Accepted Manuscript Review Synthetic approaches to the 2013 new drugs Hong X. Ding, Carolyn A. Leverett, Robert E. Kyne Jr., Kevin K.-C. Liu, Sarah J. Fink, Andrew C. Flick, Christopher J. O’Donnell PII: DOI: Reference:

S0968-0896(15)00160-1 http://dx.doi.org/10.1016/j.bmc.2015.02.056 BMC 12119

To appear in:

Bioorganic & Medicinal Chemistry

Received Date: Revised Date: Accepted Date:

5 January 2015 20 February 2015 26 February 2015

Please cite this article as: 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, Bioorganic & Medicinal Chemistry (2015), doi: http://dx.doi.org/ 10.1016/j.bmc.2015.02.056

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Synthetic Approaches to the 2013 New Drugs Hong X. Ding,1 Carolyn A. Leverett,2 Robert E. Kyne, Jr.,3 Kevin K.-C. Liu, 4 Sarah J. Fink,5 Andrew C. Flick,6 Christopher J. O’Donnell7* 1

4

Pharmacodia (Beijing) Co., LTD; Beijing, 100085, China

Lilly China Research And Development Center. Shanghai, 201203, China 5

2,3,6,7

BioDuro Co., Ltd., Shanghai 200131, China.

Pfizer Worldwide Research and Development, Groton Laboratories, 445 Eastern Point Road, Groton, CT 06340, United States

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 and also serve as leads for designing future new drugs. This annual review covers the synthesis of twenty-

1

Email: [email protected]; tel: 8610-8282-6195

2

Email: [email protected]; tel: 860-441-3936

3

Email: [email protected]; tel: 860-441-1510

4

Email: [email protected]; tel: 8621-2080-5590

5

Email: [email protected]; tel: 86-21-3175-2858

6

Email: [email protected]; tel: 860-715-0228

7

Email: [email protected]; tel: 860-715-4118

1

four NCEs that were approved for the first time in 2013 and two 2012 drugs which were not covered during the previous edition of this review. Key Words: Synthesis, New Drug Molecules, New Chemical Entities, Medicine, Therapeutic Agents. 1. Introduction “The most fruitful basis for the discovery of a new drug is to start with an old drug.” ─ Sir James Whyte Black, winner of the 1988 Nobel Prize in medicine.1 This annual review was inaugurated twelve years ago2-12 and presents synthetic methods for molecular entities that were approved for the first time in various countries during the past year. Given that drugs tend to have structural homology across similar biological targets, it is widely believed that the knowledge of new chemical entities and their syntheses will greatly enhance the ability to design new drugs more efficiently. The pharmaceutical industry enjoyed a banner year in 2013, with a total of 56 new products including new chemical entities, biological drugs, and diagnostic agents having reached the worldwide market for the first time. Although an additional 19 new products were approved for the first time in 2013, some were not launched before the end of the year,13 and therefore this review focuses on the syntheses of twenty-four NCEs that were approved and launched for the first time in 2013. It also includes two additional drugs that although were initially approved in 2012, were not included in our prior review (Fig. 1).12 New indications for previously launched medications, new combinations, new formulations of existing drugs, and drugs synthesized purely via bio-processes or peptide synthesizers have been excluded from this review. Although the scale of the synthetic routes were not explicitly disclosed in most cases, this review covers, perceptibly, the most scalable routes that have been disclosed within published or patent literature beginning from commercially available starting materials. Drugs presented in this review are ordered alphabetically by generic name. Figure 1. Structures of 26 NCEs covered in this review

2

3

4

5

2. Acotiamide hydrochloride hydrate (Acofide®) Acotiamide hydrochloride trihydrate is the first drug to be approved in Japan for the treatment of functional dyspepsia (FD). The drug was discovered by Zeria Pharmaceuticals and jointly developed with Astellas Pharmaceuticals.14

The drug blocks muscarinic receptors and inhibits peripheral

acetylcholine esterases, thereby increasing the concentration of acetylcholine,14 ultimately improving the impaired gastric motility and delayed gastric emptying along with the additional symptoms associated with FD, such as post prandial fullness, upper abdominal bloating and early satiation.14-16 Although multiple synthetic approaches to the drug have been reported,17, 18 the synthesis highlighted in Scheme 1 and described below represents the largest scale reported to date in a patent application.18 Commercial 3,4,5-trimethoxybenzoic acid (1) was first converted to the corresponding acid chloride 2, which was isolated by co-distillation with hexane. In refluxing dichloroethane (DCE), the acid chloride was coupled with the commercially available thiazole amine (3) to give the desired amidothiazole 4 in 89% yield. From this intermediate, amide linkage, selective demethylation of the 2methoxy group, salt formation, and recrystallization were accomplished in the following sequence: the thiazole ester 4 was reacted with N,N-diisopropyl ethylenediamine (5) in DMA at elevated temperatures. Upon cooling, the mixture was dissolved in n-butanol and washed with aqueous sodium hydroxide. Subsequent treatment with HCl gas in isopropanol gave the corresponding HCl salt as crystals that could be collected by filtration. The product obtained was further crystallized from 4:1 isopropanol and water to give the desired product acotimide (I) as the hydrochloride trihydrate in 71% yield. Scheme 1. Synthesis of acotiamide hydrochloride hydrate (I)

6

3. Afatinib dimaleate (Giotrif®, Gilotrif®) Afatinib dimaleate was approved by the U. S. Food and Drug Administration (FDA) in 2013 for the treatment of non-small cell lung cancer (NSCLC).19 Specifically, it was approved for patients presenting with metastatic NSCLC tumors which contain epidermal growth factor receptor (EGFR) exon 19 deletions or exon 21 mutations.19 Afatinib dimaleate is a covalent inhibitor of ErbB tyrosine kinases (tyk), which downregulates ErbB signaling by irreversible binding of EGFR tyk binding sites.19 While no manufacturing route has been disclosed to date,20-24 the most scalable published route likely derives from two Boehringer Ingelheim patents (Scheme 2).25, 26 Nitroquinazolinone (6), which is commercially available, was first chlorinated with phosphorous oxychloride (POCl3) followed by treatment with commercial 3-chloro-4-fluoroaniline (7) to afford SNAr adduct 8 in 90% yield over two steps. Sulfonylation to afford 9 (86%) and subsequent displacement with (S)-tetrahydrofuran-3-ol gave 10 in 90% yield.25

Raney-Nickel reduction of the nitro group

delivered 11 in 97% yield, which set the stage for the final side-chain functionalization.

2-

(Diethoxyphosphoryl)acetic acid and 1,1′-carbonyldiimidazole (CDI) were pre-mixed and added to aniline 11 to afford 12 in 70% isolated yield. Next, a Horner-Wadsworth-Emmons homologation gave

7

the (E)-olefin 13 in quantitative yield, followed by maleate salt formation (92%) to deliver the final API. The final five steps of this synthesis have been successfully demonstrated on multi-kilogram scale.24, 25

Scheme 2. Synthesis of afatinib dimaleate (II) F O NO2

HN N

Cl

1. POCl 3, Et 3N CH3CN, 80 °C 2. F

Cl

NH

N

NH 2

Cl

PhSO 2Na

NO 2

N

7

Cl

6

F NH

DMF, rt to 90 °C 86%

Cl

NO 2

N N

SO 2Ph

8

9

90% for 2 steps

F

F HO

Cl

Cl

NH

O

NO2

N N

t-BuOH, DMF, THF KOt-Bu, 20 °C to 45 °C 90%

NH NH2

N

Raney-Ni, DMF

N

NH4Cl, 40 °C 97%

O

HO

O

O

OEt P OEt O

CDI, THF, 40 °C to 20 °C 70%

O

O 10

11

F

F

Cl

NH N N

H N O

1. i. LiCl, THF, -7 °C ii. KOH/H 2O, -5 °C

OEt P OEt O

Cl

NH N

2. HCl (aq), then OEt NMe 2 EtO -5 °C to 30 °C

O

O

N

O

N O

O

100% for 2 steps

12

H N

13

F Cl maleic acid EtOH, 70 °C, 92%

NH N N

H N O

N O

O

COOH COOH

2

II Afatinib dimaleate

8

4. Canagliflozin hydrate (Invokana®) Canagliflozin, an orally active and selective sodium-glucose cotransporter 2 (SGLT2) inhibitor, was co-developed by Mitsubishi Tanabe Pharma and Johnson & Johnson (J&J) for the treatment of type 2 diabetes mellitus (T2DM) and obesity. The drug was approved in March by the U. S. FDA and launched in April 2013 in the U.S. SGLT2 is involved in the glucose re-absorption pathway in the kidney, and its inhibition increases urinary glucose excretion, and reduces plasma glucose and HbA1c levels.27 In addition, canagliflozin is safe in combination with other commonly used antidiabetic agents and has a significant effect on body weight reduction.28 A recently published process patent from ScinoPharm Taiwan describes the synthesis of canagliflozin. The preparation of the drug involves a convergent strategy whereby the union of the aglycone and glycoside components of the molecule ultimately secure the atomic framework of the API—the synthesis of each region and their union are described in Scheme 3.29 Synthesis of the aglycone region of canagliflozin was described in a separate patent by first condensing commercially available 5-bromo-2-methylbenzoyl chloride (14) and 2-(4-fluorophenyl)thiophene (15) under Friedel-Crafts acylation conditions to give ketone 16 in 69% yield as a crystalline solid.29 Ketone 16 was then reduced with triethylsilyl hydride in the presence of BF3·Et2O at low temperature to give aglycone bromide 17 in 70% yield. The precursor for the glycoside moiety, commercially available glycoside triol 18, was selectively treated with t-butyldiphenylsilyl chloride (TBDPSCl) in THF in the presence of imidazole to give the bis-silyl ether 19 in 81% yield. Next, a unique, stereospecific β-C-arylglucosidation was developed to secure the union of the aglyone- and glycoside-containing portions of canagliflozin. Bromide 17 was subjected to magnesium powder under standard Grignard conditions prior to treatment with AlCl3 in THF in situ. This resulting mixture was then exposed to a solution of compound 19 in PhOMe which had been pre-treated with n-BuLi, and the entire mixture was then warmed to 150 oC for 5 hours to ultimately give the β-anomer 20 in 56% yield.

9

Finally, removal of the silyl groups within 20 with tetrabutyl ammonium fluoride (TBAF) in THF delivered canagliflozin hydrate (III) in 73% yield (Scheme 3).

Scheme 3. Synthesis of canagliflozin hydrate (III)

5. Cetilistat (Oblean®) Cetilistat is a selective pancreatic lipase inhibitor which was approved in Japan in September 2013 for the treatment of obesity. The drug was discovered by Alizyme PLC and later co-developed with Takeda. Cetilistat demonstrated a lower incidence of adverse gastrointestinal events during a 12 week

10

clinical trial, and the degree of weight loss associated with cetilistat is comparable to that of other approved antiobesity therapies.30 The most likely process-scale preparation of cetilistat is described below in Scheme. 4.31 Commercially available hexadecanol (21) was treated with phosgene in THF/toluene to give the corresponding chloroformate (22), which was immediately subjected to commercial 2-amino-5methylbenzoic acid (23) in pyridine. Subsequent slow addition of methyl chloroformate at room temperature resulted in the formation of cetilistat (IV), which was produced in 31% overall yield from hexadecanol.31 Scheme 4. Synthesis of cetilistat (IV)

6. Cobicistat (Tybost®) Cobicistat, a selective, mechanism-based CYP3A inhibitor, was discovered and developed by Gilead Sciences, Inc. In 2013, European Medicines Agency (EMA) approved cobicistat (Tybost®) for the treatment of HIV-1 infection in combination with protease inhibitors (PIs) atazanavir or darunavir. Interestingly, cobicistat does not interact with HIV directly, but instead serves as a pharmacokinetic enhancer to boost the anti-HIV effect of atazanavir or darunavir through blockade of CYP3A.32

11

Cobicistat slows CYP-mediated metabolism of atazanavir and darunavir, resulting in prolonged systemic exposure of the drug(s).32 Cobicistat is also available as part of a fixed-dose combination tablet (Stribild®) of four additional drugs with CYP3A liabilities (elvitegravir, cobicistat, emtricitabine and tenofovir disoproxil fumarate), which was approved in U.S. in 2012, and subsequently approved in Europe and Japan in 2013. Although several synthetic routes have been reported,33-37 the improved process route by Gilead Sciences is described in Scheme 5 and 6.37 Commercial L-methionine (24) was treated with bromoacetic acid at elevated temperatures to afford aminolactone salt 25 in 70% yield. This material was then reacted with methyl aminomethylthiazole (26) in the presence of CDI and diisopropylethylamine to arrive at urea 27 in 91% yield. Next, lactone 27 underwent a ring-opening sequence upon exposure to trimethylsilyl iodide (TMSI) giving intermediate 28. The iodide was then displaced by morpholine, followed by treatment with oxalic acid to deliver the L-thiazole morpholine ethyl ester as the oxalate salt 29 in 71% yield for the sequence. Base-mediated hydrolysis of ethyl ester 29, followed by treatment of carboxylate 30 with monocarbonate hydrochloride 31 in the presence of EDCI and HOBT, provided cobicistat (V) in 76% yield for two steps. Of note, the preparation of mono-carbonate hydrochloride 31 arose from commercially available (S)-2-benzylaziridine (32) which was first condensed with N,N-dimethylsulfamoyl chloride to obtain Ntosyl-protected aziridine 33 in 77% yield (Scheme 6).

