You are on page 1of 10

FULL PAPER

DOI: 10.1002/ajoc.201200014

Practical Large-Scale Synthesis of the Hepatitis C Virus Protease Inhibitor


BI 201335

Carl A. Busacca,*[a] Xudong Wei,*[a] Nizar Haddad,[a] Suresh Kapadia,[a]


Jon. C. Lorenz,[a] Anjan K. Saha,[a] Richard J. Varsolona,[a] Thilo Berkenbusch,[b]
Scot C. Campbell,[a] Vittorio Farina,[a] XuWu Feng,[a] Nina C. Gonnella,[a]
Nelu Grinberg,[a] Paul-James Jones,[a] Heewon Lee,[a] Zhibin Li,[a] Oliver Niemeier,[b]
Wendelin Samstag,[b] Max Sarvestani,[a] Juergen Schroeder,[b] John Smoliga,[a]
Earl M. Spinelli,[a] Jana Vitous,[a] and Chris H. Senanayake[a]

Abstract: A concise synthetic route then required for the final assembly practical process is suitable for large
for large-scale manufacture of the of this complex target. The first step is scale production of BI 201335, re-
hepatitis C virus (HCV) protease in- a critical C O bond formation to join quires no protecting groups or chro-
hibitor BI 201335 has been developed. the heterocycle and the peptidic por- matography, and every isolated inter-
A convergent synthetic design was tion of the molecule, which was done mediate is a crystalline solid.
achieved by using three advanced in- by using a newly-designed heterocy-
termediates: a densely functionalized clic sulfone. The second step uses an
thiazole-quinoline, a hydroxyproline- inexpensive peptide coupling of the
Keywords: antiviral agents · BI
containing dipeptide acid, and an opti- aminocyclopropane and ester saponi-
201335 · HCV protease · nucleo-
cally pure vinylcyclopropane. Only fication, then recrystallization com-
philic substitution
three subsequent synthetic steps were pletes the route. This economical and

Introduction drug candidate was ultimately discontinued. This finding


then led to the identification of the nonmacrocyclic inhibi-
Hepatitis C virus (HCV) infection is believed to affect tor of HCV NS3 protease BI 201335,[3] a molecule which
about 3 % of the worlds population, and potential thera- possesses the key features of in vivo potency, safety, and
pies that involve attacking various components of the viral bioavailability needed for a HCV therapy. This inhibitor is
machinery is a very active research area. One of the most now in late-stage human clinical trials.[4] A coherent effort
promising targets has been HCV NS3 protease, an enzyme by a team of chemists was required to develop a route that
which is essential to viral replication.[1] The first small-mol- is suitable for large scale production and commercializa-
ecule inhibitor of this target was developed by Boehringer– tion.
Ingelheim with proof-of-principle in humans achieved with BI 201335 is an architecturally complex molecule with
the macrocycle BILN 2061.[2] However, development of this a molecular weight of nearly 900 amu and five stereogenic
centers. We required an economical and practical synthesis
of this important target, free of protecting groups, cryogenic
conditions, and chromatography. The retrosynthetic route
[a] C. A. Busacca, X. Wei, N. Haddad, S. Kapadia, J. C. Lorenz,
to BI 201335 that was ultimately developed is shown in
A. K. Saha, R. J. Varsolona, S. C. Campbell, V. Farina, X. Feng, Scheme 1. The assembly was performed with the three ad-
N. C. Gonnella, N. Grinberg, P.-J. Jones, H. Lee, Z. Li, vanced intermediates shown: dipeptide acid 1, aminoester
M. Sarvestani, J. Smoliga, E. M. Spinelli, J. Vitous, C. H. Senanayake cyclopropane[5] 2, and thiazole-quinoline 3. Unlike the
Chemical Development and Analytical Development
double SN2 approach used in the discovery synthesis,[4] the
Boehringer-Ingelheim Pharmaceuticals, Inc.
900 Ridgebury Rd. Ridgefield, CT 06877 (USA) strategy for developing an efficient large-scale synthesis
Fax: (+ 1) 203-791-6130 was to join 1, derived from trans-hydroxyproline, and 3 by
E-mail: carl.busacca@boehringer-ingelheim.com a more straightforward SNAr reaction. The selection of the
xudong.wei@boehringer-ingelheim.com optimum leaving group (LG) in 3 had not been finalized at
[b] T. Berkenbusch, O. Niemeier, W. Samstag, J. Schroeder this early stage. The SNAr reaction would then be followed
Process Development Chemicals
Boehringer-Ingelheim Pharma GmbH
by a coupling with the more expensive building block 2,
Binger Strasse, 173, 55216 Ingelheim (Germany) thereby avoiding tedious transformations and greatly reduc-
Supporting information for this article is available on the WWW ing the waste generated.
under http://dx.doi.org/10.1002/ajoc.201200014.

80  2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim Asian J. Org. Chem. 2012, 1, 80 – 89
the coupling to effect the
saponification. The target
dipeptide acid 1 was then
crystallized directly after ad-
justing the pH value in 91 %
overall yield for the one-pot
sequence, which achieves all
of the process goals detailed
above.
The synthesis of “north-
ern” heterocyclic fragment 3
is shown in Scheme 3 a.
Lithiation of pivalimide 7
and subsequent bromination
with dibromotetrachloro-
Scheme 1. BI 201335 retrosynthesis. LG = leaving group. ethane gave the brominated
intermediate, which was
then hydrolyzed in situ with
Results and Discussion 6 n HCl to furnish the HCl salt 8 directly in 91 % overall
yield. A variety of brominating reagents could be used in
The synthesis commences with the development of a practi- the synthesis of 8, provided that the anion solution was
cal and economical route to 1, and a protecting-group-free added to the electrophile and the temperature was main-
process with isolation of crystalline intermediates was also tained at under 5 8C, though yields were not high. Table 1
highly preferred. The chloroformate 4, derived from cyclo- shows the results with different brominating reagents. 1,2-
pentanol, was prepared by using triphosgene in tetrahydro- Dibromoethane gave a promising 51 % yield of 8, although
furan (THF) with pyridine as a base (Scheme 2). Stability toxicity concerns with this reagent led to the search for
a better electrophile.
One of the most effective brominating reagents was bro-
moperfluorooctane, which does not appear to have been
previously applied to this type of transformation. Initial re-
search quantities of 8 were prepared in 81 % yield by using
this reagent, although its commercial availability at larger
scales later proved to be problematic. A search for a readily
available and economical reagent for this reaction was un-
dertaken. It was found that 1,2-dibromotetrachloroethane
was superior to all brominating reagents tested in this trans-
formation, and it was selected based upon its improved
yield, better atom economy, and ready availability on multi-
kilogram scale as a high melting point crystalline solid. One
caveat for the use of this electrophile was competing chlori-
nation, which required careful temperature control to
reduce the chlorinated impurity 9 (Scheme 4) in bromide 8
Scheme 2. Synthesis of dipeptide acid 1. to less than 0.5 %.
Considerable optimization was required for the subse-
quent Sugasawa[8] acylation step to install the methyl
studies with 4 showed little decomposition at approximately ketone. The conditions that were ultimately developed for
10 8C in the presence of hydroxide bases, and the coupling this Sugasawa acylation involve a combination of BCl3 and
with tert-leucine was thus achieved in a Schotten–Bau- AlCl3 (Scheme 3 a), and include several key parameters.
mann-type reaction without measurable racemization.[6] Direct use of the HCl salt 7 was successful, so that a salt
The capped tert-leucine 5 was obtained as a crystalline solid break to give the free base became unnecessary. Unwanted
in 87 % overall yield from cyclopentanol. cleavage of the methyl ether by the Lewis acids occurred
Although many different types of peptide-coupling proto- initially to give 13, yet the cleavage could be effectively
cols could be applied to 5, coupling with the hydroxyproline minimized by charging MeCN prior to addition of the
ester was conveniently carried out with the inexpensive ac- second Lewis acid (AlCl3), and by using only 0.5 equiva-
tivator tosyl chloride[7] and N-methylmorpholine (NMM) as lents. The use of only five equivalents of MeCN, rather
base. The intermediate ester 6 was not isolated, rather than an excess, helped to reduce the amount of the amidine
aqueous lithium hydroxide was charged at the conclusion of impurity 14 that was formed from MeCN. Several different

