Download as pdf or txt
Download as pdf or txt
You are on page 1of 28

Bioorganic & Medicinal Chemistry 22 (2014) 2005–2032

Contents lists available at ScienceDirect

Bioorganic & Medicinal Chemistry


journal homepage: www.elsevier.com/locate/bmc

Review

Synthetic approaches to the 2012 new drugs


Hong X. Ding a, , Carolyn A. Leverett b,à, Robert E. Kyne Jr. b,§, Kevin K.-C. Liu c,–, Subas M. Sakya d,k,
Andrew C. Flick b,  , Christopher J. O’Donnell b,⇑
a
PharmaPhase Co., Ltd, Beijing 100193, China
b
Pfizer Worldwide Research and Development, Groton Laboratories, 445 Eastern Point Road, Groton, CT 06340, United States
c
Lilly China Research and Development Center, Shanghai 201203, China
d
BioDuro Co., Ltd, Shanghai 200131, China

a r t i c l e i n f o a b s t r a c t

Article history: New drugs introduced to the market every year represent a privileged structure for a particular biological
Received 27 December 2013 target. These new chemical entities (NCEs) provide insights into molecular recognition and also serve as
Revised 11 February 2014 leads for designing future new drugs. This review covers the synthesis of twenty-six NCEs that were
Accepted 13 February 2014
launched or approved worldwide in 2012 and two additional drugs which were launched at the end of
Available online 25 February 2014
2011.
Ó 2014 Elsevier Ltd. All rights reserved.
Keywords:
Synthesis
New drug molecules
New chemical entities
Medicine
Therapeutic agents

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2006
2. Aclidinium bromide (Tudorza PressairÒ, Eklira GenuairÒ, Bretaris GenuaiÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2006
3. Allisartan isoproxil . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2006
4. Anagliptin (BeskoaÒ, SuinyÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2006
5. Axitinib (InlytaÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2009
6. Azilsartan (AzilvaÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2010
7. Bedaquiline fumarate (SirturoÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2012

Abbreviations: 1,2-DAP, 1,2-diaminopropane; 1,2-DCE, 1,2-dichloroethane; Ac, acetyl; aq, aqueous; B2(pin)2, bis(pinacolato)diboron; BINAP, 2,20 -bis(diphenylphosphino)-
1,10 -binaphthyl; BOP, benzotriazol-1-yloxytris(dimethylamino)phosphonium hexafluorophospate; CDI, N,N0 -carbonyldiimidazole; DAP, diaminopropane; DBU, 1,5-diazabi-
cycolo[4.3.0]non-5-ene; DCC, 1,3-dicyclohexylcarbodiimide; DCM, dichloromethane; DIC, 1,3-diisopropylcarbodiimide; DIEA/DIPEA, diisopropylethylamine; DMA, dimeth-
ylacetamide; DMAP, 4-dimethylaminopyridine; DME, dimethoxyethane; DMF, N,N-dimethylformamide; DMSO, dimethyl sulfoxide; DPPA, diphenylphosphoryl azide; EDC,
N-(3-dimethylaminopropal)-N0 -ethylcarbodiimide; Fmoc, 9-fluorenylmethoxycarbonyl; HBTU, 2-(1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate;
HOBT, 1-hydroxybenzotriazole hydrate; IPAc, isopropyl acetate; LAH, lithium aluminum hydride; LHMDS, lithium bis(trimethylsilyl)amide; LDA, lithium diisopropylamide;
MEK, methyl ethyl ketone; MIBK, 4-methyl-2-pentanone; NBS, N-bromosuccinimide; NMM, N-methylmorpholine; NMP, N-methyl-2-pyrrolidone; Pd2(dba)3, tris(dibenzyl-
ideneacetone)dipalladium(0); Pd(dppf)Cl2, [1,10 -bis(diphenylphosphino)ferrocene]dichloropalladium(II); Pd(PPh3)4, tetrakis(triphenylphosphine)palladium(0); pin, pinacol;
Py, pyridine; RT, room temperature; STAB-H, sodium triacetoxyborohydride; TBAF, t-butyl ammonium fluoride; TFA, trifluoroacetic acid; THF, tetrahydrofuran; TMSCl,
trimethylsilyl chloride; XantPhos, 9,9-dimethyl-4,5-bis(diphenylphosphino)xanthene.
⇑ Corresponding author. Tel.: +1 860 715 4118.
E-mail addresses: Hongxia.ding@gmail.com (H.X. Ding), carolyn.a.leverett@pfizer.com (C.A. Leverett), robert.kynejr@pfizer.com (R.E. Kyne), Liu_kang_zhi_kevin@lilly.com
(K.K.-C. Liu), subas.sakya@bioduro.com (S.M. Sakya), andrew.flick@pfizer.com (A.C. Flick), christopher.j.odonnell@pfizer.com (C.J. O’Donnell).
 
Tel.: +86 10 8484 8357.
à
Tel.: +1 860 441 3936.
§
Tel.: +1 860 441 1510.

Tel.: +86 21 2080 5590.
k
Tel.: +86 38139788x3904.
  
Tel.: +1 860 715 0228.

http://dx.doi.org/10.1016/j.bmc.2014.02.017
0968-0896/Ó 2014 Elsevier Ltd. All rights reserved.
2006 H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032

8. Bosutinib hydrate (BosulifÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2013


9. Cabozantinib (S)-malate (CometriqÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2015
10. Carfilzomib (KyprolisÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2015
11. Dapagliflozin propanediol hydrate (ForxigaÒ, EmplicitiÒ, EdistrideÒ, AppebbÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2016
12. Enzalutamide (XtandiÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2016
13. Iguratimod (CareramÒ, IremodÒ). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2017
14. Imrecoxib (HengyangÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2019
15. Ingenol mebutate (PicatoÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2020
16. Ivacaftor (KalydecoÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2021
17. Lorcaserin hydrochloride hydrate (BelviqÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2021
18. Omacetaxine mepesuccinate (SynriboÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2022
19. Pasireotide (SigniforÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2022
20. Perampanel hydrate (FycompaÒ). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2024
21. Pixantrone dimaleate (PixuvriÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2024
22. Ponatinib hydrochloride (IclusigÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2025
23. Radotinib hydrochloride (SupectÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2026
24. Regorafenib hydrate (StivargaÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2026
25. Tafamidis meglumine (VyndaqelÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2027
26. Teneligliptin hydrobromide hydrate (TeneliaÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2028
27. Teriflunomide (AubagioÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2028
28. Tofacitinib citrate (XeljanzÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2029
29. Vismodegib (ErivedgeÒ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2029
References and notes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2029

1. Introduction Scheme 1.16 Dimethyl oxalate (1) was initially treated with two
equivalents of Grignard 2 to give bis-thiophenoate 3 in 36% yield.
‘The most fruitful basis for the discovery of a new drug is to start Subsequent transesterification with (R)-quinuclidinol (4) gave rise
with an old drug.’ – Sir James Whyte Black, winner of the 1988 No- to the quinuclidine-containing ester 5 in 50% yield. Aclidinium bro-
bel Prize in medicine.1 mide (I) could be accessed by two different methods involving bro-
This annual review was inaugurated eleven years ago2–11 and moalkyl phenyl ether 6: an excess of bromide in the presence of an
presents synthetic methods for molecular entities that were acetonitrile/chloroform mixture gave the drug in 89% isolated
launched in various countries during the past year. Given that yield, or with fewer equivalents of electrophile (1.25 equiv) during
drugs tend to have structural homology across similar biological exposure to refluxing acetophenone, has reportedly delivered (I)
targets, it is widely believed that the knowledge of new chemical quantitatively on multi-gram scale.17 From commercial 2,18 the
entities and their syntheses will greatly enhance the ability to de- multi-gram synthesis of Aclidinium bromide (I) was completed
sign new drugs in shorter periods of time. The pharmaceutical in 17.8% over three steps.
industry enjoyed a banner year in 2012, with a total of 36 new
products, including new chemical entities, biological drugs and 3. Allisartan isoproxil
diagnostic agents having reached the worldwide market for the
first time. Although an additional 22 new products were approved Allisartan isoproxil, a member of a new class of selective angio-
for the first time in 2012, these were not launched before year tensin II-1 receptor antagonists, was approved by the Chinese Food
end,12 and therefore this review focuses on the syntheses of 26 and Drug Administration (CFDA) for the treatment of hypertension
drugs that were launched or approved in 2012 and two additional in July 2012.19 At time of publication, there is no trade name asso-
drugs that was launched at the end of 2011 (Fig. 1). New indica- ciated with this drug. Allisartan was discovered and developed by
tions for previously launched medications, new combinations, the Chinese biomedical company Allist Pharmaceuticals. Allisartan
new formulations of existing drugs, and drugs synthesized purely isoproxil is a prodrug which is readily hydrolyzed to active
via bio-processes or peptide synthesizers have been excluded from metabolite EXP3174, which is also the active metabolite of losartan
this review. Although the scale of the synthetic routes were not (des-triphenylmethyl-9, Scheme 2).20 Although several synthetic
explicitly disclosed in most cases, this review covers, perceptibly, routes have been reported within two patents,21,22 the most likely
the most scalable routes based on published or patent literature. scalable process route is described in Scheme 2. Commercial
Drugs are covered in alphabetical order by the drug’s generic name. 2-butyl-4-chloro-5-(hydroxymethyl)-imidazole (7) was alkylated
with N-triphenylmethyl-5-(40 -bromomethylbiphenyl-2-yl)tetra-
2. Aclidinium bromide (Tudorza PressairÒ, Eklira GenuairÒ, zole (8) under basic conditions in warm DMF, providing alcohol 9
Bretaris GenuaiÒ) in 90% yield. This alcohol was then oxidized to the corresponding
carboxylic acid 10 with KMnO4 in 88% yield. Etherification of
Aclidinium bromide was approved by the U.S. Food and Drug acid 10 with isopropyl chloromethyl carbonate (11) followed by
Administration (FDA) in July 2012 for the treatment of chronic de-tritylation of the tetrazole group under acidic conditions gave
obstructive pulmonary disease (COPD).13 Marketed by Forest Phar- allisartan isoproxil (II) in 69% yield.22
maceuticals, aclidinium bromide selectively binds to five human
muscarinic receptors (M1–M5), and posesses a subnanomolar bind- 4. Anagliptin (BeskoaÒ, SuinyÒ)
ing affinity for these particular targets. Administered by inhalation,
this medicine has demonstrated favorable onset and duration of Anagliptin, which is marketed as Beskoa or Suiny, is a dipeptidyl
action, and its safety profile is an improvement over competitor peptidase-IV (DPP-4) inhibitor which was approved in September
therapies.14 While no manufacturing route has been disclosed to 2012 and launched in November 2012 in Japan for the treatment
date,15 the most scalable published synthesis is described in of Type II diabetes. The drug was co-developed by three Japanese
H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032 2007

Cl
N O
O
O
S N O O O CN
H
OH O N
O N N N
S H
N+
O N
O N N
Br -
N
N NH
I Aclidinium bromide II Allisartan isoproxil III Anagliptin

N
O NH
N CO2 H
H O
S N H OH
N O O Ph
N HO2 C
HO N NH
O O
H3 C
N
N
Br

IV Axitinib V Azilsartan VI Bedaquiline fumarate

Cl Cl H H
N N

HN O O O
O F
O CN
O O
N O N HO
OH
O N
N O OH
H 2O

VII Bosutinib hydrate VIII Cabozantinib (S)-malate

Cl
O O O O
H H HO
N N OH
N N N
H H HO OH OH
O O O O
OH H 2O
O

IX Carfilzomib X Dapagliflozin propanediol hydrate

F O O
H
N O S
F3C N S O
S O
H O O
N
NC N O N H
N H
O
O
O

XI Enzalutamide XII Iguratimod XIII Imrecoxib

OH
O
O O
H Cl
N NH HCl
O H
O HO 1/2 H 2O
N
HO H
HO

XIV Ingenol mebutate XV Ivacaftor XVI Lorcaserin hydrochloride hydrate

Figure 1. Structures of 28 NCEs marketed/approved in 2012.


2008 H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032

OBn

Ph
O H
N H2 N N
( )3 HN
O
H O
O O N
O O
HN O O O
O O
O O HN
OH NH
N
O H
Ph H2 N
H N

HO

XVII Omacetaxine mepesuccinate XVIII Pasireotide

NH2
O HN
N N
COOH
O
3/4 H2O N 2
CN COOH
O HN
NH2

XIX Perampanel hydrate XX Pixantrone dimaleate

N
N
N
HCl N N
H
N N N N
H N N
N H
N O
N
O 2 HCl CF3
CF3

XXI Ponatinib hydrochloride XXII Radotinib dihydrochloride

CF3 O Cl O
Cl O O
O N OH
H
N
N N N OH OH
H H Cl H
F H 2O N
HO
OH OH

XXIII Regorafenib hydrate XXIV Tafamidis meglumine

NH O

N N
N CF3
OH O
S
N N 2.5 HBr N
x H 2O H
CN

XXV Teneligliptin hydrobromide hydrate XXVI Teriflunomide

N
N
N CN Cl
O Cl
O OH
N
HOOC COOH N
N N H
H HOOC SO 2Me
XXVII Tofacitinib citrate XXVIII Vismodegib

Fig. 1 (continued)
H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032 2009

S
OH
O MgBr S
2 N 4
MeO OH
OMe MeO S
Et2 O, −30 °C NaH, toluene, ↑↓
O
36% O 50%

1 3

S
S
Br OPh 6 OH
OH O
O S CH3 CN : CHCl3 (2:3), RT N+
O S
N O 89%
O Br -

5 I Aclidinium bromide

Br OPh , Acetophenone, ↑↓
99%

Scheme 1. Synthesis of aclidinium bromide (I).

Cl
Br N OH

Cl N
N OH K2 CO3 , DMF, 90 °C
+
N N 90%
H N
N
N N
CPh3 N N
N CPh3
7 8 9

Cl O
OH Cl O
N
N O O
N O 1. K2 CO 3, DME, RT
1% KMnO 4 2. 11, 50 °C N O
0 °C to 50 °C, 88% 3. 4 N HCl, dioxane, RT
69%
N
N N
N Cl O O
N CPh3 N
NH
N
O

10 11 II Allisartan isoproxil

Scheme 2. Synthesis of allisartan isoproxil (II).

companies; Kowa, Sanwa Kagaku and JW pharmaceutical. Anaglip- 5. Axitinib (InlytaÒ)


tin, which is more selective against several recombinant human
proteases by comparison to sitagliptin and vildagliptin,23 has more Sold under the brand name InlytaÒ by Pfizer, Inc., axitinib was
than 10,000-fold selectivity over the structurally homologous DPP- approved by the FDA in January 2012 for the treatment of ad-
8 and DPP-9 enzymes. vanced renal cell carcinoma (RCC), specifically after the failure of
The most likely process-scale synthesis is depicted in Scheme 3.24 other systemic treatments.25 Axitinib slows cancer cell prolifera-
Commercially available (S)-1-(2-chloroacetyl)-pyrrolidine-2-carboni- tion by inhibition of the vascular endothelial growth factor
trile (12) was alkylated with t-butyl (2-amino-2-methyl-1-propyl) (VEGF)/VEGF receptor tyrosine (RTK) signaling pathway. In partic-
carbamate (13), giving rise to (S)-t-butyl (2-((2-(2-cyanopyrrolidin-1- ular, axitinib is a potent inhibitor of VEGF/RTK 1–3, which selec-
yl)-2-oxoethyl)amino)-2-methylpropyl)carbamate (14). This Boc-pro- tively slows angiogenesis, vascular permeability, and blood flow
tected system was subsequently treated with strong acid to give the in solid tumors.26,27 While numerous patents and papers have been
ethylene diamine derivative 15 in 96% yield. Activation of 15 with disclosed on the synthesis of axitinib,28–37 a recently published
CDI followed by coupling with commercially available 2-methylpy- manuscript details the development of the manufacturing route,
razolo[1,5-a] pyrimidine-6-carboxylic acid (16) gave anagliptin (III) and this route is depicted in Scheme 4.38 The synthesis began with
in 90% yield. Migita coupling of commercial iodide 17 with thiophenol 18.
2010 H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032

O CN O O CN
13, NaI, K2CO3 , acetone H 4 N HCl/Dioxane
Cl N
N O N N
0 °C to RT, 91% H 0 °C to RT, 96%

12 14

O O CN
H
O CN N
H 1. 16, CDI, THF N N N
N H
H 2N N
2. Et 3N, THF, 0 °C to RT N
90% N
15 III Anagliptin

O
O N OH
NH 2
O N N
H
N

13 16

Scheme 3. Synthesis of anagliptin (III).

