Process for the manufacture of pharmaceutically active compounds

Information

  • Patent Grant
  • 8741920
  • Patent Number
    8,741,920
  • Date Filed
    Thursday, September 13, 2012
    11 years ago
  • Date Issued
    Tuesday, June 3, 2014
    10 years ago
Abstract
The present invention relates to a composition comprising the compound of formula (1),
Description
FIELD OF THE INVENTION

The present invention is related to synthesis routes to obtain the compound propane-1-sulfonic acid {3-[5-(4-chloro-phenyl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl}-amide (formula 1).




embedded image


The synthesis of the compound of formula (1) has been described before in WO 2007002433 and WO 2007002325.


However, the known synthesis routes may not be readily adapted for use on an industrial scale.


SUMMARY OF THE INVENTION

The present invention relates in part to a process for the manufacture of the compound of formula (1),




embedded image



comprising the steps of reacting the compound of formula (5),




embedded image



with 4-chlorophenylboronic acid in the presence of a palladium catalyst to produce the compound of formula (6),




embedded image



and cleaving the 2,6-dichlorobenzamide group in said compound of formula (6) to produce the compound of formula (1).


The present invention also relates in part to a compound of formula (A),




embedded image


The present invention further relates in part to a compound of formula (B)




embedded image


In addition, the present invention relates to a composition comprising the compound of formula (1) and the compound of formula (B).


The present invention also relates to an analytical method for the detection of whether the process as described above has been used in the manufacture of the compound of formula (1), said method comprising obtaining a sample from a medicament primarily containing the compound of formula (1) as active ingredient, and applying a suitable analytical method in order to detect whether said sample contains the compound of formula (B), wherein the presence of any amount of the compound of formula (B) indicates that the process has been used.


The present invention further relates to a process for the manufacture of compound (I),




embedded image



comprising the steps of:

  • aa) reacting the compound of formula (II),




embedded image



with the compound of formula (III),




embedded image



in the presence of a catalyst, copper(I)iodide and a base, to produce the compound of formula (IV),




embedded image


  • bb) reacting said compound of formula (IV) in the presence of aqueous alkali metal hydroxide at 50-90° C. and under reduced pressure to produce the compound of formula (V),





embedded image



and

  • cc) reacting said compound of formula (V) in the presence of aqueous alkali metal hydroxide or a strong base to produce the compound of formula (I);
  • wherein
  • R1 and R2 are each independently a C1-C4 alkyl, and
  • X is —Br or —Cl.


In addition, the present invention relates to a compound of formula (IV),




embedded image



wherein

  • R1 and R2 are each independently a C1-C4 alkyl, and
  • X is —Br or —Cl.







DETAILED DESCRIPTION OF THE INVENTION

The present invention provides new and improved synthesis routes for the compound of formula (1), which employ reaction conditions which are particularly amenable to being carried out on an industrial scale.


According to the present invention, there is provided a process for the manufacture of the compound of formula (1),




embedded image



comprising the steps of reacting the compound of formula (5),




embedded image



with 4-chlorophenylboronic acid (5a, scheme 1) in the presence of a palladium catalyst to produce the compound of formula (6),




embedded image



and

  • cleaving the 2,6-dichlorobenzamide group in said compound of formula (6), for example using ammonia, to produce the compound of formula (1).


The process according to the present invention efficiently uses starting materials, avoids formation of high quantities of undesirable by-products, provides high throughput and good product yields and can safely be carried out in large scale production. It is also less expensive, and, due to the efficient use of starting materials, environmentally friendlier than processes disclosed in the prior art.


The term “palladium catalyst” as used herein means any suitable palladium (Pd) catalyst, preferably bis(triphenylphosphin)palladium(II)dichloride ((PPh3)2PdCl2) and Pd on charcoal. Preferably, the amount of (PPh3)2PdCl2 is about 0.0025 equivalents with respect to compound of formula (5).


The compound of formula (5) can be obtained according to methods known to the person of skill in the art of organic chemistry. In a particularly preferred embodiment according to the present invention, the compound of formula (5) is obtained by reacting the compound of formula (2),




embedded image



with a suitable activating agent (e.g., oxalylchloride ((COCl)2)) to produce the corresponding acid chloride, then reacting said acid chloride with 5-Bromo-7-azaindole (formula 3) and a suitable coupling agent (e.g., aluminium trichloride) to produce the compound of formula (4)




embedded image



and reacting said compound of formula (4) with 2,6-dichlorobenzoylchloride under conditions suitable to produce the compound of formula (5).


In another particularly preferred embodiment according to the present invention there is provided the process for the manufacture of the compound of formula (1), wherein


a) the compound of formula (2)




embedded image



is reacted in a suitable solvent (e.g., methylenechloride (CH2Cl2)) with a suitable activating agent (e.g., Vilsmeier's salt prepared in situ from oxalylchloride ((COCl)2 and N,N-Dimethylformamide (DMF)) to produce the corresponding acid chloride, then reacting said acid chloride with 5-bromo-7-azaindole (formula 3) in the presence of a suitable activating agent (e.g., aluminium trichloride) to produce the compound of formula (4),




embedded image


b) reacting said compound of formula (4) i with 2,6-dichlorobenzoylchloride in the presence of a suitable base (e. g. n-Tripropylamine (n-Pr3N)) and suitable catalyst (e.g. N,N-Dimethylaminopyridine (DMAP)) to produce the compound of formula (5),




embedded image


c) reacting said compound of formula (5) with 4-chlorophenylboronic acid in the presence of a suitable palladium catalyst (e.g., (PPh3)2PdCl2) to produce the compound of formula (6)




embedded image



and


d) reacting said compound of formula (6) with ammonia (NH3) or a primary or secondary amine (e.g. dimethylamine) in an alcohol (methanol or ethanol) solution diluted with a polar aprotic solvent like DMA to produce the compound of formula (1).


In a preferred embodiment according to the present invention, the above-mentioned reaction step c) is carried out in a 2 phase reaction mixture comprising a non-polar aprotic solvent such as anisole or toluene and aqueous sodium or potassium carbonate (Na2CO3, K2CO3).


The compounds (4), (5) and (6) are novel and each form an embodiment of the present invention.


The process according to the present invention can be summarized according to the following reaction scheme (scheme 1) wherein, unless explicitly otherwise stated, all abbreviations and expressions have the meanings well known to the person of skill in the art of organic chemistry. All reaction partners and auxiliary agents (like i.e. catalysts, solvents) are commercially available.




embedded image


According to the present invention, the first reaction under step a) is the formation of an acid chloride of the carboxylic acid of formula (2). The reaction can be carried out employing a suspension of the compound of formula (2) in suitable medium, e.g., methylenechloride (CH2Cl2) containing catalytic amounts of DMF, to which a solution of oxalylchloride ((COCl)2) in CH2Cl2 is slowly added. The addition is typically carried out at room temperature (RT) and the reaction mixture is further stirred at RT until reaction completion (ca 6h). The next reaction is a Friedel-Crafts-Acylation. The freshly obtained solution of acid chloride is added into a cooled (T=−12 to 5° C., preferably 0-5° C.) suspension of AlCl3 and 5-Bromo-7-azaindole (3) in CH2Cl2. The reaction mixture is allowed to heat up till RT due to released heat of the exothermic reaction and stirred for about 8h to complete the reaction. The work-up and isolation procedure involves pouring of the reaction mixture into iced water, followed by extractive work-up and crystallization of the compound of formula (4) from THF/methylenchloride/heptane. The compound of formula (4) can be further purified by washing with i.e. heptane.


Reaction step b) starts with the preparation of a suspension of the compound of formula (4) in a suitable aprotic solvent (e.g., toluene). Subsequently dimethylaminopyridine (DMAP), n-tripropylamine and 2,6-dichlorobenzoylchloride are added at RT. The reaction mixture is stirred for about 60 minutes. After completion of the reaction an aqueous work-up is performed. Subsequently the toluene is slowly evaporated from the organic solution, leading to the crystallization of the compound of formula (5) which can finally be isolated and further purified through additional washing with e.g., cooled toluene.


Reaction step c) is a Pd-catalyzed Suzuki-coupling reaction of the compound of formula (5) with 4-chlorophenylboronic acid (5a). The reaction starts with the preparation of a suspension of 4-chlorophenylboronic acid (5a) and the compound of formula (5) in an aprotic solvent, such as anisole, to which an aqueous solution of Na2CO3 is added. The reaction mixture is heated slowly up to a temperature of about 85° C. During heating the Pd-catalyst is added at a temperature between RT and around 70° C. Any suitable Pd-catalyst can be used, with bis(triphenylphosphin) palladium(II)dichloride ((PPh3)2PdCl2) being especially preferred. The reaction mixture is stirred for about 120 minutes at a temperature of about 85° C. to complete the reaction. The hot bi-phasic reaction mixture is filtered to remove potentially precipitated Pd. After aqueous work-up the organic (anisole) phase is concentrated by evaporation, diluted with methanol and cooled down to about 0° C. in order to initiate crystallization of the compound of formula (6). The precipitate is isolated, washed several times with cooled methanol and subsequently dried in vacuum. If needed the compound of formula (6) can be further purified by re-crystallization from Toluene.


Reaction step d) is the removal of the 2,6-dichlorobenzamide protection group. The reaction is carried out in a suspension/solution of the compound of formula (6) in DMA/methanol (about 1:1-2:1) by addition of about 3-7 equivalents of a saturated ammonia solution (NH3 in Methanol) at RT. The reaction mixture is stirred at about 55° C. until the protection group is entirely cleaved (about 10 h). The resulting solution is diluted with methanol and subsequently the ammonia is removed via azeotropic distillation under reduced pressure. The residue is diluted with methanol and cooled to RT in order to initiate crystallization of the compound of formula (1) which can then be isolated from the reaction mixture by filtration. The compound of formula (1) can be further purified by washing with e.g., methanol.


In another embodiment of the present invention there are identified specific compounds (compounds (A) and (B) as specified below) which are by-products of the present synthesis method.




embedded image


The compound of formula (A) may be formed during the reaction of the compound of formula (5) with 4-Chlorophenylboronic acid (step c), scheme 1). The compound of formula (B) is the de-protected form of formula (A) as obtained during reaction step d) as defined above (see also scheme 1). These compounds, in particular the compound of formula (B) can serve as characteristic fingerprints for the use of the present process.


Trace amounts of the compound of formula (B) are typically found in the final product (formula (1)) as a pharmaceutically acceptable impurity in an amount of less than 0.30%, and usually in an amount of from about 0.02% to about 0.15%, as identified by HPLC, provided said compound of formula (1) had been synthesized according to the present method. Therefore, while the impurity/trace amounts of the compound of formula (B) will not affect the pharmacological- or toxicity profile of any potential future medicament or pharmaceutical preparation containing the compound of formula (1), said compound may nevertheless serve as a fingerprint in order to detect whether the present process has been used to manufacture the compound of formula (1). The presence of the compound of formula (B) will therefore allow for an unambiguous identification as to whether the present process has been used in the manufacture of the compound of formula (1).


The compounds of formulas (A) and (B), in particular the compound of formula (B), may be detected in amounts of from about 0.02% to about 0.15% by weight in a composition which otherwise contains >99% by weight of the compound of formula (1) formed using the process according to the present invention, and even following conventional purification methods known to the person of skill in the art. Therefore, trace amounts of the compound of formula (B) may even be detectable in pharmaceutical preparations containing the compound of formula (1) when obtained according to the present process. The compounds of formula (A) and (B) show the following NMR signals:


Compound A:


1H-NMR (500 MHz, d6-DMSO): δ0.99 (t, J=7.5 Hz, 3H), 1.72-1.82 (m, 2H), 3.15-3.21 (m, 2H), 7.37 (t, J=9.0 Hz, 1H), 7.52-7.58 (m, 2H), 7.65-7.74 (m, 4H), 7.76-7.85 (m, 4H), 7.87-7.92 (m, 2H), 8.58 (br. s, 1H), 8.82 (d, J=2.5 Hz, 1H), 8.90 (br. s, 1H), 9.85 (br. s, 1H).


Compound B:


1H-NMR (600 MHz, d6-DMSO): δ0.97 (t, J=7.5 Hz, 3H), 1.70-1.80 (m, 2H), 3.10-3.15 (m, 2H), 7.29 (t, J=8.7 Hz, 1H), 7.54-7.63 (m, 3H), 7.78-7.82 (m, 2H), 7.83-7.90 (m, 4H), 8.25 (s, 1H), 8.70 (br. s, 1H), 8.79 (d, J=1.8 Hz, 1H), 9.78 (br. s, 1H), 13.02 (br. s, 1H).


Therefore, as a further embodiment according to the present invention, there are provided the compounds of formulae (A) and (B) as such.


