Quantcast
Channel: New Drug Approvals
Viewing all 2025 articles
Browse latest View live

Tadalafil (cialis)

$
0
0

Tadalafil

GF-196960, IC-351, Cialis

6R-trans)-6-(1,3-benzodioxol-5-yl)- 2,3,6,7,12,12a-hexahydro-2-methyl-pyrazino [1', 2':1,6] pyrido[3,4-b]indole-1,4-dione

Pyrazino[1',2':1,6]pyrido[3,4-b]indole-1,4-dione,6-(1,3-benzodioxol-5-yl)-2,3,6,7,12,12a-hexahydro-2-methyl-, (6R-trans)-; (6R,12aR)-6-(1,3-benzodioxol-5-yl)-2,3,6,7,12,12a-hexahydro-2-ethylpyrazino[1',2':1,6]pyrido[3,4-b]indole-1,4-dione; GF 196960;  Adcirca;

171596-29-5  casno

Molecular Weight:
389.40

Molecular Formula:C22H19N3O4

GlaxoSmithKline (Originator), Lilly Icos (Marketer), Lilly (Licensee), Lilly Icos (Licensee)

Launched-2003

Tadalafil is currently marketed as Cialis. Cialis was developed by Eli Lilly as a treatment for impotence. In this capacity, it is reported that tadalafil functions by inhibiting the formation of cyclic guanosine monophosphate (cGMP)-specific phosphodiesterase type 5 (PDE5). The inhibition of PDE5 presumably lessens impotence by increasing the amount ot c(iMP, resulting in smooth muscle relaxation and increased blood flow.

Tadalafil is a PDE5 inhibitor marketed in pill form for treating erectile dysfunction (ED) under the name Cialis, and under the name Adcirca for the treatment of pulmonary arterial hypertension. In October 2011 the U.S. Food and Drug Administration (FDA) approved Cialis for treating the signs and symptoms of benign prostatic hyperplasia (BPH) as well as a combination of BPH and erectile dysfunction (ED) when the conditions coincide. It initially was developed by the biotechnology company ICOS, and then again developed and marketed world-wide by Lilly ICOS, LLC, the joint venture of ICOS Corporation and Eli Lilly and CompanyCialis tablets, in 2.5 mg, 5 mg, 10 mg, and 20 mg doses, are yellow, film-coated, and almond-shaped. The approved dose for pulmonary arterial hypertension is 40 mg (two 20-mg tablets) once daily.

Tadalafil can be prepared via a series of intermediates. One synthesis scheme is illustrated in Scheme 1: Scheme 1

Figure imgf000003_0001

U.S. Patent No. 5,859,006 describes the synthesis of the tadalafil intermediate (Compound III) from D-tryptophan methyl ester (Compound II) and piperonal (Compound I) using trifluoroacetic acid and dichloromethane, a halogenated solvent. Compound III is then reacted with chloroacetyl chloride (Compound IV) and chloroform, providing another intermediate of tadalafil (Compound V). WO 04/011463 describes a process of preparing tadalafil intermediates from D-tryptophan methyl ester HCl salt and piperonal by refluxing the reagents in isopropyl alcohol; the obtained intermediate is reacted with chloroacetyl chloride and THF, resulting in another intermediate of tadalafil.

Tadalafil is also manufactured and sold under the name of Tadacip by the Indian pharmaceutical company Cipla in doses of 10 mg and 20 mg.

On November 21, 2003 the FDA approved tadalafil (as Cialis) for sale in the United States as the third ED prescription drug pill (after sildenafil citrate(Viagra) and vardenafil (Levitra)). Like sildenafil and vardenafil, tadalafil is recommended as an ‘as needed’ medication. Cialis is the only one of the three that is also offered as a once-daily medication.

Moreover, tadalafil was approved in May 2009 in the United States for the treatment of pulmonary arterial hypertension and is under regulatory review in other regions for this condition. In late November 2008, Eli Lilly sold the exclusive rights to commercialize tadalafil for pulmonary arterial hypertension in the United States to United Therapeutics for an upfront payment of $150 million.

The FDA’s approval of Viagra (Sildenafil) on March 27, 1998 was a ground-breaking commercial event for the treatment of ED, with sales exceedingUS$1 billion. Subsequently, the FDA approved Levitra (vardenafil) on August 19, 2003, and Cialis (tadalafil) on November 21, 2003.

Cialis was discovered by Glaxo Wellcome (now GlaxoSmithKline) under a partnership between Glaxo and ICOS to develop new drugs that began in August 1991. [1][2] In 1993, the Bothell, Washington biotechnology company ICOS Corporation began studying compound IC351, a phosphodiesterase type 5 (PDE5) enzyme inhibitor. In 1994, Pfizer scientists discovered that sildenafil, which also inhibits the PDE5 enzyme, caused penile erection in men participating in a clinical study of a heart medicine. Although ICOS scientists were not testing compound IC351 for treating ED, they recognized its potential usefulness for treating that disorder. Soon, in 1994, ICOS received a patent for compound IC351 (structurally unlike sildenafil and vardenafil), and Phase 1 clinical trials began in 1995. In 1997, the Phase 2 clinical studies were initiated for men experiencing ED, then progressed to the Phase 3 trials that supported the drug’s FDA approval. Although Glaxo had an agreement with ICOS to share profits 50/50 for drugs resulting from the partnership, Glaxo let the agreement lapse in 1996 as the drugs developed were not in the company’s core markets.[3]

In 1998, ICOS Corporation and Eli Lilly and Company formed the Lilly ICOS, LLC, joint venture company to further develop and commercialize tadalafil as a treatment for ED. Two years later, Lilly ICOS, LLC, filed a new drug application with the FDA for compound IC351 (under the tadalafil generic name, and the Cialis brand name). In May 2002, Lilly ICOS reported to the American Urological Association that clinical trial testing demonstrated that tadalafil was effective for up to 36 hours, and one year later, the FDA approved tadalafil. One advantage Cialis has over Viagra and Levitra is its 17.5-hour half-life (thus Cialis is advertised to work for up to 36 hours, after which time there remains approximately 25 percent of the absorbed dose in the body) when compared to the four-hour half–life of sildenafil (Viagra).

In 2007, Eli Lilly and Company bought the ICOS Corporation for $2.3 billion. As a result, Eli Lilly owned Cialis and then closed the ICOS operations, ending the joint venture and firing most of ICOS’s approximately 500 employees, except for 127 employees of the ICOS biologics facility, which subsequently was bought by CMC Biopharmaceuticals A/S (CMC).

Tadalafil Molecule

Persons surnamed “Cialis” objected to Eli Lilly and Company’s so naming the drug, but the company has maintained that the drug’s trade name is unrelated to the surname.[4]

On October 6, 2011, the U.S. FDA approved tadalafil [5] to treat the signs and symptoms of benign prostatic hyperplasia (BPH). BPH is a condition in males in which the prostate gland becomes enlarged, obstructing the free flow of urine. Symptoms may include sudden urges to urinate (urgency), difficulty in starting urination (hesitancy), a weak urine stream, and more frequent urination- especially at night. The FDA has also approved tadalafil for treatment of both BPH and erectile dysfunction (ED) where the two conditions co-exist.

 

Although available since 2003 in 5, 10, 20 mg dosage, in late 2008/early 2009, the U.S. FDA approved the commercial sale of Cialis in 2.5 mg dosage as a one-a-day treatment for ED. The 2.5 mg dose avoids earlier dispensing restrictions on higher dosages. The price of the 5 mg and 2.5 mg are often similar, so some people score and split the pill.[6] The manufacturer does not recommend splitting.

Moreover, tadalafil (Adcirca) 40 mg was approved in 2009 in the United States and Europe (and 2010 in Canada and Japan) as a once-daily therapy to improve exercise ability in patients withpulmonary arterial hypertension. In patients with pulmonary arterial hypertension, the pulmonary vascular lumen is decreased as a result of vasoconstriction and vascular remodeling, resulting in increased pulmonary artery pressure and pulmonary vascular resistance. Tadalafil is believed to increase pulmonary artery vasodilation, and inhibit vascular remodeling, thus lowering pulmonary arterial pressure and pulmonary vascular resistance. Right heart failure is the principal consequence of pulmonary arterial hypertension.

On October 6, 2011, the U.S. FDA approved tadalafil [6] to treat the signs and symptoms of benign prostatic hyperplasia (BPH). BPH is a condition in males in which the prostate gland becomes enlarged, obstructing the free flow of urine. Symptoms may include sudden urges to urinate (urgency), difficulty in starting urination (hesitancy), a weak urine stream, and more frequent urination- especially at night. The FDA has also approved tadalafil for treatment of both BPH and erectile dysfunction (ED) where the two conditions co-exist.

 

Tadalafil has been used in approximately 15,000 men participating in clinical trials, and over eight million men worldwide (primarily in the post-approval/post-marketing setting). The most commonside effects when using tadalafil are headache, stomach discomfort or pain, indigestion, burping, acid reflux. back pain, muscle aches, flushing, and stuffy or runny nose. These side effects reflect the ability of PDE5 inhibition to cause vasodilation (cause blood vessels to widen), and usually go away after a few hours. Back pain and muscle aches can occur 12 to 24 hours after taking the drug, and the symptom usually disappears after 48 hours.
In May 2005, the U.S. Food and Drug Administration found that tadalafil (along with other PDE5 inhibitors) was associated with vision impairment related to NAION (nonarteritic anterior ischemic optic neuropathy) in certain patients taking these drugs in the post-marketing (outside of clinical trials) setting. Most, but not all, of these patients had underlying anatomic or vascular risk factors for development of NAION unrelated to PDE5 use, including: low cup to disc ratio (“crowded disc”), age over 50, diabetes, hypertension, coronary artery disease, hyperlipidemia and smoking. Given the small number of NAION events with PDE5 use (fewer than one in one million), the large number of users of PDE5 inhibitors (millions) and the fact that this event occurs in a similar population to those who do not take these medicines, the FDA concluded that they were not able to draw a cause and effect relationship, given these patients underlying vascular risk factors or anatomical defects. However, the label of all three PDE5 inhibitors was changed to alert clinicians to a possible association.

In October 2007, the FDA announced that the labeling for all PDE5 inhibitors, including tadalafil, requires a more prominent warning of the potential risk of sudden hearing loss as the result of postmarketing reports of deafness associated with use of PDE5 inhibitors.[7]

 

Selectivity compared with other PDE5 inhibitors

Tadalafil, sildenafil, and vardenafil all act by inhibiting the PDE5 enzyme. These drugs also inhibit other PDE enzymes. Sildenafil and vardenafil inhibit PDE6, an enzyme found in the eye, more than tadalafil.[9] Some sildenafil users see a bluish tinge and have a heightened sensitivity to light because of PDE6 inhibition.[3] Sildenafil and vardenafil also inhibit PDE1 more than tadalafil.[9]PDE1 is found in the brain, heart, and vascular smooth muscle.[9] It is thought that the inhibition of PDE1 by sildenafil and vardenafil leads to vasodilationflushing, and tachycardia.[9] Tadalafil inhibits PDE11 more than sildenafil or vardenafil.[9] PDE11 is expressed in skeletal muscle, the prostate, the liver, the kidney, the pituitary gland, and the testes.[9] The effects on the body of inhibiting PDE11 are not known.[9]

 

20 mg Cialis tablet

In the United States, the FDA relaxed rules on prescription drug marketing in 1997, allowing advertisements targeted directly to consumers.[10] Lilly-ICOS hired the Grey Worldwide Agency in New York, part of the Grey Global Group, to run the Cialis advertising campaign.[11] Marketers for Cialis has taken advantage of its greater duration compared to its competitors in advertisements for the drug; Stuart Elliot of The New York Times opined: “The continuous presence of women in Cialis ads is a subtle signal that the drug makes it easier for them to set the pace with their men, in contrast to the primarily male-driven imagery for Levitra and Viagra.”[11] Iconic themes in Cialis ads include couples in bathtubs and the slogan “When the moment is right, will you be ready?”[11] Cialis ads were unique among the ED drugs in mentioning specifics of the drug.[12] As a result, Cialis ads were also the first to describe the side effects in an advertisement, as the FDA requires advertisements with specifics to mention side effects. One of the first Cialis ads aired at the 2004 Super Bowl.[12] Just weeks before the Super Bowl, the FDA required more possible side effects to be listed in the advertisement, including priapism.[12] Although many parents objected to the Cialis ad being aired during the Super Bowl, Janet Jackson‘s halftime “wardrobe malfunction” overshadowed Cialis.[12] In January 2006, the Cialis ads were tweaked, adding a doctor on screen to describe side effects and only running ads where more than 90 percent of the audience are adults, effectively ending Super Bowl ads.[10] In 2004, Lilly-ICOS, Pfizer, and GlaxoSmithKline spent a combined $373.1 million to advertise Cialis, Viagra, and Levitra respectively.[12] Cialis has sponsored many golf events, including the America’s Cup and the PGA Tour, once being title sponsor of the PGA Tour Western Open tournament.[13]

CIALIS (tadalafil) is a selective inhibitor of cyclic guanosine monophosphate (cGMP)-specific phosphodiesterase type 5 (PDE5). Tadalafil has the empirical formula C22H19N3O4 representing a molecular weight of 389.41. The structural formula is:

CIALIS (tadalafil) Structural Formula Illustration

The chemical designation is pyrazino[1',2':1,6]pyrido[3,4-b]indole-1,4-dione, 6-(1,3-benzodioxol-5-yl)2,3,6,7,12,12a-hexahydro-2-methyl-, (6R,12aR)-. It is a crystalline solid that is practically insoluble in water and very slightly soluble in ethanol.

CIALIS is available as almond-shaped tablets for oral administration. Each tablet contains 2.5, 5, 10, or 20 mg of tadalafil and the following inactive ingredients: croscarmellose sodium, hydroxypropyl cellulose, hypromellose, iron oxide, lactose monohydrate, magnesium stearate, microcrystalline cellulose, sodium lauryl sulfate, talc, titanium dioxide, and triacetin.

Tadalafil, (6R-trans)-6-(l,3-benzodioxol-5-yl)-2,3,6,7,12,12a-hexahydro-2- methyl-pyrazino[r,2':l,6]pyrido[3,4-b]indole-l,4-dione, with the structural formula shown below, is a white crystalline powder. (CAS# 171596-29-5). Tadalafil is a potent and selective inhibitor of the cyclic guanosine monophosphate (cGMP) – specific phosphodiesterase enzyme, PDE5. The inhibition of PDE5 increases the amount of cGMP, resulting in smooth muscle relaxation and increased blood flow. Tadalafil is therefore currently used in the treatment of male erectile dysfunction, and is commercially available as CIALIS ®.

