1. Trang chủ
  2. » Giáo Dục - Đào Tạo

Synthetic Approaches To The New Drugs 2018

53 94 0
Tài liệu đã được kiểm tra trùng lặp

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

THÔNG TIN TÀI LIỆU

pubs.acs.org/jmc Perspective Synthetic Approaches to New Drugs Approved during 2018 Andrew C Flick, Carolyn A Leverett, Hong X Ding, Emma McInturff, Sarah J Fink, Christopher J Helal, Jacob C DeForest, Peter D Morse, Subham Mahapatra, and Christopher J O’Donnell* Downloaded via UNIV OF EXETER on July 15, 2020 at 16:28:41 (UTC) See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles Cite This: https://dx.doi.org/10.1021/acs.jmedchem.0c00345 ACCESS Read Online Metrics & More Article Recommendations ABSTRACT: New drugs introduced to the market every year represent privileged structures for particular biological targets These new chemical entities (NCEs) provide insight into molecular recognition while serving as leads for designing future new drugs This annual review describes the most likely process-scale synthetic approaches to 39 new chemical entities approved for the first time globally in 2018 INTRODUCTION The most fruitful basis for the discovery of a new drug is to start with an old drug only a discovery-scale or a general synthetic approach capable of delivering the active pharmaceutical ingredient (API) has been made available Nonetheless, the synthetic sequences described in this review have all been previously reported in either patent or public chemical literature and, to the best of our assessment, represent scalable routes originating from commercially available starting materials (as determined by explicit statement or inferred by experimental detail) Sir James Whyte Black, Winner of the 1988 Nobel Prize in Medicine1 Because drugs can have structural homology across similar biological targets, it is widely believed that the knowledge of new chemical entities and approaches to their construction will enhance the ability to discover new drugs more efficiently This annual review, which is now in its 17th installment,2 presents synthetic routes for 39 new molecular entities that were approved for the first time by a governing body anywhere in the world during the 2018 calendar year (Figure 1).3 Each drug is prefaced by a brief introduction summarizing the relevant pharmacology or differentiating features of the medicine.4 New indications for previously launched medications, new combinations or formulations of existing drugs, and drugs synthesized entirely by biological processes or peptide synthesizers have been excluded from coverage For organizational purposes, drugs presented in this review are categorized into the following therapeutic areas: antibiotic and antifungal, anti-infective, cardiovascular and hematologic, gastrointestinal, inflammation and immunology, metabolic, oncology, ophthalmologic, rare disease, reproductive, and urinary tract Within each of these therapeutic areas, drugs are ordered alphabetically by generic name It is important to note that a drug’s process-scale synthetic approach is often not explicitly disclosed at the time of this review’s publication In some cases, © XXXX American Chemical Society ANTIBIOTIC AND ANTIFUNGAL DRUGS 2.1 Eravacycline (Xerava) Eravacycline belongs to the tetracycline class of antibiotics and was approved by the United States Food and Drug Administration (USFDA) for the treatment of complicated intra-abdominal infections in patients aged 18 years and older Eravacycline is a fully synthetic broad-spectrum antibiotic that exhibits potent activity against both Gram-positive and Gram-negative bacterial strains, including many that have acquired tetracycline-specific resistant mechanisms.5 Eravacycline was discovered and developed by Tetraphase Pharmaceuticals and was Received: February 26, 2020 Published: April 27, 2020 A https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective Figure continued B https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective Figure continued C https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective Figure Structures of 39 NCEs approved in 2018 licensed to Everest Medicines for commercialization in many eastern Asian countries Two other tetracycline antibiotics, sarecycline and omadacycline, were also approved this year, but both molecules were prepared from previously approved tetracyclines that were ultimately obtained via fermentation Eravacycline is a fully synthetic tetracycline, and a highly convergent route for its preparation was first described by the laboratory of Professor D https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective Figure Retrosynthetic approach to eravacycline Scheme Preparation of Eravacycline Isoxazole (4·Tartrate) Andrew Myers at Harvard University.6 This route was later refined at Tetraphase Pharmaceuticals, leading to the discovery and development of eravacycline.7 The route described has been published in the primary and patent literature on multikilogram scale.8 Retrosynthetically, eravacycline (I) was envisioned to be derived via a Michael addition−Dieckmann cyclization reaction between the anion of compound and Michael acceptor (Figure 2) Tricyclic intermediate was envisioned to come from addition of the anion of isoxazole to the aldehyde 3, followed by an intramolecular Diels−Alder reaction to set the carbon framework of Isoxazole was envisioned to come from dimethyl maleate, and the chiral vinyl amine stereocenter was set via an Ellman sulfinamide auxiliary The preparation of the chiral isoxazole is described in Scheme 1.8c Dimethyl maleate (5) was treated with bromine in the presence of azo-bis(isobutyronitrile) (AIBN) and ultraviolet light to give dibromide in 92% yield Condensation of with hydroxyurea in the presence of potassium tert-butoxide provided isoxazole in 66% yield Benzylation of the hydroxy group of followed by DIBAL reduction of the ester gave aldehyde in high yield over the two steps Condensation of with (S)-tert-butylsulfinylamide (Ellman’s auxiliary) in the presence of copper(II) sulfate provided chiral sulfinimine in 85% yield After reaction optimization, was treated with vinylmagnesium chloride in the presence of methyllithium and zinc chloride to give 10 in 95% yield (99.3:0.7 dr) The tertbutylsulfinyl group was removed under acidic conditions The resulting primary amine was treated with formaldehyde in the presence of sodium acetate and then reduced using a picoline− borane complex to give the dimethylamine coupling partner in 88% yield for the two-step sequence (96.0% ee) The ee was enhanced to 99.0% by tartrate salt formation, giving 4·tartrate in 91% yield The preparation of the tricyclic Michael acceptor enone is described in Scheme 2.8b Treating 4·tartrate with sodium hydroxide provided the free base 4, which was reacted with tetramethylpiperidine (TMP) magnesium chloride−lithium chloride complex to effect the direct magnesiation of on the oxazole ring, with no competing allylic metalation This intermediate was reacted with aldehyde to give alcohols 11a/ 11b in 95% yield (3.57:1 dr) Heating the mixture of 11a/11b in DMSO, DIPEA, butylated hydroxytoluene, and isopropyl acetate effected the intramolecular Diels−Alder reaction to give a mixture of endo products (12a/12b), arising from 11a, and a mixture of exo products (12c/12d), arising from 11b This mixture of alcohols 12a−d was oxidized with sulfur trioxide pyridine complex to give ketones 13a/13b in 99.1:0.9 dr and 74% overall yield from 11a/b Treating 13a/13b with boron trichloride efficiently effected demethylation of the methyl enol ether, which spontaneously underwent ring E https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective Scheme Construction of Eravacycline Tricyclic Michael Acceptor Enone Scheme Preparation of Dibenzyl Amine Protected Coupling Partner ether gave nitroarene 18 Reduction of the nitro group with sodium bisulfite provided aniline 19 in 83% from compound 15 Amine 19 was reacted with benzyl bromide to produce the dibenzylamine-protected coupling partner in 80% yield The completion of the synthesis of eravacycline (I) is described in Scheme 4.8a Compound was treated with LDA followed by compound to promote the desired intermolecular Michael addition The resulting Michael adduct was then treated with lithium bis(trimethylsilyl)amide (LHMDS) to induce an intramolecular Dieckmann cyclization, which gave compound 20 in 94% yield The silyl protecting group was opening to provide enone 14 in high yield Protection of the resulting alcohol as its tributylsilyl ether followed by recrystallization from isopropyl alcohol provided the tricyclic Michael acceptor coupling partner in 88% yield The preparation of intermediate is described in Scheme 3.6c Compound 15 was treated initially with LDA and then quenched with methyl iodide to give arene 16 Formation of the acid chloride of 16 followed by reaction with phenol provided the corresponding ester, making way for methyl ether cleavage with boron tribromide to provide phenol 17 Nitration of 17 and protection of the phenol as a benzyl F https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective Scheme Final Assembly of Eravacycline (I) Scheme Synthesis of Ravuconazole (35) removed using hydrofluoric acid, and the resulting intermediate was treated with hydrogen and palladium on carbon These conditions resulted in the removal of the dibenzylamine and benzyl ether protecting groups, which further gave rise to G https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective Scheme Synthesis of Fosravuconazole L-Lysine Ethanolate (II) Scheme Synthesis of Omadacycline III the opening of the oxazole ring to ultimately arrive at compound 21 in 89% yield for the two step sequence Compound 21 was then reacted with acid chloride 22 to furnish eravacycline I in 89% yield 2.2 Fosravuconazole L-Lysine Ethanolate (Nailin) Fosravuconazole L-lysine ethanolate (F-RVCZ) is an orally administered, broad-spectrum antifungal drug approved in Japan for the treatment of onychomycosis in 2018.9 F-RVCZ is a prodrug of ravuconazole with improved solubility and oral bioavailability.10 Originally discovered by Eisai,11 ravuconazole was licensed to Bristol-Myers Squibb (BMS) for worldwide development, excluding Japan, in 1996 However, BMS terminated development of the drug in 2004, and Eisai reacquired the worldwide development, manufacturing, and marketing rights The antifungal activities of ravuconazole, like other azole drugs, derive from the inhibition of ergosterol biosynthesis and block the 14α-demethylation pathway present in many strains of yeasts and molds.10 The lowering of ergosterol levels leads to accumulation of 14α-methyl sterols, which impairs normal structure and functions of cell membranes, ultimately resulting in growth inhibition or death of fungal cells F-RCVZ exhibited higher efficacy (higher initial cure rates and lower recurrence rates), an improved safety-profile (lower hepatic functional disorders), and improved dosing regimen (once daily for 12 weeks) over existing standards of care such as terbinafine and itraconazole.12 In addition to several disclosures describing the gram-scale synthesis of ravuconazole and related precursors,13 a robust plant-scale preparation has been described by researchers at BMS (Scheme 5).14 This route utilized lactate 23 as a starting material for the preparation of arylpropanone 26 First, methyl ester 23 was converted to a morpholine amide in the presence of catalytic sodium methoxide The alcohol was subsequently protected to generate tetrahydropyranyl ether 24 Use of realtime infrared reaction monitoring allowed for safe formation of Grignard reagent 25 from the corresponding bromide, which was then reacted with amide 24 to furnish aryl ketone 26 after aqueous acetic acid quench Corey−Chaykovsky epoxidation and subsequent epoxide opening were performed in a singlestep, telescoped process Once epoxidation was complete, heating the reaction mixture to 90 °C triggered a triazolemediated epoxide-opening sequence to form alcohol 28 The stereochemical outcome of the epoxide-forming step is H https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective Scheme Synthesis of Plazomicin (IV) Paratek Pharmaceuticals, omadacycline was licensed to Bayer, Merck, and Novartis over the course of its clinical development Ultimately the rights were returned to Paratek, who collaborated with Zai Lab (Shanghai) Co., Ltd to commercialize the drug in China.17 A number of syntheses of omadacycline have been published, and the largest scale route is described in Scheme This route advantageously began with minocycline (38, Scheme 7) which is a tetracyclic antibiotic drug first patented in 1961 Minocycline was condensed with N(hydroxymethyl)phthalimide (39) in the presence of triflic acid to give a mixture of the 9-phthalimidomethyl analogs 40 and 41 in approximately a 60:40 ratio This mixture was treated with methylamine, which resulted in hydrolysis of the phthalimide to give an unreported distribution of methylamine analogs 42 and 43 This mixture was then treated with pivaldehyde under catalytic hydrogenation conditions to affect the reductive amination at position N-9, followed by concomitant removal of the hemiaminal group on the amide, furnishing omadacycline (III) in 15−18% yield for the overall process after conversion to the tosylate salt 2.4 Plazomicin (Zemdri) Originally discovered by California-based Ionis Pharmaceuticals and later developed by Achaogen, plazomicin was approved by the USFDA in 2018 for the treatment of patients 18 years of age or older with complicated urinary tract infections (cUTI), including pyelonephritis The drug, a next-generation aminoglycoside dictated by the adjacent chiral center, providing 27 in 8.6:1 dr Removal of the tetrahydropyranyl protecting group within 28 generated an intermediate diol which was converted to trisubstituted epoxide 29 via selective mesylation of the secondary alcohol Generation of lithium cyanide in situ from acetone cyanohydrin 30 and LHMDS followed by subsequent addition to epoxide 29 delivered the α-cyano alcohol 31 in 90% yield, which was subsequently converted to thioamide monohydrate salt 33 by treatment with diethyl dithiophosphate 32 and sulfuric acid Condensation of the thioamide 33 with 2-bromo-4′-cyanoacetophenone 34 in hot ethanol resulted in thiazole formation which completed the preparation of ravuconazole (35) Conversion of ravuconazole to the highly water-soluble prodrug fosravuconazole L-lysine ethanolate (II) has been described by the scientists at Eisai (Scheme 6).15 