H O NH O NH
I N Pd 2(dba)3 , XantPhos
H
N SH S N
NaHCO3, NMP, 50 °C
N

17 18 19

O NH
1. Ac 2O, DIPEA, NMP, 60 °C
H
O NH 2. Pd(OAc)2 , 2-vinylpyridine S N
I2, NMP, aq. KOH H XantPhos, 90 °C N
S N
85-90% f or 2 steps N 3. 1,2-DAP, THF, polishing filter
4. NMP, THF, 62% for 4 steps
I N

20 IV Axitinib

Scheme 4. Synthesis of axitinib (IV).

Interestingly, this transformation’s efficiency relied upon attention The most likely process-scale synthetic route mimics that
to the number of equivalents of base and an inert atmosphere in which is disclosed in Takeda’s patents, and this is described in
the reaction vessel, conditions which minimized catalyst poisoning Scheme 541,42. Commercially available benzoic acid 21 was acti-
during the reaction. Without isolation, indazole 19 was iodinated vated as the corresponding acyl azide and underwent a Curtius
to afford diarylthioether 20 in 85–90% yield over the two steps. rearrangement to give carbamate 22 in 57% yield (three steps from
Protection of the indazole within 20 as its acetamide preceeded a compound 21). The resulting aniline 22 was alkylated with com-
Heck reaction with 2-vinylpyridine, and then subsequent removal mercial 4-(bromomethyl)-2’-cyanobiphenyl (23) to give benzyl-
of the indazole protection followed by a series of recrystallizations amine 24 in 85% yield. Nitroamine 24 was then exposed to
yielded axitinib (IV) in a combined 62% yield over the final 4 steps. mildly acidic conditions to affect Boc-removal prior to reduction
via ferric chloride hydrate in the presence of hydrazine hydrate.
6. Azilsartan (AzilvaÒ) The resulting diamine 25 arose in 64% yield across the two-step se-
quence. Interestingly, it was found that metal catalysts under con-
Azilsartan is an orally active angiotensin II blocker which was ventional hydrogenation conditions caused partial debenzylation,
approved and launched in Japan for the treatment of arterial which led the authors to arrive at the hydrazine/ferric chloride
hypertension in May 2012.39 Azilsartan, which is marketed under conditions. Next, benzimidazole formation was achieved upon
the trade name AzilvaÒ, was discovered and developed by Take- treatment of diamine 25 with ethyl orthocarbonate in acetic acid.
da—the same firm which had developed and launched a prodrug The resulting ethoxylbenzimidazole 26 was procured in 86% yield,
of azilsartan (azilsartan kamedoxomil, EdarbiÒ) in 2010. Azilsartan and this benzonitrile was further reacted with hydroxylamine
exhibits higher potency and slower off-rate kinetics for type 1 hydrochloride and sodium methoxide to provide amidoxime 27
angiotensin II receptors, which contributes to azilsartan’s compar- in 90% as a white powder. Next, activation with ethyl chlorocar-
atively improved blood pressure lowering effect.40 bonate gave 28 followed by heating in refluxing xylene to give
H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032 2011

NO 2 NO2
1. SOCl2, DMF, toluene, ↑↓
CO 2H 2. NaN 3, H2 O, 0 °C NHBoc

O O 3. t-BuOH, ↑↓, 57% for 3 steps O O

21 22

Br CN NO2 NH2

Boc 1. 1 N HCl
N NH
23 EtOAc, RT, 77%
K2 CO3, CH 3 CN, ↑↓, 85% O O CN 2. FeCl3•6 H2 O O O CN
0 °C, THF/MeOH
↑↓; NH 2NH2 •H2 O
↑↓, 64%
24 25

N N

N O N O OH
(EtO)4 C, AcOH O NHOH•HCl, NaOMe/MeOH O
O N NH2
CN O
80 °C, 86% DMSO, 90 °C, 90%

26 27

N O

N O O OEt
ClCO2 Et, CH 2Cl2, THF O N NH2
O
Et3N, RT

28

N N
O O
N O O N O O
xylenes, ↑↓ O 2 N LiOH, MeOH, ↑↓ HO
N NH N NH
O O
23% for 2 steps 84%

29 V Azilsartan

Scheme 5. Synthesis of azilsartan (V).

N N
O O
N O OH N O O
O NaOMe, O O O
N NH2 N NH
O O
DMSO, RT, 85-90%

30 29

N
O
N O O
aq. NaOH HO N NH
O
50 °C, 88-90%

V Azilsartan

Scheme 6. Improved synthesis of azilsartan (V).


2012 H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032

oxadiazolone 29 in 23% yield from hydroxyamidine 27. Finally, hydrolysis converted this methyl ester 29 to azilsartan (V) in
ester 29 was saponified with 2 N LiOH in methanol to give 88–90% yield.
azilsartan (V) in 84% yield.
An improved scalable route (Scheme 6) to azilsartan was re- 7. Bedaquiline fumarate (SirturoÒ)
ported and features reproducibly better yields.43 Hydroxyamidine
30 was treated with dimethyl carbonate and sodium methoxide, Bedaquiline fumarate is a diarylquinone drug developed by
which triggered they key cyclization along with concomitant Janssen Pharmaceutical which is marketed under the trade name
transesterification to deliver 29. Milder aqueous sodium hydroxide SirturoÒ.44 The drug, which was approved in 2012 for the

• HCl
O N O N O
aq. NaOH, 60 °C OH
P
98% O
O

31 32
34

N N
CO2 H
Ph OH H OH
Ph • Ph
Br HO2 C
O O
CH3 1. LDA, THF, -70 °C H3 C H 3C
N O 2. 32, THF, -70 °C fumaric acid, i-PrOH
N N
3. AcOH, THF, -10 °C 50-80 °C
4. 34, DMSO, ↑↓ Br 82% Br
5. 10% aq. K2CO3
33 35 VI Bedaquiline fumarate
toluene, 85 °C
39% for five steps

Scheme 7. Synthesis of bedaquiline fumarate (VI).

H 3CO Cl Br 37 H3 CO N NH
39
i-PrOH, 1 N NaOH
HO NO2 70 °C, 82% Cl O NO2 NaI, DME, ↑↓, 77%

36 38

N N
H2 , Pd/C 42, HC(OEt) 3, i-PrOH
O O
N N
i-PrOH, RT ↑↓, 93% for 2 steps
O NO2 O NH 2

40 41
Cl Cl Cl Cl

O NH HN O
O NC O CN
O POCl3, sulfolane

N O N 70 °C to 105 °C N O N
H
N 75-82%, >99% purity N
H 2O
43 VII Bosutinib monohydrate

O
45 Cl Cl
Cl Cl NC O
OH
NC
DIC, THF, ↑↓, 88% N O
H2 N O H

44 42

Scheme 8. Synthesis of bosutinib hydrate (VII).


H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032 2013

treatment of multidrug-resistant tuberculosis (MDR-TB), was treatment with fumaric acid in the presence of isopropanol, and
developed in partnership with Johnson & Johnson and represents this salt formation delivered bedaquiline fumarate (VI) in 82%
the first new tuberculosis therapy approved in over four decades.44 yield.49
Bedaquiline is the first member of a new class of diarylquinoline
compounds whose mechanism of action inhibits Mycobaterium 8. Bosutinib hydrate (BosulifÒ)
tuberculosis ATP synthase which deprives bacterium of energy.44
Of the relatively few synthetic approaches to bedaquiline (or its BosulifÒ (Bosutinib hydrate), also known as (SKI-606), is a novel
fumarate salt) that have been reported,45–47 the most likely pro- 4-phenylamino-3-quinolinecarbonitrile kinase inhibitor approved
cess-scale route is that described by Porstmann and co-workers for treatment of adults with chronic, accelerated, or blast phase
from Janssen Pharmaceutical, and this route is outlined in Philadelphia chromosome-positive chronic myeloid leukemia
Scheme 7.48 The synthesis was initiated by first treating commer- (Ph+CML).50 Bosutinib is an orally-dosed, dual Src/Abl kinase inhib-
cially available dimethylaminoketone 31 with sodium hydroxide itor51,52 which provides an alternative treatment to patients exhib-
to provide naphthylone 32 in nearly quantitative yield. Treatment iting immunity to imatinib and other kinase inhibitors utilized for
of commercially available quinoline 33 with LDA and subsequent this treatment.53,54 In contrast to competitor tyrosine inhibitors,
trapping with naphthylone 32 provided a mixture of diastereo- bosutinib inhibits autophosphorylation of both Srs and Abl kinases,
mers, whereby the major diastereomer obtained from this reaction leading to decreased cell growth and apoptosis.51 Bosutinib was
corresponded to the bedaquiline geometry. The minor diastereo- originally developed by Wyeth and continues to be marketed by
mer was resolved through multiple recrystallizations and seeding Pfizer after the merger of Wyeth and Pfizer in 2009.55
techniques.48 This racemate of the major diastereomer subse- Several synthetic routes to bosutinib have been reported,
quently underwent a chiral resolution upon treatment with BINAP including synthetic work for scale up and processing to obtain pure
derivative 34 in refluxing DMSO. Cooling and subjection to salt forms of bosutinib for pharmaceutical applications.56–59 The
aqueous base in warm toluene furnished bedaquiline 35, bearing current manufacturing route begins with reaction of 2-methoxy-
the requisite (R,S)-configuration of the two vicinal chiral centers 5-nitrophenol (36) and 1-bromo-3-chloropropane (37) to provide
corresponding to that of the drug. The overall yield of the aryl chloroether 38 in 82% yield.58 Reaction of 38 with N-methylpi-
conversion of 33 to enantiopure 35 was 39%. Aminoquinolinol 35 perazine (39) and NaI in refluxing DME provided the functionalized
was then prepared as the corresponding fumarate salt upon aryl-nitro-piperazine 40 (77% yield), which was converted directly

NH 2
OH Cl H 2N OH
O POCl3 , CH 3 CN O O
77 °C, 70% t-BuONa, DMA, 105 °C O
O N O N 72%
O N
46 47 48

H H
N N

51, K2CO3 , THF, H 2O, RT O O


O F
96% O

O N
52

H H
N N

(S)-malic acid O O
O F
MEK, H 2O, 55 °C to RT O O
HO
75% OH
O N O OH

VIII Cabozantinib (S)-malate

O O F F
1. SOCl2, Et 3 N, THF, 5 °C O O oxalyl chloride O O
HO OH HO N DMF, THF, RT Cl N
F H H
2. , THF, 10 °C
49 H 2N 50 51
70% for two steps

Scheme 9. Synthesis of cabozantinib (S)-malate (VIII).


2014 H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032

to aniline 41 under hydrogenolysis conditions. Aniline 41 was then (93% over 2 steps).58,59 Finally, conversion of 43 to bosutinib was
reacted with triethyl orthoformate and aryl cyanoamide 42, which facilitated by a POCl3-promoted cyclization in the presence of sul-
was generated in one step from 2,4-dichloro-5-methoxy-aniline folane. As shown in Scheme 8, employment of carefully optimized
(44), 1,3-diisopropylcabodiimide (DIC), and cyanoacetic acid (45) conditions for the isolation of bosutinib hydrate (VII) provided
under refluxing conditions, to yield advanced intermediate 43 material in 75–82% yields and >99% purity.59

BOP, HOBT, DMF H


N CO2 Me
BocHN
DIPEA, 0 °C, 93%
BocHN CO2H O
H2 N CO2 Me

53 54 55

O
H
BocHN N CO2 Me
1. TFA, DCM, 0 °C N
H
2. Boc-Homophe-OH, BOP O
HOBT, DMF, DIPEA, 0 °C
85% for 2 steps

56

Cl O O
H H 1. KI, THF H H
N N CO2 Me N N CO2 H
1. TFA, DCM N morpholine N N
H H
O O O O O
2. ClCH2C(O)Cl 2. LiOH, MeOH
DMF, DIPEA, 0 °C 0 °C to 5 °C
67% for 2 steps 87% for 2 steps

57 58

O O O
H H
N N
1. 59, HBTU, HOBT, DMF, DIPEA, 0 °C N N N
H H
O O O O
2. Recrystallization from MeOH/H 2 O
75% for 2 steps

O
IX Carfilzomib
TFA H2 N
O
59

Scheme 10. Synthesis of carfilzomib (IX).

i-BuOC(O)Cl, DCM OMe MgBr 61


N
MeONHMe•HCl, NMM BocHN THF, 5 °C, 81% BocHN
BocHN CO2H
Et3N, −20 °C, 94% O O

54 60 62

1. Ca(OCl)2 , NMP, H2 O
−10 °C to −5 °C, 41% O
2. TFA, CH 2 Cl2 , 0 °C, 92% TFA H 2 N
O
59

Scheme 11. Synthesis of fragment 59 of carfilzomib (IX).


H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032 2015

9. Cabozantinib (S)-malate (CometriqÒ) 10. Carfilzomib (KyprolisÒ)

Cabozantinib (S)-malate (CometriqÒ), which was discovered Carfilzomib is an irreversible inhibitor of the chymotrypsin-like
and developed by Exelixis, gained approval by the U.S. FDA in protease in the proteasome and was approved in the U.S. for the
November 2012. The drug’s indication is for the treatment of med- treatment of multiple myeloma.67,68 Carfilzomib was discovered
ullary thyroid cancer (MTC), and is the second drug for this disease by Proteolix, which was later acquired by Onyx Therapeutics,
after AstraZeneca’s vandetanib (CaprelsaÒ). The drug was success- who completed the development of this drug. Carfilzomib is also
fully launched on January 24, 2013.60,61 Cabozantinib inhibits mul- undergoing clinical evaluation for additional oncology indications
tiple receptor tyrosine kinases including RET, MET, VEGFR-1, -2 such as relapsed solid tumors, lymphoma, prolymphocytic leuke-
and -3, KIT, TRKB, FLT-3, AXL, and TIE-2.62 It is currently also mia, acute myeloid leukemia and acute lymphocytic leukemia. Car-
undergoing clinical trials for the treatment of prostate, ovarian, filzomib is an analog of the natural product epoxomicin which was
brain, melanoma, breast, non-small cell lung, pancreatic, hepato- first synthesized in the laboratories of Professor Crews at Yale Uni-
cellular and kidney cancers. Of the three syntheses of cabozantinib versity.69 Subsequent development of the SAR led to the discovery
reported,63–66 the kilo-gram scale process route65,66 is described in of YU-101 in which 3 of the amino acids of this pentapeptide were
Scheme 9. modified to improve the potency of the molecule.70 After licensing
The preparation began with 6,7-dimethoxy-quinoline-4-ol (46) the molecule to Proteolix, the introduction of the morpholino
which upon treatment with POCl3 provided chloride 47 in 70% group was found to improve the solubility of the drug while main-
yield. Exposure of 47 to 4-aminophenol under basic conditions taining efficient interaction with the target. The most scalable
using t-BuONa furnished diaryl ether 48 in 72% yield. This aniline route to carfilzomib closely resembles the original route developed
was then coupled with amidoacid chloride 51 (which arose from toward epoximicin and is described herein.71,72
the activation of commercial diacid 49 to the corresponding mono- The synthesis was initiated with the amide coupling of phenyl
chloride, coupling with p-fluoroaniline, and subsequent exposure alanine methyl ester (53) and N-Boc leucine (54) using standard
to oxalyl chloride to furnish the transient acid chloride) to con- coupling reagents to afford dipeptide 55 in high yield (Scheme 10).
struct cabozantinib as the free base 52 in 95% yield. Salt formation Acidic removal of the amine protecting group, followed by a second
of cabozantinib 52 was carried out with (S)-malic acid, which ulti- amide coupling reaction with N-Boc homophenyl alanine, provided
mately delivered the final product of cabozantinib (S)-malate (VIII) tripeptide 56 in 85% yield for the two steps. Acidic removal of the
in 75% yield.65,66 amine protecting group and subsequent acylation with chloroace-

O O O O
HO TMSO
TMSCl, NMM, THF
HO OH 0 °C to 35 °C, then -20 °C, 99% TMSO OTMS
OH OTMS

63 64

Cl Cl

Cl 1. (COCl) 2, DCM, DMF O


Br Br
20 °C, 100% Et3 SiH, BF3 •OEt2
O
Br 2. phenetole, AlCl3 , DCM CH 3CN, 10 °C, 75%
OH 0 °C to 5 °C, 91%
O O

65 66 67

Cl

O
HO
1. n-BuLi, 64, THF, toluene, -78 °C OMe
2. MeOH, CH3 SO3 H, 85% HO OH
OH

68

1. aq. LiOH
Cl Cl
1. Et3 SiH, BF3 •OEt2 THF, H2 O
DCM, CH3 CN O MeOH O OH
AcO HO
-10 °C RT, 100% OH
2. Ac 2O, pyridine AcO OAc 2. propanediol HO OH
H 2O
DMAP, DCM OAc H 2O OH
55% f or 2 steps O O
69
X Dapagliflozin propanediol hydrate

Scheme 12. Synthesis of dapagliflozin propanediol hydrate (X).