Next, a unique base-induced dimerization

reaction was employed to convert aziridine 33 to alkene 34. Presumably this proceeded through initial deprotonation at the methylene carbon within aziridine 33 upon exposure to lithium 2,2,6,6tetramethylpiperidine (LiTMP) resulting in an unstable trans-R-lithiated terminal aziridine.

This

lithiatethen underwent nucleophilic attack onto another molecule of 33 followed by elimination to give the 2-ene-1,4-diamine 34 in 72% yield.37-39 Removal of the sulfonyl groups with 1,3-diaminopropane followed by hydrogenation of the alkene provided diamine 36 in quantitative yield. Conversion to the diamine-dihydrogen chloride 37 through the use of HCl in dioxane was followed by a treatment with a single equivalent of base and 5-thiazolylmethyl carbonate 38 (prepared from bis-(4-nitrophenyl)-

12

carbonate (39) with 5-hydroxymethylthiazole). This sequence furnished amino carbamate 31, which then

participated in the coupling with carboxylate fragment 30 to prepare cobicistat as described above.

Scheme 5. Synthesis of cobicistat (V)

Scheme 6. Synthesis of intermediate 31 of cobicistat (V)

13

7. Dabrafenib Mesylate (Tafinlar®) Dabrafenib mesylate, sold by GlaxoSmithKline under the trade name Tafinlar®, was approved by the U.S. FDA in May 2013 for the treatment of metastatic BRAF-mutant melanoma. Dabrafenib reversibly inhibits the BRAF(V600E) mutant kinase as a selective ATP-competitive inhibitor which results in tumor regression.40 While the process-scale route has not yet been disclosed,41-43 the largest scale route to date is represented in Scheme 7.44 Commercially available fluoroaniline 4042 was first converted to sulfonamide 42 in 91% yield by treatment with 2,5-difluorobenzenesulfonyl chloride (41) in the presence of pyridine. Next, deprotonation of 2-chloro-4-methylpyrimidine (43) with lithium bis(trimethylsilyl)amide (LHMDS)

14

followed by addition to ester 42 afforded chloropyrimidine 44 in 72% yield. Bromination followed by thiazole formation through the use of 2,2-dimethylpropanethioamide gave the penultimate target 45 in 80% over two steps. Chloropyrimidine 45 was subjected to SNAr conditions with ammonium hydroxide to furnish the aminopyrimidine in 88% yield, and this was followed by exposure to methanesulfonic acid to afford dabrafenib mesylate (VI) in 85% yield.44 Scheme 7. Synthesis of dabrafenib mesylate (VI) F SO2Cl

F H2N

F

CO2Me

F

41

pyridine, CH2Cl2 15 °C to 25 °C 91%

F

O H N S O

N CO2Me

O H N S O

43 Cl

42

F F

N

LHMDS, THF, 0 °C 72%

F

40

Me

F

O

N N

S O

1. NBS, CH2Cl2, 10 to 20 °C Cl

F 44

2.

O

S

F

H N

N S N

F N

H2N t-Bu , DMA 20 °C to 75 °C

Cl

45

80% for 2 steps

1. NH4OH, 98 °C to 103 °C, 88% 2. MsOH, MeCN, 20 °C to 60 °C, 85%

F

H N

O

F

N S

S O

. CH SO H

N

F

3

N

3

NH2

VI Dabrafenib mesylate

8. Dolutegravir Sodium (Tivicay®)

15

Dolutegravir sodium (Tivicay®), developed and marketed by GlaxoSmithKline,45 was approved by the FDA in August 2013 as a novel integrase inhibitor for the treatment of HIV infection.46 Dolutegravir was fast-tracked by the FDA in February 2012,47 and joins an important class of drugs known as Integrase Strand Transfer inhibitors (INSTi’s).48 INSTi’s are characterized by their two-metal-chelating scaffolds, which are known to chelate Mg2+ cofactors in the enzyme active site,49, 50 interrupting function of HIV-1 integrase, which is essential for replication of viral DNA into host chromatin.49-51,52 Other drugs in this class, raltegravir and elvitegravir, are known to require either high dosages53 or PK boosting agents,54 respectively, with raltegravir also exhibiting substantial loss of potency in several major HIV-1 integrase mutation pathways.55 Dolutegravir was pursued with the goal of developing a novel INSTi with a once-daily, low-dosage treatment with improved resistance profile and without the need for the use of a PK boosting agent.51, 56 Dolutegravir sodium has been approved for treating a broad population of HIV-infected patients, including adults undergoing their first treatment as well as those who have been treated with other integrase transfer strand inhibiting agents.46 The most likely process-scale synthesis of dolutegravir sodium, as described in Scheme 8, began with benzyl protection and alkylation of pyrone 46 with benzaldehyde, yielding alcohol 47 in 74% over 2 steps (Scheme 8).57, 58 Alcohol mesylation and in-situ elimination provided the styrenyl olefin 48 in 94% yield, which further underwent an oxidative cleavage of the olefin to generate 49 by sequential addition of RuCl3/NaIO4 and NaClO2 (56% overall yield). Treatment of pyranone 49 with 3-amino-propane-2diol (50) in ethanol at elevated temperatures delivered the corresponding pyridinone in 83% yield, and this was followed by esterification and sodium periodate-mediated diol cleavage to furnish intermediate 51 in 71% overall yield across the two-step sequence.57, 58 Next, the key ring-forming step in the synthesis of dolutegravir sodium consisted of cyclization of 51 with (R)-3-amino-butan-1-ol, a process which relies on substrate control to provide the desired tricyclic carbamoylpyridone system 52 in high stereoselectivity (20/1 in favor of the desired isomer).51 Previously, cyclization of systems such as 51 with unsubstituted amino alcohols were found to yield a mixture of diastereomeric products, therefore indicating the pivotal role of the chiral amino alcohol in influencing stereochemical bias during the

16

overall cyclization step.51, 56 In practice, reaction of 51 with (R)-3-amino-butan-1-ol at 90 °C led to isolation of a single cyclization product 52, after recrystallization from EtOAc.57, 58

From 52, N-

bromosuccinimide (NBS) bromination and subsequent treatment with amine 53 under palladiumcatalyzed amidocarbonylative conditions led to amide 54 in 75% yield over 2 steps. Finally, removal of the benzyl group and subsequent crystallization using sodium hydroxide in water and ethanol provided dolutegravir sodium (VII) in 99% yield.57, 58 Scheme 8. Synthesis of dolutegravir sodium (VII)

17

9. Efinaconazole (Jublia®) Efinaconazole, marketed and developed by Valeant Pharmaceuticals International, was first approved for use in Canada in October 2013 under the brand name Jublia® for the treatment of onychomycosis, a fungal infection of the nail. Efinaconazole is believed to work by 14α-demethylase inhibition, which is a key pathway in ergosterol synthesis.59 Inhibition of ergosterol prevents secondary degenerative changes in the nail bed, plate, and surrounding tissue.59 Although several syntheses of efinaconazole have been reported, none have reported on kilogram-scale.60-64 However, as preparation of the penultimate epoxide (60) has been described on hundred-kilogram scale in the synthesis of ravuconazole,65 and final production of efinaconazole has been disclosed on a 24 g scale route, the presumed scale route is described in Scheme 9.66 Commercially available (R)-methyl lactate (55) was first converted to THP protected alcohol 57 in 4 steps and 78% yield via morpholino amide 56. Grignard displacement of the morpholine afforded ketone 58 in 81% yield. Next, ketone 58 was epoxidized by means of the Corey ylide followed by ringopening of the epoxide by triazole which had been activated by exposure to sodium t-butoxide. Finally, subjection to methanesulfonic acid furnished diol 59 in 51% yield as the corresponding mesylate salt. Diol 59 was then converted to epoxide 60 through the use of mesyl chloride and triethylamine in 78% yield and >99% ee. Finally, treatment of epoxide 60 with 4-methylene piperidine-HBr in the presence of lithium hydroxide afforded efinaconazole (VIII) in 87% yield. Scheme 9. Synthesis of efinaconazole (VIII)

18

O

1. MTBE, 60 °C 2. morpholine, 10 °C to 20 °C MeO2C

OH

O

O N

3. NaOMe, MeOH 5 °C to 20 °C

N

OH

MsOH, THF, 5 °C

O

78% for 4 steps

55

56

F MgBr

N

F F

57

N NH

N

1. Me3SOI, NaOt-Bu, THF OTHP

THF, 25 °C to 35 °C 81%, 98% ee

F

O

N

HO N

OH F

N

DMF, 99% ee F

OTHP O

HN

. HBr

LiOH, CH3CN 100 °C 87%

60

N

3

F

HO N

N

F

F VIII Efinaconazole

10. Elvitegravir (Viteka®) Elvitegravir is a quinolone-containing HIV integrase inhibitor discovered by Japan Tobacco and licensed to Gilead Pharmaceuticals for worldwide development with the exception of Japan.67 It was approved in Europe in 2013 for the treatment of HIV infection in adults having no known mutations associated with resistance to the drug.67 The drug interferes with HIV replication by preventing the virus from integrating into the DNA of human cells.67, 68 In addition to several patent applications that have been filed for the synthesis of elvitegravir, the discovery synthesis69 has also been published.69-80 The process route that has been reported on kilogram scale is highlighted in Scheme 10. None of the synthetic intermediates reportedly were isolated but instead were carried forward in the sequence. Therefore, no yields were provided in the lead reference.76

19

Commercial 2,4-dimethoxy-5-bromo benzoic acid (61) was reacted with 0.5 equiv of butylethylmagnesium to generate the dimagnesium salt in THF, which was then lithiated at -20 °C to give the aryl lithium species. The aryl lithium species was then reacted with the 2-fluoro-3-chloro benzaldehyde (62) to give alcohol 63. Treatment with triethylsilane in TFA resulted in removal of the hydroxyl functionality to provide benzoic acid 64. This acid was then reacted with carbonyldiimidazole and subsequently magnesium malonate 65 to give ketoester 66 after workup. Next, condensation with DMF-DMA converted ketoester 66 to the vinylogous amide 67, and this material was immediately subjected to an addition-elimination reaction involving (S)-valinol (68) in toluene at ambient temperature to provide intermediate 69. Warming the resulting intermediate 69 in the presence of N,Obistrimethylsilyl acetamide (BSA) and potassium chloride in DMF furnished the ring-closed quinolone 70. The ester 70 was saponified with potassium hydroxide in aqueous isopropanol and then acidified and crystallized with the use of seed crystals. Upon cooling, the crystalline product elvitegravir (IX) was collected by filtration. Scheme 10. Synthesis of Elvitegravir (IX)

20

11. Gemigliptin L-tartrate hydrate (Zemiglo®)

21

Gemigliptin is a prolyl-specific dipeptidyl aminopeptidase IV (DPP IV, DPP-4, CD26) inhibitor approved for the treatment of type 2 diabetes mellitus by the Korean Food and Drug Administration in 2012. Gemigliptin was discovered and developed by LG Life Sciences81 and is now the sixth DPP-4 inhibitor approved for the treatment of type 2 diabetes.82 At the time this review was prepared, there were no publications describing the discovery strategy and preclinical data that led to the advancement of gemigliptin to the clinic. Additionally, the synthesis of the drug has only been described in the patent literature.83-85 The molecule was prepared via a convergent route and the synthesis of the dihydropyridopyrimidine fragment is described in Scheme 11.85 Commercial N-Boc-3-piperidone (71) was treated with lithium hexamethyldisilazane (LHMDS) followed by ethyl trifluoroacetate to effect a Claisen condensation, producing diketone 72 in 81% yield. Cyclization of 72 with 2,2,2-trifluoroacetamide (73) gave bistrifluoromethyl dihydropyridopyrimidine 74 in 23% yield.

Removal of the Boc protecting group

efficiently provided amine 75 in 96% yield. Scheme 11. Synthesis of intermediate 75 of gemigliptin L-tartrate hydrate (X)

The synthesis of the carbon skeleton of the difluoropyridone fragment 80 is described in Scheme 12.84 1,4-Addition of ethyl bromodifluoroacetate (76) to ethyl acrylate (77) in the presence of copper

22

powder and tetramethylethylenediamine (TMEDA) gave diester 78, which was selectively reduced with sodium borohydride (NaBH4) to give alcohol 79 in 90% overall yield for the two-step procedure. Alcohol 79 was then treated with perfluorobutanesulfonyl chloride and triethylamine to give activated alcohol 80 in 75% yield. Scheme 12. Synthesis of intermediate 80 of gemigliptin L-tartrate hydrate (X)

The completion of the synthesis of gemigliptin is described in Scheme 13.83, 84 Boc-L-aspartic acid 4-tert-butyl ester (81) was treated with ammonium bicarbonate and pyridine in the presence of di-tertbutyl dicarbonate to give formamide 82. Dehydration of 82 to give nitrile 83 was accomplished through reaction with cyanuric chloride in 95% overall yield for the two-step sequence. Hydrogenation of 83 in the presence of Pearlman’s catalyst provided butyl amine 84. Alkylation of 84 with activated alcohol 80 in triethylamine followed by cyclization in acetic acid afforded difluoropyridone 85. Acidic hydrolysis of the ester proceeded with concomitant removal of the Boc protecting group, and was followed by reprotection of the amine with di-tert-butyl dicarbonate to give acid 86 in 84% overall yield for the three-step procedure in >97% ee.