First, ravuconazole (35) was O-alkylated with di-tert-butyl chloromethylphosphate 36 to furnish phosphate ester 37 Subjection of ester 37 to trifluoroacetic acid (TFA) and aqueous sodium hydroxide provided the free acid, which was subsequently converted to fosravuconazole L-lysine ethanolate (II) 2.3 Omadacycline (Nuzyra) Omadacycline belongs to the aminomethylcycline class of tetracycline antibiotics and was approved by the USFDA for the treatment of acute bacterial skin and skin structure infections and communityacquired bacterial pneumonia.16 Discovered and developed by I https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective Scheme Synthesis of Sarecycline Hydrochloride (V) Scheme 10 Assembly of Baloxavir Piperazine Heterocyclic Core 59 completed the development and launch of the drug before rights were acquired by Almarall S.A in 2018.22 To date, there are no publications describing the discovery of sarecycline The only reported synthetic route to the drug, performed on small scale by Paratek, is described in Scheme 9.23 Iodination of commercially available sancycline 51 with Niodosuccinimide followed by HPLC purification provided iodosancycline 52 as the trifluoroacetate salt (no yield reported) Carbonylation of 52 in the presence of palladium acetate and Xantphos followed by treatment with triethylsilane provided the corresponding aldehyde, which was treated with trifluoracetic acid to provide formylsancycline trifluoroacetate 53.24 Condensation of 53 with N,O-dimethylhydroxylamine hydrochloride, reduction with sodium cyanoborohydride, and treatment with hydrochloric acid provided sarecycline hydrochloride (V) in 23% yield for the three-step sequence that is delivered by injection, was acquired by Cipla as part of an auction of Achaogen assets after the American firm filed for chapter 11 bankruptcy.19 As a structural derivative of the antiinfective aminoglycoside sisomycin, plazomicin is a neoglycoside that is highly active against a variety of bacterial pathogens, including many of the Gram-negative rods that are implicated in cUTIs Plazomicin is not affected by most aminoglycoside-modifying enzymes and retains activity against multidrug-resistant (MDR) isolates known as carbapenemresistant enterobacteriaceae (CRE).20 Two synthetic routes to plazomicin have been reported, both of which originate from commercial sisomicin (44, Scheme 8) and vary only by differential protection of the sisomicin amines.21 Sisomicin was treated with an ionexchange resin which furnished trifluoroacetamide 45 after reaction with ethyl trifluorothioacetate Zinc acetate and benzyloxycarbonyl succinimide provided the corresponding Cbz-protected intermediate 46 in a 35% yield from 44 Amide coupling and removal of the trifluoroacetate group yielded glycoside 48, which then underwent reductive amination, benzoyl ester cleavage, and global Cbz removal under hydrogenative conditions to give rise to plazomicin (IV).21c,d 2.5 Sarecycline Hydrochloride (Seysara) Sarecycline belongs to the tetracycline class of antibiotics and was approved by the USFDA for the oral treatment of inflammatory lesions of non-nodular, moderate-to-severe acne vulgaris in patients at least years old Sarecycline was discovered at Paratek Pharmaceuticals and licensed to Warner Chilcott, which was later acquired by Allergan Allergan ANTI-INFECTIVE DRUGS 3.1 Baloxavir Marboxil (Xofluza) In 2018, baloxavir marboxil received its first approval by the Pharmaceuticals and Medical Devices Agency of Japan (PMDA) for the treatment of influenza A or B virus infections.25 Later the same year, the drug was also approved by the USFDA for the treatment of acute uncomplicated influenza (flu) in patients 12 years of age and older who have been symptomatic for no more than 48 h.26 Baloxavir marboxil was discovered by Shionogi, who licensed their rights to Roche in February 2016 for development and commercialization except in Taiwan and Japan (Shionogi maintained its rights in these two J https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective Scheme 64 Synthesis of Omidenepag Isopropyl (XXXVI) Scheme 65 Synthesis of Omidenepag Isopropyl Chloromethylpyridine 361 OPHTHALMOLOGIC DRUGS 9.1 Omidenepag Isopropyl (Eybelis) Omidenepag isopropyl, which is administered as a 0.002% ophthalmic solution, was approved in Japan in 2018 for the treatment of glaucoma and ocular hypertension.194 Discovered and developed by Santen, the drug was designed as an isopropyl ester to allow for increased passive permeability to the corneal epithelium, where the physiologic pH of this compartment would convert the molecule to the corresponding carboxylate, revealing a potent and selective prostaglandin E2 (h-EP2) receptor agonist.195 Omidenpag isopropyl, also referred to as OMDI, lowered intraocular pressure (IOP) in preclinical trials at significantly low concentrations (0.0006%) in a dosedependent manner in monkeys, suggesting that the drug could offer an alternative treatment option to prostaglandin F (FP) receptor agonists.195 The drug, whose structure evolved from a small-molecule EP2 agonist originally published by researchers at Pfizer,196 is currently undergoing clinical trial investigation in the United States as a 0.002% once-daily solution, in comparison to a twice-daily 0.5% timolol maleate solution, for the treatment of ocular hypertension.194 Two patent applications disclosed by the Japanese firm Ube Industries, Ltd have described a process synthesis of omidenepag isopropyl.197 Reacting p-cyanofluorobenzene (357) with pyrazole in the presence of base, borane reduction of the nitrile, and acid−base workup led to 358 in 83% across the three-step sequence (Scheme 64) Next, subjection of sulfonic acid 359 to triflic anhydride prior to exposure to 358 under basic conditions yielded sulfonamine 360 Lastly, reaction with 361 (arising from exposure of 362 in Scheme 65 to sulfonic acid in warm aqueous isopropanol, giving 363 and subsequent treatment with thionyl chloride and aqueous base) under basic conditions in refluxing acetonitrile furnished omidenepag isopropyl (XXXVI) in an impressive 98% yield It should be noted in Scheme 65 that although tert-butyl nicotinate 362 is commercially available, the source of this starting material was not specified in the Ube patent.197b 10 RARE DISEASES DRUGS 10.1 Tezacaftor (Symdeko) In 2018, the USFDA approved tezacaftor as a combination therapy with ivacaftor (a USFDA approved drug in 2012)2k,198 for the treatment of patients with cystic fibrosis (CF) aged 12 years and older who are homozygous for the F508del gene mutation or who have at least one mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) gene that is responsive to tezacaftor/ivacaftor.199 Discovered and developed by Vertex Pharmaceuticals, tezacaftor is a broad-acting CFTR corrector that facilitates the cellular processing and trafficking of normal CFTR and multiple mutant CFTR, including F508del, thereby increasing the amount of CFTR protein at the cell surface.200 Ivacaftor is a CFTR potentiator that enhances chloride ion transport by potentiating the channel gating activity of CFTR protein Ivacaftor potentiates the CFTR protein delivered to the cell surface by tezacaftor, leading to further enhancement of chloride ion transport than either agent can achieve alone.200a One of the major differentiators of tezacaftor over other CFTR correctors is that tezacaftor is not a CYP3A4 enzyme inducer and does not interfere with ivacaftor’s metabolic pathways.201,202 In June 2019, the USFDA approved tezacaftor/ivacaftor combination therapy for the treatment of pediatric patients aged years and older with CF who have certain genetic mutations.203 Multiple patent applications have been filed by scientists at Vertex describing the preparation of tezacaftor, and all of which outline similar reaction sequences.204 The route depicted here has been chosen based on higher reported scale, better efficiency, and lesser step counts.204f The convergent synthetic strategy utilizes a late-stage amide bond formation to unite two key fragments Preparation of the acid AM https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective Scheme 66 Synthesis of Tezacaftor Cyclopropylacetic Acid 368 Scheme 67 Synthesis of Tezacaftor (XXXVII) Scheme 68 Synthesis of Tezacaftor Alkyne 373 authors A Sonogashira coupling between aryl bromide 372 and terminal alkyne 373 (whose synthesis is described in Scheme 68) followed by a Larock-type cyclization completed the preparation of aminoindole subunit 374 To unite key subunits 368 and 374, the carboxylic acid 368 was first converted to an acid chloride and then reacted with aminoindole 374 to deliver bis-benzyl protected tezacaftor Finally, global deprotection via palladium-catalyzed hydrogenation furnished tezacaftor (XXXVII) in 68−84% yield (based on 374) Preparation of alkyne 373 is described in Scheme 68 First, propargyl alcohol 375 was converted to propargyl chloride 376 by treatment with aqueous HCl A Grignard reagent was generated from chloride 376 and subsequently alkylated with benzyl chloromethyl ether 377 Removal of the trimethylsilyl moiety generated the terminal alkyne 373 fragment 368 commenced with the palladium-catalyzed decarboxylative arylation involving ethyl cyanoacetate 365 with aryl bromide 364 in the presence of a bulky phosphine ligand (Scheme 66) The reaction yielded benzonitrile 366 in 66% yield after acidification Cyclopropanation of 366 via double alkylation of ethylene fragment 367 in the presence of a phase transfer catalyst followed by basic hydrolysis delivered carboxylic acid 368, which was purified by recrystallization from hot toluene Synthesis of the indole subunit began with regioselective bromination of 3-fluoro-4-nitroaniline (369) to arene 370 (Scheme 67) Lewis acid-catalyzed epoxide ring opening of (R)-glycidyl benzyl ether (371) by aniline 370 followed by reduction of the nitro group furnished hydroxy p-phenylenediamine 372, which was isolated as the p-TsOH salt The salt was treated with aqueous NaHCO3 in DCM to generate free base 372 prior to the next step It should be noted that the yields for conversion of 370 to 372 were not reported by the AN https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective Scheme 69 Synthesis of Segesterone Acetate (XXXVIII) Scheme 70 Synthesis of Vibegron Progenitor Aminoalcohol 386 11 REPRODUCTIVE DRUGS 11.1 Segesterone Acetate (Annovera) Segesterone acetate is a progestin hormonal contraceptive that was developed by The Population Council and has been available since 2000 In 2018, it was approved as a combination therapy with ethinyl estradiol, marketed under the brand name Annovera.205 This drug represents the first in a new class of contraceptives, as it is administered as a silicone ring that can be inserted by the patient and does not require refrigeration and whose duration of action lasts for an entire year.206 These factors are all especially beneficial for providing effective contraception in limited-resource settings and developing nations.207 Although detailed synthetic routes to segesterone acetate are sparsely available, a 1997 paper from The Population Council208 cited earlier work by Mehrof et al.209 and Schwars et al.210 for a general synthetic approach to the compound A 2013 patent by the same group disclosed an updated and concise synthesis involving nucleophilic addition of an acetylene equivalent into a ketone followed by hydration to obtain the desired functionality.211 The route began with 19norandrostenedione (378, Scheme 69) In a two-step, one-pot procedure, the enone was converted to the enol ether using trimethyl orthoformate, followed by formylation of the remaining ketone to provide 379 in an 80% yield over two steps Next, trimethysilylacetylene was deprotonated using LiHMDS in THF prior to subjection to 379 After the addition was complete, the reaction was allowed to warm to room temperature, and the TMS group was removed with potassium carbonate to generate 380 The alkyne was then hydrated under acidic conditions to the corresponding ketone 381 The next series of reactions enabled inversion of configuration at the congested C-17 center Treatment of alcohol 381 with phenylsulfuryl chloride resulted in chloride displacement and a 2,3-sigmatropic rearrangement to arrive at sulfoxide 382 Subjection of the enone of 382 to trimethoxyphosphine and triethylamine, followed by treatment with hypochlorous acetic anhydride conditions, induced a Mislow−Evans rearrangement to complete the preparation of segesterone acetate (XXXVIII) 12 URINARY TRACT DRUGS 12.1 Vibegron (Beova) Vibegron is a selective β3 adrenergic receptor (β3AR) agonist approved in Japan during 2018 for the treatment of overactive bladder (OAB).212 The drug, originally discovered by Merck,213 was developed jointly by Kyorin Pharmaceutical Co., Ltd and Kissei Pharmaceutical Co., Ltd and is currently undergoing clinical trials for OAB in the United States and Europe.212 Vibegron represents the third AO https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective Scheme 71 Synthesis of Vibegron (XXXIX) β3AR agonist to be examined in clinical trials for OAB, the other two being Astellas’ mirabegron (approved in 2012) and GSK and AltheRx’s solabegron (currently in phase II trials).213 Vibegron differentiates from mirabegron such that it offers an enhanced safety profile (reportedly driven by its selectivity against other adrenergic receptors and CYP3A4 and CYP2D6 inhibition/induction) and offers an improved dosing regimen in comparison to solabegron (once daily versus twice daily, which was used in a phase II efficacy study).213,214 Vibegron evolved from structural modifications to a previously discovered Merck β3AR agonist, MK-0634, which progressed into preclinical studies for the treatment of obesity in the early 2000s and has since been discontinued Vibegron differs from previously reported acyclic β-hydroxylamine β3AR agonists in that it possesses a unique cis-pyrrolidine moiety linking the C-2 and C-5 substituents, resulting in improved pharmacological properties,213 but simultaneously increases the complexity of the drug structure and synthesis Several reports describing the construction of vibegron have been published.213,215 A concise manufacturing-scale synthesis of the drug was reported by Merck in 2018 in which the authors engineered a dynamic kinetic resolution (DKR) that efficiently established a challenging amino alcohol stereorelationship within an early stage intermediate (386, Scheme 70) and later influenced the incumbent C-2 and C-5 pyrrolidine