Figure imgf000002_0001

Tadalafil U.S. Patent No. 5,859,006 describes the synthesis of tadalafil via the cyclization of TDCL (i.e., cis-methyl l,2,3,4-tetrahydro-2-chloroacetyl-l-(3,4- methylenedioxyphenyl)-9H-pyrido[3,4-b]mdole-3-carboxylate) using methylamine by purification by flash chromatography, followed by subsequent crystallization from methanol. Crude tadalafil typically requires additional purification steps, such as multiple extractions, crystallization, and/or flash chromatography, to remove the impurities present in the compound after synthesis is complete. Such purification processes increase the cost of producing tadalafil. Also, when repeating the US ’006 process, about 250 volumes of methanol were necessary for the crystallization step

Tadalafil can be prepared via a series of intermediates. One synthesis for preparing tadalafil is illustrated below in Scheme I:

SCHEME I

Figure imgf000003_0001

U.S. Patent No. 5,859,006 discloses the synthesis of a tadalafil intermediate

(Compound III) from D-tryptophan methyl ester (Compound II) and piperonal (Compound

I) using trifluoroacetic acid and dichloromethane, a halogenated solvent. Compound III is then reacted with chloroacetyl chloride (Compound IV) and chloroform to provide another intermediate of tadalafil (Compound V).

WO 2004/011463 discloses a process of preparing tadalafil intermediates from D-tryptophan methyl ester HCl salt and piperonal by refluxing the reagents in isopropyl alcohol, reacting the intermediate thus obtained with chloroacetyl chloride and tetrahydrofuran (THF) to provide another intermediate of tadalafil.

WO 2006/110893 discloses a process for the preparation of methyl ester intermediate (Compound III), and tadalafil using the methyl ester intermediate (CompoundII).

U.S. Patent Application Publication No. 2006/0258865 Al discloses a synthesis of the tadalafil intermediate (Compound III) from D-tryptophan methyl ester

(Compound II) and piperonal (Compound I) using a dehydrating agent selected from Na2SO4, K2SO4, MgSO4, CaSO4, CaCl2, molecular sieve or mixtures thereof and a high boiling solvent such as N,N-Dimethyl acetamide. Compound III is then reacted with chloroacetyl chloride (Compound IV) in the presence of a base such as NaHCO3 and an organic solvent such as dichloromethane, providing another intermediate of tadalafil (Compound V), which is further reacted with aqueous methyl amine solution to provide tadalafil.

………………………………………….

EP2004644A1

WO2007110734A1

Scheme II and III.

Figure imgf000011_0001
Figure imgf000011_0002
Figure imgf000011_0003

……………………………………………………………………………………………………a compound of .Formula I

SCHEME III

Figure imgf000011_0004
 
 

SCHEME IV

Figure imgf000013_0001
Compound – 1                                                 Compound – II

 

EXAMPLE l

The reaction scheme of this example is generally shown below in SchemeIV.

SCHEME IV

Figure imgf000013_0001

Compound – 1                                           Compound – II

Into a clean dry glass flask charged with ethanol (250 ml) under a nitrogen atmosphere was added Compound 1 (25 g) under stirring. The reaction mass was cooled to 0 to 50C and monomethylamine gas was purged into the reaction mixture for about 2 hours while maintaining the temperature between 0 to 50C. The temperature was raised to 75 to 8O 0C and the reaction mixture was stirred under reflux for 2 hours. The reaction mixture was then cooled to 0 to 5°C and monomethylamine gas was again purged into the reaction mixture at 0 to 5°C. The temperature was again raised to 75 to 800C and stirred for about 1 hour. The reaction mixture was concentrated under vacuum to about 1/3 its original volume, cooled to 5 to 1O0C and stirred for 1 hour at this temperature. The solids were filtered and washed with chilled ethanol (50 ml). The wet solids were dried under vacuum for 6 hours.

Yield: 25g; Mp: 202-206.70C

Specific rotation (25°C) :+44.0 ( C=l% in DMSO)

13C NMR, DMSO-D6 : 25.78, 25.92, 57.89, 57.98, 101.17, 108.09, 108.32,

109.08, 111.48, 117.82, 118.62, 122.23, 122.97, 126.97, 135.97, 136.22, 136.55, 146.99,

147.48, 173.13

1H NMR, DMSO-D6, 300 MHz, Delta values: 2.6(m,lH), 2.7(m,3H),

2.8(d,lH), 3.0(d,lH), 3.6(bs,lH), 5.1(m,lH), 6.0(s,3H), 6.9-7.1(m, 5H), 7.2(d,lH),

7.4(d,lH), 7.8(bs, IH), 10.3(s, IH)

EXAMPLE 2

The reaction scheme of this example is generally shown below in SchemeV.

SCHEME V

Figure imgf000014_0001

Formula III                                                                                     Formula II

Into a clean dry flask charged with dichloromethane (200 ml) was added

Compound II (25 g) obtained in Example 1 under stirring at 25 to 300C. Next, triethylamine (16.11 g) was added to the reaction mixture and stirred for 30 minutes at 20 to 300C. The reaction mixture was cooled to 0 to 5°C and a solution of chloroacetyl chloride (12.93 g) in chloroform (50 ml) was added to the reaction mixture while maintaining temperature between -5 to 50C. The reaction mixture was stirred at -5 to 5°C for about 2 hours. Saturated aqueous sodium bicarbonate solution (50 ml) was added to the reaction mass slowly and the temperature of the reaction mixture was raised to 25 to 300C. The lower organic layer was separated and washed twice with water (75 ml). The chloroform extract was dried over anhydrous sodium sulfate. The organic layer was concentrated under vacuum until a thick yellow slurry was obtained. The slurry was cooled to 0 to5°C. The solids obtained were filtered and washed with 50 ml chilled chloroform. The wet product was dried at 750C under vacuum for 6 hours.

Yield: 22.5 g; HPLC Purity: 97%; Mp: 180-1820C

Specific rotation(25°C): -154.3(C=1% in DMSO)

13C. NMR(DMSO-Do, 300 MHZ)= 21.11, 25.88, 44.207, 51.60, 53.95,

101.16,107.66 109.56, 111.38, 118.36, 118.75, 121.58,122.74, 126.30, 130.31, 134.13,

136.57, 146.66, 147.03,167.43, 168.45

1H. NMR (CDC13, 300 MHZ):2.4(bs,3H), 3.1(m,lH), 3.8(m,lH),

4.3(bs,2H), 4.9(m,lH), 5.4(m,lH), 5.9(s,2H), 6.6-6.8(m,3H), 6.9(bs,lH), 7.1-7.3(m,3H),

7.6(d, IH), 7.7(bs,lH)

1H. NMR (DMSO-D6, 300 MHZ): 2.0 (bs,3H), 2.9(m,lH), 3.4(m,lH),

4.5(m,lH), 4.8(m,lH), 4.9(m,lH), 6.0(m,2H), 6.4-6.8(m,4H), 6.9-7.2(m,2H), 7.3(d, IH),

7.4(bs,lH), 7.5(d,lH), 10.8(s,lH)

EXAMPLE 3

The reaction scheme of this example is generally shown below in SchemeVI.

SCHEME VI

Figure imgf000015_0001

Formula II                                                                         Formula I

Into a clean dry round bottom (RB) flask was charged tetrahydrofuran

(THF) (175 ml) under a nitrogen blanket and then cooled to -35 to -400C. Next 92 ml n- butyllithium (1.6 m solution in hexane) was added while maintaining the temperature between -35 to -400C. After the addition was complete, the reaction mixture was stirred at -35 to -400C for 15 minutes. A solution of compound of formula II (22.5 g) obtained in Example 2 in THF (75 ml) was prepared and slowly added to the reaction mixture while maintaining the temperature between -35 to -400C. After the addition was complete, the reaction mixture was stirred at -35 to -400C for 2.5 hours. Saturated aqueous ammonium chloride solution (25 ml) and 50 ml ethyl acetate was added to the reaction mixture at -35 to -400C. The temperature was raised to 25 to 300C and the two layers formed were separated. The upper organic layer was collected. The lower aqueous layer was thrice extracted with ethyl acetate (25 ml). The organic layers were combined together and washed with water (50 ml). The organic extract was dried over anhydrous sodium sulfate and concentrated under vacuum to obtain crude tadalafil as a dark brown solid. [0058] Yield: 22 g; HPLC Purity: 50%.

EXAMPLE 4

Purification of crude tadalafil

The crude tadalafil (22 g) obtained in Example 3 was suspended in 110 ml methanol and stirred for 1 hour at 25 to 300C. The solids obtained were filtered and washed with 25 ml chilled methanol. The wet product was dried at 600C under vacuum for 6 hours. This was further purified by using isopropyl alcohol. Yield: 9 g; HPLC Purity: >99.5%.

EXAMPLE 5

The reaction scheme of this example is generally shown below in SchemeVII.

Scheme VII

Figure imgf000016_0001

Formula VI where R = -OCH3 Formula VIA [0062] Into a clean dry RB flask charged with methanol (1900 ml) was added D- tryptophan methyl ester (190 g) under stirring at 25 to 300C. The reaction mixture was cooled to 0 to 50C. Monomethylamine gas was purged into the reaction mixture at 0 to 5°C for about 5-7 hours under stirring. The temperature of the reaction mixture was slowly raised to about 25 to 3O0C and stirred at this temperature for 5-7 hours. The reaction mixture was concentrated under vacuum to distill out the solvent. Diisopropyl ether (950 ml) was added and cooled to 25 to 3O0C under stirring for 1-2 hrs. The solids obtained were filtered, washed with Diisopropyl ether and dried under vacuum. [0063] MP: 122.4-1240C; Yield: 150 g (78.9 % w/w).

Specific rotation(25°C): +12.5 (C=I % in DMSO)

13 C NMR (300 MHZ,DMSO-D6): 25.71, 31.40, 55.67, 110.93, 111.55,

118.42, 118.73, 121.09, 123.95, 127.66,136.44, 175.39.

1H NMR (300 MHZ,DMSO-D6): 1.6(bs,2H), 2.5(m,3H), 2.8(m,lH),

3.1(m,lH), 3.4(m, IH), 6.9-7.2(m,3H), 7.3(d,lH), 7.5(d,lH), 7.8(bs,lH), 10.8(bs,lH)

EXAMPLE 6

The reaction scheme of this example is generally shown below in Scheme VIII.

SCHEME VIII

Figure imgf000017_0001

Formula VIA                                Formula VII

Figure imgf000017_0002

Into a clean, dry flask charged with methylene dichloride (MDC) (1000 ml) was added D-tryptophan methyl amide, the compound of Formula VIA (50 g), and piperonal, the compound of Formula VII (31.09 g), under stirring at 25 to 300C. The reaction mixture was cooled to 0 to 5°Cunder nitrogen atmosphere. Trifluoroacetic acid (85.3 g) was dissolved in MDC (250 ml) and the solution was slowly added to the reaction mixture at 0 to 5°C. The temperature of the reaction mixture was raised to 20 to 300C and stirred at this temperature for 14-16 hours. The reaction was monitored by TLC, workup was done as follows, the pH of the reaction mixture was adjusted to 8-9 using sodium carbonate solution under stirring, the two layers were settled, separated and the lower MDC layer was washed with water. The MDC layer was then dried over anhydrous sodium sulfate. The reaction mass was concentrated under vacuum at 40 to 5O0C to remove the solvent. The compound was precipitated using ethyl acetate, the solids were filtered, washed with ethyl acetate and dried.

Yield: 52.5 g; Yield: 105% w/w, HPLC Purity: 71% cis and 27% trans isomer (HPLC).

EXAMPLE 7

The reaction scheme of this example is generally shown below in Scheme IX.

SCHEME IX

1]CICOCH2C1 2]crystn

Figure imgf000018_0001
 
Formula m
Figure imgf000018_0002

Formula H

Into a clean dry flask charge with dichloromethane (400 ml) under a nitrogen atmosphere was added the compound of Formula III obtained in Example 6 and triethylamine (28.96 g) under stirring at 20 to 3O0C. The reaction mixture was then cooled to 0 to 50C. A mixture of chloroacetyl chloride (25.85 g) in dichloromethane (100 ml) was prepared and slowly added to the reaction mixture while maintaining the temperature between -5 to 50C in 1-2 hrs. The reaction mixture was stirred at 0 to 50C for 30 min and then saturated sodium bicarbonate solution (100 ml) was added at 5 to 100C under stirring. The temperature of the reaction mixture was raised to 25 to 300C and stirred at this temperature for 15 minutes. The layers were then separated. The lower MDC layer was collected, washed twice with 100 ml water and dried over anhydrous sodium sulfate. The

MDC layer was concentrated to distill out MDC until a stirrable mass was left behind. The mass was cooled to 25-3O0C and filtered, washed, to yield off-white to light yellow colored solids. The resulted product was the cis isomer, the trans isomer left behind in the mother liquor.

Yield = 25.5 g (50%w/w); HPLC Purity: > 97%.

The physical and spectral data was similar to that obtained in Example 2.

EXAMPLE 8

The reaction scheme of this example is generally shown below in SchemeX.

SCHEME X

Figure imgf000019_0001

Formula II

Into a clean dry round bottom (RB) flask was charged THF (1625 ml) under a nitrogen blanket and then cooled to -35 to -400C. Next, 505 ml n-butyllithium (1.6 m solution in hexane) was added while maintaining the temperature between -35 to -4O0C. After the addition was complete, the reaction mixture was stirred at -35 to -4O0C for 15 minutes. 72 ml diisopropyl amine was then added at -35 to -400C and then stirred at 0-50C for 1 hr. A solution of Compound of formula II (125 g) obtained in Example 7 in THF (625 ml) was prepared and slowly added to the reaction mixture while maintaining the temperature between -40 to -5O0C. After the addition was complete, the reaction mixture was stirred at -35 to -400C for 2-6 hours. Saturated aqueous ammonium chloride solution (250 ml) and ethyl acetate (125 ml) was added to the reaction mixture at -35 to -400C. The temperature was raised to 25 to 300C and the two layers formed were separated. The upper organic layer was collected. The lower aqueous layer was extracted with ethyl acetate (65 ml). The organic layers were combined together and distilled. Isopropyl alcohol (1250 ml) was added and the distillation was continued. A mixture of methanol (250 ml) and isopropanol (375 ml) were added and crude tadalafϊl was obtained upon cooling. The crude product was filtered, washed with water and dried. [0076] Yield: 60 g; (48% w/w); HPLC Purity: >99%.

EXAMPLE 9

Purification of crude Tadalafil

The crude tadalafil obtained in Example 8 was suspended in methanol (600 ml) and stirred for 1 hour at reflux. The mixture was cooled and the solids obtained were filtered and washed with chilled methanol (60 ml). The wet product was dried at under vacuum.

