Classics in Chemical Neuroscience: Chlorpromazine - ACS Publications

Jun 21, 2018 - Classics in Chemical Neuroscience: Chlorpromazine. Debra Boyd-Kimball* , Katelyn Gonczy , Benjamin Lewis , Thomas Mason , Nicole Siliko...
3 downloads 0 Views 534KB Size
Subscriber access provided by - Access paid by the | UCSB Libraries

Review

Classics in Chemical Neuroscience: Chlorpromazine Debra Boyd-Kimball, Katelyn Gonczy, Benjamin Lewis, Thomas Mason, Nicole Siliko, and Jacob Wolfe ACS Chem. Neurosci., Just Accepted Manuscript • DOI: 10.1021/acschemneuro.8b00258 • Publication Date (Web): 21 Jun 2018 Downloaded from http://pubs.acs.org on June 22, 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 30 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

Classics in Chemical Neuroscience: Chlorpromazine

Debra Boyd-Kimball*, Katelyn Gonczy, Benjamin Lewis, Thomas Mason, Nicole Siliko, and Jacob Wolfe

Department of Chemistry and Biochemistry, University of Mount Union, Alliance, OH 44601, USA

*Address correspondence to: Debra Boyd-Kimball, Department of Chemistry and Biochemistry, University of Mount Union, Alliance, OH 44601, USA Phone: 330-823-3674; Fax: 330-823-8531; E-mail: [email protected]

Keywords: chlorpromazine, antipsychotic, schizophrenia, psychopharmacology, dopamine, history

ACS Paragon Plus Environment

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 30

Abstract The discovery of chlorpromazine in the early 1950s revolutionized the clinical treatment of schizophrenia, galvanized the development of psychopharmacology, and standardized protocols used for testing the clinical efficacy of antipsychotics. Furthermore, chlorpromazine expanded our understanding of the role of chemical messaging in neurotransmission and reduced the stigma associated with mental illness facilitating deinstitutionalization in the 1960s and 1970s. In this review, we will discuss the synthesis, manufacturing, metabolism and pharmacokinetics, pharmacology, structure-activity relationship, and adverse effects of chlorpromazine. In conclusion, we summarize the history and significant contributions of chlorpromazine which have resulted in this potent first-generation antipsychotic persisting in clinical relevance for nearly 70 years.

Introduction Schizophrenia is a severe, chronic psychological disorder characterized by the presence of positive, negative, and cognitive symptoms.1-4 Up until the mid-twentieth century a diagnosis of schizophrenia historically meant a lifetime of confinement to an asylum.5-7 Standard treatments of the time were electroconvulsive therapy (ECT) and prolonged sleep therapy which were employed with agitated patients.5,7,810

The introduction of chlorpromazine (CPZ) in the early 1950s catalyzed the development of

psychopharmacology11 and expanded our understanding of chemical signaling in neurotransmission.12 Additionally, CPZ helped to reduce the public stigma surrounding mental illness leading to the deinstitutionalization of those with schizophrenia.6,7,10,13 In this review, we attempt to synthesize a portion of the literature on CPZ to describe the synthesis and basic properties of this first-generation antipsychotic that has persisted for almost 70 years as the “gold standard” to which all other antipsychotics are compared.

ACS Paragon Plus Environment

Page 3 of 30 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

Chemical Synthesis CPZ, 3-(2-chlorophenothiazin-10-yl)-N, N-dimethylpropan-1-amine (CAS No. [50-53-3]), is a low molecular weight phenothiazine derivative (MW = 318.1). The first synthetic route to CPZ was reported from the laboratories of Rhône-Poulenc by Paul Charpentier and US patent number US 2,645,640 A was issued on July 14, 1953 (Scheme 1).14,15 In the original synthesis, the chlorophenothiazine (2) was prepared by the cyclization of 3-chlorodiphenylamine (1) with sulfur in the presence of an iodine catalyst and heat. The cyclization yielded the 2- and 4-chlorophenothiazine isomers which were separated by fractional crystallization. Deprotonation of (2) with sodium amide was then followed by addition of N,N-dimethyl-3-chloropropylamine. Distillation of the ethereal extract yielded CPZ (3) which was then recrystallized as the hydrochloride.14,15 Since its first synthesis in 1951, various other routes to produce CPZ have been recognized.16-21 One of the more notable modern synthetic routes to chlorpromazine hydrochloride was patented by Wang in 2012 under patent CN102617509 (A). This synthesis which includes the substitution of 2-chlorophenothiazine with N, N-dimethyl-3-chloropropylamine followed by neutralization with hydrochloric acid, provides higher yields than traditional methods (above 90%). This higher yield is attributed to the use of sodium hydroxide and tetrabutyl ammonium bromide as the condensing agents, strict control of the raw material ratio, and an indicator for the end point of the salification reaction.22 Scheme 1: Original synthesis of chlorpromazine (3) reported by Charpentier.14,15

ACS Paragon Plus Environment

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 30

Scheme 2: Recent synthesis of chlorpromazine hydrochloride.22

Manufacturing Information CPZ, produced in the United States under the brand name Thorazine®, was approved by the FDA for treatment of schizophrenia in 1957.23 CPZ was primarily manufactured by GlaxoSmithKline under the brand name Thorazine in tablet form, but its production was discontinued in 2016.24 USL Pharma was granted FDA approval for production and sale of CPZ on July 9th, 1974. Two weeks later, West-Ward Pharmaceuticals was granted FDA approval on July 25th, 1974.25 Many other U.S. manufacturers had gained FDA approval for the sale of CPZ but have since discontinued production of the drug. It is currently manufactured by companies in the United States under the generic name, Chlorpromazine Hydrochloride, by USL Pharma in tablet form and West-Ward Pharmaceuticals in intravenous form. In tablet form, CPZ is available in dosages of 25 mg, 50 mg, 100 mg, and 200 mg.26 In injection form, CPZ is available at the strength of 25 mg/mL.27

Drug Metabolism and Pharmacokinetics CPZ has no hydrogen bond donors, two hydrogen bond acceptors, and a cLogP of 5.80.28 Together, these properties conform to Lipinski’s rules and are consistent with the acceptable drug metabolism and pharmacokinetic (DMPK) parameters and central nervous system (CNS) penetration of CPZ. The metabolism of CPZ varies based on its method of administration. If administered orally, CPZ undergoes extensive presystemic first-pass metabolism.29 This metabolism can be quantified by the bioavailability of unchanged CPZ that reaches circulation (F%). This value has been reported as low and variable ranging from 4-38%.30 It has

ACS Paragon Plus Environment

Page 5 of 30 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

been seen that orally administered CPZ begins to appear in systemic circulation after a mean lag time of 0.4 hours and is subsequently absorbed for an average of 2.9 hours. The average peak concentration of unaltered CPZ after this absorption was calculated to be 0.079 µg/ml, occurring two to four hours after ingestion.31 Alternatively, when CPZ is intravenously administered, the drug avoids extensive first-pass metabolism and the average clearance rate has been calculated to be 1.06 l·hr-1 kg-1 . Additionally, the average volume of distribution at a steady state was found to be 8.88 l·kg-1 .30 As expected, when comparing the oral and IV administration of CPZ, the data gathered on its pharmacokinetics varies. The mean half-life and AUC values are shown in Table 1. AUC, or area under the plasma drug concentration versus time curve, indicates the bodily exposure after administration of a drug.32 Table 1: Comparison of mean half-life and AUC values (SD) between varying administration methods.30 Mean Half-Life

AUC

(hr)

(ng·hr-1 ·ml-1)

Oral- 25 mg

5.48 (1.72)

27.80 (14.90)

Oral- 50 mg

9.52 (4.55)

81.80 (79.20)

Oral- 100 mg

11.05 (2.80)

247.00 (127.00)

IV- 10 mg

11.10 (4.33)

135.00 (24.50)

Formulation

Numbers in parentheses indicate standard deviation values.