stereocenters within the API structure.215a This elegant synthetic approach is described in Schemes 70 and 71 Oxidation of commercial pentynol 383 (Scheme 70) followed by a Strecker reaction and Boc-protection furnished racemic aminonitrile 384.216 Grignard addition to the nitrile followed by acidic workup generated racemic ketones 385 in 95% Because epimerization of the Boc-protected amine stereocenter within 385 was observed under basic conditions, a ketoreductase (KRED-p301) was engineered to selectively facilitate reaction of cofactor sodium nicotinamide adenine dinucleotide phosphate (NADPNa) with (R)-385 within a pH range capable of facilitating both epimerization and reduction of the ketone.215a In practice, the catalytic conditions developed efficiently converted (R)-385 to amino alcohol 386 The reaction proceeded in 95% conversion with >99.4% ee and with a diastereomeric ratio greater than 100:1 Subjection of 386 to Sonagashira conditions followed by treatment with acid delivered disubstituted alkyne 387 as the corresponding HCl salt Warming 387 in the presence of Hunig’s base facilitated an intramolecular cyclization reaction to produce crude pyrrolidinol 388 which was converted to the corresponding silyl ether prior to hydrogenation The authors reason that the bulky silyl ether imparted good selectivity (95:5 dr) during the hydrogenation step Upon workup of the hydrogenation, the silyl functionality was removed, resulting in an overall conversion of 387 to 390 in 80% yield.215a Reaction of aniline 390 with commercial carboxylate 391 under conventional amide-bond forming conditions secured vibegron (XXXIX) in 93% yield 13 CONCLUSION In summary, the pharmaceutical industry continued its trend of considerable productivity with respect to annual small molecule drug approvals These 39 new medicines (which include highly functionalized macrocycles, compounds containing multiple asymmetric centers, and complex heterocyclic systems) represent an architecturally diverse set of molecules that address disease states worldwide across a wide range of therapeutic areas Fascinatingly, the scale preparation of these drugs relied upon synthetic methodologies spanning the full breadth of known chemical reactions ranging from engineered biocatalytic reactions aided by molecular modeling technologies to transformations first discovered in the 19th century Clever utility of metal-catalyzed reactions proved critical to establishing structural motifs within the 2018 class of drugs, further emphasizing the expanding importance of this technology particularly toward the scale preparation of API and to an expanded scope of bonds that are capable of being fashioned in this manner The architectural complexity within these drugs gives clear evidence that medicinal chemists are becoming empowered to explore structurally challenging motifs in the early discovery stages, further reinforcing the industry’s embrace of complex chemical space As we embark upon a new decade of drug discovery, new biological targets and opportunities will emerge As this expansion continues, the demands to reach higher levels of complexity more quickly will be asked of the modern medicinal and process chemist AP https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Innovative synthetic technologies and creative, artistic approaches will continue to inspire and deliver the medicines of the future Emory University in Atlanta, GA, working on total synthesis of several piperidine-based natural products and the alkaloid minfiensine Prior to joining Pfizer she was a Postdoctoral Fellow working with Professor Daniel Romo at Texas A&M University, exploring new applications of nucleophile-catalyzed aldol lactonization reactions ■ AUTHOR INFORMATION Corresponding Author Perspective Hong X Ding obtained a B.S in Pharmaceutics in 2001 and a Ph.D in Medicinal Chemistry in 2006 from Zhejiang University in Hangzhou, China Hongxia is the cofounder and Chief Executive Officer of Pharmacodia, a leading pharmaceutical big data company in China, which is an online platform (http://www.pharmacodia.com) providing big data and information service in the pharmaceutical R&D field In 2010−2013, Hong Xia joined Shenogen Pharma Group, a China-based biotech company as a senior director of the R&D department Before Shenogen, Hong Xia worked in BioDuro since 2006, as senior group leader and senior research scientist Christopher J O’Donnell − Groton Laboratories, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States; orcid.org/0000-0003-1004-7139; Phone: 860-405-4976; Email: christopher.j.odonnell@ pfizer.com Authors Andrew C Flick − Takeda California, Inc., San Diego, California 92121, United States Carolyn A Leverett − Groton Laboratories, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States Hong X Ding − Pharmacodia (Beijing) Co., Ltd., Beijing 100085, China Emma McInturff − Groton Laboratories, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States; orcid.org/0000-0003-0426-3089 Sarah J Fink − Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts 02142, United States Christopher J Helal − Biogen, Cambridge, Massachusetts 02142, United States Jacob C DeForest − Groton Laboratories, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States Peter D Morse − Groton Laboratories, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States Subham Mahapatra − Groton Laboratories, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States Complete contact information is available at: https://pubs.acs.org/10.1021/acs.jmedchem.0c00345 Emma McInturff obtained a B.S in Chemistry from Boise State University and a Ph.D from the University of Texas at Austin, working in the laboratory of Professor Mike Krische on ruthenium catalyzed carbonyl addition methodology development She joined Pfizer process chemistry in Groton, CT, in 2014 Sarah J Fink obtained a B.A in Chemistry and English literature from Williams College, followed by a Ph.D in Organic Chemistry from the University of Cambridge with Professor Ian Paterson Her thesis work focused on the total synthesis of aplyronine C After a fellowship for young international scientists at Shanghai Institute of Materia Medica, Sarah joined BioDuro in Shanghai in 2014, where she was a scientist and chemistry group leader for integrated drug discovery projects in multiple therapeutic areas She relocated with BioDuro to Boston in early 2017, where she provided medicinal chemistry and scientific project management support for collaborations with pharma and biotech as an associate director for integrated programs Sarah recently joined Takeda’s Rare Disease Drug Discovery Unit Christopher J Helal received his B.S in Chemistry at the Ohio State University in 1991 and carried out his doctoral studies at Harvard University with Professor Elias J Corey He joined Pfizer in 1997 in Groton, CT, working in neuroscience medicinal chemistry He supported the metabolic disease research area and led an enabling technologies team that included biocatalysis, flow chemistry, singleelectron chemistry, reaction optimization, and parallel synthesis He currently leads the medicinal chemistry group at Biogen, focusing on neuroscience indications Notes The authors declare no competing financial interest Biographies Andrew C Flick earned a B.A in Chemistry from Lake Forest College After associate-level appointments at Abbott Laboratories and Array BioPharma, he joined Professor Albert Padwa’s laboratory at Emory University After completing his Ph.D studies on dipolar cycloaddition approaches to alkaloid natural product total synthesis, he joined Pfizer where he was involved with numerous medicinal chemistry projects within the Neurosciences, Rare Diseases, and Inflammation & Immunology therapeutic areas As a synthesis team lead, his contributions led to the discovery of the transcription factor inhibitor PF-06763809, which is currently undergoing clinical trials for the treatment of psoriasis Andy recently joined Takeda California’s GI Drug Discovery Unit and has authored over 40 peer-reviewed publications and patents Jacob C DeForest received his B.A in Chemistry from Pepperdine University under the supervision of Professor Matt Joyner in 2012 He completed his Ph.D in Chemistry at the University of California, Irvine, in 2017 working with Professor Scott Rychnovsky on the total synthesis of alkaloid natural products He is currently a process chemist at Pfizer Peter D Morse obtained a B.Sc in Structural Biology and Chemistry from the University of Connecticut in 2010 He then obtained his Ph.D at the University of North Carolina at Chapel Hill, working with Professor David Nicewicz in 2015 After postdoctoral studies with Professor Timothy Jamison at MIT, he joined the Applied Synthesis Technology group at Pfizer in 2017 Carolyn Leverett began her career at Pfizer in 2012, focusing on the development of microtubule inhibitor-based payloads for use as antibody−drug conjugates She currently works in the Discovery Sciences research group, exploring the use of protein degrader-based therapies for treating a variety of disease areas Carolyn obtained her B.S in Chemistry from North Carolina State University She completed her doctoral studies with Professor Albert Padwa at Subham Mahapatra received his M.Sc in Chemistry from Indian Institute of Technology Bombay, Mumbai, and earned his Ph.D from Oregon State University, Corvallis, where he completed the first total syntheses of natural products amphidinolides C and F Subsequently, Subham’s postdoctoral research with Professor Barry Trost at Stanford University was focused on the development of a new C− AQ https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc H activation method for the generation of π-allyl intermediates Later, Subham picked up a second postdoctoral position in the laboratory of Professor Seth Herzon at Yale University where he worked on the total synthesis of lomaiviticin A Subham joined Pfizer in 2017 Currently, he is a Senior Scientist (medicinal chemistry) in the Internal Medicine Department and his research focuses on cardiovascular and metabolic disease areas NIS, N-iodosuccinimide; NMM, N-methylmorpholine; NMO, N-methylmorpholine N-oxide; NMP, N-methyl-2-pyrrolidone; PBS, phosphate buffered saline; PCC, pyridinium chlorochromate; Ph, phenyl; PhCH3, toluene; Pin, pinacolato; PLP, pyridoxal-5-phosphate; PPA, polyphosphoric acid; PPTS, pyridinium p-toluenesulfonate; p-TsOH, p-toluenesulfonic acid; py, pyridine; rt, room temperature; Su, succinimide; SFC, supercritical fluid chromatography; T3P, 1-propanephosphonic acid cyclic; TBAB, tetrabutylammonium bromide; TBAC, tetrabutylammonium chloride; TBAF, tetrabutylammonium fluoride; TBAHS, tetrabutylammonium hydrogen sulfate; TBAI, tetrabutylammonium iodide; TBS, tert-butyldimethylsilyl; TBTU, O-(benzotriazol-1-yl)-N,N,N,N-tetramethyluronium tetrafluoroborate; t-Bu, tert-butyl; TEMPO, 2,2,6,6-tetramethylpiperidine 1-oxyl; Tf, triflic, trifluoromethanesulfonyl; TFA, trifluoroacetic acid; TFAA, trifluoroacetic acid anhydride; TFE, 2,2,2-trifluoroethanol; THF, tetrahydrofuran; THP, tetrahydropyranyl; TMEDA, N,N,N′,N′tetramethylethylenediamine; TMP, tetramethylpiperidine; TMS, trimethylsilyl; Tr, trityl, triphenylmethyl; Ts, ptoluenesulfonyl Christopher J O’Donnell obtained a B.S in Chemistry from the University of Illinois UrbanaChampaign and a Ph.D in Organic Chemistry from the University of Wisconsin After postdoctoral research at the University of CaliforniaIrvine, he joined Pfizer in 1999 in the Neuroscience Medicinal Chemistry group As a scientist, project leader, and manager in these areas, he has led project teams to the nomination of over 10 clinical candidates In 2010, Chris moved to the Oncology Medicinal Chemistry group to build the Antibody Drug Conjugate chemistry group, and his team has nominated 11 conjugates for clinical development In 2018, Chris moved to the Pfizer Ventures team where he makes and manages equity investments for Pfizer Chris is an author/inventor of 75 peerreviewed journal articles and patent applications ■ ■ ABBREVIATIONS USED Ac, acetyl; ADDP, 1,1′-(azodicarbonyl)dipiperidine; AIBN, 2,2′-azobis(isobutyronitrile); aq, aqueous; BHT, butylated hydroxytoluene; BINAP, 2,2′-bis(diphenylphosphino)-1,1′dinaphthalene; Bn, benzyl; Boc, N-tert-butoxycarbonyl; 2,2′bpy, 2,2′-bipyridyl; Bu, butyl; n-Bu, n-butyl; TTBP·HBF4, tritert-butylphosphonium tetrafluoroborate; cat., catalytic; Cbz, benzyloxycarbonyl; CDI, N,N′-carbonyldiimidazole; COD, 1,5-cyclooctadiene; conc, concentrated; CPME, cyclopentyl methyl ether; CSA, camphorsulfonic acid; Cy, cyclohexyl; dba, dibenzylideneacetone; DBU, 1,8-diazabicyclo[5.4.0]undec-7ene; DCC, 1,3-dicyclohexylcarbodiimide; DCE, dichloroethane; DCM, dichloromethane; de, diastereomeric excess; dr, diastereomeric ratio; DIBAL, diisobutylaluminum hydride; DIC, N,N′-diisopropylcarbodiimide; DIPEA, diisopropylethylamine; DIP-Cl, B-chlorodiisopinocampheylborane, (+)-Ipc2BCl; DMA, dimethylacetamide; DMAP, 4-dimethylaminopyridine; DME, 1,2-dimethoxyethane; DMF, N,N-dimethylformamide; DMF−DMA, dimethylformamide−dimethylacetal; DMSO, dimethyl sulfoxide; DPPA, diphenylphosphoryl azide; dppb, 1,4-bis(diphenylphosphino)butane; dppf, 1,1′ferrocenediyl-bis(diphenylphosphine); dr, diastereomeric ratio; dtbpf, 1,1′-bis(di-tert-butylphosphino)ferrocene; EDCI, N-(3dimethylaminopropal)-N-ethylcarbodiimide; ee, enantiomeric excess; EEDQ, 2-ethoxy-1-ethoxycarbonyl-1,2-dihydroquinoline; Et, ethyl; HATU, 1-[bis(dimethylamino)methylene]-1H1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate; HBTU, (2-(1H-benzotriazol-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate; Hex, hexyl; n-Hex, n-hexyl; HMDS, hexamethyldisilazane; HOAc, acetic acid; HOAt, 1-hydroxy-7azabenzotriazole; HOBt, 1-hydroxybenzotriazole hydrate; i-Pr, isopropyl; i-PrOH, isopropyl alcohol; KOTMS, potassium trimethylsilanolate; LAH, lithium aluminum hydride; LHMDS, lithium bis(trimethylsilyl)amide; LDA, lithium diisopropylamide; m-CPBA, 3-chloroperoxybenzoic acid; Me, methyl; MeCN, acetonitrile; MEK, methyl ethyl ketone; MeTHF, 2methyltetrahydrofuran; MIBK, methyl isobutyl ketone; Ms, methylsulfonyl, mesyl; MS, molecular sieves; MTBE, methyl tert-butyl ether; n-Pr, n-propyl; NADP, nicotinamide adenine dinucleotide phosphate; NADPNa, sodium nicotinamide adenine dinucleotide phosphate; NBS, N-bromosuccinimide; Perspective REFERENCES (1) Raju, T N K The Nobel Chronicles 1988: James Whyte Black, (B 1924), Gertrude Elion (1918-99), and George H Hitchings (190598) Lancet 2000, 355, 1022 (2) (a) Li, J.; Liu, K K.