Yield: 56 g; HPLC Purity: 99.8%.

……………………………………………………………………………………

Beilstein J. Org. Chem. 2011, 7, 442–495.

http://www.beilstein-journals.org/bjoc/single/articleFullText.htm?publicId=1860-5397-7-57#S28

A different approach was used in the synthesis of the phosphodiesterase inhibitor tadalafil (132, Cialis) starting from commercially available (D)-tryptophan methyl ester to form the indolopiperidine motif 135 via a Pictet–Spengler reaction followed by a double condensation to install the additional diketopiperazine ring (Scheme 28[38,39].

[1860-5397-7-57-i28]
Scheme 28: Optimised Pictet–Spengler reaction towards tadalafil.

To achieve the high levels of cis selectivity required from the Pictet–Spengler reaction, an extensive investigation of solvents and the influence of additives was undertaken [40]. It was identified that the use of a specific 23 mol % of benzoic acid significantly increased the cis/trans ratio from a base level of 55:45 to 92:8 (16 h reaction time at ambient temperature) in an overall yield of 86%. It was also determined that more polar solvents such as acetonitrile and nitromethane preferentially solvated the trans product and thereby allowed the isolation of the ciscompound by precipitation. It was also shown that by heating the reaction mixture under reflux the product distribution could be driven to the thermodynamically more favoured cis isomer having both the ester and the piperonyl moiety in equatorial positions. Hence, after heating under reflux for 8 h the cis/trans ratio was found to be 99:1 and the product could be isolated in an overall yield of 91%. This work represents an impressive example of a well considered and executed process optimisation study.

………………………………

The process disclosed in the patent US 5 859 006 (Scheme 1) involves condensation of D-tryptophan methyl ester with a piperonal derivative to yield a compound of formula (II). After conversion into a thioamide derivative of formula (III), cyclization occurs in presence of both an alkylating and reducing agents to provide a tetrahydro-β-carboline derivative of formula (IV), which on treatment with chloroacetyl chloride and methyl amine, gives Tadalafil. The compound of formula (IV) can also be obtained in one step, after separation of the other diastereoisomer, by a Pictet Spengler reaction between D-tryptophan methyl ester and piperonal in presence of an acid, such as trifluoroacetic acid.

  • Figure imgb0002
  • The patent application WO2007/10038 discloses the reaction of D-tryptophan with piperonal to provide a tetrahydro-β-carboline acid that was cyclised to Tadalafil in presence of a sarcosine derivative.
    The patent application WO2007/1107 discloses the reaction of D-tryptophan methyl amide with piperonal, to provide an intermediate that after reaction with chloroacetyl chloride cyclises to Tadalafil in presence of butyllithium.
    Thus, the active substance prepared by the processes known up till now can only be obtained in a satisfactory quality after running through a large number of process steps. Moreover a toxic alkylating agent, such as methylamine, is often used.

EP2181997A1

Example 1

  • (1R,3R)-methyl-1,2,3,4-tetrahydro-2-(2-(benzyl(methyl)amino)acetyl)-1-(3,4-methylenedioxyphenyl)-9H-pyrido[3,4-b]indole-3-carboxylate (VII)
    Figure imgb0007
  •  
    A 50 mL three-necked flask fitted with thermometer and reflux condenser was charged with (1R,3R)-methyl 1,2,3,4-tetrahydro-2-chloroacetyl-1-(3,4-methylenedioxyphenyl)-9H-pyrido [3,4-b] indole-3-carboxylate (VI) (1.39 g, 3.26 mmol), DMA (5.33 mL), K2CO3 (0.5 g, 3.6 mmol) and N-benzylmethylamine (0.41 mL, 3.26 mmol). The resultant solution was stirred at room temperature. After 2 hours, the mixture was poured in brine (20 mL) and extracted with isopropyl acetate. The combined organic phases were washed with brine (3 x 5 mL), dried over sodium sulfate and concentrated to a residue under reduced pressure, affording 1.5 g of the desired product (VII), as a white solid. Yield: 70%.
    1H NMR (d6-DMSO 300 MHz, 298K) 2.24 (s, 3H), 2.94-3.00 (m, 5H), 3.44-3.68 (m, 3H), 5.56 (bd, J = 6.4, 1H), 5.95 (s, 1H), 5.96 (s, 1H), 6.55 (bd, J = 7.4, 1H), 6.75 (bs, 1H), 6.77 (d, J = 8.0, 1H), 6.84 (bs, 1H), 7.05 (td, J = 7.4, 0.9, 1H), 7.12 (td, J = 7.5, 1.2, 1H), 7.17-7.32 (m, 6H), 7.56 (d, J = 7.7, 1H), 10.76 (bs, 1H)
    13C NMR (d6-DMSO 75.4 MHz, 298K) 21.9, 42.5, 51.3, 51.9, 52.4, 61.0, 61.7, 101.5, 107.0, 108.0, 109.8, 111.8, 118.5, 119.2, 122.0, 123.0, 126.7, 127.7, 128.7, 129.6, 131.1, 134.7, 137.1, 138.6, 147.1, 147.5, 170.6, 171.5

Example 2

  • (6R-trans)-6-(1,3-benzodioxol-5-yl)-2,3,6,7,12,12a-hexahydro-2-methyl-pyrazino [1',2':1,6] pyrido [3,4-b] indole-1,4-dione (Tadalafil) (I)
    Figure imgb0008

    Under H2 atmosphere (3 atm) and magnetic stirring, Raney® Ni (2800 slurry in water, 0.0276 g, 0.47 mmol), previously washed with methanol (3 times), was added to a solution of (1R,3R)-methyl-1,2,3,4-tetrahydro-2-(2-(benzyl(methyl)amino)acetyl)-1-(3,4-methylenedioxyphenyl)-9H-pyrido[3,4-b]indole-3-carboxylate (VII) (3.00 g, 4.70 mmol) in DMA (21.3 mL). The mixture was heated at 90°C for 17 hours and then cooled to room temperature. The suspension was filtered over a pad of Celite® and the resulting solutionand the resulting solution was concentrated until 6 mL. Methanol (12 mL) was added and the solid which was so obtained was filtered over Buchner, washed with methanol (4 mL) and oven-dried under reduced pressure for 2 hours, affording 1.3 g of the title compound, as a white solid. Yield: 70%
    1H NMR (d6-DMSO 300 MHz, 298K): 2.91-3.00 (m, 4H), 3.32 (s, 1H), 3.47-3.54 (dd, J = 4.6, 11.3, 1H), 3.93 (d, J = 17.1, 1H), 4.17 (d, J = 17.1, 1H), 4.35-4.40 (dd, J = 4.27, 11.6, 1H), 5.91 (s, 2H), 6.11 (s, 1H), 6.76 (s, 2H), 6.85 (s, 1H), 6.98-7.06 (m, 2H), 7.28 (d, J = 7.9, 1H), 7.52 (d, J = 7.3, 1H), 11.0 (s, 1H)
    13C NMR (d6-DMSO 75.4 MHz, 298K) 23.8, 33.4, 52.0, 55.9, 56.1, 101.5, 105.3, 107.6, 108.6, 111.9, 118.7, 119.5, 119.9, 121.8, 126.4, 134.5, 136.8, 137.6, 146.7, 147.6, 167.1, 167.5

References

  1.  Daugan, A; Grondin P, Ruault C, Le Monnier de Gouville AC, Coste H, Kirilovsky J, Hyafil F, Labaudinière R (October 9, 2003). “The discovery of tadalafil: a novel and highly selective PDE5 inhibitor. 1: 5,6,11,11a-tetrahydro-1H-imidazo[1',5':1,6]pyrido[3,4-b]indole-1,3(2H)-dione analogues”. Journal of Medicinal Chemistry 46 (21): 4525–32. doi:10.1021/jm030056e . PMID 14521414.
  2.  Richards, Rhonda (September 17, 1991). “ICOS At A Crest On Roller Coaster”. USA Today. p. 3B.
  3.  Ervin, Keith (June 21, 1998). “Deep Pockets + Intense Research + Total Control = The Formula — Bothell Biotech Icos Keeps The Pipeline Full Of Promise”The Seattle Times. p. F1. Retrieved January 10, 2009.
  4.  Revill, Jo (February 2, 2003). “Drugs giant says its new pill will pack more punch than rival Viagra”The Observer. Retrieved 2007-04-06.
  5.  http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm274642.htm
  6.  https://www.consumerreports.org/health/resources/pdf/best-buy-drugs/money-saving-guides/english/PillSplitting-FINAL.pdf
  7.  “FDA Announces Revisions to Labels for Cialis, Levitra and Viagra”Food and Drug Administration. 2007-10-18. Retrieved 2009-09-28.
  8.  “Cialis: Warnings, Precautions, Pregnancy, Nursing, Abuse”. RxList. 2007. Retrieved 2007-04-06.
  9.  Bischoff, E (June 2004). “Potency, selectivity, and consequences of nonselectivity of PDE inhibition”International Journal of Impotence Research 16: S11–4.doi:10.1038/sj.ijir.3901208 . PMID 15224129. Retrieved January 19, 2009.
  10.  Elliott, Stuart (January 10, 2006). “For Impotence Drugs, Less Wink-Wink”The New York Times. p. C2. Retrieved January 15, 2009.
  11.  Elliott, Stuart (April 25, 2004). “Viagra and the Battle of the Awkward Ads”The New York Times. p. 1. Retrieved January 15, 2009.
  12. McCarthy, Shawn (March 5, 2005). “First they tried to play it safe; Ads for erectile dysfunction drug Cialis bared all – including a scary potential side effect. It was risky but it has paid off”. The Globe and Mail. p. B4.
  13.  Loyd, Linda (July 6, 2003). “Two Pills Look to Topple Viagra’s Reign in Market; Levitra Expects Approval Next Month, Cialis Later This Year”. The Philadelphia Inquirer. p. E01.
  14. 38  is 1 below
  15. 39 is 2 below
  16. 40 is 3 below
    1. daugan, A. C.-M. Tetracyclic Derivatives; Process of Preparation and Use. U.S. Patent 5,859,006, Jan 12, 1999.
    2. Daugan, A. C.-M. Tetracyclic Derivatives, Process of Preparation and Use. U.S. Patent 6,025,494, Feb 15, 2000.
    3. Shi, X.-X.; Liu, S.-L.; Xu, W.; Xu, Y.-L. Tetrahedron: Asymmetry 2008, 19, 435–442.doi:10.1016/j.tetasy.2007.12.017

DAUGAN A ET AL: “THE DISCOVERY OF TADALAFIL: A NOVEL AND HIGHLY SELECTIVE PDE5 INHIBITOR. 2: 2,3,6,7,12,12A-HEXAHYDROPYRAZINO[1′,2′:1,6 ÜPYRIDO[3,4-B ÜINDOLE-1,4-DIONE” JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. WASHINGTON, US, vol. 46, no. 21, 2003, pages 4533-4542, XP008052656 ISSN: 0022-2623

WO2009004557A2 * Jun 28, 2008 Jan 8, 2009 Ranbaxy Lab Ltd A process for the preparation of intermediates of tetracyclic compounds
WO2009148341A1 Jun 3, 2009 Dec 10, 2009 Zaklady Farmaceutyczne Polpharma Sa Process for preparation of tadalafil
WO2012107549A1 Feb 10, 2012 Aug 16, 2012 Interquim, S.A. PROCESS FOR OBTAINING COMPOUNDS DERIVED FROM TETRAHYDRO-ß-CARBOLINE
EP2107059A1 Mar 31, 2008 Oct 7, 2009 LEK Pharmaceuticals D.D. Conversion of tryptophan into ß-carboline derivatives
US8445698 Jun 28, 2008 May 21, 2013 Ranbaxy Laboratories Limited Process for the preparation of an intermediate of tadalafil

Filed under: Uncategorized Tagged: cialis, Tadalafil

Grape Seeds Fight Bowel Cancer

$
0
0

Originally posted on lyranara.me:

University of Adelaide research has shown for the first time that grape seed can aid the effectiveness of chemotherapy in killing colon cancer cells as well as reducing the chemotherapy’s side effects.

Published in the prestigious journal PLOS ONE, the researchers say that combining grape seed extracts with chemotherapy has potential as a new approach for bowel cancer treatment – to both reduce intestinal damage commonly caused by cancer chemotherapy and to enhance its effect.

Lead author Dr Amy Cheah says there is a growing body of evidence about the antioxidant health benefits of grape seed tannins or polyphenols as anti-inflammatory agents and, more recently, for their anti-cancer properties.

“This is the first study showing that grape seed can enhance the potency of one of the major chemotherapy drugs in its action against colon cancer cells,” says Dr Cheah, researcher in the School of Agriculture, Food and Wine.

View original


Filed under: Uncategorized

FDA Approves Vimizim to Treat Mucopolysaccharidosis Type IVA

$
0
0

STRUCTURAL FORMULA
Monomer
APQPPNILLL LMDDMGWGDL GVYGEPSRET PNLDRMAAEG LLFPNFYSAN 50
PLCSPSRAAL LTGRLPIRNG FYTTNAHARN AYTPQEIVGG IPDSEQLLPE 100
LLKKAGYVSK IVGKWHLGHR PQFHPLKHGF DEWFGSPNCH FGPYDNKARP 150
NIPVYRDWEM VGRYYEEFPI NLKTGEANLT QIYLQEALDF IKRQARHHPF 200
FLYWAVDATH APVYASKPFL GTSQRGRYGD AVREIDDSIG KILELLQDLH 250
VADNTFVFFT SDNGAALISA PEQGGSNGPF LCGKQTTFEG GMREPALAWW 300
PGHVTAGQVS HQLGSIMDLF TTSLALAGLT PPSDRAIDGL NLLPTLLQGR 350
LMDRPIFYYR GDTLMAATLG QHKAHFWTWT NSWENFRQGI DFCPGQNVSG 400
VTTHNLEDHT KLPLIFHLGR DPGERFPLSF ASAEYQEALS RITSVVQQHQ 450
EALVPAQPQL NVCNWAVMNW APPGCEKLGK CLTPPESIPK KCLWSH 496
Disulfide bridges
139-139′ 282-393 282′-393′ 463-492 463′-492′ 475-481 475′-481′
Modified residues
C
53 , 53′
3-oxoAla
O
CO2H
H NH2

Glycosylation sites (N)
Asn-178 Asn-178′ Asn-397 Asn-397′

Vimizim (elosufase alfa)

Elosulfase alfa nonproprietary drug name  GET STRUCTURE

MOLECULAR FORMULA C5020H7588N1364O1418S34

MOLECULAR WEIGHT 110.8 kDa (peptide)

SPONSOR BioMarin Pharmaceutical Inc.