Coupling of 86 with fragment 75 in the presence of

hydroxybenzotriazole (HOBT) and 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC) gave amide 87 in 51% yield. Removal of the Boc group with thionyl chloride in ethanol followed by neutralization with aqueous sodium hydroxide and salt formation with L-tartaric acid provided gemigliptin L-tartrate hydrate (X) in 97.5% yield.83

23

Scheme 13. Synthesis of gemigliptin L-tartrate hydrate (X)

12. Ibrutinib (Imbruvica®)

24

Ibrutinib is an irreversible inhibitor of Bruton’s tyrosine kinase (BTK) which was granted breakthrough status by the U.S. Food and Drug Administration in 2013 for the treatment of mantle cell lymphoma (MCL) and in 2014 for chronic lymphocytic leukemia (CLL).86 In preclinical studies involving CLL cells, the drug effectively promoted apoptosis, inhibited proliferation, and also prevented CLL cells from responding to survival stimuli provided by the microenvironment.87

Ibrutinib

demonstrated superiority over an anti-CD20 antibody (atumumab) in terms of disease progression measurements and overall survival of the patient.87

The drug, which was discovered by Celera

Pharmaceuticals and acquired by Pharmacyclics, was developed in partnership with Johnson & Johnson’s Janssen Pharmaceutical division. Although several different synthetic approaches to ibrutinib have been described in the patent literature,88-91 the most likely scale route is described in Scheme 14.9295

Condensation of commercially available 4-phenoxybenzoyl chloride (88) with malononitrile followed by acidic quench and O-methylation with dimethyl sulfate furnished vinyl dinitrile 89 in 84% yield over the three-step sequence. Next, treatment with hydrazine hydrate in refluxing ethanol secured aminopyrazole 90 and this was followed by treatment with neat formamide at elevated temperature to furnish pyrimidopyrazole 91 in excellent conversion. Selective alkylation of the pyrazole nitrogen with commercially-available (S)-piperidinyl tosylate (92) proceeded in 32% yield.95 Finally, liberation of the amide followed by pH adjustment and amide bond formation with acrolyl chloride furnished ibrutinib (XI) in 50% over the three-step sequence.

Scheme 14. Synthesis of Ibrutinib (XI)

25

13. Istradefylline (Nouriast®) Istradefylline (Nouriast®), a selective adenosine A2A inhibitor developed by Kyowa Pharmaceuticals, was approved in Japan in 2013 as an adjunctive therapy for the treatment of Parkinson’s disease (PD).96,97 A majority of therapies for PD, including the primary treatment, Levodopa,98 function via dopamine replacement.99 These treatments are very effective in the early stages of PD but they often exhibit dyskinesias symptoms in long-term treatment, leading to the inability to control motor fluctuations and therefore resulting in involuntary movements in patients.100-102 In contrast, istradefylline has been shown to reverse motor disability in monkeys and provide anti-Parkinsonian effects without exhibiting traditional symptoms of dyskinesias.103 While the ability to completely reproduce these

26

results in human PD patients with istradefylline therapy exclusively are still inconclusive,104 this oncedaily oral treatment has shown great potential for improving the quality of life for PD patients because of its effectiveness when used with other dopamine replacement treatments.102, 105 Numerous synthetic approaches to istradefylline have been developed, with a large majority of these methods employing 5,6-amino-1,3-diethyluracil 97 as a key intermediate.106-109 Despite the commercial availability of 96, most reported routes to istradefylline rely on sourcing of this intermediate via a wellestablished four-step synthesis from N,N-diethylurea (94) and cyanoacetic acid (95).106,

110, 111

Specifically, 6-amino-1,3-diethyluracil (96) can be formed by sequential treatment of 94 and 95 with Ac2O and NaOH.

Nitrosation of 96 with NaNO2/AcOH/H2O, followed by Na2S2O4/NH3-mediated

nitroso reduction provided 5,6-amino-1,3-diethyluracil (97).110, 111 Even though other groups have recently reported modified scale routes to istradefylline,106 the route described herein will focus on the sequence outlined by Kyowa Hakko Kogyo research laboratories during their initial development of istradefylline.109, 112-114 EDC-mediated amine coupling involving 97 and 3,4-dimethoxycinnamic acid (98) led to the corresponding amide intermediate. After aqueous workup, this crude amide intermediate underwent cyclization with aqueous sodium hydroxide to yield the desired purine dione 99 in 47% yield over 2 steps. Methylation of 99 with MeI/K2CO3 provided istradefylline (XII) in 68% yield (Scheme 15).109 Scheme 15. Synthesis of istradefylline (XII)

27

14. Levomilnacipran hydrochloride (Fetzima®) Levomilnacipran (Fetzima®) is a dual serotonin-norepinephrine reuptake inhibitor (SNRI) approved by the FDA in 2013 for the treatment of major depressive disorders (MDD).115-118 Levomilnacipran is the most active enantiomer of the racemate Milnacipran,119 which is currently used to treat pain associated with fibromyalgia.120, 121 The drug was developed by Forest Laboratories and the Pierre Fabre group.116, 122 Although initial enantiopure samples of levomilnacipran could be obtained by chromatographic separation of Milnacipran, this method was unable to provide sufficient quantities of material for pharmaceutical applications.123-125 Enantioselective routes have also been pursued,119, 126-128 but most rely on the use of sodium azide, which suffers from toxicity and stability issues on large scale.129 However, development of a scalable route to levomilnacipran has now been accomplished via optically active intermediate 102.119, 126, 128 This synthetic approach is described in Scheme 16.129

28

Reaction of phenylacetonitrile (100) and commercially available (R)-epichlorohydrin (101) with NaNH2 led to chloride displacement and intramolecular cyclopropanation, yielding lactone 102 after a one-pot nitrile hydrolysis and acid-promoted lactonization (75% yield over 3 steps). Lactone ringopening with Et2NH-AlCl3 complex provided amido-alcohol 103, which was converted to its phthalimido derivative 104 by sequential treatment with thionyl chloride and potassium phthalimide in 80% over three steps. Finally, levomilnacipran hydrochloride (XIII) was obtained in >95% optical purity after phthalimide cleavage, HCl salt formation, and crystallization from HCl/i-PrOH/i-PrAc. This sequence represents a highly efficient route to levomilnacipran, requiring no isolation of intermediates, resulting in >40% overall yield, and allowing use of the same solvent solution (toluene) for all steps. Scheme 16. Synthesis of levomilnacipran HCl (XIII)

15. Lomitapide mesylate (Juxtapid®) Lomitapide is an orally active microsomal triglyceride transfer protein (MTP) inhibitor for the treatment of hypercholesterolemia.130 The drug was developed by Aegerion Pharmaceuticals Inc. and licensed to Bristol-Myers Squibb Co. and the University of Pennsylvania.130 Lomitapide effectively

29

lowered LDL-cholesterol, both as a single agent and in combination with commonly prescribed lipidlowering therapies.130 Sold under the trade name Juxtapid®, the drug offers a new treatment option to patients who cannot tolerate statin therapy or who experience insufficient LDL-cholesterol reduction with the currently available therapies, such as patients with homozygous familial hypercholesterolemia caused by mutations in the LDLR gene.130 The most likely scale synthetic route to lomitapide mesylate is described in Scheme 17.131 Commercial 9H-fluorene-9-carboxylic acid (105) was alkylated with 1,4-dibromobutane in the presence of n-butyl lithium in THF to give 9-(4-bromobutyl)-9H-fluorene-9-carboxylic acid (106) in 85% yield. Next, activation of the acid as the acid chloride followed by coupling with (2,2,2trifluoroethylamine) provided amide 107 in 71% yield for the two-step sequence. Displacement of the terminal bromide with the appropriate 4-carbamoyl piperidine followed by removal of the Boc group furnished piperidinyl fluorine 108 in high yield. Amine 108 was then reacted with the acid chloride derived from acid 109 (derived from the Suzuki coupling of boronic acid 110 and o-iodobenzoic acid 111)132 to give lomitapide, and this was followed by salt formation with methanesulfonic acid to afford lomitapide mesylate (XIV).131 Scheme 17. Synthesis of lomitapide mesylate (XIV)

30

16. Macitentan (Opsumit®) Macitentan (Opsumit®) is an endothelin receptor antagonist marketed by Actelion,133 and was first approved in the U. S. in October 2013 for the treatmant of Pulmonary Arterial Hypertension (PAH).134, 135 Soon after, the drug obtained approval in Canada, and is currently under regulatory review in other countries.136 Macitentan exhibits greater inhibitory action of ETA versus ETB receptor agonists,137-139 with

31

higher potency than bosentan,138 allowing for once-daily dosing at significantly lower levels.140 The most likely scaleworthy route of macitentan is described in Scheme 18.139, 141 The preparation of the drug began with reaction of commercial 4-bromophenylacetate (112) with dimethylcarbonate (113) under basic conditions to yield the malonate ester 114.139, 141 Treatment of this diester with sodium methoxide and formamidine hydrochloride 115 provided the desired intermediate 4,6-dihydroxypyrimidine as a tautomeric mixture; from this system, dichloride 116 was generated in 6080% yield upon treatment with warm phosphorus oxychloride in N,N-dimethylaniline. Reaction of 116 with excess sulfonyl urea potassium salt 117139 provided chloropyrimidine 118 in high yield (83-93%). This was reacted with bromochloropyrimidine 119 in an SNAr reaction to provide macitentan (XV) in 88% yield.141 Synthesis of sulfamide potassium salt 117 was accomplished via sequential reaction of chlorosulfonyl isocyanate (120) with t-BuOH and propylamine/Et3N to provide ester sulfamide 121, followed by Boc removal and treatment with potassium t-butoxide to yield 117. This material could be isolated by trituration with diethyl ether. Scheme 18. Synthesis of macitentan (XV)

32

17. Olodaterol hydrochloride (Striverdi Respimat®) Olodaterol hydrochloride was approved for long-term, once-daily maintenance treatment of chronic obstructive pulmonary disease (COPD) in 2013 in the following countries: Canada, Russia, United Kingdom, Denmark, and Iceland.142, 143 The drug has been recommended by a federal advisory panel for approval by the FDA.142, 143 Developed and marketed by Boehringer Ingelheim, olodaterol is a longacting β2-adrenergic receptor agonist with high selectivity over the β1- and β3-receptors (219- and 1622-

33

fold, respectively).144 Upon binding to and activating the β2-adrenergic receptor in the airway, olodaterol stimulates adenyl cyclase to synthesize cAMP, leading to the relaxation of smooth muscle cells in the airway.

Administered by inhalation using the Respimat® Soft Mist inhaler, it delivers significant

bronchodilator effects within five minutes of the first dose and provides sustained improvement in forced expiratory volume (FEV1) for over 24 hours.143 While several routes have been reported in the patent and published literature,144-146 the manufacturing route for olodaterol hydrochloride disclosed in 2011 is summarized in Scheme 19 below.147 Commercial 2’,5’-dihydroxyacetophenone (122) was treated with one equivalent of benzyl bromide and potassium carbonate in methylisobutylketone (MIBK) to give the 5’-monobenzylated product in 76% yield. Subsequent nitration occurred at the 4’-position to provide nitrophenol 123 in 87% yield. Reduction of the nitro group followed by subjection to chloroacetyl chloride resulted in the construction of benzoxazine 124 in 82% yield. Next, monobromination through the use of tetrabutylammonium tribromide occurred at the acetophenone carbon to provide bromoketone 125, and this was followed by asymmetric reduction of the ketone employing (−)-DIP chloride to afford an intermediate bromohydrin, which underwent conversion to the corresponding epoxide 126 in situ upon treatment with aqueous NaOH. This epoxide was efficiently formed in 85% yield and 98.3% enantiomeric excess. Epoxide 126 underwent ring-opening upon subjection to amine 127 to provide amino-alcohol 128 in in 84-90% yield and 89.5-99.5% enantiomeric purity following salt formation with HCl. Tertiary amine 127 was itself prepared in three steps by reaction of ketone 129 with methylmagnesium chloride, Ritter reaction of the tertiary alcohol with acetonitrile, and hydrolysis of the resultant acetamide with ethanolic potassium hydroxide.

Hydrogenative removal of the benzyl ether within 128 followed by

recrystallization with methanolic isopropanol furnished olodaterol hydrochloride (XVI) in 63-70% yield.