-C Synthetic Approaches to the 2002 New Drugs Mini-Rev Med Chem 2004, 4, 207−233 (b) Liu, K K.-C.; Li, J.; Sakya, S Synthetic Approaches to the 2003 New Drugs Mini-Rev Med Chem 2004, 4, 1105−1125 (c) Li, J.; Liu, K K.-C.; Sakya, S Synthetic Approaches to the 2004 New Drugs Mini-Rev Med Chem 2005, 5, 1133−1144 (d) Sakya, S M.; Li, J.; Liu, K K.-C Synthetic Approaches to the 2005 New Drugs Mini-Rev Med Chem 2007, 7, 429−450 (e) Liu, K K.-C.; Sakya, S M.; Li, J Synthetic Approaches to the 2006 New Drugs Mini-Rev Med Chem 2007, 7, 1255−1269 (f) Liu, K K.-C.; Sakya, S M.; O’Donnell, C J.; Li, J Synthetic Approaches to the 2007 New Drugs Mini-Rev Med Chem 2008, 8, 1526−1548 (g) Liu, K K.-C.; Sakya, S M.; O’Donnell, C J.; Li, J Synthetic Approaches to the 2008 New Drugs Mini-Rev Med Chem 2009, 9, 1655−1675 (h) Liu, K K.-C.; Sakya, S M.; O’Donnell, C J.; Flick, A C.; Li, J Synthetic Approaches to the 2009 New Drugs Bioorg Med Chem 2011, 19, 1136−1154 (i) Liu, K K.-C.; Sakya, S M.; O’Donnell, C J.; Flick, A C.; Ding, H X Synthetic Approaches to the 2010 New Drugs Bioorg Med Chem 2012, 20, 1155−1174 (j) Ding, H X.; Liu, K K.; Sakya, S M.; Flick, A C.; O’Donnell, C J Synthetic Approaches to the 2011 New Drugs Bioorg Med Chem 2013, 21, 2795−2825 (k) Ding, H X.; Leverett, C A.; Kyne, R E., Jr.; Liu, K K.; Sakya, S M.; Flick, A C.; O’Donnell, C J Synthetic Approaches to the 2012 New Drugs Bioorg Med Chem 2014, 22, 2005−2032 (l) Ding, H X.; Leverett, C A.; Kyne, R E., Jr.; Liu, K K.; Fink, S J.; Flick, A C.; O’Donnell, C J Synthetic Approaches to the 2013 New Drugs Bioorg Med Chem 2015, 23, 1895−1922 (m) Flick, A C.; Ding, H X.; Leverett, C A.; Kyne, R E., Jr.; Liu, K K.; Fink, S J.; O’Donnell, C J Synthetic Approaches to the 2014 New Drugs Bioorg Med Chem 2016, 24, 1937−1980 (n) Flick, A C.; Ding, H X.; Leverett, C A.; Kyne, R E.; Liu, K K C.; Fink, S J.; O’Donnell, C J Synthetic Approaches to the New Drugs Approved During 2015 J Med Chem 2017, 60, 6480−6515 (o) Flick, A C.; Ding, H X.; Leverett, C A.; Fink, S J.; O’Donnell, C J Synthetic Approaches to New Drugs Approved During 2016 J Med Chem 2018, 61, 7004−7031 (p) Flick, A C.; Leverett, C A.; Ding, H X.; McInturff, E.; Fink, S J.; Helal, C J.; O’Donnell, C J Synthetic Approaches to the New Drugs Approved During 2017 J Med Chem 2019, 62, 7340−7382 (3) Graul, A I.; Pina, P.; Cruces, E.; Stringer, M The Year’s New Drugs & Biologics 2018: Part I Drugs Today 2019, 55, 35 AR https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc (4) For a more detailed description of the medicinal chemistry strategy, preclinical and clinical pharmacology of these drugs, see the following: To Market, To Market2018 In 2019 Medicinal Chemistry Reviews; Bronson, J J., Ed.; ACS Division of Medicinal Chemistry: Cambridge, MA, 2019; Vol 54, pp 469−596 (5) (a) Chopra, S.; Dasgupta, A Eravacycline Drugs Future 2014, 39, 247−256 (b) Scott, L J Eravacycline: A Review in Complicated Intra-Abdominal Infections Drugs 2019, 79, 315−324 (c) Zhanel, G G.; Cheung, D.; Adam, H.; Zelenitsky, S.; Golden, A.; Schweizer, F.; Gorityala, B.; Lagace-Wiens, P R S.; Walkty, A.; Gin, A S.; Hoban, D J.; Karlowsky, J A Review of Eravacycline, a Novel Fluorocycline Antibacterial Agent Drugs 2016, 76, 567−588 (6) (a) Brubaker, J D.; Myers, A G A Practical, Enantioselective Synthetic Route to a Key Precursor to the Tetracycline Antibiotics Org Lett 2007, 9, 3523−3525 (b) Charest, M G.; Lerner, C D.; Brubaker, J D.; Siegel, D R.; Myers, A G A Convergent Enantioselective Route to Structurally Diverse 6-Deoxytetracycline Antibiotics Science 2005, 308, 395−398 (c) Myers, A G.; Kummer, D A.; Li, D.; Hecker, E.; Dion, A.; Wright, P M Synthesis of Tetracyclines and Intermediates for the Treatment of Infection WO 2010126607A2, 2010 (7) (a) Xiao, X.-Y.; Hunt, D K.; Zhou, J.; Clark, R B.; Dunwoody, N.; Fyfe, C.; Grossman, T H.; O’Brien, W J.; Plamondon, L.; Ronn, M.; Sun, C.; Zhang, W.-Y.; Sutcliffe, J A Fluorocyclines 7-Fluoro9-Pyrrolidinoacetamido-6-Demethyl-6-Deoxytetracycline: A Potent, Broad Spectrum Antibacterial Agent J Med Chem 2012, 55, 597− 605 (b) Zhang, W.-Y.; Che, Q.; Crawford, S.; Ronn, M.; Dunwoody, N A Divergent Route to Eravacycline J Org Chem 2017, 82, 936− 943 (c) Zhou, J.; Xiao, X.-Y.; Plamondon, L.; Hunt, D K.; Clark, R B.; Zahler, R B Preparation of C7-Fluoro Substituted Tetracycline Compounds as Antibacterial Agents WO2010017470A1, 2010 (8) (a) Ronn, M.; Zhu, Z.; Hogan, P C.; Zhang, W.-Y.; Niu, J.; Katz, C E.; Dunwoody, N.; Gilicky, O.; Deng, Y.; Hunt, D K.; He, M.; Chen, C.-L.; Sun, C.; Clark, R B.; Xiao, X.-Y Process R&D of Eravacycline: The First Fully Synthetic Fluorocycline in Clinical Development Org Process Res Dev 2013, 17, 838−845 (b) Zhang, W.-Y.; Chen, C.-L.; He, M.; Zhu, Z.; Hogan, P.; Gilicky, O.; Dunwoody, N.; Ronn, M Process Research and Development of TP808: A Key Intermediate for the Manufacture of Synthetic Tetracyclines Org Process Res Dev 2017, 21, 377−386 (c) Zhang, W.-Y.; Hogan, P C.; Chen, C.-L.; Niu, J.; Wang, Z.; Lafrance, D.; Gilicky, O.; Dunwoody, N.; Ronn, M Process Research and Development of an Enantiomerically Enriched Allylic Amine, One of the Key Intermediates for the Manufacture of Synthetic Tetracyclines Org Process Res Dev 2015, 19, 1784−1795 (9) https://www.eisai.com/news/2018/news201806.html (accessed Dec 3, 2019) (10) Yamaguchi, H Potential of Ravuconazole and Its Prodrugs as the New Oraltherapeutics for Onychomycosis Med Mycol J 2016, 57, E93−E110 (11) Hata, K.; Kimura, J.; Miki, H.; Toyosawa, T.; Nakamura, T.; Katsu, K In Vitro and in Vivo Antifungal Activities of ER-30346, a Novel Oral Triazole with a Broad Antifungal Spectrum Antimicrob Agents Chemother 1996, 40, 2237−2242 (12) (a) Watanabe, S.; Tsubouchi, I.; Okubo, A Efficacy and Safety of Fosravuconazole L-Lysine Ethanolate, a Novel Oral Triazole Antifungal Agent, for the Treatment of Onychomycosis: A Multicenter, Double-Blind, Randomized Phase III Study J Dermatol 2018, 45, 1151−1159 (b) Torrico, F.; Gascon, J.; Ortiz, L.; Alonso-Vega, C.; Pinazo, M.-J.; Schijman, A.; Almeida, I C.; Alves, F.; StrubWourgaft, N.; Ribeiro, I Treatment of Adult Chronic Indeterminate Chagas Disease with Benznidazole and Three E1224 Dosing Regimens: A Proof-of-Concept, Randomised, Placebo-Controlled Trial Lancet Infect Dis 2018, 18, 419−430 (13) (a) Xu, L.; Muller, M R.; Yu, X.; Zhu, B.-Q Improved Chiral Synthesis of Ravuconazole Synth Commun 2009, 39, 1611−1625 (b) Tsuruoka, A.; Kaku, Y.; Kakinuma, H.; Tsukada, I.; Yanagisawa, M.; Nara, K.; Naito, T Synthesis and Antifungal Activity of Novel Thiazole-Containing Triazole Antifungals II Optically Active ER- 30346 and Its Derivatives Chem Pharm Bull 1998, 46, 623−630 (c) Tamura, K.; Kumagai, N.; Shibasaki, M An Enantioselective Synthesis of the Key Intermediate for Triazole Antifungal Agents; Application to the Catalytic Asymmetric Synthesis of Efinaconazole (Jublia) J Org Chem 2014, 79, 3272−3278 (d) Soukup, M Process for the Manufacture of Enantiomerically Pure Antifungal Azoles as Ravuconazole and Isavuconazole US20110087030A1, 2011 (e) Soukup, M Intermediate Halophenyl Derivatives and Their Use in a Process for Preparing Azole Derivatives WO2003002498A1, 2003 (f) Souza, A S D Process for Selectively Preparing (1H-1,2,4Triazol-1-yl)alkanols, Hydrazinyl Alkanol Compound Produced by Said Process and Use Thereof WO2015058272A1, 2015 (g) Souza, A S D.; Mendes, B L D M E Process for Selectively Preparing (1H-1,2,4-Triazol-1-yl)alkanols, Hydrazinyl Alkanol Compound Produced by Said Process and Use Thereof BR102013027313B1, 2013 (h) Shalini, K.; Kumar, N.; Drabu, S.; Sharma, P K Advances in Synthetic Approach to and Antifungal Activity of Triazoles Beilstein J Org Chem 2011, 7, 668−677 (i) Naito, T.; Hata, K.; Tsuruoka, A ER-30346 Triazole Antifungal Drugs Future 1996, 21, 20−24 (j) Lindrud, M D.; Kim, S.; Wei, C Sonic Impinging Jet Crystallization Apparatus and Process WO2000044468A1, 2000 (14) Pesti, J.; Chen, C.-K.; Spangler, L.; DelMonte, A J.; Benoit, S.; Berglund, D.; Bien, J.; Brodfuehrer, P.; Chan, Y.; Corbett, E.; Costello, C.; DeMena, P.; Discordia, R P.; Doubleday, W.; Gao, Z.; Gingras, S.; Grosso, J.; Haas, O.; Kacsur, D.; Lai, C.; Leung, S.; Miller, M.; Muslehiddinoglu, J.; Nguyen, N.; Qiu, J.; Olzog, M.; Reiff, E.; Thoraval, D.; Totleben, M.; Vanyo, D.; Vemishetti, P.; Wasylak, J.; Wei, C The Process Development of Ravuconazole: An Efficient Multikilogram Scale Preparation of an Antifungal Agent Org Process Res Dev 2009, 13, 716−728 (15) Gao, Q.; Chen, C.-P H.; Fakes, M G.; Pendri, Y R.; Kiau, S.; Vakkalagadda, B Preparation of Mono-Lysine Salts of Azole Compounds as Fungicides WO2006118351A1, 2006 (16) Wang, Y.; Castaner, R.; Bolos, J.; Estivill, C Omadacycline: Tetracycline Antibiotic Drugs Future 2009, 34, 11−15 (17) Markham, A.; Keam, S J Omadacycline: First Global Approval Drugs 2018, 78, 1931−1937 (18) (a) Chung, J Y L.; Hartner, F W.; Cvetovich, R J Synthesis Development of an Aminomethylcycline Antibiotic Via an Electronically Tuned Acyliminium Friedel-Crafts Reaction Tetrahedron Lett 2008, 49, 6095−6100 (b) Johnston, S.; Warchol, T Methods for Synthesizing and Purifying Aminoalkyl Tetracycline Compounds WO2008134048A1, 2008 (c) Tanaka, S K.; Steenbergen, J.; Villano, S Discovery, Pharmacology, and Clinical Profile of Omadacycline, a Novel Aminomethylcycline Antibiotic Bioorg Med Chem 2016, 24, 6409−6419 (19) (a) https://urldefense.proofpoint.com/v2/url?u=https-3A www.fda.gov_drugs_new-2Ddrugs-2Dfda-2Dcders-2Dnew2Dmolecular-2Dentities-2Dand-2Dnew-2Dtherapeutic-2Dbiological2Dproducts_novel-2Ddrug-2Dapprovals-2D2018&d=DwMFoQ&c= UE1eNsedaKncO0Yl_u8bfw&r=MKA0UqpxRyImwzO_ tEhpkCczC9cjy5h9oz_Ouk5Cc5s&m= CVuOqytT8wbiBPnC7O4FpCDUO9pblLXS3us5ofOd6v4&s= R3DCQACVOkMOAMrTxYddEgl3XJs1w-cCwklKJy_myRo&e= (accessed Dec 16, 2019) (b) https://www.globenewswire.com/newsrelease/2019/06/06/1865709/0/en/Achaogen-Inc-AnnouncesResults-of-Auction-for-Substantially-All-Company-Assets.html (accessed Jan 3, 2020) (20) (a) Galani, I Plazomicin: Aminoglycoside Antibiotic Drugs Future 2014, 39, 25−35 (b) McCarthy, M W Plazomicin for the Treatment of Patients with Complicated Urinary Tract Infection Drugs Today 2017, 54, 513−518 (c) Eljaaly, K.; Alharbi, A.; Alshehri, S.; Ortwine, J K.; Pogue, J M Plazomicin: A Novel Aminoglycoside for the Treatment of Resistant Gram-Negative Bacterial Infections Drugs 2019, 79, 243−269 (d) Jean, S.-S.; Gould, I M.; Lee, W.-S.; Hsueh, P.-R New Drugs for Multidrug-Resistant Gram-Negative Organisms: Time for Stewardship Drugs 2019, 79, 705−714 (21) (a) Nagabhushan, T L Process for the Manufacture of 6′-NAlkyl Derivatives of Sisomicin and Verdamicin; Novel Intermediates AS Perspective https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Useful Therein, and Novel 6′-N-Alkylverdamicins Prepared Thereby US3997524A, 1976 (b) Aggen, J.; Goldblum, A A.; Linsell, M.; Dozzo, P.; Moser, H E.; Hildebrandt, D.; Gliedt, M Antibacterial Aminoglycoside Analogs WO2009067692A1, 2010 (c) Aggen, J B.; Armstrong, E S.; Goldblum, A A.; Dozzo, P.; Linsell, M S.; Gliedt, M J.; Hildebrandt, D J.; Feeney, L A.; Kubo, A.; Matias, R D.; Lopez, S.; Gomez, M.; Wlasichuk, K B.; Diokno, R.; Miller, G H.; Moser, H E Synthesis and Spectrum of the Neoglycoside ACHN490 Antimicrob Agents Chemother 2010, 54, 4636−4642 (d) Bruss, J B.; Miller, G H.; Aggen, J B.; Armstrong, E S Treatment of Urinary Tract Infections with Antibacterial Aminoglycoside Compounds WO2010132777A2, 2010 (e) Bruss, J B.; Miller, G H.; Aggen, J B.; Armstrong, E S Treatment of Klebsiella Pneumoniae Infections with Antibacterial Aminoglycoside Compounds WO2010132770A1, 2010 (f) Trend, R.; Dappen, M.; Henry, C E.; Goldblum, A A.; Aggen, J B.; Mendonca, R F d J G.; Sardinha, J C F Synthesis of Antibacterial Aminoglycoside Analogs WO2019079613A1, 2019 (22) Deeks, E D Sarecycline: First Global Approval Drugs 2019, 79, 325−329 (23) (a) Abato, P.; Assefa, H.; Berniac, J.; Bhatia, B.; Bowser, T.; Grier, M.; Honeyman, L.; Ismail, M.; Kim, O K.; Nelson, M.; Pan, J.; Verma, A Substituted Tetracycline Compounds for Treatment of Bacterial Infections and Neoplasms WO2008079339A1, 2008 (b) Nelson, M L.; McIntyre, L Preparation of 7-Substituted Fused Ring Tetracycline Compounds as Antibiotics WO2001087824A1, 2001 (24) Seyedi, F.; Warchol, T.; Grier, M Methods for Synthesizing Substituted Tetracycline Compounds WO2009009042A1, 2009 (25) Heo, Y.-A Baloxavir: First Global Approval Drugs 2018, 78, 693−697 (26) https://www.fda.gov/news-events/press-announcements/fdaapproves-new-drug-treat-influenza (accessed Dec 3, 2019) (27) (a) Yang, T Baloxavir Marboxil: The First Cap-Dependent Endonuclease Inhibitor for the Treatment of Influenza Ann Pharmacother 2019, 53, 754−759 (b) Locke, S C.; Splawn, L M.; Cho, J C Baloxavir Marboxil: A Novel Cap-Dependent Endonuclease (CEN) Inhibitor for the Treatment of Acute Uncomplicated Influenza Drugs Today 2019, 55, 359−366 (28) (a) Kawai, M.; Tomita, K.; Akiyama, T.; Okano, A.; Miyagawa, M Substituted Polycyclic Pyridone Derivative and Prodrug Thereof WO2016175224A1, 2016 (b) Shibahara, S.; Fukui, N.; Maki, T.; Anan, K Method for Producing Substituted Polycyclic Pyridone Derivative and Crystal of Same WO2017221869A1, 2017 (c) Shishido, T.; Noshi, T.; Yamamoto, A.; Kitano, M Medicine for Treating Influenza Characterized by Comprising Combination of Cap-Dependent Endonuclease Inhibitor with Anti-Influenza Drug WO2017104691A1, 2017 (d) Kawai, M.; Tomita, K.; Akiyama, T.; Okano, A.; Miyagawa, M Pharmaceutical Compositions Containing Substituted Polycyclic Pyridone Derivatives and Prodrug Thereof US2019169206A1, 2019 (29) Tsiang, M.; Jones, G S.; Goldsmith, J.; Mulato, A.; Hansen, D.; Kan, E.; Tsai, L.; Bam, R A.; Stepan, G.; Stray, K M.; NiedzielaMajka, A.; Yant, S R.; Yu, H.; Kukolj, G.; Cihlar, T.; Lazerwith, S E.; White, K L.; Jin, H Antiviral Activity of Bictegravir (GS-9883), a Novel Potent HIV-1 Integrase Strand Transfer Inhibitor with an Improved Resistance Profile Antimicrob Agents Chemother 2016, 60, 7086−7097 (30) Deeks, E D Bictegravir/Emtricitabine/Tenofovir Alafenamide: A Review in HIV-1 Infection Drugs 2018, 78, 1817−1828 (31) (a) Bacchi, A.; Biemmi, M.; Carcelli, M.; Carta, F.; Compari, C.; Fisicaro, E.; Rogolino, D.; Sechi, M.; Sippel, M.; Sotriffer, C A.; Sanchez, T W.; Neamati, N From Ligand to Complexes Part Remarks on Human Immunodeficiency Virus Type Integrase Inhibition by Beta-Diketo Acid Metal Complexes J Med Chem 2008, 51, 7253−7264 (b) Sechi, M.; Bacchi, A.; Carcelli, M.; Compari, C.; Duce, E.; Fisicaro, E.; Rogolino, D.; Gates, P.; Derudas, M.; AlMawsawi, L