CODE DESIGNATION BMN 110, rhGALNS

CAS REGISTRY NUMBER 9025-60-9

THERAPEUTIC CLAIM Treatment of Morquio Syndrome

CHEMICAL NAMES

1. Sulfatase, chondroitin

2. Human N-acetylgalactosamine-6-sulfatase (chondroitinsulfatase, galactose-6-sulfate
sulfatase, EC=3.1.6.4) dimer (139-139′)-disulfide glycosylated (produced by CHO cells)

Company: BioMarin Pharmaceutical Inc.
Date of FDA Approval: February 14, 2014
Treatment for: Mucopolysaccharidosis Type IVA

  • BMN 110
  • Chondroitin 6-sulfatase
  • Chondroitin sulfatase
  • Chondroitin sulfate sulfatase
  • Chondroitinase
  • Chondrosulfatase
  • E.C. 3.1.6.4
  • Elosulfase alfa
  • rhGALNS
  • UNII-ODJ69JZG85
  • Vimizim

CLINICAL….http://clinicaltrials.gov/search/intervention=Elosulfase%20alfa%20OR%20bmn%20110

Vimizim (elosufase alfa) is an enzyme replacement therapy for patients with Mucopolysaccharidosis Type IVA (Morquio A syndrome).

  • FDA Advisory Committee Recommends Approval for BioMarin’s Vimizim for the Treatment of Patients With Morquio A Syndrome - November 20, 2013

Feb 16, 2014 Approval FDA Approves Vimizim to Treat Mucopolysaccharidosis Type IVA

The U.S. Food and Drug Administration today approved Vimizim (elosulfase alfa), the first FDA-approved treatment for Mucopolysaccharidosis Type IVA (Morquio A syndrome). Morquio A syndrome is a rare, autosomal recessive lysosomal storage disease caused by a deficiency in N-acetylgalactosamine-6-sulfate sulfatase (GALNS). Vimizim is intended to replace the missing GALNS enzyme involved in an important metabolic pathway. Absence of this enzyme leads to problems with bone development, growth and mobility. There are approximately 800 patients with Morquio A syndrome in the United States.

Vimizim was granted priority review. An FDA priority review provides for an expedited review of drugs for serious diseases or conditions that may offer major advances in treatment. Vimizim is also the first drug to receive the Rare Pediatric Disease Priority Review Voucher – a provision that aims to encourage development of new drugs and biologics for the prevention and treatment of rare pediatric diseases.

“This approval and rare pediatric disease priority review voucher underscores the agency’s commitment to making treatments available to patients with rare diseases,” said Andrew E. Mulberg, M.D., deputy director, Division of Gastroenterology and Inborn Errors Products in the FDA’s Center for Drug Evaluation and Research (CDER). “Prior to today’s approval, patients with this rare disease have had no approved drug treatment options.”

The safety and effectiveness of Vimizim were established in a clinical trial involving 176 participants with Morquio A syndrome, ranging in age from 5 to 57 years. Participants treated with Vimizim showed greater improvement in a 6-minute walk test than participants treated with placebo. On average, patients treated with Vimizim in the trial walked 22.5 meters farther in 6 minutes compared to the patients who received placebo.

The most common side effects in patients treated with Vimizim during clinical trials included fever, vomiting, headache, nausea, abdominal pain, chills and fatigue. The safety and effectiveness of Vimizim have not been established in pediatric patients less than 5 years of age. Vimizim is being approved with a boxed warning to include the risk of anaphylaxis. During clinical trials, life-threatening anaphylactic reactions occurred in some patients during Vimizim infusions.

Vimizim is marketed by Novato, Calif.-based BioMarin Pharmaceutical Inc.

Elosulfase alfa (GALNS), a proposed treatment for Morqio A syndrome. Morquio A syndrome is an inherited, autosomal recessive disease caused by a deficiency of a particular lysosomal enzyme, N- acetylgalactosamine- 6 sulfatase. BioMarin’s experimental drug for Morquio A syndrome is an enzyme replacement of elosulfase alfa (called BMN 110), which is designed to clear keratan sulfate from the lysosome. BMN 110 is being studied to determine if it is safe, if it will slow the progression of the disease and if it will improve some of the symptoms.

BioMarin started BMN 110 clinical studies in humans in 2009 to evaluate safety and efficacy. In a phase III Multicenter, Multinational, Extension Studythe Long-Term Efficacy and Safety of BMN 110 in Patients With Mucopolysaccharidosis IVA (Morquio A Syndrome) MOR-005 was evaluated. Participants will receive 2 mg/kg weekly or every other weekly dosing of study drug via infusion until the MOR- 004 study is unblinded and the optimal dose is selected. All subjects will then be treated with the optimal dose for up to approximately 5 years or until the drug is approved.


Filed under: FDA 2014, NEW DRUGS, Uncategorized Tagged: BIOMARIN, Elosulfase alfa, Vimizim

IkB kinase inhibitors , SANOFI, for osteoarthritis

$
0
0

Figure imgf000002_0001

K salt monohydrate, N-​[[2-​[2-​(methylamino)​-​4-​pyrimidinyl]​-​1H-​indol-​5-​yl]​carbonyl]​-​3-​(phenyl-​2-​pyridinylamino)​-   L-​Alanine,

2-{[2-(2-methylamino-pyrimidin-4-yl)-lH-indole-5- carbonyl]-amino}-3-(phenylpyridin-2-yl-amino)-propionic acid, as the monopotassium monohydrate salt., 899418-66-7 , C28 H25 N7 O3 . H2 O . K

IC 50= 0.4 nm

K SALT

L-​Alanine, N-​[[2-​[2-​(methylamino)​-​4-​pyrimidinyl]​-​1H-​indol-​5-​yl]​carbonyl]​-​3-​(phenyl-​2-​pyridinylamino)​-​, monopotassium salt , 899418-65-6, C28 H25 N7 O3 . K

Free acid

  • C28 H25 N7 O3
  • N-​[[2-​[2-​(methylamino)​-​4-​pyrimidinyl]​-​1H-​indol-​5-​yl]​carbonyl]​-​3-​(phenyl-​2-​pyridinylamino)​-   L-​Alanine,
  •     869796-50-9

As an inhibitor of IKB kinase, the compound of the invention, functions via the selective inhibition of IKK, particularly an IKK-2 inhibitor; as well as exhibiting localized activity, as opposed to a systemic activity. Such an inhibitor is particularly useful for treating a patient suffering from or subject to IKK- 2 mediated pathological diseases or conditions, e.g., asthma, rhinitis, chronic obstructive pulmonary disorder (COPD), or COPD exacerbations, that could be ameliorated by the targeted administering of the inhibitor.

Sanofi.. INNOVATOR

SANOFI LISTS http://clinicaltrials.gov/show/NCT01463488 SAR113945 AS  IkB kinase inhibitors IN PHASE II…. BUT I AM NOT SURE OF THIS….Protein Kinases as Small Molecule Inhibitor Targets  - ResearchGa click here to see  see table 7 (cont)……2227

EMAIL ME amcrasto@gmail.com

WO 2005113554

………………….

Synthesis

WO2006076318A1

EXAMPLES

Example 1, Step 1

Synthesis of 2-{[2-(2-Methylamino-pyrimidin-4-yl)-lH-indole-5-carbonyl]amino}-3-(phenyl-pyridin-

2-yl-amino)-propionic acid

Figure imgf000016_0001

6.04 mmol of the 2-{[2-(2-methylamino-pyrimidin-4-yl)-lH-indole-5-carbonyl]-amino}-3-(phenyl- ρyridin-2-yl-amino)-propionic acid, methyl ester prepared essentially as described in patent application WO2005/113544, is dissolved in 70 mL of ethanol. 24.2 mL of 0.5 N aqueous ΝaOΗ is added and the mixture is stirred at room temperature for 2 h. After the reaction is complete, the pH is adjusted to ~5 using 1 N HCl. Water is added slowly and the resulting precipitate is filtered off and washed with water. After drying under reduced pressure of about 1 mbar at 400C, 2.49 g of 2-{[2-(2-methylamino- pyrimidin-4-yl)-lH-indole-5-carbonyl]-arnino}-3-(phenyl-pyridin-2-yl-amino)-propionic acid is isolated. Empirical formula C28H25N7O3; M. W. = 507.56; MS (M+H) 508.3. 1H NMR (DMSO-^6) 2.95 (s, 3 H), 4.32-4.50 (m, 2 H), 4.65-4.72 (m, 1 H), 6.29-6.36 (d? 1 H), 6.70- 6.79 (m, 1 H), 6.90-7.10 (sb, 1 H), 7.13-7.19 (m, 1 H), 7.22-7.38 (m, 4 H), 7.40-7.48 (m, 3 H), 7.50-7.55 (m, 1 H), 7.57-7.60 (m, 1 H), 7.96 (bs, 1 H), 8.34-8.40(m, 2 H), 8.80-8.90 (d, 1 H), 11.80 (s, 1 H) 12.8 (bs, IH). Chiral HPLC shows 94% ee.

Example 1, Step 2

Enantiomeric Purification of 2-{[2-(2-Methylaminopyrimidin-4-yl)-lH-indole-5-carbonyl]amino}-3-

(phenylpyridin-2-yl-amino)-propionic acid

Figure imgf000017_0001

2- { [2-(2-methylaminopyrimidin-4-yl)- lH-indole-5-carbonyl]amino} -3-(phenylpyridin-2-yl-amino)- propionic acid, prepared essentially according to Example 1, Step 1 above, is heated under reflux for 15 minutes. The insoluble racemic compound is removed by hot filtration. The TΗF of the resulting filtrate is removed by distillation and the residue is precipitated by the addition of isopropanol. After drying under reduced pressure of about 1 mbar at 400C, the desired 2-{[2-(2-methylaminopyrimidin-4- yl)-lH-indole-5-carbonyl]amino}-3-(phenylpyridin-2-yl-amino)-propionic acid is isolated with an ee = 98.5%.

Example 1, Step 3

Synthesis of 2-{[2-(2-Methylamino-pyrimidin-4-yl)-lH-indole-5-carbonyl]-amino}-3-(phenyl-pjτidin- 2-yl-amino)-propionic acid monopotassium monohydrate salt

Figure imgf000018_0001

To a slurry of 2-{[2-(2-methylaminopyrimidin-4-yl)-lH-indole-5-carbonyl]amino}-3-(phenylpyridin- 2-yl-amino)-propionic acid (50.8 mmol from Example 1, Step 2 above) in H2O and EtOH is added 1.02 M KOH (2.00 equiv) with vigorous swirling. The mixture is heated to 670C with swirling on a steam bath to dissolve the starting material, while braking up any remaining clumps. After several minutes the clear orange solution is filtered and the flask containing the filtrate is wrapped in aluminum foil and allowed to cool slowly to room temperature in the hot water remaining in the steam bath. After 17 hours, the mixture is cooled in an ice-bath and the salt is collected by filtration and washed 4 times with ice-cold H2O. The last two washes have a pH of 8. The salt is dried in a vacuum oven at 45 0C with an N2 bleed to yield the desired compound as fine needles:1H NMR (DMSO-«k) 2.95 (s,3 H)5 3.95-4.05 (m, 1 H), 4.35-4.40 (m, IH), 4.55-4.62 (m, 1 H), 6.35-6.39 (d, 1 H), 6.58-6.60 (m, IH), 6.90-7.10 (sb, 1 H), 7.13-7.19 (m, 1 H), 7.22-7.38 (m, 6 H), 7.40-7.48 (m, 3 H), 7.57-7.60 (m,l H), 7.70 (s, 1 H), 8.10-8.15(d, 1 H), 8.30 (bs, 1 H), 11.80 (s, 1 H); LC-MS m/z 509 (M+ + 2), 508 (M+ H- I), 275, 254 (100). Anal. Calcd for C28H24KN7O3-H2O (563.66): C, 59.67; H, 4.65; N, 17.39; K. 6.94; H2O (Karl Fischer), 3.20. Found: C, 59.59; H, 4.66; N, 17.39; K5 6.44; H2O (Karl Fischer), 3.16. Chiral HPLC showed 99.5% S-enantiomer.

Example 2 Synthesis of 2-{[2-(2-Methylammo-pyrimidin-4-yl)-lH-indole-5-carbonyl]-amino}-3-(phenyl-pyridin-

2-yl-amino)-propionic acid monopotassium monohydrate salt

Figure imgf000018_0002

As an alternative procedure for preparing the compound of formula Ha3 (3.8 mmol) of methyl ester 1 is dissolved in ethanol and water and 2 N aqueous KOH is added and the mixture is stirred at room temperature for 4 h. The product starts to crystallize and the mixture is diluted with additional water. The resulting crystalline precipitate is filtered off and washed with water. After drying under reduced pressure of about 1 mbar at 400C, the monopotassium monohydrate salt π is isolated. Empirical formula C28H24KN7O3-H2O M.W. = 563.65; MS (free acid, M+H) 508.3. 1H ΝMR (DMSO-J6) 2.95 (s, 3 H), 3.95-4.05 (m, 1 H), 4.35-4.40 (m, IH), 4.55-4.62 (m, 1 H), 6.35-6.39 (d, 1 H), 6.58-6.60 (m, 1 H), 6.90-7.10 (sb, 1 H), 7.13-7.19 (m, 1 H), 7.22-7.38 (m, 6 H), 7.40-7.48 (m, 3 H), 7.57-7.60 (m, 1 H), 7.70 (s, 1 H), 8.10-8.15(d, 1 H), 8.30 (bs, 1 H), 11.80 (s, 1 H). Water (Karl-Fischer): 3.2% (Monohydrate). XRPD (2 theta): 5.28, 6.45, 7.97, 9.46, 10.18, 10.93, 13.23, 13.66, 14.94, 15.94, 16.71, 18.15, 19.49, 20.38, 21.04, 21.42, 23.76, 24.38, 25.36, 25.71, 26.19, 27.13, 27.67, 28.13, 28.61, 29.12, 29.75, 30.95, 31.37, 32.94. ee: 99.8% (Chiralpak AD-H, 250 x 4.6mm, Heptane : EtOH : MeOH 5 : 1 : 1, RT).