Asian J. Org. Chem. 2012, 1, 80 – 89  2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.AsianJOC.org 81
Carl A. Busacca, Xudong Wei et al.
FULL PAPER
The order of addition of
the reagents was crucial,
with sequential charges of
aniline HCl salt 8, BCl3,
MeCN, and then AlCl3
giving optimum results.
Under these carefully opti-
mized conditions, Sugasawa
product 9 was formed in
80 % isolated yield. Aniline
9 was then acylated with the
acid chloride derived from
thiazole acid 10, itself pre-
pared in two steps from
thiourea by a Hantzsch[9]
synthesis, as shown in
Scheme 3 b. The initial acy-
lation was most efficiently
performed without added
base of any kind. The inter-
mediate 11 was not isolated,
but was cyclized in situ to
quinolone 12 by using
tBuOK at 100 8C, then
chlorinated with POCl3 to
chloroquinoline 3 a (LG =
Scheme 3. a) Synthesis of the “northern” heterocycle. b) Synthesis of acid 10.
Cl) in 81 % yield.
The key consideration in
the assembly of BI 201335
Table 1. Screen of brominating reagents for the conversion of 7 into 8. was how to carry out the C O bond forming reaction be-
Brominating reagent Yield of 8 [%] tween the “southern” peptide and the “northern” heterocy-
C3H5Br trace cle. A double SN2 approach[4] had been successfully used
NBS 21 for related compounds, yet a direct SNAr reaction was
Br2 25 chosen at this juncture, as it is a more straightforward and
BnBr 44 less wasteful strategy. The SNAr reaction in turn required
BrC2H4Br 51
BrC8F17 81
the synthesis of a thiazole-quinoline bearing a leaving
BrACHTUNGRE(CCl2)2Br 91 group at the 4-position. Several leaving groups were exam-
NBS = N-bromosuccinimide; Bn = benzyl.
ined, and chloride was chosen for the first SNAr studies.
The initial research thus focused on 3 a, which could be pre-
pared from 12 by using POCl3.
The use of any peptide esters as nucleophiles in this reac-
tion led to very complex reaction mixtures, and reasonable
conversions were obtained only with the free carboxylic
acids. This may reflect partial racemization at the hydroxy-
Scheme 4. Bromide 8 and chloride impurity 9.
proline ester a position under the strongly basic conditions
of the reaction.[10] Another major SNAr side product was
Table 2. Optimization of the solvent for the Sugasawa reaction.
the quinolinyl methyl ether 16 (Scheme 5) formed by reac-
Solvent Yield of 8 [%]
tion of 3 a with methoxide generated from the decomposi-
PhMe 89 tion of the hydroxyproline methyl ester. Further optimiza-
PhCl 78
1,2-DCE 94
tion of the SNAr reaction between 3 a and 1 revealed that
PhF 93 the reaction could be performed at 18–22 8C with 4.5 equiv-
DCE = dichloroethane alents of tBuOK in dimethylsulfoxide (DMSO), and re-
quired about five hours to reach completion. In a process
solvents could be used (Table 2), yet fluorobenzene proved safety evaluation, however, this strong base/DMSO system
to be the optimal solvent, as a reaction temperature of proved to be hazardous. The decomposition onset tempera-
about 75 8C was needed for full conversion and the use of ture was found to be 120 8C for the reaction mixture, which
solvents with lower boiling points thus became problematic. was presumably as a result of the deprotonation of DMSO

82 www.AsianJOC.org  2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim Asian J. Org. Chem. 2012, 1, 80 – 89
Synthesis of HCV Protease Inhibitor

Scheme 6. Synthesis of sulfone 3 b.

Scheme 5. trans-Carbamoylation and ether impurities.


ing the use of excess POCl3 as chlorinating reagent, which
can present safety issues. Competition experiments between
sulfone 3 b and chloride 3 a were carried out. The sulfone
and further decomposition, possibly generating the methyl- gave a cleaner reaction and was also significantly more re-
ene carbene. Potassium dimethyloctanoxide (KDMO) has active than 3 a. Addition of acetic acid to the SNAr reaction
far greater stability in N,N-dimethylacetamide (DMAc) and mixture was beneficial. The unusual role of HOAc here
N,N-dimethylformamide (DMF) than in DMSO, yet when may be as a proton donor to the sulfonate leaving group
a less-polar solvent such as THF was used, the SNAr reac- oxygen or the quinoline nitrogen, thereby accelerating the
tion was sluggish and more complicated mixtures were ob- reaction rate. HOAc is an extremely weak acid in dipolar
tained. Additional base (ca. 4.8–5.0 equivalents total) was aprotic solvents relative to its acidity in water.[12] Complete
then needed to drive the reaction to completion, yet 5– protonation of the sulfinate nucleophile is not likely to
10 % of the quinolone impurity 12 was always formed, and occur in the solvent system used for the SNAr reaction.
this species was very difficult to remove by crystallization. The more economical base tBuOK could then again be
In the initial scale-up work, this sparingly soluble impurity used because of the greater reactivity of the sulfone in
had to be removed by filtration. a THF/DMF mixture. These optimized conditions com-
An additional problem associated with this process was bined with the superior sulfone coupling partner furnished
the formation of trans-carbamoylation species 17, the tert- SNAr adduct 20 in 86 % isolated yield at room temperature
butyl carbamate analogue of 20 (Scheme 5), in which the by using 3.8 equivalents of base, as shown in Scheme 7. The
cyclopentyl group was replaced by the tert-butyl group from intermediate was then crystallized from nPrOH after sol-
tBuOK. This species was present at levels of approximately vent switching prior to the final isolation.
5 %. The use of KDMO largely eliminated the trans-carba- The initial peptide coupling of acid 20 with the vinylcy-
moylation problem, as the corresponding DMO-carbamate clopropane aminoester 2 was carried out with 1-ethyl-3-(3-
18 was much more soluble and could be easily removed dimethylaminopropyl)carbodiimide (EDC) and N-hydroxy-
during the crystallization of 20. Applying 4.25 equivalents benzotriazole (HOBT), to give final intermediate 21 in
of KDMO in DMSO at room temperature gave the desired high yield. HOBT is relatively expensive and is also classi-
SNAr product 20 in 87 % isolated yield. Presumably all fied as shock-sensitive, which creates likely supply-chain
acidic protons on each coupling partner must be deproton- and transportation issues. We examined several HOBT sub-
ated for efficient SNAr reactions. stitutes,[13] yet detectable amounts of the diastereomer de-
Initial multikilogram quantities of BI 201335 were pre- rived from racemization of the proline stereocenter were
pared by using this sequence, yet a more acceptable solvent obtained. Isobutylchloroformate (IBCF) was identified as
than DMSO was desirable, as were an increase in the fun- an inexpensive activating reagent for this transformation[14]
damental reaction rate of the SNAr reaction and a more that could provide optically pure 20 provided that careful
economical base. control of the reaction temperature was maintained. Slow
Sulfones are known to be competent leaving groups for addition of NMM to 20 at 10 8C then slow addition of
nucleophilic substitution.[11] Crystalline sulfone 3 b was pre- IBCF and holding the reaction at 10 8C generated the acti-
pared from 12 by a one-pot tosylation (to give 19) and sub- vated intermediate. Peptide coupling was then achieved by
sequent sulfinate addition in 87 % yield, as shown in adding 2 to the intermediate described then adding of
Scheme 6. This transformation has the advantage of avoid- a second quantity of NMM while maintaining the reaction

Asian J. Org. Chem. 2012, 1, 80 – 89  2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.AsianJOC.org 83
Carl A. Busacca, Xudong Wei et al.
FULL PAPER
Acyl thiourea 15

Thiourea (15 kg, 197 mol, 1 equiv)


and toluene (150 L) were charged
to a 500 L glass-lined reactor. Iso-
butyryl chloride (21 kg, 197 mol,
1 equiv) was then added by a dia-
phragm pump over 2 h. The resul-
tant mixture was then gradually
heated to reflux, and significant
foaming of the batch was ob-
served. The batch was maintained
at reflux for 48 h, cooled to 60 8C,
then saturated NaHCO3 (45 kg)
was added. The mixture was then
cooled to 0 8C, left to stand for
12 h, and filtered. The filter cake
was washed with H2O (2  30 L),
then toluene (2  30 L), and the
filter cake was dried at 40 8C
under vacuum for 72 h to give
20.1 kg (70 %) of acyl thiourea 15
as a white solid; m.p. 110–111 8C.
1
H NMR (500 MHz, [D6]DMSO,
30 8C): d = 9.66 (br s, 1 H, N-H),
9.27 (br s, 1 H, N-H), 2.67 (septet,
J = 7 Hz, 1 H, C-H), 1.02 ppm (d,
J = 7 Hz, 6 H, Me). 13C NMR
Scheme 7. Synthesis of BI 201335.
(125 MHz, [D6]DMSO, 30 8C): d =
181.9 (s), 178.6 (s), 34.3 (d),
18.8 ppm (q). IR: ~ n = 651, 722,
temperature below 8 8C, and finally warming the reaction 881, 936, 1018, 1094, 1112, 1141, 1188, 1285, 1390, 1411, 1456, 1525,
to 10 8C. Ester 21 was then crystallized from denatured eth- 1601, 1701, 3168, 3310 cm 1. HRMS: m/z: calcd for C5H11N2OS:
147.0587 [M + H] + ; found: 147.0580; error = 4.497 ppm. Elemental anal-
anol and isolated. Under these optimized conditions, final
ysis calcd (%) for C5H10N2OS: C 41.07; H 6.89; N 19.16; found: C 40.90;
intermediate 21 was obtained in 88 % isolated yield with no H 7.11; N 18.99.
detectable racemization. Synthesis of the active pharma-
ceutical ingredient was then achieved by simple ester sapo- Thiazole acid 10
nification and crystallization, again from denatured ethanol, Acyl thiourea 15 (17.5 kg, 120 mol, 1 equiv) and dioxane (180 kg) were
in 85 % yield. Verification of the absolute configuration of charged to a 500 L reactor. Bromopyruvic acid (20 kg, 120 mol, 1 equiv)
all stereocenters was achieved through single crystal x-ray was then charged portionwise, which caused an exotherm to 40 8C. The
resultant slurry was heated to 100 8C and left to stand at that tempera-
structure analysis. ture for 3 h. The batch was concentrated to a final volume of 50 L under
vacuum, and then H2O (150 L) was charged. The resulting mixture was
agitated at 23 8C for 12 h, cooled to 0 8C, and then filtered. The resultant
Conclusions filter cake was then washed with H2O (3  125 L), until the filtrate
reached pH 3.0. The filter cake was dried in a vacuum oven for 72 h to
give 19.0 kg (74 %) of thiazole acid 10 as a white solid; m.p. 246–248 8C.
In summary, a multikilogram route to the new HCV drug 1
H NMR (500 MHz, [D6]DMSO): d = 7.95 (s, 1 H, Ar-H), 2.71 (septet,
BI 201335 has been developed. It is a robust and conver- J = 7 Hz, 1 H, C-H), 1.10 ppm (d, J = 7 Hz, 6 H, Me). 13C NMR
gent synthesis without the need for protecting groups or (125 MHz, [D6]DMSO, 30 8C): d = 175.7 (s), 162.4 (s), 157.9 (s), 141.9 (s),
chromatography. Each raw material used is inexpensive, all n = 655, 717, 733, 850, 868, 938, 981,
122.1 (d), 33.9 (d), 19.0 ppm (q). IR: ~
1095, 1105, 1149, 1190, 1222, 1292, 1350, 1416, 1461, 1552, 1699, 2508,
peptide couplings are achieved without racemization, and 2969, 3134 cm 1. HRMS: m/z: calcd for C8H11N2O3S: 215.0485 [M + H] + ;
every intermediate is produced as a single enantiomer or found: 215.0476; error = 4.141 ppm. Elemental analysis calcd (%) for
diastereomer. Each isolated intermediate is a stable crystal- C8H10BrN2O3S·0.25 H2O: C 43.93; H 4.84; N 12.81; found: C 43.68; H
line solid, and all process safety concerns were addressed 4.45; N 12.57.
by choosing inherently safer alternatives for scale-up. The Bromoanisidine HCl 8
chemistry described here has now been successfully per-
Part 1: Piv-m-anisidine 7 (2.5 kg, 12.1 mol, 1 equiv) and THF (17 L)
formed on large scale in chemical production to support
were charged to a 50 L reactor and the solution cooled to 25 8C.
the final clinical development phases of BI 201335. nBuLi/hexane (2.5 m, 12 L, 30 mol, 2.5 equiv) was then slowly charged
(< 10 8C). This mixture was agitated at ~ 15 8C for 3 h. In a separate
reactor dibromotetrachloroethane (5.9 kg, 18.2 mol, 1.5 equiv) and THF
(4 L) were charged, and the mixture was cooled to 30 8C. The cold dia-
Experimental Section nion solution from the first reactor was then slowly added to the electro-
phile solution over approximately 3 h and the internal temperature was
Unless otherwise specified, all reactions were performed in equipment maintained at approximately 20 8C. After 1 h, the reaction was
pre-purged with N2 before charging the reagents. quenched by the slow addition of HCl (1 n, 5 L), and the phases were