Q.; Neamati, N From Ligand to Complexes: Inhibition of Human Immunodeficiency Virus Type Integrase by Beta-Diketo Acid Metal Complexes J Med Chem 2006, 49, 4248−4260 (32) (a) Chiu, A.; Enquist, J.; Griggs, N.; Hale, C.; Ikemoto, N.; Keaton, K A.; Kraft, M.; Lazerwith, S E.; Leeman, M.; Peng, Z.; Schrier, K.; Trinidad, J.; van Herpt, J.; Waltman, A W Synthesis of Polycyclic-Carbamoylpyridone Compounds US20150368264A1, 2015 (b) Hughes, D L Review of Synthetic Routes and Final Forms of Integrase Inhibitors Dolutegravir, Cabotegravir, and Bictegravir Org Process Res Dev 2019, 23, 716−729 (33) Ye, F.; Zeng, Q Method for Preparing (1r,3s)-3-Aminocyclopentanol Hydrochloride CN108774145A, 2018 (34) Michelin, R A.; Sgarbossa, P.; Scarso, A.; Strukul, G The Baeyer-Villiger Oxidation of Ketones: A Paradigm for the Role of Soft Lewis Acidity in Homogeneous Catalysis Coord Chem Rev 2010, 254, 646−660 (35) Markham, A.; Keam, S J Danoprevir: First Global Approval Drugs 2018, 78, 1271−1276 (36) Gower, E.; Estes, C.; Blach, S.; Razavi-Shearer, K.; Razavi, H Global Epidemiology and Genotype Distribution of the Hepatitis C Virus Infection J Hepatol 2014, 61, S45−S57 (37) Rajagopalan, R.; Misialek, S.; Stevens, S K.; Myszka, D G.; Brandhuber, B J.; Ballard, J A.; Andrews, S W.; Seiwert, S D.; Kossen, K Inhibition and Binding Kinetics of the Hepatitis C Virus NS3 Protease Inhibitor ITMN-191 Reveals Tight Binding and Slow Dissociative Behavior Biochemistry 2009, 48, 2559−2568 (38) (a) Blatt, L M.; Andrews, S W.; Condroski, K R.; Doherty, G A.; Jiang, Y.; Josey, J A.; Kennedy, A L.; Madduru, M R.; Stengel, P J.; Wenglowsky, S M.; Woodard, B T.; Woodard, L Preparation of Macrocyclic Carboxylic Acid Derivatives as Inhibitors of HCV Replication US20050267018A1, 2005 (b) Blatt, L M.; Wenglowsky, S M.; Andrews, S W.; Condroski, K R.; Jiang, Y.; Kennedy, A L.; Doherty, G A.; Josey, J A.; Stengel, P J.; Woodard, B T.; Madduru, M R Preparation of Macrocyclic Carboxylic Acids or Sulfonamides as Inhibitors of HCV Replication WO2005095403A2, 2005 (c) Blatt, L M.; Wenglowsky, S M.; Andrews, S W.; Jiang, Y.; Kennedy, A L.; Condroski, K R.; Josey, J A.; Stengel, P J.; Madduru, M R.; Doherty, G A.; Woodard, B T Preparation of Macrocyclic Carboxylic Acids and Acylsulfonamides as Inhibitors of HCV Replication WO2005037214A2, 2005 (39) Jiang, Y.; Andrews, S W.; Condroski, K R.; Buckman, B.; Serebryany, V.; Wenglowsky, S.; Kennedy, A L.; Madduru, M R.; Wang, B.; Lyon, M.; Doherty, G A.; Woodard, B T.; Lemieux, C.; Do, M G.; Zhang, H.; Ballard, J.; Vigers, G.; Brandhuber, B J.; Stengel, P.; Josey, J A.; Beigelman, L.; Blatt, L.; Seiwert, S D Discovery of Danoprevir (ITMN-191/R7227), a Highly Selective and Potent Inhibitor of Hepatitis C Virus (HCV) NS3/4a Protease J Med Chem 2014, 57, 1753−1769 (40) Beaulieu, P L.; Gillard, J.; Bailey, M D.; Boucher, C.; Duceppe, J.-S.; Simoneau, B.; Wang, X.-J.; Zhang, L.; Grozinger, K.; Houpis, I.; Farina, V.; Heimroth, H.; Krueger, T.; Schnaubelt, J Synthesis of (1r,2s)-1-Amino-2-Vinylcyclopropanecarboxylic Acid Vinyl-ACCA) Derivatives: Key Intermediates for the Preparation of Inhibitors of the Hepatitis C Virus NS3 Protease J Org Chem 2005, 70, 5869− 5879 (41) Burch, J D.; Sherry, B D.; Gauthier, D R.; Campeau, L.-C Discovery and Development of Doravirine: An Investigational Next Generation Non-Nucleside Reverse Transcriptase Inhibitor (NNRTI) for the Treatment of HIV In Comprehensive Accounts of Pharmaceutical Research and Development: From Discovery to LateStage Process Development Volume 1; Abdel-Magid, A F.; Pesti, J.; Vaidyanathan, R., Eds.; American Chemical Society: Washington, DC, 2016; Vol 1239, pp 175−205 (42) Colombier, M A.; Molina, J M Doravirine: A Review Curr Opin HIV AIDS 2018, 13, 308−314 (43) (a) Zhang, L K.; Yang, R.; Sheng, H.; Helmy, R.; Zheng, J.; Cao, Y.; Gauthier, D R., Jr Characterization of Impurities of HIV NNRTI Doravirine by UHPLC-High Resolution Ms and Tandem Ms Analysis J Mass Spectrom 2016, 51, 959−968 (b) Gauthier, D R., Jr.; Sherry, B D.; Cao, Y.; Journet, M.; Humphrey, G.; Itoh, T.; AT Perspective https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Mangion, I.; Tschaen, D M Highly Efficient Synthesis of HIV NNRTI Doravirine Org Lett 2015, 17, 1353−1356 (c) Cao, Y.; Donald, R.; Gauthier, J.; Humphrey, G R.; Itoh, T.; Journet, M.; Qian, G.; Sherry, B D.; Tschaen, D M Process for Making Reverse Transcriptase Inhibitors WO2015084763A2, 2015 (d) Itoh, T.; Jeon, I.; Mangion, I.; Qian, G.; Sherry, B D.; Gauthier, D R.; Cao, Y Process for Making Reverse Transcriptase Inhibitors WO2014089140A1, 2014 (e) Cote, B.; Burch, J D.; AsanteAppiah, E.; Bayly, C.; Bedard, L.; Blouin, M.; Campeau, L C.; Cauchon, E.; Chan, M.; Chefson, A.; Coulombe, N.; Cromlish, W.; Debnath, S.; Deschenes, D.; Dupont-Gaudet, K.; Falgueyret, J P.; Forget, R.; Gagne, S.; Gauvreau, D.; Girardin, M.; Guiral, S.; Langlois, E.; Li, C S.; Nguyen, N.; Papp, R.; Plamondon, S.; Roy, A.; Roy, S.; Seliniotakis, R.; St-Onge, M.; Ouellet, S.; Tawa, P.; Truchon, J F.; Vacca, J.; Wrona, M.; Yan, Y.; Ducharme, Y Discovery of MK-1439, an Orally Bioavailable Non-Nucleoside Reverse Transcriptase Inhibitor Potent against a Wide Range of Resistant Mutant HIV Viruses Bioorg Med Chem Lett 2014, 24, 917−922 (f) Burch, J.; Cote, B.; Nguyen, N.; Li, C S.; St-Onge, M.; Gauvreau, D NonNucleoside Reverse Transcriptase Inhibitors WO2011120133A1, 2011 (44) Campeau, L.-C.; Chen, Q.; Gauvreau, D.; Girardin, M l.; Belyk, K.; Maligres, P.; Zhou, G.; Gu, C.; Zhang, W.; Tan, L.; O’Shea, P D A Robust Kilo-Scale Synthesis of Doravirine Org Process Res Dev 2016, 20, 1476−1481 (45) (a) Yang, W.; Xu, L.; Makha, M.; Sandoval, C A.; Huang, C Synthesis Method of 3-Chloromethyl-1,2,4-triazoline-5-one CN104628663A, 2015 (b) Cowden, C J.; Wilson, R D.; Bishop, B C.; Cottrell, I F.; Davies, A J.; Dolling, U.-H A New Synthesis of 1,2,4-Triazolin-5-ones: Application to the Convergent Synthesis of an NK1 Antagonist Tetrahedron Lett 2000, 41, 8661−8664 (46) (a) Wilby, K J.; Eissa, N A Clinical Pharmacokinetics and Drug Interactions of Doravirine Eur Eur J Drug Metab Pharmacokinet 2018, 43, 637−644 (b) https://www.accessdata.fda gov/scripts/cder/daf/index.cfm?event=overview.process&ApplNo= 210867 (accessed Dec 3, 2019) (47) (a) Opoku, N O; Bakajika, D K; Kanza, E M; Howard, H.; Mambandu, G L; Nyathirombo, A.; Nigo, M M; Kasonia, K.; Masembe, S L; Mumbere, M.; Kataliko, K.; Larbelee, J P; Kpawor, M.; Bolay, K M; Bolay, F.; Asare, S.; Attah, S K; Olipoh, G.; Vaillant, M.; Halleux, C M; Kuesel, A C Single Dose Moxidectin Versus Ivermectin for Onchocerca Volvulus Infection in Ghana, Liberia, and the Democratic Republic of the Congo: A Randomised, Controlled, Double-Blind Phase Trial Lancet 2018, 392, 1207−1216 (b) Awadzi, K.; Opoku, N O.; Attah, S K.; Lazdins-Helds, J.; Kuesel, A C A Randomized, Single-Ascending-Dose, IvermectinControlled, Double-Blind Study of Moxidectin in Onchocerca Volvulus Infection PLoS Neglected Trop Dis 2014, 8, e2953 (18 pp) (48) Cobb, R.; Boeckh, A Moxidectin: A Review of Chemistry, Pharmacokinetics and Use in Horses Parasites Vectors 2009, 2, S5 (49) Prichard, R.; Menez, C.; Lespine, A Moxidectin and the Avermectins: Consanguinity but Not Identity Int J Parasitol.: Drugs Drug Resist 2012, 2, 134−153 (50) Lumaret, J.-P.; Errouissi, F.; Floate, K.; Rombke, J.; Wardhaugh, K A Review on the Toxicity and Non-Target Effects of Macrocyclic Lactones in Terrestrial and Aquatic Environments Curr Pharm Biotechnol 2012, 13, 1004−1060 (51) (a) Tsou, H R.; Ahmed, Z H.; Fiala, R R.; Bullock, M W.; Carter, G T.; Goodman, J J.; Borders, D B Biosynthetic Origin of the Carbon Skeleton and Oxygen Atoms of the LL-F28249α, a Potent Antiparasitic Macrolide J Antibiot 1989, 42, 398−406 (b) Song, X.; Zhang, Y.; Xue, J.; Li, C.; Wang, Z.; Wang, Y Enhancing Nemadectin Production by Streptomyces Cyaneogriseus Ssp Noncyanogenus through Quantitative Evaluation and Optimization of Dissolved Oxygen and Shear Force Bioresour Technol 2018, 255, 180−188 (52) (a) Dai, Y.; Li, H.; Wang, R Process for Eliminating Byproduct Dimethyl Sulfide from Production of Moxidectin CN105294729A, 2016 (b) Li, H.; Dai, Y.; Wang, W.; Zhao, D.; Wang, R Method for Preparing High-Purity Moxidectin Using Membrane Separation CN104356140A, 2015 (c) He, Y.; Wen, J.; Zhang, H Method for Preparing Moxidectin CN104277050A, 2015 (d) Dai, Y.; Du, Z.; Wang, R Preparation of Moxidectin CN104017001B, 2016 (53) Pfitzner, K E.; Moffatt, J G A New and Selective Oxidation of Alcohols J Am Chem Soc 1963, 85, 3027−3028 (54) Awasthi, A.; Razzak, M.; Al-Kassas, R.; Harvey, J.; Garg, S Analytical Profile of Moxidectin Profiles Drug Subst., Excipients, Relat Methodol 2013, 38, 315−366 (55) White, N J Tafenoquine - a Radical Improvement? N Engl J Med 2019, 380, 285−286 (56) Blumbergs, P.; LaMontagne, M P 4-Methyl-5-[(Un)Substituted Phenoxy]-2,6-dimethoxy-8-(aminoalkylamino)quinolines US4617394A, 1986 (57) Vennerstrom, J L.; Nuzum, E O.; Miller, R E.; Dorn, A.; Gerena, L.; Dande, P A.; Ellis, W Y.; Ridley, R G.; Milhous, W K 8Aminoquinolines Active against Blood Stage Plasmodium Falciparum in Vitro Inhibit Hematin Polymerization Antimicrob Agents Chemother 1999, 43, 598−602 (58) (a) Carvalho, L.; Luque-Ortega, J R.; Manzano, J I.; Castanys, S.; Rivas, L.; Gamarro, F Tafenoquine, an Antiplasmodial 8Aminoquinoline, Targets Leishmania Respiratory Complex III and Induces Apoptosis Antimicrob Agents Chemother 2010, 54, 5344− 5351 (b) Lanners, H N Effect of the 8-Aminoquinoline Primaquine on Culture-Derived Gametocytes of the Malaria Parasite Plasmodium Falciparum Z Parasitenkd 1991, 77, 478−481 (59) McIntyre, J A.; Castaner, J.; Bayes, M Tafenoquine Succinate: Antimalarial Drugs Future 2003, 28, 859−869 (60) Bell, D.; Davies, B J.; Kincey, P M Process for the Preparation of Quinoline Derivatives WO2003093239A2, 2003 (61) Jordan, R.; Leeds, J M.; Tyavanagimatt, S.; Hruby, D E Development of ST-246 for Treatment of Poxvirus Infections Viruses 2010, 2, 2409−2435 (62) Hoy, S M Tecovirimat: First Global Approval Drugs 2018, 78, 1377−1382 (63) (a) Merchlinsky, M.; Albright, A.; Olson, V.; Schiltz, H.; Merkeley, T.; Hughes, C.; Petersen, B.; Challberg, M The Development and Approval of Tecoviromat (TPOXX®), the First Antiviral against Smallpox Antiviral Res 2019, 168, 168−174 (b) Grosenbach, D W.; Honeychurch, K.; Rose, E A.; Chinsangaram, J.; Frimm, A.; Maiti, B.; Lovejoy, C.; Meara, I.; Long, P.; Hruby, D E Oral Tecovirimat for the Treatment of Smallpox N Engl J Med 2018, 379, 44−53 (64) Hughes, D L Review of the Patent Literature: Synthesis and Final Forms of Antiviral Drugs Tecovirimat and Baloxavir Marboxil Org Process Res Dev 2019, 23, 1298−1307 (65) (a) Jordan, R.; Bailey, T R.; Rippin, S R.; Dai, D Preparation of Isoindole Derivatives for Treatment and Prevention of Orthopoxvirus Infections WO2008130348A1, 2008 (b) Jordan, R.; Bailey, T R.; Rippin, S R.; Dai, D Compounds, Compositions and Methods for Treatment and Prevention of Orthopoxvirus Infections and Associated Diseases US20120020922A1, 2012 (66) Tyavanagimatt, S R.; Stone, M A C L.; Weimers, W C.; Nelson, D.; Bolken, T C.; Hruby, D E.; O’Neill, M H.; Sweetapple, G.; McCloughan, K A Polymorphic Forms ST-246 WO2011119698A1, 2011 (67) https://www.fda.gov/drugs/resources-information-approveddrugs/fda-approves-avatrombopag-thrombocytopenia-adults-chronicliver-disease (accessed Dec 3, 2019) (68) https://www.ema.europa.eu/documents/smop-initial/chmpsummary-positive-opinion-doptelet_en.pdf (accessed Dec 3, 2019) (69) Shirley, M Avatrombopag: First Global Approval Drugs 2018, 78, 1163−1168 (70) (a) Sugasawa, K.; Watanuki, S.; Koga, Y.; Nagata, H.; Obitsu, K.; Wakayama, R.; Hirayama, F.; Suzuki, K.-I 2-Acylaminothiazole Derivative or Salt Thereof WO2003062233A1, 2003 (b) Sugasawa, K.; Koga, Y.; Hirayama, F.; Suzuki, K.-I.; Awamura, Y Novel Salt of 2Acylaminothiazole Derivative WO2004029049A1, 2004 (c) Sugasawa, K.; Miyafuji, A.; Suzuki, K.; Awamura, Y 2-Acylaminothiazole Compound Crystals WO2013018362A1, 2013 AU Perspective https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc (71) Heo, Y.-A Revefenacin: First Global Approval Drugs 2019, 79, 85−91 (72) Li, F.; Yang, J Revefenacin for the Treatment of Chronic Obstructive Pulmonary Disease Expert Rev Clin Pharmacol 2019, 12, 293−298 (73) (a) Axt, S.; Church, T J.; Malathong, V Crystalline Forms of a Biphenyl Compound WO2006099165A1, 2006 (b) Colson, P.-J Process for Preparing a Biphenyl-2-Ylcarbamic Acid WO2012009166A1, 2012 (c) Mammen, M.; Ji, Y.-H.; Mu, Y.; Husfeld, C.; Li, L Biphenyl Compounds Useful as Muscarinic Receptor Antagonists US20050203133A1, 2005 (74) Dhillon, S Roxadustat: First Global Approval Drugs 2019, 79, 563−572 (75) (a) Provenzano, R.; Besarab, A.; Sun, C H.; Diamond, S A.; Durham, J H.; Cangiano, J L.; Aiello, J R.; Novak, J E.; Lee, T.; Leong, R.; Roberts, B K.; Saikali, K G.; Hemmerich, S.; Szczech, L A.; Yu, K.-H P.; Neff, T B Oral Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitor Roxadustat (FG-4592) for the Treatment of Anemia in Patients with CKD Clin J Am Soc Nephrol 2016, 11, 982−991 (b) Yang, Y.; Yu, X.; Zhang, Y.; Ding, G.; Zhu, C.; Huang, S.; Jia, Z.; Zhang, A Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitor Roxadustat (FG-4592) Protects against Cisplatin-Induced Acute Kidney Injury Clin Sci 2018, 132, 825−838 (76) (a) Zhou, F.; Jin, H.; Zheng, Y.; Huang, M.; Meng, X Preparation Method of Roxadustat Intermediate 4-Hydroxy-1-methyl7-phenoxyisoquinoline-3-carboxylate for Treating Chronic Anemia CN106478504A, 2017 (b) Sun, T Process for Preparation of Roxadustat for Treatment of Chronic Nephrosis and Terminal Stage Nephrosis-Related Anaemia from 5-Nitrophenylethylamine CN107602466A, 2018 (c) Sun, T Preparation Method of Roxadustat for Treating Chronic Kidney Disease and Anemia Associated with End-Stage Renal Disease CN107698505A, 2018 (d) Yang, M.; Liu, W.; Wei, X.; Wang, Y.; Song, Y Preparation Method of Roxadustat WO2018072662A1, 2018 (e) Han, R.; Li, C.; Qiao, D Method for Preparing Roxadustat CN108017583A, 2018 (f) Wang, Z Method for Preparing Roxadustat CN108383787A, 2018 (g) Zheng, X.; Zhang, Y Preparation Method of Roxadustat (Fg-459) for Treating Chronic Anaemia CN108424388A, 2018 (h) Xu, Y Synthesis Method of Roxadustat, and Intermediate Compound Thereof CN108794397A, 2018 (77) Arend, M P.; Flippin, L A.; Guenzler-Pukall, V.; Ho, W.