2016 H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032

tyl chloride yielded b-chloro amide 57 in 67% yield. Reaction of 57 5-bromo-2-chlorobenzoyl acid (65) was converted to the corre-
with morpholine in the presence of catalytic amounts of potassium sponding acid chloride with oxalyl chloride. Subsequently, this
iodide followed by saponification of the methyl ester with lithium acid chloride was subjected to Friedel–Crafts acylation with ethyl
hydroxide led to acid 58 in 87% yield for the two steps. Finally, phenyl ether (‘phenetole’) in the presence of aluminum trichloride
amide coupling between acid 58 and keto-epoxyamine 59 (whose at low temperature to give benzophenone 66 in 91% yield. Next,
preparation is described in Scheme 11) using HOBT as the coupling the carbonyl functionality within 66 was removed upon treatment
reagent and recrystallization of the resulting product ultimately with triethylsilane and boron trifluoride-etherate, producing
gave carfilzomib (IX) in 75% yield. 5-bromo-2-chloro-40 -ethoxydiphenylmethane (67) in 75% yield as
Keto-epoxyamine 59 was prepared from N-Boc leucine (54) as the aglycon partner. Aryl bromide 67 was subjected to lithium hal-
described in Scheme 11. Reaction of 54 with isobutyl chlorofor- ogen exchange conditions and subsequent exposure to lactone 64,
mate followed by N,O-dimethylhydroxylamine provided Weinreb provided a mixture of lactols which were then immediately sub-
amide 60 in 94% yield. Grignard addition of isopropenylmagnesium jected to methanesulfonic acid, leading to glucol 68 in 85% yield.
bromide (60) provided enone 62 in 81% yield. Epoxidation of 62 The anomeric methoxy group of 68 was reduced with triethylsilane
with calcium hypochlorite provided a mixture of epoxides giving and boron trifluoride-etherate followed by peracetylation to deli-
41% yield of the desired isomer (presumably isolated by chroma- ver a-C-glycoside tetraacetate 69 in 55% (two steps) after recrys-
tography), and subsequent treatment with TFA liberated the talliaztion in ethanol. Hydrolysis of polyacetate 69 with lithium
amine, providing the TFA salt of ketoepoxy amine 59 in 92% yield. hydroxide gave dapagliflozin in quantitative yield, and upon treat-
ment with propanediol in water, dapagliflozin propanediol hydrate
11. Dapagliflozin propanediol hydrate (ForxigaÒ, EmplicitiÒ, (X) was produced.
EdistrideÒ, AppebbÒ)
12. Enzalutamide (XtandiÒ)
Dapagliflozin propanediol hydrate, an orally active sodium glu-
cose cotransporter type 2 (SGLT-2) inhibitor, was developed by In August 2012, the FDA approved enzalutamide, marketed by
Bristol–Myers Squibb (BMS) and AstraZeneca for the once-daily Medivation and Astellas Pharma U.S. for the treatment of meta-
treatment of type 2 diabetes. As opposed to competitor SGLT-2 static castration-resistant prostate cancer (CRPC), specifically for
inhibitors, dapagliflozin was not associated with renal toxicity or those patients who had previously received docetaxel.76 Enzaluta-
long-term deterioration of renal function in phase III clinical tri- mide is an inhibitor of androgen receptors (AR)—whose increased
als.73 The drug exhibits excellent SGLT-2 potency with more than expression has been closely linked with castration-resistant pros-
1200-fold selectivity over the SGLT-1 enzyme.74 tate cancer (CRPC), thus, AR inhibitors have seen increased recent
The most likely process-scale synthesis has been described in a attention from the medicinal chemistry community. Phase I/II
literature publication and patent, and this is summarized in trials were particularly promising for enzalutamide, as 43% of
Scheme 12 below.74,75 The synthesis began with global silylation patients showed >50% sustained suppression of a key serum
glucolactone 63 to form tetrasiloxide 64. In parallel, commercial biomarker.77 Of the several patents and papers describing

O O
CO 2H
SOCl2, IPAc Cl CH3 NH2 , IPAc, 2 °C to 35 °C NHMe
Br F DMF, 60 °C to 72 °C 90% for 2 steps
Br F Br F

70 71 72

O
H2 N CO2 H 73 NHMe K2 CO3 , DMF, H 2 O
HO MeI, 30 °C to 40 °C
CuCl, K2 CO3, DMF, H 2O, 30 °C N F
H 95%
then 2-acetylcyclohexanone, 105 °C O
76%
74

CF3
O
NC
NHMe 76, DMSO S
O
MeO IPAc, 84 °C N
N F N
H 78% NHMe
O O F
75 XI Enzalutamide

S
CF3 CF3
NC Cl Cl NC
Heptane, H 2O, 5 °C to 40 °C
NH2 NCS
84%
77 76

Scheme 13. Synthesis of enzalutamide (XI).


H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032 2017

synthetic approaches,78–81 a 2011 patent represents the most likely was also independently developed by Simcere Pharmaceutical
scale production route to enzalutamide, and this is described in Group and is marketed as IremodÒ in China. The drug exhibited
Scheme 13.82 inhibitory effects on granuloma inflammation, and was shown to
Commercially available carboxylic acid 70 was first converted be efficacious for the prevention of joint destruction in adjuvant
to the corresponding acid chloride 71, followed by amide forma- arthritis.84,85 While several synthesis of iguratimod have been pub-
tion with methylamine to furnish benzamide 72 in 90% yield over lished,86 the most likely scale synthesis, which does not require
two steps. Bromide 72 was then coupled with amine 73 using cop- chromatographic purification, is described in Scheme 14.87
per(I) catalysis to afford trisubstituted benzene 74 in 76% isolated The synthesis began with commercially available 3-nitro-4-
yield. Esterification of 74 to 75 with iodomethane furnished one chloro anisole (78) which was reacted with potassium phenoxide
fragment for the key ring-forming event. Isothiocyanate 76, avail- (generated from phenol and potassium t-butoxide at 110 °C) to
able in one step from the corresponding aniline 77, was then ex- provide the corresponding nitrophenyl ether which was subse-
posed to aminoester 75 in the presence of warm isopropyl quently reduced and sulfonylated to furnish sulfonamide 79. Next,
acetate, resulting in construction of the lynchpin thiohydantoin this diphenyl ether was submitted to a Friedel–Crafts reaction with
and delivering enzalutamide (XI) in an impressive 78% yield. This aminoacetonitrile hydrochloride which gave rise to aminomethy-
5-step process has successfully generated multi-gram quantities lacetophenone 80 in 90% yield. This aminoketone was then formy-
of the drug in 50.7% overall yield. lated with formic trimethylacetic anhydride 81 at room
temperature to afford formamide 82 in 91% yield, and this material
13. Iguratimod (CareramÒ, IremodÒ) was immediately subjected to O-demethylation conditions with
aluminum trichloride and sodium iodide in acetonitrile to give
Iguratimod, which was discovered by Toyama Pharmaceuticals the phenol 83 in 95% yield. Finally, treatment of the aminomethyl
and jointly co-developed with Eisai in Japan, was approved by the acetophenone phenol 83 with N,N-dimethylformamide dimethyl-
PMDA (Pharmaceuticals and Medical Devices Agency) of Japan on acetal in DMF at low temperatures furnished iguratimod (XII) in
June 29, 2012 for the treatment of rheumatoid arthritis.83 This drug 87% yield.

H
N O
1. PhOH, t-BuOK, DMF, 110 °C, 84% S
O 2N O O O
2. Fe/4 N HCl, EtOH, 65 °C to 70 °C, 72%
O
Cl 3. MeSO 2Cl•Py, 0 °C to RT, 82%

78 79

H O O H
N O N O
1. NCCH2 NH2 •HCl, AlCl3 , 40 °C S S O
O O O H O O
2. dry HCl, 25-30 °C, PhNO2 81
O NH 2 O N H
90% for 2 steps HCl acetone, RT, 91% H
O O

80
82

H H
N OH O N O
S O N S O
AlCl3 , NaI O O O O O
O N H O N H
CH3 CN, RT, 95% H DMF, 15 °C, 87% H
O O

83 XII Iguratimod

Scheme 14. Synthesis of iguratimod (XII).

COOH

85 O O
O O S S
S O NH 2 O
1. 85, Et 3N, CH 3CN, RT O , HOAc N

Br 2. Et 3N, CH3 CN, 75 °C 50 °C to 120 °C, 85%


O O
O 72% for two steps

84 86 XIII Imrecoxib

Scheme 15. Synthesis of imrecoxib (XIII).


2018 H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032

O O
H H
acetone, cat. p-TsOH, RT

HO HO 69% HO HO
HO O
HO O

87; ingenol 88

O O
O O
O
89 H H3 PO4, RT H

LHMDS, THF, RT, 73% O 71% O


O HO O HO
O HO
O HO

90 XIV Ingenol mebutate

Scheme 16. Synthesis of ingenol mebutate (XIV).

O O
OH O O O
EtO O H2 SO4 , HNO 3 EtO O
EtO Cl DCM, 0 °C to RT Cl
Et3N, EtOAc 57%
N
0 °C to RT, 99% O2 N H
94

91 92 93

O
OH
EtO O
O O
1. Pd/C, H 2, MeOH O O KOH, MeOH
2. 94, Et3N, DCM, RT 96% N
N H
53% f or 2 steps H N
N H
H
95 XV Ivacaftor

Scheme 17. Synthesis of ivacaftor (XV).

OH PBr 3, 0 °C to 85 °C Br 1. 1-amino-2-propanol, 85 °C to 100 °C


95% 2. SOCl2 , DMA, PhCH3 , 65 °C
Cl Cl
71% for 2 steps
96 97

1. AlCl3, 1,2-dichlorobenzene, 128 °C O OH


NH • HCl 2. aq. NaOH, cyclohexane NH • OH
Cl Cl HO
3. L-(+)-tartaric acid, acetone /H2 O
4. Recrystallization from acetone/H 2O OH O
Cl
27% for 4 steps 2
98 99

1. K2 CO3, H2O, RT
2. EtOAc NH • HCl
Cl • 1/2 H 2O
3. HCl (gas), 0 °C to 5 °C
90% for 3 steps
XVI Lorcaserin hydrochloride hydrate

Scheme 18. Synthesis of lorcaserin hydrochloride hydrate (XVI).


H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032 2019

Br 1. i. Mg, THF HO CO2 Et 1. p-TsOH, toluene, 65 °C, 69%


ii. diethyl oxalate, ↑↓, 70% 2. KOH, EtOH, ↑↓, 98%
CH 2CO2 Me
2. 101 , LHMDS, THF 3. BF3, MeOH, RT, 69%
100 -78 °C, 72% 102

O O R* O R*
1. 104 , Et3N, DMAP, toluene, RT
CO2 H +
2. 105 , RT, 84% O O
MeO2 C (2 diastereomers, 1:1) MeO 2C MeO2 C

103 106a (desired) 106b (undesired)

OMe OMe
Cl O
N N
O Cl R* =
OH O
O Cl Cl N N

101 104 105 quinine

Scheme 19. Synthesis of fragment 106a of omacetaxine mepesuccinate (XVII).

O
N
O
O OH 1. 104 , Et N, toluene , 30 °C H
H2 , Pd/C 3
106a O
EtOAc, RT, 50% O 2. 108 , RT, 43% O OMe
MeO2C O

CO2Me
107 109

O
N
O
H O
N
O
1. HBr/HOAc, DCM, -10 °C, 87% OMe O
O H
O
2. 5% aq. NaHCO3 , acetone, RT, 47%
O OH HO
OMe
108 cephalotaxine
HO

XVII Omacetaxine mepesuccinate

Scheme 20. Synthesis of omacetaxine mepesuccinate (XVII).

O 1. Fmoc-OSu, aq. Na2CO3 , THF, RT Fmoc


H
N 2. triphosgene, N-Boc-diaminoethane N
O OH
O THF, RT
BocHN
3. 0.1 N HCl in THF N O O
HO H
49% f or 3 steps

110 111

Scheme 21. Synthesis of fragment 111 of pasireotide (XVIII).

14. Imrecoxib (HengyangÒ) Medica (IMM) of the Chinese Academy of Medical Sciences in col-
laboration with Hengrui Pharmaceuticals.88 Imrecoxib, which is a
Imrecoxib, a new non-steroid anti-inflammtory drug (NSAID), moderately selective COX-2 inhibitor (with IC50 values against
was launched in China with the trade name of HengyangÒ for the COX-1 and COX-2 being 115 ± 28 and 18 ± 4 nM, respectively),89
treatment of osteoarthritis in 2012. It was originally designed is the subject of two synthetic routes reported across several
and synthesized by Guo and co-workers at the Institute of Materia publications.90–93
2020 H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032

OBn O 1. 20% piperidine in


DMF, 0 ºC to RT, 0.25 h
OBn O O
Fmoc-Lys(Boc)-OH 2. Fmoc-DTrp(Boc)-OH
O DIC/HOBt, DMF, RT, 16 h DIC/HOBt, DMF, RT, 16 h
O
O
O
O O NH
HN
R NH
( )4
BocHN Fmoc
20% piperidine,
112; R = Fmoc
DMF, 0 ºC to rt, 114
113; R = H
0.25 h

OBn O
O OBn O
1. 20% piperidine in
DMF, 0 ºC to RT, 0.25 h O
O 2. Fmoc-PhG-OH
DIC/HOBt, DMF, RT, 16 h O
O
O NH O
O O NH
NHFmoc O Ph
( )4 N H
H N
BocHN ( )4 N NHFmoc
H
Boc N BocHN O
Boc N

115
116

Scheme 22. Synthesis of pasireotide intermediate 116. (XVIII).

The most likely process-scale route to this drug is described in Euphorbia pelpus.95 From natural extractions, 17 kg of fresh E. pel-
Scheme 15,93 which began with 2-bromo-40 -(methylsulfonyl)-ace- pus afforded 7 g of ingenol 3-angelate as an oil, which upon further
tophenone (84) and p-tolylacetic acid (85) as starting materials. In purification was deemed insufficient for process-scale produc-
the presence of base, a-bromoketone 84 was treated with acid 85 tion.96 Although several synthetic approaches to the ingenol family
which resulted in lactone 86 in 72% yield across the two-step se- of terpenes have been reported,97–113 Liang and coworkers at LEO
quence. Exposure of lactone 86 with propylamine triggered a Pharma have reported a semisynthesis of the API from naturally-
ring-opening-ring closing reaction, which resulted in imrecoxib occurring ingenol. This natural product’s accessibility from the
(XIII) directly in 85% yield.93 seeds of E. lathyris renders it widely commercial on scale. The con-
version of ingenol to ingenol mebutate involves a protection, ester-
15. Ingenol mebutate (PicatoÒ) ification, and deprotection strategy to procure scale quantities of
the drug (Scheme 16).114 Conversion of ingenol (87) to the corre-
Ingenol mebutate is a diterpene ester which was approved in sponding 5, 20-acetonide 88 proceeded in good yield using a pro-
the U.S., EU, Australia, and Brazil for the treatment of actinic kera- tocol modified from the original conditions described by Hecker.115
tosis, a disease stage associated with sun exposure which can A considerable amount of study was conducted by Liang to affect
potentially develop into cancer.94 The drug, which is marketed by efficient angeloylation with minimal isomerization of the double-
LEO Pharma A/S as PicatoÒ, is administered as a topical gel bond to the corresponding Z-isomer (tiglate). It was found that an-
(0.015%, 0.05%) which has been proven effective in treating face-, gelic anhydride 89 (which is a commercially available reagent, but
scalp-, and trunk-localized actinic keratosis in four randomized, for process scale was prepared immediately prior to usage from the
double-blind, vehicle-controlled, multicenter studies.94 The drug self-condensation of 99.5% pure angelic acid with 0.5 equivalents
exhibits mild side effects limited to application-site conditions of DCC) in the presence of LHMDS gave acetonide 90 in over 95%
(e.g. irritation, pain, pruritus), and no detectable concentrations conversion and was practically free of the undesired tiglate bi-
of ingenol mebutate or two of its metabolites were found in blood product after recrystallization (73% yield).96 Deprotection of the
samples.94 Traditionally used as a home remedy for various skin acetonide 90 was affected using phosphoric acid and after three
conditions, the ingenol mebutate, also referred to as ingenol recrystallizations, ingenol mebutate (XIV) was produced on multi-
3-angelate, is the main active constituent of sap from the plant gram scale in a combined yield of 37% starting from ingenol 87.96
H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032 2021