In yet another embodiment the present invention provides an analytical method for detecting whether the process according to the present invention has been used, characterized in that detectable levels of the compounds of formula (A) and/or (B) as disclosed herein are generally present in any commercial pharmaceutical preparation comprising primarily the compound of formula (1) prepared employing the synthetic process disclosed herein. Preferably the analytical method is used to detect the compound of formula (B). Any suitable analytical method known to the Organic Chemist may be applied, such as for example IR-spectroscopy, NMR-spectroscopy, Mass Spectrometry (MS) or High Performance Liquid Chromatography (HPLC). More preferably the analytical method is based on High Performance Liquid Chromatography (HPLC), which may optionally be combined with a second analytical method such as for example MS (HPLC-MS).


An aspect of the present invention is an analytical method for the detection of whether the process described above has been used in the manufacture of the compound of formula (1), said method comprising obtaining a sample from a medicament primarily containing the compound of formula (1) as active ingredient, and applying a suitable analytical method in order to detect whether said sample contains the compound of formula (B), wherein the presence of any amount of the compound of formula (B) indicates that said process has been used.


The compound of formula (1) shows potential as an active pharmaceutical ingredient, as inter alia described in WO 2007002433 and WO 2007002325. Consequently, in a further embodiment of the present invention, there is provided a pharmaceutical preparation comprising the compound of formula (1) together with detectable amounts of at least compound of formula (B). More particularly there is provided a composition comprising the compound of formula (1) in an amount greater than 99% by weight and the compound of formula (B) in amounts from about 0.01% to about 0.15% by weight. Said composition may be further processed with pharmaceutically acceptable adjuvants to give any kind of pharmaceutical preparations as inter alia described in WO 2007002433 and WO 2007002325.


The starting materials, solvents, catalysts and auxiliary reagents used in the method according to the present invention (see i.e. scheme 1) are commercially available. However, when produced on a large industrial scale, there remains a need to also obtain large amounts of starting materials in good quality and high yields.


Consequently, according to the present invention, there are also provided improved methods of making azaindole derivatives which are halogenated at their 5-position, in particular 5-bromo-7-azaindole ((3), CAS 183208-35-7) and 5-chloro-7-azaindole (CAS 866546-07-8). 5-Bromo-7-azaindole is a useful starting material in the above-mentioned process according to scheme 1.


Consequently, in a further embodiment according to the present invention there is provided a process for the manufacture of compound (I),




embedded image



comprising the steps of:

  • aa) reacting the compound of formula (II),




embedded image



with the compound of formula (III),




embedded image



in the presence of a catalyst, copper(I)iodide and a base, to produce the compound of formula (IV),




embedded image


  • bb) reacting said compound of formula (IV) in the presence of aqueous alkali metal hydroxide at 50-90° C. and under reduced pressure to produce the compound of formula (V)





embedded image



and

  • cc) reacting said compound of formula (V) in the presence of aqueous alkali metal hydroxide or a strong base to produce the compound of formula (I);
  • wherein
  • R1 and R2 are each independently selected from a C1-C4 alkyl, and
  • X is —Br or —Cl.


In one particularly preferred embodiment according to the present invention, the above process aa) to cc) is carried out with X being —Br.


In another particularly preferred embodiment according to the present invention, the above process aa) to cc) is carried out with X being —Cl.


In still another preferred embodiment according to the present invention R1 and R2 are both methyl.


The synthesis of compounds of formula (I) as described above is a sequential process. After each reaction described in steps aa) to cc), aqueous work-up procedures are applied and the formed intermediate product is optionally being isolated. In an alternative setup it is also possible to carry out the reaction steps bb) and cc) almost simultaneously. This means the conversion of compound (V) into compound (I) starts as soon as compound (V) is formed, and before the reaction to obtain compound (V) (step bb)) is terminated. Therefore, the two reaction steps bb) and cc) may run under the same reaction conditions. Consequently, no work-up of compound (V) prior to its further reaction to compound (I) is required according to this alternative method.


Therefore, in yet another embodiment there is provided a method to obtain the compound of formula (I) directly from formula (IV) without the need to apply an aqueous work-up on the stage of the compound of formula (V). This can be achieved by adding to the compound of formula (IV), dissolved in an approximately 1:1 (w/w) mixture of water and N-methylpyrrolidone (NMP), 5 to 10 equivalents of aqueous sodium hydroxide at about 75 to about 85° C. while applying a pressure of about 350 mbar, followed by stirring at about 75 to about 85° C. and under reduced pressure (<400 mbar) for 15 to 20 hours. During the entire reaction time distilled acetone/water is continuously replaced by water. The termination of the reaction can be monitored by taking a sample out of the reaction mixture after 15 to 20 hours and analyzing said sample with HPLC. A specific procedure for this reaction is disclosed in Example 7b.


The term “C1-C4 alkyl” as used herein means a linear or branched saturated hydrocarbon, such as for example methyl, ethyl, propyl, i-propyl, n-butyl, 2-butyl, tert-butyl, with methyl being especially preferred.


The term “alkali metal hydroxide” as used herein means sodium-, lithium- or potassium hydroxide, with lithium- and sodium hydroxide being especially preferred.


The term “reduced pressure” as used herein means <500 mbar, preferably <400 mbar.


The term “catalyst” as used herein in step aa) means any Pd(II)- or Pd(0)-catalyst usually used in this type of Sonogashira reaction. Examples of such catalysts are Pd(PPh3)4, PdCl2(PPh3)2, Pd(OAc)2, Pd2(dba)3 with bis-(triphenylphosphine)-palladium(II)-dichloride (PdCl2(PPh3)2) being especially preferred.


The term “base” as used herein in step aa) means weak bases as for example N-methylmorpholine or trialkyl-amines, with triethylamine being especially preferred.


The term “strong base” as used herein in step cc) means alkali metal alcoholates, preferably potassium tert.-butylate.


In an embodiment of the invention, step bb) is carried out under reduced pressure below 100 mbar.


In an embodiment, the catalyst in step aa) is PdCl2(PPh3)2 and the alkali metal hydroxide in step bb) is lithium- or sodium hydroxide.


In an embodiment, the compound of formula (IV) is dissolved in a mixture of water and N-methylpyrrolidone, and is reacted in the presence of aqueous sodium hydroxide at about 75 to about 85° C. and under reduced pressure for 15 to 20 hours to give directly the compound of formula (I) without the need to apply an aqueous work-up of the compound of formula (V).


In a particularly preferred embodiment according to the present invention, the above-mentioned reaction step aa) is carried out in the presence of bis-(triphenylphosphine)-palladium(II)-dichloride, copper(I)iodide and triethylamine, the reaction step bb) is carried out with aqueous lithium- or sodium hydroxide and the reaction step cc) is carried out in the presence of potassium tert.-butylate or aqueous sodium hydroxide. The elimination of acetone (step bb) and subsequent ring formation of step cc) are preferably carried out in N-methylpyrrolidone or in a mixture of N-methylpyrrolidone and water as solvent.


The process according to the reaction steps aa) to cc) is preferably carried out according to the specific conditions and parameters given in Example 7a) or b). Consequently, the specific process according to Example 7a) or b) form each another preferred embodiment according to the present invention.


The compounds of formula (II) can be obtained for example by iodination of the corresponding 5-halogenated 2-amino-pyridines. Among the many iodination conditions known to the skilled in the art a mixture of iodine and periodic acid proved to be especially suitable for the mentioned transformation.


The above described synthesis route via the compound of formula (IV) is a novel, alternative method for the production of the otherwise well known compounds of formula (I), in particular the 5-bromo-7-azaindole (3). A particular advantage of this process is the use of the intermediate compound of formula (IV) which can be easily purified and isolated from the reaction mixture. In addition, the use of compound (IV) avoids the release of environmentally hazardous Si by-products upon cyclization, which are formed if the corresponding trimethylsilyl-ethynyl derivative is used, which is usually known for this type of reaction (Sonogashira reaction) as inter alia described in WO 2009/016460 and US 2006/0183758.


The compounds of formula (IV) are thus valuable and novel intermediates in the above-described synthesis to obtain 5-halogenated 7-azaindoles (I). Therefore, in yet another embodiment according to the present invention there is provided the compounds of formula (IV) as such.


The present invention is now illustrated by the following accompanying working examples. In some examples, the term “In-process control” is used. This term means that a sample is taken from the reaction mixture while the process is running, and said sample is being analyzed by standard techniques known to the skilled person, preferably by HPLC, in order to detect the degree of conversion of starting material into product.


EXAMPLES
Example 1
(Step a))











Formation of Carboxylic Acid Chloride (step a) 1) according to scheme 1)

















55.8
g
Sulfonamide Acid (2) was placed into a dried 1st reaction vessel kept under




nitrogen atmosphere, to which


280
mL
methylenchloride were added. Then


619
μL
DMF were added to the obtained suspension and the resulting mixture was




kept at a temperature between 18-22° C. Then,


25.4
g
oxalylchloride were dissolved in


66
mL
methylenchloride, and this solution was slowly added over approximately 30









minutes to the above-mentioned suspension whereby the temperature of said



suspension was kept between 18-22° C. The formation of CO2 and CO could



be observed during said addition. The reaction mixture was then further



stirred for about 4 to 6 hours and further kept at a temperature between



18-22° C. until the suspension almost entirely turned into a solution and no gas



formation could be observed any more.







Friedel-Crafts-Acylation









In parallel to the above described formation of the acid-chloride, a 2nd



reaction vessel was prepared, wherein









106.7
g
aluminiumtrichloride were mixed together with


266
mL
methylenchloride to produce a suspension which was then cooled down to




about −12 to 2° C. In parallel, a suspension of


39.4
g
5-bromo-7-azaindole in


66
mL
methylenchloride was prepared in a 3rd dried reaction vessel under nitrogen




atmosphere. Said bromoazaindole suspension was added to said









aluminiumtrichloride suspension over approximately 30 minutes and at a



temperature between −12 to 2° C.



The obtained suspension was immediately further reacted by addition of the



acid-chloride solution as obtained according to the procedure described



above over approximately 30 minutes, whereby the reaction mixture was



allowed to warm up to about 20-25° C. using the spontaneous heat release



observed upon addition of the said acid chloride solution. After addition of



the acid-chloride solution, the reaction mixture was further stirred over



approximately 8 to 10 hours; whereby the mixture was kept at a temperature



between 20-25° C. During this time a separation into 2 phases was observed.



Meanwhile, a 4th reaction vessel was prepared, containing









400
ml
water which was cooled to a temperature between 0-5° C. The two phase









reaction mixture as obtained according to the preceding steps was added



slowly, over approximately 30 minutes, to said cooled water into said 4th



reaction vessel whereby the resulting mixture was kept at a temperature



between 0-20° C. This resulted in an exothermic reaction and precipitation of



the compound of formula (4) from the resulting biphasic reaction mixture.



Methylene chloride was widely removed by distillation under reduced



pressure from the heterogeneous mixture. Then the aqueous suspension of



the compound of formula (4) was diluted with methylene chloride and THF.



A clear biphasic mixture was obtained by heating the reaction mixture to



about 50° C. After phase separation the organic phase was washed twice



with 400 ml semi-saturated brine at about 50° C. The organic phase was



concentrated under reduced pressure at about 50° C. to a volume of about 400



ml whereby crystallization of the compound of formula (4) started.









600
ml
Heptane was added within about 30 min at about 50° C. The resulting









suspension was cooled in about 3-5 h to ca. 0° C. After stirring for at least



one additional hour at ca. 0° C. to complete the crystallization, the suspension



was filtered and the wet precipitate was washed two times with









120
mL
n-heptane. The wet product was dried in vacuum and at a temperature









between 50-60° C.









Yield: 85 g (=90%) light beige colored, crystalline azaindole (4).


Example 2
Step b











Formation of compound (5)

















45.8
g
of the compound (4) as obtained according to Example 1 were




suspended in


600
ml
toluene. Water as contained in the suspension was removed at a




temperature between 60-80° C. and under reduced pressure of 450-400 mbar. Subsequently,


200
ml
toluene were newly added and the suspension was cooled to 20-




25° C. Then, a solution of


1.22
g
dimethylaminopyridine in


20
ml
toluene was added, prior to the addition of


15.8
g
n-tripropylamine. Subsequently,


22.0
g
2,6-dichlorobenzoylchloride were slowly added via a dropping




funnel over approximately 15 minutes while the mixture was kept between 20 and 25° C.









The reaction mixture was stirred for about 1-2 hours at a temperature between 20-25 ° C., whereby the color of the mixture turned into brown.