CPZ undergoes dramatic metabolism that ultimately results in at least 17 recoverable metabolites from urine or plasma samples.33 The five main metabolites of CPZ include chlorpromazine 5-sulfoxide (A), monoN-demethylchlorpromazine (B), di-N-demethylchlorpromazine (C), 7-hydroxychlorpromazine (D), and glucuronic acid conjugates (Figure 1).30, 34-36 These metabolites are produced by specific hepatic cytochrome P450 (CYP) isozymes. For example, CYP1A2 and CYP3A4 have been found to catalyze the mono-Ndemethylation, di-N-demethylation, and 5-sulfoxidation of CPZ. Additionally, CYP2D6, with the assistance of CYP1A2, has been shown to be the primary isozyme that carries out the 7-hydroxylation of CPZ.29, 35-37

ACS Paragon Plus Environment

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 30

A genetic component of pharmaceutical drug metabolism has been reported as a contributing factor to the wide range of pharmacokinetic values for CPZ. Genetic polymorphisms among the P450 enzymes, specifically CYP2D6, have been shown to lead to the emergence of two different phenotypes: extensive metabolizers and poor metabolizers. More specifically, the CYP2D6 polymorphism is known as the debrisoquine/sparteine polymorphism.29,38 Such polymorphisms could account for the variation in pharmacokinetic values among experimental patients. Genetic testing can be done before experimentation to eliminate this variable.29 Additionally, care must be taken when comparing reported pharmacokinetic data, particularly with respect to historical studies, in light of such subject specific metabolic variation.

Figure 1: Scheme of primary metabolites found in urine samples.34-37

ACS Paragon Plus Environment

Page 7 of 30 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

Pharmacology CPZ demonstrates high affinity for dopamine (DA) receptors and acts as a receptor antagonist by inhibiting adenylate cyclase activity. The first in its class, CPZ has become the “gold standard” against which other first-generation, and even second-generation, antipsychotics were compared.39-40 While CPZ has several active metabolites which cross the blood-brain barrier, including 7-hydroxychlorpromazine, it has been shown that unaltered CPZ is primarily responsible for the CNS response.41 The primary pharmaceutical activity of CPZ is dopamine D2 receptor (D2R) antagonism with therapeutic concentrations occupying a range of 60-80%. Therapeutic concentrations resulting in levels of D2R occupancy above 78% account for extrapyramidal symptoms (EPS) as explained by the fast-off D2R theory of antipsychotic action. Positron Emission Tomography (PET) imaging has shown that many conventional antipsychotics, such as CPZ, occupy D2Rs for a longer period of time than many atypical antipsychotics, which wear off quickly following oral dose. This increased duration of occupancy is due to the tighter binding of CPZ, and other typical antipsychotics, to D2Rs in comparison to atypical antipsychotics. Such prolonged stimulation of D2Rs by CPZ contributes to the high rates of EPS observed in patients.42 While the binding affinity of CPZ for other dopamine receptors has been tested, the D1, D3, and D4 receptors show low affinity, designating low likelihood of these receptors playing a role in the antipsychotic activity (Table 2).41-43 For a majority of its history, CPZ was mainly studied as a D2R antagonist. More recently, study of the binding interactions of CPZ have been expanded beyond the DA system to include other receptor sites. In particular, the serotonin (5-HT) system has been extensively investigated; however, there remains some question as to the efficacy and blockage of the 5-HT neuronal system by CPZ. For most 5-HT receptors a low binding affinity is reported.41,44 A notable exception to this is the 5-HT2A receptor subtype which exhibits Ki values similar to the D2R (Table 2).42-45 However, the high 5-HT2A occupancy does not necessarily provide a unique clinical profile for CPZ because atypical antipsychotics, like clozapine, share similar blockage of this receptor, signifying that D2Rs most likely play the largest role in the clinical profile of the antipsychotic activity of the drug. Therefore, at typical clinical dosage, blockage of the 5-HT2A receptor by CPZ appears to be just as

ACS Paragon Plus Environment

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 30

likely as blockage by atypical antipsychotics.45 Additionally, contrasting studies report that while CPZ demonstrates 5-HT2A receptor inhibition in vitro, this pharmacological profile is lost in vivo.46 CPZ is also a known adrenoceptor antagonist, which contributes to its antipsychotic effect. CPZ has shown high affinity towards the α1A and α1B subtypes of this receptor, which is coexpressed in the frontal cortex and thalamus.47 Within rats, CPZ has a 4-fold higher affinity for the α1A adrenoceptor subtype, which is higher than that exhibited at D2Rs.47-49 The inhibitor α1A adrenoceptor could account for the antipsychotic activity of CPZ since psychosis is a symptom due to hyperactivity of adrenergic receptors.49 Additionally, at high doses, CPZ has also been shown to increase hippocampal acetylcholine release. While several atypical antipsychotics can induce this response at a lower dose, it is suggested that CPZ increases acetylcholine release through inhibition of terminal muscarinic M2 autoreceptors.50 Table 2: Summary of Select Inhibitor Constants Associated with Chlorpromazine

Receptor Serotonin44 5-HT1A 5-HT1B 5-HT1D 5-HT1E 5-HT2A 5-HT2C 5-HT3 5-HT5A 5-HT6 5-HT7 Dopamine44 D1 D2 D3 D4 D4.2 D4.4 D5 Adrenoceptor47 α1A α1B

Ki Value (nM) 3115 1489 452 344 3.32 15.55 977 118 12 21 112 2 5 10.8 26.2 15.9 133 1.5 5.6

ACS Paragon Plus Environment

Page 9 of 30 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

Structure-Activity Relationship (SAR) Although CPZ has been extensively characterized as binding to a variety of targets (Table 2), relatively few structural binding mechanisms have been elucidated owing to the age of CPZ. First crystallized from solution by McDowell in 1969, the functional propylamine moiety present on CPZ was found to be facing the A-ring with its chlorine substituent.51 This conformation was initially of some debate, but simple overlay studies of this A-ring facing conformation with DA placed both the A-ring and protonatable tertiary amine in similar positions to those of DA (Figure 2).52 These observations were eventually validated by potential energy calculations and molecular dynamics simulations with phenothiazine, xanthine, and thioxanthene derivatives that indicated critical van der Waals interactions between the side chain and the chlorine A-ring substituent existed. Increasing such van der Waals forces was clinically associated with increased D2R binding, antipsychotic efficacy, and EPS.53,54 Although a co-crystal structure is sorely lacking, one fairly well understood interaction is between CPZ and the D2R where the drug exerts its greatest clinical effects. Earlier work suggested that CPZ must contain a protonatable amine to effectively bind the D2R as both quaternary ammonium and dimethylsulfonium derivatives showed substantially decreased D2R affinity.55 Schetz and Floresca concluded that the cationic amine group of CPZ bound to a highly conserved aspartate residue on the 3rd transmembrane helix of the D2R. This SAR was most likely responsible for bringing CPZ into the ligand binding site crevice of the D2R.56 A tryptophan residue in the 6th transmembrane helix that sits on one side of the binding crevice in a cluster of aromatic residues is considered a molecular switch responsible for D2R activation. DA binding to the D2R results in flipping of this molecular switch and conformational change whereas CPZ binding does not.56,57 This most likely owes to a lack of or improper steric clashing between the bulky aromatic residues that are thought to cause D2R activation upon DA binding.56 Thus, in a primitive sense, CPZ does “blockade” the D2R in order to exert its effects. An additional area of which some characterization has occurred in is the binding of CPZ to cationselective pentameric ligand gated ion channels (pLGICs), such as nAChRs and the 5-HT3R.58 These receptors