16. Ivacaftor (KalydecoÒ) 17. Lorcaserin hydrochloride hydrate (BelviqÒ)

Vertex’s ivacaftor was granted breakthrough therapy desig- Lorcaserin hydrochloride is a selective serotonin 5HT2C agonist
nation by the FDA in January 2012 for cystic fibrosis (CF) approved in the U.S. for the treatment of obesity. Lorcaserin hydro-
patients who bear the G551D mutation in the Cystic Fibrosis chloride was discovered and developed by Arena Pharmaceuticals,
Transmembrane Regulator (CFTR) gene. This CFTR mutation Inc. and licensed to Eisai. Lorcaserin hydrochloride is reported to be
occurs in roughly 4% of the 30,000 people living with CF in approximately 100 fold more active at the 5HT2C receptor than the
the United States. While the compound has been identified 5HT2B receptor.135 The significance of this selectivity is that 5HT2B
as a potentiator in cell-based assays, its mechanism of action activation is hypothesized to be associated with the cardiac valvul-
is currently unknown.116–118 While several patents describe a opathy side effect of the infamous ‘fen–phen’ (fenfluramine + dex-
synthesis of ivacaftor,119–133 only one demonstrates the synthe- fenfluramine) combination treatment for obesity.136 Numerous
sis on scale and includes yields, which is depicted in syntheses of lorcaserin hydrochloride have been reported135,137,138
Scheme 17.134 and the process scale route is highlighted in Scheme 18.139
Beginning with treatment of commercial di-tert-butylphenol Commercial 2-(40 -chlorophenyl)ethanol (96) was treated with
derivative 91 with ethyl chloroformate, the synthesis of carbon- phosphorus tribromide to give 2-(40 -chlorophenyl)ethyl bromide
ate 92 was achieved in quantitative yield. Nitration of 92 pro- (97) in 95% yield. Alkylation of 1-amino-2-propanol with 97 fol-
vided the desired nitroarene regioisomer 93 in 57% yield which lowed by treatment of the corresponding alcohol with thionyl
was isolated by recrystallization. Reduction of the newly-in- chloride gave chloroamine 98 in 71% yield. Friedel–Crafts acylation
stalled nitro group and subsequent amide bond formation via of 98 with aluminum trichloride followed by a classical resolution
reaction with commercially available acid chloride 94 produced with L-(+)-tartaric acid gave the desired (R)-enantiomer tartrate
amide 95 in 53% yield over the two step sequence. Finally, cleav- salt of lorcaserin 99 in 27% overall yield from 98. The free base of
age of the carbonate unmasked the phenol to furnish ivacaftor 99 was liberated upon treatment with aqueous potassium carbon-
(XV) in 96% yield. ate and this material was then immediately extracted into ethyl

OBn O 1. 20% piperidine in


O DMF, 0 ºC to RT, 0.25 h
2. Fmoc-Phe-OH,
O DIC/HOBt, DMF, RT, 16 h
1. 20% piperidine in 3. 20% piperidine in
DMF, 0 ºC to RT, 0.25 h O DMF, 0 ºC to RT, 0.25 h
2. 111, DIC/HOBt O NH
O Ph O Fmoc 4. 2% TFA in CH2 Cl2
DMF, RT, 16 h H
N N RT, 1 h
116 ( )4 N N (S)
H H
BocHN O (R)
Boc N
O
HN
O
BocHN
117

OBn

OBn
O
Ph
OH NH 2 H
1. DPPA, DIEA, H2 N N
O NH O DMF, 0 ºC, 16 h ( ) 3 HN
O Ph O Ph
H 2. aq. TFA, RT, 40 O
N N mins O O N
( )4 N N O
H H HN O O
BocHN O O O
Boc N 20% from 112 HN
O NH
HN N
H H2 N
O Ph
BocHN H N

119 XVIII Pasireotide

Scheme 23. Conversion of pasireotide intermediate 116 to pasireotide (XVIII).


2022 H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032

acetate and treated with HCl gas to give lorcaserin hydrochloride provided the active homoharringtonine product, which is de-
hydrate (XVI) in 90% yield. scribed in Scheme 19.152,153
The method for large scale synthesis of homoharringtonine be-
18. Omacetaxine mepesuccinate (SynriboÒ) gins with derivatization of commercial 5-bromo-2-methyl-pent-2-
ene (100) with diethyl oxalate and the pre-formed enolate of
SynriboÒ (Omacetaxine mepesuccinate) was approved by the methyl acetate (101), generating diester 102 in 50% overall yield
FDA for the treatment of adult patients with chronic or accelerated (Scheme 19).153 Acid-promoted pyran formation, followed by uni-
phase chronic myeloid leukemia (CML) exhibiting resistance or versal ester saponification and selective re-esterification provided
intolerance to tyrosine kinase inhibitors (TKI’s). Omacetaxine the desired racemic pyran acid 103. Activation of acid 103 with
mepesuccinate inhibits protein synthesis and prevents amino- 2,4,6-trichlorobenzoyl chloride (104) and subsequent addition of
acyl-tRNA binding during the elongation phase and targets myelo- quinine (105) led to a mixture of diastereomers 106a/106b (1:1)
ma-promoting molecules Mcl-1, XIAP, and b-catenin,140–142 which in 84% yield, which were separable by chromatographic methods.
are particularly important in the survival of myeloma cells.143 Diastereomer 106a was then carried on to the synthesis of homo-
Omacetaxine mepesuccinate is also known as homoharringtonine, harringtonine (Omacetaxine mepesuccinate) as described in
an alkaloid originally discovered144 and structurally identified145–147 Scheme 20.153
from Cephalotaxus harringtonia, which occurs naturally in Japan From isomer 106a, hydrogenolysis with Pd/C and H2 provided
and eastern Asia. Because of its leukemic activity and interesting acid 107 in 50% yield. Activation of 107 with 2,4,6-trichlorobenzoyl
chemical structure, the core and ester side chains of the cephalo- chloride (104) followed by addition of cephalotaxine (CET) (108)
taxine alkaloids have been the focus of numerous synthetic stud- provided the desired cephalotaxine-coupled product 109 in 43%
ies.148 However, large-scale production often utilizes a yield. Sequential treatment with HBr/HOAc and 5% aqueous
semisynthetic route which relies upon cephalotaxine (CET) derived NaHCO3 completed the synthesis, providing omacetaxine mepe-
from natural sources149 coupled with a synthetically obtained ester succinate (XVII) in 41% yield (over two steps).153
side chain.150,151 The challenges associated with direct esterifica-
tion of cephalotaxine with the homoharringtonine and other re- 19. Pasireotide (SigniforÒ)
lated ester side chains are the basis of ongoing research aimed at
identification of improved side-chain coupling methods.148,152 Pasireotide, also known as SOM230, is a cyclic, hexameric
The most likely process-scale synthetic route features the coupling peptide developed by Novartis which exhibits somatostatin-like
of the homoharringtonine side chain with the cephalotaxine core, activity as an antisecretory agent used in the treatment of
and a subsequent conversion of the a-hydroxy moiety to a bridged Cushing’s disease.154 Pasireotide activates a broad range of
heterocyclic species. Following this coupling, ring opening somatostatin receptors, and in particular displays a significantly

N
Br 1. n-BuLi, hexanes
Br2 , NaOAc, EtOAc N THF, –76 °C N N
OMe 10 °C to 50 °C, 86% OMe 2. 122
OMe
120 121 123

HCl, H 2O 125, Cu(OAc)2 , H2 O NBS, DMF


N NH N N
↑↓, 60% pyridine, DMF 30 °C
O 28 °C to 40 °C, 91% O

124 126

1. 128, Pd(OAc) 2, PPh 3, CuI N N


N N K2CO3 , DME, ↑↓
O 3/4 H2 O
2. Recrystallization from acetone /H2 O
O CN
86% for 3 steps
Br

127 XIX Perampanel hydrate

O N
B B
O
N S O O
B O
O B
O

122 125 128

Scheme 24. Synthesis of perampanel hydrate (XIX).


H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032 2023

higher binding affinity for somatostatin receptors 1, 3, and 5 than Beginning from (2S,4R)-4-hydroxyproline methyl ester (110) in
its competitor somatostatin-mimic octreotide in vitro, as well as a Scheme 21 above, this pyrrolidine nitrogen was first Fmoc-pro-
comparable binding affinity for somatostatin receptor 2.154 tected in 85% yield, followed by treatment with triphosgene and
Pasireotide is more potent than somatostatin in inhibiting the N-Boc diaminoethane to provide the prolino carbamate. 111 in
secretion of human growth hormone (HGH), glucagon, and 49% yield over the two step sequence after a recrystallization with
insulin.155 ethyl acetate.156
The synthesis of pasireotide is relatively straightforward, given Next, commercially available Fmoc-Tyr(Bzl)-O-CH2-Ph(3-
that the chemical entity is a cyclic peptide. The most likely scalable OCH3)-O-CH2-SASRIN157 resin (112) was used as starting material
route closely mimics that described by the discovery authors in a manually operated reactor and carried through a standard
involving a series of conventional couplings and deprotection steps protocol consisting of repetitive cycles of Na deprotection (piper-
to arrive at a linear peptide which then underwent sequential re- idine/DMF, 2:8), repeated washings with DMF, and coupling using
lease from solid support, macrocyclization, and a global deprotec- DIC/HOBT in DMF (Schemes 22 and 23). The following amino acid
tion step.156 derivatives were sequentially coupled: Fmoc-Lys(Boc)-OH,

O O O F O F
F F
OH Ac 2O 131 + N
O
N OH ↑↓, 76% N AlCl3, ↑↓, 84% N
CO2 H CO2 H
O O F F
129 130 132a 133b

NH2
O F NH2 O HN O
1.
H2N 134
20% H 2SO4 THF, 55 °C OH
•2
N N OH
140 °C, 81% 2. AcOH, H 2O
O F 3. 3 M aq. maleic acid O HN O
40 °C to 50 °C NH 2
92% for 3 steps
133 XX Pixantrone dimaleate

Scheme 25. Synthesis of pixantrone dimaleate (XX).

N
Si(CH 3) 3 136 TBAF, THF N
N N
N N
N Pd(PPh3) 4, CuI, DIPEA RT, 94% N
N
Br CH 3 CN RT, 71%
Si(CH 3) 3
135 137 138

N
N
N
HCl
1. 139, Pd(PPh3 )4 , CuI N
EtOAc, Et3 N, RT H
N N
2. EtOH, HCl, RT
53% for 2 steps O
CF3

XXI Ponatinib hydrochloride

I I

1. SOCl2, DCM, RT O
CF 3
OH 2. H 2N
N HN
O N
N
140 141 CF3 139
N
DCM, DIPEA, DMAP, RT, 65%

Scheme 26. Synthesis of ponatinib hydrochloride (XXI).


2024 H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032

Fmoc-D-Trp(Boc)-OH, Fmoc-PhG-OH, proline derivative 111 Bromination of commercial 2-methoxypyridine (120) gave
above, and finally Fmoc-Phe-OH. Couplings were continued or 5-bromo-2-methoxypridine 121 in 86% yield (Scheme 24). Lithium
repeated until complete disappearance of residual amino groups halogen exchange was then accomplished by treating 121 with n-
as monitored with a ninhydrin stain test. Before cleavage of the butyllithium, followed by reaction with 2-benzenesulfonylpyridine
protected linear peptide from its resin support, the Fmoc group (122) to provide bi-aryl 123. Hydrolysis of 123 under acidic condi-
was removed. After washings with dichloromethane, the peptide tions gave pyridinone 124 in 60% overall yield. N-Arylation with
resin was transferred into a column and the peptide fragment triphenylboroxine (125) in the presence of copper acetate afforded
was cleaved from solid support upon subjection to 2% TFA in N-aryl pyridinone 126 in 91% yield. Pyridone 126 was reacted with
dichloromethane. The eluant was immediately neutralized with N-bromosuccinimide to give bromopyridine 127, which was
a saturated NaHCO3 solution which resulted in the side coupled with 2-(1,3,2-dioxaborinan-2-yl)benzonitrile (128) under
chain protected fragment 119 (Scheme 23). This material was palladium-catalyzed conditions to give perampanel hydrate (XIX)
obtained in 93% homogeneity and cyclized without further in 86% yield after recrystallization from acetone/H2O.
purification. For cyclization, the linear fragment was dissolved
in DMF and subsequently treated with DIPEA and 1.5 equiv of 21. Pixantrone dimaleate (PixuvriÒ)
diphenylphosphoryl azide, resulting in the protected cyclized
product in good yield. For complete deprotection, the residue Pixuvri (Pixantrone dimaleate) is a novel aza-anthracenedione
was dissolved at 0 °C in aqueous TFA, and the mixture was derivative approved in Europe for the treatment of adult patients
stirred at this temperature for 30 min. The product was then with non-Hodgkin B-cell lymphoma.163 It is also being pursued
precipitated with ether containing ca. 10 equiv of HCl, then as a treatment for various cancers, and specifically as an alternative
filtered and washed with ether, and finally dried. The entire to other structurally-related drugs like mitoxantrone, employed for
sequence produced pasireotide (XVIII) in 20% yield from treatment of breast cancer, acute myeloid leukemia (AML), and
resin-bound 112.156 non-Hodgkins lymphoma.164 Pixantrone dimaleate has been de-
signed to maintain antitumor efficacy while decreasing highly car-
20. Perampanel hydrate (FycompaÒ) diotoxic side effects observed during treatment with other related
anti-tumor anthracenedione derivatives.164–166 Like many anthra-
Perampanel is a selective, non-competitive a-amino-3-hydro- cenedione drugs, the mechanism of action for pixantrone dimal-
xy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) antago- eate likely includes a number of pathways and processes, with
nist approved for partial-onset seizures in patients with studies suggesting intercalation into DNA and/or interference with
epilepsy.158,159 Perampanel was discovered and developed by Eisai. DNA—Topoisomerase II activity, leading to subsequent protein
A number of synthetic routes to perampanel have been reported160 associated-DNA strand breaks and eventually to cell death.167,168
and the process scale route is described herein.161,162 Pixantrone dimaleate, also known as BBR 2778, was originally

O
O N O
O
N N 144, NaOH, i-PrOH, ↑↓
N

N Et 3 N, 110 °C, 81% N

142 143

N N
N H
N N N
N O 146, t -BuOK, THF N N
N N H
H -25 °C to RT, 85% O
O N
N
CF3
145 147

N N
H
N N N
35% aq. HCl, acetone N N
H
O
0°C to RT N
2 HCl CF3
XXII Radotinib dihydrochloride

N
NH H2N N
O
H 2N N
H
O
HNO 3 CF3
144 146

Scheme 27. Synthesis of radotinib dihydrochloride (XXII).


H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032 2025

synthesized by Professors Krapcho and Hacker at the University of AlCl3-promoted Friedel–Crafts reaction of 1,4-difluorobenzene
Vermont,169 and determination of in vitro tumor cell cytotoxicity (131) with 130 under reflux conditions provided a mixture of
was co-identified by the Boehringer Mannheim Italia research nicotinic acid isomers 132a/132b in 84% yield, which were carried
center and the University of Vermont.170 After the merger of directly to the next step. Cyclization with fuming H2SO4 yielded
Boehringer Mannheim with La-Roche, Novuspharm, and Cell the desired difluorobenzo-isoquinoline-dione core 133, which
Therapeautics, Inc., pixantrone dimaleate has been developed and was further functionalized with ethylenediamine (134) to provide
marketed by Cell Therapeutics, Inc. the free base of pixantrone. Subjection of the pixantrone free base
The manufacturing scale synthesis of pixantrone dimaleate re- to aqueous acetic anhydride and maleic acid provided pixantrone
lies on several process modifications171,172 from the original syn- dimaleate (XX) in 92% yield over 3 steps.171
thesis reported by Krapcho in 1994.169 This modified procedure
has provided active pharmaceutical ingredient (API) in high purity 22. Ponatinib hydrochloride (IclusigÒ)
(>99%) and is acceptable for use in pharmaceutical applications
(Scheme 25).171 Beginning with pyridine 3,4-dicarboxylic acid Ponatinib hydrochloride (IclusigÒ), previously known as
(129), generation of the corresponding anhydride 130 proceeded AP24534, is a multi-targeted tyrosine kinase inhibitor approved
in 76% yield upon treatment with refluxing Ac2O. Next, an in the U.S. as an oral treatment for resistant or intolerant chronic

O 1. SOCl2, DMF, chlorobenzene O


70 °C to 90 °C Cl 1. toluene, NaOH
OH N
2. methylamine H 2. 150, t-BuOK, NMP
N N
toluene, H 2O, 20 °C 100 °C, 84%
3. acetyl chloride HCl
EtOH, toluene, RT, quant.
148 149

CF3
Cl 152
O CF3 O
O Cl O
N N C O O N
H H
N N
H 2N THF, RT, 83% N N
H H
F F H2 O

151 XXIII Regorafenib hydrate

H 2N OH
N OH
cyclohexane, ↑↓
F
F
153 150

Scheme 28. Synthesis of regorafenib hydrate (XXIII).