The brownish reaction mixture as obtained by the last step above, was diluted with
















275
ml
water and subsequently with


29.6
g
hydrochloric acid (37%). The resulting two phase mixture was




heated to 65-70° C. The two phases were allowed to separate after about 10 minutes. The toluene




phase was washed at a temperature between 65 and 70° C., first with


300
ml
of an aqueous solution containing 10% sodium hydrogencarbonate,




and then with


300
ml
water. The organic (toluene) phase was concentrated by




evaporation at temperatures between 55 and 60° C. and at reduced pressure (200-80 mbar) to a




volume of about 200 ml. During this procedure the crude product (5) precipitated due to




crystallization. The resulting suspension was then slowly cooled down (within about 5 h) to −5 to




0° C. and further stirred at that temperature for 1 h. The crude product was separated by filtration,




washed twice with


30
ml
toluene (0° C.), and was subsequently dried at 50-55° C. and 26-13




mbar.









Yield: 57 g (90%) of compound of formula (5).


Example 3
Step c











Formation of compound (6), Suzuki-Coupling

















23.16
g
4-Chlorphenylboronic acid (5a) were mixed with


85.00
g
of compound (5) as obtained according to Example 2 in a dried 1st reaction




vessel under nitrogen atmosphere. To the resulting suspension were further




added


395
ml
anisole.




The suspension was kept at room temperature (20-25° C.) and mixed with a




solution of


57.08
g
sodium carbonate in


337
ml
water. The reaction mixture was then heated to a temperature of 70 +/− 2° C.




At this temperature,


0.236
g
bis-(triphenylphosphin)-palladium(II)-dichloride together with


110
ml
anisole were added to the reaction mixture, which was subsequently slowly




(within about 60 minutes) heated up to a temperature between 80-88° C.




(Heating Temperature outside did not exceed 110° C.) and stirred for about 2




h. Towards the end of the reaction the formation of a clear two phase




reaction mixture with a dark red upper (organic) phase was obtained.




After completion of the reaction, the reaction mixture was cooled down to




60-80° C. and transferred via a filter into a 2nd reaction vessel. The 1st




reaction vessel and the filter were washed with


110
ml
warm anisole which was added to the 2nd reaction vessel. The obtained two




phases were separated and the organic phase was washed at 60-80° C., first




with


375
ml
0.1N sulfuric acid containing 10% (w/w) sodium sulfate, and subsequently




with


375
ml
water. During the last extraction, reversal of the two phases was observed.









The organic phase was now the lower phase. In order to obtain complete



phase separation, the last extraction was carried out at a temperature of at



least 70° C. The obtained orange-yellow solution was concentrated at



reduced pressure to about 225 mL (+/−10%), while the temperature of the



solution was kept between 60-80° C. Subsequently, the resulting suspension



was cooled to about 60° C. Then, over a period of about 30 minutes,









628
ml
methanol were continuously added. Subsequently, the suspension was slowly









(within about 4 hours) cooled to 0° C. (+/−2° C.), and stirred for another 3



hours at that temperature until complete crystallization of the product of



formula (6) occurred. The suspension was separated by filtration, and the wet



solid residue was washed two times with









112
ml
cold methanol.









The wet chlorophenylbenzamide (6) was dried in a rotary evaporator under



reduced pressure and at a water bath temperature between 70-80° C.









Yield: 74-76 g (82-85%) almost colorless, crystalline Chlorophenylbenzamide (6). The product can be optionally re-crystallized from toluene for further purification.


1H-NMR (400 MHz, CDCl3): δ ppm 1.09 (t, J=7.5 Hz, 3H), 1.86-1.99 (m, 2H), 3.10-3.19 (m, 2H), 6.53 (s, 1H), 7.11 (dt, J=8.6, 1.5 Hz, 1H), 7.39-7.49 (m, 5H), 7.50-7.59 (m, 2H), 7.79 (td, J=9.0, 5.6 Hz, 1H), 8.32 (br. s, 1H), 8.43 (br. s, 1H), 8.84 (d, J=2.3 Hz, 1H).


Example 4
Step d











Formation of compound (1)

















70.0
g
Chlorphenylbenzamide (6),


175
ml
DMA and


88
ml
methanol were placed into a dried reaction vessel under nitrogen




atmosphere.




The resulting suspension was kept at 20-25° C., and mixed with


48.0
g
of a solution of ammonia in methanol (15%). The autoclave was then closed




and the reaction mixture heated to 50-55° C., which resulted in the




formation of a clear solution. The temperature was maintained while stirring




the reaction mixture for about 10-20 hours.




Subsequently, the clear solution was transferred into a double jacket reaction




vessel, and further diluted with


254
ml
methanol. The reaction mixture was then concentrated to its original volume




under reduced pressure and (600-500 mbar) at a jacket-temperature of




maximum 60° C. Subsequently,


508
ml
methanol were newly added slowly over a period of about 20-30 minutes,




whereby the temperature of the reaction mixture was kept between




45-55° C. The resulting suspension was slowly cooled down (within about 2 hours)




to 20° C. (±3° C.) and subsequently was further stirred for at least 1 hour




prior to separation of the solid reaction product (1) by filtration. The filter




cake was washed two times with


120
mL
methanol. The wet product was dried in rotary evaporator under reduced









pressure and while applying a water bath temperature of 50 to 60° C.









Yield: 49 g (95%) of the white, crystalline compound formula (1).


1H-NMR (600 MHz, CDCl3): δ ppm 1.07 (t, J=7.5 Hz, 3H), 1.84-1.98 (m, 2H), 3.07-3.17 (m, 2H), 6.41 (s, 1H), 7.06 (dt, J=8.6 Hz, 1.5 Hz, 1H), 7.46-7.51 (m, 2H), 7.60-7.64 (m, 2H), 7.70 (td, J=9.0, 5.5 Hz, 1H), 7.73 (d, J=2.7 Hz, 1H), 8.65 (d, J=2.2 Hz, 1H), 8.88 (d, J=1.9 Hz, 1H), 9.55 (br. s, 1H).


Purity: ≧99% (m/m, HPLC); Palladium content ≦5 ppm; compound (B): about 0.1%


Example 5
Preparation of 2-amino-5-bromo-3-iodopyridine (IIa)



embedded image












In a 1000 mL double-jacket reactor (under a nitrogen atmosphere)

















38.28
g
iodine is suspended in


21
g
acetic acid and


62
g
acetonitrile. To the brown mixture is added at 20 to 40° C.


14.6
g
sulfuric acid 96%. The addition is strongly exothermic. The dropping funnel is




rinsed with


20
g
water. The resulting mixture is heated with a jacket temperature of 90° C. When




the temperature of the mixture is 70° C., the mixture is treated within 3 to 6




minutes with


45.20
g
periodic acid (50% in water). The addition is endothermic. The funnel is rinsed




with


10
g
water. The solution is then treated at 65 to 75° C. within 5 to 10 minutes with a




previously prepared solution of


58.00
g
2-amino-5-bromopyridine in


67
g
acetonitrile and


31.5
g
acetic acid. The dropping funnel is rinsed with


15
g
acetonitrile. The resulting solution is heated to 77 to 82° C. and stirred under




slightly reflux conditions for 3 to 4 hours (approx. 90° C. jacket temperature). In-




process control (proposed target value: <2.0% starting material). Upon




complete conversion the mixture is immediately cooled down and treated at 60




to 70° C. drop wise with


66
g
sodium hydrogen sulfite (38-40% in water). Immediately after the addition, the




mixture is diluted at 60 to 70° C. within 30 to 60 minutes with


360
g
water. The mixture is then treated at 60 to 70° C. within 50 to 90 minutes with




approximately


~202
g
sodium hydroxide 28% to adjust the pH to 7.3 to 7.6. When the desired pH is




reached the suspension is stirred at 60 to 70° C. for 30 to 60 minutes. The




suspension is cooled to 20 to 25° C. within 2 to 5 hours and then stirred at this




temperature for 2 to 5 hours. The crystals are filtered off and washed in two




portions with a mixture of


270
g
water and


23
g
acetonitrile. The wet crystals (approx. 120 g) are dried at 40 to 50° C./<30 mbar




until constant weight.









Yield: 90.5 g of slightly brownish crystals with an assay of 95.0% (m/m). This corresponds to a corrected yield of 86%.


Example 6
a) Preparation of 4-(2-Amino-5-bromo-pyridin-3-yl)-2-methyl-but-3-Yn-2-Ol.



embedded image


A suspension of 2-amino-5-bromo-3-iodopyridine (10.0 g, 33.5 mmol), bis-(triphenyl-phosphine)-palladium(II)-dichloride (117 mg, 0.17 mmol), copper(I)iodide (79 mg, 0.41 mmol) and triethylamine (6.1 mL, 43.5 mmol) in dichloromethane (40 mL) was treated at 23 to 30° C. within 1 to 2 hours with a solution of 1,1-dimethyl-2-propyn-1-ol (3.70 g, 44 mmol) in dichloromethane (10 mL) and the resulting mixture was stirred at 25° C. for 3 hours. The mixture was diluted with dichloromethane (20 mL) and washed with water (2×50 mL). The organic phase was then treated with 1 M HCl (80 mL). The layers were separated and the organic layer was extracted with 1 M HCl (20 mL). The combined product containing aqueous layers were washed with dichloromethane (2×10 mL). The pH of the aqueous layer was adjusted to pH 7-9 by the drop wise addition of sodium hydroxide solution (28% in water, 18 g). The resulting suspension was stirred at 20° C. for 2 hours and the crystals were then filtered off and washed with water (2×20 mL). The wet crystals were dried at 50° C./<30 mbar affording 6.99 g (82%) of 4-(2-amino-5-bromo-pyridin-3-yl)-2-methyl-but-3-yn-2-ol as a melted mass with a purity of 99.5% (HPLC, area%). 1H NMR (400 MHz, CDCl3): δ ppm 1.63 (s, 6H); 4.0 (br., 3H); 7.59 (d, J=2.4, 1H); 8.03 (d, J=2.4, 1H).


b) Alternative Preparation of 4-(2-Amino-5-bromo-pyridin-3-Yl)-2-methyl-but-3-Yn-2-Ol.











In a 1000 mL double-jacket reactor (under a nitrogen atmosphere)

















84.0
g
2-amino-5-bromo-3-iodopyridine (96.4% (m/m)) is suspended in


900
g
dichloromethane. The suspension is heated to reflux (45° C. jacket temperature)









and stirred at slightly reflux for 15 to 45 minutes. The dimmished solution is



cooled to 30 to 35° C. and then polish-filtered (using a Zeta plus filter plate



charged with Decalite Speedex). The first reactor and the transfer pipe are rinsed



with









130
g
dichloromethane (pre-heated to 30 to 35° C.). The clear filtrate is concentrated to









a residual volume of 260 to 300 mL. The resulting suspension is treated at ca.



30° C. with









600
mg
bis-(triphenylphosphine)-palladium(II)-dichloride,


400
mg
copper(I)-iodide and


38.0
g
triethylamine. The used receivers are rinsed with


10
g
dichloromethane. The brown suspension is treated at 30 to 34° C. within 1 to 2




hours with a solution of


32.0
g
2-methyl-3-butin-2-ol in


120
g
dichloromethane. The dropping funnel is rinsed with


15
g
dichloromethane. The mixture is stirred for 10 hours at 30 to 34° C. In-process




control. Upon complete conversion the mixture is diluted at 30 to 34° C. with


240
g
dichloromethane and


200
g
water and treated at 28 to 34° C. within 10 to 20 minutes with


100
g
ammonium hydroxide solution (25% in water). The biphasic solution (ca. 950




mL) is stirred for 15 to 30 minutes at 28 to 34° C. and the layers are then allowed




to separate for 15 to 30 minutes. The organic layer is separated and the aqueous




layer is extracted at 28 to 34° C. with


80
g
dichloromethane. The combined organic layers are diluted at 30 to 34° C. with


100
g
water and then treated at 28 to 34° C. within 10 to 20 minutes with


50
g
ammonium hydroxide solution (25% in water). The biphasic solution is stirred









for 15 to 30 minutes at 28 to 34° C. and the layers are then allowed to separate for



15 to 30 minutes. The organic layer is separated and then washed at 28 to 34° C.



with









100
g
water. The organic layer is concentrated under reduced pressure and at a









maximum temperature of 34° C. to a volume of 550 to 600 mL. The organic layer



is diluted at 25 to 32° C. with









400
g
water and treated at 25 to 32° C. within 15 to 30 minutes with


45
g
hydrochloric acid (37% in water). The biphasic solution (980 mL) is stirred for









15 to 30 minutes at 25 to 32° C. and the layers are then allowed to separate for 30



to 60 minutes. The layers are separated and the organic layer is diluted with









225
g
water and then treated at 25 to 32° C. within 15 to 30 minutes with


25
g
hydrochloric acid (37% in water). The combined product containing aqueous









layers are washed at 25 to 32° C. with









100
g
dichloromethane. From the aqueous layer dichloromethane is then azeotropically









removed with water under reduced pressure and at a maximum internal



temperature of 32° C. At the end of the distillation a volume of 550 to 600 mL is



adjusted. The resulting aqueous solution is polish-filtered (using a Zeta plus



filter plate). The first reactor and the transfer pipes are rinsed with









40
g
water. The clear solution is then treated at 22 to 30° C. within 60 to 120 minutes









with approximately









54
g
sodium hydroxide (28% in water) until the pH is adjusted to pH 7.5 to 9.5. This









forces the product to precipitate. After the dosing the resulting suspension is



stirred at 20 to 25° C. for 4 to 16 hours. The crystals are filtered off and washed



in two portions with









300
g
water. The wet crystals (ca. 73 g) are dried at 40 to 50° C. and <30 mbar until









constant weight.