Overall, the synthesis of olodaterol hydrochloride required 10 total steps (7 linear) from

commercially available acetophenone 122. Scheme 19. Synthesis of olodaterol hydrochloride (XVI)

34

18. Ospemifene (Osphena®) Ospemifene was approved by the U.S. FDA in February 2013 for treatment of moderate to severe dyspareunia (painful intercourse), a symptom of menopause-related vulvovaginal atrophy (VVA); it is the first non-hormonal treatment approved for this indication.148 Ospemifene was developed by QuatRx Pharmaceuticals, which acquired the drug as part of a merger with Hormos Medical in 2005, and was

35

licensed to Shionogi for regulatory filing and worldwide commercialization.148 It is a selective estrogen receptor modulator (SERM), and although it possesses a similar structure to tamoxifen and toremifene, ospemifene displays a unique set of tissue-specific estrogenic agonist/antagonist effects which includes an estrogen-like effect on vaginal epithelium.149 Although several synthetic routes have been reported,150-154 no manufacturing route for ospemifene has been disclosed to date. The shortest and largest scale route disclosed in the patent literature is summarized in Scheme 20.155 The drug can be synthesized succinctly in two steps. First, alkylation of commercially available 4hydroxybenzophenone (130) with ethylene carbonate and catalytic sodium iodide in refluxing toluene provided benzophenone 131 in 94% yield. This was followed by a McMurry coupling involving benzophenone 131 with chloropropiophenone 132 in the presence of zinc powder and titanium tetrachloride in 2-methyltetrahydrofuran. This reaction gave rise to a mixture of triphenylethylenes directly as a 5.5:1 ratio of Z to E isomers which could be separated by crystallization in aqueous methanol to give a mixture of olefins, 98% of which was comprised of the desired Z-isomer corresponding to ospemifene (XVII). The product purity was further improved by recrystallization to give 99.9% of the Z-isomer in 46% yield from 131. Thus, ospemifene was synthesized in two steps and 43% overall yield. Scheme 20. Synthesis of ospemifene (XVII)

19. Pomalidomide (Pomalyst®)

36

Pomalidomide, an anti-angiogenic derivative of thalidomide marketed by Celgene as Pomalyst®, was originally discovered in the early 1990s by D’Amato and co-workers at Boston Children’s Hospital.156 The drug was approved in February 2013 by the U.S. Food and Drug Administration (FDA) for the treatment of relapsed and refractory multiple myeloma, and received similar approval from the European Commission in August 2013 (the drug will be marketed in Europe under the name Imnovid®).157 Interestingly, the structural optimization of thalidomide and the related therapeutic agent lenalidomide led to the discovery of pomalidomide (XVIII), which is 10-fold more potent than lenalidomide as a TNF-α inhibitor inhibitor and and IL-2 IL stimulator, and has been shown to be effective in overcoming resistance to lenalidomide and thalidomide as well as the proteosome inhibitor bortezomib.158 A scalable preparation of pomalidomide (which has been developed as a racemate due to rapid interconversion of the R- and S- enantiomers in vivo)159, 160 involves the sequence described in Scheme 21.161 First, condensation of commercially available 3-nitrophthalic anhydride (133) and L-glutamine in warm DMF gave nitrophthalimide 134.161 Although the authors from Celgene do not explicitly describe the racemization of the stereocenter derived from L-glutamine, scrambling of the stereocenter has been reported during this step under neutral conditions at elevated temperatures.161 Next, hydrogenative reduction of the nitro group furnished the anilinophthalimide 135, and this was followed by treatment with CDI in refluxing acetonitrile to secure the piperidone dione and ultimately furnish pomalidomide (XVIII) as the racemate in 87% overall yield from 134. Scheme 21. Synthesis of pomalidomide (XVIII)

37

20. Riociguat (Adempas®) Riociguat is a potent, oral stimulator of soluble guanylate cyclase (sGC).162 Riociguat can sensitize sGC to endogenous nitric oxide (NO) by stabilizing NO-sGC binding, and also directly stimulate sGC in a NO-independent manner to increase generation of cGMP to affect subsequent vasodilation.162, 163 Discovered by Bayer Healthcare, riociguat obtained approval in Canada for the treatment of adults with persistent/recurrent Chronic Thromboembolic Pulmonary Hypertension (CTEPH), and was later approved by the U.S. FDA in 2013 for the treatments of both CTEPH and Pulmonary Arterial Hypertension (PAH).164 Several strategies for the assembly of the drug have been reported,165-171 and the process route is described below in Scheme 22.171 The sequence began with condensation of commericial 2-fluorobenzylhydrazine (136) with sodium ethyl cyanopyruvate (137), which derives from diethyl oxalate to generate aminopyrazole 138. This was followed by the cyclocondensation with 3-dimethylaminoacrolein (139) to access pyrazolopyridine 140 in 50% yield for the two-step operation. Next, ester 140 was transformed to the corresponding primary amide 141, which was subsequently dehydrated upon treatment with trifluoroacetic acid anhydride (TFAA) to construct nitrile 142 in quantitative yield from 140. Subjection of cyanopyrazole 142 to Pinner conditions using methoxide and ammonium chloride in refluxing acetic acid generated

38

amidine 143, and this was followed by condensation with the malononitrile derivative 144 in base to provide pyrimidine 145 in 73% yield. Hydrogenative cleavage of the phenyldiazine converted 145 to the pyrimidyl triamine 146, which underwent carbamoylation at the 4’ position to produce the penultimate carbamate 147. This carbamate was then selectively methylated through deprotonation of the carbamate N-H proton followed by quench with methyl iodide. Sequential recrystallization from warm DMSO and refluxing ethyl acetate produced riociguat (XIX) in 64% yield from 147. Scheme 22. Synthesis of riociguat (XIX)

39

21. Saroglitazar (Lipaglyn ®) Saroglitazar is an orally active PPAR-α and -γ dual agonist in a class of drugs referred to as glitazars. Saroglitazar was the first glitazar to be approved by the Drug Controller General of India, and was approved for the treatment of type II diabetes.172, 173 The drug was developed by Zydua Cadila, an

40

India-based pharmaceutical firm. Saroglitazar has been found to regulate lipid levels and exhibits improved glycemic HbA1c control in comparison to pioglitazone.172, 173 In addition, saroglitazar has been found to display no adverse effects associated with cardiac safety.172, 173 A scalable synthesis of this drug is described in Scheme 23.174 The sequence began with a Paal-Knorr pyrrole synthesis starting from commercial 1-[4(methylthio)phenyl]pentane-1,4-dione (148). Subjection of this diketone to ethanolamine in warm pivalic acid furnished pyrrole 149, which was taken forward as the crude product. Next, the alcohol was mesylated to afford 150, which was used without further purification. Williamson ether conditions were employed to convert mesylate 150 to the corresponding aryl ether 152 through the use of commercial phenol 151 and anhydrous potassium carbonate. This reaction proceeded in 80% yield. Saponification of the terminal ethyl ester using sodium hydroxide followed by acidic pH adjustment ultimately delivered the carboxylic acid drug saroglitazar (XX) in 98% yield from 152. Scheme 23. Synthesis of saroglitazar (XX)

41

22. Simeprevir (Olysio®; Sovriad®) Simeprevir is a hepatitis C viral (HCV) NS3/4A protease inhibitor approved in Japan, Canada and the U.S. for the treatment of chronic HCV infection in combination with ribavirin and pegylated interferon-α.175,

176

Simeprevir was discovered and developed at Medivir,177,

178

which was later

acquired by Janssen and co-developed by Tibotec, a subsidiary of Johnson & Johnson and Pharmasset (now Gilead). Simeprevir contains a 14-membered ring macrocycle and its process-scale synthesis was nicely described in a recent full paper on its discovery and development179, with individual process improvements reported throughout the patent literature.180-184 The synthesis of the quinolinol fragment 158 is described in Scheme 24.177, 179

42

Commercial 2-methyl-3-methoxybenzoic acid (153) was treated with diphenylphosphorylazide (DPPA) and triethylamine to affect a Curtius rearrangement and the resulting isocyanate was trapped with t-butanol to produce the Boc-protected aniline 154 in quantitative yield. Upon removal of the Boc protecting group with TFA, the resulting aniline was reacted with boron trichloride followed by the addition of acetonitrile and aluminum trichloride to affect Friedel-Crafts acylation to give aminoacetophenone 155 in 40% yield.

Acylation of the amino group with 4-isopropylthiazole-2-

carbonyl chloride (156) gave ketoamide 157 in 90% yield, which was treated with potassium tertbutoxide in t-butanol at 100 ˚C to furnish quinolinol 158 in 88% yield. Scheme 24. Synthesis of fragment 158 of simeprevir (XXI)

Use of a ring closing metathesis approach, enabling the synthesis of the macrocyclic portion of the drug and ultimately simeprevir, is described in Scheme 25.179-184 Hydrogenation of commercial transcyclopentanone-3,4-dicarboxylic acid (159) over Raney Ni in the presence of triethylamine followed by cyclization

to

the

lactone

using

2-chloro-4,6-dimethoxy-1,3,5-triazine

(CDMT)

and

N-

methylmorpholine (NMM), and subsequent cinchonidine salt formation gave lactone acid 160 in 26% yield over the 3 steps in 97% ee. Next, amide coupling with N-methylhexenylamine using Nethoxycarbonyl-2-ethoxy-1,2-dihydroquinoline

(EEDQ),

Fischer

esterification,

and

subsequent

43

introduction of the quinolinol fragment 158 under Mitsunobu conditions using triphenylphosphine (PPh3) and diisopropyl azodicarboxylate (DIAD) provided methyl ester 161 in 65% overall yield for the three steps. Saponification of the ester with lithium hydroxide followed by EEDQ-promoted coupling to (1R,2S)-1-amino-2-vinyl-cyclopropane ethyl ester (162)185 and Boc protection of the resulting amide gave the RCM substrate, diene 163 in 95% yield for the two steps. Macrocyclization of 163 using the second generation M2 catalyst186, 187 under dilute concentration in refluxing toluene followed by acidic removal of the amide protecting group gave cycloalkene ester 164 in high yield. Saponification of the ester, activation of the resulting acid with 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDCI), and coupling with cyclopropylsulfonamide led to simeprevir (XXI) in high overall yield. Scheme 25. Synthesis of simeprevir (XXI)

44

45

23. Sofosbuvir (Sovaldi®) Sofosbuvir is a NS5B polymerase inhibitor approved for the treatment of the hepatitis C virus (HCV) across several genotypes, demonstrating the ability to prevent virus replication within the body.188

Sofosbuvir was discovered at Pharmasset and developed by Gilead Sciences, and was

approved by the FDA in 2013.

Although the preparation of this unique nucleotide monotherapy

presents a variety of synthetic challenges,189-201 the penultimate target has been demonstrated on kilogram-scale according to reports in the published literature.

202, 203

Interestingly, the final step has

only been disclosed on a gram-scale, and this overall sequence is described in Scheme 26.204 The enantiopure unsaturated ester 166, which readily arises from olefination of the commerciallyavailable aldehyde 165, was subjected to ethylene glycol-promoted permanganate dihydroxylation conditions to afford diol 167 in 60% yield over the two steps.205 A three-step sequence was then employed to generate lactone 169. Diol 167 was converted to the cyclic sulfite and then oxidized with bleach to give the corresponding cyclic sulfate. Treatment with nucleophilic fluorine gave intermediate ammonium sulfonate 168, which, upon acidic hydrolysis of both the acetonide and sulfonate, underwent cyclization to give lactone 169. Next, bis-protection of diol 169 furnished 170 in 71% yield for the four-step sequence. Reduction and chlorination through the use of Red-Al® and sulfuryl chloride, respectively, constructed chlorotetrahydrofuran 171, which was subsequently reacted with commercial N-(2-oxo-1,2-dihydropyrimidin-4-yl)benzamide 172 in the presence of base and Lewis acid to afford 173 in 57% yield over the two steps. Treatment of 173 with AcOH and then ammonia in MeOH removed all benzoyl protection to give rise to diol 174 in 78% yield. Finally, treatment of 174 with pentafluorophenolic phosphonate ester 175 and tert-butylmagnesium chloride generated sofosbuvir (XXII) in 68% yield. The final step proceeds with excellent chirality transfer from 175 (99.7% ee). Notably, the preparation of the key phosphonate fragment was achieved in a simple two-step sequence beginning with alanine isopropyl ester 176. Phosphorylation in the presence of base at cryogenic temperatures, followed by treatment with pentafluorophenol, delivered scale quantities of 176 in 34% isolated yield and high enantiopurity (>98% ee) after recrystallization.