Cl
Cl
O
H2 N CO2Me O
pyridine, DCM
HN CO2Me
HO Cl
Cl
Cl
HO
154 155 156

Cl
O CO2 Me 1. LiOH, THF, H2 O, 40 °C
p-TsOH, toluene , ↑↓

N 2. N-methyl-D-glucamine
Cl i-PrOH, H2 O, 82%
157

Cl O
O OH
N OH OH
Cl H
N
HO
OH OH

XXIV Tafamidis meglumine

Scheme 29. Synthesis of tafamidis meglumine (XXIV).


2026 H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032

myeloid leukemia (CML) and Philadelphia chromosome-positive 23. Radotinib hydrochloride (SupectÒ)
(Ph+) acute lymphoblastic leukemia (ALL).173 Ponatinib hydrochlo-
ride was designed for treatment of tumors containing the T351I In January 2012, radotinib hydrochloride (marketed as SupectÒ)
mutation which are present in some forms of CML and resistant obtained its approval from the KFDA (Korea Food and Drug Admin-
to traditional therapies such as imatinib.173–175 Ponatinib hydro- istration) for the treatment of patients with Philadelphia chromo-
chloride was developed by Ariad Pharmaceauticals, and operates some-positive chronic myeloid leukemia (CML) who have
by a similar mechanism of action as other tyrosine kinase inhibi- become resistant to existing drugs such as Gleevec, Tasigna and
tors, inhibiting the enzymatic activity of BCR-ABL, an abnormal Sprycel.181 Originally developed by IL-YANG pharmaceuticals of
tyrosine kinase responsible for unregulated and excess white blood South Korea as an oral second-generation tyrosine kinase inhibitor,
cell production by bone marrow.176 However, the ability of ponati- the drug inhibits both Bcr-Abl fusion protein and the platelet-de-
nib hydrochloride to target isoforms of the BCR-ABL gene typically rived growth factor receptor (PDGFR).182 Because of the structural
leading to resistance in other known tyrosine kinase inhibitors pro- similarity of radotinib to that of nilotinib (TasignaÒ), the process-
vides an alternate form of therapy not previously available.175 scale synthetic route (which is depicted in Scheme 27) is capable
A significant amount of research has been devoted towards of furnishing both drugs.183–185
identification of a manufacturing synthesis of ponatinib hydrochlo- Claisen condensation of commerical 2-acetylpyrazine (142)
ride.177–180 A majority of methods rely on two key Sonagashira with N,N-dimethylformamide dimethylacetal gave rise to the ena-
couplings to generate the imidazo[1,2-b]pyridazin-3-ylethynyl mino ketone 143 in 81% yield.186 Under basic conditions, vinylo-
framework. The most likely process-scale method begins with 3- gous amide 143 was coupled with commercial guanidine nitrate
bromo-imidazo[1,2-b]pyridazine (135) (Scheme 26).177,180 Direct 144187 to produce aminopyridine 145.184 Subsequent condensation
Sonogashira coupling of (135) with ethynyltrimethylsilane (136) with commercial aniline (146) by means of potassium t-butoxide
in the presence of Pd(PPh3)4 and CuI, followed by treatment with in THF constructed radotinib 147 in 85% yield as the free base,
TBAF/THF led to the desired alkynyl imidazo[1,2-b]pyridazine and this material could be converted to the radotinib dihydrochlo-
138 in 71% and 94% yields, respectively. Alkyne 138 was then cou- ride (XXII) upon exposure to concentrated hydrochloric acid in
pled under similar Sonogashira conditions with functionalized aryl chilled acetone.185
iodide 139 (generated in two steps from 3-iodo-4-methylbenzoic
acid (140) and commercially available piperazinyl aniline 141) 24. Regorafenib hydrate (StivargaÒ)
providing ponatinib free base, which was then immediately treated
with EtOH/HCl at room temperature to ultimately furnish ponati- Regorafenib was approved by the U.S. Food and Drug Adminis-
nib hydrochloride (XXI).177,180 tration (FDA) in September 2012 for the treatment of metastatic

O
O O NHNH 2
159 1. MsOH, RT
Boc N NH Boc N N O
DMF, RT, 86% 2. pyridine, POCl3 , RT, 12%

158 160

N N Boc N NH
N N TFA, DCM N N

88%

NaBH(OAc)3 , AcOH
161 162 1,2-DCE, RT, 50%
Boc O
H O H
N Boc N
HN 1. HOBt, EDC, DMF, RT, 62% N
OH + S
S 2. DMSO, SO3 Py, RT, 55%
HO
O
163 164 165

Boc
N O NH O

N N N N
N N
S 1. TFA, DCM, RT, 93% S
N N 2. 48% HBr, ↑↓, EtOH, 90% N N 2.5 HBr
x H 2O

166 XXV Teneligliptin hydrobromide hydrate

Scheme 30. Synthesis of teneligliptin hydrobromide hydrate (XXV).


H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032 2027

colorectal cancer in patients who have previously undergone flu- which was subsequently reacted with aqueous methylamine in tol-
oropyrimidine-, oxaliplatin-, and irinotecan-based therapies.188 uene to give 4-chloro-2-methylcarboxamide as its hydrochloride
The FDA expanded the approved use of the drug to include patients salt 149 in quantitative yield after treatment with acetyl chloride
with advanced gastrointestinal stromal tumors (GIST) that cannot in toluene and ethanol. Conversion of 149 to its free base form
be surgically removed and no longer respond to imatinib and sun- was performed with sodium hydroxide, and this intermediate
itinib, two other drugs approved for treatment of GIST. Regorafe- was immediately reacted with imine 150 (formed upon exposure
nib, marketed under the trade name StivargaÒ, was discovered to 4-amino-3-fluorophenol (153) in refluxing 3-methyl-2-buta-
and developed by Bayer Pharmaceuticals and marketed jointly none) in base to provide diaryl ether 151 in 85% yield. Reaction
with Onyx Pharmaceuticals.189 The active metabolites of the drug of amine 151 with the commercially available isocyanate 152 ulti-
inhibit multiple targets within a variety of kinase families includ- mately delivered regorafenib hydrate (XXIII) in 83% yield.
ing those in the RET, VEGF, FGFR, PTK, and Abl pathways.190,191
Among several published synthesis,192,193 the most likely pro- 25. Tafamidis meglumine (VyndaqelÒ)
cess scale synthesis will be highlighted from the two published
syntheses, and this is described in Scheme 28.194 Commercially Tafamidis meglumine is a transthyretin amyloid inhibitor that
available picolinic acid (148) was heated with thionyl chloride to was approved for the treatment of transthyretin amyloid polyneu-
provide the crude intermediate 4-chloro-2-pyridyl acid chloride ropathy (ATTR-PN) and transthyretin familial amyloid polyneropa-

CF3 CF3
O O O O
xylene KBr, H 2O 2, toluene
+ O N
H 2N ↑↓, 51% H
HCl (conc.), RT, 67%

167 168 169

CF3 CF3
O O OH O
NaCN, DMSO, RT
N 85% N
H H
Br CN

170 XXVI Teriflunomide

Scheme 31. Synthesis of teriflunomide (XXVI).

1. H2 , 5% Rh/C (JM C101023-5)


KOt -Bu, (MeO 2C)2 O O AcOH O
N N NBn
H 2N 2-MeTHF, toluene O N 2. PhCHO, NaHB(OAc) 3 O N
20 °C to 35 °C, 87% H toluene, 73% H

171 172 173

1. LAH, THF, RT 1. 2 N NaOH, i-PrOH, MeOH HO2 C O


NBn
2. HCl, i-PrOH, 87% N 2. di-p-toluoyl-L-tartaric acid NBn
H N
2HCl 42% for 2 steps H HO2 C O
2
174 175 98.6% ee O

Cl

N 176
NBn 1. H2 , Pd(OH)2 /C N OH
Cl N N N H 2 O, 70 °C to 75 °C N CN
H HOOC COOH
O
K2 CO 3, H 2 O, 95 °C to 105 °C N 2. DBU, MeOCCH 2CN N
quant. 1-butanol, 40 °C HOOC
Cl N N 3. citric acid, 90% N N
H H
177 XXVII Tofacitinib citrate

OH Cl

N POCl3, DIPEA N
N toluene, 106 °C N
HO N 52% Cl N
H H

178 176

Scheme 32. Synthesis of tofacitinib citrate (XXVII).


2028 H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032

thy (TTR-FAP).195–197 These diseases represent a rare autosomal give pyrazole 161 in 12% yield. The t-butyl carbamate was then re-
neurodegenerative disorder characterized by autonomic, sensory moved with TFA in dichloromethane to give amine 162 in 88%
and motor impairment which are typically fatal. Tafamidis was dis- yield. This amine was then subjected to butyrolactam 165 (which
covered at The Scripps Research Institute and developed by Pfizer. was prepared from N-Boc-trans-4-hydroxy-L-proline (163) coupled
Numerous synthetic routes have been reported including the use of with thiazolidine (164) under conventional amide-forming condi-
direct CH activation to form the key biaryl bond.198,199 Although tions using EDC) in the presence of sodium triacetoxy borohydride
only reported on small scale, the most likely production route is (STAB-H) in acetic acid.205 This reductive amination reaction affor-
detailed in Scheme 29.200–202 ded the cis-aminopyrrolidine 166 exclusively in 50% yield. Removal
Condensation of methyl 4-amino-3-hydroxybenzoic acid (154) of the t-butyl carbamate group with TFA afforded the teneligliptin
with 3,5-dichlorobenzoyl chloride (155) in refluxing pyridine gave free amine in 93% yield, which was then subsequently treated with
intermediate amide 156, which underwent cyclization upon treat- 48% hydrobromic acid in refluxing ethanol to give teleligliptin
ment with p-TsOH in refluxing toluene, producing benzoxazole hydrobromide hydrate (XXV) in 90% yield.
157. Saponification of the methyl ester with LiOH (aq) afforded
tafamidis. The free acid was treated with N-methyl-D-glutamine 27. Teriflunomide (AubagioÒ)
to provide tafamidis meglumine (XXIV) in 82% yield.
Teriflunomide (AubagioÒ), also known as A77 1726, is an imm-
26. Teneligliptin hydrobromide hydrate (TeneliaÒ) unosupressant marketed by Sanofi for the teatment of multiple
sclerosis (MS).206 Teriflunomide is the active metabolite of lefluno-
Teneligliptin is a DPP-4 inhibitor which was approved in Japan mide, used for treatment of patients diagnosed with rheumatoid
in 2012 for the treatment of type II diabetes.203 It was discovered arthritis, and therefore simultaneously can be used as a treatment
and developed by Mitsubishi Tanabe Pharma under the trade name for rheumatoid arthritis.207 Teriflunomide acts as an inhibitor of
TeneliaÒ. Similar to other marketed DPP-4 inhibitors, teneligliptin the mitochondrial enzyme dihydrorotate dehydrogenase,208–210
was well tolerated in all studies and QD dosing produced a long- inhibiting pyrimidine formation,211 and resulting in reduced B
lasting inhibitory action against DPP-4 and an increase in active and T cell proliferation.210
GLP-1 levels, with very low rates of renal excretion.204 Numerous syntheses of teriflunomide have been developed to
The only reported synthesis of teneligliptin is described in date,212–217 most relying on the use of 4-trifluoromethyl aniline
Scheme 30.203 Reaction of commercially available N-Boc-pipera- (167).212 The current optimized method for scale-up synthesis of
zine (158) with diketene (159) in DMF at room temperature gave teriflunomide, developed by Keshav and coworkers, begins with
acetoacetamide 160 in 86% yield, and this material was immedi- reaction of commercial 4-trifluoromethyl aniline 167 and ethylace-
ately condensed with phenylhydrazine in methanesulfonic acid toacetate (168) in refluxing xylenes, providing acetoamidate 169 in
followed by a cyclodehydration with phosphorus oxychloride to 51% yield (Scheme 31).215 The resulting acetoamidate 169 was

I I
Cl
H2 SO4 (conc.), NaIO4 , KI, RT Cl t-BuOH, DPPA Cl
O
73% Et 3 N, ↓↑, 84%
CO2 H N Ot-Bu
CO2 H
H
179 180 181

O O N
B2(pin) 2 , Pd(dppf)Cl 2 B 2-bromopyridine, Pd(PPh 3 )4
Cl Cl
KOAc, DMSO, 110 °C, 91% O NaHCO3 , H 2O, DME O
↑↓, quant.
N Ot-Bu N Ot-Bu
H H
182 183

N
N
TFA, DCM, 0 °C 185 , Et 3N, DCM Cl
Cl O Cl
98% 0°C to 5 °C, 99%
N
NH2 H
SO 2Me
184 XXVIII Vismodegib

Cl O Cl
HO 2C 10% NaOH, toluene, Et3 N
Cl
DMF, SOCl2 , 0 °C to 50 °C
SO2 Me SO2 Me
186 185

Scheme 33. Synthesis of vismodegib (XXVIII).