Yield: 65.04 g (93%) of yellow crystals with a content of 98.6% (m/m).


This intermediate can optionally be re-crystallized from isopropanol/water for further purification.



1H NMR (400 MHz, CDCl3): δ ppm 1.61 (s, 6H); 2.3 (br., 1H); 4.9 (br., 2H); 7.57 (d, J=2.4, 1H); 8.01 (d, J=2.4, 1H).


Example 7
a) Preparation of 5-bromo-7-azaindole (3 or Ia) from isolated 2-amino-5-bromo-3-iodopyridine



embedded image


A suspension of 2-amino-5-bromo-3-iodopyridine (5.0 g, 16.7 mmol), bis-(triphenyl-phosphine)-palladium(II)-dichloride (43 mg, 0.061 mmol), copper(I)iodide (29.4 mg, 0.15 mmol) and triethylamine (2.21 g, 21.8 mmol) in dichloromethane (20 mL) was treated at 23 to 30° C. within 1 to 2 hours with a solution of 1,1-dimethyl-2-propyn-1-ol (1.85 g, 21.7 mmol) in dichloromethane (10 mL) and the resulting mixture was stirred at 25° C. for 4 hours. The mixture was diluted with dichloromethane (10 mL) and washed with water (2×25 mL). The organic phase was then treated with 1 M HCl (40 mL). The layers were separated and the organic layer was extracted with 1 M HCl (15 mL). The combined product containing aqueous layers were washed with dichloromethane (2×8 mL). The pH of the aqueous layer was adjusted to pH 7-9 by the drop wise addition of sodium hydroxide solution (28% in water). The resulting suspension was stirred at 20° C. over night and the crystals were then filtered off and washed with water (2×5 mL). The wet crystals were dissolved in N-methylpyrrolidone (50 mL) and treated within 2 hours at 60° C. and 50-100 mbar with an aqueous solution of lithium hydroxide (2.4 M, 32 mL). The resulting mixture was heated to 75° C. and stirred at this temperature and under reduced pressure (50-100 mbar) for 15-20 hours. Toluene (20 mL) and water (20 mL) were then added and the layers were separated. The aqueous layer was extracted with toluene (3×25 mL). The combined organic layers were washed with water (3×10 mL) and then concentrated to dryness. The residue was dissolved in N-methylpyrrolidone (50 mL) and treated at 60° C. with potassium tert.-butylate (3.52 g, 30.7 mmol). After stirring for 3 hours at 60° C., the mixture was cooled to ambient temperature and diluted with toluene (40 mL) and water (40 mL). The aqueous layer was separated and back extracted with toluene (3×50 mL). The combined toluene layers were washed with water (3×10 mL) and then concentrated to dryness. The residue was dissolved in a hot mixture of toluene and n-heptane (20 mL). The clear solution was cooled to −5° C. within 4 to 6 hours whereupon crystals precipitated. The suspension was stirred at −5° C. for 2-4 hours. The crystals were filtered off, washed with heptane and dried at 45° C./<30 mbars over night to afford 5-bromo-7-azaindole (2.05 g, 62% yield) as slightly yellow crystals with a purity of 99.6% (HPLC, area%).


b) Preparation of 5-bromo-7-azaindole from isolated 4-(2-Amino-5-bromo-pyridin-3-yl)-2-methyl-but-3-yn-2-ol.



embedded image












A 1000 mL double-jacket reactor (under a nitrogen atmosphere) is charged with

















80.0
g
4-(2-amino-5-bromo-pyridin-3-yl)-2-methyl-but-3-yn-2-ol,


320
mL
N-methylpyrrolidone and


330
mL
water. The mixture is heated to 75 to 80° C. and a vacuum of ca 350 mbar is




applied. The solution is then treated at 75 to 80° C. within 30 to 45 minutes with


181
mL
sodium hydroxide (28% in water). The dropping funnel is rinsed with


5
mL
water and the mixture stirred at 78 to 81° C. for 15 to 20 hours. During stirring




the jacket temperature and the vacuum have to be adjusted such that the internal




temperature is 78 to 81° C. and a slight steadily distillate flow is guaranteed.




When the volume in the reactor has reached approx. 800 mL water is




continuously added to keep the volume constant for the rest of the reaction time.




In-process control. Upon complete conversion, the reaction mixture is




concentrated to a volume of approx. 700 mL and then cooled to 50 to 55° C. The




mixture is treated at this temperature with


200
mL
toluene. The biphasic mixture (ca. 900 mL) is stirred at 50 to 55° C. for 15 to 30




minutes and the layers are then allowed to separate for 15 to 30 minutes. The




aqueous layer is separated and then extracted at 50 to 55° C. with 3 × 140 mL,




totally with


420
mL
toluene. The combined toluene layers are washed at 50 to 55° C. with 2 × 100 mL,




totally with


200
mL
water. The toluene layer is concentrated under reduced pressure at 45 to 55° C.




until a residual volume of 450 to 500 mL is obtained. The residue is treated at 50




to 55° C. with


225
g
ethyl acetate and the resulting solution is washed at 50 to 55° C. with 3 × 150 mL,




totally with


450
mL
water. From the organic layer, water and ethyl acetate are azeotropically distilled




off with toluene under reduced pressure at 45 to 55° C. At the end of the




distillation a volume of 600 to 700 mL is adjusted. The mixture is heated to 90




to 95° C. and stirred until a clear solution is obtained. The solution is treated with


2.0
g
activated charcoal (Norit SX) and the resulting mixture stirred for 15 to 30




minutes at 90 to 95° C. The charcoal is removed by a hot filtration at 90 to 95° C.




The first reactor, the filter and the transfer pipes are washed with 3 × 100 mL,




totally with


300
mL
toluene. The filtrate is concentrated under reduced pressure to a volume of




approx. 400 mL. The resulting suspension is heated to 90 to 100° C. to obtain a




clear solution. The solution is cooled to −5 to −10° C. within 7 to 10 hours and the




resulting suspension stirred at this temperature for additional 3 to 5 hours. The




crystals are filtered off and washed in two portions with


120
mL
toluene (pre-cooled to <0° C.). The wet crystals are dried at 55 to 65° C./<30









mbar until constant weight.









Yield: 46.5 g (75%) of slightly yellow crystals with an assay of 100.1% (m/m).


Example 8
Preparation of 5-Chloro-7-Azaindol (Ib)
Step 1: Synthesis of 2-Amino-5-chloro-3-iodopyridine (IIb)



embedded image












In a 1000 mL double-jacket reactor under a nitrogen atmosphere

















38.28
g
iodine is suspended in


21
g
acetic acid and


62
g
acetonitrile. To the brown mixture is added at 20 to 40° C.


14.6
g
sulfuric acid 96%. The addition is strongly exothermic. The dropping funnel is




rinsed with


20
g
water. The resulting mixture is heated with a jacket temperature of 90° C. When




the temperature of the mixture is 70° C., the mixture is treated within 3 to 6




minutes with


45.20
g
periodic acid (50% in water). The addition is endothermic. The funnel is rinsed




with


10
g
water. The solution is then treated at 65 to 75° C. within 5 to 10 minutes with a




previously prepared solution of


43.1
g
2-amino-5-chloropyridine in


67
g
acetonitrile and


31.5
g
acetic acid. The dropping funnel is rinsed with


15
g
acetonitrile. The resulting solution is heated to 77 to 82° C. and stirred under




slightly reflux conditions for 4 hours (approx. 90° C. jacket temperature). The




mixture is then cooled 60-65° C. and treated with


66
g
sodium hydrogen sulfite (39% in water). After the addition, the mixture is




diluted at 60 to 70° C. within 10 to 20 minutes with


360
g
water. The mixture is then treated with


162
mL
sodium hydroxide 28% to adjust the pH to pH 7.4 and the resulting suspension




is stirred at 50° C. for 30 to 60 minutes. The suspension is cooled to 20 to 25° C.




within 2 hours and then stirred at this temperature over night. The crystals are




filtered off and washed in two portions with a mixture of


270
g
water and


23
g
acetonitrile. The wet crystals (approx. 110 g) are dried at 40-50° C./<30 mbar









to afford 72.5 g (85%) of slightly brownish crystals with a purity of 94.8



% (area).









Step 2: Synthesis of 4-(2-Amino-5-chloro-pyridin-3-yl)-2-methyl-but-3-yn-2-ol



embedded image












In a 1000 mL double-jacket reactor

















38.0
g
2-amino-5-chloro-3-iodopyridine is suspended in


120
mL
dichloromethane. The suspension is treated at ca 30° C. with


0.60
g
bis-(triphenylphosphine)-palladium(II)-dichloride,


0.41
g
copper(I)-iodide and


27.5
mL
triethylamine. The used receivers are rinsed with


10
g
dichloromethane. The brown suspension is treated at 30 to 34° C. within 1 to 2




hours with a solution of


16.8
g
2-methyl-3-butin-2-ol in


60
mL
dichloromethane. The dropping funnel is rinsed with


5
mL
dichloromethane. The mixture is stirred for 3 hours at 30 to 34° C. and then




treated at 30 to 34° C. with


100
mL
dichloromethane and


150
mL
ammonium hydroxide solution (10% in water). The biphasic solution is stirred




for 10 to 20 minutes at 30 to 34° C. and the layers are then allowed to separate for




15 to 45 minutes. The organic layer is separated and the aqueous layer is




extracted at 30 to 34° C. with


40
mL
dichloromethane. The combined organic layers are washed at 28 to 34° C. with


150
mL
ammonium hydroxide solution (10% in water) and then with


150
mL
water. The organic layer is then treated at 25 to 32° C. with


300
mL
hydrochloric acid (1.0M in water). The biphasic solution is stirred for 20 to 30




minutes at 25 to 32° C. and the layers are then allowed to separate for 30 to 60




minutes. The organic layer is separated and extracted at 25 to 32° C. with


100
mL
hydrochloric acid (1.0M in water). The combined product containing aqueous




layers are washed at 25 to 32° C. with


100
mL
dichloromethane. From the aqueous layer dichloromethane is then azeotropically




removed with water under reduced pressure and at a maximum internal




temperature of 30° C. The aqueous solution is then treated at 22 to 30° C. within




60 to 120 minutes with approximately


32
mL
sodium hydroxide (28% in water) until the pH is adjusted to pH 9. This forces




the product to precipitate. After the dosing the resulting suspension is stirred at




20 to 25° C. over night. The crystals are filtered off and washed in two portions




with


150
g
water. The wet crystals (40.2 g) are dried at 40 to 50° C. and <30 mbar until









constant weight to afford 29.2 g (92%) of slightly brownish crystals with a



purity of 98.7% (area).









Step 3: Synthesis of 5-chloro-7-azaindol (Ib)



embedded image












A 500 mL double-jacket reactor (under a nitrogen atmosphere) is charged with

















25.0
g
4-(2-amino-5-chloro-pyridin-3-yl)-2-methyl-but-3-yn-2-ol,


120
mL
N-methylpyrrolidone and


130
mL
water. The mixture is heated to 75 to 80° C. (ca. 95° C. jacket temperature) and a









vacuum of ca. 350 mbar is applied. The solution is then treated at 75 to 80° C.



within 30 to 45 minutes with









85
mL
sodium hydroxide (28% in water). The dropping funnel is rinsed with


5
mL
water and the mixture stirred at 78 to 81° C. over night. During stirring the jacket









temperature and the vacuum have to be adjusted such that a slight steadily



distillate flow is guaranteed. In a typical lab experiment approx. 50 mL of



water/acetone are distilled off in 2 hours. During the reaction, water is conti-



nuously added to keep the volume constant at approx. 270 mL. Upon complete



conversion, the reaction mixture is cooled to 50 to 55° C. The mixture is treated at



this temperature with









60
mL
toluene. The biphasic mixture is stirred at 50 to 55° C. for 15 to 30 minutes and the









layers are then allowed to separate for 15 to 30 minutes. The aqueous layer is



separated and then extracted at 50 to 55° C. with 3 × 50 mL with



toluene. The combined toluene layers are washed at 50 to 55° C. with 5 × 40 mL with



water. The toluene layer is concentrated to dryness. The residue (17.3 g) is



crystallized from









90
mL
toluene to afford 13.0 g (71%) of 5 chloro-7-azaindol (Ib) as slightly yellow









crystals with a purity of 96.7% (area).