ACS Paragon Plus Environment

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 30

allow for the fast conversion of a chemical signal to an electrical signal at synapses and CPZ appears to modulate their activity in different ways. In nAChRs, CPZ physically obstructs the ion channel.59 Photoaffinity studies have indicated that CPZ can bind at sites near both the cytoplasmic and extracellular portions of the transmembrane pore.60 In contrast, CPZ binds directly to the 5-HT3R ligand binding site and antagonizes serotonin function.61,62 In addition to binding with multiple receptors, CPZ has been shown to interact with lipid bilayers. Owing to its bulk phenothiazine ring structure, CPZ easily diffuses into bilayers.63 This is stabilized by the hydrogen bonding of the phosphate group from membrane phospholipids with the propylamine moiety of CPZ.54 CPZ was found to be preferentially sequestered on the inner leaflet of membranes due to its ionic interactions with the anionic phospholipids, phosphatidylinositol (PI) and phosphatidylserine (PS); however, it has been demonstrated that CPZ is preferentially bound to phosphoinositides present on the inner leaflet when compared to PS.63 This claim was furthered by work that determined polyphosphoinositide levels in erythrocytes were upregulated in the presence of CPZ.64 This points to the possible physical sequestration of CPZ by the these densely charged phospholipids which could ultimately lead to the disruption of intracellular signals by phospholipase C generated second messengers, inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG).63,64 Although these studies focused on erythrocytes due to their easily quantifiable morphological index, this principle could be applied to other lipid bilayers due to their universally anionic inner leaflets.

ACS Paragon Plus Environment

Page 11 of 30 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: Structural similarities between DA and CPZ. An early overlay study by Horn and Snyder 52 made the astute observation that if the propylamine arm of CPZ faces the A-ring, this would cause the A-ring and propylamine to line up with the aromatic ring and amine found in DA. This was later confirmed as the biologically active form of CPZ by further work.53,54

ACS Paragon Plus Environment

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 30

Adverse Effects CPZ is an antipsychotic medication that works at all levels of the central nervous system, with subcortical levels being the primary target.8 CPZ was at one time the main drug for the treatment of schizophrenia but has since been discontinued by many pharmaceutical companies.23,24 Studies have shown that use of CPZ can result in weight gain, sedation, movement disorders (tremors and shaking), and orthostatic hypotension. Orthostatic hypotension is usually observed when CPZ is administered in higher doses.65 Some of these EPS side effects are seen with many antipsychotic drugs and are the main concern when prescribing CPZ.66 EPS most often occur when D2Rs are highly occupied, resulting most often in acute dystonic reactions or tardive dyskinesia.42 These are usually seen when dosage of CPZ is changed, especially in children and young adults.67 Additionally, CPZ can cause drug-induced parkinsonism.68 This side effect is often seen in elderly patients within the first few months of treatment.69 The presence of EPS in schizophrenic patients using CPZ were initially thought to be indicators that proper dosages had been achieved. Further evidence and research showed that EPS could worsen psychotic symptoms to the point that they were irreversible and lethal in some cases.70 Research has shown that decreasing the dose of CPZ to less than 600 mg/day reduced the risk of the onset of EPS to levels equivalent with second-generation drugs for schizophrenia treatment.66 The most commonly reported side effects of CPZ include drowsiness, sedation, dry mouth, and nasal discomfort. CPZ negatively affects various systems in the body including: cardiovascular, respiratory, endocrine, metabolic and immune systems. It also commonly has negative effects on many organs, such as the: eyes, intestines, liver, and skin.71 Many of these negative effects are typical of first-generation antipsychotics and are the reason that second-generation drugs were developed.72

ACS Paragon Plus Environment

Page 13 of 30 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

History and Significance in Chemical Neuroscience The serendipitous discovery of the neuroleptic properties of chlorpromazine grew out of the simultaneous investigation of phenothiazine derivatives for a variety of clinical applications. Phenothiazines are a class of molecules that were originally developed in the late 19th century in the dye and textile industries; during the early 20th century, however, phenothiazines were recognized for their antiseptic and antiparasitic properties and explored for antihistaminic properties, but research on known phenothiazines was halted due to toxicity concerns. During World War II the Allied forces in southeast Asia experienced a restricted supply of quinine for the treatment of malaria and researchers turned again to phenothiazines.73 Aminoalkyl derivatives of phenothiazine were synthesized74 but were devoid of antimalarial activity.73 The lack of antimalarial properties was confirmed by researchers at Rhône-Poulenc Laboratories in France who continued to study these derivatives for their antihistaminic properties, notably bringing promethazine (Phenergan®) to the market as a clinical allergy treatment in 1947.73,75 Concurrently, other researchers including Daniel Bovet and Henri-Marie Laborit were studying the ability of these antihistaminic compounds to alleviate stress and shock reactions. Laborit, a French army surgeon, was particularly interested in identifying a pharmacologically active agent with hypothermic and sedative effects to induce an artificial hibernation that could prevent surgical shock.76 Laborit used promethazine in combination with dolantine creating his “lytic cocktail” and noted that anxious patients were relaxed and calm following surgery77 consistent with the reported use of promethazine as a sedative to calm patients with manic depression.78 At Rhône-Poulenc, Simone Courvoisier analyzed the sedative properties of all the antihistaminic agents Paul Charpentier and his team had synthesized since 1944 and determined promazine appeared to be a promising option;73 thus, Charpentier undertook the synthesis of promazine derivatizes and, in December 1950, synthesized CPZ. Based on the promising results of in vitro and in vivo laboratory tests,79,80 Laborit tested the activity of CPZ in his lytic cocktail and observed that patients were relaxed and calm not only after surgery, but also before surgery. Based on such observations, Laborit predicted the implication of these agents in treating psychiatric disorders.81 Pharmacodynamic and pharmacokinetic studies showed that CPZ possessed a broad

ACS Paragon Plus Environment

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 30

range of pharmaceutical activities including, but not limited to, antipyretic, anticonvulsant, adrenolytic, antiemetic, and antifibrillatory.80 Based on this wide range of pharmacological activity, CPZ was commercialized in 1952 in France by Rhône-Poulenc under the commercial name Largactil® meaning large in action.73,82,83 CPZ was first administered to a hospitalized manic patient in January 1952 as an adjunct therapy. The patient rapidly calmed and, as therapy continued over a three-week period, remained calm ultimately being discharged from the hospital.84 Jean Delay and Pierre Deniker were the first to administer CPZ alone as a pharmaceutical agent in psychiatric patients at Hôpital Sainte-Anne in Paris. Delay and Deniker confirmed the efficacy of the agent in calming agitated or psychotic patients but, noted that a significantly higher dose than Laborit reported was necessary to achieve these effects when CPZ was administered alone.85 Based on the observed effects, Delay proposed the term neuroleptic to classify CPZ and any agent producing a similar slowing in motor activity.86 The use of CPZ in psychiatry spread quickly through Europe to the Psychiatrische Universitätsklinik in Switzerland where Felix Labhardt published on the efficacy of CPZ in treating schizophrenia87,88 and to the University of Birmingham in England where Joel and Charmain Elkes conducted and published the first controlled test of CPZ.89 The Elkes’ study was blind and self-controlled and notably introduced randomized trials and placebo-controlled research methodology into psychiatry.73,82,90 While the use of CPZ spread quickly across Europe, Heinz Lehmann (Montreal) and Smith Kline & French Laboratories (Philadelphia), now GlaxoSmithKline, can be credited with introducing CPZ to North America.73,91 Lehmann initially tested the sedative effects of CPZ compared to secobarbital in healthy volunteers and, concluding that CPZ induced “selective inhibition,” or a sedation that did not impair cognitive function, Lehmann set up a clinical trial of CPZ on 71 psychiatric patients which was the first published in North America.8,92,93 Smith Kline & French Laboratories obtained the license from Rhône-Poulenc to market CPZ in the US and brought CPZ to market in 1954 under the name Thorazine®. Originally approved by the US Food and Drug Administration (FDA) as an antiemetic, CPZ was first used in the US as a hypothermic agent during surgery rather than in psychiatric use;73,91 however, in 1953, Frank Ayd (Baltimore) had become the first ACS Paragon Plus Environment