H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032 2029

then treated with H2O2, KBr, and concentrated HCl at room tem- References and notes
perature, providing bromide 170 in 67% yield. Bromide 170 was re-
acted with NaCN in DMSO, generating teriflunomide (XXVI) in 85% 1. Raju, T. N. K. Lancet 2000, 355, 1022.
2. Li, J.; Liu, K. K.-C. Mini-Rev. Med. Chem. 2004, 4, 207.
yield.215 3. Liu, K. K.-C.; Li, J.; Sakya, S. Mini-Rev. Med. Chem. 2004, 4, 1105.
4. Li, J.; Liu, K. K.-C.; Sakya, S. Mini-Rev. Med. Chem. 2005, 5, 1133.
5. Sakya, S. M.; Li, J.; Liu, K. K.-C. Mini-Rev. Med. Chem. 2007, 7, 429.
28. Tofacitinib citrate (XeljanzÒ)
6. Liu, K. K.-C.; Sakya, S. M.; Li, J. Mini-Rev. Med. Chem. 2007, 7, 1255.
7. Liu, K. K.-C.; Sakya, S. M.; O’Donnell, C. J.; Li, J. Mini-Rev. Med. Chem. 2008, 8,
Tofacitinib, a pan-Janus Kinase (JAK) inhibitor, was approved by 1526.
the FDA for the treatment of moderate to serious cases of rheuma- 8. Liu, K. K.-C.; Sakya, S. M.; O’Donnell, C. J.; Li, J. Mini-Rev. Med. Chem. 2009, 9,
1655.
toid arthritis (RA) in patients that have not responded to mono 9. Liu, K. K.-C.; Sakya, S. M.; O’Donnell, C. J.; Flick, A. C.; Li, J. Bioorg. Med. Chem.
therapy with methotrexate therapy.188 Tofacitinib, which was dis- 2011, 19, 1136.
covered and developed by Pfizer Inc.,218 is a potent and selective 10. Liu, K. K.-C.; Sakya, S. M.; O’Donnell, C. J.; Flick, A. C.; Ding, H. X. Bioorg. Med.
Chem. 2012, 20, 1155.
inhibitor of the JAK family of kinases and has shown potential for 11. Ding, H. X.; Liu, K. K.; Sakya, S. M.; Flick, A. C.; O’Donnell, C. J. Bioorg. Med.
treating psoriasis and organ transplant in addition to RA.219 Both Chem. 2013, 21, 2795.
the initial discovery synthetic route220,221 and the process scale 12. Graul, A. I.; Lupone, B.; Cruces, E.; Stringer, M. Drugs Today (Barc) 2013, 49, 33.
13. FDA approves Tudorza Pressair to treat chronic obstructive pulmonary
synthetic route have been disclosed in the chemical literature, disease. http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/
and the process approach is described in Scheme 32 below.222,223 ucm313052.htm; 2012, [Access Date: 2012-July-23].
Commercially available aminopyridine 171 was reacted with 14. Gavalda, A.; Miralpeix, M.; Ramos, I.; Otal, R.; Carreno, C.; Vinals, M.;
Domenech, T.; Carcasona, C.; Reyes, B.; Vilella, D.; Gras, J.; Cortijo, J.; Morcillo,
dimethyl dicarbonate in the presence of potassium t-butoxide to
E.; Llenas, J.; Ryder, H.; Beleta, J. J. Pharmacol. Exp. Ther. 2009, 331, 740.
give the methyl carbamate 172 in 87% yield. Hydrogenation of this 15. Fernandez Forner, D.; Prat Quinones, M.; Buil Albero, M. A. WO Patent 01/
carbamate 172 in the presence of 20 wt % of 5% Rh/C (JM type 04118 A2, 2001.
16. Prat, M.; Fernandez, D.; Antonia Buil, M.; Crespo, M. I.; Casals, G.; Ferrer, M.;
C101023-5) in acetic acid followed by reductive amination with
Tort, L.; Castro, J.; Monleon, J. M.; Gavalda, A.; Miralpeix, M.; Ramos, I.;
benzaldehyde and sodium triacetoxy borohydride furnished the Domenech, T.; Vilella, D.; Anton, F.; Huerta, J. M.; Espinosa, S.; Lopez, M.;
cis-benzyl protected piperidine 173 in 73% yield. Reduction of the Sentellas, S.; Gonzalez, M.; Alberti, J.; Segarra, V.; Cardenas, A.; Beleta, J.;
methyl carbamate within 173 with lithium aluminum hydride Ryder, H. J. Med. Chem. 2009, 52, 5076.
17. Busquets Baque, N.; Pajuelo Lorenzo, F. WO Patent 2008/009397 A1, 2008.
(LAH) in THF gave the corresponding methyl amino piperidine 18. Nyberg, K.; Östman, B.; Wallerberg, G. Acta Chem. Scand. 1970, 24, 7.
which was isolated as the dihydrochloride salt 174 in 87% yield. 19. 2012 China Drug Review Annual Report, 2013, http://www.cde.org.cn/linshi/
Enantiomeric resolution of the methyl amino piperidine was regulatEn/newsShow.jsp.
20. Wu, M. Y.; Ma, X. J.; Yang, C.; Tao, X.; Liu, A. J.; Su, D. F.; Liu, J. G. Acta
achieved by preparation of the free base of 174 with sodium Pharmacol. Sin. 2009, 30, 307.
hydroxide, conversion to the di-toluol-L-tartaric acid salt, and sub- 21. Guo, J. H.; An, D. WO Patent 2007/095789 A1, 2007.
sequent crystallization to give 175 in 42% yield and 98.6% ee. The 22. An, D.; Guo, J. H. CN Patent 101367795 B, 2012.
23. Tsubamoto, Y.; Goto, M. Folia Pharmacol. Jpn. 2013, 141, 339.
enantioenriched tartrate salt 175 was then directly reacted with 24. Kato, N.; Oka, M.; Murase, T.; Yoshida, M.; Sakairi, M.; Yamashita, S.; Yasuda,
dichloride 176 (obtained from reaction of commercial 7H-pyrrol- Y.; Yoshikawa, A.; Hayashi, Y.; Makino, M.; Takeda, M.; Mirensha, Y.;
o[2,3-d]pyrimidine-2,4-diol (178) with phosphorous oxychloride) Kakigami, K. Bioorg. Med. Chem. 2011, 19, 7221.
25. Yang, L. P. H.; McKeage, K. Drugs 2012, 72, 2375.
in the presence of potassium carbonate in water to give the cou-
26. Hu-Lowe, D. D.; Zou, H. Y.; Grazzini, M. L.; Hallin, M. E.; Wickman, G. R.;
pled product 177 in essentially quantitative yield. Hydrogenation Amundson, K.; Chen, J. H.; Rewolinski, D. A.; Yamazaki, S.; Wu, E. Y.; McTigue,
of intermediate 177 with DeGussa’s catalyst triggered concomitant M. A.; Murray, B. W.; Kania, R. S.; O’Connor, P.; Shalinsky, D. R.; Bender, S. L.
Clin. Cancer Res. 2008, 14, 7272.
debenzylation and chloride removal, and this was followed by
27. Rini, B. I.; Escudier, B.; Tomczak, P.; Kaprin, A.; Szczylik, C.; Hutson, T. E.;
installation of the cyanoacetate group and subsequent treatment Michaelson, M. D.; Gorbunova, V. A.; Gore, M. E.; Rusakov, I. G.; Negrier, S.; Ou,
with citric acid to provide tofacitinib citrate (XXVII) in 90% Y.-C.; Castellano, D.; Lim, H. Y.; Uemura, H.; Tarazi, J.; Cella, D.; Chen, C.;
yield.224 Rosbrook, B.; Kim, S.; Motzer, R. J. Lancet 2011, 378, 1931.
28. Kania, R. S.; Bender, S. L.; Borchardt, A. J.; Braganza, J. F.; Cripps, S. J.; Hua, Y.;
Johnson, M. D.; Johnson, T. O., Jr.; Luu, H. T.; Palmer, C. L.; Reich, S. H.;
29. Vismodegib (ErivedgeÒ) Tempczyk-russell, A. M.; Teng, M.; Thomas, C.; Varney, M. D.; Wallace, M. B.
WO Patent 01/02369 A2, 2001.
29. Kania, R. S.; Bender, S. L.; Borchardt, A. J.; Cripps, S. J.; Palmer, C. L.; Tempczyk-
Until the approval of Genentech’s visomodegib, the only treat- russell, A. M.; Varney, M. D.; Collins, M. R. US Patent 6531491 B1, 2003.
ment options for patients with advanced or metastatic basal-cell 30. Bender, S.; Kania, R.; Mctigue, M.; Palmer, C.; Pinko, C.; Wickersham, J. WO
Patent 2004/092217 A1, 2004.
carcinoma (BCC) were surgery or radiation therapy. Because it rep-
31. Flahive, E.; Ewanicki, B.; Yu, S.; Higginson, P. D.; Sach, N. W.; Morao, I. QSAR
resented a first-in-class treatment, vismodegib received priority Comb. Sci. 2007, 26, 679.
review from the FDA as the drug progressed through clinical trials. 32. Ewanicki, B. L.; Flahive, E. J.; Kasparian, A. J.; Mitchell, M. B.; Perry, M. D.;
The drug, which is marketed as ErivedgeÒ, derives its efficacy O’Neill-Slawecki, S. A.; Sach, N. W.; Saenz, J. E.; Shi, B.; Stankovic, N. S. US
Patent 2006/0094881 A1, 2006.
through inhibition of the hedgehog signaling pathway which exists 33. Ye, Q.; Hart, R. M.; Kania, R.; Ouellette, M.; Wu, Z. P.; Zook, S. E. US Patent
in most BCC’s.225,226 While there are several patents that cover the 2006/0094763 A1, 2006.
synthesis of vismodegib,227–229 the scale route has been published, 34. Babu, S.; Dagnino Jr, R.; Ouellette, M.; Shi, B.; Tian, Q.; Zook, S. WO Patent
2006/048745 A1, 2006.
and this route is depicted in Scheme 33.230 35. Friesen, D. T.; Lorenz, D. A.; Smith, S. W. WO Patent 2006/123223 A1, 2006.
The synthesis began with selective iodination of commercial 36. Flahive, E. J.; Ewanicki, B. L.; Sach, N. W.; O’Neill-Slawecki, S. A.; Stankovic, N.
carboxylic acid 179, affording trisubstituted arene 180 in 73% yield. S.; Yu, S.; Guinness, S. M.; Dunn, J. Org. Process Res. Dev. 2008, 12, 637.
37. Ewanicki, B. L.; Flahive, E. J.; Kasparian, A. J.; Mitchell, M. B.; Perry, M. D.;
A Curtius reaction then converted 180 to carbamate 181 in 84% O’Neill-Slawecki, S. A.; Sach, N. W.; Saenz, J. E.; Shi, B.; Stankovic, N. S.;
yield, and this was followed by a palladium(0)-catalyzed boryla- Srirangam, J. K.; Tian, Q.; Yu, S. EP Patent 2163544 A1, 2010.
tion of 181 which furnished Suzuki coupling partner 182 in 91% 38. Chekal, B. P.; Guinness, S. M.; Lillie, B. M.; McLaughlin, R. W.; Palmer, C. W.;
Post, R. J.; Sieser, J. E.; Singer, R. A.; Sluggett, G. W.; Vaidyanathan, R.;
yield. Pinacol borane 182 was exposed to commercial 2-bromopyr- Withbroe, G. J. Org. Process Res. Dev. 2013.
idine under conventional cross-coupling conditions to furnish biar- 39. Buclin, T.; Biollaz, J.; Kung, S.; Appenzeller, M.; Nussberger, J.; Higgins, T.;
yl 183, which underwent Boc-deprotection in quantitative Obayashi, M.; Brunner, H. R. Clin. Pharmacol. Ther. 1995. Westline Industrial
Dr, St Louis, MO 63146-3318, Abstract 204.
conversion to generate 184. Amide bond formation with acid chlo-
40. Ojima, M.; Igata, H.; Tanaka, M.; Sakamoto, H.; Kuroita, T.; Kohara, Y.; Kubo,
ride 185 (readily available from the corresponding commercial K.; Fuse, H.; Imura, Y.; Kusumoto, K.; Nagaya, H. J. Pharmacol. Exp. Ther. 2011,
acid)231 produced vismodegib (XXVIII) in 99% yield. 336, 801.
2030 H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032