Claims
  • 1. A composition comprising compound (B),
  • 2. The composition according to claim 1, wherein said compound (B) is present in an amount of less than 0.30% by weight of the composition.
  • 3. The composition according to claim 1, wherein said compound (B) is present in an amount of from about 0.01% to about 0.15% by weight of the composition.
  • 4. The composition according to claim 1, wherein said compound (B) is present in an amount of from about 0.02% to about 0.15% by weight of the composition.
  • 5. The composition according to claim 1, wherein said compound (B) in the composition is detectable by high performance liquid chromatography, mass spectrometry, proton nuclear magnetic resonance spectroscopy, infrared spectroscopy, or a combination thereof.
  • 6. The composition according to claim 5, wherein said compound (B) in the composition is detectable by high performance liquid chromatography.
  • 7. The composition according to claim 5, wherein said compound (B) in the composition is detectable by high performance liquid chromatography in combination with mass spectrometry.
  • 8. The composition according to claim 2, wherein said compound (B) in the composition is detectable by high performance liquid chromatography, mass spectrometry, proton nuclear magnetic resonance spectroscopy, infrared spectroscopy, or a combination thereof.
  • 9. The composition according to claim 8, wherein said compound (B) in the composition is detectable by high performance liquid chromatography.
  • 10. The composition according to claim 8, wherein said compound (B) in the composition is detectable by high performance liquid chromatography in combination with mass spectrometry.
  • 11. The composition according to claim 3, wherein said compound (B) in the composition is detectable by high performance liquid chromatography, mass spectrometry, proton nuclear magnetic resonance spectroscopy, infrared spectroscopy, or a combination thereof.
  • 12. The composition according to claim 11, wherein said compound (B) in the composition is detectable by high performance liquid chromatography.
  • 13. The composition according to claim 11, wherein said compound (B) in the composition is detectable by high performance liquid chromatography in combination with mass spectrometry.
  • 14. The composition according to claim 4, wherein said compound (B) in the composition is detectable by high performance liquid chromatography, mass spectrometry, proton nuclear magnetic resonance spectroscopy, infrared spectroscopy, or a combination thereof.
  • 15. The composition according to claim 14, wherein said compound (B) in the composition is detectable by high performance liquid chromatography.
  • 16. The composition according to claim 14, wherein said compound (B) in the composition is detectable by high performance liquid chromatography in combination with mass spectrometry.
  • 17. A composition comprising compound (B):
  • 18. The composition according to claim 17, wherein said compound (B) is present in an amount of less than 0.30% by weight of the composition.
  • 19. The composition according to claim 17, wherein said compound (B) is present in an amount of from about 0.01% to about 0.15% by weight of the composition.
  • 20. The composition according to claim 17, wherein said compound (B) is present in an amount of from about 0.2% to about 0.15% by weight of the composition.
  • 21. The composition according to claim 17, wherein said propane-1-sulfonic acid {3-[5-(4-chloro-phenyl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl}-amide is present in an amount of greater than 99% by weight of the composition.
  • 22. The composition according to claim 18, wherein said propane-1-sulfonic acid {3-[5-(4-chloro-phenyl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl}-amide is present in an amount of greater than 99% by weight of the composition.
  • 23. The composition according to claim 19, wherein said propane-1-sulfonic acid {3-[5-(4-chloro-phenyl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl}-amide is present in an amount of greater than 99% by weight of the composition.
  • 24. The composition according to claim 20, wherein said propane-1-sulfonic acid {3-[5-(4-chloro-phenyl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl}-amide is present in an amount of greater than 99% by weight of the composition.
  • 25. A composition comprising: (a) a mixture of compound (B):
  • 26. The composition according to claim 25, wherein said compound (B) is present in an amount of less than 0.30% by weight of the mixture.
  • 27. The composition according to claim 25, wherein said compound (B) is present in an amount of from about 0.01% to about 0.15% by weight of the mixture.
  • 28. The composition according to claim 25, wherein said compound (B) is present in an amount of from about 0.02% to about 0.15% by weight of the mixture.
  • 29. A composition comprising compound (B):
  • 30. The composition according to claim 29, wherein said compound (B) is present in an amount of less than 0.30% by weight of the composition.
  • 31. The composition according to claim 29, wherein said compound (B) is present in an amount of from about 0.01% to about 0.15% by weight of the composition.
  • 32. The composition according to claim 29, wherein said compound (B) is present in an amount of from about 0.02% to about 0.15% by weight of the composition.
  • 33. The composition according to claim 29, wherein said compound (B) in the composition is detectable by high performance liquid chromatography, mass spectrometry, proton nuclear magnetic resonance spectroscopy, infrared spectroscopy, or a combination thereof.
  • 34. The composition according to claim 33, wherein said compound (B) in the composition is detectable by high performance liquid chromatography.
  • 35. The composition according to claim 33, wherein said compound (B) in the composition is detectable by high performance liquid chromatography in combination with mass spectrometry.
  • 36. A compound that is compound (B):
Priority Claims (2)
Number Date Country Kind
09167054 Aug 2009 EP regional
09175101 Nov 2009 EP regional
PRIORITY TO RELATED APPLICATION(S)

This application is a divisional application of U.S. application Ser. No. 12/843,908, filed Jul. 27, 2010, and claims the benefit of European Patent Application No. 09167054.7, filed Aug. 3, 2009, and European Patent Application No. 09175101.6, filed Nov. 5, 2009, both of which are hereby incorporated by reference in their entirety.