Page 15 of 30 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

psychiatrist authorized by the FDA to study CPZ. Nevertheless, in 1954, N. William Winkelman (Philadelphia) and Willis H. Bower (Boston) independently published on the clinical use of CPZ in a range of psychiatric disorders while Ayd published his findings on CPZ in 1955. Collectively, these publications prompted the general acceptance of CPZ in the management of psychiatric illness in North America.73,94-97 It is estimated that 2 million patients were prescribed CPZ in the first 8 months5 resulting in 75 million dollars in gross sales for Smith Kline & French in 1955.73 Agranulocytosis98, liver toxicity, and EPS were observed and reported, but the neuroleptic effect of CPZ was determined to outweigh the risks for most patients.8,68,92,99-106 Haase later suggested that a “neuroleptic threshold” in which the dosage could be therapeutically useful without producing EPS would be optimum;107 however, therapeutic dosages of CPZ could not be disentangled from EPS effects.108-110 The first meta-analysis on the clinical efficacy of chlorpromazine in the treatment of schizophrenia was published in 1960 and highlighted the need for standardized methods of scientific inquiry to be used in the clinical study of CPZ and any other such agent.111 Indeed, the first reports on the use of CPZ were small in scale and many lacked the use of appropriate controls. This is an issue that Lehmann himself highlights in retrospect93 and, while the Elkes’ contribution to the development of these methods was significant73,82,90,112 the Elkes’ study reported the effect of CPZ in a number of psychotic conditions which included only 13 patients with schizophrenia.89 The first large-scale controlled studies of CPZ in schizophrenic patients were conducted in numerous US Veterans Administration (VA) neuropsychiatric hospitals113,114 and through the National Institute of Mental Health Psychopharmacology Service Center (NIMH-PSC).115 In the first VA study, CPZ was tested against promazine, phenobarbital, and a lactose placebo in a double-blind, cross-over study that lasted 24 weeks in total and found the efficacy of CPZ was significantly greater than promazine and the two control agents.113 The second VA study used CPZ as a positive control in a double-blind, 12-week study to test therapeutic efficacy of other phenothiazine derivatives.114 These studies focused solely on the therapeutic efficacy of CPZ in men with both studies largely dominated by chronic schizophrenic patients.113,114 The NIMH-PSC study attempted to address these limitations in the experimental design by limiting the patient pool ACS Paragon Plus Environment

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 30

to those newly admitted with no hospitalization in the prior 12 months and utilizing mixed-gender groups for each treatment at each of the 9 collaborating hospitals. The therapeutic efficacy of thioridazine and fluphenazine were tested against CPZ and while this study found no significant difference in the therapeutic effects of the phenothiazines tested, it did begin to cast doubt on the hypothesis that extrapyramidal symptoms were necessary for clinical effectiveness.115 While CPZ was broadly prescribed worldwide by the 1960, a mode of action was still unknown, and studies had failed to reach a consensus on recommended dosage. In 1962, it was reported that discrepancies in earlier CPZ studies could have been due to the hypothermic effect of the drug.116 When the hypothermic effect of the drug was controlled, Carlsson and Lindqvist reported that CPZ blocked dopamine receptors in mice resulting in a feedback mechanism that increased the synthesis and metabolism of catecholamines117 which was further supported by reports of increased dopamine synthesis and metabolism induced by CPZ118-122 and led to the proposal of the dopamine hypothesis of schizophrenia.123 Evidence of CPZ blockade of dopamine receptors was reported in the mid-1970s124-126 lending further support to the mode of action of CPZ and the dopamine hypothesis of schizophrenia.127-130 Prior to the introduction of CPZ, patients diagnosed with schizophrenia may have been secluded from society living out the remainder of their lives in the confines of a hospital; however, CPZ made the symptoms of schizophrenia more manageable and more individuals with schizophrenia found a life outside of the institutional care system. This helped to break the stigma surrounding mental health as these patients integrated into society and normalized mental health care within communities.6,7,10,131 In the US, with the help of federal funding of deinstitutionalization,132 it is estimated that the number of institutionalized patients declined from more than 500,000 in the mid- 1950s to 60,000 by 2000.7 The clinical development of CPZ as a therapeutic agent for schizophrenia was recognized in 1957 when Deniker, Laborit, and Lehmann were recognized with the American Public Health Association’s Albert Lasker Clinical Medical Research Award.73,133,134 It should also be noted that Daniel Bovet was also recognized in the

ACS Paragon Plus Environment

Page 17 of 30 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

same year with the Nobel Prize in Medicine for his contributions to the synthesis of antihistamines135 which subsequently led to the synthesis of promazine and, ultimately, CPZ.134 Overall, the discovery of chlorpromazine revolutionized our understanding of mental illness and introduced the field of psychopharmacology90,93,112,136,137 including the development of standardized screening methods and protocols for large-scale, multi-hospital double-blind controlled tests which brought psychiatry into the medical mainstream.73,82,89,111,113-115,138 To facilitate communication, the study of CPZ led to the founding of the Collegium Internationale Neuro-Psychopharmacologicum (CINP) in 1957139 and the American College of Neuropsychopharmacology (ACNP) in 1960.140 Studies of CPZ provided evidence that neurons communicate by chemical messengers further contributing to our broader understanding of neurochemistry.12,141,142 Additionally, chlorpromazine changed the way that we think about and treat mental illness leading to deinstitutionalization around the world.6-7,10,131 CPZ remains the “gold standard” positive control to test the clinical efficacy of new antipsychotic agents143 and, while the introduction of second-generation antipsychotics has diminished the clinical use of CPZ, it remains reliable in clinical efficacy and on the World Health Organization Model List of Essential Medicines.144 For almost 70 years, CPZ has withstood the test of time and it is only fitting that CPZ holds a special place as a classic in chemical neuroscience.

ACS Paragon Plus Environment

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 30

Author Contributions K.G. wrote the section on drug metabolism and pharmacokinetics, B. L. wrote the section on chemical synthesis, T. M. wrote the section on structure activity relationships, N.S. wrote the section on pharmacology and J.W. contributed sections on manufacturing information and adverse effects. D.B.K. wrote the abstract, introduction, and history and significance in chemical neuroscience sections. K.G., B. L., T. M., N.S., and D.B.K. prepared figures and tables. All authors contributed to the editing of the final manuscript.

Conflict of Interest The authors report no conflict of interest.

Funding Sources The authors have no funding sources to declare.