84 www.AsianJOC.org  2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim Asian J. Org. Chem. 2012, 1, 80 – 89
Synthesis of HCV Protease Inhibitor

separated. The organic phase was washed with H2O (1  10 L) and con- d = 198.6 (s), 160.0 (s), 149.0 (s), 132.9 (d), 113.9 (s), 99.4 (d), 98.4 (s),
centrated to a minimal stirrable volume at 30 8C/10 mmHg. This material 56.3 (q), 27.6 ppm (q). IR: 615, 641, 784, 931, 972, 1022, 1046, 1131,
was then used directly for the subsequent hydrolysis. 1H NMR 1187, 1256, 1308, 1361, 1422, 1463, 1484, 1522, 1564, 1604, 1631, 1640,
(500 MHz, CDCl3, 30 8C): d = 8.15 (br s, 1 H, N-H), 8.08 (d, J = 8.5 Hz, 1631, 1640, 3285, 3426 cm 1. HRMS: m/z: calcd for C9H11BrNO2 :
1 H, Ar-H), 7.28 (m, 1 H, Ar-H), 6.68 (d, J = 8 Hz, 1 H, Ar-H), 3.91 (s, 243.9968 [M + H] + ; found: 243.9968; error = 0.138 ppm. Elemental anal-
3 H, OMe), 1.68 ppm (s, 9 H, tBu). 13C NMR (125 MHz, CDCl3): d = ysis calcd (%) for C9H10BrNO2 : C 44.29; H 4.13; N 5.74; found: C 44.36;
176.7 (s), 155.9 (s), 137.1 (s), 128.2 (d), 113.7 (d), 103.2 (d), 103.2 (s), H 3.79; N 5.48.
56.4 (q), 40.3 (s), 27.5 ppm (q). IR: 650, 772, 946, 1030, 1077, 1158, 1179,
1227, 1259, 1292, 1366, 1410, 1470, 1522, 1596, 1690, 2962 cm 1. HRMS: Quinolone 12
m/z: calcd for C12H17BrNO2 : 286.0437 [M + H] + ; found: 286.0433; A 1 L jacketed reactor fitted with a mechanical stirrer, thermocouple,
error = 1.46 ppm. Elemental analysis calcd (%) for C12H16BrNO2 : C N2 inlet, and addition funnel was charged with thiazole carboxylic acid
50.37; H 5.64; N 4.89; found: C 50.03; H 5.21; N 4.68. 10 (25.0 g, 117 mmol, 1 equiv). The solid was then dissolved in 180 mL
of N-methylpyrrolidone (NMP). Thionyl chloride (8.94 mL, 122 mmol,
Bromoanisidine HCl 8
1.05 equiv) was added dropwise by syringe and the internal temperature
Part 2: The crude solution of the bromoanisyl amide described above was maintained below 25 8C. After 30 min, a sample was removed and
was diluted with ethanol (16 L), and HCl (12 n, 12 L) was slowly added. quenched into diethylamine to confirm conversion to the acid chloride.
The resulting mixture was heated to reflux for 24 h, then allowed to cool A solution of anilinoketone 9 in 70 mL NMP was then added all at
slowly to 23 8C, which caused a precipitate of the product to form. The once, and the charge port was rinsed with 5 mL of NMP. The reaction
slurry was cooled to 0 8C, left to stand for 2 h, and filtered. The filter was stirred at 25 8C, and after 10 h a sample was taken for HPLC analy-
cake was then washed with n-heptane/methyl tert-butyl ether (MTBE, sis to confirm formation of the intermediate amide. Additional NMP
1:1, 4 L), and then dried under vacuum at 35 8C until H2O < 1.0 % (as was then charged to the reactor (58 mL) followed by slow addition of
monitored by Karl Fischer (KF) coulometry) was obtained, to give tBuOK (13.1 g, 117 mmol, 1 equiv). The internal temperature was main-
2.55 kg (89 %) of bromoanisidine HCl 8 as a light brown solid; tained below 30 8C. The reaction was then heated to 80 8C, and the re-
m.p. 85 8C (dec). 1H NMR (500 MHz, [D6]DMSO, 30 8C): d = 8.15 (br s, maining tBuOK (65.5 g, 584 mmol, 5 equiv) was added slowly over
3 H, NH2 + HCl), 7.21 (dd, J = 8, 8 Hz, 1 H, Ar-H), 6.86 (d, J = 8 Hz, 1 H, about 30 min through a solid addition funnel. The internal temperature
Ar-H), 6.69 (d, J = 8 Hz, 1 H, Ar-H), 3.82 ppm (s, 3 H, OMe). 13C NMR was maintained below 100 8C. The reaction darkened to almost black.
(125 MHz, [D6]DMSO, 30 8C): d = 156.4 (s), 139.8 (s), 128.7 (d), 112.3 After the addition was complete, the reaction was heated to 100 8C.
(d), 105.7 (d), 101.5 (s), 56.3 ppm (q). IR: ~n = 621, 648, 705, 750, 770, After 1 h, HPLC analysis indicated complete conversion of 10 into 12.
883, 913, 962, 1036, 1063, 1102, 1115, 1163, 1257, 1289, 1443, 1467, 1508, The reaction was cooled to 60 8C, then HCl (6 m, 124 g, 628 mmol,
1551, 1580, 1610, 1936, 2598, 2798 cm 1. HRMS: m/z: calcd for 5.4 equiv) was charged through the addition funnel. The internal temper-
C7H9BrNO: 201.9862 [M + H] + ; found: 201.9857; error = 2.48 ppm. Ele- ature was maintained below 70 8C, which caused a solid to precipitate
mental analysis calcd (%) for C7H9BrClNO: C 35.25; H 3.80; N 5.87; out of solution. The reaction mixture was held at 65 8C for 30 min as
found: C 35.10; H 3.56; N 5.83. a thick slurry developed. A small sample was taken from the reaction,
diluted with H2O, filtered, and the pH value was measured to be
Anilinoketone 9 pH 1.5–2.0. A peristaltic pump was used to charge 350 g of H2O over ap-
BCl3 in a solution of fluorobenzene (1.886 kg, 4.182 mol, 1.33 equiv) was proximately 60 min, and the reaction was then cooled to 40 8C over ap-
added to a stirring suspension of bromoanisidine HCl 8 (0.750 kg, proximately 30 min, then held at this temperature for 1 h. The solid was
3.145 mol, 1 equiv) in fluorobenzene (1.50 L) at 0–5 8C over 15 min and collected by vacuum filtration by using a Bchner funnel. The filter cake
the internal temperature was maintained below 5 8C. The reaction mix- was washed with NMP/H2O (1:1 w/w, 80 g), H2O (120 g), and finally
ture was allowed to slowly warm up to 22 8C over 0.5 h, and stirred at EtOAc (80 g). The filter cake was dried on the funnel until the light
ambient temp (20–22 8C) for 1 h. MeCN (0.723 kg, 17.61 mol, 5.6 equiv) gray solid could be transferred to a dish and dried in a vacuum oven at
was then slowly added to the reaction mixture over 20 min and the inter- 45 8C/70 mmHg. After drying to a constant weight, 41.9 g (81.5 % over
nal temperature was maintained at 25 8C or below by using a cold water two steps corrected for both water content and purity) of 12 was ob-
bath (18–20 8C). The mixture was stirred at 20–22 8C for 1 h. Fluoroben- tained. HPLC purity: 98.8 %; H2O = 3.05 % (KF); m.p. 283–284 8C.
1
zene (1.68 L) was added to the reaction mixture, a water bath (18–20 8C) H NMR (400 MHz, [D6]DMSO, 30 8C): d = 12.42 (s, 1 H, N-H), 8.21 (s,
was placed under the flask, and AlCl3 (0.236 kg, 1.77 mol, 0.56 equiv) 1 H, Ar-H), 8.13 (d, J = 9 Hz, 1 H, Ar-H), 7.31 (d, J = 9 Hz, 1 H, Ar-H),
was added in one portion (internal temperature rose from 22 to 32 8C). 6.91 (br s, 1 H, N-H), 4.02 (s, 3 H, OMe), 2.84 (septet, J = 7 Hz, 1 H, C-
The reaction mixture was then stirred at approximately 22 8C for 1 h, H), 1.16 ppm (s, 6 H, Me). 13C NMR (125 MHz, [D6]DMSO, 30 8C): d =
then stirred at 75–77 8C (reflux) for 3.5 h. The resulting mixture was 175.9 (s), 158.8 (s), 158.4 (s), 125.6 (d), 118.9 (s), 115.4 (d), 110.3 (d),
then cooled to approximately 10 8C (ice-water bath) and quenched by n = 677, 708, 798, 826, 876,
103.3 (d), 57.0 (q), 33.7 (d), 19.0 ppm (q). IR: ~
slow addition of H2O (3.75 L) over 20 min and the internal temperature 951, 1006, 1066, 1105, 1159, 1182, 1195, 1236, 1268, 1288, 1367, 1401,
was maintained at 25 8C or below. The reaction was then stirred at 50 8C 1433, 1456, 1479, 1528, 1548, 1568, 1602, 1632, 1682, 2970, 3073,
for 2 h. The reaction mixture was cooled to 22 8C and stirred for 10 h. 3291 cm 1. HRMS: m/z: calcd for C17H17BrN3O3S: 422.0169 [M + H] + ;
Celite (0.325 kg) was charged to the reactor, stirred for 15 min., and fil- found: 422.016158; error = 1.779 ppm. Elemental analysis calcd (%) for
tered. The flask and filter cake were washed with additional fluoroben- C17H16BrN3O3S·H2O: C 46.37; H 4.12; N 9.54; found: C 46.01; H 3.70; N
zene (2.25 L), the layers were separated, and the aqueous layer was ex- 9.42.
tracted with fluorobenzene (1.0 L). The combined organic extracts were
Chloroquinoline 3 a
washed with H2O (3.0 L). The organic extract was concentrated to
1.75 L by distillation, and isoproyl alcohol (IPA, 3.0 L) was added. The CAUTION! This compound is Ames positive. Handle with proper per-
mixture was again concentrated to 2.40 L. At this stage analysis of the sonal protective equipment. A 20 gallon reactor was charged with quino-
supernatant by NMR spectroscopy (or GC) showed the complete ab- lone 12 (2.70 kg, 6.4 mol, 1.0 equiv), 1,4-dioxane (19.2 L), then POCl3
sence of fluorobenzene. H2O (3.0 L) was added over 15 min (ratio of (2.94 kg, 19.2 mol, 3 equiv). The resulting mixture was heated to 85 8C
H2O/IPA ca. 3:2), then the mixture stirred for 2 h and filtered. The flask over 1 h and kept at this temperature for 2 h. HPLC analysis showed
and filter cake were washed with H2O/IPA (3:2, 1.50 L), and dried at that the reaction was complete (99.4 % conversion). The reaction mix-
22 8C under a flow of N2 at 80 mmHg to give 0.615 kg (80.0 %) of prod- ture was cooled to room temperature. In a separate 50 gallon reactor,
uct 9 as an off-white solid; m.p. 95–96 8C. HPLC purity: 99.9 %; H2O = NaHCO3 (8.72 kg) was dissolved in H2O (90.8 L), and about 20 L of the
0.16 % (KF); 1H NMR (400 MHz, CDCl3, 30 8C): d = 7.72 (d, J = 8 Hz, solution was taken out for use in a rinse afterwards. The reaction mix-
1 H, Ar-H), 7.10 (br s, 2 H, NH2), 6.28 (d, J = 8 Hz, 1 H, Ar-H), 3.94 (s, ture from the 20 gallon reactor was slowly transferred to the aqueous
3 H, OMe), 2.56 ppm (s, 3 H, Me). 13C NMR (100 MHz, CDCl3, 30 8C): NaHCO3 solution at such a rate that the foaming of CO2 was under con-