US Referenced Citations (160)
Number Name Date Kind
2234705 Normington et al. Apr 1940 A
2413258 Soday Dec 1946 A
3524860 Alberstson et al. Aug 1970 A
4150949 Smith Apr 1979 A
4301159 Ogata et al. Nov 1981 A
4439444 Nisato et al. Mar 1984 A
4568649 Bertoglio-Matte Feb 1986 A
4595780 Ogata et al. Jun 1986 A
4626513 Burton et al. Dec 1986 A
4634701 De Vincentiis Jan 1987 A
4714693 Targos Dec 1987 A
4727395 Oda et al. Feb 1988 A
4863945 Friebe et al. Sep 1989 A
5120782 Hubsch et al. Jun 1992 A
5124335 Patchett et al. Jun 1992 A
5143854 Pirrung et al. Sep 1992 A
5338849 Festal et al. Aug 1994 A
5360822 Morino et al. Nov 1994 A
5426039 Wallace et al. Jun 1995 A
5432177 Baker et al. Jul 1995 A
5434049 Okano et al. Jul 1995 A
5449614 Danos et al. Sep 1995 A
5474935 Chattejee et al. Dec 1995 A
5486525 Summers et al. Jan 1996 A
5556752 Lockhart et al. Sep 1996 A
5576319 Baker et al. Nov 1996 A
5580859 Felgner et al. Dec 1996 A
5631236 Woo et al. May 1997 A
5632957 Heller et al. May 1997 A
5658775 Gilboa Aug 1997 A
5681959 Bishop et al. Oct 1997 A
5700637 Southern Dec 1997 A
5700809 Leeson et al. Dec 1997 A
5712285 Curtis et al. Jan 1998 A
5721118 Scheffler Feb 1998 A
5721246 Yoshino et al. Feb 1998 A
5744305 Fodor et al. Apr 1998 A
5747276 Hoch et al. May 1998 A
5763198 Hirth et al. Jun 1998 A
5770456 Holmes Jun 1998 A
5800992 Fodor et al. Sep 1998 A
5807522 Brown et al. Sep 1998 A
5830645 Pinkel et al. Nov 1998 A
5840485 Lebl et al. Nov 1998 A
5856174 Lipshutz et al. Jan 1999 A
5858995 Kawai et al. Jan 1999 A
5877007 Housey Mar 1999 A
5952362 Cournoyer et al. Sep 1999 A
5958944 Arita et al. Sep 1999 A
5959098 Goldberg et al. Sep 1999 A
5965452 Kovacs Oct 1999 A
6013440 Lipshutz et al. Jan 2000 A
6022963 McGall et al. Feb 2000 A
6025155 Hadlaczkv et al. Feb 2000 A
6045996 Cronin et al. Apr 2000 A
6048695 Bradley et al. Apr 2000 A
6054270 Southern Apr 2000 A
6090912 Lebl et al. Jul 2000 A
6096718 Weitzman et al. Aug 2000 A
6107478 Pedersen et al. Aug 2000 A
6110456 Durina Aug 2000 A
6110458 Freeman et al. Aug 2000 A
6113913 Brough et al. Sep 2000 A
6117681 Salmons et al. Sep 2000 A
6161776 Byles Dec 2000 A
6178384 Kolossvary Jan 2001 B1
6235769 Clary May 2001 B1
6243980 Bronstein et al. Jun 2001 B1
6258606 Kovacs Jul 2001 B1
6261776 Pirrung et al. Jul 2001 B1
6277489 Abbott et al. Aug 2001 B1
6277628 Johann et al. Aug 2001 B1
6288234 Griffin Sep 2001 B1
6294330 Michnick et al. Sep 2001 B1
6310074 Depreux et al. Oct 2001 B1
6348474 Kayakiri et al. Feb 2002 B1
6350786 Albano et al. Feb 2002 B1
6451825 Masayoshi et al. Sep 2002 B1
6476045 Dinnell et al. Nov 2002 B1
6545014 Verner Apr 2003 B2
6653309 Saunders et al. Nov 2003 B1
6673812 Azuma et al. Jan 2004 B1
6787534 Haneda et al. Sep 2004 B2
6858860 Hosono et al. Feb 2005 B2
6897207 Cox et al. May 2005 B2
7109208 Takayama et al. Sep 2006 B2
7122318 Ono et al. Oct 2006 B2
7259165 Bernotas et al. Aug 2007 B2
7361763 Arnold et al. Apr 2008 B2
7361764 Arnold et al. Apr 2008 B2
7452993 Arnold et al. Nov 2008 B2
7498342 Ibrahim et al. Mar 2009 B2
7504509 Ibrahim et al. Mar 2009 B2
7582637 Arnold et al. Sep 2009 B2
7601839 Arnold et al. Oct 2009 B2
7626021 Arnold et al. Dec 2009 B2
7846941 Zhang et al. Dec 2010 B2
7863288 Ibrahim et al. Jan 2011 B2
7863289 Spevak et al. Jan 2011 B2
7893075 Zhang et al. Feb 2011 B2
7947706 Tabart et al. May 2011 B2
7994185 Rheault Aug 2011 B2
8067434 Ibrahim et al. Nov 2011 B2
8067638 Kai et al. Nov 2011 B2
8143271 Ibrahim et al. Mar 2012 B2
8198273 Ibrahim et al. Jun 2012 B2
8329724 Hildbrand et al. Dec 2012 B2
8415469 Ibrahim et al. Apr 2013 B2
8470818 Ibrahim et al. Jun 2013 B2
8530661 Hildbrand et al. Sep 2013 B2
20010008765 Shinoki et al. Jul 2001 A1
20010012537 Anderson et al. Aug 2001 A1
20010014448 Chappa et al. Aug 2001 A1
20010014449 Nerenberg et al. Aug 2001 A1
20010016322 Caren et al. Aug 2001 A1
20010018642 Balaban et al. Aug 2001 A1
20010019827 Dawson et al. Sep 2001 A1
20020028809 Imoto et al. Mar 2002 A1
20030003004 Stones et al. Jan 2003 A1
20030219489 Curatolo et al. Nov 2003 A1
20040002534 Lipson et al. Jan 2004 A1
20040022534 Amano et al. Feb 2004 A1
20040077595 Cheng et al. Apr 2004 A1
20040142864 Bremer et al. Jul 2004 A1
20040167030 Bernotas et al. Aug 2004 A1
20040180947 Kayakiri et al. Sep 2004 A1
20050014942 Maruyama et al. Jan 2005 A1
20050026792 Cartwright Feb 2005 A1
20050085463 Weiner et al. Apr 2005 A1
20050154014 Bloxham et al. Jul 2005 A1
20050164300 Artis et al. Jul 2005 A1
20050170431 Ibrahim et al. Aug 2005 A1
20050256151 Salom et al. Nov 2005 A1
20060018726 Hall Jan 2006 A1
20060024361 Odidi et al. Feb 2006 A1
20060035898 Arnold et al. Feb 2006 A1
20060058339 Ibrahim et al. Mar 2006 A1
20060058340 Ibrahim et al. Mar 2006 A1
20060183758 Beard et al. Aug 2006 A1
20070032519 Zhang et al. Feb 2007 A1
20070049615 Ibrahim et al. Mar 2007 A1
20070066641 Ibrahim et al. Mar 2007 A1
20070072862 Dimauro et al. Mar 2007 A1
20070218138 Bittorf et al. Sep 2007 A1
20070287711 Arnold et al. Dec 2007 A1
20080167338 Spevak et al. Jul 2008 A1
20080176892 Heinrich et al. Jul 2008 A1
20080188514 Wu et al. Aug 2008 A1
20080207700 Werz et al. Aug 2008 A1
20090005356 Blaney et al. Jan 2009 A1
20090076046 Zhang et al. Mar 2009 A1
20090143352 Arnold et al. Jun 2009 A1
20090306056 Arnold et al. Dec 2009 A1
20100310659 Desai et al. Dec 2010 A1
20120022258 Brumsted et al. Jan 2012 A1
20120053177 Ibrahim et al. Mar 2012 A1
20120122860 Visor et al. May 2012 A1
20120245174 Ibrahim et al. Sep 2012 A1
20130261117 Ibrahim et al. Oct 2013 A1
20130303534 Ibrahim et al. Nov 2013 A1
Foreign Referenced Citations (180)
Number Date Country
2550361 Jul 2005 CA
2413258 Oct 1975 DE
0 148 725 Jul 1985 EP
0 154 734 Sep 1985 EP
0 344 603 Dec 1989 EP
0 465 970 Jan 1992 EP
0 580 860 Feb 1994 EP
0 596 406 May 1994 EP
0 870 768 Oct 1998 EP
0 901 786 Mar 1999 EP
0 988 863 Mar 2000 EP
1 057 826 Dec 2000 EP
1 267 111 Dec 2002 EP
1 368 001 Oct 2005 EP
1 749 829 Feb 2007 EP
2 264 804 Oct 1975 FR
1 198 301 Jul 1970 GB
1 451 299 Sep 1976 GB
2 292 143 Feb 1996 GB
2 292 145 Feb 1996 GB
2 298 198 Aug 1996 GB
2 299 581 Oct 1996 GB
6-135946 May 1994 JP
10-130269 May 1998 JP
11-284336 Oct 1999 JP
2001-278886 Oct 2001 JP
2003-73357 Mar 2003 JP
WO-9313099 Jul 1993 WO
WO-9414808 Jul 1994 WO
WO-9420459 Sep 1994 WO
WO-9420497 Sep 1994 WO
WO-9504742 Feb 1995 WO
WO-9507910 Mar 1995 WO
WO-9528387 Oct 1995 WO
WO-9600226 Jan 1996 WO
WO-9605200 Feb 1996 WO
WO-9611929 Apr 1996 WO
WO-9617958 Jun 1996 WO
WO-9618738 Jun 1996 WO
WO-9638131 Dec 1996 WO
WO-9703967 Feb 1997 WO
WO-9746313 Dec 1997 WO
WO-9746558 Dec 1997 WO
WO-9749703 Dec 1997 WO
WO-9806433 Feb 1998 WO
WO-9822457 May 1998 WO
WO-9847899 Oct 1998 WO
WO-9858935 Dec 1998 WO
WO-9900386 Jan 1999 WO
WO-9909217 Feb 1999 WO
WO-9932433 Jul 1999 WO
WO-9951231 Oct 1999 WO
WO-9951232 Oct 1999 WO
WO-9951233 Oct 1999 WO
WO-9951234 Oct 1999 WO
WO-9951595 Oct 1999 WO
WO-9951596 Oct 1999 WO
WO-9951773 Oct 1999 WO
WO-0009162 Feb 2000 WO
WO-0012074 Mar 2000 WO
WO-0012514 Mar 2000 WO
WO-0017202 Mar 2000 WO
WO-0029411 May 2000 WO
WO-0053582 Sep 2000 WO
WO-0055153 Sep 2000 WO
WO-0064898 Nov 2000 WO
WO-0071506 Nov 2000 WO
WO-0071537 Nov 2000 WO
WO-0075139 Dec 2000 WO
WO-0109121 Feb 2001 WO
WO-0124236 Apr 2001 WO
WO-0129036 Apr 2001 WO
WO-0146196 Jun 2001 WO
WO-0160822 Aug 2001 WO
WO-0162255 Aug 2001 WO
WO-0174786 Oct 2001 WO
WO-0198299 Dec 2001 WO
WO-0200657 Jan 2002 WO
WO-0218346 Mar 2002 WO
WO-02083175 Oct 2002 WO
WO-02085896 Oct 2002 WO
WO-02102783 Dec 2002 WO
WO-03000258 Jan 2003 WO
WO-03000267 Jan 2003 WO
WO-03003004 Jan 2003 WO
WO-03004472 Jan 2003 WO
WO-03006459 Jan 2003 WO
WO-03008422 Jan 2003 WO
WO-03011868 Feb 2003 WO
WO-03020698 Mar 2003 WO
WO-03028724 Apr 2003 WO
WO-03037862 May 2003 WO
WO-03051838 Jun 2003 WO
WO-03064413 Aug 2003 WO
WO-03068221 Aug 2003 WO
WO-03082289 Oct 2003 WO
WO-03082868 Oct 2003 WO
WO-03082869 Oct 2003 WO
WO-03087087 Oct 2003 WO
WO-03101990 Dec 2003 WO
WO-2004005283 Jan 2004 WO
WO-2004009600 Jan 2004 WO
WO-2004009601 Jan 2004 WO
WO-2004014369 Feb 2004 WO
WO-2004016609 Feb 2004 WO
WO-2004016610 Feb 2004 WO
WO-2004024895 Mar 2004 WO
WO-2004052880 Jun 2004 WO
WO-2004054581 Jul 2004 WO
WO-2004056830 Jul 2004 WO
WO-2004065393 Aug 2004 WO
WO-2004065394 Aug 2004 WO
WO-2004069138 Aug 2004 WO
WO-2004074286 Sep 2004 WO
WO-2004078756 Sep 2004 WO
WO-2004078923 Sep 2004 WO
WO-2004101565 Nov 2004 WO
WO-2005028475 Mar 2005 WO
WO-2005028624 Mar 2005 WO
WO-2005030128 Apr 2005 WO
WO-2005034869 Apr 2005 WO
WO-2005044181 May 2005 WO
WO-2005058891 Jun 2005 WO
WO-2005062795 Jul 2005 WO
WO-2005063746 Jul 2005 WO
WO-2005063747 Jul 2005 WO
WO-2005066347 Jul 2005 WO
WO-2005082367 Sep 2005 WO
WO-2005085244 Sep 2005 WO
WO-2005086904 Sep 2005 WO
WO-2005092896 Oct 2005 WO
WO-2005095400 Oct 2005 WO
WO-2005103050 Nov 2005 WO
WO-2005115363 Dec 2005 WO
WO-2005115374 Dec 2005 WO
WO-2006004984 Jan 2006 WO
WO-2006009755 Jan 2006 WO
WO-2006009797 Jan 2006 WO
WO-2006010637 Feb 2006 WO
WO-2006010637 Feb 2006 WO
WO-2006015123 Feb 2006 WO
WO-2006015124 Feb 2006 WO
WO-2006063167 Jun 2006 WO
WO-2006114180 Nov 2006 WO
WO-2006114520 Nov 2006 WO
WO-2006114520 Nov 2006 WO
WO-2006127587 Nov 2006 WO
WO-2006137376 Dec 2006 WO
2007002325 Jan 2007 WO
2007002433 Jan 2007 WO
2007002325 Jan 2007 WO
WO-2007002325 Jan 2007 WO
WO-2007002433 Jan 2007 WO
WO-2007009799 Jan 2007 WO
WO-2007013896 Feb 2007 WO
WO-2007013896 Feb 2007 WO
WO-2007022380 Feb 2007 WO
WO-2007022380 Feb 2007 WO
WO-2007106236 Sep 2007 WO
WO-2007106236 Sep 2007 WO
WO-2008058341 May 2008 WO
WO-2008064265 May 2008 WO
WO-2008064265 May 2008 WO
WO-2008065417 Jun 2008 WO
WO-2008079903 Jul 2008 WO
WO-2008079906 Jul 2008 WO
WO-2008079909 Jul 2008 WO
WO-2008138755 Nov 2008 WO
WO-2008138755 Nov 2008 WO
WO-2009012283 Jan 2009 WO
WO-2009012791 Jan 2009 WO
2009016460 Feb 2009 WO
WO-2009111277 Sep 2009 WO
WO-2009111277 Sep 2009 WO
WO-2009111278 Sep 2009 WO
WO-2009111278 Sep 2009 WO
WO-2009111279 Sep 2009 WO
WO-2009111280 Sep 2009 WO
WO-2010114928 Oct 2010 WO
WO-2010114928 Oct 2010 WO
Non-Patent Literature Citations (156)
Entry
Intl Search Report for PCT/EP2010/061079 dated Feb. 17, 2011.
International Search Report mailed on Dec. 17, 2012 for PCT Patent Application No. PCT/EP2011/062207 filed on Jul. 18, 2011, eight pages.
Written Opinion of the International Searching Authority mailed on Dec. 17, 2012 for PCT Patent Application No. PCT/EP2011/062207 filed on Jul. 18, 2011, ten pages.
Kumar, V. et al. (Dec. 4, 1992). “Synthesis of 7-Azaindole and 7-Azaoxindole Derivatives Through a Palladium-Catalyzed Cross-Coupling Reaction,” Journal of Organic Chemistry 57(25):6995-6998.
Pillard, C. et al. (May 2008). “Synthesis of 4-,5- and 6-Benzoylated 7-Azaindoles,” Synthesis 13:2049-2054.
Satoh, M. et al. (Apr. 1987). “Palladium-Catalyzed Cross-Coupling Reaction of (1-Ethoxy-1-alken-2-yl)Boranes with Ortho-Functionalized lodoarenes. A Novel and Convenient Synthesis of Benzo-Fused Heteroaromatic Compounds,” Synthesis 373-377.
Whelligan, D.K. et al. (2010, e-pub. Dec. 1, 2009). “Two-Step Synthesis of Aza- and Diazaindoles from Chloroamino-N-Heterocycles Using Ethoxyvinylborolane,” J. Org. Chem. 75(1):11-15.
Zhao, S-B. et al. (2010, e-pub. Dec. 31, 2009). “New Bimetallic Reactivity in Pt2II,II/Pt2IV,IV Transformation Mediated by a Benzene Ring,” Organometallics 29(4): 998-1003.