41. Naka, T.; Inada, Y. US Patent 5583141 A, 1996. 84. Tanaka, K.; Shimotori, T.; Makino, S.; Aikawa, Y.; Inaba, T.; Yoshida, C.; Takano,
42. Naka, T.; Inada, Y. EP Patent 0520423 A2, 2003. S. Arzneim.-Forsch. 1992, 42, 935.
43. Rádl, S.; Cerný, J.; Stach, J.; Gablíková, Z. Org. Process Res. Dev. 2013, 17, 77. 85. Tanaka, K.; Makino, S.; Shimotori, T.; Aikawa, Y.; Inaba, T.; Yoshida, C.
44. Jones, D. Nat. Rev. Drug Disc. 2013, 12, 175. Arzneim.-Forsch. 1992, 42, 945.
45. Van Gestel, J. F. E.; Guillemont, J. E. G.; Venet, M. G.; Poignet, H. J. J.; Decrane, L. 86. Takano, S.; Yoshida, C.; Inaba, T.; Tanaka, K.; Takeno, R.; Nagaki, H.; Shimotori,
F. B.; Vernier, D. F. J.; Odds, F. C. US Patent 2005/0148581 A1, 2005. T.; Makino, S. US Patent 4954518 A, 1990.
46. Saga, Y.; Motoki, R.; Makino, S.; Shimizu, Y.; Kanai, M.; Shibasaki, M. J. Am. 87. Inaba, T.; Tanaka, K.; Takeno, R.; Nagaki, H.; Yoshida, C.; Takano, S. Chem.
Chem. Soc. 2010, 132, 7905. Pharm. Bull. 2000, 48, 131.
47. Shibazaki, M.; Kanai, M.; Saga, H. JP patent 2011–168519 A, 2011. 88. Guo, Z. R. Chin. J. New Drugs 2012, 21, 223.
48. Porstmann, F. R.; Horns, S.; Bader, T. WO Patent 2006/125769 A1, 2006. 89. Chen, X. H.; Bai, J. Y.; Shen, F.; Bai, A. P.; Guo, Z. R.; Cheng, G. F. Acta Pharmacol.
49. Hegyi, J. F., Alexandre, Lucas; Aelterman, W., Albert, Alex; Lang, Y. L.; Sin. 2004, 25, 927.
Stokbroekx, S., Carl, Maria; Leys, C.; Van Remoortere, P., Jozef, Maria; Faure, A. 90. Bai, A. P.; Guo, Z. R.; Hu, W. H.; Shen, F.; Cheng, G. F. Chin. Chem. Lett. 2001, 12,
WO Patent 2008/068231 A1, 2008. 775.
50. Keller-von Amsberg, G.; Koschmieder, S. OncoTargets Ther. 2013, 6, 99. 91. Guo, Z.; Cheng, G.; Chu, F.; Yang, G.; Xu, B. CN Patent 1134413 C, 2001.
51. Golas, J. M.; Arndt, K.; Etienne, C.; Lucas, J.; Nardin, D.; Gibbons, J.; Frost, P.; Ye, 92. Guo, Z.; Cheng, G.; Chu, F. US Patent 2004/0029951 A1, 2004.
F.; Boschelli, D. H.; Boschelli, F. Cancer Res. 2003, 63, 375. 93. Zhang, F. Y.; Shen, X. M.; Sun, P. Y. CN Patent 102206178 A, 2011.
52. Boschelli, D. H.; Wang, Y. D.; Johnson, S.; Wu, B.; Ye, F.; Sosa, A. C. B.; Golas, J. 94. Keating, G. M. Drugs 2012, 72, 2397.
M.; Boschelli, F. J. Med. Chem. 2004, 47, 1599. 95. Aylward, J. H.; Parsons, P. G.; Suhrbier, A.; Turner, K. A. US Patent 7449492 B2,
53. Khoury, H. J.; Cortes, J. E.; Kantarjian, H. M.; Gambacorti-Passerini, C.; 2008.
Baccarani, M.; Kim, D.-W.; Zaritskey, A.; Countouriotis, A.; Besson, N.; Leip, E.; 96. Liang, X.; Grue-Sørensen, G.; Petersen, A. K.; Högberg, T. Synlett 2012, 2647.
Kelly, V.; Brummendorf, T. H. Blood 2012, 119, 3403. 97. Liang, X.; Grue-Sørensen, G.; Mansson, K.; Vedsø, P.; Soor, A.; Stahlhut, M.;
54. Cortes, J. E.; Kantarjian, H. M.; Brummendorf, T. H.; Kim, D.-W.; Turkina, A. G.; Bertelsen, M.; Engell, K. M.; Högberg, T. Bioorg. Med. Chem. Lett. 2013, 23,
Shen, Z.-X.; Pasquini, R.; Khoury, H. J.; Arkin, S.; Volkert, A.; Besson, N.; Abbas, 5624.
R.; Wang, J.; Leip, E.; Gambacorti-Passerini, C. Blood 2011, 118, 4567. 98. Jørgensen, L.; McKerrall, S. J.; Kuttruff, C. A.; Ungeheuer, F.; Felding, J.; Baran,
55. Keller, G.; Schafhausen, P.; Bruemmendorf, T. H. Recent Results Cancer Res. P. S. Science (New York, N.Y.) 2013, 341, 878.
2010, 184, 119. 99. Winkler, J. D.; Lee, E. C. Y.; Nevels, L. I. Org. Lett. 2005, 7, 1489.
56. Yin, X. J.; Xu, G. H.; Sun, X.; Peng, Y.; Ji, X.; Jiang, K.; Li, F. Molecules 2010, 15, 100. Kuwajima, I.; Tanino, K. Chem. Rev. 2005, 105, 4661.
4261. 101. Nickel, A.; Maruyama, T.; Tang, H.; Murphy, P. D.; Greene, B.; Yusuff, N.;
57. Li, F.; Yin, X. J.; Jiang, K.; Sun, X.; Xu, G. H. CN Patent 101792416 A, 2010. Wood, J. L. J. Am. Chem. Soc. 2004, 126, 16300.
58. Sutherland, K. W.; Feigelson, G. B.; Boschelli, D. H.; Blum, D. M.; Strong, H. L. 102. Tanino, K.; Onuki, K.; Asano, K.; Miyashita, M.; Nakamura, T.; Takahashi, Y.;
US Patent 2005/0043537 A1, 2005. Kuwajima, I. J. Am. Chem. Soc. 2003, 125, 1498.
59. Withbroe, G. J.; Seadeek, C.; Girard, K. P.; Guinness, S. M.; Vanderplas, B. C.; 103. Winkler, J. D.; Rouse, M. B.; Greaney, M. F.; Harrison, S. J.; Jeon, Y. T. J. Am.
Vaidyanathan, R. Org. Process Res. Dev. 2013, 17, 500. Chem. Soc. 2002, 124, 9726.
60. Hart, C. D.; De Boer, R. H. OncoTargets Ther. 2013, 6, 1. 104. Winkler, J. D.; Harrison, S. J.; Greaney, M. F.; Rouse, M. B. Synthesis 2002, 2002,
61. Traynor, K. Am. J. Health Syst. Pharm. 2013, 70, 88. 2150.
62. Bentzien, F.; Zuzow, M.; Heald, N.; Gibson, A.; Shi, Y.; Goon, L.; Yu, P.; Engst, S.; 105. Wood, J. L.; Tang, H. Abstracts of Papers, 221st ACS National Meeting, San
Zhang, W.; Huang, S.; Zhao, L.; Vysotskaia, V.; Chu, F.; Bautista, R.; Cancilla, B.; Diego, CA, United States, April 1–5, 2001, ORGN.
Lamb, P.; Joly, A. H.; Yakes, F. M. Thyroid 2013. 106. Tanino, K.; Onuki, K.; Asano, K.; Miyashita, M.; Kuwajima, I. Conference
63. Bannen, L. C.; Chan, D. S.-M.; Chen, J.; Dalrymple, L. E.; Forsyth, T. P.; Huynh, T. proceedings from the 43rd Tennen Yuki Kagobutsu Toronkai Koen Yoshishu,
P.; Jammalamadaka, V.; Khoury, R. G.; Leahy, J. W.; Mac, M. B.; Mann, G.; 2001, 85.
Mann, L. W.; Nuss, J. M.; Parks, J. J.; Takeuchi, C. S.; Wang, Y.; Xu, W. WO 107. Tang, H.; Yusuff, N.; Wood, J. L. Org. Lett. 2001, 3, 1563.
Patent 2005/030140 A2, 2005. 108. Kigoshi, H.; Suzuki, Y.; Aoki, K.; Uemura, D. Tetrahedron Lett. 2000, 41, 3927.
64. St Clair Brown, A.; Lamb, P.; Gallagher, W. P. WO Patent 2010/083414 A1, 109. Kim, S.; Winkler, J. D. Chem. Soc. Rev. 1997, 26, 387.
2010. 110. Winkler, J. D.; Hong, B. C.; Bahador, A.; Kazanietz, M. G.; Blumberg, P. M.
65. Wilson, J. A. WO Patent 2012/109510 A1, 2012. Bioorg. Med. Chem. Lett. 1993, 3, 577.
66. Wilson, J. A.; Naganathan, S.; Pfeiffer, M.; Andersen, N. G. WO Patent 2013/ 111. Rigby, J. H.; Moore, T. L.; Rege, S. J. Org. Chem. 1986, 51, 2398.
059788 A1, 2013. 112. Satoh, T.; Kaneko, Y.; Okuda, T.; Uwaya, S.; Yamakawa, K. Chem. Pharm. Bull.
67. Kim, K. B.; Crews, C. M. Nat. Prod. Rep. 2013, 30, 600. 1984, 32, 3452.
68. Davies, S.; Pandian, R.; Bolos, J.; Castaner, R. Drugs Future 2009, 34, 708. 113. Seip, E. H.; Hecker, E. Planta Med. 1982, 46, 215.
69. Sin, N.; Kim, K. B.; Elofsson, M.; Meng, L.; Auth, H.; Kwok, B. H. B.; Crews, C. M. 114. Grue-Sørensena, G.; Liang, X.; Högberga, T.; Mansson, K.; Vedsø, P.; Vifian, T.
Bioorg. Med. Chem. Lett. 1999, 9, 2283. WO Patent 2012/083953 A1, 2012.
70. Elofsson, M.; Splittgerber, U.; Myung, J.; Mohan, R.; Crews, C. M. Chem. Biol. 115. Sorg, B.; Hecker, E. Z. Naturforsch., B: Anorg. Chem. Org. Chem. 1982, 37B, 1640.
1999, 6, 811. 116. FDA approves Kalydeco to treat rare form of cystic fibrosis, http://www.fda.gov/
71. Phiasivongsa, P.; Sehl, L. C.; Fuller, W. D.; Laidig, G. J. WO Patent 2009/045497 NewsEvents/Newsroom/PressAnnouncements/ucm289633.htm, [Access Date:
A1, 2009. 2012-Jan-31].
72. Laidig, G. J.; Radel, P. A.; Smyth, M. S. US Patent 2005/0256324 A1, 2005. 117. Van Goor, F.; Hadida, S.; Grootenhuis, P. D.; Burton, B.; Stack, J. H.; Straley, K.
73. Ptaszynska, A.; Chalamandaris, A.-G.; Sugg, J.; Johnsson, K.; Parikh, S.; List, J. F. S.; Decker, C. J.; Miller, M.; McCartney, J.; Olson, E. R.; Wine, J. J.; Frizzell, R. A.;
European Association for the Study of Diabetes Annual Meeting 2012 (48th), Ashlock, M.; Negulescu, P. A. Proc. Natl. Acad. Sci. U.S.A. 2011, 108, 18843.
2012, Berlin, German, Abstract 242. 118. Eckford, P. D. W.; Li, C.; Ramjeesingh, M.; Bear, C. E. J. Biol. Chem. 2012, 287,
74. Meng, W.; Ellsworth, B. A.; Nirschl, A. A.; McCann, P. J.; Patel, M.; Girotra, R. N.; 36639.
Wu, G.; Sher, P. M.; Morrison, E. P.; Biller, S. A.; Zahler, R.; Deshpande, P. P.; 119. Van Goor, F. F.; Burton, W. L. WO Patent 2011/050325 A1, 2011.
Pullockaran, A.; Hagan, D. L.; Morgan, N.; Taylor, J. R.; Obermeier, M. T.; 120. Hadida Ruah, S. S.; Hazlewood, A. R.; Grootenhuis, P. D. J.; Van Goor, F. F.;
Humphreys, W. G.; Khanna, A.; Discenza, L.; Robertson, J. G.; Wang, A.; Han, Singh, A. K.; Zhou, J.; Mccartney, J. WO Patent 2006/002421 A2, 2006.
S.; Wetterau, J. R.; Janovitz, E. B.; Flint, O. P.; Whaley, J. M.; Washburn, W. N. J. 121. Singh, A.; Van Goor, F.; Worley III, J. F.; Knapp, T. WO Patent 2007/075946 A1,
Med. Chem. 2008, 51, 1145. 2007.
75. Deshpande, P. P.; Ellsworth, B. A.; Singh, J.; Lai, C.; Crispino, G.; Randazzo, M. 122. Hurter, P. WO Patent 2007/079139 A2, 2007.
E.; Gougoutas, J. Z.; Denzel, T. W. WO Patent 2004/063209 A2, 2004. 123. Young, C. R.; Rowe, C. W. WO Patent 2007/134279 A2, 2007.
76. FDA approves new treatment for a type of late stage prostate cancer, http:// 124. Young, C. R.; Rowe, C. W. US Patent 2008/0090864 A1, 2008.
www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm317838.htm, 125. Demattei, J.; Feng, Y.; Harrison, C.; Looker, A.; Mudunuri, P.; Roeper, S.; Zhang,
[Access Date: 2012-Aug-31]. Y. WO Patent 2009/038683 A2, 2009.
77. Tran, C.; Ouk, S.; Clegg, N. J.; Chen, Y.; Watson, P. A.; Arora, V.; Wongvipat, J.; 126. Sheth, U.; Fanning, L. T. D.; Numa, M. M. D.; Binch, H.; Hurley, D.; Zhou, J.;
Smith-Jones, P. M.; Yoo, D.; Kwon, A.; Wasielewska, T.; Welsbie, D.; Chen, C. Hadida Ruah, S. S.; Hazlewood, A.; Silina, A.; Vairagoundar, R. US Patent 2010/
D.; Higano, C. S.; Beer, T. M.; Hung, D. T.; Scher, H. I.; Jung, M. E.; Sawyers, C. L. 0184739 A1, 2010.
Science (New York, N.Y.) 2009, 324, 787. 127. Demattei, J.; Looker, A. R.; Neubert-Langille, B.; Trudeau, M.; Roeper, S.; Ryan,
78. Sawyers, C. L.; Jung, M. E.; Chen, C. D.; Ouk, S.; Welsbie, D.; Tran, C.; M. P.; YAP, D. M. L.; Krueger, B. R.; Grootenhuis, P.; Vac Goor, F. F.; Botfield, M.
Wongvipat, J.; Yoo, D. WO Patent 2006/124118 A1, 2006. C.; Zlokarnik, G. WO Patent 2010/108162 A1, 2010.
79. Jung, M.; Yoo, D.; Sawyers, C. L.; Tran, C. US Patent 2007/0254933 A1, 2007. 128. Sheth, U.; Fanning, L. T. D.; Numa, M.; Binch, H.; Hurley, D. J.; Zhou, J.; Hadida
80. Jung, M. E.; Ouk, S.; Yoo, D.; Sawyers, C. L.; Chen, C.; Tran, C.; Wongvipat, J. J. Ruah, S. S.; Hazlewood, A. R.; Silina, A.; Vairagoundar, R.; Van Goor, F. F.;
Med. Chem. 2010, 53, 2779. Grootenhuis, P. D. J.; Botfield, M. C. WO Patent 2011/072241 A1, 2011.
81. Chen, Y. WO Patent 2013/087004 A1, 2013. 129. Arekar, S. G.; Johnston, S. C.; Krawiec, M.; Medek, A.; Mudunuri, P.; Sullivan,
82. Jain, R. P.; Angelaud, R.; Thompson, A.; Lamberson, C.; Greenfield, S. WO M. J. US Patent 2011/0230519 A1, 2011.
Patent 2011/106570 A1, 2011. 130. Van Goor, F.; Alargova, R. G.; Alcacio, T. E.; Arekar, S. G.; Johnston, S. C.;
83. Eisai and Toyama Chemical Receive Approval to Market Anti-rheumatic Agent Kadiyala, I. N.; Keshavarz-Shokri, A.; Krawiec, M.; Lee, E. C.; Medek, A. WO
Iguratimod in Japan, 2012, http://www.eisai.com/news/news201239.html, Patent 2011/133951 A1, 2011.
[Access Date: 2012-July-29]. 131. Van Goor, F.; Alargova, R. G.; Alcacio, T. E.; Arekar, S. G.; Binch, H. M.; Botfield,
M. C.; Fanning, L. T. D.; Grootenhuis, P. D. J.; Hurley, D. J.; Johnson, S. C.;
H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032 2031