Asian J. Org. Chem. 2012, 1, 80 – 89  2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.AsianJOC.org 85
Carl A. Busacca, Xudong Wei et al.
FULL PAPER
trol and the temperature remained below 20 8C. Reactor 1 was rinsed Sulfone 3 b
with 1,4-dioxane (5 L) and sodium bicarbonate solution (20 L), and the
Tosylate 19 was prepared as described above but was not distilled or iso-
rinse was transferred to reactor 2. The reaction mixture was stirred at
lated. Instead, after the reaction mixture was left to stand at 32  2 8C
approximately 20 8C for 1 h, and the slurry was filtered with a Nutsche
for 0.5 h then cooled to 22 8C over 0.5 h, sodium benzenesulfinate
filter. The filter cake was washed with H2O (35 L, pH 7) and n-heptane
(15 L). The wet product was dried under vacuum (80 mm) at 40 8C for (0.777 kg, 4.73 mol, 2.0 equiv) was charged in one portion at 22 8C to the
reaction mixture, and subsequently acetic acid (1.50 L) was also added.
2 days (H2O = ca. 0.1 % (KF)). In total, 2.44 kg (90 %) of 3 a was ob-
The reaction mixture was then heated to 56  1 8C and stirred for ap-
tained as a pink solid, which was directly used in the next step. An
HPLC assay showed the product to be 98.5 % pure. A crystalline sample proximately 3 h. HPLC analysis showed that 0.1 % of unreacted 19 re-
was obtained by suspending the material in DMAc at 50 8C and agitating mained. The reaction mixture was then cooled to 22 8C over 0.5 h and
left to stand at this temperature for 0.5 h. The reaction mixture was then
for 1 h. The hot slurry was filtered, the filter cake was washed with H2O,
quenched by addition of H2O (1.75 L) over 30 min and stirred for 0.5 h
then dried on the frit to give 3 a as the DMAc monosolvate; m.p. >
250 8C. 1H NMR (500 MHz, [D6]DMSO, 30 8C): d = 12.33 (s, 1 H, N-H), at 22 8C. Test filtration showed that the mother liquor losses at this time
were normal, at 5.7 mg mL 1. The product was then filtered and the
8.21 (d, J = 9.3 Hz, 1 H, Ar-H), 8.15 (s, 1 H, Ar-H), 8.10 (s, 1 H, Ar-H),
filter cake was washed with MeCN/H2O (3:1 v/v, 2.84 L), then with H2O
7.70 (d, J = 9.3 Hz, 1 H, Ar-H), 4.08 (s, 3 H, OMe), 2.95 (s, 3 H, DMAc
solvate), 2.82 (m, 1 H, C-H), 2.79 (s, 3 H, DMAc solvate), 1.96 (s, 3 H, (3.0 L). The crude sulfone was then dried under vacuum at 50 8C/
DMAc solvate), 1.16 ppm (d, J = 6.8 Hz, 6 H, iPr). 13C NMR (125 MHz, 80 mmHg for 12 h until H2O = 7.8 % (KF). The moist filter cake
(1.22 kg) was charged back to the reactor and DMF (5.92 L) was added.
[D6]DMSO, 30 8C): d = 175.7 (s), 158.7 (s), 158.0 (s), 153.0 (s), 147.7 (s),
The mixture was then heated to 70 8C, which caused all solids to dissolve
146.2 (s), 142.3 (s), 124.4 (d), 120.6 (s), 117.1 (d), 115.4 (d), 114.8 (d),
109.3 (s), 57.1 (q), 37.4 (q), 34.5 (q), 33.8 (d), 21.4 (q), 19.1 ppm (q). IR: and a dark brown solution was obtained. The resultant mixture was
n = 656, 671, 710, 718, 740, 753, 764, 806, 825, 871, 907, 948, 981, 1007,
~ cooled to 50 8C and left to stand for 1 h, which caused some solid to
crystallize out. H2O (2.0 L) was then added over 15–20 min and the in-
1057, 1091, 1141, 1161, 1182, 1200, 1218, 1257, 1301, 1360, 1398, 1426,
1445, 1463, 1489, 1519, 1537, 1585, 1606, 1661, 2979, 3175 cm 1. HRMS: ternal temperature was maintained at 50–55 8C. The reaction mixture
m/z: calcd for C17H16BrClN3O2S: 439.9830 [M + H] + ; found: 439.9823; was then cooled to 22 8C over 0.5 h and left to stand at this temperature
error = 1.509 ppm. Elemental analysis calcd (%) for C17H15BrClN3O2S: for 1 h. Aqueous NaOH (1 m, 0.625 L) was then charged until a pH 12.5
was reached. The mixture was then stirred for 1 h at 22 8C. The resultant
C 46.33; H 3.43; N 9.53; found: C 46.08; H 3.38; N 9.23.
slurry was filtered and the filter cake washed with DMF/H2O (2:1 v/v,
Tosylate 19 5.33 L) until the pH value of the filtrate was 7–8. Sulfone 3 b was then
dried for 12 h at 70 8C/80 mmHg with an N2 bleed to constant weight, to
Quinolone 12 (1.25 kg, H2O = 4.25 % (KF), 2.37 mol) was charged to
give 1.10 kg (78.5 %) of 3 b as the mono-DMF solvate. HPLC purity:
a 20 L three-neck flask fitted with a thermocouple, a condenser, and me-
99.0 %; wt % purity: 91.7 %; H2O = 0.25 % (KF); m.p. > 250 8C. 1H NMR
chanical stirrer. n-Propanol (12.5 L, H2O = 0.054 % (KF)) was then
(500 MHz, [D6]DMSO, 30 8C): d = 8.89 (s, 1 H, N-H), 8.48 (d, J = 9.2 Hz,
charged to the reactor and stirred at moderate speed. The suspension
1 H, Ar-H), 8.09 (s, 1 H, Ar-H), 8.06 (d, J = 7.2 Hz, 2 H, Ar-H), 7.95 (s,
was agitated at 95 8C for 2 h, then cooled to 22 8C linearly over 2 h and
1 H, DMF), 7.73 (t, J = 6.6 Hz, 1 H, Ar-H), 7.66 (m, 3 H, Ar-H), 4.02 (s,