It is known that indole derivatives are used as units for the synthesis of active pharmaceutical ingredients. For example, 2-(2-aminopyrimidin-4-yl)-1H-indole-5-carboxylic acids or their salts are important units for the preparation of IkB kinase inhibitors (see WO 01/30774 A1):

Figure US08232395-20120731-C00002

2-(2-Aminopyrimidin-4-yl)-1H-indole-5-carboxylic acids can be prepared by classical Fischer indole synthesis starting from the corresponding 4-acetylpyrimidines (III) and 4-hydrazinobenzoic acid (II) (see scheme 1):

Figure US08232395-20120731-C00003

One disadvantage here is the severe reaction conditions which are required for a full conversion. Secondly, the products of this reaction are obtained in a mixture with the corresponding oligomers, which leads to a poor isolability, especially with regard to the filtration times. Moreover, these oligomers, owing to the low solubility of 2-(2-aminopyrimidin-4-yl)-1H-indole-5-carboxylic acids in organic solvents, can only be removed with difficulty and are entrained as an impurity in the further reactions, in some cases up to the active ingredient.

Here are two ways to make a kinase inhibitor intermediates. 

http://www.google.com/patents/US8232395

J. Graeser and co-inventors describe indole derivatives such as 4 and 12 as intermediates for preparingIκB kinase inhibitors. Although indoles can be prepared by the classical Fisher synthesis, the inventors state that this method is not satisfactory when it is used for making the desired compounds. Severe reaction conditions are needed, and oligomeric compounds are formed that are difficult to remove.

The inventors describe two routes for preparing the desired compounds. The first route (Figure 1, top) begins with the reaction of indoleboronic acid 1 and chloropyrimidine 2in the presence of (Ph3P)4Pd to form 3, which is isolated in 93% yield and 96% purity. Compound 3 is converted to amine derivative 4 by treating it with MeNH2. The product was isolated in quantitative yield and with 97.6% purity. If desired, the ester group in 4can be hydrolyzed with NaOH to produce sodium salt 5.

Indoleboronic acid 1 is obtained by treating tert-butoxycarbonyl (Boc)–protected indole6 with B(O-i-Pr)3 in the presence of LiN-i-Pr2 (Figure 1, bottom) The reaction initially forms Boc-protected compound 7. After acid hydrolysis, 1 is isolated in 61% yield with 92.7% purity.

The inventors mention the advantage of using unprotected indole 1 in the reaction with2 rather than the N-protected compound. Their explanation is that although some 6 is formed by the loss of the boronate group from 1 during the coupling reaction with 26does not subsequently react with 2. Hence the yield of 3 in the coupling step is not reduced.

The second route to the desired compound is quite different from the first. Figure 2 outlines the process for preparing 12, the methyl ester analogue of 4. This route starts with the preparation of silylated acetylene compound 8, isolated in 90% yield with 99% purity after what is described as an aqueous workup. In the next step, the silyl group is removed, and primary alkyne 9 is isolated in quantitative yield. Alkyne 9 is treated with chloropyrimidine 10 in the presence of CuI and a palladium catalyst in DMF to give 11, which is isolated after aqueous workup in 85% yield and 99.7% purity. The cyclization of 11 to form 12 is carried out with a strong base such as KO-t-Bu. The product is isolated after an aqueous workup in 58% yield and 92.3% purity.

Although the inventors do not provide details for preparing 10, they state that it can be synthesized by the route shown at the bottom of Figure 2. The reaction produces isomers 10 and 13, which can be separated by chromatographic methods or steam distillation.

The inventors describe an alternative route to 4 in which 1 reacts with 10 in place of 2. They point out that 1 reacts with a mixture of 10 and 13 to give 4. Although it may be expected that 13 would react to give an isomer of 4, they claim that this reaction does not take place. No examples of the reaction of 1 and 10 with or without 13 are given Also, the inventors mention “aqueous workup” several times but do not explain what this means.

These processes provide alternative routes to a drug intermediate that overcome product isolation problems. (Sanofi [Paris]. US Patent 8,232,395, July 31, 2012;

US8546567

EXAMPLE 1 Synthesis of ethyl 2-(2-chloropyrimidin-4-yl)-1H-indole-5-carboxylate

Figure US08546567-20131001-C00026

28 g (114 mmol) of 2-borono-5-ethoxycarbonylindole, 12 g (113 mmol) of sodium carbonate and 17.2 g of 2,4-(113 mmol) dichloropyrimidine were initially charged in 412 ml of ethanol. The clear solution was freed of oxygen by vigorous stirring and passing argon through (20 minutes). At RT, 2.67 g of tetrakis(triphenylphosphine)palladium(0) were added. The mixture was heated to from 65° C. to 70° C. for 2 hours (h). Subsequently, 112 ml of water and 112 ml of 30% hydrochloric acid were added and the mixture was cooled to 0° C. After filtration and drying under reduced pressure, 37.3 g (93% of theory) of ethyl 2-(2-chloropyrimidin-4-yl)-1H-indole-5-carboxylate were obtained (HPLC >96%).

The purity was determined by high-pressure liquid chromatography (HPLC):

Column: Waters Symetry Shield RP8 3.9 * 150
Temperature: 40° C.
Flow rate: 1 ml/min Injection volume: 10 μl
Pressure: 90 bar UV: 254 nm
Eluent: A: Water/trifluoroacetic acid (0.05%)
B: Acetonitrile/trifluoroacetic acid (0.05%)
Time (min)  0 15 20 25 30
A (%) 80 25 25 80 80
B (%) 20 75 75 20 20
Retention time of 12.6 min
title compound:

EXAMPLE 2 Synthesis of ethyl 2-(2-methylaminopyrimidin-4-yl)-1H-indole-5-carboxylate

Figure US08546567-20131001-C00027

30 g (95.4 mmol) of ethyl 2-(2-chloropyrimidin-4-yl)-1H-indole-5-carboxylate were initially charged and suspended in 150 ml of ethanol. 53.9 g of methylamine solution in ethanol (8 M) were added to this suspension which was heated to from 75° C. to 80° C. in an autoclave for 4 h. After concentration and washing with ethanol, 29.7 g of ethyl 2-(2-methylamino-pyrimidin-4-yl)-1H-indole-5-carboxylate were obtained (97.6 HPLC area %). LCMS: [M+H]⊕ 297.12

HPLC method as in example 1; retention time of title compound: 5.8 min

EXAMPLE 3 Synthesis of 2-(2-methylaminopyrimidin-4-yl)-1H-indole-5-carboxylic acid sodium salt

Figure US08546567-20131001-C00028

25 g of ethyl 2-(2-methylaminopyrimidin-4-yl)-1H-indole-5-carboxylate were admixed with 200 ml of ethanol and 24.5 g of 33% sodium hydroxide solution, and heated to from 65° C. to 70° C. for 4 h. After cooling, the mixture was filtered with suction and the precipitate was washed with 15 ml of ethanol/water (9:1). 24.5 g (87.6% of theory) of 2-(2-methylaminopyrimidin-4-yl)-1H-indole-5-carboxylic acid sodium salt were obtained (98.1 HPLC area %). LCMS: [M+H]⊕ 269.10

HPLC method as in example 1; retention time of title compound: 3.3 min

EXAMPLE 4 Synthesis of methyl 4-amino-3-trimethylsilylethynylbenzoate

Figure US08546567-20131001-C00029

5.83 g (20 mmol) of methyl 4-aminobenzoate, 20.2 g (198 mmol) of triethylamine and 80 ml of toluene were initially charged. The clear solution was freed of oxygen by vigorous stirring and passing argon through (20 minutes). At an internal temperature of 20° C., 3.2 g (33 mmol) of trimethylsilylacetylene, 76 mg of copper(I) iodide and 52 mg of triphenylphosphine were added. After aqueous workup, 5.45 g of 4-amino-3-trimethylsilylethynylbenzoate were obtained (HPLC: >99 area %). HPLC method as in example 1.

EXAMPLE 5 Synthesis of methyl 4-amino-3-ethynylbenzoate

Figure US08546567-20131001-C00030

1.9 g (7.7 mmol) of methyl 4-amino-3-trimethylsilylethynylbenzoate were initially charged in 20 ml of tetrahydrofuran (THF). At from 5° C. to 8° C., 8.45 ml (8.5 mmol) of tetrabutylammonium fluoride solution (1 M in THF) were added dropwise within 5 minutes. After 25 min at 2° C., 438 ml of acetic acid were added. After addition of water and extraction with dichloromethane, and after removal of the solvent, 1.35 g of methyl 4-amino-3-ethynylbenzoate were obtained. HPLC method as in example 1.

EXAMPLE 6 Synthesis of methyl 4-amino-3-(1-methylaminopyrimidin-4-yl)-ethynylbenzoate

Figure US08546567-20131001-C00031

3.0 g (17 mmol) of methyl 4-amino-3-ethynylbenzoate and 2.6 g (19 mmol) of 4-chloro-2-methylaminopyrimidine were initially charged in 20 ml of dimethylformamide (DMF) and 8.7 g (85 mmol) of triethylamine, and degassed with argon while stirring for 5 min. Subsequently, 65 mg of copper(I) iodide and 20 mg of tetrakis(triphenylamine)palladium(0) were added and the mixture was heated to 71° C. for 3 h. After aqueous workup, 4.1 g of methyl 4-amino-3-(1-methylaminopyrimidin-4-yl)ethynylbenzoate were obtained. (HPLC: 99.7 area %) HPLC method as in example 1.

EXAMPLE 7 Synthesis of methyl 2-(2-methylaminopyrimidin-4-yl)-1H-indole-5-carboxylate by cyclizing methyl 4-amino-3-(1-methylaminopyrimidin-4-yl)ethynylbenzoate

Figure US08546567-20131001-C00032

73 mg (0.7 mmol) of potassium tert-butoxide were dissolved in 1 ml of NMP and admixed with a solution of 140 mg (0.5 mmol) of methyl 4-amino-3-(1-methylaminopyrimidin-4-yl)ethynylbenzoate in 1 ml of NMP. Subsequently, stirring was continued at RT for 24 h. Aqueous workup afforded 115 mg of methyl 2-(2-methylaminopyrimidin-4-yl)-1H-indole-5-carboxylate (HPLC: 92.3 area %).

EXAMPLE 8 Synthesis of 2-borono-5-ethoxycarbonylindole

Figure US08546567-20131001-C00033

150 g (519 mmol) of N-Boc-5-ethoxycarbonylindole and 192 ml (833 mmol) of triisopropyl borate in 350 ml of toluene were admixed at from 5° C. to 10° C. with 350 ml of a 1.8 molar solution of LDA in THF. The mixture was stirred for a further 5 min and the reaction mixture was added to a solution of 278 g of 30% hydrochloric acid and 940 ml of water. Subsequently, the mixture was stirred at from 5° C. to 10° C. for 30 min. Thereafter, the mixture was filtered and the filtercake was suspended in 530 ml of ethanol. This suspension was added at 40° C. to a solution of 500 ml of 30% hydrochloric acid and 224 ml of ethanol. Subsequently, the mixture was stirred at from 40° C. to 45° C. for 2.5 h and admixed at 30° C. with 380 ml of water. The mixture was then cooled to from 10° C. to 15° C., stirred at this temperature for 30 min and filtered. Drying under reduced pressure afforded 79.5 g (61% of theory) of 2-borono-5-ethoxycarbonylindole (HPLC: 92.7 area %).

…………………………………………….

WO2004022553A1

C) Synthesis of the heterocyclic base

Figure imgf000017_0002

C.1) indole base synthesis. Of 2 – (2-methylamino-pyrimidin-4-yl) -1 H-indole-5-carboxylic acid (20) C.1.1) 1-Dimethylamino-4 ,4-dimethoxy-pent. -1-en-3-one (18)

100 g (0.76 mol) of 3,3-dimethoxy-2-butanone (16) of (17) (0.76 mol) at 120 ° C with stirring 90.2 g of 48 N, N-dimethylformamide dimethyl acetal h. The methanol formed during the reaction was continuously removed from the reaction solution by distillation. On cooling, the solution became a spontaneous crystallization, which was brought by adding a little heptane to completion. This gave 128.24 g of crude 18 (90% yield), which was reacted without further purification. Molecular formula C 9 Hι 7 N0 3, MW = 187.24, MS (M + H) 188.2 i H NMR (DMSO-de) 1.22 (s, 3H), 2.80 (s, 3H), 3.10 (s, 9H), 5.39. (d, J = 15 Hz, 1 H), 7:59 (d, J = 15 Hz, 1 H). . . . . . . .

C.1.2). [4 - (1,1-Dimethoxy-ethyl)-pyrimidin-2-yl]-methyl-amine (19)

1:22 g (53 mmol) of sodium were dissolved in 100 ml absolute ethanol. This was

Stirring 5.8 g (53 mmol) Methylguanidinhydrochlorid and 10 g (53 mmol) of 1-dimethylamino-4,4-dimethoxy-penM-en-3-one (18) and heated to boiling for 4 h. To stop the reaction, the ethanol was evaporated. The product 19 thus obtained was used without further purification for the subsequent reaction. Yield 11.5 g (58 mmol, quantitative) Molecular Formula C9H15N3O2, MW = 197.24, MS (M + H) 198.2 1 H NMR (DMSO-de) 1.45 (s, 3H), 2.78 (s, 3H), 3.10 (s,. 6H), 6.75 (d, J = .3 Hz, 1 H), 7.0 – 7.1 (s (b), 1 H), 8.30 (d, J = 3 Hz, 1 H).

C.1.3) 2 -. (2-methylamino-pyrimidin-4-yl) -1 H-indole-5-carboxylic acid (20) Into 150 ml of 50% sulfuric acid at room temperature 5 g (25 mmol) [4 - ( 1, 1 - dimethoxy-ethyl)-pyrimidin-2-yl]-methyl-amine (19) and, 3.85 g of 4-hydrazinobenzoic acid with stirring and heated 4 h at 130 ° C. The methanol formed during the reaction was continuously removed from the reaction solution by distillation. After cooling to 10 ° C the reaction mixture was poured into 200 mL of ice and adjusted to a pH of about 5.5 with concentrated sodium hydroxide solution. The precipitate formed from sodium sulfate, and the product mixture was filtered and the filter residue was extracted several times with methanol. The combined methanol extracts were concentrated and the product 20 by flash chromatography (DCM / methanol 9:1). Yield: 0.76 g (11%) Molecular formula Oι Hι3 N 4 4 0 2, MW = 268.28, MS (M + H) 269.1.

1 H NMR (DMSO-de) 2.95 (s, 3H), 6.90 – 7.10 (s (b), 1 H), 7.18 (d, J = 3 Hz, 1H), 7.4 (s, 1 H), 7:58 (d, J = 4.5 Hz, 1H), 7.80 (d, J = 4.5 Hz, 1H), 8.30 (s, 1H), 7.80 (d, J = 4.5 Hz, 1H), 8:38 (d, J = 3 Hz, 1H), 11.85 (s, 1H), 12:40 – 12.60 (s (b), 1 H).