ACS Paragon Plus Environment

Page 19 of 30 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. Volk, D. W., and Lewis, D. A. (2014) Schizophrenia. In Rosenberg’s Molecular and Genetic Basis of Neurological and Psychiatric Disease. (Rosenberg, R.N. and Pascaul, J.M., Ed.), 5th ed., pp. 12931299, Academic Press, New York. 2. Konopaske, G. T. and Coyle, J. T. (2015) Schizophrenia. In Neurobiology of Brain Disorders: Biological Basis of Neurological and Psychiatric Disorders. (Zigmond, M.J., Coyle, J.T., and Rowland, L., Ed.), pp. 639-654, Academic Press, New York. 3. Fröhlich, F. (2016) Schizophrenia. In Network Neuroscience. pp. 309-318, Academic Press, New York. 4. Zielasek, J. and Gaebel, W. (2015) Schizophrenia. In International Encyclopedia of the Social & Behavioral Sciences. (Wright, J.D., Ed.), 2nd ed., pp. 9-15, Elsevier, Cambridge. 5. Mitchell, P. (1993) Chlorpromazine turns forty. Aust. N. Z. J. Psychiatry. 27, 370-373. 6. Rosenbloom, M. (2002) Chlorpromazine and the Psychopharmacologic Revolution. J. Am. Med. Assoc. 287, 1860-1861. 7. Goldstein, J. (2016) Community-Based Care Can Reduce the Stigma of Mental Illness. National Public Radio Public Health Invisibilia. https://www.npr.org/sections/healthshots/2016/07/02/484055668/community-based-care-can-reduce-the-stigma-of-mental-illness (accessed 24 April 2018) 8. Lehmann, H. E. and Hanrahan, G. E. (1954) Chlorpromazine New Inhibiting Agent for Psychomotor Excitement and Manic States. AMA Arch. Neurol. Psychiatry. 71, 227-237. 9. Maxwell, R. A. and Eckhardt, S. B. (1990) Chlorpromazine. In Drug Discovery A Casebook and Analysis. pp. 111-132, Humana Press, Clifton. 10. Haddad, P., Kirk, R., and Green, R. (2016) Chlorpromazine, the first antipsychotic medication: history, controversy and legacy. British Association for Psychopharmacology. https://www.bap.org.uk/articles/chlorpromazine-the-first-antipsychotic/ (accessed 24 April 2018) 11. Lehmann, H. E., and Ban, T. A. (1997) The History of Psychopharmacology of Schizophrenia. Can. J. Psychiatry. 42, 152-162. 12. Carlsson, A. (2004) The Discovery of Chlorpromazine – Impact on Basic Research. Int. J. Neuropsycholpharmacol. 7 (supplement 2), S22. 13. Goldberg, S. E., Klerman, G. L., and Cole, J. O. (1965) Changes in Schizophrenia Psychopathology and Ward Behavior as a Function of Phenothiazine Treatment. Br. J. Psychiatry. 111, 120-133. 14. Charpentier, P. Phenthiazine Derivatives. U.S. Patent 2,645,640, July 14, 1953.

ACS Paragon Plus Environment

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 30

15. Charpentier, P., Gailliot, P., Jacob, R., Gaudechon, J. and Buisson, P. (1952) Recherches sur les diméthylaminopropyl-N phénothiazines substituées. C. R. Acad. Sci. 235, 59–60. 16. Dahl, T., Tornøe, C. W., Bang-Andersen, B., Nielsen, P., and Jørgensen, M. (2008) PalladiumCatalyzed Three-Component Approach to Promazine with Formation of One Carbon–Sulfur and Two Carbon–Nitrogen Bonds. Angew. Chem. Int. Ed. 47, 1726-1728. 17. Galons, H., Miocque, M., Combet-Farnoux, C., Bensaid, Y., Decodts, G., and Bram, G. A. (1985) Convenient Procedure for the Synthesis of Phenothiazine Drugs. Chem. Pharm. Bull. 33, 5108-5109. 18. Jaworski, T. J., Sardessai, M. S., Aravagiri, M., Lin, G., Shi, Y. Y., Hawes, E. M., Hubbard, J. W., McKay, G., and Midha, K. K. (1993) Synthesis of the N-oxides of phenothiazine antipsychotic agents. J. Pharm. Sci. 82, 330-333. 19. Ma, D., Geng, Q., Zhang, H., and Jiang, Y. (2010) Assembly of Substituted Phenothiazines by a Sequentially Controlled CuI/L-Proline-Catalyzed Cascade C-S and C-N Bond Formation. Angew. Chem. Int. Ed. 49, 1291-1294. 20. Ram, S. and Spicer, L. D. (1989) Regio- and Chemoselective N-C Bond Formation Via Carbon Dioxide: A New Source of the Methyl Group. Applications to N-Methylated Secondary and Tertiary Amines. Synth. Commun. 19, 3561-3571. 21. Schmolka, S. J. and Zimmer, H. (1984) N -Dimethylaminopropylation in a Solid-Liquid Two Phase System: Synthesis of Chlorpromazine, its Analogs, and Related Compound. Synthesis 1984, 29-31. 22. Wang, S. and Tang, J. Jiangsu Province/China Patent CN102617509 (A), 2012. 23. U.S. Food and Drug (1957) Administration. FDA Approved Drug Products All Approvals November 1957. https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=overview.process&ApplNo=009149 (accessed Nov 13, 2017). 24. U.S. Food and Drug Administration and Health and Human Services Agencies. (2016) Determination That THORAZINE (Chlorpromazine Hydrochloride) Tablets and Other Drug Products Were Not Withdrawn From Sale for Reasons of Safety or Effectiveness. Fed. Regist. 81, 3430-3431. 25. U.S. Food and Drug Administration. (1974) FDA Approved Drug Products: All Approvals July 1974. https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=reportsSearch.process&rptName=1&r eportSelectMonth=7&reportSelectYear=1974&nav (accessed Nov 13, 2017). 26. Upsher-Smith Laboratories. Our Products: Chlorpromazine Hydrochloride http://www.upshersmith.com/our-products/products-a-z/ (accessed Nov 13, 2017). 27. West-Ward Pharmaceuticals. Products: Chlorpromazine Hydrochloride Injection, USP. http://www.westward.com/sitecore/content/Sites/WestWardPublicSite/Home/Products/ProductsRoot/Chlorpromazine%2 0Hydrochloride%20Injection%20USP (accessed Nov 13, 2017).

ACS Paragon Plus Environment

Page 21 of 30 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

28. Kasim, N. A., Whitehouse, M., Ramachandran, C., Bermejo, M., Lennernäs, H., Hussain, A. S., Junginger, H. E., Stavchansky, S. A., Midha, K. K., Shah, V. P., and Amidon, G. L. (2004) Molecular Properties of WHO Essential Drugs and Provisional Biopharmaceutical Classification. Mol. Pharm. 1, 85-96. 29. Muralidharan, G., Cooper, J. K., Hawes, E. M., Korchinski, E. D., and Midha, K. K. (1996) Quinidine inhibits the 7-hydroxylation of chlorpromazine in extensive metabolisers of debrisoquine. Eur J Clin Pharmacol. 50, 121-128. 30. Yeung, P. K., Hubbard, J. W., Korchinski, E. D., and Midha, K. K. (1993) Pharmacokinetics of chlorpromazine and key metabolites. Eur. J. Clin. Pharmacol. 45, 563-569. 31. Whitfield, L. R., Kaul, P. N., and Clark, M. L. (1978) Chlorpromazine metabolism. IX. Pharmacokinetics of chlorpromazine following oral administration in man. J. Pharmacokinet. Biopharm. 6, 187-196. 32. UNIL Area Under The Plasma Drug Concentration-Time Curve. https://sepia.unil.ch/pharmacology/index.php?id=66 (accessed November 19, 2017). 33. Chan, T. L., and Gershon, S. (1973) Chlorpromazine metabolism in humans. Part I. Quantitation of chlorpromazine and its metabolites in human plasma and urine by direct scan spectrodensitometry. Mikrochim Acta. 61, 435-452. 34. Beckett, A. H., Beaven, M. A., and Robinson, A. E. (1963) Metabolism of chlorpromazine in humans. Biochem. Pharmacol. 12, 779-794. 35. Wojcikowski, J., Boksa, J., and Daniel, W. A. (2010) Main contribution of the cytochrome P450 isoenzyme 1A2 (CYP1A2) to N-demethylation and 5-sulfoxidation of the phenothiazine neuroleptic chlorpromazine in human liver-A comparison with other phenothiazines. Biochem. Pharmacol. 80, 1252-1259. 36. Wojcikowski, J., and Daniel, W. A. (2010) Influence of antidepressant drugs on chlorpromazine metabolism in human liver - an in vitro study. Pharmacol. Rep. 62, 1062-1069. 37. Zanger, U. M. and Schwab, M. (2013) Cytochrome P450 enzymes in drug metabolism: Regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol. Ther. 138, 103-141. 38. Eichelbaum, M., Kroemer, H. K., and Mikus, G. (1992) Genetically determined differences in drug metabolism as a risk factor in drug toxicity. Toxicol. Lett. 64-65, 115-122. 39. Dazzan, P., Morgan, K. D., Orr, K., Hutchinson, G., Chitnis, X., Suckling, J., Fearon, P., McGuire, P. K., Mallett, R. M., Jones, P. B., Leff, J., and Murray, R. M. (2005) Different Effects of Typical and Atypical Antipsychotics on Grey Matter in First Episode Psychosis: the AESOP Study. Neuropsychopharmacology 30, 765-774.

