologische Studie, Archiv fur. 140. Fink, M., Klein, D. F., and Kramer, J. C. (1965) Clinical efficacy of chlorpromazine-procyclidine combination, imipramine and placebo in depressive disorders, Psychopharmacologia 7, 27-36. 141. Goldsmith, S. R., Frank, I., and Ungerleider, J. T. (1968) Poisoning from ingestion of a stramoniumbelladonna mixture: flower power gone sour, JAMA 204, 169-170. 142. Affaticati, A., Gerra, M. L., Amerio, A., Inglese, M., Antonioni, M. C., and Marchesi, C. (2015) The Controversial Case of Biperiden: From Prescription Drug to Drug of Abuse, Journal of clinical psychopharmacology 35, 749-750. 143. Caplan, J. P., Epstein, L. A., Quinn, D. K., Stevens, J. R., and Stern, T. A. (2007) Neuropsychiatric effects of prescription drug abuse, Neuropsychology review 17, 363-380. 144. Clancy, D. (2016) Potential abuse from unexpected sources (Part 2), Australian Pharmacist 35, 67. 145. Ramos, A., Dieckmann, L., and Galduróz, J. (2017) Biperiden in the Treatment of Cocaine/Crack Dependence: Clinical Perspectives. In The Neuroscience of Cocaine pp 655-662, Elsevier. 146. Jalali, F., Afshari, R., and Babaei, A. (2014) Smoking crushed hyoscine/scopolamine tablets as drug abuse, Substance use & misuse 49, 793-797. 147. Capasso, A., and De Feo, V. (2002) Central nervous system pharmacological effects of plants from Northern Peruvian Andes: Valeriana adscendens, Iresine herbstii and Brugmansia arborea, Pharm Biol 40, 274-293. 148. Milhorn, H. T. (2018) Hallucinogen Dependence. In Substance Use Disorders: A Guide for the Primary Care Provider pp 167-177, Springer International Publishing, Cham. 149. Nikolaou, P., Papoutsis, I., Stefanidou, M., Dona, A., Maravelias, C., Spiliopoulou, C., and Athanaselis, S. (2012) Accidental poisoning after ingestion of "aphrodisiac" berries: diagnosis by analytical toxicology, J Emerg Med 42, 662-665. 150. Wallace, M., and Safer, M. (2009) Hypersexuality among cognitively impaired older adults, Geriatric Nursing 30, 230-237. ACS Paragon Plus Environment
34
Page 35 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
151. Bechtel, L. K., and Holstege, C. P. (2007) Criminal poisoning: drug-facilitated sexual assault, Emergency medicine clinics of North America 25, 499-525. 152. Assault, D. F. S. (2015) DRUG FACILITATED SEXUAL ASSAULT, Sexual Offending: Predisposing Antecedents, Assessments and Management, 67. 153. King, L., QC, R. F., Ujvary, I., Ramsey, J., and Nutt, D. (2014) Scopolamine: Useful medicine or dangerous drug?, Science and Justice 54, 321-322. 154. Series, H., and Degano, P. (2005) Hypersexuality in dementia, Advances in Psychiatric Treatment 11, 424-431. 155. Floody, O. R., and Lusk, L. G. (2013) Responses to central oxotremorine and scopolamine support the cholinergic control of male mating behavior in hamsters, Pharmacol Biochem Behav 105, 1-11. 156. Dohanich, G. P., Daniel, J. M., Fader, A. J., Wolff, S. C., Gallogly, P. M., and Overstreet, D. M. (1998) Sexual behavior of Flinders Line female rats bred for differential cholinergic sensitivities, Horm Behav 33, 7784. 157. Lynch, C. S., Ruppel, T. E., Domingue, R. N., and Ponthier, G. L. (1999) Scopolamine inhibition of female sexual behavior in rhesus monkeys (Macaca mulatta), Pharmacol Biochem Behav 63, 655-661. 158. Wee, B. E., Francis, T. J., Lee, C. Y., Lee, J. M., and Dohanich, G. P. (1995) Mate preference and avoidance in female rats following treatment with scopolamine, Physiol Behav 58, 97-100. 159. Leavitt, F. I. (1969) Drug-induced modifications in sexual behavior and open field locomotion of male rats. 160. Sanz, C., Zamberlan, F., Erowid, E., and Tagliazucchi, E. (2018) The experience elicited by hallucinogens presents the highest similarity to dreaming within a large database of psychoactive substance reports, Frontiers in neuroscience 12, 7. 