US7285560 Aug 18, 2003 Oct 23, 2007 Sanofi-Aventis Deutschland Gmbh Indole derivatives or benzimidazole derivatives for modulating IκB kinase
US7342029 Jul 22, 2005 Mar 11, 2008 Sanofi-Aventis Deutschland Gmbh Substituted indoles
US7462638 Aug 18, 2003 Dec 9, 2008 Sanofi-Aventis Deutschland Gmbh Use of IκB-kinase inhibitors in pain therapy
US20030119820 Oct 4, 2002 Jun 26, 2003 Aventis Pharma Deutschland Gmbh Substituted indoles
US20040116494 Aug 18, 2003 Jun 17, 2004 Aventis Pharma Deutschland Gmbh Use of IkappaB-kinase inhibitors in pain therapy
US20040209868 May 11, 2004 Oct 21, 2004 Aventis Pharma Deutschland Gmbh Substituted indoles
US20070244139 Jun 6, 2007 Oct 18, 2007 Sanofi-Aventis Deutschland Gmbh Indole Derivatives or Benzimidazole Derivatives for Modulating IkB Kinase
US20090069358 Nov 6, 2008 Mar 12, 2009 Sanofi-Aventis Deutschland Gmbh Use of IKappaB-Kinase Inhibitors in Pain Therapy
JP2003519101A Title not available
WO1998040380A1 Feb 27, 1998 Sep 17, 1998 Alessio Roberto D Indolyl-pyrrolydenemethylpyrrole derivatives and process for their preparation
WO2001030774A1 Oct 17, 2000 May 3, 2001 Aventis Pharma Gmbh Substituted indoles for modulating nfkb activity
WO2003066629A2 Feb 6, 2003 Aug 14, 2003 Michael J Arnost Heteroaryl compounds useful as inhibitors of gsk-3
WO2004022057A1 Aug 5, 2003 Mar 18, 2004 Aventis Pharma Gmbh USE OF IκB KINASE INHIBITORS FOR THE TREATMENT OF PAIN
WO2004022553A1 Aug 5, 2003 Mar 18, 2004 Aventis Pharma Gmbh INDOLE OR BENZIMIDAZOLE DERIVATIVES FOR MODULATING IκB KINASE
WO2004089913A1 Apr 8, 2004 Oct 21, 2004 Novartis Ag Aminopyrimidine derivatives and their medical use
WO2005040133A1 Oct 11, 2004 May 6, 2005 Michael Clare Pyrimidine compounds for the treatment of inflammation
WO2004022553A1 * Aug 5, 2003 Mar 18, 2004 Aventis Pharma Gmbh INDOLE OR BENZIMIDAZOLE DERIVATIVES FOR MODULATING IκB KINASE

Filed under: Phase2 drugs, Uncategorized Tagged: IkB kinase inhibitors, SANOFI

Researchers discover how cancer ‘invisibility cloak’ works

$
0
0

Originally posted on lyranara.me:

Researchers discover how cancer 'invisibility cloak' works

Researchers at National Jewish Health have discovered how a lipid secreted by cancer tumors prevents the immune system from mounting an immune response against it. When lysophosphatidic acid (LPA) binds to killer T cells, it acts almost like an “invisibility cloak,” preventing T cells from recognizing and attacking nascent tumors.

“In recent years, several therapeutic medicines have been developed that spur a person’s own immune system to fight ,” said Raul Torres, PhD, professor of immunology at National Jewish Health, and senior author on the paper, published in the October issue of Cancer Immunology Research. “Our findings suggest new targets and strategies for enlisting the immune system’s help in fighting cancer.”

View original


Filed under: Uncategorized

The US FDA has issued full approval for Israeli drugmaker Teva’s Synribo (omacetaxine mepesuccinate)高三尖杉酯碱 for chronic myeloid leukaemia (CML).

$
0
0


Omacetaxine mepesuccinate 高三尖杉酯碱

Alkaloid from Cephalotaxus harringtonia; FDA approved orphan drug status for Ceflatonin in the treatment of chronic myeloid leukemia due to being an inducer of apoptosis in myeloid cells and inhibitor of angiogenesis.
26833-87-4 CAS NO

1-((1S,3aR,14bS)-2-Methoxy-1,5,6,8,9,14b-hexahydro-4H-cyclopenta(a)(1,3)dioxolo(4,5-h)pyrrolo(2,1-b)(3)benzazepin-1-yl) 4-methyl (2R)-2-hydroxy-2-(4-hydroxy-4-methylpentyl)butanedioate

1-((11bS,12S,14aR)-13-methoxy-2,3,5,6,11b,12-hexahydro-1H-[1,3]dioxolo[4',5':4,5]benzo[1,2-d]cyclopenta[b]pyrrolo[1,2-a]azepin-12-yl) 4-methyl 2-hydroxy-2-(4-hydroxy-4-methylpentyl)succinate

Also known as:  NSC-141633,

  • BRN 5687925
  • Ceflatonin
  • CGX-635
  • Homoharringtonine
  • Myelostat
  • NSC 141633
  • Omacetaxine mepesuccinate
  • Omapro
  • Synribo
  • UNII-6FG8041S5B
  • 高三尖杉酯碱

CGX-635-14 (formulation), CGX-635, HHT, ZJ-C, Myelostat, Ceflatonin

 USFDA on 26th October 2012  APPROVED

US FDA:    link

Formula C29H39NO9 
Mol. mass 545.62 g/mol
Melting Point: 144-146 °C
 FEBRUARY 17, 2014

The US Food and Drug Administration has now issued full approval for Israeli drugmaker Teva’s Synribo (omacetaxine mepesuccinate) for chronic myeloid leukaemia (CML).

Synribo is indicated for adult patients with chronic phase (CP) or accelerated phase (AP) CML with resistance and/or intolerance to two or more tyrosine kinase inhibitors (TKIs).

Read more at: http://www.pharmatimes.com/Article/14-02-17/US_green_light_for_Teva_s_CML_drug_Synribo.aspx#ixzz2tdkbGFcw

Homoharringtonine is an angiogenesis-inhibiting and apoptosis-inducing alkaloid which was approved in October 2012 by the FDA for the treatment of adult patients with chronic or accelerated phase chronic myeloid leukemia (CML) with resistance and/or intolerance to two or more tyrosine kinase inhibitors (TKI). In November 2012, the product was commercialized as Synribo(R) on the U.S. market by Teva.

The original developer, ChemGenex, selected homoharringtonine for the combination trials due to its complementary mechanism of action that can reduce Bcr-Abl protein expression associated with resistance to imatinib mesylate.

In 2004, the compound received orphan drug designation from the EMEA for the treatment of AML and CML. Orphan drug designation was granted by the FDA for the treatment of CML in 2006 and for the treatment of myelodysplasia in 2009. Fast track designation was assigned to homoharringtonine for CML in 2006. In 2009, the product was licensed to Hospira by ChemGenex Pharmaceuticals for development and marketing in Europe, the Middle East and parts of Africa.

Homoharringtonine, AKA HHT or omacetaxine mepesuccinate, is a cephalotaxine ester and protein synthesis inhibitor with established clinical activity as a single agent in hematological malignancies. Homoharringtonine is synthesized from cephalotaxine, which is an extract from the leaves of the plant, Cephalotaxus species. In October 2005, homoharringtonine received Orphan Drug designation from the EMEA for the treatment of chronic myeloid leukemia (CML). Then in March 2006, homoharringtonine received Orphan Drug status from the FDA for the treatment of CML. In November 2006, homoharringtonine, for the treatment of CML, was granted Fast Track designation by the FDA. Most recently, in October 2012, homoharringtonine was marketed under the brand name Synribo” and FDA approved for patients who are intolerant and/or resistant to two or more tyrosine kinase inhibitors used to treat accelerated or chronic phase CML

Omacetaxine mepesuccinate is administered subcutaneously and acts differently from TKIs. It may have a therapeutic advantage for patients who have failed TKIs. Omacetaxine is currently in global phase 2/3 clinical trials for CML and has been granted Orphan Drug designations by the U.S. Food and Drug Administration (FDA) and European Medicines Agency (EMEA) as well as Fast Track status by the FDA. In vitro and animal model trails are promising and recent results showed that omacetaxine has potential to treat resistant leukemia mainly CML and ALL.

 PATENT
3-25-2011
CEPHALOTAXUS ESTERS, METHODS OF SYNTHESIS, AND USES THEREOF

Tetrahedron Letters,Vo1.23,No.34,pp 3431-3434  - Brock University

Omacetaxine mepesuccinate

Omacetaxine mepesuccinate (INN, trade name Synribo) is a semi-synthetic analogue of an alkaloid from Cephalotaxus harringtonia that is indicated for treatment of chronic myelogenous leukemia (CML). It was approved by the US FDA in October 2012 for the treatment of adult patients with CML with resistance and/or intolerance to two or more tyrosine kinase inhibitors (TKIs).[1]

Omacetaxine mepesuccinate is a semisynthetic derivative of the cytotoxic plant alkaloid homoharringtonine isolated from the evergreen tree Cephalotaxus with potential antineoplastic activity. Omacetaxine mepesuccinate binds to the 80S ribosome in eukaryotic cells and inhibits protein synthesis by interfering with chain elongation. This agent also induces differentiation and apoptosis in some cancer cell types. Omacetaxine mepesuccinate (INN, or homoharringtonine, trade name Synribo) is an alkaloid from Cephalotaxus harringtonia that is indicated for treatment of Chronic Myelogenous Leukemia. It was approved by the USFDA on 26th October 2012 for the treatment of adult patients with chronic myeloid leukemia (CML) with resistance and/or intolerance to two or more tyrosine kinase inhibitors (TKIs)

Omacetaxine is indicated for use as a treatment for patients with chronic myeloid leukaemia who are intolerant of tyrosine kinase inhibitors.[2][3]

In June 2009, results of a long-term open label Phase II study were published, which investigated the use of omacetaxine infusions in CML patients. After twelve months of treatment, about one third of patients showed a cytogenetic response.[4] A study in patients who had failed imatinib and who had the drug resistant T315I mutation achieved cytogenetic response in 28% of patients and haematological response in 80% of patients, according to preliminary data.[5]

Phase I studies including a small number of patients have shown benefit in treating myelodysplastic syndrome (MDS, 25 patients)[6] and acute myelogenous leukaemia (AML, 76 patients).[7] Patients with solid tumors did not benefit from omacetaxine.[8]

Omacetaxine is a protein translation inhibitor. It inhibits protein translation by preventing the initial elongation step of protein synthesis. It interacts with the ribosomal A-site and prevents the correct positioning of amino acid side chains of incoming aminoacyl-tRNAs. Omacetaxine acts only on the initial step of protein translation and does not inhibit protein synthesis from mRNAs that have already commenced translation.[9]

Omacetaxine mepesuccinate

SYNRIBO contains the active ingredient omacetaxine mepesuccinate, a cephalotaxine ester. It is a protein synthesis inhibitor. Omacetaxine mepesuccinate is prepared by a semi-synthetic process from cephalotaxine, an extract from the leaves of Cephalotaxus sp. The chemical name of omacetaxine mepesuccinate is cephalotaxine, 4-methyl (2R)-hydroxyl-2-(4-hydroxyl-4-methylpentyl) butanedioate (ester).

Omacetaxine mepesuccinate has the following chemical structure:

SYNRIBO™ (omacetaxine mepesuccinate)  Structural Formula Illustration

The molecular formula is C29H39NO9 with a molecular weight of 545.6 g/mol. SYNRIBO for injection is a sterile, preservative-free, white to off-white, lyophilized powder in a single-use vial. Each vial contains 3.5 mg omacetaxine mepesuccinate and mannitol.

SYNRIBO is intended for subcutaneous administration after reconstitution with 1.0 mL of 0.9% Sodium Chloride Injection, USP. The pH of the reconstituted solution is between 5.5 and 7.0.

…………………………………..

INTRODUCTION

Harringtonines 3 are particular cephalotaxanes formed by attachement of a branched hydroxyacyloxy side-chain at the 3-position of various cephalotaxines moieties. Harringtoriines are natural esters of cephalotaxines exhibiting generally a strong cytotoxic activity. However the lost only one atom of this minimal structure lead to a dramatic lost of activity (see below). Some example of harringtonines are harringtonine

3a, homoharringtonine 3b, drupangtonine 3c, anhydroharringtonine 3d and neoharringtonine 3e.

SCHEME 1 DEFINITION NOMENCLATURE AND NUMBERING OF CEPHALOTAXANES

Figure imgf000003_0001
Figure imgf000003_0002

Examples of harringtonines

Figure imgf000003_0003

Examples of cephalotaxines

Figure imgf000003_0004

Harringtonine 3a (n = 2) Anhydroharringtonine 3d Homoharringtonine 3b (n = 3)

Figure imgf000003_0006

(-)-Cephalotaxine 2a

Figure imgf000003_0008
Figure imgf000003_0007

Drupacine 2b Drupangtonine 3c Neoharringtonine 3e (n = 2)

…………………………………

The term “cephalotaxanes” refers to compounds or salts thereof which have a basic skeleton of formula

Figure US06831180-20041214-C00001

where p is equal to 1 or 2 (it being possible for the two units to be identical or different and linked via a single bond or an oxygen atom), which can contain various oxygenated substituents (aliphatic or aromatic ethers, free or esterified alcohols, substituted or free enols and/or phenols, bridged ethers, and more generally any substituent usually encountered in the natural state on compounds of this type).

Harringtonines are alkaloids which are of high interest in anticancer chemotherapy, in particular on certain haematosarcomas which are multi-resistant to the existing therapies. The selectivity of harringtonines, which is based on a novel mechanism of action relating to protein synthesis, is such that this series is favoured with a great future in anticancer therapy.

Several literature compilations give a seemingly exhaustive review of all of the knowledge relating to cephalotaxanes, these compilations being, chronologically: [C. R. Smith, Jr, R. G. Powell and K. L. Mikolajczack, Cancer Treat. Rep., Vol. 60, 1157 (1976); C. R. Smith, Jr, L. Kenneth, K. L. Mikolajczack and R. G. Powell in “Anticancer Agent Based on Natural Product Model”, 391 (1980); Liang Huang and Zhi Xue in “The Alkaloids”, Vol. XXIII (A. Brossi Ed.), 157 (1984); M. Suffness and G. A. Cordell in “The Alkaloids, Chemistry and Pharmacology” (A. Brossi Ed.), Vol. 25, 57-69, 295-298 (1'987); P. J. O'Dwyer, S. A. King, D. F. Hoth, M. Suffness and B. Leyland-Jones, Journal of Clinical Oncology, 1563 (1986); T. Hudlicky, L. D. Kwart and J. W. Reed, in “Alkaloid: Chemical and Biological Perspectives” (S. W. Pelletier Ed.), Vol. 5, 639 (1987); M. A. Miah, T. Hudlicky and J. Reed in “The Alkaloids”, Vol. 51, 199 (1998)].