-B.; Turtle, E D.; Du, X Preparation of Isoquinolinecarboxamides and Their Use in Mediating Hypoxia Inducible Factor and Increasing Endogenous Erythropoietin WO2004108681A1, 2004 (78) Witschi, C.; Park, J M.; Thompson, M D.; Martinelli, M J.; Yeowell, D A.; Arend, M P Crystalline Forms of a Isoquinolinecarbonylaminoacetic Acid Derivative as a Prolyl Hydroxylase Inhibitor WO2014014835A2, 2014 (79) Chedid, V.; Vijayvargiya, P.; Camilleri, M Elobixibat for the Treatment of Constipation Expert Rev Gastroenterol Hepatol 2018, 12, 951−960 (80) Nelson, A D.; Camilleri, M.; Chirapongsathorn, S.; Vijayvargiya, P.; Valentin, N.; Shin, A.; Erwin, P J.; Wang, Z.; Murad, M H Comparison of Efficacy of Pharmacological Treatments for Chronic Idiopathic Constipation: A Systematic Review and Network Meta-Analysis Gut 2017, 66, 1611 (81) Miner, P B., Jr Elobixibat, the First-in-Class Ileal Bile Acid Transporter Inhibitor, for the Treatment of Chronic Idiopathic Constipation Expert Opin Pharmacother 2018, 19, 1381−1388 (82) (a) Starke, I.; Dahlstrom, M.; Blomberg, D Preparation of N[(8-Benzothiazepinyloxy)acetyl]phenylglycinates and Analogs as Ileal Bile Acid Transport Inhibitors WO2002050051A1, 2002 (b) Starke, I.; Dahlstrom, M.; Blomberg, D Preparation of 1,5-Benzothiazepines for Use as Hypolipidemics WO2001066533A1, 2001 (c) Starke, I.; Dahlstrom, M U J.; Blomberg, D.; Alenfalk, S.; Nordberg, P.; Wallberg, A C.; Bostrom, S J Preparation of Benzothiazepine Derivatives for Potential Use as Ileal Bile Acid Transport Inhibitors for the Treatment of Hyperlipidemia WO2003020710A1, 2003 (83) Takahashi, N.; Take, Y Tegoprazan, a Novel PotassiumCompetitive Acid Blocker to Control Gastric Acid Secretion and Motility J Pharmacol Exp Ther 2018, 364, 275−286 (84) Lee, K J.; Son, B K.; Kim, G H.; Jung, H.-K.; Jung, H.-Y.; Chung, I.-K.; Sung, I.-K.; Kim, J I.; Kim, J H.; Lee, J S.; Kwon, J G.; Park, J H.; Huh, K C.; Park, K S.; Park, M.-I.; Kim, N.; Lee, O Y.; Jee, S R.; Lee, S K.; Youn, S J.; Kim, S K.; Lee, S T.; Hong, S J.; Choi, S C.; Kim, T N.; Youn, Y H.; Park, H J.; Kang, M J.; Park, C H.; Kim, B T.; Youn, S.; Song, G S.; Rhee, P.-L Randomised Phase Trial: Tegoprazan, a Novel Potassium-Competitive Acid Blocker, Vs Esomeprazole in Patients with Erosive Oesophagitis Aliment Pharmacol Ther 2019, 49, 864−872 (85) Hanazawa, T.; Koike, H Chromane Substituted Benzimidazole Derivatives US20070142448A1, 2007 (86) Xiaolong, Q.; Lin, H.; Wenbo, L.; Ping, Z.; Lingling, C.; Xingang, Z.; Ping, W.; Donghui, W.; Lei, C.; Jun, C Method for Synthetizing Tegoprazan Chiral Alcohol CN109320485, 2019 (87) Lamb, Y N Elagolix: First Global Approval Drugs 2018, 78, 1501−1508 (88) Barra, F.; Grandi, G.; Tantari, M.; Scala, C.; Facchinetti, F.; Ferrero, S A Comprehensive Review of Hormonal and Biological Therapies for Endometriosis: Latest Developments Expert Opin Biol Ther 2019, 19, 343−360 (89) (a) Pokrzywinski, R M.; Soliman, A M.; Chen, J.; Snabes, M.; Diamond, M P.; Surrey, E.; Coyne, K S Impact of Elagolix on Work Loss Due to Endometriosis-Associated Pain: Estimates Based on the Results of Two Phase III Clinical Trials Fertil Steril 2019, 112, 545− 551 (b) Sidduri, A.; Tilley, J W.; Hull, K.; Ping Lou, J.; Kaplan, G.; Sheffron, A.; Chen, L.; Campbell, R.; Guthrie, R.; Huang, T.-N.; Huby, N.; Rowan, K.; Schwinge, V.; Renzetti, L M N-CycloalkanoylL-Phenylalanine Derivatives as VCAM/VLA-4 Antagonists Bioorg Med Chem Lett 2002, 12, 2475−2478 (90) (a) Chen, C.; Wu, D.; Guo, Z.; Xie, Q.; Reinhart, G J.; Madan, A.; Wen, J.; Chen, T.; Huang, C Q.; Chen, M.; Chen, Y.; Tucci, F C.; Rowbottom, M.; Pontillo, J.; Zhu, Y.-F.; Wade, W.; Saunders, J.; Bozigian, H.; Struthers, R S Discovery of Sodium R-(+)-4-{2-[5-(2Fluoro-3-Methoxyphenyl)-3-(2-Fluoro-6-[Trifluoromethyl]Benzyl)-4Methyl-2,6-Dioxo-3,6-Dihydro-2h-Pyrimidin-1-Yl]-1Phenylethylamino}Butyrate (Elagolix), a Potent and Orally Available Nonpeptide Antagonist of the Human Gonadotropin-Releasing Hormone Receptor J Med Chem 2008, 51, 7478−7485 (b) Gallagher, D J.; Treiber, L R.; Hughes, R M.; Campopiano, O.; Wang, P.; Zhao, Y.; Chou, S K.; Ouellette, M A.; Hettinger, D N Processes for the Preparation of Uracil Derivatives US8765948B2, 2014 (91) (a) Friedman-Kien, A E Disseminated Kaposi’s Sarcoma Syndrome in Young Homosexual Men J Am Acad Dermatol 1981, 5, 468 (b) Markham, A Fostamatinib: First Global Approval Drugs 2018, 78, 959−963 (92) (a) Hymes, K B.; Greene, J B.; Marcus, A.; William, D C.; Cheung, T.; Prose, N S.; Ballard, H.; Laubenstein, L J Kaposi’s Sarcoma in Homosexual Men-a Report of Eight Cases Lancet 1981, 318, 598−600 (b) Gallo, R C.; Sarin, P S.; Gelmann, E P.; RobertGuroff, M.; Richardson, E.; Kalyanaraman, V S.; Mann, D.; Sidhu, G D.; Stahl, R E.; Zolla-Pazner, S.; Leibowitch, J.; Popovic, M Isolation of Human T-Cell Leukemia Virus in Acquired Immune Deficiency Syndrome (AIDS) Science 1983, 220, 865 (93) Bussel, J.; Arnold, D M.; Grossbard, E.; Mayer, J.; Trelinski, J.; Homenda, W.; Hellmann, A.; Windyga, J.; Sivcheva, L.; Khalafallah, A A.; Zaja, F.; Cooper, N.; Markovtsov, V.; Zayed, H.; Duliege, A.-M Fostamatinib for the Treatment of Adult Persistent and Chronic Immune Thrombocytopenia: Results of Two Phase 3, Randomized, Placebo-Controlled Trials Am J Hematol 2018, 93, 921−930 (94) (a) Singh, R.; Masuda, E S.; Payan, D G Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors J Med Chem 2012, 55, 3614−3643 (b) Mocsai, A.; Ruland, J.; Tybulewicz, V L J The SYK Tyrosine Kinase: A Crucial Player in Diverse Biological Functions Nat Rev Immunol 2010, 10, 387−402 AV Perspective https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc (95) Newland, A.; Lee, E.-J.; McDonald, V.; Bussel, J B Fostamatinib for Persistent/Chronic Adult Immune Thrombocytopenia Immunotherapy 2018, 10, 9−25 (96) Sweeny, D J.; Li, W.; Clough, J.; Bhamidipati, S.; Singh, R.; Park, G.; Baluom, M.; Grossbard, E.; Lau, D T W Metabolism of Fostamatinib, the Oral Methylene Phosphate Prodrug of the Spleen Tyrosine Kinase Inhibitor R406 in Humans: Contribution of Hepatic and Gut Bacterial Processes to the Overall Biotransformation Drug Metab Dispos 2010, 38, 1166−1176 (97) McKeever, B.; Diorazio, L J.; Jones, M F.; Ferris, L.; Janbon, S L M.; Siedlecki, P S.; Churchill, G H.; Crafts, P A Preparation of Pharmaceutical 2,4-Pyrimidinediamines in Large Scale WO2015095765A1, 2015 (98) Hart, R.; Herring, A.; Howell, G P.; McKeever-Abbas, B.; Pedge, N.; Woodward, R Real-Time Monitoring and Control of Critical Process Impurities During the Manufacture of Fostamatinib Disodium Org Process Res Dev 2015, 19, 537−542 (99) Miyazaki, H.; Ikeda, Y.; Sakurai, O.; Miyake, T.; Tsubota, R.; Okabe, J.; Kuroda, M.; Hisada, Y.; Yanagida, T.; Yoneda, H.; Tsukumo, Y.; Tokunaga, S.; Kawata, T.; Ohashi, R.; Fukuda, H.; Kojima, K.; Kannami, A.; Kifuji, T.; Sato, N.; Idei, A.; Iguchi, T.; Sakairi, T.; Moritani, Y Discovery of Evocalcet, a Next-Generation Calcium-Sensing Receptor Agonist for the Treatment of Hyperparathyroidism Bioorg Med Chem Lett 2018, 28, 2055−2060 (100) Sekercioglu, N.; Busse, J W.; Mustafa, R A.; Guyatt, G H.; Thabane, L Cinacalcet Versus Standard Treatment for Chronic Kidney Disease: A Protocol for a Systematic Review and MetaAnalysis Syst Rev 2016, 5, (101) (a) Fukagawa, M.; Shimazaki, R.; Akizawa, T Evocalcet study, g Head-to-Head Comparison of the New Calcimimetic Agent Evocalcet with Cinacalcet in Japanese Hemodialysis Patients with Secondary Hyperparathyroidism Kidney Int 2018, 94, 818−825 (b) Nemeth, E F.; Van Wagenen, B C.; Balandrin, M F Discovery and Development of Calcimimetic and Calcilytic Compounds Prog Med Chem 2018, 57, 1−86 (102) Tomoyuki, N.; Yuusuke, M.; Michihiro, M.; Atsushi, H Polyethylene Glycol Derivative WO2018174283A1, 2018 (103) Syed, Y Y Anlotinib: First Global Approval Drugs 2018, 78, 1057−1062 (104) Shen, G.; Zheng, F.; Ren, D.; Du, F.; Dong, Q.; Wang, Z.; Zhao, F.; Ahmad, R.; Zhao, J Anlotinib: A Novel Multi-Targeting Tyrosine Kinase Inhibitor in Clinical Development J Hematol Oncol 2018, 11, 120 (105) (a) Xie, C.; Wan, X.; Quan, H.; Zheng, M.; Fu, L.; Li, Y.; Lou, L Preclinical Characterization of Anlotinib, a Highly Potent and Selective Vascular Endothelial Growth Factor Receptor-2 Inhibitor Cancer Sci 2018, 109, 1207−1219 (b) Sun, Y.; Niu, W.; Du, F.; Du, C.; Li, S.; Wang, J.; Li, L.; Wang, F.; Hao, Y.; Li, C.; Chi, Y Safety, Pharmacokinetics, and Antitumor Properties of Anlotinib, an Oral Multi-Target Tyrosine Kinase Inhibitor, in Patients with Advanced Refractory Solid Tumors J Hematol Oncol 2016, 9, 105 (106) Syrigos, K N.; Saif, M W.; Karapanagiotou, E M.; Oikonomopoulos, G.; De Marinis, F The Need for Third-Line Treatment in Non-Small Cell Lung Cancer: An Overview of New Options Anticancer Res 2011, 31, 649−659 (107) Lin, B.; Song, X.; Yang, D.; Bai, D.; Yao, Y.; Lu, N Anlotinib Inhibits Angiogenesis Via Suppressing the Activation of VEGFR2, PDGFRβ and FGFR1 Gene 2018, 654, 77−86 (108) Han, B.; Li, K.; Zhao, Y.; Li, B.; Cheng, Y.; Zhou, J.; Lu, Y.; Shi, Y.; Wang, Z.; Jiang, L.; Luo, Y.; Zhang, Y.; Huang, C.; Li, Q.; Wu, G Anlotinib as a Third-Line Therapy in Patients with Refractory Advanced Non-Small-Cell Lung Cancer: A Multicentre, Randomised Phase II Trial (ALTER0302) Br J Cancer 2018, 118, 654−661 (109) (a) Chen, G P Substituted Quinoline Compounds as Angiogenesis Inhibitors and Their Preparation and Use in the Treatment of Angiogenesis and Cancer WO2008112407A1, 2008 (b) Chen, G P C Kinase Inhibitors for the Treatment of Disease Associated with Angiogenesis WO2010021918A1, 2010 (c) Chen, G P.; Yan, C Process for Preparing an Anti-Cancer Agent, 1-{[4-(4- Fluoro-2-methyl-1H-indol-5-yloxy)-6-methoxyquinolin-7-yloxy]methyl}cyclopropanamine, Its Crystalline Form and Its Salts WO2016179123A1, 2016 (110) (a) Zhao, R.; Zhang, X.; Gao, Y.; Chen, G.; Wang, Q.; Chen, Z.; Dong, P.; Jin, J Crystal of Quinoline Derivative and Preparation Method Thereof CN102344438A, 2012 (b) Zhang, X.; Wang, X.; Jiang, H.; Tian, X.; Yang, L Quinoline Derivative for Treatment of Neuroendocrine Tumors WO2019052520A1, 2019 (111) Mitsuya, H.; Weinhold, K J.; Furman, P A.; St Claire, M H.; Lehrman, S N.; Gallo, R C.; Bolognesi, D.; Barry, D W.; Broder, S 3′-Azido-3′-Deoxythymidine (BW A509U): An Antiviral Agent That Inhibits the Infectivity and Cytopathic Effect of Human TLymphotropic Virus Type III/Lymphadenopathy-Associated Virus in Vitro Proc Natl Acad Sci U S A 1985, 82, 7096 (112) Al-Salama, Z T Apalutamide: First Global Approval Drugs 2018, 78, 699−705 (113) Jung, M.; Yoo, D.; Sawyers, C L.; Tran, C Preparation of Diarylthiohydantoins as Androgen Receptor Antagonists for the T r e a t m en t o f Ho r m o n e R e fr a c t o r y P r o st a t e C a n c e r US20070254933A1, 2007 (114) Roberts, J D.; Bebenek, K.; Kunkel, T A The Accuracy of Reverse Transcriptase from HIV-1 Science 1988, 242, 1171 (115) Isaacsson Velho, P.; Carducci, M A Investigational Therapies Targeting the Androgen Signaling Axis and the Androgen Receptor and in Prostate Cancer - Recent Developments and Future Directions Expert Opin Invest Drugs 2018, 27, 811−822 (116) Bambury, R M.; Rathkopf, D E Novel and Next-Generation Androgen Receptor-Directed Therapies for Prostate Cancer: Beyond Abiraterone and Enzalutamide Urol Oncol.: Semin Orig Invest 2016, 34, 348−355 (117) Smith, M R.; Saad, F.; Chowdhury, S.; Oudard, S.; Hadaschik, B A.; Graff, J N.; Olmos, D.; Mainwaring, P N.; Lee, J Y.; Uemura, H.; Lopez-Gitlitz, A.; Trudel, G C.; Espina, B M.; Shu, Y.; Park, Y C.; Rackoff, W R.; Yu, M K.; Small, E J Apalutamide Treatment and Metastasis-Free Survival in Prostate Cancer N Engl J Med 2018, 378, 1408−1418 (118) (a) Qin, Y.; Chen, Y.; Bian, J.; Yu, J.; Wang, M.; Dai, X Preparation of Apalutamide CN107501237A, 2017 (b) Bian, H.; Yang, J.; Nie, F.; Yang, Y.; Jiang, X Preparation of Diaryl Thiohydantoin Compound Apalutamide CN108047200A, 2018 (c) Hu, Z.; Wei, D.; Zhu, X.; Wang, S Preparing Method of Apalutamide and Intermediates CN108069869A, 2018 (d) Muthusamy, A R.; Kanniah, S L.; Arote, N D.; Bhagwat, O V.; Sonar, J K.; Poundkar, V B.; Wagh, Y D Solid State Forms of Apalutamide WO2018112001A1, 2018 (e) Chen, S.-H.; Guo, J.-C.; Shih, W.-L Process for Preparing Apalutamide US20180201601A1, 2018 (f) Pawar, A A.; Subba Reddy, P R.; Mudapaka, V K.; Gopi, S P.; Reddy, K R S.; Verma, H Amorphous Solid Dispersions of Apalutamide and Process for the Preparation Thereof WO2019016747A1, 2019 (119) (a) Smith, N D.; Bonnefous, C.; Julien, J D Preparation of Spiro Thiohydantoin Compounds as Androgen Receptor Modulators WO2011103202A2, 2011 (b) Haim, C B.; Horvath, A.; Weerts, J E E Process for the Preparation of Apalutamide WO2016100652A2, 2016 (c) Haim, C B.; Horvath, A.; Weerts, J E E.; Albaneze-Walker, J Processes for the Preparation of a Diarylthiohydantoin Compound WO2016100645A1, 2016 (d) Bignan, G.; Connolly, P J.; Hickson, I.; Meerpoel, L.; Pande, V.; Zhang, Z.; Branch, J.; Rocaboy, C.; Trabalon Escolar, L B Preparation of Substituted Thiohydantoin Derivatives as Androgen Receptor Antagonists WO2017123542A1, 2017 (120) Blaser, H.-U.; Steiner, H.; Studer, M Selective Catalytic Hydrogenation of Functionalized Nitroarenes: An Update ChemCatChem 2009, 1, 210−221 (121) Rose, A A N Encorafenib and Binimetinib for the Treatment of BRAF V600E/K-Mutated Melanoma Drugs Today 2019, 55, 247− 264 AW Perspective https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc (122) Richman, J.; Martin-Liberal, J.; Diem, S.; Larkin, J BRAF and MEK Inhibition for the Treatment of Advanced BRAF Mutant Melanoma Expert Opin Pharmacother 2015, 16, 1285−1297 (123) Dummer, R.; Ascierto, P A.; Gogas, H J.; Arance, A.; Mandala, M.; Liszkay, G.; Garbe, C.; Schadendorf, D.; Krajsova, I.; Gutzmer, R.; Chiarion-Sileni, V.; Dutriaux, C.; de Groot, J W B.; Yamazaki, N.; Loquai, C.; Moutouh-de Parseval, L A.; Pickard, M D.; Sandor, V.; Robert, C.; Flaherty, K T Encorafenib Plus Binimetinib Versus Vemurafenib or Encorafenib in Patients with BRAF-Mutant Melanoma (COLUMBUS): A Multicentre, Open-Label, Randomised Phase Trial Lancet Oncol 2018, 19, 603−615 (124) Chen, J Synthesizing Method for Binimetinib CN105820124A, 2016 (125) Krell, C M.; Misun, M.; Niederer, D A.; Pachinger, W H.; Wolf, M.-C.; Zimmermann, D.; Liu, W.; Stengel, P J.; Nichols, P Preparation of and Formulation Comprising a Mek Inhibitor WO2014063024A1, 2014 (126) Shirley, M Dacomitinib: First Global Approval Drugs 2018, 78, 1947−1953 (127) Díaz-Serrano, A.; Gella, P.; Jiménez, E.; Zugazagoitia, J.; Rodríguez, L P.-A Targeting EGFR in Lung Cancer: Current Standards and Developments Drugs 2018, 78, 893−911 (128) Wu, Y.