filtered. (filtrate H2O = 0.61 % (KF)). The solid was washed with n-prop-
3 H, OMe), 2.89 (s, 3 H, DMF), 2.80 (quintet, J = 6.8 Hz, 1 H, C-H), 2.73
anol (3.0 L) and dried under vacuum (80 mmHg) at 80 8C for 17 h (with
(s, 3 H, DMF), 1.17 ppm (d, J = 6.6 Hz, 6 H, iPr). 1H NMR (500 MHz,
N2 bleed) to give 1.124 kg (90 %) of anhydrous quinolone 12 (H2O =
CDCl3): d = 9.82 (s, 1 H, N-H), 8.87 (s, 1 H, Ar-H), 8.57 (d, J = 9.4 Hz,
0.76 % (KF)). Residual n-propanol was determined to be 0.03 mol % by
1 H, Ar-H), 8.15 (s, 1 H, Ar-H), 8.11 (s, 1 H, Ar-H), 7.97 (d, J = 8.2 Hz,
GC. The anhydrous quinolone 12 (1.00 kg, 2.367 mol, H2O = 0.76 %
2 H, Ar-H), 7.56 (m, 1 H, Ar-H), 7.48 (m, 2 H, Ar-H), 7.32 (d, J = 9.4 Hz,
(KF), 1 equiv) was charged to a 20 L three neck flask fitted with a ther-
1 H, Ar-H), 4.04 (s, 3 H, OMe), 2.99 (s, 3 H, DMF), 2.92 (s, 3 H, DMF),
mocouple, a condenser, and mechanical stirrer. MeCN was then charged
2.81 (quintet, J = 6.8 Hz, 1 H, C-H), 1.35 ppm (d, J = 6.9 Hz, 6 H, iPr).
(7.48 L, H2O = 0.05 %(KF)) then N,N-dimethylacetamide (1.11 L, H2O = 13
C NMR (125 MHz, [D6]DMSO, 30 8C): d = 175.8 (s), 162.3 (d, DMF),
0.05 %(KF)) and the mixture was stirred at medium speed. N-methylpyr-
159.0 (s), 157.7 (s), 152.8 (s), 147.6 (s), 146.6 (s), 145.2 (s), 139.6 (s),
rolidine (0.32 L, 0.262 kg, 3.08 mol, 1.3 equiv) was added in one portion
134.5 (d), 130.0 (d), 127.4 (d), 124.1 (d), 117.8 (d), 116.29 (d), 116.23 (d),
at 22 8C and left to stand for approximately 10 min. p-Toluenesulfonyl
115.2 (d), 110.0 (s), 57.0 (q), 35.7 (q, DMF), 33.9 (d), 30.7 (q, DMF),
chloride (0.519 kg, 2.72 mol, 1.15 equiv) was then charged at 25 8C over
19.0 ppm (q). 13C NMR (125 MHz, CDCl3): d = 175.3 (s), 162.8 (d,
15–20 min, which caused an exotherm to 29–30 8C. The reaction mixture
DMF), 158.2 (s), 157.7 (s), 153.1 (s), 148.5 (s), 147.5 (s), 145.8 (s), 140.5
was then left to stand at 32  2 8C for 0.5 h, then cooled to 22 8C over
(s), 133.9 (d), 129.5 (d), 127.7 (d), 124.3 (d), 118.5 (d), 117.3 (d), 115.6
0.5 h. The batch was then distilled at 40 8C/20 mm to remove most of the
(d), 115.0 (d), 111.5 (s), 57.0 (q), 36.6 (q, DMF), 35.5 (d), 31.5 (q, DMF),
MeCN. The batch was then diluted with H2O (7 L), the resultant slurry
19.2 ppm (q). IR: 618, 662, 672, 687, 710, 728, 744, 754, 763, 774, 806,
filtered, and the filter cake washed with additional H2O. Solid 19 was
823, 856, 871, 901, 947, 980, 995, 1008, 1057, 1082, 1094, 1140, 1154,
then dried in a tray drier (60 8C) to constant weight of 1.26 kg (92 %);
1186, 1199, 1259, 1297, 1321, 1355, 1384, 1398, 1423, 1446, 1461, 1491,
m.p. 216–217 8C. 1H NMR (400 MHz, [D6]DMSO, 30 8C): d = 12.38 (s,
1516, 1556, 1579, 1604, 1651, 1687, 2974 cm 1. HRMS: m/z: calcd for
1 H, N-H), 8.06 (d, J = 4.9 Hz, 2 H, Ar-H), 7.90 (d, J = 8.3 Hz, 2 H, Ar-H),
C23H21BrN3O4S2 : 546.0151 [M + H] + ; found: 546.0150; error = 0.252 ppm.
7.80 (d, J = 9.2 Hz, 1 H, Ar-H), 7.51 (d, J = 9.3 Hz, 1 H, Ar-H), 7.46 (d,
Elemental analysis calcd (%) for C26H27BrN4O5S2 : C 50.40; H 4.39; N
J = 8.2 Hz, 2 H, Ar-H), 4.02 (s, 3 H, OMe), 2.83 (quint, J = 6.8 Hz, 1 H, C-
9.04; found: C 50.46; H 4.26; N 8.93.
H), 2.39 (s, 3 H, Ar-Me), 1.16 ppm (d, J = 6.9 Hz, 6 H, iPr). 13C NMR
(125 MHz, [D6]DMSO, 30 8C): d = 174.9 (s), 162.7 (d, DMF), 157.9 (s),
157.8 (s), 154.4 (s), 153.9 (s), 149.0 (s), 147.9 (s), 146.1 (s), 132.0 (s), Capped tert-leucine 5
130.0 (d), 128.4 (d), 121.8 (d), 117.3 (s), 115.0 (d), 113.7 (d), 110.4 (s), Cyclopentanol (3.45 g, 40 mmol) was slowly added over approximately
108.8 (d), 57.0 (q), 36.5 (q), 35.6 (d), 31.5 (q), 21.7 (q), 19.2 ppm (q). IR: 5 min to a solution of triphosgene (4.45 g, 15 mmol) in THF (40 mL) at
n = 663, 677, 708, 725, 741, 769, 809, 832, 874, 944, 993, 1008, 1034, 1070,
~ 0–5 8C, and the resultant mixture was stirred for 5 min to dissolve the
1101, 1122, 1178, 1269, 1382, 1408, 1462, 1498, 1519, 1547, 1600, 1661, solids. Pyridine (3.16 g, 40 mmol) was then added to the reaction mix-
1695, 2970 cm 1. HRMS: m/z: calcd for C17H17BrN3O3S: 422.0169 ture over 25 min and the internal temperature was maintained below
[M OTs + H] + ; found: 422.0168; error = 0.121 ppm. Elemental analysis 5 8C. The cold bath was then removed and the reaction mixture was
calcd (%) for C24H22BrN3O5S2·0.35 H2O: C 49.46; H 3.93; N 7.21. stirred at 23 8C for 3 h. The reaction mixture was then filtered and the
Found: C 49.11; H 4.15; N 7.49. filter cake was washed with THF (7 mL). The filtrate containing the
chloroformate was assayed by HPLC and used as such in the next step.
1
H NMR (400 MHz, CDCl3): d = 1.40–1.90 (m, 8 H), 5.25–5.35 ppm (m,
1 H).