Antiparasitic activities, in particular on the haematozoon of malaria, have also been recognized [J. M. Whaun and N. D. Brown, Ann Trop. Med. Par., Vol. 84, 229 (1990)].

Homo-harringtonine (HHT), the most active member of the series, is active at and above daily doses of 2.5 mg/mof body area per 24 hours, i.e., as a guide, at doses twenty times lower than that for Taxol. HHT has already undergone fourteen phase I and II clinical trials and it is the only known product capable of a 70% reinduction of full haematological remissions in patients suffering from chronic myeloid leukaemias that have become resistant to alpha-interferon [S. O'Brien, H. Kantarjian, M. Keating, M. Beran, C. Koler, L. E. Robertson, J. Hester, M. Rios, M. Andreeff and M. Talpaz, Blood, 332 (1995); Leukemia Insights, Vol. 3, No. 1 (1998)].

Harringtonines were extracted over 35 years ago from an exclusively Asiatic cephalotaxacea known as Cephalotaxus harringtonia, following the programme of research into novel anticancer agents in the plant kingdom developed by the National Cancer Institute. In fact, the Cephalotaxus alkaloids consist essentially (at least 50%) of cephalotaxine, a biosynthetic precursor of the harringtonines, the latter individually representing only a few percent of the total alkaloids.

Besides their low concentration in the natural state in plant starting material, harringtonines are mixed with many congeners which have very similar chemical structures. Thus, in a high resolution high performance liquid chromatography (HPLC) chromatogram of a semi-purified alkaloid extract, no less than several tens of cephalotaxine esters are counted.

Numerous antileukemia drugs have been investigated but so far, there is no single drug that is effective and safe. As discussed in U.S. 3,497,593, an alkaloid from Tylophora plant is said to have antitumor activity against mouse leukemia (L-1210). U.S. 3,928,584 discloses an organic composition derived from tree sap and is said to have activity against mouse leukemia P-388. Also U.S. 4,431,639 discloses that an extract of Rhisoma Stractylis promotes the production of lymphocytes in the circulating blood, consequently eliminating cancer growth

  • Harringtonine or Homoharringtonine, hereinafter referred to as HH, has been known to be effective against acute chronic granulocytic and monocytic leukemia (Journal of Chinese Internal Medicine 3:162-164, 1978). However, it is highly toxic and causes damage to heart and hematopoietic organs. The results of experiments in animals, such as mice, rabbits and dogs, indicate that most of them die from cardiotoxicity after receiving the drug. Therefore, there is a need to improve the HH drug for safe use against leukemia. This drug is of special importance in that all known antileukemia drugs are effective against lymphatic leukemia and there are no effective drugs for treating nonlymphatic leukemia

All the literature from 1972 to the present date [Mikolajczack et al., Tetrahedron, 1995 (1972); T. Hudlicky, L. D. Kwart and J. W. Reed in “Alkaloid: Chemical and Biological Perspectives” (S. W. Pelletier Ed.), Vol. 5, 639 (1987); M. A. Miah, T. Hudlicky and J. Reed in “The Alkaloids”, Vol. 51, p. 236 (1998)] mention the impossibility hitherto of esterifying the highly sterically hindered secondary hydroxyl of cephalotaxane 2a with the tertiary carboxyl of the alkanoyl chain of harringtonic acid 3 totally preformed to give a harringtonine 4b, i.e. the conversion 2a+3e(4b as described in the example featured in the scheme below

Figure US06831180-20041214-C00002
  • ……………………………………………………..

SYNTHESIS

Tetrahedron Lett 1982,23(34),3431,  J Org Chem 1983,48(26),5321

The oxidation of 2-methyl-1-cyclopentene-1-carbaldehyde (I) with O3 and Ag2O gives 2,6-dioxoheptanoic acid (II), which is esterified with cephalotaxine (III) by means of (COCl)2, yielding the ester (IV). Reformatsky reaction of (IV) with methyl bromoacetate (V) and Zn affords the adduct (VI), which is treated with an excess of methylmagnesium iodide to provide the target homoharringtonine (as a single diastereomer), along with some starting cephalotaxine that is separated by chromatography.

………………………………

SYNTHESIS

EP 1064285; FR 2776292; WO 9948894, Tetrahedron Lett 1999,402931

The intermediate (racemic)-2-(methoxycarbonylmethyl)-6,6-dimethyltetrahydropyran-2-carboxylic acid (VIII) has been obtained by several related methods: 1. The Grignard condensation of 4-methyl-3-pentenyl bromide (I) with diethyl oxalate (II) in HF gives the 2-oxoheptenoate (III), which is condensed with methyl acetate (IV) by means of LiHMDS in THF to yield 3-(ethoxycarbonyl)-3-hydroxy-7-methyl-6-octenoic acid methyl ester (V).

The cyclization of (V) by means of Ts-OH in hot toluene or by means of hot aqueous formic acid affords 2-(methoxycarbonylmethyl)-6,6-dimethyltetrahydropyran-2-carboxylic acid ethyl ester (VI), which is hydrolyzed with KOH in boiling water to provide the corresponding dicarboxylic acid (VII). Finally, this compound is regioselectively monoesterified by means of BF3/MeOH in methanol to furnish the intermediate (racemic)-2-(methoxycarbonylmethyl)-6,6-dimethyltetrahydropyran-2-carboxylic acid (VIII). 2.

The reaction of 3-(ethoxycarbonyl)-3-hydroxy-7-methyl-6-octenoic acid methyl ester (V) with HCl in hot methanol gives 3-(ethoxycarbonyl)-3,7-dihydroxy-7-methyloctanoic acid methyl ester (IX), which is then cyclized by means of ZnCl2 in hot dichloroethane to yield the previously described intermediate (VIII). 3. The hydrolysis of 3-(ethoxycarbonyl)-3-hydroxy-7-methyl-6-octenoic acid methyl ester (V) with KOH in refluxing methanol/water gives the corresponding diacid (X), which is regioselectively monoesterified by means of BF3/MeOH in methanol to yield 3-carboxy-3-hydroxy-7-methyl-6-octenoic acid methyl ester (XI).

Finally, this compound is cyclized by means of Ts-OH in hot toluene to afford the previously described carboxylic intermediate (VIII). The racemic acid (VIII) is submitted to optical resolution by esterification with quinine (XII) by means of 2,4,6-trichlorobenzoyl chloride and TEA or DCC to give a diastereomeric mixture of esters (XIII) that is separated by preparative HPLC to obtain the desired diastereomer (XIV).

The hydrolysis of (XIV) with KOH in refluxing ethanol/water gives the corresponding chiral dicarboxylic acid (XV), which is regioselectively monoesterified with BF3/MeOH in methanol to yield the chiral (R)-2-(methoxycarbonylmethyl)-6,6-dimethyltetrahydropyran-2-carboxylic acid (XVI).

The esterification of (XVI) with cephalotaxine (XVII) by means of 2,4,6-trichlorobenzoyl chloride and TEA in toluene affords the corresponding ester (XVIII), which is treated with HBr in dichloromethane/HOAc, providing the bromoester (XIX). Finally, this compound is treated with NaHCO3, CaCO3 or BaCO3 in acetone/water to give the target hydroxyester.

………………………………………….

EXTRACTION

EP0203386B1

  • Throughout the specification, the concentration of the solvent is the same as first given unless stated otherwise. Redeuced pressure means about 2,27 kPa (17 mm Hg. abs), l is liter, kg is kilogram. ml is milliliter. Yield in weight %.
    Example 1. HH is extracted from the skins, stems, leaves and seeds of Cephalotaxus fortunel Hook and other related species, such as Cephalotaxus sinensis Li, C. hainanensis, and C. wilsoniana, including C.oliveri mast and C.harringtonia.
  • 1 kg of finely ground Cephalotaxus fortunel Hook is extracted with 8 l of 90% ethanol at room temperature for 24 hrs. The solution is filtered to yield a filtrate A and filtercake. The filtercake is percolated with ethanol and filtered again to yield filtrate B. A and B are combined and distilled under reduced pressure to recover ethanol and an aqueous residue. To this residue, 2% HCl is added to adjust the pH to 2.5. The solids are separated from the solution by filtration to yield a filtrate C. The solids are washed once with 2% HCl and filtered to yield a filtrate D. C and D are combined and the pH adjusted to 9.5 by adding saturated sodium carbonate solution. The alkaline filtrate is extracted with chloroform and the chloroform layer separated from the aqueous layer. This extration process is repeated five times. All the chloroform extracts are combined and distilled at reduced pressure to recover chloroform and alkaloid as a solid residue respectively.
  • The solid alkaloid is then dissolved in 20 ml. of 6% citric acid in water. The solution is divided into three equal portions. These are adjusted to pH 7,8 and 9 by adding saturated sodium carbonate solution.
  • The portions having pH 8 and 9 are combined and extracted with chloroform. The chloroform extracts are distilled under reduced pressure, whereby chloroform is removed and recovered and a solid residue of crude Harringtonine is obtained.
  • The crude Harringtonine is dissolved in pure ethanol i.e. alkaloid : anhydrous ethanol 1:10 , and crystallized. The crystals are refined by recrystalliation in diethyl ether. Overall yield of Harringtonine is about 0.1% including yield from mixed HH from the subsequent process.
    Harringtonine has the following chemical structure:
    Figure imgb0001

    wherein R is

    Figure imgb0002
    melting point:
    135° – 137°C
    crystal:
    colorless
    infrared spectrum:
    3750, 1660, 1505, 1490, 1050, and 945 cm⁻¹.
    Figure imgb0003
  • The portion having a pH of 7 and the mother liquors from the foregoing crystallization of Harringtonine are combined and passed through a liquid chromatographic column of diameter to height ratio 1:50 packed with alumina. The column is finally flushed with chloroform and followed by chloroform-methanol of 9:1 mixture. The resulting alkaloids are mixture of HH. The mixed HH is then separated from each other by countercurrent distribution employing chloroform and pH 5 buffer. The first fraction of the countercurrent distribution is Homoharringtonine and the last fraction of the countercurrent distribution is Harringtonine. Homoharringtonine is purified by crystallization in methyl alcohol.
    Homoharringtonine has the following chemical structure:
    Figure imgb0004

    wherein R is

    Figure imgb0005
    yield:
    0.02%
    melting point:
    144° – 146°C
    infrared spectrum:
    3500∼3400, 1750, 1665, 1030 and 940 cm⁻¹.
    Figure imgb0006

…………………………………………………………………………..

EXTRACTION

EP1064285B1

All the literature from 1972 to the present date [Mikolajczack et al.,Tetrahedron, 1995 (1972); T. Hudlicky, L.D. Kwart and J.W. Reed in "Alkaloid: Chemical and Biological Perspectives" (S.W. Pelletier Ed.), Vol. 5, 639 (1987); M.A. Miah, T. Hudlicky and J. Reed in "The Alkaloids", Vol. 51, p. 236 (1998)] mention the impossibility hitherto of esterifying the highly sterically hindered secondary hydroxyl of cephalotaxine 2a with the tertiary carboxyl of the alkanoyl chain of harringtonic acid 3e totally preformed to give a harringtonine 4b , i.e. the conversion 2a 3e ( 4b as described in the example featured in the scheme below

Figure 00080001

Example 46

Preparation of purified (-) cephalotaxine from total alkaloidic extract of Cephalotaxus sp

    • [0319]
      Figure 01280001
    • Partially racemized cephalotaxine [H. Wenkui; L. Yulin; P. Xinfu, Scientia Sinica,; 23; 7; 835 (1980)]
    • 1H NMR of two batches of cephalotaxine (extracted in the same conditions as above) with the optically active NMR shift reagent europium(III) tris[3-(heptafluoropropylhydroxymethylene)-(+)-camphorate (1 éq) showed the following results:

      • Batch A: 1H NMR 400 MHz (CDCl3)(δ ppm): 6.06 (1H, OCH2O (+)-cephalotaxine) and 5.82 (1H, OCH2O (+)-cephalotaxine) ; 5.99 (1H, OCH2O (-)-cephalotaxine) and 5.76 (1H, OCH2O (-)-cephalotaxine).
        Presence of 11 ± 5 % de (+)-cephalotaxine.
        [α]22 = -134,0° (c = 0,214; CHCl3) : calculated rate 25 ± 5 %
      • Batch B: slightly racemized (1%)
        [α]19 = -173,3° (c = 0,208; CHCl3)

Enantiomeric enrichment of the natural cephalotaxine:

    • Crude chromatographied cephalotaxine (20g) was dissolved at 55°C in dry methanol (100 ml). Crystallization occurs by cooling with rotary evaporator and after filtration the product thus obtained showed 99.9% of HPLC purity.
      [α]20 D =-130° (C1, CHD3) corresponding to 10 % of racemization. The crystallized product thus obtained (20g) was dissolved again in hot methanol (100 ml).
      Slowly cooling the solution allows translucent prisms composed of pure enantiomeric (-)-cephalotaxine [α]20 D= -185°(C1,CHCl3).
      After filtration, the mother liquors was allowed to slowly evaporate at room temperature and crystals in the form of macled needles exclusively composed of racemic cephalotaxine [α]D 20 = 0,5° (C1 ; CHCl3) were obtained.
      After filtration, the second mother liquors allowed prisms composed of (-)-cephalotaxine identical to this obtained at the first crystallization.
      After filtration, the third mother liquors still allowed macled needles (urchins) composed of (±)-cephalotaxine.
      The cycle is repeated three times. The combined prismatic crystals was recrystallized once to give enantiomerically pure (-)-cephalotaxine, while the combined macled needles treated in the same way gives 100% racemic cephalotaxine.

Chemical evaluation of the enantiomeric purity of natural cephalotaxine:

  • A sample of partially racemized natural cephalotaxine was inserted in the process, which sequence is described in the Examples 1,2,3,4,5,6,15,19 and 21, by using a pure (2R)-homoharrintonic acid resulting from Example 19.
    The HPLC analysis of the diastereomeric mixture of anhydro-homoharrintonine thus obtained showed a significant enantio-epi-homoharringtonine rate (11% ± 3%) corresponding to the (+)-cephalotaxine content in the racemic mixture of origin (it has been demonstrated that the two antipodes of the homoharringtonic acid react in a stoechiometric way comparable to the pure enantiomeric cephalotaxine).