161. Battaglia, J. (2005) Pharmacological management of acute agitation, Drugs 65, 1207-1222. 162. Kaminer, Y., Munitz, H., and Wijsenbeek, H. (1982) Trihexyphenidyl (Artane) abuse: euphoriant and anxiolytic, The British Journal of Psychiatry 140, 473-474. 163. Ramírez, J., Piñero, J., and Baptista, T. (2017) Treating ‘Devil ׳s Breath ׳intoxication: Use of rivastigmine in six patients with toxic psychoses due to non pharmaceutical scopolamine, European Neuropsychopharmacology 27, 833-834. 164. Ham, S., Kim, T. K., Chung, S., and Im, H.-I. (2017) Drug abuse and psychosis: new insights into druginduced psychosis, Experimental neurobiology 26, 11-24. 165. Weinstein, M. R., and Fischer, A. (1972) Benztropine toxicity and atropine psychosis, JAMA 220, 16161617. 166. Bergman, K. R., Pearson, C., Waltz, G. W., and Evans, R. (1980) Atropine-induced psychosis: an unusual complication of therapy with inhaled atropine sulfate, Chest 78, 891-893. 167. Ziskind, A. A. (1988) Transdermal scopolamine—induced psychosis, Postgraduate medicine 84, 73-76. 168. Minagar, A., Shulman, L. M., and Weiner, W. J. (1999) Transderm-induced psychosis in Parkinson’s disease, Neurology 53, 433-433-a. 169. Birkhimer, L., Jacobson, P., Olson, J., and Goyette, D. (1984) Ocular scopolamine-induced psychosis, The Journal of family practice 18, 464, 466, 469. 170. Marneros, A., Gutmann, P., and Uhlmann, F. (2006) Self-amputation of penis and tongue after use of Angel’s Trumpet, European archives of psychiatry and clinical neuroscience 256, 458-459. 171. Göpel, C., Laufer, C., and Marcus, A. (2002) Three cases of angel's trumpet tea-induced psychosis in adolescent substance abusers, Nordic Journal of Psychiatry 56, 49-52. 172. Babu, S., Chandrika, D., and Kulkarni, M. (2016) Plant poisoning—an observational study in a tristate region emergency department, International Journal of Medical Science and Public Health 5, 2158-2161. 173. Drevets, W. C., Zarate Jr, C. A., and Furey, M. L. (2013) Antidepressant effects of the muscarinic cholinergic receptor antagonist scopolamine: a review, Biological psychiatry 73, 1156-1163. 174. LI, Y.-h., and TIAN, S.-w. (2014) Progress in research of the antidepressant effect of scopolamine, Chinese Journal of New Drugs 21, 014. 175. Pae, C.-U. (2014) The Potential Role of Scopolamine as Antidepressant for Mood Disorder, Clinical neuropharmacology 37, 67-68. 176. Duman, R. S., Aghajanian, G. K., Sanacora, G., and Krystal, J. H. (2016) Synaptic plasticity and depression: new insights from stress and rapid-acting antidepressants, Nat Med 22, 238-249. 177. Czeh, B., Fuchs, E., Wiborg, O., and Simon, M. (2016) Animal models of major depression and their clinical implications, Prog Neuropsychopharmacol Biol Psychiatry 64, 293-310. ACS Paragon Plus Environment
35
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 36 of 38
178. Rahmati, B., Kiasalari, Z., Roghani, M., Khalili, M., and Ansari, F. (2017) Antidepressant and anxiolytic activity of Lavandula officinalis aerial parts hydroalcoholic extract in scopolamine-treated rats, Pharmaceutical biology 55, 958-965. 179. Aydin, E., Hritcu, L., Dogan, G., Hayta, S., and Bagci, E. (2016) The effects of inhaled Pimpinella peregrina essential oil on scopolamine-induced memory impairment, anxiety, and depression in laboratory rats, Molecular neurobiology 53, 6557-6567. 180. Gazdag, G., Bitter, I., Ungvári, G., and Gerevich, J. (2005) Atropine coma: a historical note, The journal of ECT 21, 203-206. 181. Volgin, A. D., Yakovlev, O. V., Demin, K. A., Alekseeva, P. A., Kyzar, E., Collins, C., Nichols, D. E., and Kalueff, A. (2018) Understanding CNS effects of deliriant hallucinogenic drugs through experimental animal models, ACS chemical neuroscience. 