86 www.AsianJOC.org  2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim Asian J. Org. Chem. 2012, 1, 80 – 89
Synthesis of HCV Protease Inhibitor

NaOH (2 n, 38 mL) was added to a stirring suspension of tert-leucine OH), 4.31 (d, J = 9.2 Hz, 1 H, CHN), 4.01 (d, J = 11.2 Hz, 1 H, CHN),
(3.00 g, 22.80 mmol) in THF (15 mL, ACS grade) at ~ 10 8C over 5 min 3.68 (dd, J = 3.6, 11.6 Hz, 1 H, CHO), 3.06 (s, 1 H, 0.33 MTBE), 2.08 (m,
and the internal temperature was maintained below 15 8C. The mixture 1 H, C-H2), 1.87–1.48 (m, 9 H, C-H2), 1.09 (s, 3 H, 0.33 MTBE), 0.92 ppm
was stirred at approximately 15 8C for 15 min, and a solution of the (s, 9 H, tBu). 13C NMR (100 MHz, CDCl3, both rotamers, 30 8C): d =
chloroformate described above (5.75 g, 38.8 mmol) in THF (15 mL) was 173.5 (s), 172.5 (s), 157.1 (s), 78.5 (d), 69.8 (d), 59.1 (d), 58.2 (d), 56.8
then added to the stirring reaction mixture over 15–20 min at 10–15 8C. (t), 49.6 (MTBE), 36.7 (t), 35.5 (s), 32.8 (t), 32.7 (t), 26.3 (d), 23.7 ppm
The cold bath was then removed and the reaction mixture was stirred at (t). IR: ~n = 681, 704, 870, 961, 989, 1015, 1051, 1069, 1085, 1181, 1213,
23 8C for 3 h. MTBE (25 mL) was added, the mixture was stirred vigo- 1276, 1309, 1361, 1433, 1501, 1630, 1716, 2950, 3442 cm 1. HRMS: m/z:
rously for 10 min, then the layers were separated. The aqueous layer calcd for C17H29N2O6 : 357.2020 [M + H] + ; found: 357.2010; error =
was acidified with concentrated HCl to approximately pH 2 and the 0.038 ppm. Elemental analysis calcd (%) for C17H28N2O6 : C 57.29; H
product was extracted with ethyl acetate (2  25 mL). The combined or- 7.92; N 7.86; found: C 56.89; H 8.36; N 7.76.
ganic layers were concentrated, n-heptane was added to the residue, and
it was again concentrated. The slurry thus obtained was filtered, washed SNAr product 20
with n-heptane, and dried to give 4.55 g (82 %) of 5 as a white solid; A 200 L reactor was charged with DMF (89 L), then sulfone 3 b·DMF
m.p. 155–156 8C. 1H NMR (400 MHz, CDCl3): d = 11.2 (br s, 1 H, O-H), (15.09 kg, 24.21 mol, 0.98 equiv), and dipeptide acid 1 (8.95 kg,
5.20 (m, 1 H, C-H-O), 5.10 (br s, 1 H, N-H), 4.20 (d, J = 10 Hz, 1 H, C-H- 24.69 mol, 1.0 equiv). The mixture was cooled to 4 8C, then tBuOK/
N), 1.84–1.57 (m, 8 H, C-H2), 1.03 ppm (s, 9 H, tBu). 1H NMR (500 MHz, THF (0.97 m, 84.63 kg, 74.01 mol, 3.8 equiv) was added over 1 h and the
[D6]DMSO, 30 8C): d = 12.5 (br s, 1 H, O-H), 7.06 (d, J = 9.0 Hz, 1 H, N- internal temperature was maintained at 1– 4 8C. The batch was left to
H), 4.94 (m, 1 H, C-H-O), 3.79 (d, J = 8.8 Hz, 1 H, C-H-N), 1.80 (m, 2 H, stand for 10 min at this temperature, then warmed to 19 8C over 1 h. The
C-H2), 1.69–1.48 (m, 6 H, C-H2), 0.94 ppm (s, 9 H, tBu). 13C NMR mixture was then left to stand for a further 3 h at 19 8C, at which time
(125 MHz, [D6]DMSO, 30 8C): d = 172.8 (s), 156.3 (s), 76.3 (d), 62.6 (d), HPLC analysis indicated that 0.6 % of 3 b remained. The batch was then
n = 581, 654, 696,
33.2 (s), 32.31 (t), 32.28 (t), 26.7 (q), 23.3 ppm (t). IR: ~ cooled to 0 8C and quenched by the addition of HOAc (7.45 kg, 124 mol,
965, 1018, 1063, 1172, 1213, 1261, 1329, 1369, 1416, 1477, 1529, 1663, 5 equiv) over approximately 30 min. The internal temperature was main-
1727, 2963, 3336 cm 1. HRMS: m/z: calcd for C12H22NO4 : 244.1543 [M + tained at 0–10 8C. The reactor was then configured for distillation and
H] + ; found: 244.1541; error = 0.962 ppm. Elemental analysis calcd (%) 90 L of THF distillate was obtained at 35–40 8C/300 mmHg over 90 min.
for C12H21NO4 : C 59.24; H 8.70; N 5.76; found: C 58.71; H 8.61; N 5.68. GC analysis of an aliquot showed a THF/DMF ratio of 1:13 (target
< 1:10 w/w). The batch was cooled to 21 8C and 2-MeTHF (148 L) and
Dipeptide acid 1 HCl (0.6 n, 120 L) were added. The pH value of the aqueous layer was
A 100 gallon glass reactor was charged with acid 5 (15.0 kg, 61.65 mol, pH 4.65. The phases were separated, and the aqueous phase was discard-
1 equiv) and MeCN (30.0 L). The mixture was agitated at 150 RPM for ed. H2O (148 L) was then charged to the organic phase, and agitation
15 min, and then cooled to 0 8C. NMM (31.4 kg, 308.3 mol, 5.0 equiv) was continued for 10 min. After agitation was stopped, the aqueous
was then added over 20 min to give a colorless solution. The internal phase was discarded. The organic phase was then clarified by filtration
temperature was then reduced to 2 8C and a solution of tosyl chloride through a Seitz filter containing 0.5 kg Celite, and the reactor was
(12.15 kg, 63.75 mol, 1.03 equiv) in MeCN (30.0 L) was added over washed with an additional small charge of 2-MeTHF. The filtered organ-
80 min. The internal temperature was maintained below 5 8C. The result- ic solution was then charged back to the reactor and distillation per-
ing mixture was left to stand for 1 h from the end of the addition at 0 8C, formed at 45–55 8C/200 mmHg. 97 L of distillate was collected. nPrOH
then cooled to 2 8C. A slurry of 4-hydroxyproline methylester HCl was then charged (74 L) and distillation at 50–65 8C/200 mmHg per-
(12.25 kg, 67.50 mol, 1.1 equiv) in MeCN (30.0 L) was then added over formed. 24 L of distillate was collected. An additional charge of nPrOH
25 min. The temperature was maintained below 10 8C during the addi- (25.55 kg) was then made, to give an approximately 20 % w/w solution
tion. The mixture was held for 1.5 h at 0–15 8C, and HPLC analysis of SNAr product 20. The batch was heated to 75 8C, to give a clear solu-
showed that less than 0.22 % of 5 remained (checked as the benzylamide tion. The mixture was then cooled to 22 8C over 2 h, then left to stand at
derivative). LiOH (3.2 m, 90.0 L, 12.10 kg in 85.5 L H2O; 288.0 mol, 22 8C for 1.5 h. The resultant slurry was filtered and the filter cake
4.67 equiv) was then added to this mixture of ester 6 over 40 min, and washed with nPrOH (36 L). The wet filter cake was dried at 70 8C/
the internal temperature was maintained at 15 8C or below. The mixture 100 mmHg with an N2 bleed for 5 h to give 16.0 kg (86 %) of 20; HPLC
was then warmed to 23 8C and left to stand for 1 h, at which time HPLC purity: 99.4 %. Residual solvent analysis showed 0.03 % H2O and 0.06 %
analysis showed complete consumption of the intermediate ester. The nPrOH; m.p. > 250 8C. 1H NMR (500 MHz, [D6]DMSO, 30 8C): d = 12.33
MeCN wasremoved by distillation under reduced pressure (30–45 8C/ (s, 1 H, O-H), 8.16 (d, J = 9.1 Hz, 1 H, Ar-H), 8.05 (br s, 1 H, N-H), 7.46
30 mm), then HCl (6 n, 25.5 L, 28.4 kg, 153.0 mol) was added over (s, 1 H, Ar-H), 7.36 (d, J = 9.2 Hz, 1 H, Ar-H), 6.98 (d, J = 9.2 Hz, 1 H,
25 min and the internal temperature was maintained below 35 8C. The Ar-H), 5.41 (br s, 1 H, C-H-O), 4.51–4.42 (m, 1 H, C-H-O), 4.10 (d, J =
final pH value was 7–8 (pH 7.98 was measured). Residual MeCN was 8.6 Hz, 1 H, N-H), 4.02 (m, 1 H, C-H-N), 4.01 (s, 3 H, OMe), 3.91 (dd,
then removed by distillation (45 8C/30 mm) to below 1 % w/w. The mix- J = 2.6, 12 Hz, 1 H, C-H-N), 3.35 (br s, 2 H, C-H-N), 2.84 (sep, J = 6.8 Hz,
ture was heated to 60 8C and HCl (6 n, 28.75 kg, 154 mol) was slowly 1 H, C-H-iPr), 2.71 (dd, J = 7.5, 14 Hz, 1 H, C-H2), 2.33 (m, 1 H, C-H2),
added to reach pH 3.6, then the batch was seeded with 75 g dipeptide 1.68–1.35 (m, 7 H, C-H2), 1.24 (br m, 1 H, C-H2), 1.16 (d, J = 6.8 Hz, 6 H,
acid 1 seeds in 1 L H2O as a slurry. The agitation was continued at 60 8C iPr), 1.00–0.93 ppm (br s, 9 H, tBu). 13C NMR (125 MHz, [D6]DMSO,
for 30 min to establish a seed bed. An additional charge of HCl (6 n, 30 8C): d = 175.7 (s), 172.7 (s), 170.5 (s), 160.2 (s), 158.4 (s), 157.3 (s),
11.05 kg) was then added over 1 h to reach pH 0.99 at 60 8C (pH 1.3 at 156.3 (s), 154.1 (s), 149.3 (s), 146.7 (s), 122.6 (d), 116.4 (s), 113.8 (d),
23 8C). After standing for 1 h at 60 8C, the mixture was cooled to 23 8C 112.5 (d), 108.7 (s), 97.9 (d), 76.7 (d), 76.3 (d), 59.1 (d), 57.4 (d), 56.7
over 1 h, and then left to stand at that temperature for a further 1 h. (q), 52.9 (t), 34.1 (t), 34.2 (s), 34.1 (t), 33.7 (d), 32.2 (t), 31.9 (t), 26.2 (q),
The slurry was then filtered (50 min), and the filter cake was washed 23.14 (t), 23.06 (t), 19.13 (q), 19.06 ppm (q). IR: ~ n = 648, 772, 824, 840,
with HCl (0.1 n, 2  25.0 L), then finally washed with H2O (25.0 L). The 965, 1060, 1095, 1124, 1148, 1168, 1188, 1217, 1263, 1291, 1355, 1387,
solid was dried at 50 8C/30 mm with an N2 bleed for 36 h to give 20.65 kg 1425, 1437, 1502, 1556, 1592, 1609, 1658, 1708, 2954 cm 1. HRMS: m/z:
(91 % over two steps) of dipeptide acid 1 as a white crystalline solid. calcd for C34H43BrN5O8S: 760.2010 [M + H] + ; found: 760.1995; error =
HPLC purity: 99.91 %; wt % purity: 101.14 %; H2O = 0.093 % (KF); 2.004 ppm. Elemental analysis calcd (%) for C34H42BrN5O8S: C 53.68; H
thermogravimetric analysis loss = 0.854 %. m.p. 117–124 8C; aD = 58.6 5.57; N 9.21; found: C 53.84; H 5.57; N 9.16.
(c 2.17, MeOH). A crystalline sample for further characterization was
Ester 21
obtained by recrystallization from hot MTBE, which provided the 0.33
MTBE solvate crystalline form: m.p. 184–185 8C. 1H NMR (400 MHz, A clean, dry 200 L glass lined steel reactor, capable of maintaining a re-
CDCl3, major rotamer reported, 30 8C): d = 5.51 (d, J = 9.2 Hz, 1 H, N- action at 15 8C, was dried under heat and vacuum. The reactor was
H), 5.02 (br s, 1 H, O-H), 4.70 (t, J = 8.8 Hz,1 H, CHO), 4.52 (s, 1 H, charged with acid 20 (15.7 kg, 20.6 mol, 1.0 equiv) and THF (110 L,