-L.; Cheng, Y.; Zhou, X.; Lee, K H.; Nakagawa, K.; Niho, S.; Tsuji, F.; Linke, R.; Rosell, R.; Corral, J.; Migliorino, M R.; Pluzanski, A.; Sbar, E I.; Wang, T.; White, J L.; Nadanaciva, S.; Sandin, R.; Mok, T S Dacomitinib Versus Gefitinib as First-Line Treatment for Patients with EGFR-Mutation-Positive Non-Small-Cell Lung Cancer (ARCHER 1050): A Randomised, Open-Label, Phase Trial Lancet Oncol 2017, 18, 1454−1466 (129) (a) Smaill, J B.; Gonzales, A J.; Spicer, J A.; Lee, H.; Reed, J E.; Sexton, K.; Althaus, I W.; Zhu, T.; Black, S L.; Blaser, A.; Denny, W A.; Ellis, P A.; Fakhoury, S.; Harvey, P J.; Hook, K.; McCarthy, F O J.; Palmer, B D.; Rivault, F.; Schlosser, K.; Ellis, T.; Thompson, A M.; Trachet, E.; Winters, R T.; Tecle, H.; Bridges, A Tyrosine Kinase Inhibitors 20 Optimization of Substituted Quinazoline and Pyrido[3,4-D]Pyrimidine Derivatives as Orally Active, Irreversible Inhibitors of the Epidermal Growth Factor Receptor Family J Med Chem 2016, 59, 8103−8124 (b) Reed, J E.; Smaill, J B The Discovery of Dacomitinib, a Potent Irreversible EGFR Inhibitor In Comprehensive Accounts of Pharmaceutical Research and Development: From Discovery to Late-Stage Process Development Volume 1; Abdel-Magid, A F., Pesti, J A., Vaidyanathan, R., Eds.; ACS Publications: Washington, DC, 2016; pp 207−233 (130) Yu, S.; Dirat, O Early and Late Stage Process Development for the Manufacture of Dacomitinib In Comprehensive Accounts of Pharmaceutical Research and Development: From Discovery to Late-Stage Process Development Volume 1; Abdel-Magid, A F., Pesti, J A., Vaidyanathan, R., Eds.; ACS Publications: Washington, DC, 2016; pp 235−252 (131) Lin, K.; Mo, G Synthesis Method of EGFR (Epidermal Growth Factor Receptor) Inhibitor Dacomitinib CN103304492A, 2013 (132) (a) Krajczyk, A.; Boryski, J Dimroth Rearrangement-Old but Not Outdated Curr Org Chem 2017, 21, 2515−2529 (b) El Ashry, E S H.; Nadeem, S.; Shah, M R.; El Kilany, Y Recent Advances in the Dimroth Rearrangement: A Valuable Tool for the Synthesis of Heterocycles Adv Heterocycl Chem 2010, 101, 161−228 (c) El Ashry, E S H.; El Kilany, Y.; Rashed, N.; Assafir, H Dimroth Rearrangement: Translocation of Heteroatoms in Heterocyclic Rings and Its Role in Ring Transformations of Heterocycles Adv Heterocycl Chem 1999, 75, 79−165 (133) Wang, Q.; Wang, Q.; Su, R.; Li, H.; Huo, X.; Wang, L.; Wang, J Dacomitinib Solvate, Novel Crystalline Form of Dacomitinib and Preparation Method and Use of Dacomitinib in Novel Crystalline Form CN107793368A, 2018 (134) (a) Rodrigues, D A.; Sagrillo, F S.; Fraga, C A M Duvelisib: A 2018 Novel FDA-Approved Small Molecule Inhibiting Phosphoinositide 3-Kinases Pharmaceuticals 2019, 12, 69 (b) Vangapandu, H V.; Jain, N.; Gandhi, V Duvelisib: A Phosphoinositide-3 Kinase Delta/Gamma Inhibitor for Chronic Lymphocytic Leukemia Expert Opin Invest Drugs 2017, 26, 625−632 (135) Kaneda, M M.; Messer, K S.; Ralainirina, N.; Li, H.; Leem, C J.; Gorjestani, S.; Woo, G.; Nguyen, A V.; Figueiredo, C C.; Foubert, P.; Schmid, M C.; Pink, M.; Winkler, D G.; Rausch, M.; Palombella, V J.; Kutok, J.; McGovern, K.; Frazer, K A.; Wu, X.; Karin, M.; Sasik, R.; Cohen, E E.; Varner, J A PI3Kgamma Is a Molecular Switch That Controls Immune Suppression Nature 2016, 539, 437−442 (136) (a) Hennessy, B T.; Smith, D L.; Ram, P T.; Lu, Y.; Mills, G B Exploiting the PI3K/AKT Pathway for Cancer Drug Discovery Nat Rev Drug Discovery 2005, 4, 988−1004 (b) Liu, P.; Cheng, H.; Roberts, T M.; Zhao, J J Targeting the Phosphoinositide 3-Kinase Pathway in Cancer Nat Rev Drug Discovery 2009, 8, 627−644 (137) Anastasia, A.; Rossi, G Novel Drugs in Follicular Lymphoma Mediterr J Hematol Infect Dis 2015, 8, e2016061 (138) Ren, P.; Martin, M.; Isbester, P.; Lane, B.; Kropp, J Process for Preparing Isoquinolines and Solid Forms of Isoquinolines WO2012097000A1, 2011 (139) Shirley, M Encorafenib and Binimetinib: First Global Approvals Drugs 2018, 78, 1277−1284 (140) Cancer Genome Atlas Network Genomic Classification of Cutaneous Melanoma Cell 2015, 161, 1681−1696 (141) Mackiewicz, J.; Mackiewicz, A BRAF and MEK Inhibitors in the Era of Immunotherapy in Melanoma Patients Wspolczesna Onkol 2018, 22, 68−72 (142) Huang, S.; Jin, X.; Liu, Z.; Poon, D.; Tellew, J.; Wan, Y.; Wang, X.; Xie, Y Compounds and Compositions as Protein Kinase Inhibitors WO2011025927A1, 2011 (143) Sun, Q.; Zhou, J.; Zhang, Z.; Guo, M.; Liang, J.; Zhou, F.; Long, J.; Zhang, W.; Yin, F.; Cai, H.; Yang, H.; Zhang, W.; Gu, Y.; Ni, L.; Sai, Y.; Cui, Y.; Zhang, M.; Hong, M.; Sun, J.; Yang, Z.; Qing, W.; Su, W.; Ren, Y Discovery of Fruquintinib, a Potent and Highly Selective Small Molecule Inhibitor of Vegfr 1, 2, Tyrosine Kinases for Cancer Therapy Cancer Biol Ther 2014, 15, 1635−1645 (144) Shirley, M Fruquintinib: First Global Approval Drugs 2018, 78, 1757−1761 (145) Wu, Z.; Li, W.; Chu, Y Crystalline Forms of 6-((6,7Dimethoxyquinazolin-4-yl)oxy)-N,2-dimethylbenzofuran-3-carboxamide WO2016037550A1, 2016 (146) (a) Wu, Z.; Li, W.; Chu, Y 6-(6,7-Dimethoxyquinazoline-4yloxy)-N, 2-Dimethyl Benzofuran-3-Methane Amide Crystal Form CN105461702A, 2016 (b) Yu, Y.; Chen, J.; Yi, D Preparation of Quinazoline Derivative Crystal Form B CN105777723A, 2016 (c) Yu, Y.; Chen, J.; Yi, D Preparation and Application of Fruquintinib Crystal Form C CN105777722A, 2016 (d) Yu, Y.; Chen, J.; Yi, D Preparation of Crystal Form a of 6-[(6,7-Dimethoxy4-quinazolinyl)oxy]-N,2-Dimethyl-3-benzofurancarboxamide CN105777721A, 2016 (147) (a) Su, W.-G.; Zhang, W.; Yan, X.; Cui, Y.; Ren, Y.; Duan, J Preparation of Quinazoline Derivatives as KDR Inhibitors WO2009137797A2, 2009 (b) Su, W.; Zhang, W.; Yan, X.; Cui, Y.; Ren, Y.; Duan, J Preparation of Quinazoline Derivatives as Angiogenesis Inhibitors CN101575333A, 2009 (148) Dhillon, S Gilteritinib: First Global Approval Drugs 2019, 79, 331−339 (149) (a) Mori, M.; Kaneko, N.; Ueno, Y.; Yamada, M.; Tanaka, R.; Saito, R.; Shimada, I.; Mori, K.; Kuromitsu, S Gilteritinib, a FLT3/ Axl Inhibitor, Shows Antileukemic Activity in Mouse Models of FLT3 Mutated Acute Myeloid Leukemia Invest New Drugs 2017, 35, 556− 565 (b) Wang, E S Gilteritinib for the Treatment of Patients with FLT3 Mutated Relapsed or Refractory Acute Myeloid Leukemia Expert Rev Precis Med Drug Dev 2019, 4, 105−112 (150) Shimada, I.; Kurosawa, K.; Matsuya, T.; Iikubo, K.; Kondoh, Y.; Kamikawa, A.; Tomiyama, H.; Iwai, Y Diamino Heterocyclic Carboxamide Compound US8969336, 2012 (151) Yue, Q.; Zhou, Z.; Gao, Q.; Zheng, B Synthesis Method of 3,5-Disubstituted-Pyrazine-2-formamide Compound CN106083821A, 2016 AX Perspective https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc (152) Huang, W.-S.; Liu, S.; Zou, D.; Thomas, M.; Wang, Y.; Zhou, T.; Romero, J.; Kohlmann, A.; Li, F.; Qi, J.; Cai, L.; Dwight, T A.; Xu, Y.; Xu, R.; Dodd, R.; Toms, A.; Parillon, L.; Lu, X.; Anjum, R.; Zhang, S.; Wang, F.; Keats, J.; Wardwell, S D.; Ning, Y.; Xu, Q.; Moran, L E.; Mohemmad, Q K.; Jang, H G.; Clackson, T.; Narasimhan, N I.; Rivera, V M.; Zhu, X.; Dalgarno, D.; Shakespeare, W C Discovery of Brigatinib (AP26113), a Phosphine Oxide-Containing, Potent, Orally Active Inhibitor of Anaplastic Lymphoma Kinase J Med Chem 2016, 59, 4948−4964 (153) (a) Hoy, S M Glasdegib: First Global Approval Drugs 2019, 79, 207−213 (b) https://www.fda.gov/Drugs/Fda-ApprovesGlasdegib-Aml-Adults-Age-75-or-Older-or-Who-Have-Comorbidities (accessed Dec 3, 2019) (154) Cortes, J E.; Heidel, F H.; Hellmann, A.; Fiedler, W.; Smith, B D.; Robak, T.; Montesinos, P.; Pollyea, D A.; DesJardins, P.; Ottmann, O.; Ma, W W.; Shaik, M N.; Laird, A D.; Zeremski, M.; O’Connell, A.; Chan, G.; Heuser, M Randomized Comparison of Low Dose Cytarabine with or without Glasdegib in Patients with Newly Diagnosed Acute Myeloid Leukemia or High-Risk Myelodysplastic Syndrome Leukemia 2019, 33, 379−389 (155) Munchhof, M J.; Li, Q.; Shavnya, A.; Borzillo, G V.; Boyden, T L.; Jones, C S.; LaGreca, S D.; Martinez-Alsina, L.; Patel, N.; Pelletier, K.; Reiter, L A.; Robbins, M D.; Tkalcevic, G T Discovery of PF-04449913, a Potent and Orally Bioavailable Inhibitor of Smoothened ACS Med Chem Lett 2012, 3, 106−111 (156) (a) Chaudhry, P.; Singh, M.; Triche, T J.; Guzman, M.; Merchant, A A GLI3 Repressor Determines Hedgehog Pathway Activation and Is Required for Response to SMO Antagonist Glasdegib in AML Blood 2017, 129, 3465−3475 (b) Long, B.; Zhu, H.; Zhu, C.; Liu, T.; Meng, W Activation of the Hedgehog Pathway in Chronic Myelogeneous Leukemia Patients J Exp Clin Cancer Res 2011, 30, (157) Heretsch, P.; Tzagkaroulaki, L.; Giannis, A Modulators of the Hedgehog Signaling Pathway Bioorg Med Chem 2010, 18, 6613− 6624 (158) (a) Jager, T.; Ocker, M.; Kiesslich, T.; Neureiter, E.; Neureiter, D Thoughts on Investigational Hedgehog Pathway Inhibitors for the Treatment of Cancer Expert Opin Invest Drugs 2017, 26, 133−136 (b) Peukert, S.; Miller-Moslin, K Small-Molecule Inhibitors of the Hedgehog Signaling Pathway as Cancer Therapeutics ChemMedChem 2010, 5, 500−512 (159) Peng, Z.; Wong, J W.; Hansen, E C.; Puchlopek-Dermenci, A L A.; Clarke, H J Development of a Concise, Asymmetric Synthesis of a Smoothened Receptor (SMO) Inhibitor: Enzymatic Transamination of a 4-Piperidinone with Dynamic Kinetic Resolution Org Lett 2014, 16, 860−863 (160) Hansen, E C.; Seadeek, C S Process for Preparing Crystalline Forms of 1-((2r,4r)-2-(1h-Benzo[D]imidazol-2-yl)-1-methylpiperidin-4-yl)-3-(4-cyanophenyl)urea Maleate WO2016170451A1, 2016 (161) Dhillon, S Ivosidenib: First Global Approval Drugs 2018, 78, 1509−1516 (162) (a) Boddu, P.; Borthakur, G Therapeutic Targeting of Isocitrate Dehydrogenase Mutant AML Expert Opin Invest Drugs 2017, 26, 525−530 (b) Nassereddine, S.; Lap, C J.; Haroun, F.; Tabbara, I The Role of Mutant IDH1 and IDH2 Inhibitors in the Treatment of Acute Myeloid Leukemia Ann Hematol 2017, 96, 1983−1991 (163) (a) Hansen, E.; Quivoron, C.; Straley, K.; Lemieux, R M.; Popovici-Muller, J.; Sadrzadeh, H.; Fathi, A T.; Gliser, C.; David, M.; Saada, V.; Micol, J.; Bernard, O.; Dorsch, M.; Yang, H.; Su, M.; Agresta, S.; de Botton, S.; Penard-Lacronique, V.; Yen, K AG-120, an Oral, Selective, First-in-Class, Potent Inhibitor of Mutant IDH1, Reduces Intracellular 2HG and Induces Cellular Differentiation in TF-1 R132H Cells and Primary Human IDH1 Mutant AML Patient Samples Treated Ex Vivo Blood 2014, 124, 3734 (b) DiNardo, C D.; Stein, E M.; de Botton, S.; Roboz, G J.; Altman, J K.; Mims, A S.; Swords, R.; Collins, R H.; Mannis, G N.; Pollyea, D A.; Donnellan, W.; Fathi, A T.; Pigneux, A.; Erba, H P.; Prince, G T.; Stein, A S.; Uy, G L.; Foran, J M.; Traer, E.; Stuart, R K.; Arellano, M L.; Slack, J L.; Sekeres, M A.; Willekens, C.; Choe, S.; Wang, H.; Zhang, V.; Yen, K E.; Kapsalis, S M.; Yang, H.; Dai, D.; Fan, B.; Goldwasser, M.; Liu, H.; Agresta, S.; Wu, B.; Attar, E C.; Tallman, M S.; Stone, R M.; Kantarjian, H M.; et al Durable Remissions with Ivosidenib in IDH1Mutated Relapsed or Refractory AML N Engl J Med 2018, 378, 2386−2398 (164) (a) Lemieux, R M.; Popovici-Muller, J.; Travins, J.; Cai, Z.; Cui, D.; Ding, Z Therapeutically Active Compounds and Their Methods of Use WO2013107291A1, 2013 (b) Lemieux, R M.; Popovici-Muller, J.; Travins, J.; Cai, Z.; Cui, D.; Ding, Z Therapeutically Active Compounds and Their Methods of Use WO2015010297A1, 2015 (c) Popovici-Muller, J.; Lemieux, R M.; Artin, E.; Saunders, J O.; Salituro, F G.; Travins, J.; Cianchetta, G.; Cai, Z.; Zhou, D.; Cui, D.; Chen, P.; Straley, K.; Tobin, E.; Wang, F.; David, M D.; Penard-Lacronique, V.; Quivoron, C.; Saada, V.; de Botton, S.; Gross, S.; Dang, L.; Yang, H.; Utley, L.; Chen, Y.; Kim, H.; Jin, S.; Gu, Z.; Yao, G.; Luo, Z.; Lv, X.; Fang, C.; Yan, L.; Olaharski, A.; Silverman, L.; Biller, S.; Su, S.-S M.; Yen, K Discovery of AG-120 (Ivosidenib): A First-in-Class Mutant IDH1 Inhibitor for the Treatment of IDH1 Mutant Cancers ACS Med Chem Lett 2018, 9, 300−305 (165) Muthusamy, A R.; Singh, A.; Yazali, V S.; Luthra, P K.; Vasoya, S L.; Patil, B T.; Taneja, A K.; Srivastav, N C.; Singh, R.; Rengasamy, V.; Tyagi, A Solid State Forms of Ivosidenib WO2019104318A1, 2019 (166) Scott, L J Larotrectinib: First Global Approval Drugs 2019, 79, 201−206 (167) Cocco, E.; Scaltriti, M.; Drilon, A NTRK Fusion-Positive Cancers and TRK Inhibitor Therapy Nat Rev Clin Oncol 2018, 15, 731−747 (168) Wilson, F H.; Herbst, R S Larotrectinib in NTRKRearranged Solid Tumors Biochemistry 2019, 58, 1555−1557 (169) Huang, F W.; Feng, F Y A Tumor-Agnostic NTRK (TRK) Inhibitor Cell 2019, 177, (170) Reynolds, M.; Eary, C T.; Spencer, S.; Juengst, D.; Hache, B.; Jiang, Y.; Haas, J.; Andrews, S W Preparation of (S)-N-(5-((R)-2(2,5-Difluorophenyl)pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidin-3-yl)3-hydroxypyrrolidine-1-carboxamide WO2017201241A1, 2017 (171) Ryser, C O.; Diebold, J.; Gautschi, O Treatment of Anaplastic Lymphoma Kinase-Positive Non-Small Cell Lung Cancer: Update and Perspectives Curr Opin Oncol 2019, 31, 8−12 (172) Syed, Y Y Lorlatinib: First Global Approval Drugs 2019, 79, 93−98 (173) Johnson, T W.; Richardson, P F.; Bailey, S.; Brooun, A.; Burke, B J.; Collins, M R.; Cui, J J.; Deal, J G.; Deng, Y.-L.; Dinh, D.; Engstrom, L D.; He, M.; Hoffman, J.; Hoffman, R L.; Huang, Q.; Kania, R S.; Kath, J C.; Lam, H.; Lam, J L.; Le, P T.; Lingardo, L.; Liu, W.; McTigue, M.; Palmer, C L.; Sach, N W.; Smeal, T.; Smith, G L.; Stewart, A E.; Timofeevski, S.; Zhu, H.; Zhu, J.; Zou, H Y.; Edwards, M P Discovery of (10r)-7-Amino-12-Fluoro-2,10,16Trimethyl-15-Oxo-10,15,16,17-Tetrahydro-2h-8,4-(Metheno)Pyrazolo[4,3-H][2,5,11]-Benzoxadiazacyclotetradecine-3-Carbonitrile (PF-06463922), a Macrocyclic Inhibitor of Anaplastic Lymphoma Kinase (ALK) and C-Ros Oncogene (Ros1) with Preclinical Brain Exposure and Broad-Spectrum Potency against ALK-Resistant Mutations J Med Chem 2014, 57, 4720−4744 (174) Shaw, A T.; Solomon, B J.; Besse, B.; Bauer, T M.; Lin, C.C.; Soo, R A.; Riely, G J.; Ou, S.-H I.; Clancy, J S.; Li, S.; Abbattista, A.; Thurm, H.; Satouchi, M.; Camidge, D R.; Kao, S.; Chiari, R.; Gadgeel, S M.; Felip, E.; Martini, J.-F ALK Resistance Mutations and Efficacy of Lorlatinib in Advanced Anaplastic Lymphoma KinasePositive Non-Small-Cell Lung Cancer J Clin Oncol 2019, 37, 1370− 1379 (175) Solomon, B J.; Besse, B.; Bauer, T M.; Felip, E.; Soo, R A.; Camidge, D R.; Chiari, R.; Bearz, A.; Lin, C.-C.; Gadgeel, S M.; Riely, G J.; Tan, E H.; Seto, T.; James, L P.; Clancy, J S.; Abbattista, A.; Martini, J.-F.; Chen, J.; Peltz, G.; Thurm, H.; Ou, S.