46

Scheme 26. Synthesis of sofosbuvir (XXII)

47

24. Topiroxostat (Uriadec®; Topiloric®) Topiroxostat is an orally-administered, non-purine, selective xanthine oxidase (XO) inhibitor developed for the treatment of hyperuricemia specifically for patients with gout in Japan.206 The drug was discovered and developed by Fuji Yakuhin.207 In contrast to conventional XO inhibitors such as febuxostat, topiroxostat interacts with key amino acid residues of the solvent channel.207 Of the few disclosed preparative approaches to the drug, the most likely scale assembly of topiroxostat is represented in Scheme 27.208, 209 The synthesis commenced with the reaction of two commercially available components, nitrile oxide 177 and isonicotinohydrazide (178). Condensation of these two subunits in the presence of sodium methoxide followed by acidic quench gave rise to 1,2,4-triazole 179 in 89% yield. Next, utilization of the N-oxide for installation of the nitrile functionality was required to furnish the drug, but it is interesting to note that this step has been the subject of study by Yamamoto and co-workers at Tohoku University in Japan.208 Although the process preparation describes the formation of the drug using sodium

cyanide

and

dimethylcarbamoyl

chloride

followed

by

isolation

through

a

salt

formation/freebasing process to deliver topiroxostat (XXIII) in 66% yield,209 Yamamoto has described this same sequence using zinc cyanide and tosylate salt formation (freebasing of the drug was not attempted).208 Scheme 27. Synthesis of topiroxostat (XXIII)

48

25. Trametinib dimethyl sulfoxide (Mekinist®) Trametinib, approved by the U.S. FDA in May 2013 under the brand name Mekinist®, is a drug discovered by Japan Tobacco and developed by GlaxoSmithKline for the treatment of metastatic BRAF-mutant melanoma.210 Trametinib is a reversible inhibitor of mitogen-activated protein kinase (MAPK), kinase MEK1 and MEK2, which are downstream from BRAF in the MAPK pathway, resulting in an inhibition of growth factor-mediated cell signaling and cellular proliferation in various cancers.211 In January 2014, the U. S. FDA also granted an accelerated approval for the combination of trametinib and darafenib for the treatment of patients with BRAF V600E/K-mutant metastatic melanoma.210 Two synthetic strategies have been reported for trametinib;212, 213 and the scalable route is shown in Scheme 28.213 Commericial 2-fluoro-4-iodoaniline (180) was sequentially subjected to CDI and cyclopropylamine to generate urea 181 in 96% yield. This was followed by coupling with cyanoacetic acid in the presence of mesyl chloride and DMF to furnish imide 182 in 96% yield. Under basic conditions, imide 182 underwent an intramolecular cyclization reaction to produce pyrimidine-2,4-dione 183 in 88% yield. Next, condensation with DMF-DMA generated formamidine 184 in 92% yield, and this was followed by NaBH4-mediated reduction and subsequent annulation with 2-methyl-malonic acid (186) to arrive at

49

trione 187 in 58% from 184. Trione 187 was then treated with p-toluenesulfonyl chloride in Et3N, and the resulting tosylate was exposed to 3’-aminoacetanilide (189) in the presence of 2,6-lutidine and DMA, inducing an addition-elimination reaction to give pyrido[2,3-d]pyrimidine 190 in 93% yield. The rearrangement of pyrido[2,3-d]pyrimidine 190 with sodium methoxide in THF/MeOH gave pyrido[4,3d]pyrimidine (trametinib) in 89% yield. This was then complexed with a single equivalent of DMSO to produce trametinib DMSO (XXIV) in 92% yield.

Scheme 28. Synthesis of trametinib dimethyl sulfoxide (XXIV)

50

26. Trastuzumab emtansine (Kadcyla®)

51

Trastuzumab emtansine is an antibody drug conjugate that is comprised of an anti-human epidermal growth factor receptor 2 (HER2) antibody, trastuzumab, and the potent tubulin based inhibitor, maytansine DM1.214,

215

These two entities are connected together via a linker that attaches the

cytotoxin through surface exposed lysine side chains on the antibody. The linker is stable and does not contain a cleavage element that has been present in other ADCs such as Acetris® or Mylotarg®. Instead, after the ADC is internalized it is catabolized within the tumor cell and releases the maytansinebased linker payload with the lysine from the antibody still attached; this molecule is ultimately responsible for destruction of the tumor cell. Trastuzumab emtansine was discovered and developed through a collaboration between Immunogen214, 216 and Genentech, and it has been approved for the treatment of patients with HER2-positive metastatic breast cancer who have previously received trastuzumab and a taxane.

The synthesis of trastuzumab emtansine has only been described on small

scale, so it is unclear if these methods have translated to a production scale. The synthesis of the Nacyl-N-methyl-L-alanine precursor that will be connected to maytansinol is described in Scheme 29.216, 217

First, commercial 3-mercaptopropanoic acid (191) was treated with methanethiolsufonate to give the corresponding methyldithio analog 192 in 90% yield.

Activation of the acid with N-

hydroxysuccinimide in the presence of 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDCI) provided the activated ester 193, which was reacted with N-methyl-L-alanine (194) to give acid 195 in 60% yield from compound 192. Scheme 29. Synthesis of fragment 195 of trastuzumab emtansine (XXV)

52

Preparation of the DM1 linker-payload is described in Scheme 30. The starting material used for the production of DM1 is ansamitocin P-3 (196), which is produced via fermentation of the microorganism Actinosynnema pretiosum. The ester group of 196 was removed using a reductive process in the presence of lithium trimethoxyaluminum hydride to give maytansinol 197 in 85% yield.216, 218 The use of reductive conditions was required to avoid subsequent elimination to the α,βunsaturated amide. Esterification with 195 in the presence of dicyclohexylcarbodiimide (DCC) and zinc chloride provided DM1-SMe 198 in 32% yield.216,

217

Reductive removal of the dithiane using

dithiothreitol (DTT) in aqueous buffer at pH 7.5 gave DM1 thiol 199 in 76% yield, which was utilized in the conjugation to trastuzumab (200). Scheme 30. Synthesis of fragment 199 of trastuzumab emtansine (XXV)

53

Completion of the synthesis of trastuzumab emtansine is described in Scheme 31. The surface accessible lysine residues of trastuzumab (200) were treated with succinimidyl-4-(N-maleimidomethyl)cyclohexane-carboxylate (SMCC, 201) in pH 7.0 buffer to give amide 202 with approximately four SMCC molecules added per antibody in 88% yield.216, 219, 220 Next, the free thiol group of DM1 (199) was conjugated to the maleimide groups present on 202 to give trastuzumab emtansine (XXV) with an average 3.5 drug molecules loaded per antibody. Scheme 31. Synthesis of trastuzumab emtansine (XXV)

54

27. Vortioxetine (Brintellix®) Vortioxetine is an antidepressant developed by Lundbeck and Takeda and approved in 2013 by the FDA for the treatment of major depressive disorder (MDD) in adult patients. This multi-modal acting drug, which is marketed in North America under the trade name Brintellix®, exhibits a high affinity for a range of serotonergic targets.221 The anti-depressant effects of vortioxetine are thought to be mediated through three serotonergic targets: inhibition of the 5-HT re-uptake which leads to an increase in extracellular 5-HT levels in the brain (in analogy to previous antidepressants), agonism of 5-HT1AR (which is believed to shorten the time to onset of clinical effects), and antagonism of 5-HT3R.222 Preclinical studies have shown that antagonism of 5-HT3R could have positive effects on mood and cognitive dysfunction in patients with depression. 223 Although several approaches to the synthesis of vortioxetine have been reported,224-229 Bang-Andersen and co-workers at Lundbeck describe three

55

approaches to producing vortioextine on scale.221 The most practical approach according to the authors, which has been executed on multigram scale, is depicted in Scheme 32.224 The sequence involves iterative palladium-catalyzed carbon-heteroatom bond formations, the first establishing the thioethereal bond between commercially available thiol (213) and o-iodobromobenzene (214) employing conditions described by Schopfer and Schlapbach.230

Next, Buchwald-Hartwig

conditions were employed to establish the piperazine linkage,231, 232 and this was followed by subjection to warm hydrobromic acid to furnish vortioxetine hydrobromide (XXVI) in 75% yield across the entire three-step sequence.224 Scheme 32. Synthesis of vortioxetine hydrobromide (XXVI)

Conclusion In summary, the examples cited in this brief account illustrate the ever-increasing power and potential of synthetic innovation as a major driver in providing access to the 26 drugs approved in late 2012 and 2013. The structures and synthetic routes described herein may be extended to the preparation of additional collections of unique compounds aimed at providing novel therapies or treatments, or as improvements upon current ones. The presumed scalable synthesis discussed within not only serves to provide the larger community with a means to access these chemical architectures, but to raise the awareness of modern process-scale synthetic capability. Furthermore, the work presented herein and in

56

subsequent editions of this review will serve as a catalyst for future inspiration in the synthesis of medicinally-relevant tool compounds, natural products, and drugs of current interest to the larger scientific community and society.

ABBREVIATIONS 1,2-DAP = 1,2-Diaminopropane 1,2-DCE = 1,2-Dichloroethane Ac = Acetyl aq. = Aqueous Bn = Benzyl Bz = Benzoyl Boc = t-Butoxycarbonyl B2(pin)2 = Bis(pinacolato)diboron BINAP = 2,2'-bis(diphenylphosphino)-1,1'-binaphthyl BSA = N,O-bistrimethylsilyl acetamide CDI = N,N'-Carbonyldiimidazole CDMT = 2-Chloro-4,6-dimethoxy-1,3,5-triazine DAP = Diaminopropane Dba = Dibenzylideneacetone DBU =1,5-Diazabicycolo[4.3.0]non-5-ene DCC = 1,3-Dicyclohexylcarbodiimide DCE = Dichloroethane DCM = Dichloromethane DIAD = Diisopropyl azodicarboxylate DIC = 1,3-Diisopropylcarbodiimide DIEA/DIPEA = Diisopropylethylamine (-)-DIP-Chloride = (-)-Diisopinocampheyl chloroborane DMA = Dimethylacetamide DMAP = 4-Dimethylaminopyridine DME = Dimethoxyethane DMF = N,N-Dimethylformamide DMSO = Dimethyl sulfoxide DPPA = Diphenylphosphoryl azide EDCI = N-(3-Dimethylaminopropal)-N'-ethylcarbodiimide EDTA = Ethylenediaminetrteaacetic acid EEDQ = N-ethoxycarbonyl-2-ethoxy-1,2-dihydroquinoline HBTU = 2-(1H-Benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate HMDS = Bis(trimethylsilyl)amide HOBT = 1-Hydroxybenzotriazole hydrate IPA = Isopropyl alcohol IPAc = Isopropyl acetate LAH = Lithium aluminum hydride LHMDS = Lithium bis(trimethylsilyl)amide MIBK = Methyl isobutyl ketone MsOH = Methansulfonic acid MsCl = Methanesulfonic chloride

57

MTBE = Methyl tert-butyl ether NaHMDS = Sodium bis(trimethylsilyl)amide NBS = N-Bromosuccinimide NMM = N-Methylmorpholine NMP = N-methyl-2-pyrrolidone pin = Pinacol Py = Pyridine rt = Room temperature TBAB = Tetrabutylammonium bromide TBAF = t-Butyl ammonium fluoride TFA = Trifluoroacetic acid TFAA = Trifluoroacetic anhydride THF = Tetrahydrofuran TMEDA = Tetramethylethylenediamine TMP = 2,2,6,6-tetramethylpiperidine TMSCl = Trimethylsilyl chloride TMSI = Trimethylsilyl iodide TBDPS = tert-Butyl diisopropylsilyl Ts = Tosyl (p-toluenesulfonyl)

References and Notes

1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19.

Raju, T. N. K. Lancet 2000, 355, 1022. Li, J.; Liu, K. K.-C. Mini-Rev. Med. Chem. 2004, 4, 207. Liu, K. K.-C.; Li, J.; Sakya, S. Mini-Rev. Med. Chem. 2004, 4, 1105. Li, J.; Liu, K. K.-C.; Sakya, S. Mini-Rev. Med. Chem. 2005, 5, 1133. Sakya, S. M.; Li, J.; Liu, K. K.-C. Mini-Rev. Med. Chem. 2007, 7, 429. Liu, K. K.-C.; Sakya, S. M.; Li, J. Mini-Rev. Med. Chem. 2007, 7, 1255. Liu, K. K.-C.; Sakya, S. M.; O'Donnell, C. J.; Li, J. Mini-Rev. Med. Chem. 2008, 8, 1526. Liu, K. K.-C.; Sakya, S. M.; O'Donnell, C. J.; Li, J. Mini-Rev. Med. Chem. 2009, 9, 1655. Liu, K. K.-C.; Sakya, S. M.; O'Donnell, C. J.; Flick, A. C.; Li, J. Bioorg. Med. Chem. 2011, 19, 1136. Liu, K. K.-C.; Sakya, S. M.; O’Donnell, C. J.; Flick, A. C.; Ding, H. X. Bioorg. Med. Chem. 2012, 20, 1155. Ding, H. X.; Liu, K. K.; Sakya, S. M.; Flick, A. C.; O'Donnell, C. J. Bioorg. Med. Chem. 2013, 21, 2795. Ding, H. X.; Leverett, C. A.; Kyne, R. E., Jr.; Liu, K. K.; Sakya, S. M.; Flick, A. C.; O'Donnell, C. J. Bioorg. Med. Chem 2014, 22, 2005. Graul, A. I.; Cruces, E.; Stringer, M. Drugs Today (Barc) 2014, 50, 51. Nowlan, M. L.; Scott, L. J. Drugs 2013, 73, 1377. Sorbera, L. A.; Castañer, J.; Leeson, P. A. Drugs Future 2003, 28, 26. Tack, J.; Janssen, P. Expert Opin. Investig. Drugs 2011, 20, 701. Nagasawa, M.; Murata, M.; Nishioka, H.; Kurimoto, T.; Ueki, S.; Kitagawa, O. WO Patent 9636619A1, 1996. Nagasawa, M.; Nishioka, H.; Suzuki, T.; Nagano, E.; Ishii, K.; Nakao, R. WO Patent 9858918A1, 1998. Keating, G. M. Drugs 2014, 74, 207.