Asian J. Org. Chem. 2012, 1, 80 – 89  2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.AsianJOC.org 87
Carl A. Busacca, Xudong Wei et al.
FULL PAPER
H2O = 45 ppm (KF)). The mixture was agitated until a solution was BI 201335
formed, then the jacket was adjusted so that the internal temperature
A dry 400 L reactor was charged with ester 21 (16.32 kg, 15.76 kg cor-
was approximately 15 8C. N-methylmorpholine (2.31 kg, 22.9 mol,
rected for volatiles and purity, 17.83 mol, 1 equiv), purged with N2, then
1.1 eq) was then slowly added at a rate that maintained the internal tem-
THF (100 L) was charged. The mixture was agitated for 5 min to give
perature at 10 8C. By using a pre-calibrated metering valve, isobutyl
chloroformate (3.13 kg, 22.9 mol, 1.1 eq) was slowly added at a rate that a clear solution, which was then cooled to 2 8C. A solution of
LiOH·H2O (2.24 kg, 53.38 mol, 3.0 equiv) in H2O (32 L) was added to
maintained the internal temperature at 10 8C. On this scale the addi-
the cold mixture over 30 min and the internal temperature was main-
tion took 50 min. Note: controlling the temperature is critical for the
tained below 0 8C. The batch was then warmed to 21 8C over 1 h, then
success of this reaction. If the internal temperature goes above 10 8C
during the addition the yield will drop and additional impurities will be held at 21 8C for 8 h, at which time HPLC analysis of an aliquot showed
that less than 1 % of 21 remained. Isopropyl acetate (IPAc, 89 L) and
formed. The internal temperature was adjusted to 10 8C and held for
a solution of NaCl (28.1 kg) in H2O (79 L) were then charged. The re-
90 min. Aminocyclopropane salt 2 (7.12 kg, 22.9 mol, 1.1 eq) was then
sulting mixture was cooled to 5 8C, and 21.6 kg (ca. 80 % of the total) of
added through a solid-charging funnel and the internal temperature was
aqueous HCl (conc. HCl (5.06 kg) in H2O (22 L)) was added over
maintained at 8 8C. A second charge of N-methylmorpholine (2.29 kg,
30 min. The internal temperature was maintained below 6 8C. The
22.7 mol, 1.1 eq) was then added at a rate that maintained the internal
pH value was pH 7.0. Three additional small charges of the aqueous
temperature at 8 8C. The reaction was held at 10 8C for 2 h, then
HCl solution (1–2 kg) were then added and the pH value was checked
warmed to 10 8C, and held at that temperature until conversion was at
after each addition, until all of the HCl solution (27.1 kg) had been
least 95 % by HPLC analysis (this batch required 12 h). The reaction
added, and the final pH value was pH 4.3. The agitation was then
mixture was then warmed to 20 8C. 10 % aqueous NaCl (61 L) and 2-
stopped, and the aqueous phase was separated into a drum. The organic
MeTHF (63 L) were then charged, agitated for 10 min, and the layers al-
phase was drained into a separate drum. The aqueous phase was
lowed to separate. The aqueous layer was discarded. The organic layer
charged back to the reactor, then IPAc (89 L) was charged, and the mix-
was then distilled under reduced pressure to a minimum stirrable
ture agitated for 5 min. The agitation was stopped and the phases sepa-
volume (ca. 15 % of the original volume). Ethanol (126 L, denatured
rated. The aqueous phase was then discarded, and the first organic ex-
with 0.5 % toluene, SDA2B-3) was then added and the mixture again
tract was charged back to the reactor. H2O (79 L) was then charged, and
distilled to a minimum stirrable volume (ca. 15 % of the original
the mixture agitated for 5 min. The agitation was stopped once more
volume). An additional charge of SDA2B-3 ethanol (50 kg) was added
and the phases separated. The aqueous phase was again discarded. The
to make the solution 20 wt % in ester 21 based on an HPLC assay, and
organic solution was clarified by filtration through a Seitz filter contain-
then the solution was heated to 50 8C. H2O (36 L) was added over about
40 min and the internal temperature was maintained at 50 8C, to make ing 0.5 kg of Celite, and the reactor was washed out with additional
charge of IPAc (5 kg). This solution was stored for two days while
the final solution 29 wt % in H2O. Note: if the H2O is added too fast,
a second, identical hydrolysis reaction (17.4 kg) was carried out. The
there is the possibility of spontaneous crystallization. The reaction mix-
clarified organic solution from the second batch was combined with that
ture was held at 50 8C for 20 min. The resultant mixture was then seeded
with type A seed crystals of ester 21 (1 wt %, 157 g) as a slurry in etha- of the first batch and the IPAc was removed by distillation at a jacket
temperature of 61 8C and a pressure of 280 torr. After 6 h, a total of
nol/H2O (1:1). The reaction mixture was then cooled at 15 8C per hour
415 L of distillate was collected. The H2O content of the product was
to 25 8C and held at 25 8C for 1 h. The slurry was then filtered by using
0.25 % (KF). Distillation was stopped and the batch was heated to 70 8C,
14“ and an 18” Sparkler filters with polypropylene filter cloths. The
system was washed with SDA2B-3 ethanol/H2O (1/1, 56 kg) and blown then ethanol (12.9 kg) was charged. At 70 8C, a slurry of BI 201335 etha-
nol solvate seed crystals (332 g) in ethanol (4.0 L) was charged by N2
dry with N2. The solid product was dried to constant weight in a vacuum
pressure. The batch temperature was lowered to 65 8C and held at that
oven at 40 8C/100 mmHg with a N2 sweep to give 15.43 kg (85 %) of
temperature for 1 h. A visible seed bed developed. The batch tempera-
ester 21 (corrected for volatiles) as a light yellow solid; HPLC purity:
98.7 %; m.p. 182–184 8C. 1H NMR (400 MHz, [D6]DMSO, major rotamer ture was then lowered to 20 8C over 5 h, and held at 20 8C for 7 h. The
slurry was then centrifuged, and the filter cake washed with two volumes
reported, 30 8C): d = 12.32 (s, 1 H, N-H), 8.69 (s, 1 H, Ar-H), 8.14 (d, J =
of ethanol (31 L and 35 L). The filter cake was then dried at 80 8C/
9.2 Hz, 1 H, Ar-H), 8.03 (s, 1 H, Ar-H), 7.45 (s, 1 H), 7.33 (d, J = 9.4 Hz,
100 mmHg with an N2 bleed for 10 h. TGA showed 0.65 % weight loss.
1 H, Ar-H), 6.97 (d, J = 8.6 Hz, 1 H, N-H), 5.65 (m, 1 H, CH = CH2), 5.40
(br s, 1 H, CHOAr), 5.20 (dd, J = 1.5, 17 Hz, 1 H, CH=CH2), 5.06 (dd, J = The dry weight of the first crop of BI 201335 was 27.3 kg (85 %) HPLC
purity: 99.5 %; m.p. 235 8C. Detailed 1H, 13C, and 15N NMR assignments
1.6, 10.2 Hz, 1 H, CH=C-H2), 4.51 (m, 1 H, CHOACHTUNGRE(C=O)), 4.39 (t, J =
are given in the Supporting Information. IR: ~ n = 657, 772, 796, 810, 837,
8.4 Hz, 1 H, CHNACHTUNGRE(C=O)), 4.31 (d, J = 11.6 Hz, 1 H, NCHtBu), (4.08 (m,
865, 880, 900, 907, 934, 961, 997, 1012, 1039, 1059, 1094, 1154, 1170,
1 H, CH2N), 3.99 (s, 3 H, OMe), 3.90 (m, 1 H, CH2N), 3.56 (s, 3 H,
CO2Me), 2.81 (m, 1 H, NH), 2.51 (m, 1 H, CH2CHOAr), 2.25 (m, 1 H, 1203, 1217, 1264, 1283, 1317, 1354, 1417, 1448, 1479, 1503, 1544, 1587,
1610, 1699, 1720, 2874, 2962, 3229 cm 1. Elemental analysis calcd (%)
CH2-CHOAr), 2.07 (m, 1 H, CH (cyclopropane)), 1.70–1.32 (m, 7 H,
for C40H49BrN6O9S·0.5 H2O: C 54.67; H 5.73; N 9.56; found: C 54.74; H
CH2-CH2), 1.30 (m, 3 H, CH2 (cyclopropane) + CH2-CH2), 1.15 (d, J =
5.85; N 9.54.
8.1 Hz, 6 H, iPr), 0.95 ppm (s, 9 H, tBu). 13C NMR (100 MHz,
[D6]DMSO, all rotamers, 30 8C): d = 175.7 (s), 171.7 (s), 170.4 (s), 170.3 Crystal structure: CCDC 875522 contains the supplementary crystallo-
(s), 160.3 (s), 160.2 (s), 158.5 (s), 158.4 (s), 157.5 (s), 157.3 (s), 156.4 (s), graphic data for this paper. These data can be obtained free of charge
155.5 (s), 154.2 (s), 154.1 (s), 149.3 (s), 149.1 (s), 146.8 (s), 146.7 (s), from The Cambridge Crystallographic Data Centre via www.ccdc.cam.a-
134.7 (d), 134.2 (d), 128.0 (s), 125.5 (s), 122.5 (d), 122.0 (d), 117.4 (t), c.uk/data_request/cif.
117.1 (t), 116.4 (s), 115.8 (s), 114.0 (d), 113.9 (d), 113.0 (d), 112.5 (d),
108.82 (s), 108.78 (s), 98.0 (d), 97.9 (d), 76.8 (d), 76.33 (d), 76.28 (d),
74.9 (d), 59.4 (d), 58.5 (d), 58.1 (d), 56.9 (q), 56.7 (q), 53.2 (t), 52.1 (q),
[1] a) M. A. Joyce, D. L. J. Tyrrell, Microbes Infect. 2010, 12, 263 – 271;
51.9 (q), 48.7 (s), 35.7 (s), 34.2 (s), 34.0 (t), 33.8 (d), 33.1 (d), 32.9 (d),
b) H. Tang, H. Grise, Clin. Sci. 2009, 117, 49 – 65.
32.4 (t), 32.3 (t), 32.2 (t), 32.0 (t), 26.8 (s), 26.3 (q), 26.2 (q), 23.24 (t),
[2] a) L. Chatel-Chaix, M. Baril, D. Lamarre, Viruses 2010, 2, 1752 –
23.20 (t), 23.16 (t), 23.08 (t), 22.4 (t), 19.11 (q), 19.09 (q), 19.07 ppm (q).
1765; b) K. D. Raney, S. D. Sharma, I. M. Moustafa, C. E. Cameron,
IR: ~n = 455, 481, 521, 585, 666, 728, 771, 811, 831, 854, 913, 972, 997,
J. Biol. Chem. 2010, 285, 22725 – 22731.
1013, 1056, 1092, 1124, 1156, 1221, 1265, 1292, 1327, 1348, 1415, 1445,
[3] a) M. Llinas-Brunet, M. D. Bailey, G. Bolger, C. Brochu, A.-M.
1508, 1545, 1585, 1609, 1699, 1716, 2973, 3276 cm 1. HRMS: m/z: calcd
Faucher, J. M. Ferland, M. Garneau, E. Ghiro, V. Gorys, C.
for C41H52BrN6O9S: 883.2694 [M + H] + ; found: 883.2675; error =
Grande-Matre, T. Halmos, N. Lapeyre-Paquette, F. Liard, M. Poiri-
2.193 ppm. Elemental analysis calcd (%) for C41H51BrN6O9S.2H2O: C
er, M. Rhaume, Y. S. Tsantrizos, D. Lamarre, J. Med. Chem. 2004,
53.23; H 6.03; N 9.08; found: C 53.33; H 5.58; N 8.95.
47, 1605 – 1608; b) D. Lamarre, P. C. Anderson, M. Bailey, P. Beau-
lieu, G. Bolger, P. Bonneau, D. Lamarre, P. C. Anderson, M. Bailey,