ACS Paragon Plus Environment

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 22 of 30

40. Oades, R. D., Rao, M. L., Bender, S., Sartory, G., and Muller, B. W. (2000) Neuropsychological and conditioned blocking performance in patients with schizophrenia: assessment of the contribution of neuroleptic dose, serum levels and dopamine D2-receptor occupancy. Behav. Pharmacol. 11, 317-330. 41. Alfredsson, G., Wiesel, F., and Skett, P. (1977) Levels of chlorpromazine and its active metabolites in rat brain and the relationship to central monoamine metabolism and prolactin secretion. Psychopharmacology (Berl.) 53, 13-18. 42. Seeman, P. (2006) Targeting the dopamine D2 receptor in schizophrenia. Expert Opin. Ther. Targets 10, 515-531. 43. Roth, B. L., Tandra, S., Burgess, L. H., Sibley, D. R., and Meltzer, H. Y. (1995) D4 dopamine receptor binding affinity does not distinguish between typical and atypical antipsychotic drugs. Psychopharmacology (Berl.) 120, 365-368. 44. Richtand, N. M., Welge, J. A., Logue, A.D., Keck, Jr, P.E., Strakowski, S.M., and McNamara, R. K. (2007) Dopamine and Serotonin Receptor Binding and Antipsychotic Efficacy. Neuropsychopharmacology 32, 1715-1726. 45. Trichard, C., Paillére-Martinot, M., Attar-Levy, D., Recassens, C., Monnet, F., and Martinot, J. (1998) Binding of Antipsychotic Drugs to Cortical 5-HT2A Receptors: A PET Study of Chlorpromazine, Clozapine, and Amisulpride in Schizophrenic Patients. Am. J. Psychiatry 155, 505-508. 46. Suzuki, H., Gen, K., and Inoue, Y. (2013) Comparison of the anti-dopamine D(2) and anti-serotonin 5HT(2A) activities of chlorpromazine, bromperidol, haloperidol and second-generation antipsychotics parent compounds and metabolites thereof. J. Psychopharmacol. 27, 396-400. 47. Cahir, M. and King, D. J. (2005) Antipsychotics lack α1A/B adrenoceptor subtype selectivity in the rat. Eur. Neuropsychopharmacol. 15, 231-234. 48. Huerta-Bahena, J., Villalobos-Molina, R., and García-Sáinz, J. A. (1983) Trifluoperazine and chlorpromazine antagonize alpha 1- but not alpha2- adrenergic effects. Mol. Pharmacol. 23, 67-70. 49. Cohen, B. M. and Lipinski, J. F. (1986) In vivo potencies of antipsychotic drugs in blocking alpha 1 noradrenergic and dopamine D2 receptors: Implications for drug mechanisms of action. Life Sci. 39, 2571-2580. 50. Johnson, D. E., Nedza, F. M., Spracklin, D. K., Ward, K. M., Schmidt, A. W., Iredale, P. A., Godek, D. M., and Rollema, H. (2005) The role of muscarinic receptor antagonism in antipsychotic-induced hippocampal acetylcholine release. Eur. J. Pharmacol. 506, 209-219. 51. McDowell, J. (1969) The crystal and molecular structure of chlorpromazine. Acta Crystallogr. Sect. B 25, 2175-2181.

ACS Paragon Plus Environment

Page 23 of 30 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

52. Horn, A. S. and Snyder, S. H. (1971) Chlorpromazine and Dopamine: Conformational Similarities that Correlate with the Antischizophrenic Activity of Phenothiazine Drugs. Proc. Natl. Acad. Sci. USA 68, 2325-2328. 53. Feinberg, A. P. and Snyder, S. H. (1975) Phenothiazine drugs: structure-activity relationships explained by a conformation that mimics dopamine. Proc. Natl. Acad. Sci. USA 72, 1899-1903. 54. Pickholz, M., Oliveira, Jr., O. N., and Skaf, M. S. (2007) Interactions of chlorpromazine with phospholipid monolayers: effects of the ionization state of the drug. Biophys. Chem. 125, 425-434. 55. Harrold, M. W., Chang, Y. A., Wallace, R. A., Farooqui, T., Wallace, L. J., Uretsky, N., and Miller, D. D. (1987) Charged analogues of chlorpromazine as dopamine antagonists. J. Med. Chem. 30, 16311635. 56. Floresca, C. Z. and Schetz, J.A. (2004) Dopamine Receptor Microdomains Involved in Molecular Recognition and the Regulation of Drug Affinity and Function. J. Recept. Signal Tranduct. Res. 24, 20739. 57. Visiers, I., Ballesteros, J. A., and Weinstein, H. (2002) Three-dimensional representations of G proteincoupled receptor structures and mechanisms. Methods Enzymol. 343, 329-371. 58. Nys, M., Kesters, D., and Ulens, C. (2013) Structural insights into Cys-loop receptor function and ligand recognition. Biochem. Pharmacol. 86, 1042-1053. 59. Revah, F., Galzi, J. L., Giraudat, J., Haumont, P. Y., Lederer, F., and Changeux, J. P. (1990) The noncompetitive blocker [3H]chlorpromazine labels three amino acids of the acetylcholine receptor gamma subunit: implications for the alpha-helical organization of regions MII and for the structure of the ion channel. Proc. Natl. Acad. Sci. U. S. A. 87, 4675-4679. 60. Chiara, D. C., Hamouda, A. K., Ziebell, M. R., Mejia, L. A., Garcia, G.,3rd, and Cohen, J. B. (2009) (3)H]chlorpromazine photolabeling of the torpedo nicotinic acetylcholine receptor identifies two statedependent binding sites in the ion channel. Biochemistry 48, 10066-10077. 61. Lummis, S. C. and Baker, J. (1997) Radioligand binding and photoaffinity labelling studies show a direct interaction of phenothiazines at 5-HT3 receptors. Neuropharmacology 36, 665-670. 62. Nys, M., Wijckmans, E., Farinha, A., Yoluk, O., Andersson, M., Brams, M., Spurny, R., Peigneur, S., Tytgat, J., Lindahl, E., and Ulens, C. (2016) Allosteric binding site in a Cys-loop receptor ligandbinding domain unveiled in the crystal structure of ELIC in complex with chlorpromazine. Proc. Natl. Acad. Sci. U. S. A. 113, E6696-E6703. 63. Chen, J. Y., Brunauer, L. S., Chu, F. C., Helsel, C. M., Gedde, M. M., and Huestis, W. H. (2003) Selective amphipathic nature of chlorpromazine binding to plasma membrane bilayers. Biochim. Biophys. Acta 1616, 95-105.