182. Lipowski, Z. (1987) Delirium (acute confusional states), Jama 258, 1789-1792. 183. Liptzin, B., and Levkoff, S. E. (1992) An empirical study of delirium subtypes, The British Journal of Psychiatry 161, 843-845. 184. Caraceni, A., and Grassi, L. (2011) Delirium: acute confusional states in palliative medicine, Oxford University Press. 185. Tamura, Y., Chiba, S., Takasaki, H., Tabata, K., Ishimaru, Y., and Ishimoto, T. (2006) Biperiden-induced delirium model in rats: a behavioral and electroencephalographic study, Brain research 1115, 194-199. 186. Trzepacz, P. (2000) Is there a final common neural pathway in delirium? Focus on acetylcholine and dopamine, In Seminars in clinical neuropsychiatry, pp 132-148. 187. Meagher, D. J. (2001) Regular review: Delirium: optimising management, BMJ: British Medical Journal 322, 144. 188. RUDBERG, M. A., POMPEI, P., FOREMAN, M. D., ROSS, R. E., and CASSEL, C. K. (1997) The natural history of delirium in older hospitalized patients: a syndrome of heterogeneity, Age and Ageing 26, 169-174. 189. Wohleb, E. S., and Delpech, J.-C. (2017) Dynamic cross-talk between microglia and peripheral monocytes underlies stress-induced neuroinflammation and behavioral consequences, Progress in NeuroPsychopharmacology and Biological Psychiatry 79, 40-48. 190. Ma, L., Demin, K. A., Kolesnikova, T. O., Kharsko, S. L., Zhu, X., Yuan, X., Song, C., Meshalkina, D. A., Leonard, B. E., Tian, L., and Kalueff, A. V. (2017) Animal inflammation-based models of depression and their application to drug discovery, Expert Opin Drug Discov 12, 995-1009. 191. Van Gool, W. A., Van de Beek, D., and Eikelenboom, P. (2010) Systemic infection and delirium: when cytokines and acetylcholine collide, The Lancet 375, 773-775. 192. Berizzi, A. E., Gentry, P. R., Rueda, P., Den Hoedt, S., Sexton, P. M., Langmead, C. J., and Christopoulos, A. (2016) Molecular Mechanisms of Action of M5 Muscarinic Acetylcholine Receptor Allosteric Modulators, Mol Pharmacol 90, 427-436. 193. Carr, B. J., Mihara, K., Ramachandran, R., Saifeddine, M., Nathanson, N. M., Stell, W. K., and Hollenberg, M. D. (2018) Myopia-Inhibiting Concentrations of Muscarinic Receptor Antagonists Block Activation of Alpha2A-Adrenoceptors In Vitro, Invest Ophthalmol Vis Sci 59, 2778-2791. 194. Dulawa, S. C., and Janowsky, D. S. (2018) Cholinergic regulation of mood: from basic and clinical studies to emerging therapeutics, Mol Psychiatry. 195. Říčný, J., Gualtieri, F., and Tuček, S. (2002) Constitutive inhibitory action of muscarinic receptors on adenylyl cyclase in cardiac membranes and its stereospecific suppression by hyoscyamine, Physiol Res 51, 131-137. 196. Fields, R. D., Dutta, D. J., Belgrad, J., and Robnett, M. (2017) Cholinergic signaling in myelination, Glia 65, 687-698. 197. Svoboda, J., Popelikova, A., and Stuchlik, A. (2017) Drugs Interfering with Muscarinic Acetylcholine Receptors and Their Effects on Place Navigation, Front Psychiatry 8, 215. 198. Witkin, J. M., Overshiner, C., Li, X., Catlow, J. T., Wishart, G. N., Schober, D. A., Heinz, B. A., Nikolayev, A., Tolstikov, V. V., Anderson, W. H., Higgs, R. E., Kuo, M. S., and Felder, C. C. (2014) M1 and m2 muscarinic receptor subtypes regulate antidepressant-like effects of the rapidly acting antidepressant scopolamine, J Pharmacol Exp Ther 351, 448-456. 199. He, N., Mao, L. M., Sturich, A. W., Jin, D. Z., and Wang, J. Q. (2018) Inhibition of basal and amphetaminestimulated extracellular signal-regulated kinase (ERK) phosphorylation in the rat forebrain by muscarinic acetylcholine M4 receptors, Brain Res 1688, 103-112.