88 www.AsianJOC.org  2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim Asian J. Org. Chem. 2012, 1, 80 – 89
Synthesis of HCV Protease Inhibitor

P. Beaulieu, G. Bolger, P. Bonneau, M. Bçs, D. R. Cameron, M. A.-M. Faucher, C. Brochu, B. Hache, J.-S. Duceppe, P. Beaulieu,
Cartier, M. G. Cordingley, A.-M. Faucher, N. Goudreau, S. H. Synthesis 2006, 2563; c) A. W. Douglas, N. L. Abramson, I. N.
Kawai, G. Kukolj, L. Lagac, S. R. LaPlante, H. Narjes, M.-A. Pou- Houpis, S. Karady, A. Molina, L. C. Xavier, N. Yasuda, Tetrahedron
part, J. Rancourt, R. E. Sentjens, R. St. George, B. Simoneau, G. Lett. 1994, 35, 6807 – 6810; d) I. N. Houpis, A. Molina, A. W. Doug-
Steinmann, D. Thibeault, Y. S. Tsantrizos, S. M. Weldon, C.-L. las, L. Xavier, J. Lynch, R. P. Volante, P. J. Reider, Tetrahedron
Yong, M. Llins-Brunet, Nature 2003, 426, 186 – 189; c) S. R. LaP- Lett. 1994, 35, 6811 – 6814; e) K. Prasad, G. T. Lee, A. Chaudhary,
lante, M. Llinas-Brunet, Curr. Med. Chem.: Anti-Infect. Agents M. J. Girgis, J. W. Streemke, O. Repic, Org. Process Res. Dev. 2003,
2005, 4, 111 – 132; d) Y. S. Tsantrizos, Bioploymers 2004, 76, 309 – 7, 723 – 732.
323. [10] T. Eicher, S. Hauptmann in The Chemistry of Heterocycles, 2nd ed.,
[4] a) P. W. White, M. Llinas-Brunet, M. Amad, R. B. Bethell, G. Wiley-VCH, Weinheim, 2003, pp. 149 – 155.
Bolger, M. Cordingley, J. Duan, M. Garneau, L. Lagac, D. Thi- [11] C. Reichard in Solvents and Solvent Effects in Organic Chemistry,
beault, G. Kukolj, Antimicrob. Agents Chemother. 2010, 54, 4611 – 3rd ed., Wiley-VCH, Weinheim, 2003, pp. 243 – 259.
4618; b) M. Llinas-Brunet, M. Bailey, N. Goudreau, P. K. Bhardwaj, [12] For examples, see: a) G. B. Barlin, W. V. Brown, J. Chem. Soc. B
J. Bordeleau, M. Bçs, Y. Bousquet, M. G. Cordingley, J. Duan, P. 1967, 648; b) G. B. Barlin, W. V. Brown, J. Chem. Soc. B 1967, 736;
Forgione, M. Garneau, E. Ghiro, V. Gorys, S. Goulet, T. Halmos, c) Z. Wrobel, Tetrahedron 1998, 54, 2607 – 2618; d) G. A. Gfesser,
S. H. Kawai, J. Naud, M.-A. Poupart, P. W. White, J. Med. Chem. E. K. Bayburt, M. Cowart, S. DiDomenico, A. Gomtsyan, C.-H.
2010, 53, 6466 – 6476. Lee, A. O. Stewart, M. F. Jarvis, E. A. Kowaluk, S. S. Bhagwat, Eur.
[5] J. Schltter, Nature 2011, 474, S5 – S7. J. Med. Chem. 2003, 38, 245 – 252.
[6] a) P. L. Beaulieu, J. G. Murray, D. Bailey, C. Boucher, J.-S. Du- [13] C. Reichardt in Solvents and Solvent Effects in Organic Chemistry,
ceppe, B. Simoneau, X.-J. Wang, L. Zhang, K. Grozinger, I. Houpis, 3rd ed., Wiley-VCH, Weinheim, 2003, pp. 99 – 105; C. Reichardt in
V. Farina, H. Heimroth, T. Krueger, J. Schnaubelt, J. Org. Chem. Solvents and Solvent Effects in Organic Chemistry, 3rd ed., Wiley-
2005, 70, 5869 – 5879; b) M. E. Fox, I. C. Lennon, V. Farina, Tetrahe- VCH, Weinheim, 2003, pp. 121 – 123.
dron Lett. 2007, 48, 945 – 948. [14] J. C. Lorenz, C. A. Busacca, X. Feng, N. Grinberg, N. Haddad, J.
[7] a) E. Fischer, Ber. Dtsch. Chem. Ges. 1903, 36, 2982; b) C. Schotten, Johnson, S. Kapadia, H. Lee, A. K. Saha, M. Sarvestani, E. M.
Ber. Dtsch. Chem. Ges. 1884, 17, 2544; c) E. Baumann, Ber. Dtsch. Spinelli, R. Varsolona, X. Wei, X. Zeng, C. H. Senanayake, J. Org.
Chem. Ges. 1886, 19, 3218. Chem. 2010, 75, 1155 – 1161.
[8] K. Wakasugi, A. Iida, T. Misaki, Y. Nishii, Y. Tanabe, Adv. Synth. [15] G. P. Samokhin, I. N. Filimonov, Anal. Biochem. 1985, 145, 311 –
Catal. 2003, 345, 1209 – 1214. 314.
[9] a) T. Sugasawa, T. Toyoda, M. Adachi, K. Sasakura, J. Am. Chem.
Soc. 1978, 100, 4842 – 4852; b) R. P. Frutos, N. Haddad, I. N. Houpis, Received: April 11, 2012
M. Johnson, L. L. Smith-Keenan, V. Fuchs, N. K. Yee, V. Farina, Published online: July 24, 2012

Asian J. Org. Chem. 2012, 1, 80 – 89  2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.AsianJOC.org 89

You might also like