58

20. 21.

22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. 41.

42.

43. 44. 45. 46. 47. 48. 49.

Nishino, S.; Hirotsu, K.; Shima, H.; Harada, T.; Oda, H.; Takahashi, T.; Suzuki, S. WO Patent 2003064399A1, 2003. Carmi, C.; Galvani, E.; Vacondio, F.; Rivara, S.; Lodola, A.; Russo, S.; Aiello, S.; Bordi, F.; Costantino, G.; Cavazzoni, A.; Alfieri, R. R.; Ardizzoni, A.; Petronini, P. G.; Mor, M. J. Med. Chem. 2012, 55, 2251. Xu, X. N. CN Patent 103288808A, 2013. Xu, X. N. CN Patent 103254183A, 2013. Chen, Q. C.; Zhao, J.; Zhao, X. W.; Li, Z. CN Patent 103755688A, 2014. Schroeder, J.; Dziewas, G.; Fachinger, T.; Jaeger, B.; Reichel, C.; Renner, S. WO Patent 2007085638A1, 2007. Soyka, R.; Rall, W.; Schnaubelt, J.; Sieger, P.; Kulinna, C. US Patent 20050085495A1, 2005. Sha, S.; Devineni, D.; Ghosh, A.; Polidori, D.; Chien, S.; Wexler, D.; Shalayda, K.; Demarest, K.; Rothenberg, P. Diabetes, Obes. Metab. 2011, 13, 669. Qiu, R.; Capuano, G.; Meininger, G. J. Clin. Transl. Endocrinol. 2014, 1, 54. Henschke, J. P.; Lin, C.-W.; Wu, P.-Y.; Hsiao, C.-N.; Liao, J.-H.; Hsiao, T.-Y. US Patent 20140128595A1, 2014. Kopelman, P.; Groot, G. d. H.; Rissanen, A.; Rossner, S.; Toubro, S.; Palmer, R.; Hallam, R.; Bryson, A.; Hickling, R. I. Obesity 2010, 18, 108. Hodson, H. F.; Downham, R.; Mitchell, T. J.; Carr, B. J.; Dunk, C. R.; Palmer, R. M. J. US Patent 20030027821A1, 2003. Deeks, E. D. Drugs 2014, 74, 195. Desai, M. C.; Hong, A. Y.; Hui, H. C.; Liu, H.; Vivian, R. W.; Xu, L. WO Patent 2008103949A1, 2008. Desai, M. C.; Hong, A. Y.; Liu, H.; Xu, L.; Vivian, R. W. WO Patent 2008010921A2, 2008. Koziara, J. M.; Menning, M. M.; Oliyai, R.; Strickley, R. G.; Yu, R.; Kearney, B. P.; Mathias, A. A. WO Patent 2009135179A2, 2009. Xu, L.; Liu, H.; Murray, B. P.; Callebaut, C.; Lee, M. S.; Hong, A.; Strickley, R. G.; Tsai, L. K.; Stray, K. M.; Wang, Y.; Rhodes, G. R.; Desai, M. C. ACS Med. Chem. Lett. 2010, 1, 209. Polniaszek, R.; Pfeiffer, S.; Yu, R.; Cullen, A.; Dowdy, E.; Tran, D.; Kent, K.; Zhou, Z.; Cordeau, D.; Easton, L. WO Patent 2010115000A2, 2010. Hodgson, D. M.; Humphreys, P. G.; Miles, S. M.; Brierley, C. A. J.; Ward, J. G. J. Org. Chem. 2007, 72, 10009. Hodgson, D. M.; Miles, S. M. Angew. Chem. Int. Ed. Engl. 2006, 45, 935. Menzies, A. M.; Long, G. V. Clin. Cancer. Res. 2014, 20, 2035. Adams, J. L.; Dickerson, S. H.; Johnson, N. W.; Kuntz, K.; Petrov, K.; Ralph, J. M.; Rheault, T. R.; Schaaf, G.; Stellwagen, J.; Tian, X.; Uehling, D. E.; Waterson, A. G.; Wilson, B. WO Patent 2009137391A2, 2009. Rheault, T. R.; Stellwagen, J. C.; Adjabeng, G. M.; Hornberger, K. R.; Petrov, K. G.; Waterson, A. G.; Dickerson, S. H.; Mook, R. A.; Laquerre, S. G.; King, A. J.; Rossanese, O. W.; Arnone, M. R.; Smitheman, K. N.; Kane-Carson, L. S.; Han, C.; Moorthy, G. S.; Moss, K. G.; Uehling, D. E. ACS Med. Chem. Lett. 2013, 4, 358. Xu, X. CN Patent 103588767A, 2014. Hoos, A.; Greshock, J. WO Patent 2014066606A2, 2014. Johns, B. A.; Kawasuji, T.; Taishi, T.; Taoda, Y. WO Patent 2006116764A1, 2006. http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm364744.htm. http://newdrugapprovals.org/2013/07/16/dolutegravir-biggest-rival-to-worlds-best-selling-hivdrug-atripla-may-get-fda-approval-by-august-2013/. Pendri, A.; Meanwell, N. A.; Peese, K. M.; Walker, M. A. Expert Opin. Ther. Pat. 2011, 21, 1173. Johns, B. A.; Svolto, A. C. Expert Opin. Ther. Pat. 2008, 18, 1225.

59

50. 51.

52. 53.

54. 55. 56.

57. 58. 59. 60. 61. 62. 63. 64. 65.

66. 67. 68. 69.

70. 71. 72. 73. 74. 75. 76.

Johns, B. A.; Weatherhead, J. G.; Allen, S. H.; Thompson, J. B.; Garvey, E. P.; Foster, S. A.; Jeffrey, J. L.; Miller, W. H. Bioorg. Med. Chem. Lett. 2009, 19, 1802. Johns, B. A.; Kawasuji, T.; Weatherhead, J. G.; Taishi, T.; Temelkoff, D. P.; Yoshida, H.; Akiyama, T.; Taoda, Y.; Murai, H.; Kiyama, R.; Fuji, M.; Tanimoto, N.; Jeffrey, J.; Foster, S. A.; Yoshinaga, T.; Seki, T.; Kobayashi, M.; Sato, A.; Johnson, M. N.; Garvey, E. P.; Fujiwara, T. J. Med. Chem. 2013, 56, 5901. Kawasuji, T.; Johns, B. A.; Yoshida, H.; Taishi, T.; Taoda, Y.; Murai, H.; Kiyama, R.; Fuji, M.; Yoshinaga, T.; Seki, T.; Kobayashi, M.; Sato, A.; Fujiwara, T. J. Med. Chem. 2012, 55, 8735. Lennox, J. L.; De Jesus, E.; Lazzarin, A.; Pollard, R. B.; Valdez Ramalho Madruga, J.; Berger, D. S.; Zhao, J.; Xu, X.; Williams-Diaz, A.; Rodgers, A. J.; Barnard, R. J. O.; Miller, M. D.; Di Nubile, M. J.; Nguyen, B.-Y.; Leavitt, R.; Sklar, P. Lancet 2009, 374, 796. Ramanathan, S.; Mathias, A. A.; German, P.; Kearney, B. P. Clin. Pharmacokinet. 2011, 50, 229. Ceccherini-Silberstein, F.; Malet, I.; D'Arrigo, R.; Antinori, A.; Marcelin, A.-G.; Perno, C.-F. AIDS Rev. 2009, 11, 17. Kawasuji, T.; Johns, B. A.; Yoshida, H.; Weatherhead, J. G.; Akiyama, T.; Taishi, T.; Taoda, Y.; Mikamiyama-Iwata, M.; Murai, H.; Kiyama, R.; Fuji, M.; Tanimoto, N.; Yoshinaga, T.; Seki, T.; Kobayashi, M.; Sato, A.; Garvey, E. P.; Fujiwara, T. J. Med. Chem. 2013, 56, 1124. Johns, B. A.; Duan, M.; Hakogi, T. WO Patent 2010068262A1, 2010. Yoshida, H.; Taoda, Y.; Johns, B. A. WO Patent 2010068253A1, 2010. Patel, T.; Dhillon, S. Drugs 2013, 73, 1977. Naito, T.; Kobayashi, H.; Ogura, H.; Nagai, K.; Nishida, T.; Arika, T.; Yokoo, M.; Nakahashi, S. WO Patent 9426734A1, 1994. Ogura, H.; Kobayashi, H.; Nagai, K.; Nishida, T.; Naito, T.; Tatsumi, Y.; Yokoo, M.; Arika, T. Chem. Pharm. Bull. 1999, 47, 1417. Naito, T.; Kobayashi, H.; Ogura, H.; Nagai, K.; Nishida, T.; Arika, T.; Yokoo, M.; Shusse, S. US Patent 5962476A, 1999. Mimura, M.; Watanabe, M.; Ishiyama, N.; Yamada, T. WO Patent 2012029836A1, 2012. Tamura, K.; Kumagai, N.; Shibasaki, M. J. Org. Chem. 2014, 79, 3272. 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. Org. Process Res. Dev. 2009, 13, 716. Mimura, M.; Watanabe, M.; Ishiyama, N.; Yamada, T. US Patent 20130150586A1, 2012. Deeks, E. D. Drugs 2014, 74, 687. Sorbera, L. A.; Serradell, N. Drugs Future 2006, 31, 310. Sato, M.; Motomura, T.; Aramaki, H.; Matsuda, T.; Yamashita, M.; Ito, Y.; Kawakami, H.; Matsuzaki, Y.; Watanabe, W.; Yamataka, K.; Ikeda, S.; Kodama, E.; Matsuoka, M.; Shinkai, H. J. Med. Chem. 2006, 49, 1506. Satoh, M.; Kawakami, H.; Itoh, Y.; Shinkai, H.; Motomura, T.; Aramaki, H.; Matsuzaki, Y.; Watanabe, W.; Wamaki, S. WO Patent 2004046115A1, 2004. Satoh, M.; Motomura, T.; Matsuda, T.; Kondo, K.; Ando, K.; Matsuda, K.; Miyake, S.; Uehara, H. WO Patent 2005113508A1, 2005. Matsuda, K.; Ando, K.; Ohki, S.; Yamasaki, T.; Hoshi, J.-I. US Patent 20090318702A1, 2009. Matsuda, K.; Ando, K.; Ohki, S.; Hoshi, J.-I.; Yamasaki, T. US Patent 20130310595A1, 2013. Satoh, M.; Kawakami, H.; Itoh, Y.; Shinkai, H.; Motomura, T.; Aramaki, H.; Matsuzaki, Y.; Watanabe, W.; Wamaki, S. US Patent 20130172344A1, 2013. Dowdy, E.; Chen, X.; Pfeiffer, S. WO Patent 2008033836A2, 2008. Dowdy, E.; Pfeiffer, S. WO Patent 2009036161A1, 2009.

60

77. 78. 79. 80.

81. 82. 83. 84. 85.

86. 87.

88. 89. 90. 91. 92. 93. 94.

95. 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. 106. 107. 108. 109. 110. 111. 112.