ACS Paragon Plus Environment
36
Page 37 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
ACS Chemical Neuroscience
200. Ztaou, S., Maurice, N., Camon, J., Guiraudie-Capraz, G., Kerkerian-Le Goff, L., Beurrier, C., Liberge, M., and Amalric, M. (2016) Involvement of Striatal Cholinergic Interneurons and M1 and M4 Muscarinic Receptors in Motor Symptoms of Parkinson's Disease, J Neurosci 36, 9161-9172. 201. Joseph, L., and Thomsen, M. (2017) Effects of muscarinic receptor antagonists on cocaine discrimination in wild-type mice and in muscarinic receptor M1, M2, and M4 receptor knockout mice, Behavioural brain research 329, 75-83. 202. Waelbroeck, M., Camus, J., Tastenoy, M., Lambrecht, G., Mutschler, E., Tacke, R., and Christophe, J. (1990) Stereoselectivity of procyclidine binding to muscarinic receptor subtypes M1, M2 and M4, Eur J Pharmacol 189, 135-142. 203. Soulaidopoulos, S., Sinakos, E., Dimopoulou, D., Vettas, C., Cholongitas, E., and Garyfallos, A. (2017) Anticholinergic syndrome induced by toxic plants, World J Emerg Med 8, 297-301. 204. Bozkurt, M., Karabulut, V., Evren, C., Seker, M., and Kan, H. (2015) Intravenous Abuse of Tropicamide in Opioid Use Disorder: Presentation of 2 Cases, Subst Abus 36, 170-173. 205. Kastrup, O., Eikmeier, G., and Gastpar, M. (1991) [Central anticholinergic intoxication syndrome. A contribution to the differential diagnosis of exogenous psychoses], Dtsch Med Wochenschr 116, 1748-1751. 206. Orhan, F. O., ŞENOĞLU, N., BAĞCIOĞLU, E., and DEMİRKAN, H. (2008) Central anticholinergic syndrome associated with atropine: case report, AJCI 2, 45-46. 207. Delberghe, X., and de Beyl, D. Z. (1987) Repeated delirium from homatropine eye-drops: A case report, Clinical neurology and neurosurgery 89, 53-54. 208. Fisher, C. M. (1991) Visual hallucinations on eye closure associated with atropine toxicity. A neurological analysis and comparison with other visual hallucinations, Canadian Journal of Neurological Sciences 18, 1827. 209. Rubinstein, J. S. (1979) Antiparkinsonian drug abuse: eight case reports, Psychiatric Services 30, 34-37. 210. Laski, E., and Taleporos, E. (1977) Anticholinergic psychosis in a bilingual: A case study, The American journal of psychiatry. 211. Ananth, J., and Jain, R. (1973) Benztropine psychosis, Canadian Psychiatric Association Journal 18, 409414. 212. Espi Martinez, F., Espi Forcen, F., Shapov, A., and Martinez Moya, A. (2012) Biperiden dependence: case report and literature review, Case reports in psychiatry 2012. 213. Affaticati, A., Gerra, M. L., Amerio, A., Inglese, M., Antonioni, M. C., and Marchesi, C. (2015) The Controversial Case of Biperiden: From Prescription Drug to Drug of Abuse, J Clin Psychopharmacol 35, 749750. 214. Kinay, D., and Soyata, A. Z. (2018) Biperiden-Induced Delirium In A Five-Years Old Child, Curr Drug Saf. 215. . 216. Mohamed, A., Alkhiyami, D., Rahhal, A., Del Castillo, M., Pichad, S. M., and Ahmad, M. (2018) Procyclidine overdose induced central and peripheral anti-cholinergic toxicity, Toxicology Communications 2, 42-44. 217. Simpson, N. (2003) Delirium in adults with intellectual disabilities and DC-LD, Journal of Intellectual Disability Research 47, 38-42. 218. Haleem, A., and Hag, S. E. A. (2014) The Central Anticholinergic Syndrome in a Three-Year-Old Girl by Accidental Ingestion of a High-Dose of Trihexyphenidyl= ﺛﻼﺙ ﺫﺍﺕ ﻃﻔﻠﺔ ﻓﻲ ﺍﻟﻜﻮﻟﻴﻨﻲ ﺍﻟﻔﻌﻞ ﻟﻤﻀﺎﺩﺍﺕ ﺍﻟﻤﺮﻛﺰﻳﺔ ﺍﻟﻤﺘﻼﺯﻣﺔ ﻓﻴﻨﻴﺪﺍﻳﻞ ﻫﻴﻜﺴﻲ ﺗﺮﺍﻱ ﻣﻦ ﻋﺎﻟﻴﺔ ﻟﺠﺮﻋﺔ ﺍﻟﻌﺮﺿﻲ ﺍﻟﺘﻨﺎﻭﻝ ﻧﺘﻴﺠﺔ ﺳﻨﻮﺍﺕ, Sudan Medical Journal 11, 1-8.
ACS Paragon Plus Environment
37
ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47
ACS Paragon Plus Environment
Page 38 of 38