Allegretti et al. (2001). “Palladium-Catalysed Functionalisation at 4- and 6-Position of the 7-Azaindole System,” Synlett 5:609-612.
Al-Obeidi et al. (1998). “Peptide and Peptidomimetic Libraries,” Mol. Biotechnol. 9(3):205-223.
Alvarez et al. (1999). “Synthesis of 3-Aryl- and 3-Heteroaryl-7-Azaindoles” Synthesis 4:615-620.
Anderson et al. (1998). “Cooperative Catalyst Effects in Palladium-Mediated Cyanation Reactions of Aryl Halides and Triflates,” J. Org. Chem. 63:8224-8228.
Antonini et al. (1982). “Synthesis of 4-Amino-1-β-D-ribofuranosy1-1H-pyrrolo[2,3-b]pyridine (1-Deazatubercidin) as a Potential Antitumor Agent,” J. Med. Chem. 25:1258-1261.
Arbiser (2007). “Why targeted therapy hasn't worked in advanced cancer,” J. Clin. Invest. 117(10):2762-2765.
Bartlett et al. (1989). “CAVEAT: A Program to Facilitate the Structure-derived Design of Biologically Active Molecules,” in Molecular Recognition: Chemical and Biological Problems, S.M. Roberts (ed.), Royal Society of Chemistry, pp. 182-196.
Barton et al. (1987). “The chemistry of pentavalent organobismuth reagents. Part X. Studies on the phenylation and oxidation of phenols,” Tetrahedron 43(2):323-332.
Bertolini et al. (1997). “A New Rational Hypothesis for the Pharmacophore of the Active Metabolite of Leflunomide, a Potent Immunosuppressive Drug,” J. Med. Chem. 40:2011-2016.
Bloom, A. et al. (1939). “The Preparation of 2-Alkylaminobenzimidazoles,” J. Org. Chem. 14(17):14-19.
Blundell et al. (1988). “Knowledge-Based Protein Modelling and Design,” Eur. J. Biochem. 172:513-520.
Bohm, H. (1994). “On the Use of LUDI to Search the Fine Chemicals Directory for Ligands of Proteins of Known Three-Dimensional Structure,” J. Comp. Aided Molec. Des. 8:623-632.
Brenner et al. (1992). “Encoded Combinatorial Chemistry,” PNAS 89:5381-5383.
Capon et al. (1989). “Designing CD4 Immunoadhesins for AIDS Therapy,” Nature 337:525-531.
Carell et al. (1995). “New Promise in Combinatorial Chemistry: Synthesis, Characterization, and Screening of Small-Molecule Libraries in Solution,” Chem. Biol. 3:171-183.
Chabala, J.C. (1995). “Solid-Phase Combinatorial Chemistry and Novel Tagging Methods for Identifying Leads,” Curr. Opin. Biotechnol. 6:632-639.
Chayer, et al. (2001). “Synthesis of Carboranylpyrroles,” Tetrahedron Lett. 42(44):7759-7761.
Chappell et al. (2011). “Ras/Raf/MEK/ERK and Pl3K/PTEN/Akt/mTOR Inhibitors: Rationale and Importance to Inhibiting These Pathways in Human Health,” Oncotarget. 2(3):136-164.
Chou, T. et al. (1984). “Quantitative Analysis of Dose-Effect Relationships: The Combined Effects of Multiple Drugs or Enzyme Inhibitors,” Adv. Enzyme Regul. 22:27-55.
Chou, T. et al. (1991). “Chemotherapeutic Synergism, Potentiation and Antagonism,” Encyclopedia of Human Biology, Academic Press, 2:371-379.
Chou, T. (1991). “The Median-Effect Principle and the Combination Index for Quantitation of Synergism and Antagonism,” in Synergism and Antagonism in Chemotherapy, Chou, T. and Rideout, D.C. (eds.), San Diego, CA: Academic Press, Chapter 2, pp. 61-102.
Clark et al. (1995). “PRO LIGAND: An Approach to De Novo Molecular Design. 1. Application to the Design of Organic Molecules,” J. Comp. Aided Molec. Design 9:13-32.
Coe et al. (1999). “Solution-Phase Combinatorial Chemistry,” Mol. Divers. 4(1):31-38.
Colliuod et al. (1993). “Oriented and Covalent Immobilization of Target Molecules to Solid Supports: Synthesis and Application of a Light-Activatable and Thiol-Reactive Cross-Linking Reagent,” Bioconjugate Chem. 4:528-536.
Colman, P.M. (1994). “Structure-Based Drug Design,” Curr. Opin. Struct. Biol. 4:868-874.
Coste et al. (1994). “Coupling N-Methylated Amino Acids Using PyBroP and PyCloP Halogenophosphonium Salts: Mechanism and Fields of Application,” J. Org. Chem. 59:2437-2446.
Crump, M. (2002). “Inhibition of Raf Kinase in the Treatment of Acute Myeloid Leukemia,” Curr. Pharm. Design 8:(25):2243-2248.
Curtin et al. (1998). “Discovery and Evaluation of a Series of 3-Acylindole Imidazopyridine Platelet-Activating Factor Antagonists,” J. Med. Chem. 41:74-95.
Cwirla et al. (1990). “Peptides on Phage: A Vast Library of Peptides for Identifying Ligands,” Biochemistry 87:6378-6382.
Das-Gupta et al. (1941). “Acridine Derivatives, Part IV,” J. Indian Chem. Soc. 18:25-28.
Dastych et al. (1994). “Stem Cell Factor Induces Mast Cell Adhesion to Fibronectin,” J. Immunol. 152:213-219.
Demetri (2001). “Targeting c-kit mutations in solid tumors: Scientific rationale and novel therapeutic options,” Seminars in Oncology 28(5)(Suppl. 17)19-26.
Dewar et al. (2005). “Inhibition of c-fms by Imatinib Expanding the Spectrum of Treatment,” Cell Cycle 4(7):851-853.
Dolle et al. (1999). “Comprehensive Survey of Combinatorial Library Synthesis: 1998,” J. Comb. Chem. 1(4):235-282.
Doyle et al. (1979). “Alkyl Nitrite-metal halide Deamination Reactions. 6. Direct Synthesis of Arenediazonium Tetrafluoroborate Salts from Aromatic Amines, tert-Butyl Nitrite, and Boron Trifluoride Etherate in Anhydrous Media,” J. Org. Chem. 44(9):1572-1574.
Dube et al. (1999). “Reductive N-Alkylation of Amides, Carbamates and Ureas,” Tetrahedron Lett. 40:2295-2298.
Dutcher et al. (1951). “Studies of the C11H8N2OS Degradation Product of Gliotoxin,” J. Am. Chem. Soc. 73:4139-4141.
Eklund et al. (2003). “Treatment of rheumatoid arthritis with imatinib mesylate: clinical improvements in three refractory cases,” Annals of Medicine 35:362-367.
Eliseev et al. (1999). “Dynamic Combinatorial Chemistry: Evolutionary Formation and Screening of Molecular Libraries,” Current Topics in Microbiology & Immunology 243:159-172.
Enjalbal et al. (2000). “Mass Spectrometry in Combinatorial Chemistry,” Mass Spectrom. Rev. 19:139-161.
Felder, E.R. (1994). “The Challenge of Preparing and Testing Combinatorial Compound Libraries in the Fast Lane, at the Front End of Drug Development,” Chimia 48:531-541.
Fischer et al. (2007). “Targeting receptor tyrosine kinase signalling in small cell lung cancer (SCLC): What have we learned so far?,” Cancer Treatment Reviews 33(4):391-406.
Franz et al. (1973). “Sulfuranes. X. A Reagent for the Facile Cleavage of Secondary Amides.” J. Am. Chem. Soc. 95(6):2017-2019.
Friesen et al. (2008). “Hydroxypropyl Methylcellulose Acetate Succinate-Based Spray-Dried Dispersions: An Overview,” Molecular Pharmaceutics 5(6):1003-1019.
Gallop et al. (1994). “Applications of Combinatorial Technologies to Drug Discovery. 1. Background and Peptide Combinatorial Libraries,” J. Med. Chem. 37:1233-1251.
Gassman et al. (1973). “Specific ortho substitution of aromatic heterocyclic amines. Conversions of 2-aminopyridines,” J. Am. Chem. Soc. 95(13):4453-4455.
Ghebre-Sellassie, I. et al. (2003). “Film, Sheet, and Laminates,” Chapter 12 in Pharmaceutical Extrusion Technology, Marcel Dekker, Inc., New York, Basel, CRC Press, pp. 225-244.
Girgis, N. et. al. (1989). “The Synthesis of 5-Azaindoles by Substitution-Rearrangement of 7-Azaindoles upon Treatment with Certain Primary Amines,” J. Heterocyclic Chem. 26:317-325.
Gordon et al. (1972). “Detection of Peroxides and Their Removal,” in The Chemist's Companion: A Handbook of Practical Data, Techniques, and References, p. 437.
Gordon et al. (1994). “Applications of Combinatorial Technologies to Drug Discovery. 2. Combinatorial Organic Synthesis, Library Screening Strategies, and Future Directions,” J. Med. Chem. 37:1385-1401.
Gravert et al. (1997). “Synthesis on Soluble Polymers: New Reactions and the Construction of Small Molecules,” Curr. Opin. Chem. Biol. 1:107-113.
Guida, W. (1994). “Software for Structure-Based Drug Design,” Curr. Opin. Struct. Biol. 4:777-781.
Hands et al. (Jul. 1996). “A Convenient Method for the Preparation of 5-, 6- and 7-Azaindoles and Their Derivatives,” Synthesis 877-882.
Hayashi et al. (1984). “Dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium-(II): An Effective Catalyst for Cross-Coupling of Secondary and Primary Alkyl Grignard and Alkylzinc Reagents with Organic Halides,” J. Am. Chem. Soc. 106:158-163.
Haydock et al. (1984). “Analogues of clofibrate and clobuzarit containing fluorine in the side chains,” Eur. J. Med. Chem. 19:205-214.
Heacock et al. (1960). “Orientation and Relative Reaction Rate Factors in Aromatic Substitution by the Benzenesulfonimido Radical,” J. Am. Chem. Soc. 82:3460-3463.
Heinrich, M.C. et al. (2002). “Inhibition of KIT Tyrosine Kinase Activity: A Novel Molecular Approach to the Treatment of KIT-Positive Malignancies,” J. Clin. Oncol. 20(6):1692-1703.
Hoffmann, R.V. (1981/1990). “m-Trifluoromethylbenzenesulfonyl Chloride,” Organic Syntheses Coll. vol. 7, p. 508; vol. 60, p. 121.
Houghten et al. (1991). “Generation and Use of Synthetic Peptide Combinatorial Libraries for Basic Research and Drug Discovery,” Nature 354:84-86.
Houghten, R. (2000). “Parallel Array and Mixture-Based Synthetic Combinatorial Chemistry: Tools for the Next Millennium,” Annu. Rev. Pharmacol. Toxicol. 40:273-282.
Houghten, R. (1993). “Peptide Libraries: Criteria and Trends,” Trends Genet. 9:235-239.
Ibrahim et al. (2007). Caplus, pp. 11300, two pages.
Jarugula et al. (1997). “Nonlinear pharmacokinetics of 5-fluorouracil in rats,” J. Pharm. Sci. 86(6):756-757.
Jones et al. (1984). “Antiestrogens. 2. Structure-Activity Studies in a Series of 3-Aroyl-2-arylbenzo[b]thiophene Derivatives Leading to [6-Hydroxy-2-(4-hydroxyphenyl)benzo(b)thien-3-yl][4-[2-(1-piperidinyl)ethoxy]-phenyl]methanone Hydrochloride (LY156758), a Remarkably Effective Estrogen Antagonist with Only Minimal Intrinsic Estrogenicity,” J. Med. Chem. 27:1057-1066.
Jones, R.J. (1997). “Biology and treatment of chronic myeloid leukemia,” Curr. Opin. Onc. 9:3-7.
Joseph-McCarthy, D. (1999). “Computational Approaches to Structure-Based Ligand Design,” Pharmacology & Therapeutics 84:179-191.
Katritzky et al. (2003). “Regiospecific C-Acylation of Pyrroles and Indoles Using N-Acylbenzotriazoles,” J. Org. Chem. 68:5720-5723.
Kim et al. (2000). “A Merger of Rational Drug Design and Combinatorial Chemistry: Development and Application of Peptide Secondary Structure Mimetics,” Comb. Chem. High Throughput Screen. 3:167-183.
Kim et al. (2002). “Preparation of 2-anilino-4-indolylpyrimidines as tyrosine kinase inhibitors,” 138:55974 (abstract).
Kirkpatrick et al. (1999). “Structure-Based Drug Design: Combinatorial Chemistry and Molecular Modeling,” Comb. Chem. High Throughput Screen. 2:211-221.
Kitamura et al. (2003). “Synthesis of Quinolines and 2H-Dihydropyrroles by Nucleophilic Substitution at the Nitrogen Atom of Oxime Derivatives,” Synthesis 15:2415-2426.
Kline et al. (1986). “Studies by 1H Nuclear Magnetic Resonance and Distance Geometry of the Solution Conformation of the x-Amylase Inhibitor Tendamistat,” J. Mol. Biol. 189:377-382.
Komoyira, S. et al. (2004). “Design, synthesis and biological activity of amidinobicyclic compounds (derivatives of DX-9065a) as a factor Xa inhibitors: SAR study of S1 and aryl binding sites,” Bioorg. Med. Chem. 12:2099.
Konno et al. (2006). “Influence of Different Polymers on the Crystallization Tendency of Molecularly Dispersed Amorphous Felodipine,” J. Pharm. Sci. 95(12):2692-2705.