Vellanki, S. R. P.; Dhake, V. N.; Ravi, S.; Nuchu, R.; Puliyala, R.; Shaik, M. B.; Datta, D. WO Patent 2011004389A2, 2011. Radl, S. WO Patent 2014056465A1, 2014. Pcion, D.; Tomasi, C.; Whitcomb, M. C.; Dowdy, E. D.; Fu, W.; MacLeod, P. WO Patent 2014022707A1, 2014. Satoh, M.; Matsuda, T.; Okuda, S.; Kawakami, H.; Aramaki, H.; Shinkai, H.; Matsuzaki, Y.; Watanabe, W.; Yamataka, K.; Kiyonari, S.; Wamaki, S.; Takahashi, M.; Yamada, N.; Nagao, A. WO Patent 2005113509A1, 2005. Kim, S.-H.; Lee, S.-H.; Yim, H.-J. Arch. Pharmacal Res. 2013, 36, 1185. Juillerat-Jeanneret, L. J. Med. Chem. 2014, 57, 2197. Park, K. S.; Yun, J. M.; Kim, B. C.; Kim, K. Y.; Lee, J. H. WO Patent 2012060590A2, 2012. Kim, B. C.; Kim, K. Y.; Lee, H. B.; An, J. E.; Lee, K. W. WO Patent 2012030106A2, 2012. Lee, C.-S.; Koh, J. S.; Koo, K. D.; Kim, G. T.; Kim, K.-H.; Hong, S. Y.; Kim, S.; Kim, M.-J.; Yim, H. J.; Lim, D.; Kim, H. J.; Han, H. O.; Bu, S. C.; Kwon, O. H.; Kim, S. H.; Hur, G.-C.; Kim, J. Y.; Yeom, Z.-H.; Yeo, D.-J. WO Patent 2006104356A1, 2006. Young, R. M.; Staudt, L. M. Cancer Cell 2014, 26, 11. Herman, S. E. M.; Gordon, A. L.; Hertlein, E.; Ramanunni, A.; Zhang, X.; Jaglowski, S.; Flynn, J.; Jones, J.; Blum, K. A.; Buggy, J. J.; Hamdy, A.; Johnson, A. J.; Byrd, J. C. Blood 2011, 117, 6287. Buggy, J. J.; Elias, L. US Patent 20140079690A1, 2014. Honigberg, L.; Verner, E.; Buggy, J.; Loury, D.; Chen, W. WO Patent 2010009342A2, 2010. Honigberg, L.; Verner, E.; Buggy, J. J.; Loury, D.; Chen, W. WO Patent 2008121742A2, 2008. Xu, X. CN Patent 103626774A, 2014. Buggy, J. J.; Chang, B. Y. WO Patent 2013003629A2, 2013. Honigberg, L.; Verner, E.; Pan, Z. US Patent 20080108636A1, 2008. Pan, Z.; Scheerens, H.; Li, S.-J.; Schultz, B. E.; Sprengeler, P. A.; Burrill, L. C.; Mendonca, R. V.; Sweeney, M. D.; Scott, K. C. K.; Grothaus, P. G.; Jeffery, D. A.; Spoerke, J. M.; Honigberg, L. A.; Young, P. R.; Dalrymple, S. A.; Palmer, J. T. Chem. Med. Chem. 2007, 2, 58. Goldstein, D. M.; Brameld, K. A. WO Patent 2012158764A1, 2012. http://www.kyowa-kirin.com/news_releases/2013/e20130325_04.html. Park, A.; Stacy, M. Expert Opin. Pharmacother. 2012, 13, 111. http://www.parkinson.org/Parkinson-s-Disease/Treatment/Medications-for-Motor-Symptoms-ofPD/Carbidopa-levodopa. Di Stefano, A.; Sozio, P.; Cerasa, L. S.; Iannitelli, A. Curr. Pharm. Des. 2011, 17, 3482. Dungo, R.; Deeks, E. D. Drugs 2013, 73, 875. Homayoun, H.; Goetz, C. G. Future Neurol. 2012, 7, 127. LeWitt, P. A.; Guttman, M.; Tetrud, J. W.; Tuite, P. J.; Mori, A.; Chaikin, P.; Sussman, N. M. Ann. Neurol. 2008, 63, 295. Grondin, R.; Bedard, P. J.; Hadj, T. A.; Gregoire, L.; Mori, A.; Kase, H. Neurology 1999, 52, 1673. Perez-Lloret, S.; Merello, M. Expert Opin. Pharmacother. 2014, 15, 1097. Pinna, A. CNS Drugs 2014, 28, 455. Hockemeyer, J.; Burbiel, J. C.; Muller, C. E. J. Org. Chem. 2004, 69, 3308. Jorg, M.; Shonberg, J.; Mak, F. S.; Miller, N. D.; Yuriev, E.; Scammells, P. J.; Capuano, B. Bioorg. Med. Chem. Lett. 2013, 23, 3427. Luo, F.; He, Z.; Zhu, G.; Li, Y. CN Patent 103254194A, 2013. Suzuki, F.; Shimada, J.; Koike, N.; Nakamura, J.; Shiozaki, S.; Ichikawa, S.; Nonaka, H. EP Patent 590919A1, 1994. Blicke, F. F.; Godt, H. C., Jr. J. Am. Chem. Soc. 1954, 76, 2798. Speer, J. H.; Raymond, A. L. J. Am. Chem. Soc. 1953, 75, 114. Shimada, J.; Suzuki, F.; Nonaka, H.; Ishii, A. J. Med. Chem. 1992, 35, 924.

61

113. 114. 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. 127. 128. 129. 130. 131. 132. 133. 134. 135. 136. 137. 138. 139. 140. 141. 142. 143. 144. 145. 146. 147.

Shimada, J.; Koike, N.; Nonaka, H.; Shiozaki, S.; Yanagawa, K.; Kanda, T.; Kobayashi, H.; Ichimura, M.; Nakamura, J.; Kase, H.; Suzuki, F. Bioorg. Med. Chem. Lett. 1997, 7, 2349. Suzuki, F.; Shimada, J.; Koike, N.; Nakamura, J.; Shioazaki, S.; Ichikawa, S.; Ishii, A.; Nonaka, H. US Patent 5484920A, 1996. http://www.drugs.com/newdrugs/forest-laboratories-pierre-fabre-laboratories-announce-fdaapproval-fetzima-major-depressive-3859.html. Mago, R.; Mahajan, R.; Thase, M. E. Expert Rev. Clin. Pharmacol. 2014, 7, 137. Haddley, K. Drugs Future 2012, 37, 841. Hasin Deborah, S.; Goodwin Renee, D.; Stinson Frederick, S.; Grant Bridget, F. Arch. Gen. Psychiatry 2005, 62, 1097. Viazzo, P.; Alphand, V.; Furstoss, R. Tetrahedron Lett. 1996, 37, 4519. Arnold, L. M.; Palmer, R. H.; Gendreau, R. M.; Chen, W. Psychosomatics 2012, 53, 371. Bellato, E.; Marini, E.; Castoldi, F.; Barbasetti, N.; Mattei, L.; Bonasia, D. E.; Blonna, D. Pain Res. Treat. 2012, 2012, 426130. Auclair, A. L.; Martel, J. C.; Assie, M. B.; Bardin, L.; Heusler, P.; Cussac, D.; Marien, M.; Newman-Tancredi, A.; O'Connor, J. A.; Depoortere, R. Neuropharmacology 2013, 70, 338. Bonnaud, B.; Calmel, F.; Patoiseau, J. F.; N'guyen, N. T.; Cousse, H. J. Chromatogr. 1985, 318, 398. Jagtap, V. S.; Chache, R. B.; Ranbhan, K. J.; Zunjarrao, Y. K.; Sarjekar, P.; Mandal, A. K.; Pai, G. G. US Patent 20120184774A1, 2012. Pai, G. G.; Mandal, A. K.; Ranbhan, K. J.; Jagtap, V. S.; Patil, D. G.; Zunjarrao, Y. K. WO Patent 2012059933A1, 2012. Alliot, J.; Gravel, E.; Pillon, F.; Buisson, D. A.; Nicolas, M.; Doris, E. Chem. Commun. 2012, 48, 8111. Doyle, M. P.; Hu, W. Adv. Synth. Catal. 2001, 343, 299. Shuto, S.; Ono, S.; Hase, Y.; Kamiyama, N.; Matsuda, A. Tetrahedron Lett. 1996, 37, 641. Nicolas, M.; Hellier, P.; Diard, C.; Subra, L. WO Patent 2010086394A1, 2010. Panno, M. D.; Cefalù, A. B.; Averna, M. R. Clin. Lipidol. 2014, 9, 19. Biller, S. A.; Dickson, J. K.; Lawrence, R. M.; Magnin, D. R.; Poss, M. A.; Sulsky, R. B.; Tino, J. A. US Patent 5739135, 1998. Korolev, D. N.; Bumagin, N. A. Tetrahedron Lett. 2006, 47, 4225. Bolli, M.; Boss, C.; Fischli, W.; Clozel, M.; Weller, T. WO Patent 2002053557A1, 2002. http://www1.actelion.com/en/journalists/news-archive.page?newsId=1736781. http://www.fda.gov/newsevents/newsroom/pressannouncements/ucm371362.htm. Patel, T.; McKeage, K. Drugs 2014, 74, 127. Hong Irene, S.; Coe Holly, V.; Catanzaro Linda, M. Ann. Pharmacother. 2014, 48, 538. Raja, S. G. Curr. Opin. Invest. Drugs 2010, 11, 1066. Bolli, M. H.; Boss, C.; Binkert, C.; Buchmann, S.; Bur, D.; Hess, P.; Iglarz, M.; Meyer, S.; Rein, J.; Rey, M.; Treiber, A.; Clozel, M.; Fischli, W.; Weller, T. J. Med. Chem. 2012, 55, 7849. Dingemanse, J.; Sidharta, P. N.; Maddrey, W. C.; Rubin, L. J.; Mickail, H. Expert Opin. Drug Saf. 2014, 13, 391. Boss, C.; Fischli, W.; Weller, T.; Clozel, M.; Bolli, M. WO Patent 2006051502A2, 2006. Gibb, A.; Yang, L. P. H. Drugs 2013, 73, 1841. http://www.boehringeringelheim.com/news/news_releases/press_releases/2013/18_october_2013_olodaterol.html. Bouyssou, T.; Hoenke, C.; Rudolf, K.; Lustenberger, P.; Pestel, S.; Sieger, P.; Lotz, R.; Heine, C.; Buettner, F. H.; Schnapp, A.; Konetzki, I. Bioorg. Med. Chem. Lett. 2010, 20, 1410. Trunk, M. J. F.; Schiewe, J. US Patent 20050255050A1, 2005. Lustenberger, P.; Konetzki, I.; Sieger, P. US Patent 20090137578A1, 2009. Krueger, T.; Ries, U.; Schnaubelt, J.; Rall, W.; Leuter, Z. A.; Duran, A.; Soyka, R. US Patent 20110124859A1, 2011.

62

148. 149. 150. 151. 152. 153. 154. 155. 156. 157.

158. 159. 160. 161. 162. 163. 164. 165. 166. 167. 168. 169. 170. 171. 172. 173. 174. 175. 176. 177.

178.

Elkinson, S.; Yang, L. P. H. Drugs 2013, 73, 605. McCall, J. L.; De Gregorio, M. W. Expert Opin. Drug Metab. Toxicol. 2010, 6, 773. Soedervall, M.; Eloranta, M.; Kalapudas, A. US Patent 8293947B2, 2012. Tois, J.; Pihko, A. WO Patent 2014060640A1, 2014. Soedervall, M.; Eloranta, M.; Kalapudas, A. US Patent 20110015448A1, 2011. Soedervall, M.; Eloranta, M.; Kalapudas, A. WO Patent 2008099059A1, 2008. Tois, J.; Pihko, A.; Grumann, A. WO Patent 2014060639A1, 2014. Eklund, L.; Nilsson, J. WO Patent 2011089385A1, 2011. D'Amato, R. J.; Loughnan, M. S.; Flynn, E.; Folkman, J. Proc. Natl. Acad. Sci. U. S. A. 1994, 91, 4082. Richardson, P. G.; Siegel, D. S.; Vij, R.; Hofmeister, C. C.; Baz, R.; Jagannath, S.; Chen, C.; Lonial, S.; Jakubowiak, A.; Bahlis, N.; Song, K.; Belch, A.; Raje, N.; Shustik, C.; Lentzsch, S.; Lacy, M.; Mikhael, J.; Matous, J.; Vesole, D.; Chen, M.; Zaki, M. H.; Jacques, C.; Yu, Z.; Anderson, K. C. Blood 2014, 123, 3208. Muller, G. W.; Chen, R.; Huang, S.-Y.; Corral, L. G.; Wong, L. M.; Patterson, R. T.; Chen, Y.; Kaplan, G.; Stirling, D. I. Bioorg. Med. Chem. Lett. 1999, 9, 1625. Teo, S. K.; Chen, Y.; Muller, G. W.; Chen, R. S.; Thomas, S. D.; Stirling, D. I.; Chandula, R. S. Chirality 2003, 15, 348. Robarge, M. J.; Chen, R. S.-C.; Muller, G. W.; Man, H.-W. WO Patent 2002059106A1, 2002. Ge, C.; Muller, G. W.; Chen, R.; Saindane, M. T. US Patent 20070004920A1, 2007. Conole, D.; Scott, L. J. Drugs 2013, 73, 1967. Schermuly, R. T.; Janssen, W.; Weissmann, N.; Stasch, J.-P.; Grimminger, F.; Ghofrani, H. A. Expert Opin. Invest. Drugs 2011, 20, 567. http://www.fda.gov/newsevents/newsroom/pressannouncements/ucm370866.htm. Alonso-alija, C.; Bischoff, E.; Muenter, K.; Stasch, J.-P.; Stahl, E.; Weigand, S.; Feurer, A. US Patent 7173037B2, 2007. Bischoff, H.; Stasch, J.-P.; Weigand, S. US Patent 20070225299A1, 2007. Follmann, M.; Stasch, J.-P.; Redlich, G.; Ackerstaff, J.; Griebenow, N.; Knorr, A.; Wunder, F.; Li, V. M.-J.; Baerfacker, L.; Weigand, S. WO Patent 2012028647A1, 2012. Hirth-Dietrich, C.; Sandner, P.; Stasch, J.-P.; Knorr, A.; Von Degenfeld, G.; Hahn, M.; Follmann, M. WO Patent 2011147810A1, 2011. Li, J.; Yang, X.; Zhu, J.; Yang, M.; Wu, X. WO Patent 2013086935A1, 2013. Li, L.; Li, X.; Liu, Y.; Zheng, Z.; Li, S. Chin. J. Med. Chem. 2011, 21, 120. Mais, F.-J.; Rehse, J.; Joentgen, W.; Siegel, K. US Patent 20110130410A1, 2011. Jani, R. H.; Kansagra, K.; Jain, M. R.; Patel, H. Clin. Drug Investig. 2013, 33, 809. Jani, R. H.; Pai, V.; Jha, P.; Jariwala, G.; Mukhopadhyay, S.; Bhansali, A.; Joshi, S. Diabetes Technol. Ther. 2014, 16, 63. Lohray, B. B.; Lohray, V. B.; Barot, V. K.; Raval, S. K.; Raval, P. S.; Basu, S. WO Patent 2003009841A1, 2003. Talwani, R.; Heil, E. L.; Gilliam, B. L.; Temesgen, Z. Drugs Today (Barc) 2013, 49, 769. Davies, S. Drugs Future 2009, 34, 545. Raboisson, P.; de Kock, H.; Rosenquist, A.; Nilsson, M.; Salvador-Oden, L.; Lin, T.-I.; Roue, N.; Ivanov, V.; Wahling, H.; Wickstrom, K.; Hamelink, E.; Edlund, M.; Vrang, L.; Vendeville, S.; Van de Vreken, W.; McGowan, D.; Tahri, A.; Hu, L.; Boutton, C.; Lenz, O.; Delouvroy, F.; Pille, G.; Surleraux, D.; Wigerinck, P.; Samuelsson, B.; Simmen, K. Bioorg. Med. Chem. Lett. 2008, 18, 4853. Raboisson, P. J.-M. B.; De Kock, H. A.; Hu, L.; Vendeville, S. M. H.; Tahri, A.; Surleraux, D. L. N. G.; Simmen, K. A.; Nilsson, K. M.; Samuelsson, B. B.; Rosenquist, A. A. K.; Ivanov, V.; Pelcman, M.; Belfrage, A. K. G. L.; Johansson, P.-O. M. WO Patent 2007014926A1, 2007.