Kadiyala, I. N.; Keshavarz-Shokri, A.; Krawiec, M.; Lee, E. C.; Luisi, B.; Medek, Loriaux, M. M.; Corbin, A. S.; Wardwell, S.; Ning, Y.; Keats, J. A.; Wang, Y.;
A.; Mufunuti, P.; Numa, M.; Sheth, U.; Silina, A.; Sullivan, M. J.; Verwijs, M. J.; Sundaramoorthi, R.; Thomas, M.; Zhou, D.; Snodgrass, J.; Commodore, L.;
Yang, X.; Young, C. R.; Zaman, N. T.; Zhang, B.; Zhang, Y.; Zlokarnik, G. WO Sawyer, T. K.; Dalgarno, D. C.; Deininger, M. W. N.; Druker, B. J.; Clackson, T.
Patent 2011/133953 A1, 2011. Cancer Cell 2009, 16, 401.
132. Morgan, A. J. WO Patent 2012/158885 A1, 2012. 175. Zhou, T.; Commodore, L.; Huang, W.-S.; Wang, Y.; Thomas, M.; Keats, J.; Xu,
133. Van Goor, F. F. WO Patent 2013/067410 A1, 2013. Q.; Rivera, V. M.; Shakespeare, W. C.; Clackson, T.; Dalgarno, D. C.; Zhu, X.
134. Xu, Y.; Wang, J.; He, G.; Lu, J. CN Patent 103044263 A, 2013. Chem. Biol. Drug Des. 2010, 77, 1.
135. Smith, B. M.; Smith, J. M.; Tsai, J. H.; Schultz, J. A.; Gilson, C. A.; Estrada, S. A.; 176. Gozgit, J. M.; Wong, M. J.; Wardwell, S.; Tyner, J. W.; Loriaux, M. M.;
Chen, R. R.; Park, D. M.; Prieto, E. B.; Gallardo, C. S.; Sengupta, D.; Dosa, P. I.; Mohemmad, Q. K.; Narasimhan, N. I.; Shakespeare, W. C.; Wang, F.; Druker, B.
Covel, J. A.; Ren, A.; Webb, R. R.; Beeley, N. R. A.; Martin, M.; Morgan, M.; J.; Clackson, T.; Rivera, V. M. Mol. Cancer Ther. 2011, 10, 1028.
Espitia, S.; Saldana, H. R.; Bjenning, C.; Whelan, K. T.; Grottick, A. J.; Menzaghi, 177. Huang, W.-S.; Metcalf, C. A.; Sundaramoorthi, R.; Wang, Y.; Zou, D.; Thomas,
F.; Thomsen, W. J. J. Med. Chem. 2008, 51, 305. R. M.; Zhu, X.; Cai, L.; Wen, D.; Liu, S.; Romero, J.; Qi, J.; Chen, I.; Banda, G.;
136. Fitzgerald, L. W.; Burn, T. C.; Brown, B. S.; Patterson, J. P.; Corjay, M. H.; Lentini, S. P.; Das, S.; Xu, Q.; Keats, J.; Wang, F.; Wardwell, S.; Ning, Y.;
Valentine, P. A.; Sun, J. H.; Link, J. R.; Abbaszade, I.; Hollis, J. M.; Largent, B. L.; Snodgrass, J. T.; Broudy, M. I.; Russian, K.; Zhou, T.; Commodore, L.;
Hartig, P. R.; Hollis, G. F.; Meunier, P. C.; Robichaud, A. J.; Robertson, D. W. Mol. Narasimhan, N. I.; Mohemmad, Q. K.; Iuliucci, J.; Rivera, V. M.; Dalgarno, D.
Pharmacol. 2000, 57, 75. C.; Sawyer, T. K.; Clackson, T.; Shakespeare, W. C. J. Med. Chem. 2010, 53, 4701.
137. Wang, Y.; Serradell, N.; Bolos, J. Drugs Future 2007, 32, 766. 178. Zou, D.; Huang, W.-S.; Thomas, R. M.; Romero, J. A. C.; Qi, J.; Wang, Y.; Zhu, X.;
138. Smith, B. M.; Smith, J. M.; Tsai, J. H.; Schultz, J. A.; Gilson, C. A.; Estrada, S. A.; Shakespeare, W. C.; Sundaramoorthi, R.; Metcalf III, C. A.; Dalgarno, D. C.;
Chen, R. R.; Park, D. M.; Prieto, E. B.; Gallardo, C. S.; Sengupta, D.; Thomsen, W. Sawyer, T. K. WO Patent 2007/075869 A2, 2007.
J.; Saldana, H. R.; Whelan, K. T.; Menzaghi, F.; Webb, R. R.; Beeley, N. R. A. 179. Huang, W.-S.; River, V. M.; Clackson, T. P.; Shakespeare, W. C.; Squillace, R. M.;
Bioorg. Med. Chem. Lett. 2005, 15, 1467. Gozgit, J. M. WO Patent 2011/053938 A1, 2011.
139. Weigl, U.; Porstmann, F.; Straessler, C.; Ulmer, L.; Koetz, U. WO Patent 2007/ 180. Shakespeare, W. C.; Haluska, F. G. WO Patent 2012/139027 A1, 2012.
120517 A2, 2007. 181. Droppert, P. In Biotech Strategy Blog: http://biotechstrategyblog.com/2012/01/
140. Kuroda, J.; Kamitsuji, Y.; Kimura, S.; Ashihara, E.; Kawata, E.; Nakagawa, Y.; radotinib-approved-in-south-korea-for-cml.html/, 2012.
Takeuichi, M.; Murotani, Y.; Yokota, A.; Tanaka, R.; Andreeff, M.; Taniwaki, M.; 182. Radotinib hydrochloride http://www.cancer.gov/drugdictionary?cdrid=
Maekawa, T. Int. J. Hematol. 2008, 87, 507. 723999.
141. Legros, L.; Hayette, S.; Nicolini, F. E.; Raynaud, S.; Chabane, K.; Magaud, J. P.; 183. Davies, S.; Bolos, J.; Serradell, N.; Bayes, M. Drugs Future 2007, 32, 17.
Cassuto, J. P.; Michallet, M. Leukemia 2007, 21, 2204. 184. Kim, D.-Y.; Cho, D.-J.; Lee, G.-Y.; Kim, H.-Y.; Woo, S.-H.; Kim, Y.-S.; Lee, S.-A.;
142. Chen, Y.; Hu, Y.; Michaels, S.; Segal, D.; Brown, D.; Li, S. Leukemia 2009, 23, Han, B.-C. WO Patent 2007/018325 A1, 2007.
1446. 185. Kim, D. Y.; Cho, D. J.; Lee, G. Y.; Kim, H. Y.; Woo, S. H. WO Patent 2010/018895
143. Wuilleme-Toumi, S.; Robillard, N.; Gomez, P.; Moreau, P.; Le Gouill, S.; Avet- A1, 2010.
Loiseau, H.; Harousseau, J. L.; Amiot, M.; Bataille, R. Leukemia 2005, 19, 1248. 186. Delorme, D.; Vaisburg, A.; Moradei, O.; Leit, S.; Raeppel, S.; Frechette, S.;
144. Paudler, W. W.; Kerley, G. I.; McKay, J. J. Org. Chem. 1963, 28, 2194. Bouchain, G.; Zhou, Z.; Paquin, I.; Gaudette, F.; Isakovic, L. WO Patent 2005/
145. Powell, R. G.; Rogovin, S. P.; Smith, C. R., Jr. Ind. Eng. Chem. Prod. Res. Dev. 1974, 092899 A1, 2005.
13, 129. 187. Breitenstein, W.; Furet, P.; Jacob, S.; Manley, P. W. WO Patent 2004/005281
146. Powell, R. G.; Weisleder, D.; Smith, C. R., Jr. J. Pharm. Sci. 1972, 61, 1227. A1, 2004.
147. Powell, R. G.; Weisleder, D.; Smith, C. R., Jr.; Rohwedder, W. K. Tetrahedron 188. Mullard, A. Nat. Rev. Drug Disc. 2013, 12, 87.
Lett. 1970, 815. 189. Bayer’s StivargaÒ (regorafenib) Tablets Approved by U.S. FDA for Treatment
148. Abdelkafi, H.; Nay, B. Nat. Prod. Rep. 2012, 29, 845. of Patients with Locally Advanced, Unresectable or Metastatic GIST, http://
149. Li, G. P.; Wu, X. P. CN Patent 1493572 A, 2004. www.onyx.com/view.cfm/662/bayers-stivarga-regorafenib-tablets-approved-
150. Liu, Y. US Patent 2010/0240887 A1, 2010. by-us-fda-for-treatment-of-patients-with-locally-advanced-unresectable-or-
151. Robin, J.-P.; Robin, J.; Cavoleau, S.; Chauviat, L.; Charbonnel, S.; Dhal, R.; metastatic-gist, [Access Date: 2013-Feb-25].
Dujardin, G.; Fournier, F.; Gilet, C.; Girodier, L.; Mevelec, L.; Poutot, S.; Rouaud, 190. Marrari, A.; George, S. Drugs Future 2011, 36, 17.
S. WO Patent 99/48894 A1, 1999. 191. Strumberg, D.; Schultheis, B. Expert Opin. Invest. Drugs 2012, 21, 879.
152. Robin, J.-P.; Radosevic, N.; Blanchard, J. WO Patent 2010/103405 A2, 2010. 192. Wilhelm, S.; Dumas, J.; Ladouceur, G.; Lynch, M.; Scott, W. J. WO Patent 2004/
153. Robin, J.-P.; Blanchard, J.; Chauviat, L.; Dhal, R.; Marie, J.-P.; Radosevic, N. US 113274 A2, 2004.
Patent 2005/0090484 A1, 2005. 193. Dumas, J.; Boyer, S.; Riedl, B.; Wilhelm, S. WO Patent 2005/009961 A2, 2005.
154. Kvols, L. K.; Oberg, K. E.; O’Dorisio, T. M.; Mohideen, P.; de Herder, W. W.; 194. Stiehl, J.; Heilmann, W.; Logers, M.; Rehse, J.; Gottfried, M.; Wichmann, S. WO
Arnold, R.; Hu, K.; Zhang, Y.; Hughes, G.; Anthony, L.; Wiedenmann, B. Endocr. Patent 2011/128261 A1, 2011.
Relat. Cancer 2012, 19, 657. 195. Said, G.; Grippon, S.; Kirkpatrick, P. Nat. Rev. Drug Disc. 2012, 11, 185.
155. Petersenn, S.; Unger, N.; Hu, K.; Weisshaar, B.; Zhang, Y.; Bouillaud, E.; 196. de Lartigue, J. Drugs Today (Barc) 2012, 48, 331.
Resendiz, K. H.; Wang, Y.; Mann, K. J. Clin. Pharmacol. 2012, 52, 1017. 197. Razavi, H.; Palaninathan, S. K.; Powers, E. T.; Wiseman, R. L.; Purkey, H. E.;
156. Lewis, I.; Bauer, W.; Albert, R.; Chandramouli, N.; Pless, J.; Weckbecker, G.; Mohamedmohaideen, N. N.; Deechongkit, S.; Chiang, K. P.; Dendle, M. T. A.;
Bruns, C. J. Med. Chem. 2003, 46, 2334. Sacchettini, J. C.; Kelly, J. W. Angew. Chem., Int. Ed. 2003, 42, 2758.
157. Mergler, M.; Tanner, R.; Gosteli, J.; Grogg, P. Tetrahedron Lett. 1988, 29, 4005. 198. Yamamoto, T.; Muto, K.; Komiyama, M.; Canivet, J.; Yamaguchi, J.; Itami, K.
158. Shvarts, V.; Chung, S. Expert Rev. Neurother. 2013, 13, 131. Chem. Eur. J. 2011, 17, 10113.
159. Rektor, I. Expert Opin. Pharmacother. 2013, 14, 225. 199. Wu, G.; Zhou, J.; Zhang, M.; Hu, P.; Su, W. Chem. Commun. 2012, 8964.
160. McElhinny, C. J., Jr.; Carroll, F. I.; Lewin, A. H. Synthesis 2012, 44, 57. 200. Labaudiniere, R. F.; O’Neill, M. H. WO Patent 2013/038351 A1, 2013.
161. Koyakumaru, K.; Matsuo, Y.; Satake, Y. WO Patent 2004/009553 A1, 2004. 201. Kelly, J. W.; Sekijima, Y. WO Patent 2004/056315 A2, 2004.
162. Arimoto, I.; Nagato, S.; Sugaya, Y.; Urawa, Y.; Ito, K.; Naka, H.; Omae, T.; 202. Bulawa, C. E.; Connelly, S.; DeVit, M.; Wang, L.; Weigel, C.; Fleming, J. A.;
Kayano, A.; Nishiura, K. US Patent 2007/0142640 A1, 2007. Packman, J.; Powers, E. T.; Wiseman, R. L.; Foss, T. R.; Wilson, I. A.; Kelly, J. W.;
163. Pixuvri, 2012, http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/ Labaudiniere, R. Proc. Natl. Acad. Sci. U.S.A. 2012, 109, 9629.
human/medicines/002055/human_med_001549.jsp&mid=WC0b01ac058001d124, 203. Yoshida, T.; Akahoshi, F.; Sakashita, H.; Kitajima, H.; Nakamura, M.; Sonda, S.;
[Access Date: 2012-May-31]. Takeuchi, M.; Tanaka, Y.; Ueda, N.; Sekiguchi, S.; Ishige, T.; Shima, K.; Nabeno,
164. Cavalletti, E.; Crippa, L.; Mainardi, P.; Oggioni, N.; Cavagnoli, R.; Bellini, O.; M.; Abe, Y.; Anabuki, J.; Soejima, A.; Yoshida, C.; Takashina, Y.; Ishii, S.; Kiuchi,
Sala, F. Invest. New Drugs 2007, 25, 187. S.; Fukudaa, S.; Tsutsumiuchi, R.; Kosaka, K.; Murozono, T.; Nakamaru, Y.;
165. Beaven, A. W.; Rizzieri, D. Clin. Invest. (London, U. K.) 2012, 2, 49. Utsumi, H.; Masutomi, N.; Kishida, H.; Miyaguchi, I.; Hayashi, Y. Biorg. Med.
166. Mukherji, D.; Pettengell, R. Expert Opin. Pharmacother. 2010, 11, 1915. Chem. 2012, 5, 69.
167. Evison, B. J.; Mansour, O. C.; Menta, E.; Phillips, D. R.; Cutts, S. M. Nucleic Acids 204. Abe, Y.; Anabuki, J.; Soejima, A.; Shimamura, K.; Hayashi, Y.; Sakai, K. Diabetes,
Res. 2007, 35, 3581. 65th Scientific Sessions (2005), 2005; San Diego, California, Abstract 1493.
168. de Isabella, P.; Palumbo, M.; Sissi, C.; Capranico, G.; Carenini, N.; Menta, E.; 205. Sakashit, H.; Akahoshi, F.; Kitajima, H.; Tsutsumiuchi, R.; Hayashi, Y. Biorg.
Oliva, A.; Spinelli, S.; Krapcho, A. P.; Giuliani, F. C.; Zunino, F. Mol. Pharmacol. Med. Chem. 2006, 14, 3662.
1995, 48, 30. 206. Rocklin, R. WO Patent 2012018704 A1, 2012.
169. Krapcho, A. P.; Petry, M. E.; Getahun, Z.; Landi, J. J., Jr.; Stallman, J.; Polsenberg, 207. Fox, R. I. J. Rheumatol. Suppl. 1998, 53, 20.
J. F.; Gallagher, C. E.; Maresch, M. J.; Hacker, M. P. J. Med. Chem. 1994, 37, 828. 208. Baumann, P.; Mandl-Weber, S.; Voelkl, A.; Adam, C.; Bumeder, I.; Oduncu, F.;
170. Krapcho, A. P.; Hacker, M. P.; Cavalletti, E.; Giuliani, F. C. US Patent 5587382 A, Schmidmaier, R. Mol. Cancer Ther. 2009, 8, 366.
1996. 209. Lolli, M. L.; Giorgis, M.; Tosco, P.; Foti, A.; Fruttero, R.; Gasco, A. Eur. J. Med.
171. Spinelli, S.; Didomenico, R. WO Patent 9526189 A1, 1995. Chem. 2012, 49, 102.
172. Krapcho, P. A. EP Patent 503537 A1, 1992. 210. Palmer, A. M. Curr. Opin. Invest. Drugs 2010, 11, 1313.
173. Cortes, J. E.; Kantarjian, H.; Shah, N. P.; Bixby, D.; Mauro, M. J.; Flinn, I.; O’Hare, 211. Williamson, R. A.; Yea, C. M.; Robson, P. A.; Curnock, A. P.; Gadher, S.;
T.; Hu, S.; Narasimhan, N. I.; Rivera, V. M.; Clackson, T.; Turner, C. D.; Haluska, Hambleton, A. B.; Woodward, K.; Bruneau, J.-M.; Hambleton, P.; Moss, D.;
F. G.; Druker, B. J.; Deininger, M. W. N.; Talpaz, M. N. Eng. J. Med. 2012, 367, Thomson, A.; Spinella-Jaegle, S.; Morand, P.; Courtin, O.; Saute, C.; Westwood,
2075. R.; Hercend, T.; Kuo, E. A.; Ruuth, E. J. Biol. Chem. 1995, 270, 22467.
174. O’Hare, T.; Shakespeare, W. C.; Zhu, X.; Eide, C. A.; Rivera, V. M.; Wang, F.; 212. Mulakayala, N.; Rao, P.; Iqbal, J.; Bandichhor, R.; Oruganti, S. Eur. J. Med. Chem.
Adrian, L. T.; Zhou, T.; Huang, W.-S.; Xu, Q.; Metcalf, C. A., III; Tyner, J. W.; 2013, 60, 170.
2032 H. X. Ding et al. / Bioorg. Med. Chem. 22 (2014) 2005–2032

213. Chen, G.; Sun, L. CN Patent 102786437 A, 2012. 222. Ruggeri, S. G.; Hawkins, J. M.; Makowski, T. M.; Rutherford, J. L.; Urban, F. J.
214. Deo, K.; Patel, S.; Dhol, S.; Sanghani, S.; Ray, V. WO Patent 2009/147624 A2, WO Patent 2007/012953 A2, 2007.
2009. 223. Cai, W.; Colony, J. L.; Frost, H.; Hudspeth, J. P.; Kendall, P. M.; Krishnan, A. M.;
215. Deo, K.; Patel, S.; Dhol, S.; Sanghani, S.; Ray, V. WO Patent 2010/013159 A1, Makowski, T.; Mazur, D. J.; Phillips, J.; Ripin, D. H. B.; Ruggeri, S. G.; Stearns, J.
2010. F.; White, T. D. Org. Process Res. Dev. 2005, 9, 51.
216. Metro, T.-X.; Bonnamour, J.; Reidon, T.; Sarpoulet, J.; Martinez, J.; Lamaty, F. 224. Price, K. E.; Larrivee-Aboussafy, C.; Lillie, B. M.; McLaughlin, R. W.; Mustakis,
Chem. Commun. 2012, 11781. J.; Hettenbach, K. W.; Hawkins, J. M.; Vaidyanathan, R. Org. Lett. 2009, 11,
217. Shi, J.; Zhang, Q.; Jin, Y.; Li, J. Chin. Pharm. J. 2008, 43, 1353. 2003.
218. Flanagan, M. E.; Blumenkopf, T. A.; Brissette, W. H.; Brown, M. F.; Casavant, J. 225. FDA approves new treatment for most common type of skin cancer, http://
M.; Shang-Poa, C.; Doty, J. L.; Elliott, E. A.; Fisher, M. B.; Hines, M.; Kent, C.; www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm289545.htm,
Kudlacz, E. M.; Lillie, B. M.; Magnuson, K. S.; McBride, C. E.; McCurdy, S. P.; [Access Date: 2012-Jan-30].
Munchhof, M. J.; Perry, B. D.; Sawyer, P. S.; Strelevitz, T. J.; Subramanyam, C.; 226. Sekulic, A.; Migden, M. R.; Oro, A. E.; Dirix, L.; Lewis, K. D.; Hainsworth, J. D.;
Sun, J.; Whipple, D. A.; Changelian, P. S. J. Med. Chem. 2010, 53, 8468. Solomon, J. A.; Yoo, S.; Arron, S. T.; Friedlander, P. A.; Marmur, E.; Rudin, C. M.;
219. Changelian, P. S.; Flanagan, M. E.; Ball, D. J.; Kent, C. R.; Magnuson, K. S.; Chang, A. L. S.; Low, J. A.; Mackey, H. M.; Yauch, R. L.; Graham, R. A.; Reddy, J.
Martin, W. H.; Rizzuti, B. J.; Sawyer, P. S.; Perry, B. D.; Brissette, W. H.; C.; Hauschild, A. N. Eng. J. Med. 2012, 366, 2171.
McCurdy, S. P.; Kudlacz, E. M.; Conklyn, M. J.; Elliott, E. A.; Koslov, E. R.; Fisher, 227. Gunzner, J. L.; Sutherlin, D.; Stanley, M. S.; Bao, L.; Castanedo, G. M.; Lalonde,
M. B.; Strelevitz, T. J.; Yoon, K.; Whipple, D. A.; Sun, J.; Munchhof, M. J.; Doty, J. R. L.; Wang, S.; Reynolds, M. E.; Savage, S. J.; Malesky, K.; Dina, M. S. US Patent
L.; Casavant, J. M.; Blumenkopf, T. A.; Hines, M.; Brown, M. F.; Lillie, B. M.; 2006/0063779 A1, 2006.
Subramanyam, C.; Shang-Poa, C.; Milici, A. J.; Beckius, G. E.; Moyer, J. D.; Su, 228. Gunzner, J. L.; Sutherlin, D.; Stanley, M. S.; Bao, L.; Castanedo, G. M.; Lalonde,
C.; Woodworth, T. G.; Gaweco, A. S.; Beals, C. R.; Littman, B. H.; Fisher, D. A.; R. L.; Wang, S.; Reynolds, M. E.; Savage, S. J.; Malesky, K.; Dina, M. S. WO
Smith, J. F.; Zagouras, P.; Magna, H. A.; Saltarelli, M. J.; Johnson, K. S.; Nelms, L. Patent 2009/126863 A2, 2009.
F.; Des Etages, S. G.; Hayes, L. S.; Kawabata, T. T.; Finco-Kent, D.; Baker, D. L.; 229. Cheng, D.; Han, D.; Zhang, G.; Wan, Y.; Xie, Y. F.; Jiang, J.; Gao, W.; Pan, S. WO
Larson, M.; Si, M.-S.; Paniagua, R.; Higgins, J.; Holm, B.; Reitz, B.; Zhou, Y.-J.; Patent 2010/027746 A2, 2010.
Morris, R. E.; O’Shea, J. J.; Borie, D. C. Science (New York, N.Y.) 2003, 302, 875. 230. Zhu, J.; Mao, J.; Yang, M.; Wu, X. CN Patent 102731373 A, 2012.
220. Blumenkopf, T. A.; Flanagan, M. E.; Munchhof, M. J. EP Patent 1235830 B1, 231. Neves, J.; Teixeira, L.; Bhatia, S.; Ermrich, M. WO Patent 2011/005127 A1,
2004. 2011.
221. Flanagan, M. E.; Li, Z. J. WO Patent 2003/048162 A1, 2003.

You might also like