Kundu et al. (1999). “Combinatorial Chemistry: Polymer Supported Synthesis of Peptide and Non-Peptide Libraries,” in Progress in Drug Research, Jucker, E. (ed.), Springer Verlag AG, 53:89-156.
Kuntz et al. (1994). “Structure-Based Molecular Design,” Acc. Chem. Res. 27:117-123.
Lam et al. (1991). “A New Type of Synthetic Peptide Library for Identifying Ligand-Binding Activity,” Nature 354:82-84.
Langham et al. (1941). “Metalation and Halogen-Metal Interconversion Reactions of Some Halogenated Phenyl Ethers,” J. Am. Chem. Soc. 63:545-549.
Lebl et al. (1995). “One-Bead-One-Structure Combinatorial Libraries,” Biopolymers 37:177-198.
Leuner, C. et al. (2000). “Improving drug solubility for oral delivery using solid dispersions,” Eur. J. Pharm. Biopharm. 50:47-60.
Lipinski et al. (1997). “Experimental and Computational Approaches to Estimate Solubility and Permeability in Drug Discovery and Development Settings,” Adv. Drug Deliv. Rev. 23:3-25.
Liu et al. (2006). “Sorafenib blocks the RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and induces tumor cell apoptosis in hepatocellular carcinoma model PLC/PRF/5,” Cancer Res. 66:11852-11858.
Ma et al. (2000). “Indolinone Derivatives Inhibit Constitutively Activated KIT Mutants and Kill Neoplastic Mast Cells,” J. Invest. Dermatol. 114:392-394.
Ma et al. (2002). “The c-kit mutation causing human mastocytosis is resistant to ST1571 and other KIT kinase inhibitors; kinases with enzymatic site mutations show different inhibitor sensitivity profiles than wild-type kinase and those with regulatory-type mutations,” Blood 99:1741-1744.
Madden, et al. (1994). “Synthetic Combinatorial Libraries: Views on Techniques and Their Application,” Perspectives in Drug Discovery and Design 2:269-285.
Madhusudan et al. (2004). “Tyrosine kinase inhibitors in cancer therapy,” Clin. Biochem. 37(7):618-635.
Markiewicz et al. (2000). “Synthetic Oligonucleotide Combinatorial Libraries and Their Applications,” II Farmaco 55:174-177.
Martin, Y. (1991). “Computer-Assisted Rational Drug Design,” Methods Enzymol. 203:587-613.
Matsumoto et al. (1999). “Physical properties of solid molecular dispersions of indomethacin with poly(vinyl)pyrrolidinone and poly(vinylpyrrolidinone-co-vinyl-acetate) in relation to indomethacin crystallization,” Pharm. Res. 16(11):1722-1728.
Mazeas et al. (1999). “Synthesis of New Melatoninergic Ligands Including Azaindole Moiety,” Heterocycles 50(2):1065-1080.
McPherson, A. (1990). “Current Approaches to Macromolecular Crystallization,” Eur. J. Biochem. 189:1-23.
Merour et al. (2001). “Synthesis and Reactivity of 7-Azaindoles (1H-Pyrrolo[2,3-b]pyridine),” Current Organic Chemistry 5:471-506.
Merritt, A.T. (1998). “Solution Phase Combinatorial Chemistry,” Comb. Chem. High Throughput Screen. 1(2):57-72.
Meula Pomeda et al. (2002). “Efecto de codisolventes y dispersiones solidas de polivinilpirrolidona K-30 en la solubilidad del tiabendazol,” 86 VI Congreso SEFIG y 3as Jornadas TF, pp. 85-87 (with English translation of Introduction).
Miller et al. (1994). “FLOG: A System to Select ‘Quasi-Flexible’ Ligands Complementary to a Receptor of Known Three-Dimensional Structure,” J. Comp.-Aided Mol. Des. 8:153-174.
Minakata et al. (1992). “Functionalization of 1H-Pyrrolo[2,3-b]pyridine.” Bull. Chem. Soc. Japan 65(11):2992-2997.
Minakata et al. (Jul. 1992). “Regioselective Functionalization of 1H-Pyrrolo[2,3-b]pyridine via its N-Oxide,” Synthesis pp. 661-663.
Miyaura et al. (1995). “Palladium-Catalyzed Cross-Coupling Reactions of Organoboron Compounds,” Chem. Rev. 95:2457-2483.
Mokhtari et al. (2010). “Potential utility of small tyrosine kinase inhibitors in the treatment of diabetes,” Clin. Sci. 118(4):241-247.
Mol et al. (2004). “Structural Basis for the Autoinhibition and STI-571 Inhibition of c-Kit Tyrosine Kinase,” J. Biol. Chem. 279:31655-31663.
Nagafuji et al. (1996). “A General Synthesis of Pyrroles and Fused Pyrrole Systems from Ketones and Amino Acids,” J. Org. Chem. 61:4999-5003.
Nahm et al. (1981). “N-Methoxy-N-Methylamides as Effective Acylating Agents,” Tetrahedron Lett. 22(39):3815-3818.
Nakai et al. (1988). “New Potent Antagonists of Leuokotrienes C4 and D4. 1. Synthesis and Structure-Activity Relationships,” J. Med. Chem. 31:84-91.
Neidle et al. (1991). “Molecular Modeling to Study DNA Intercalation by Anti-Tumor Drugs,” Methods Enzymol. 203:433-458.
Ng et al. (1995). “Engineering Protein-Lipid Interactions: Targeting of Histidine-Tagged Proteins to Metal-Chelating Lipid Monolayers,” Langmuir 11:4048-4055.
Olah et al. (1984). “Synthetic Methods and Reactions: Part 209. Improved Preparation of Aldehydes and Ketones from N,N-Dimethylamides and Grignard Reagents,” Synthesis pp. 228-230.
Ottoni et al. (1998). “Efficient and Simple Methods for the Introduction of the Sulfonyl, Acyl and Alkyl Protecting Groups on the Nitrogen of Indole and its Derivatives,” Tetrahedron 54:13915-13928.
Owicki et al. (1997). “Application of Fluorescence Polarization Assays in High-Throughput Screening,” Genetic Engineering News 17:28.
Panitumumab PiCCA Study (Panitumumab in Combination with Cisplatin/Gemcitabine) (2012). National Cancer Institute at the National Institutes of Health, located at http://clinicaltrials.gov/ct2/show/NCT01320254, last visited on Dec. 16, 2013, 5 pages.
Parker et al. (2000). “Development of High Throughput Screening Assays Using Fluorescence Polarization: Nuclear Receptor-Ligand-Binding and Kinase/Phosphatase Assays,” J. Biomol. Screen. 5:77-88.
Patani et al. (1996). “Bioisosterism: A rational approach in drug design,” Chem. Rev. 96:3147-3176.
Perrin, D.M. (2000). “Nucleic Acids for Recognition and Catalysis: Landmarks, Limitations, and Looking to the Future,” Comb. Chem. High Throughput Screen. 3:243-269.
Pflugrath et al. (1986). “Crystal Structure Determination, Refinement and the Molecular Model of the α-Amylase Inhibitor Hoe-467A,” J. Mol. Biol. 189:383-386.
Pierce et al. (1942). “Local anaesthetics. I. Beta-Monoalkylaminoethyl Esters of Alkoxybenzoic Acids,” J. Am. Chem. Soc. 64:1691-1694.
Plunkett et al. (1995). “A Silicon-Based Linker for Traceless Solid-Phase Synthesis,” J. Org. Chem. 60:6006-6007.
Remington. (1995). “Preformulation,” Chapter 83 in Remington: The Science and Practice of Pharmacy, Remington's Pharmaceutical Sciences, 19th ed., Mack Publishing Co., Easton, PA, vol. 2, pp. 1457-1462.
Robison et al. (1955). “7-Azaindole. I. Synthesis and Conversion to 7-Azatryptophan and Other Derivatives,” J. Am. Chem. Soc. 77:457-460.
Rodan, G. et al. (2000). “Therapeutic Approaches to Bone Diseases,” Science 289:1508-1514.
Saify et al. (1994). “Synthesis of some 7-azaindole derivatives: their cytotoxicity and antibacterial activity,” Pakistan Journal of Scientific and Industrial Research 37(10):439-441.
Saiki, R. (1990). “Amplification of Genomic DNA,” in PCR Protocols: A Guide to Methods and Applications, Academic Press, Inc.: San Diego, CA 2:13-20.
Sathornsumetee et al. (2006). “AAL881, a novel small molecule inhibitor of RAF and vascular endothelial growth factor receptor activities, blocks the growth of malignant glioma,” Cancer Res. 66:8722-8730.
Sawada et al. (2001). “4-(Benzoylindolizinyl)butyric acids: Novel nonsteroidal inhibitors of steroid 5α-reductase. III,” Chem. Pharm. Bull. 49(7):799-813.
Schiemann et al. (1943). “p-Fluorobenzoic Acid,” Org. Synth. Coll. vol. 2, 299, 4 total pages.
Schneller et al. (1980). “Synthesis of 4-Amino-1H-Pyrrolo[2,3-b]pyridine (1,7-Dideazaadenine) and 1H-Pyrrolo[2,3-b]pyridin-4-ol (1,7-Dideazahypoxanthine),” J. Org. Chem. 45:4045-4048.
Schweizer et al. (1999). “Combinatorial Synthesis of Carbohydrates,” Curr. Opin. Chem. Biol. 3(3):291-298.
Shah et al. (2006). “Analytical Techniques for Quantification of Amorphous/Crystalline Phases in Pharmaceutical Solids,” J. Pharm. Sci. 95(8):1641-1665.
Shan et al. (1997). “Prodrug strategies based on intramolecular cyclization reactions,” J. Pharm. Sci. 86(7):765-767.
Song et al. (2002). “Isomerism of Bis(7-azaindolyl)methane.” Org. Lett. 4:23:4049-4052 and Supporting information pp. 1-15.
Sorafenib Tosylate. (2012). National Cancer Institute at the National Institutes of Health, located at <http://www.cancer.gov/cancertopics/druginfo/sorafenibtosylate>, last visited on Dec. 16, 2013, two pages.
Su et al. (1960). “Synthesis of Bromo-substituted Indoxyl Esters for Cytochemical Demonstration of Enzyme Activity,” J. Am. Chem. Soc. 82:1187-1189.
Sun, C. (1999). “Recent Advances in Liquid-Phase Combinatorial Chemistry,” Comb. Chem. High Throughput Screen. 2:299-318.
Sun, C. (1999). “Design, Synthesis and Evalutions of Substituted 3-[(3- or 4-Carboxyethylpyrrol-2-yl)methylidenyl]indolin-2-ones as Inhibitors of VEGF, PGF, and PDGF Receptor Tyrosine Kinases,” J. Med. Chem. 42:5120-5130.
Tanno et al. (2004). “Evaluation of Hypromellose Acetate Succinate (HPMCAS) as a Carrier in Solid Dispersions,” Drug Dev. Ind. Pharm. 30(1):9-17.
Thibault et al. (2003). “Concise and Efficient Synthesis of 4-Fluoro-1H-pyrrolo[2,3-b]pyridine,” Org. Lett. 5:5023-5025.
Thomas et al. (2001). “Light-Emitting Carbazole Derivatives: Potential Electroluminescent Materials,” J. Am. Chem. Soc. 123:9404-9411.
Toste et al. (1995). “A Versatile Procedure for the Preparation of Aryl Thiocyanates Using N-Thiocyanatosuccinimide (NTS),” Synth. Commun. 25(8):1277-1286.
Uritskaya et al. (1973). Azaindole derivatives. XLIII. Synthesis of 1-acety1-4-methy1-7-azatryptamine, STN Accession No. 1974:27133; Document No. 80:27133; Abstract of Khimiya Geterotsiklichenskikh Soedinenii 10:1370-1373.
Vachon et al. (1997). “The influence of microencapsulation using Eudragit RS100 on the hydrolysis kinetics of acetylsalicylic acid,” J. Microencapsulation 14:281-301.
Vandelli et al. (1993). “Analysis of release data in the evaluation of the physical state of progesterone in matrix systems,” J. Microencapsulation 10(1):55-65.
Weber, P. (1991). “Physical Principles of Protein Crystallization,” Adv. Protein Chem. 41:1-36.
Wells et al. (2009). “Targeting the RET Pathway in Thyroid Cancer,” Clin. Cancer Res. 15(23):7119-7123.
Wendt et al. (2004). “Identification of novel binding interactions in the development of potent, selective 2-naphthamidine inhibitors of urokinase. Synthesis, structural analysis, and SAR of N-phenyl amide 6-substitution,” J. Med. Chem. 47(2):303-324.
Wessjohann, L. (2000). “Synthesis of Natural-Product-Based Compound Libraries,” Curr. Opin. Chem. Biol. 4:303-309.
Yakhontov et al. (1965). “Derivatives of 7-azaindole. XV. Electrophilic substitution of 4-methyl-7-azaindole and its derivatives.” Zhurnal Obshchei Khimii 1(11):2032-2040. (English abstract only).
Yang, et al. (1992). “Synthesis of Some 5-Substituted Indoles,” Heterocycles 34:1169-1175.
Yeung et at. (2002). “Friedel-Crafts acylation of indoles in acidic imidazolium chloroaluminate ionic liquid at room temperature,” Tetrahedron Lett. 43(33):5793-5795.
Zanon et al. (2003). “Copper-Catalyzed Domino Halide Exchange-Cyanation of Aryl Bromides,” J. Am. Chem. Soc. 125:2890-2891.
Zhang et al. (2002). “An effective procedure for the acylation of azaindoles at C-3,” J. Org. Chem. 67(17):6226-6227 and p. S1-S30.
Related Publications (1)
Number Date Country
20130005761 A1 Jan 2013 US
Divisions (1)
Number Date Country
Parent 12843908 Jul 2010 US
Child 13613017 US