63

179.

180. 181. 182. 183. 184. 185. 186. 187. 188. 189. 190. 191. 192. 193. 194. 195. 196.

197. 198. 199. 200. 201. 202. 203. 204. 205. 206. 207. 208. 209. 210. 211.

Rosenquist, A.; Samuelsson, B.; Johansson, P.-O.; Cummings, M. D.; Lenz, O.; Raboisson, P.; Simmen, K.; Vendeville, S.; de Kock, H.; Nilsson, M.; Horvath, A.; Kalmeijer, R.; de la Rosa, G.; Beumont-Mauviel, M. J. Med. Chem. 2014, 57, 1673. Horvath, A.; Wuyts, S.; Depre, D. P. M.; Couck, W. L. J.; Cuypers, J. L. J.; Harutyunyan, S.; Binot, G. F. S. WO Patent 2013061285A1, 2013. Depre, D. P. M.; Ormerod, D. J.; Horvath, A. WO Patent 2013041655A1, 2013. Ormerod, D. J.; Depre, D. P. M.; Horvath, A. WO Patent 2011113859A1, 2011. Horvath, A.; Ormerod, D. J.; Depre, D. P. M.; Cerpentier, V. WO Patent 2010072742A1, 2010. Horvath, A.; Depre, D. P. M.; Ormerod, D. J. WO Patent 2008092955A1, 2008. Lou, S.; Cuniere, N.; Su, B. N.; Hobson, L. A. Org. Biomol. Chem. 2013, 11, 6796. Clavier, H.; Nolan, S. P. Chem. Eur. J. 2007, 13, 8029. Monsaert, S.; Drozdzak, R.; Dragutan, V.; Dragutan, I.; Verpoort, F. Eur. J. Inorg. Chem. 2008, 2008, 432. Keating, G. M. Drugs 2014, 74, 1127. Peifer, M.; Berger, R.; Shurtleff, V. W.; Conrad, J. C.; MacMillan, D. W. C. J. Am. Chem. Soc. 2014, 136, 5900. Ray, A. S.; Watkins, W. J.; Link, J. O.; Oldach, D. W.; Delaney, W. E. I. V. WO Patent 2013040492A2, 2013. Cho, A.; Wolckenhauer, S. A. WO Patent 2012012465A1, 2012. Ross, B. S.; Sofia, M. J.; Pamulapati, G. R.; Rachakonda, S.; Zhang, H.-R. WO Patent 2011123668A2, 2011. Ross, B. S.; Sofia, M. J.; Pamulapati, G. R.; Rachakonda, S.; Zhang, H.-R.; Chun, B.-K.; Wang, P. WO Patent 2010135569A1, 2010. Mayes, B. A.; Stewart, A. J.; Moussa, A. M. WO Patent 2013177219A1, 2013. Smith, D. B.; Beigelman, L.; Wang, G.; WELCH, M. H. WO Patent 2014100498A1, 2014. Sofia, M. J.; Bao, D.; Chang, W.; Du, J.; Nagarathnam, D.; Rachakonda, S.; Reddy, P. G.; Ross, B. S.; Wang, P.; Zhang, H.-R.; Bansal, S.; Espiritu, C.; Keilman, M.; Lam, A. M.; Steuer, H. M. M.; Niu, C.; Otto, M. J.; Furman, P. A. J. Med. Chem. 2010, 53, 7202. Ross, B. S.; Sofia, M. J.; Pamulapati, G. R.; Rachakonda, S.; Zhang, H.-R.; Chun, B.-K.; Wang, P. US Patent 20110251152A1, 2011. Ross, B.; Sofia, M. J.; Pamulapati, G. R.; Rachakonda, S.; Zhang, H.-R.; Chun, B.-K.; Wang, P. US Patent 8642756B2, 2014. Smith, D. B.; Deval, J.; Dyatkina, N.; Beigelman, L.; Wang, G. US Patent 20120071434A1, 2012. Yue, X.; Zhong, X.; Wang, Z. CN Patent 103848876A, 2014. Ji, M.; Liu, H.; Cai, J.; Li, R. CN Patent 103804446A, 2014. Wang, P.; Chun, B. K.; Rachakonda, S.; Du, J.; Khan, N.; Shi, J.; Stec, W.; Cleary, D.; Ross, B. S.; Sofia, M. J. J. Org. Chem. 2009, 74, 6819. Axt, S. D.; Sarma, K.; Vitale, J.; Zhu, J.; Ross, B.; Rachakonda, S.; Jin, Q.; Chun, B. K. WO Patent 2008045419A1, 2008. Ross, B. S.; Ganapati Reddy, P.; Zhang, H.-R.; Rachakonda, S.; Sofia, M. J. J. Org. Chem. 2011, 76, 8311. Javan, M. J.; Tehrani, Z. A.; Fattahi, A.; Hashemi, M. M. J. Phys. Org. Chem. 2012, 25, 1198. Hosoya, T.; Ohno, I.; Nomura, S.; Hisatome, I.; Uchida, S.; Fujimori, S.; Yamamoto, T.; Hara, S. Clin. Exp. Nephrol. 2014, Epub with DOI: 10.1007/s10157-014-0935-8. Shimo, T.; Moto, M.; Ashizawa, N.; Matsumoto, K.; Iwanaga, T.; Saito, K. Arch. Toxicol. 2014, 88, 1035. Huo, Z.; Kosugi, T.; Yamamoto, Y. Tetrahedron Lett. 2008, 49, 4369. Nakamura, H.; Uda, J.; Ono, A.; Sato, T. WO Patent 2005009991A1, 2005. http://www.fda.gov/drugs/informationondrugs/approveddrugs/ucm381451.htm. Wright, C. J.; McCormack, P. L. Drugs 2013, 73, 1245.

64

212.

213.

214. 215. 216.

217. 218. 219. 220. 221. 222. 223. 224.

225. 226. 227. 228. 229. 230. 231. 232.

Abe, H.; Kikuchi, S.; Hayakawa, K.; Iida, T.; Nagahashi, N.; Maeda, K.; Sakamoto, J.; Matsumoto, N.; Miura, T.; Matsumura, K.; Seki, N.; Inaba, T.; Kawasaki, H.; Yamaguchi, T.; Kakefuda, R.; Nanayama, T.; Kurachi, H.; Hori, Y.; Yoshida, T.; Kakegawa, J.; Watanabe, Y.; Gilmartin, A. G.; Richter, M. C.; Moss, K. G.; Laquerre, S. G. ACS Med. Chem. Lett. 2011, 2, 320. Sakai, T.; Kawasaki, H.; Abe, H.; Hayakawa, K.; Iida, T.; Kikuchi, S.; Yamaguchi, T.; Nanayama, T.; Kurachi, H.; Tamaru, M.; Hori, Y.; Takahashi, M.; Yoshida, T. WO Patent 2005121142A1, 2005. Lambert, J. M.; Chari, R. V. J. Med. Chem. 2014, 57, 6949. Kuemler, I.; Mortensen, C. E.; Nielsen, D. L. Drugs Future 2011, 36, 825. Widdison, W. C.; Wilhelm, S. D.; Cavanagh, E. E.; Whiteman, K. R.; Leece, B. A.; Kovtun, Y.; Goldmacher, V. S.; Xie, H.; Steeves, R. M.; Lutz, R. J.; Zhao, R.; Wang, L.; Blattler, W. A.; Chari, R. V. J. Med. Chem. 2006, 49, 4392. Chari, R. V. J.; Widdison, W. C. US Patent 6333410B1, 2001. Chari, R. V. J.; Widdison, W. C. WO Patent 2007056550A2, 2007. Ebens, A. J., Jr.; Jacobson, F. S.; Polakis, P.; Schwall, R. H.; Sliwkowski, M. X.; Spencer, S. D. WO Patent 2005117986A2, 2005. Chari, R. V.; Martell, B. A.; Gross, J. L.; Cook, S. B.; Shah, S. A.; Blattler, W. A.; McKenzie, S. J.; Goldmacher, V. S. Cancer Res. 1992, 52, 127. Bang-Andersen, B.; Ruhland, T.; Jorgensen, M.; Smith, G.; Frederiksen, K.; Jensen, K. G.; Zhong, H.; Nielsen, S. M.; Hogg, S.; Mork, A.; Stensbol, T. B. J. Med. Chem. 2011, 54, 3206. Blier, P.; Ward, N. M. Biol. Psychiatry 2003, 53, 193. Ye, J. H.; Ponnudurai, R.; Schaefer, R. CNS Drug Rev. 2001, 7, 199. Bang-Andersen, B.; Faldt, A.; Moerk, A.; Lopez De Diego, H.; Holm, R.; Stensboel, T. B.; Ringgaard, L. M.; Mealy, M. J.; Rock, M. H.; Brodersen, J.; Joergensen, M.; Moore, N. WO Patent 2007144005A1, 2007. Christensen, K. L. WO Patent 2013102573A1, 2013. Moore, N.; Dragheim, M.; Batra, A. WO Patent 2009062517A1, 2009. Moore, N.; Stensboel, T. B. WO Patent 2008113359A2, 2008. Ruhland, T.; Smith, G. P.; Bang-Andersen, B.; Pueschl, A.; Moltzen, E. K.; Andersen, K. WO Patent 2003029232A1, 2003. Xu, X. CN Patent 103788020A, 2014. Schopfer, U.; Schlapbach, A. Tetrahedron 2001, 57, 3069. Louie, J.; Hartwig, J. F. Tetrahedron Lett. 1995, 36, 3609. Guram, A. S.; Rennels, R. A.; Buchwald, S. L. Angew. Chem. Int. Ed. Engl. 1995, 34, 1348.

65

Synthetic Approaches to the 2013 New Drugs Hong X. Ding,8 Carolyn A. Leverett,9 Robert E. Kyne, Jr.,10 Kevin K.-C. Liu, 11 Sarah J. Fink,12 Andrew C. Flick,13 Christopher J. O’Donnell14*

8

Email: [email protected]; tel: 8610-8282-6195

9

Email: [email protected]; tel: 860-441-3936

10

Email: [email protected]; tel: 860-441-1510

11

Email: [email protected]; tel: 8621-2080-5590

12

Email: [email protected]; tel: 86-21-3175-2858

6

14

Email: [email protected]; tel: 860-715-0228 Email: [email protected]; tel: 860-715-4118

66

Synthetic approaches to the 2013 new drugs.

New drugs introduced to the market every year represent privileged structures for particular biological targets. These new chemical entities (NCEs) pr...
1MB Sizes 17 Downloads 24 Views