ACS Paragon Plus Environment

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 30

64. Butikofer, P., Lin, Z. W., Kuypers, F. A., Scott, M. D., Xu, C. M., Wagner, G. M., Chiu, D. T., and Lubin, B. (1989) Chlorpromazine inhibits vesiculation, alters phosphoinositide turnover and changes deformability of ATP-depleted RBCs. Blood 73, 1699-1704. 65. Leucht, C., Kitzmantel, M., Chua, L., Kane, J., and Leucht, S. (2008) Haloperidol versus chlorpromazine for treatment of schizophrenia. Schizophr. Bull. 34, 813-815. 66. Leucht, S., Wahlbeck, K., Hamann, J., and Kissling, W. (2003) New generation antipsychotics versus low-potency conventional antipsychotics: a systematic review and meta-analysis. Lancet 361, 15811589. 67. Pierre, J. M. (2005) Extrapyramidal Symptoms with Atypical Antipsychotics. Drug Saf. 28, 191-208. 68. Hall, R. A., Jackson, R. B., and Swain, J. M. (1956) Neurotoxic reactions resulting from chlorpromazine administration. J. Am. Med. Assoc. 161, 214-218. 69. Caroff, S. N., Hurford, I., Lybrand, J., and Campbell, E. C. (2011) Movement Disorders Induced by Antipsychotic Drugs: Implications of the CATIE Schizophrenia Trial. Neurol. Clin. 29, 127-148. 70. Rifkin, A. (1987) Extrapyramidal side effects: a historical perspective. J. Clin. Psychiatry 48 Suppl, 3-6. 71. Solmi, M., Murru, A., Pacchiarotti, I., Undurraga, J., Veronese, N., Fornaro, M., Stubbs, B., Monaco, F., Vieta, E., Seeman, M. V., Correll, C. U., and Carvalho, A. F. (2017) Safety, tolerability, and risks associated with first- and second-generation antipsychotics: a state-of-the-art clinical review. Ther. Clin. Risk Manag. 13, 757-777. 72. Peluso, M. J., Lewis, S. W., Branes, T. R., and Jones, P.B. (2012) Extrapyramidal motor side-effects of first- and second-generation antipsychotic drugs. Br. J. Psychiatry 200, 387-392. 73. Lopez-Munoz, F., Alamo, C., Cuenca, E., Shen, W. W., Clervoy, P., and Rubio, G. (2005) History of the discovery and clinical introduction of chlorpromazine. Ann. Clin. Psychiatry. 17, 113-135. 74. Gilman, H., van Ess, P. R., and Shirley, D. A. (1944) The methylation of 10-phenylphenothiazine and of 10-ethyl-phenothiazine. J. Am. Chem. Soc. 66, 1214-1216. 75. Halpern, B. N. and Ducrot, R. (1946) Recherches expérimentales sur une Nouvelles série chimique de corps doués de propriétés antihistaminiques puissantes: les dérivés de la thiodiphenylamine. C.R. Soc. Biol. 140, 361-364. 76. Laborit, H. and Huguenard P. (1951) L’hibernation artificielle par moyens pharmacodynamiques et physiques. Presse Méd. 59, 1329. 77. Laborit, H. (1949) Étude expérimentale du syndrome d’irritation et application clique à la maladie posttraumatique. Thérapie 4, 126-139.

ACS Paragon Plus Environment

Page 25 of 30 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

78. Guiraud, P. and David, C. (1950) Traitement de l'agitation mortice par un antihistaminique (3277 R.P). C. R. Congres. Med. Alien. Neurol. France 48, 599-602. 79. Courviosier, S., Fournel, J., Ducrot, R., Kolsky, M., and Koetschet, P. (1953) Pharmacodynamic properties of 3-chloro-10-(3-dimethylaminopropul)-phenothizaine hydrochloride (R.P. 4560); experimental study of a new substance used in potentialized anesthesia and in artificial hibernation. Arch Int. Pharmacodyn. Ther. 92, 305-361. 80. Courviosier, S. (1956) Pharmacodynamic basis for the use of chlorpromazine in psychiatry. J. Clin. Exp. Psychopathol. 17, 25-37. 81. Laborit, H., Huguenard, P., and Alluaume, R. (1952) Un nouveau stabilisateur végétative (le 4560 PR). Presse Méd. 60, 206-208. 82. Ban, T. A. (2007) Fifty years chlorpromazine: a historical perspective. Neuropsychiatr. Dis. Treat. 3, 495-500. 83. Moncrieff, J. (2013) Chlorpromazine: The First Wonder Drug. In The Bitterest Pills: The Troubling Story of Antipsychotic Drugs. pp. 20-38, Palgrave MacMillian, Hamshire UK. 84. Harmon, J., Paraire, J., and Velluz, J. (1952) Remarques sur l’action du 4560RP sur l’agitation maniaque. Ann. Méd. Psychol. 110, 332-335. 85. Delay, J., Deniker, P., and Harl, J.M. (1952) Utilisation en thérapeutique d’une phénothiazine d’action centrale selective (4560 RP). Ann. Méd. Psychol. 110, 112-117. 86. Delay, J. and Deniker, P. (1952) 38 cas de psychoses traits par la cure prolongee et continue de 4560 R.P. C. R. Congres. Med. Alien. Neurol. France 50, 503-513. 87. Labhardt, F. (1953) Die largactiltherapie bei schizophrenien und anderen psychotischen störungen. Schw. Med. Wochensch. 60, 206-208. 88. Labhardt, F. (1957) Die ergebnisse der Largactil-behandlung schizophener von 1953 bis 1955 an der Basler Psychiatrischen Universitätsklinik. Schw. Arch. NeurolPsychiatr. 79, 355-389. 89. Elkes, J. and Elkes, C. (1954) Effects of chlorpromazine on the behavior of chronically overactive psychotic patients. Br. Med. J. 2, 560-565. 90. Elkes, J. (1995) Psychopharmacology: Finding One’s Way. Neuropsychopharmacology 12, 93-111. 91. Science History Institute. Paul Charpentier, Henri-Marie Laborit, Simone Courvoisier, Jean Delay, and Pierre Deniker. https://www.sciencehistory.org/historical-profile/paul-charpentier-henri-marie-laboritsimone-courvoisier-jean-delay-and-pierre (accessed 24 April 2018) 92. Lehmann, H.E. (1954) Selective Inhibition of Affective Drive by Pharmacological Means. American Journal of Psychiatry 110, 856-857.

ACS Paragon Plus Environment

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 30

93. Lehmann, H.E. (1993) Before They Called It Psychopharmacology. Neuropsychopharmacology 8, 291-303. 94. Winkelman, N.W. (1954) Chlorpromazine in the treatment of neuropsychiatric disorders. J. Am. Med. Assoc. 155, 18-21. 95. Bower, W.H. (1954) Chlorpromazine in psychiatric illness. New Eng. J. Med. 251, 689-692. 96. Ayd, Jr., F.J. (1955) Treatment of psychiatric patients with Thorazine. South. Med. J. 48, 177-186. 97. Ayd, Jr., F.J. (1955) Large doses of chlorpromazine in the treatment of psychiatric patients. Dis. Nerv. Syst. 16, 146-149. 98. Goldman, D. (1955) Agranulocytosis Associated with Administration of Chlorpromazine. AMA Arch. Intern. Med. 96, 496-499. 99. Cohen, I.M. and Archer, J.D. (1955) Liver function and hepatic complications in patients receiving chlorpromazine. J. Am. Med. Assoc. 159, 99-101. 100. Steck, H. (1954) Le syndrome extra-pyramidal et di-encephalique au cours des traitments au Largactil at au Serpasil. Annales Medico-Psychologiques 112, 737-743. 101. Haase, J.H. (1954) Occurrence and interpretation of psychomotor parkinsonism in megaphen or largactil prolonged therapy. Nervenarzt 25, 486-492. 102. Anton-Stephens, D. (1954) Preliminary observations on the psychiatric uses of chlorpromazine (Largactil). Journal of Mental Science 100, 543-557. 103. Hollister, L.E. (1957) Complications from the use of tranquilizing drugs. N. Engl. J. Med. 257, 170177. 104. Kline, N.S. (1956) Chemotherapy in psychiatry: Recent advances. Semin. Int. 5, 7-10. 105. Ayd, Jr., F.J. (1961) A Survey of Drug -Induced Extrapyramidal Reactions. J. Am. Med. Assoc. 175, 1054-1060. 106. Ayd, Jr., F.J. (1963) Chlorpromazine: Ten Years’ Experience. J. Am. Med. Assoc. 184, 51-54. 107. Haase, H.J. and Janssen, P.A.J. (1965) The Action of Neuroleptic Drugs. Chicago: Yearbook Medical Publishers. 108. Haase, H.J. (1956) Definition and mode of action of the psychomotor Parkinson syndrome therapeutically induced by serpasil and largactil. Monatsschrift Psychiatrie fur Neurologie, 131, 201214. 109. Deniker, P. (1960) Experimental neurological syndromes and the new drug therapies in psychiatry. Compr. Psychiatry 1, 92-102. ACS Paragon Plus Environment