-H I.; Shaw, A T Lorlatinib in Patients with ALK-Positive Non-Small-Cell Lung AY Perspective https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc Cancer: Results from a Global Phase Study Lancet Oncol 2018, 19, 1654−1667 (176) Duan, S.; Li, B.; Dugger, R W.; Conway, B.; Kumar, R.; Martinez, C.; Makowski, T.; Pearson, R.; Olivier, M.; Colon-Cruz, R Developing an Asymmetric Transfer Hydrogenation Process for (S)5-Fluoro-3-Methylisobenzofuran-1(3h)-One, a Key Intermediate to Lorlatinib Org Process Res Dev 2017, 21, 1340−1348 (177) Li, B.; Barnhart, R W.; Hoffman, J E.; Nematalla, A.; Raggon, J.; Richardson, P.; Sach, N.; Weaver, J Exploratory Process Development of Lorlatinib Org Process Res Dev 2018, 22, 1289− 1293 (178) (a) Bharate, S B.; Sawant, S D.; Singh, P P.; Vishwakarma, R A Kinase Inhibitors of Marine Origin Chem Rev 2013, 113, 6761− 6815 (b) Rinehart, K L.; Lithgow-Bertelloni, A M Dehydrodidemnin B WO9104985A1, 1991 (179) Urdiales, J.; Morata, P.; De Castro, I N.; Sánchez-Jiménez, F Antiproliferative Effect of Dehydrodidemnin B (DDB), a Depsipeptide Isolated from Mediterranean Tunicates Cancer Lett 1996, 102, 31−37 (180) Losada, A.; Muñoz-Alonso, M J.; García, C.; Sánchez-Murcia, P A.; Martínez-Leal, J F.; Domínguez, J M.; Lillo, M P.; Gago, F.; Galmarini, C M Translation Elongation Factor eEF1A2 Is a Novel Anticancer Target for the Marine Natural Product Plitidepsin Sci Rep 2016, 6, 35100 (181) Leisch, M.; Egle, A.; Greil, R Plitidepsin: A Potential New Treatment for Relapsed/Refractory Multiple Myeloma Future Oncol 2019, 15, 109−120 (182) Rodriguez, I.; Polanco, C.; Cuevas, F.; Mandez, P.; Cuevas, C.; Gallego, P.; Munt, S.; Manzanares, I Synthetic Methods for Aplidine and New Antitumoral Derivatives, Methods of Making and Using Them WO2002002596A2, 2002 (183) Blair, H A Pyrotinib: First Global Approval Drugs 2018, 78, 1751−1755 (184) Li, X.; Yang, C.; Wan, H.; Zhang, G.; Feng, J.; Zhang, L.; Chen, X.; Zhong, D.; Lou, L.; Tao, W.; Zhang, L Discovery and Development of Pyrotinib: A Novel Irreversible EGFR/HER2 Dual Tyrosine Kinase Inhibitor with Favorable Safety Profiles for the Treatment of Breast Cancer Eur J Pharm Sci 2017, 110, 51−61 (185) (a) Chew, W.; Cheal, G K.; Lunetta, J F Methods of Synthesizing Substituted 3-Cyanoquinolines and Intermediates Thereof WO2006127207A1, 2006 (b) Chew, W.; Papamichelakis, M Methods of Preparing 3-Cyano-Quinolines and Intermediates Made Thereby WO2006127205A2, 2006 (186) Wu, G.; Zhang, Q.; Cao, Y Method for Preparing Tyrosine Kinase Inhibitor and Derivative Thereof WO2017186140A1, 2017 (187) Li, X.; Wang, B Pharmaceutically Acceptable Salt of (E)-N[4-[[3-Chloro-4-(2-pyridylmethoxy)phenyl]amino]-3-cyano-7ethoxy-6-quinolyl]-3-[(2r)-1-methylpyrrolidin-2-yl]prop-2-enamide, Preparation Method Therefor, and Medical Use Thereof WO2012122865A2, 2012 (188) Hoy, S M Talazoparib: First Global Approval Drugs 2018, 78, 1939−1946 (189) Sulai, N H.; Tan, A R Development of Poly(ADP-ribose) Polymerase Inhibitors in the Treatment of BRCA-Mutated Breast Cancer Clin Adv Hematol Oncol 2018, 16, 491−501 (190) (a) Murai, J.; Huang, S.-y N.; Das, B B.; Renaud, A.; Zhang, Y.; Doroshow, J H.; Ji, J.; Takeda, S.; Pommier, Y Trapping of PARP1 and PARP2 by Clinical PARP Inhibitors Cancer Res 2012, 72, 5588−5599 (b) Murai, J.; Huang, S.-Y N.; Renaud, A.; Zhang, Y.; Ji, J.; Takeda, S.; Morris, J.; Teicher, B A.; Doroshow, J H.; Pommier, Y Stereospecific PARP Trapping by BMN 673 and Comparison with Olaparib and Rucaparib Mol Cancer Ther 2014, 13, 433−443 (191) Wang, B.; Chu, D.; Feng, Y.; Shen, Y.; Aoyagi-Scharber, M.; Post, L E Discovery and Characterization of (8s, 9r)-5-Fluoro-8-(4Fluorophenyl)-9-(1-Methyl-1 H-1, 2, 4-Triazol-5-Yl)-2, 7, 8, 9Tetrahydro-3 H-Pyrido [4, 3, 2-De] Phthalazin-3-One (BMN 673, Talazoparib), a Novel, Highly Potent, and Orally Efficacious Poly (ADP-Ribose) Polymerase-1/2 Inhibitor, as an Anticancer Agent J Med Chem 2016, 59, 335−357 (192) (a) Wang, B.; Chu, D.; Liu, Y.; Jiang, Q.; Lu, L Processes of S y n th es i z i n g D ih y d r o p y r id o p ht la zi no ne De ri va ti v es WO2011097602A1, 2011 (b) Wang, B.; Chu, D.; Liu, Y.; Peng, S Crystalline (8s,9r)-5-Fluoro-8-(4-fluorophenyl)-9-(1-methyl-1h-1,2,4triazol-5-yl)-8,9-dihydro-2h-pyrido[4,3,2-de]phthalazin-3(7h)-one Tosylate Salt WO2012054698A1, 2012 (c) Feng, Y.; Gutierrez, A A.; Shen, Y.; Wang, E W.; Okhamafe, A O.; Price, C P.; Chou, T Dihydropyridophthalazinone Inhibitors of Poly (Adp-Ribose) Polymerase (Parp) for the Treatment of Multiple Myeloma WO2013028495A1, 2013 (d) Henderson, M Triazole Intermediates Useful in the Synthesis of Protected N-Alkyltriazolecarbaldehydes WO2015069851A1, 2015 (193) Xu, Y.; Yohi, P W.; Xu, M.; Cruise, D.; Huang, L Synthesis of Parpinhibitor Talazoparib WO2017215166A1, 2017 (194) https://www.prnewswire.com/news-releases/santenannounces-initiation-of-phase-3-clinical-development-programspectrum-in-the-united-states-evaluating-omidenepag-isopropyl-de117-for-the-treatment-of-glaucoma-or-ocular-hypertension300735092.html (accessed Dec 3, 2019) (195) Iwamura, R.; Tanaka, M.; Okanari, E.; Kirihara, T.; OdaniKawabata, N.; Shams, N.; Yoneda, K Identification of a Selective, Non-Prostanoid EP2 Receptor Agonist for the Treatment of Glaucoma: Omidenepag and Its Prodrug Omidenepag Isopropyl J Med Chem 2018, 61, 6869−6891 (196) (a) Li, M.; Ke, H Z.; Qi, H.; Healy, D R.; Li, Y.; Crawford, D T.; Paralkar, V M.; Owen, T A.; Cameron, K O.; Lefker, B A.; Brown, T A.; Thompson, D D A Novel, Non-Prostanoid EP2 Receptor-Selective Prostaglandin E2 Agonist Stimulates Local Bone Formation and Enhances Fracture Healing J Bone Miner Res 2003, 18, 2033−2042 (b) Paralkar, V M.; Borovecki, F.; Ke, H Z.; Cameron, K O.; Lefker, B.; Grasser, W A.; Owen, T A.; Li, M.; DaSilva-Jardine, P.; Zhou, M.; Dunn, R L.; Dumont, F.; Korsmeyer, R.; Krasney, P.; Brown, T A.; Plowchalk, D.; Vukicevic, S.; Thompson, D D An EP2 Receptor-Selective Prostaglandin E2 Agonist Induces Bone Healing Proc Natl Acad Sci U S A 2003, 100, 6736−6740 (197) (a) Iwamura, R.; Tanaka, M.; Katsube, T.; Shigetomi, M.; Okanari, E.; Tokunaga, Y.; Fujiwara, H Pyridylaminoacetic Acid Compound WO2009113600A1, 2009 (b) Yamamoto, Y.; Oue, M.; Wada, Y N-Substituted Sulfonamide Compound and Method for Producing Same WO2015190507A1, 2015 (198) https://www.drugbank.ca/drugs/DB08820 (accessed Jul 28, 2019) (199) https://www.drugs.com/newdrugs/fda-approves-symdekotezacaftor-ivacaftor-ivacaftor-cystic-fibrosis-ages-12-older-certainmutations-4693.html (accessed Dec 3, 2019) (200) (a) Donaldson, S H.; Pilewski, J M.; Griese, M.; Cooke, J.; Viswanathan, L.; Tullis, E.; Davies, J C.; Lekstrom-Himes, J A.; Wang, L T Tezacaftor/Ivacaftor in Subjects with Cystic Fibrosis and F508del/F508del-CFTR or F508del/G551D-CFTR Am J Respir Crit Care Med 2018, 197, 214−224 (b) Taylor-Cousar, J L.; Munck, A.; McKone, E F.; van der Ent, C K.; Moeller, A.; Simard, C.; Wang, L T.; Ingenito, E P.; McKee, C.; Lu, Y.; Lekstrom-Himes, J.; Elborn, J S Tezacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del N Engl J Med 2017, 377, 2013−2023 (201) Garg, V.; Shen, J.; Li, C.; Agarwal, S.; Gebre, A.; Robertson, S.; Huang, J.; Han, L.; Jiang, L.; Stephan, K.; Wang, L T.; LekstromHimes, J Pharmacokinetic and Drug-Drug Interaction Profiles of the Combination of Tezacaftor/Ivacaftor Clin Transl Sci 2019, 12, 267−275 (202) https://www.accessdata.fda.gov/drugsatfda_docs/label/ 2018/210491lbl.pdf (accessed Dec 3, 2019) (203) https://www.drugs.com/newdrugs/fda-approves-symdekotezacaftor-ivacaftor-ivacaftor-underlying-cause-cf-children-ages-6-11years-4996.html (accessed Dec 3, 2019) (204) (a) Ruah, S H S.; Grootenhuis, P D J.; Van Goor, F.; Miller, M T.; McCartney, J.; Zhou, J.; Bear, B.; Numa, M M D Preparation of Indolyl Cycloalkylcarboxamides as Modulators of ATP-Binding Cassette Transporters for Use as Drugs and Biological Tools AZ Perspective https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX Journal of Medicinal Chemistry pubs.acs.org/jmc WO2010053471A1, 2010 (b) Keshavarz-Shokri, A.; Zhang, B.; Alcacio, T E.; Lee, E C.; Zhang, Y.; Krawiec, M Preparation of Benzodioxolyl-N-Indolyl Cyclopropanecarboxamide Derivatives for Use as CFTR Modulators WO2011119984A1, 2011 (c) Alargova, R G.; Kadiyala, I N.; Zaman, N T Formulations of (R)-1-(2,2Difluorobenzo[d][1,3]dioxol-5-yl)-N-(1-(2,3-dihydroxypropyl)-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)-1h-indol-5-yl)cyclopropanecarboxamide for Treatment of Cystic Fibrosis WO2012170061A1, 2012 (d) Van Goor, F F Pharmaceutical Compositions for the Treatment of CFTR-Mediated Disorders WO2013185112A1, 2013 (e) Alargova, R G.; Dunbar, C A.; Kadiyala, I N Oral Compositions of (R)-1-(2,2-Difluorobenzo[d][1,3]dioxol-5-yl)-N-(1-(2,3-dihydroxypropyl)-6-fluoro-2-(1-hydroxy2-methylpropan-2-yl)-1h-indol-5-yl)cyclopropanecarboxamide for Treatment of Cystic Fibrosis WO2014014841A1, 2014 (f) Phenix, B D.; Bagnol, L J.-C.; Brodeur, G G.; Chandran, S.; Dokou, E.; Ferris, L A.; Knezic, D.; McCarty, K L.; Medek, A.; Waggener, S A Method for Preparation of Quinolinone Carboxamides, Indole Carboxamides and Pharmaceutical Compositions Containing Them for the Treatment of Cystic Fibrosis Transmembrane Conductance Regulator Mediated Diseases WO2015160787A1, 2015 (g) Alcacio, T.; Baek, M.; Grootenhuis, P.; Hadida Ruah, S S.; Hughes, R M.; Keshavarz-Shokri, A.; McAuley-Aoki, R.; McCartney, J.; Miller, M T.; Van Goor, F.; Zhang, B.; Andreson, C.; Cleveland, T.; Frieman, B A.; Khatuya, H.; Joshi, P V.; Krenitsky, P J.; Melillo, V.; Pierre, F J D.; Termin, A P.; Uy, J.; Zhou, J.; Abela, A R.; Busch, B B.; Paraselli, P.; Siesel, D A Preparation of Benzenesulfonylpyrazolylpyrrolidinylpyridinecarboxamide Derivatives and Analogs for Use as Cystic Fibrosis Transmembrane Conductance Regulator Modulators WO2018064632A1, 2018 (205) Blakely, K K A New Year-Long Combination Hormonal Contraceptive Nurs Womens Health 2019, 23, 172−176 (206) Gemzell-Danielsson, K.; Sitruk-Ware, R.; Creinin, M D.; Thomas, M.; Barnhart, K T.; Creasy, G.; Sussman, H.; Alami, M.; Burke, A E.; Weisberg, E.; Fraser, I.; Miranda, M J.; Gilliam, M.; Liu, J.; Carr, B R.; Plagianos, M.; Roberts, K.; Blithe, D Segesterone Acetate/Ethinyl Estradiol 12-Month Contraceptive Vaginal System Safety Evaluation Contraception 2019, 99, 323−328 (207) Urquhart, L Regulatory Watch: FDA New Drug Approvals in Q3 2018 Nat Rev Drug Discovery 2018, 17, 779 (208) Li, F Preparation of Ester Derivatives of Steroids WO199713779, 1997 (209) Mehrhof, W.; Irmscher, K.; Erb, R.; Pohl, L Synthesewege Zum 17a-Hydroxy-16-Methylen-19-nor-Progesteron Und Seinen Derivaten Chem Ber 1969, 102, 643−658 (210) Schwars, V.; Zachova, J.; Syhora, K Steroid derivatives L A Synthesis of 16-Methylene-17-a-Acetoxy-nor-Progesterone Tetrahedron Lett 1967, 22, 1925−1929 (211) Fuentes, G.; Gerardo, L.; Rodriguez, S.-D.; Miguel, C Process for Alkynylating 16-Substituted 17-Keto Steroids WO2013092668A1, 2013 (212) Keam, S J Vibegron: First Global Approval Drugs 2018, 78, 1835−1839 (213) Edmondson, S D.; Zhu, C.; Kar, N F.; Di Salvo, J.; Nagabukuro, H.; Sacre-Salem, B.; Dingley, K.; Berger, R.; Goble, S D.; Morriello, G.; Harper, B.; Moyes, C R.; Shen, D.-M.; Wang, L.; Ball, R.; Fitzmaurice, A.; Frenkl, T.; Gichuru, L N.; Ha, S.; Hurley, A L.; Jochnowitz, N.; Levorse, D.; Mistry, S.; Miller, R R.; Ormes, J.; Salituro, G M.; Sanfiz, A.; Stevenson, A S.; Villa, K.; Zamlynny, B.; Green, S.; Struthers, M.; Weber, A E.; et al Discovery of Vibegron: A Potent and Selective B3 Adrenergic Receptor Agonist for the Treatment of Overactive Bladder J Med Chem 2016, 59, 609−623 (214) (a) Yoshida, M.; Takeda, M.; Gotoh, M.; Nagai, S.; Kurose, T Vibegron, a Novel Potent and Selective B3-Adrenoreceptor Agonist, for the Treatment of Patients with Overactive Bladder: A Randomized, Double-Blind, Placebo-Controlled Phase Study Eur Urol 2018, 73, 783−790 (b) Wein, A J Re: Pharmacological Characterization of a Novel Beta Adrenergic Agonist, Vibegron: Evaluation of Antimuscarinic Receptor Selectivity for Combination Therapy for Overactive Bladder J Urol 2018, 200, 33−38 (c) Di Salvo, J.; Nagabukuro, H.; Wickham, L A.; Abbadie, C.; DeMartino, J A.; Fitzmaurice, A.; Gichuru, L.; Kulick, A.; Donnelly, M J.; Jochnowitz, N.; Hurley, A L.; Pereira, A.; Sanfiz, A.; Veronin, G.; Villa, K.; Woods, J.; Zamlynny, B.; Zycband, E.; Salituro, G M.; Frenkl, T.; Weber, A E.; Edmondson, S D.; Struthers, M Pharmacological Characterization of a Novel Beta Adrenergic Agonist, Vibegron: Evaluation of Antimuscarinic Receptor Selectivity for Combination Therapy for Overactive Bladder J Pharmacol Exp Ther 2017, 360, 346−355 (215) (a) Xu, F.; Kosjek, B.; Cabirol, F L.; Chen, H.; Desmond, R.; Park, J.; Gohel, A P.; Collier, S J.; Smith, D J.; Liu, Z.; Janey, J M.; Chung, J Y L.; Alvizo, O Synthesis of Vibegron Enabled by a Ketoreductase Rationally Designed for High Ph Dynamic Kinetic Reduction Angew Chem., Int Ed 2018, 57, 6863−6867 (b) Xu, F.; Liu, Z.; Desmond, R.; Park, J.; Kalinin, A.; Kosjek, B.; Strotman, H.; Li, H.; Moncecchi, J Process for Preparing Beta Agonists and Intermediates WO2014150639A1, 2014 (c) Yasuda, N.; Liu, Z.; Zhong, Y.-L.; Chung, J Y L.; Dunn, R F.; Maloney, K M.; Campos, K.; Hoerrner, R S.; Lynch, J.; Xu, F.; Cleator, E.; Gibson, A.; Keen, S.; Lieberman, D.; Yoshikawa, N Process for Making Beta Agonists and Intermediates WO2013062881A1, 2013 (d) Berger, R.; Chang, L.; Edmondson, S D.; Goble, S D.; Ha, S N.; Kar, N F.; Kopka, I E.; Li, B.; Morriello, G J.; Moyes, C R.; Shen, D.-M.; Wang, L.; Zhu, C Hydroxymethyl Pyrrolidine as B3 Adrenoceptor Agonist JP2012020961A, 2012 (e) Nagabukuro, H.; Edmondson, S D.; Sinharoy, M S.; Denney, W S.; Frenkl, T L Combination Therapy Using a Beta Adrenergic Receptor Agonist and an Antimuscarinic Agent WO2011043942A1, 2011 (f) Berger, R.; Chang, L.; Edmondson, S D.; Goble, S D.; Ha, S N.; Kar, N F.; Kopka, I E.; Li, B.; Morriello, G J.; Moyes, C R.; Shen, D.-M.; Wang, L.; Zhu, C Hydroxymethyl Pyrrolidines as Beta Adrenergic Receptor Agonists WO2009124167A1, 2009 (216) Dyker, G Amino Acid Derivatives by Multicomponent Reactions Angew Chem., Int Ed Engl 1997, 36, 1700−1702 BA Perspective https://dx.doi.org/10.1021/acs.jmedchem.0c00345 J Med Chem XXXX, XXX, XXX−XXX ... K K.-C.; Sakya, S Synthetic Approaches to the 2004 New Drugs Mini-Rev Med Chem 2005, 5, 1133−1144 (d) Sakya, S M.; Li, J.; Liu, K K.-C Synthetic Approaches to the 2005 New Drugs Mini-Rev Med... M.; Li, J Synthetic Approaches to the 2006 New Drugs Mini-Rev Med Chem 2007, 7, 1255−1269 (f) Liu, K K.-C.; Sakya, S M.; O’Donnell, C J.; Li, J Synthetic Approaches to the 2007 New Drugs Mini-Rev... Li, J Synthetic Approaches to the 2008 New Drugs Mini-Rev Med Chem 2009, 9, 1655−1675 (h) Liu, K K.-C.; Sakya, S M.; O’Donnell, C J.; Flick, A C.; Li, J Synthetic Approaches to the 2009 New Drugs

Ngày đăng: 29/08/2021, 10:59

Xem thêm:

TỪ KHÓA LIÊN QUAN