Page 27 of 30 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

110. Freyhan, F.A. (1959) Clinical and investigative aspects. In, Psychopharmacology Frontiers: Second International Congress of Psychiatry Psychopharmacology Symposium. (Kline, N.S. ed.), pp. 7-14, J&A Churchill Ltd., London. 111. Heilizer, F. (1960) A critical review of some published experiments with chlorpromazine in schizophrenic, neurotic, and normal humans. J. Chronic Dis. 11, 102-148. 112. Ayd, Jr., F.J. (1991) The early history of modern psychopharmacology. Neuropsychopharmacology 5, 71-84. 113. Casey, J.F., Bennett, I.F., Lindley, C.J., Hollister, L.E., Gordon, M.H. and Springer, N.N. (1960) Drug Therapy in Schizophrenia: A Controlled Study of the Relative Effectiveness of Chlorpromazine, Promazine, Phenobarbital, and Placebo. AMA Arch. Gen. Psychiatry 2, 210-220. 114. Casey, J.F., Lasky, J.J., Klett, C.J., and Hollister, L.E. (1960) Treatment of schizophrenic reactions with phenothiazine derivatives. A comparative study of chlorpromazine, triflupromazine, mepazine, prochlorperazine, perphenazine, and phenobarbital. Am. J. Psychiatry 117, 97-105. 115. The National Institute of Mental Health Psychopharmacology Service Center Collaborative Study Group. (1964) Phenothiazine Treatment in Acute Schizophrenia. Arch. Gen. Psychiatry 10, 246-261. 116. Costa, E., Gessa, G.L., and Brodie, B.B. (1962) Influence of hypothermia on chlorpromazine-induced changes in brain amine levels. Life Sci. 1, 315-319. 117. Carlsson, A. and Lindqvist M. (1963) Effect of Chlorpromazine or Haloperidol on Formation of 3Methoxytyramine and Normetanephrine in Mouse Brain. Acta Pharmacol. Toxicol. 20, 140-144. 118. Andén, N., Roos, B., and Werdinius, B. (1964) Effects of chlorpromazine, haloperidol, and reserpine on the levels of phenolic acids in rabbit corpus striatum. Life Sci. 3, 149-158. 119. Prada, M. and Pletscher, A. (1966) Acceleration of cerebral dopamine turnover by chlorpromazine. Experientia 22, 465-466. 120. Gey, K.F. and Pletscher, A. (1968) Acceleration of turnover of 14C-catecholamines in rat brain by chlorpromazine. Experinetia 24, 335-336. 121. Nybäck, H. and Sedvall, G. (1968) Effect of chlorpromazine on accumulation and disappearance of catecholamines formed from tyrosine-C14 in brain. J. Pharmacol. Exp. Ther. 162, 294-301. 122. Nybäck, H. and Sedvall, G. (1970) Further studies on the accumulation and disappearance of catecholamines formed from tyrosine-C14 in mouse brain: Effect of some phenothiazine analogues. Eur. J. Pharmacol. 10, 193-205. 123. Van Rossum, J.M. (1966) The significance of dopamine-receptor blockade for the mechanism of action of neuroleptic drugs. Arch. Int. Pharmacodyn. Ther. 160, 492-494.

ACS Paragon Plus Environment

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 30

124. Seeman, P., Chau-Wong, M., Tedesco, J., and Wong, K. (1975) Brain receptors for antipsychotic drugs and dopamine: Direct binding assays. Proc. Natl. Acad. Sci. USA 72, 4376-4380. 125. Seeman, P., Lee, T., Chau-Wong, M., and Wong, K. (1976) Antipsychotic drug doses and neuroleptic/dopamine receptors. Nature 261, 717-719. 126. Creese, I., Burt, D.R., and Snyder, S.H. (1975) Dopamine receptor binding: differentiation of agonist and antagonist states with 3H-dopamine and 3H-haloperidol. Life Sci. 17, 993-1002. 127. Seeman, P. (1987) Dopamine receptors and the dopamine hypothesis of schizophrenia. Synapse 1, 133-152. 128. Baumeister, A.A. and Francis, J.L. (2002) Historical Development of the Dopamine Hypothesis of Schizophrenia. J Hist. Neurosci. 11, 265-277. 129. Carlsson, A. and Carlsson, M.L. (2006) A dopaminergic deficit hypothesis of schizophrenia: the path to discovery. Dialogues Clin. Neurosci. 8, 137-142. 130. Madras, B.K. (2013) History of the Discovery of the Antipsychotic Dopamine D2 Receptor: A Basis for the Dopamine Hypothesis of Schizophrenia. J Hist. Neurosci. 22, 62-78. 131. Moncrieff, J. (2013) Magic Bullets for Mental Disorders: The Emergence of the Concept of an “Antipsychotic” Drug. J Hist. Neurosci. 22, 30-46. 132. Toth, R. C. (1963) President Seeks Funds to Reduce Mental Illness. The New York Times. 133. Albert and Mary Lasker Foundation. 1957 Albert Lasker Clinical Medical Research Award. http://www.laskerfoundation.org/awards/show/chlorpromazine-for-treating-schizophrenia/ (accessed 7 May 2018) 134. Ban, T.A. (2006) The role of serendipity in drug discovery. Dialogues Clin. Neurosci. 8, 335-344. 135. Uvnäs, B. (1957) The Nobel Prize in Physiology or Medicine 1957 Award Ceremony Speech. https://www.nobelprize.org/nobel_prizes/medicine/laureates/1957/press.html (accessed 7 May 2018) 136. Caldwell, A. E. (1970) Origins of Psychopharmacology From CPZ to LSD. Charles C Thomas, Springfield. 137. Ban, T.A. (2001) Pharmacotherapy of Mental Illness – A Historical Analysis. Prog. NeuroPsychopharmacol. & Biol. Psychiat. 25, 709-727. 138. Cook, L. (2004) Early Pharmacology of Chlorpromazine. Int. J. Neuropsycholpharmacol. 7 (supplement 2), S21. 139. Ban, T.A. (2006) A history of the Collegium Internationale Neuro-Psychopharmacologicum (19572004). Progress in Neuro-Psychopharmacology & Biological Psychiatry, 30, 599-616.

ACS Paragon Plus Environment

Page 29 of 30 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

140. Healy, D. (1993) 100 Years of Psychopharmacology. J Psychopharmacol. 7, 207-214. 141. Valenstein, E.S. (2005) The War of the Soups and the Sparks: The Discovery of Neurotransmitters and the Dispute Over How Nerves Communicate. Columbia University Press, New York. 142. Baumeister, A.A. (2013) The Chlorpromazine Enigma. J Hist. Neurosci. 22, 14-29. 143. Saha, K.B., Bo, L., Zhao, S., Xia, J., Sampson, S., and Zaman, R.U. (2016) Chlorpromazine versus atypical antipsychotic drugs for schizophrenia. Cochrane Database of Systematic Reviews. http://cochranelibrary-wiley.com/doi/10.1002/14651858.CD010631.pub2/full (accessed 7 May 2018) 144. World Health Organization. (2017) World Health Organization Model List of Essential Medicines. 20th Edition. http://www.who.int/medicines/publications/essentialmedicines/20th_EML2017_FINAL_amendedAug2 017.pdf?ua=1 (accessed 7 May 2018)

ACS Paragon Plus Environment

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

Table of Contents Graphic (For Table of Contents Use Only)

Journal: ACS Chemical Neuroscience Manuscript ID: cn-2018-002588 Title: "Classics in Chemical Neuroscience: Chlorpromazine" Author(s): Boyd-Kimball, Debra; Gonczy, Katelyn; Lewis, Benjamin ; Mason, Thomas ; Siliko, Nicole; Wolfe, Jacob

ACS Paragon Plus Environment

Page 30 of 30