Example 47Preparation of homoharringtonine, from anhydro-homoharringtonine:

    • Figure 01300001

1)° Method A

    • A commercial solution of hydrobromic acid in acetic acid (17.4 ml, 86.6 mmol, HBr 30% w/w) was added to a stirred solution of anhydrohomoharringtonine resulting from Example 21 (50.8 g, 9.63 mmol) in anhydrous dichloromethane (25.6 ml) at -10°C. After stirring at -10°C for 3 hours was added water (240 ml) and the reaction mixture was become viscous. The temperature was allowed to rise to room temperature and after stirring for 2.5 hours was added sodium carbonate 0.76M (406 ml) to pH 8. The resulting aqueous layer was saturated with sodium chloride, then was extracted with dichloromethane (3 × 230 ml) and the combined organic layers were dried over magnesium sulfate and evaporated to dryness to afford a foam. After phase reverse chromatography below-mentioned were obtained 4.03g of homoharringtonine (77%). The product thus obtained showed identical characteristics to this resulting from Example 25.

2°) Method B

  • To a stirred solution of anhydrohomoharringtonine resulting from Example 21 (214 mg, 0.406 mmol) in anhydrous dichloromethane (1.1 ml) was added at -10°C a commercial solution of hydrobromic acid in acetic acid (0.728 ml, 3.6 mmol, HBr 30% w/w). After stirring at -10°C for 3 hours, was added water (13 ml) and then the temperature was raised to 20°C. After stirring at 20°C for 3 hours, was added a sodium carbonate solution (0.76M; 31.5 ml) up to pH 8. The resulting aqueous layer, after saturation with sodium chloride, was extracted with dichloromethane (3 × 20 ml) and the combined organic layers were dried over magnesium sulfate and evaporated to dryness. The resulting crude product was purified by phase reverse chromatography below-mentioned to provide homoharringtonine (166 mg, 75%). The product thus obtained showed identical characteristics to this resulting from Example 25.

    Figure 01320001
    Figure 01330001

……………………

SEMISYNTHESIS

US6831180

EXAMPLE 27 Preparation of homoharringtonine as a pharmaceutical use from crude semi-synthetic homoharringtonine resulting from example 25 by preparative high-performance liquid chromatography

Figure US06831180-20041214-C00126

1°) Method A

Crude homoharringtonine (35 g) is dissolved in buffer (triethylamine (1.55/1000) in deionised water and orthophosphoric acid to adjust pH to 3. The solution was filtered then injected on a preparative high-performance liquid chromatograph equipped with axial compression and high pressure pump (stationary phase: n-octadecylsilane, 15 μm, porosity 100, 1 kg; mobile phase; buffer/tetrahydrofurane 85/15). Elution was performed at a flow rate of 0.2 l/min. Fractions contain was monitored by U.V. detector and TLC. Retained fraction were finally checked by HPLC then combined, alkalinised with 2.5% aqueous ammonia and extracted with dichloromethane (4×400 ml). After concentration under reduced pressure homoharringtonine is obtained as a pale yellow resin which on trituration in a 8/2 water-methanol mixture gave pure homoharringtonine as a white crystalline solid (mp=127° C.), HPLC purity was higher than 99.8%.

2°) Method B

Same procedure of purification as method A was performed but mobile phase buffer/methanol (68/32) was used instead buffer/tetrahydrofurane.

3°) Method C

Same procedure of purification as method A was performed but mobile phase buffer/acetonitrile (85/15) was used instead buffer/tetrahydrofurane.

EXAMPLE 28 Preparation of homoharringtonine as a pharmaceutical use from semi-purified natural cephalotaxine

Crude homoharringtonine, prepared according to Example 25 from a partially racemized natural cephalotaxine and purified by chromatography and crystallisation according to the method A of Example 27, gave an homoharringtonine showing a non natural enantiomeric epi-homoharringtonine content less than 0.05%.

EXAMPLE 46 Preparation of purified (−) cephalotaxine from total alkaloidic extract of cephatotaxus sp

Figure US06831180-20041214-C00145

Partially racemized cephalotaxine [H. Wenkui; L. Yulin; P. Xinfu, Scientia Sinica; 23; 7; 835 (1980)]

1H NMR of two batches of cephalotaxine (extracted in the same conditions as above) with the optically active NMR shift reagent europium(III) tris[3-(heptafluoropropylhydroxymethylene)-(+)-camphorate (1éq) showed the following results:

Batch A: 1H NMR 400 MHz (CDCl3)(δ ppm): 6.06 (1H, OCH2O (+)-cephalotaxine) and 5.82 (1H, OCH2O (+)-cephalotaxine); 5.99 (1H, OCH2O (−)-cephalotaxine) and 5.76 (1H, OCH2O (−)-cephalotaxine). Presence of 11±5% de (+)-cephalotaxine. [α]22=−134,0°(c=0,214; CHCl3): calculated rate 25±5%

Batch B: slightly racemized (1%) [α]19=−173,3°(c=0,208; CHCl3)

Enantiomeric Enrichment of the Natural Cephalotaxine:

Crude chromatographied cephalotaxine (20 g) was dissolved at 55° C. in dry methanol (100 ml). Crystallization occurs by cooling with rotary evaporator and after filtration the product thus obtained showed 99.9% of HPLC purity, [α]20 D=−130°(C1, CHD3) corresponding to 10% of racemization. The crystallized product thus obtained (20 g) was dissolyed again in hot methanol (100 ml).

Slowly cooling the solution allows translucent prisms composed of pure enantiomeric (-−)-cephalotaxine [α]20 D=−185°(C1, CHCl3).

After filtration, the mother liquors was allowed to slowly evaporate at room temperature and crystals in the form of macled needles exclusively composed of racemic cephalotaxine [α]D 20=0,5°(C1; CHCl3) were obtained.

After filtration, the second mother liquors allowed prisms composed of (−)-cephalotaxine identical to this obtained at the first crystallization.

After filtration, the third mother liquors still allowed macled needles (urchins) composed of (±)-cephalotaxine.

The cycle is repeated three times. The combined prismatic crystals was recrystallized once to give enantiomerically pure (−)-cephalotaxine, while the combined macled needles treated in the same way gives 100% racemic cephalotaxine.

Chemical Evaluation of the Enantiomeric Purity of Natural Cephalotaxine:

A sample of partially racemized natural cephalotaxine was inserted in the process, which sequence is described in the Examples 1,2,3,4,5,6,15,19 and 21, by using a pure (2R)-homoharrintonic acid resulting from Example 19. The HPLC analysis of the diastereomeric mixture of anhydro-homoharrintonine thus obtained showed a significant enantio-epi-homoharringtonine rate (11%±3%) corresponding to the (+)-cephalotaxine content in the racemic mixture of origin (it has been demonstrated that the two antipodes of the homoharringtonic acid react in a stoechiometric way comparable to the pure enantiomeric cephalotaxine).

EXAMPLE 47

Preparation of homoharringtonine, from anhydro-homoharringtonine

Figure US06831180-20041214-C00146

1°) Method A

A commercial solution of hydrobromic acid in acetic acid (17.4 ml, 86.6 mmol, HBr 30% w/w) was added to a stirred solution of anhydrohomoharringtonine resulting from Example 21 (50.8 g, 9.63 mmol) in anhydrous dichloromethane (25.6 ml) at −10° C. After stirring at −10° C. for 3 hours was added water (240 ml) and the reaction mixture was become viscous. The temperature was allowed to rise to room temperature and after stirring for 2.5 hours was added sodium carbonate 0.76M (406 ml) to pH 8. The resulting aqueous layer was saturated with sodium chloride, then was extracted with dichloromethane (3×230 ml) and the combined organic layers were dried over magnesium sulfate and evaporated to dryness to afford a foam. After phase reverse chromatography below-mentioned were obtained 4.03 g of homoharringtonine (77%). The product thus obtained showed identical characteristics to this resulting from Example 25.

2°) Method B

To a stirred solution of anhydrohomoharringtonine resulting from Example 21 (21.4 mg, 0.406 mmol) in anhydrous dichloromethane (1.1 ml) was added at −10° C. a commercial solution of hydrobromic acid in acetic acid (0.728 ml, 3.6 mmol, HBr 30% w/w). After stirring at −10° C. for 3 hours, was added water (13 ml) and then the temperature was raised to 20° C. After stirring at 20° C. for 3 hours, was added a sodium carbonate solution (0.76M; 31.5 ml) up to pH 8. The resulting aqueous layer, after saturation with sodium chloride, was extracted with dichloromethane (3×20 ml) and the combined organic layers were dried over magnesium sulfate and evaporated to dryness. The resulting crude product was purified by phase reverse chromatography below-mentioned to provide homoharringtonine (166 mg, 75%). The product thus obtained showed identical characteristics to this resulting from Example 25.

…………………………………

EXTRACTION

US20100240887

The remarkable clinical efficacy of Homoharringtonine (HHT) resulting in lot of observations of complete remission of leukemia and other solid cancer in human being since 1988. Recently, research articles reported that the HHT efficacy in glaucoma, inhibition of Hepatities B virus replication and using in bone marrow transplantation. For example, the University of Texas M.D. Anderson Cancer Center and National Cancer Institute reported that “Ninety-two percent of patients achieved CHR with HHT.” [Susan O'Brien, at al.; Sequential homoharringtonine and interferon-α in the treatment of early chronic phase chronic myelogenous leukemia; Blood, Vol 93, No 12 (June 15), 1999: pp 4149-4153]. Another article reported that “the median number of days on HHT per month was 2 days with a median follow-up of 26 months; the estimated 2-year survival rate was 90%.” (Susan O’Brien, at al.; Simultaneous homoharringtonine and interferon-α in the treatment of patients with chronic-phase chronic myelogenous leukemia; American Cancer Society; Apr. 1, 2002, Vol 94, No. 7).

On Nov. 8, 1988, U.S. Pat. No. 4,783,454 titled Process for producing harringtonine and homoharringtonine disclosed the technique of isolation of a purified HHT from bark of Cephalotaxus. However, the natural source ofCephalotaxus is very limited. Trees of Cephalotaxus grow slowly. Bark ofCephalotaxus has very low content of HHT. Extracting HHT from bark ofCephalotaxus the yield was about 0.02% only. More important to harvest bark ofCephalotaxus will kill and destroy trees. Supply of HHT is very short now. Therefore, it is necessary to find a new manufacturing method.

DETAILED DESCRIPTION

Great progress has been made in research on Homoharringtonine (HHT) production and on future generation HHT drug since 1988. For example, the University of Texas M.D. Anderson Cancer Center and National Cancer Institute reported that “Ninety-two percent of patients achieved CHR with HHT.” Another article reported that “the median number of days on HHT per month was 2 days with a median follow-up of 26 months; the estimated 2-year survival rate was 90%.”

The good clinical results of HHT in treating cancer brought to the major problem, which is the supply of HHT both short term and long term. It is apparent that a huge amount of bark of Cephalotaxus is needed for collection, extraction and purification of HHT. It is clear that due to the slow growth of the trees ofCephalotaxus, which is a nature source of HHT, and the killing of trees by harvesting bark is not a sustainable resource for HHT production.

Present invention disclosed new methods for producing HHT. The new methods of producing HHT are shown as follows.

1. Tissue Culture (Plant Cell Culture):

Culture manipulation to promote secretion of HHT is a new way for an extracellular product HHT. The biosynthetic methods can yield more HHT through precursor of HHT feeding. The production of HHT increased significantly after the addition of the precursors and special biochemical agents. Content of precursor of HHT abounds in tree and it is very cheap. The present methods include several significant developments in technique of culture plant tissues that are

    • (a) yields of HHT selected from rapid growth, resistance to infections organisms; and
    • (b) HHT can excrete into media.

Traditional method of plant culture is very difficult to overcome the problem of high cost. Therefore, traditional method appears too long to have commercial value. HHT is secondary metabolite of Cephalotaxus. Secondary compound acts in defense against the harmful effects of toxins, carcinogens or mutagens found in the plant. In fact, traditional method is very difficult to increase HHT contenting in plant tissues. The present new method uses a special biochemical agent for increasing content of HHT and more easily to purify HHT from other metabolites.

More important is that the key of the present new technique for producing high content of HHT in plant cell culture is to increase production of HHT by directed fermentation through precursor of HHT feeding. The present new methods are used special metabolite of Cephalotaxus for markedly enhance production of HHT. Therefore, the present invention disclosed a new source for the long term of producing HHT.

2. Using Precursor of HHT:

Recent research’s results have established that direct production of HHT from its precursor and advances in biosynthetic understanding for HHT metabolism. Biosynthesis or semisynthesis of HHT from major nonactivity ingredients is well established through great advances in special biochemistry reactions. Using precursor of HHT for semisynthesis and increase of production in plant cell culture are new developing methods for producing HHT.

3. Using Leaves:

Our new method use leaves of tree of Cephalotaxus not use the bark. So far, the extraction of HHT is used bark. The leaves are harvested from the trees ofCephalotaxus, which grow in mountains of South China. The natural source of leaves is very abundance. The new methods do not use bark. Therefore, it can avoid destroy trees. The natural source of Cephalotaxus tree is very limited and slow growing. In fact, bark of Cephalotaxus has very low yield of HHT. The yield of HHT from Cephalotaxus bark is about 50-100 mg/kg of dried bark. The present new method, therefore, has a great economic and environmental value.

4. Semisynthesis:

HHT has received important chemical studies particularly in regard to structure and anticancer activity relationship and semisynthesis.

A great progress in biochemistry allows semisynthesis to use precursor of HHT from leaves of Cephalotaxus and to produce HHT. The total chemical synthesis of HHT appears too long to have commercial value too. Semisynthesis method can yield a high efficient conversion of precursor to HHT. It is other better biological source for manufacturing HHT. This new method uses closing chemical analogues to convert to HHT. This analogue is produced from leaves or other organ of Cephalotaxus. The present invention disclosed that new methods and techniques of manufacturing HHT could avoid chopping down Cephalotaxus trees which governmental environmentalists are trying to have declared a threatened species.

5. Using Taxol Residual

The anticancer drug Taxol is the most promising new chemotherapeutic agents that developed for cancer treatment in the past twenty years. Taxol has a unique mechanism of action. It has been shown to promote tubulin polymerization and stabilize microtubules against depolymerization. The FDA approved the clinical use of Taxol for several types of cancer. So far, annual sales of Taxol are more than $2 billion in market. Taxol is extracted from bark or leaves of an evergreen tree named Taxus species including Taxus brevifolia (or called Pacific yew). After Taxol has been extracted from bark or leaves, all residual materials of Taxus brecifolia named Taxus residual, which are waste.

Both taxol and HHT can be extracted from yew tree. The content of taxol is less than 0.01% in yew tree. The content of HHT in yew tree is about 0.01% -0.22%. The content of HHT is much higher than content of Taxol. Taxol extracted from bark of yew is difficult and expensive. One reason is that the presences of closely related congeners are similar to Taxol. A major congener is Cephalomannine (CPM), which is a waster of process in manufacturing of Taxol.

The chemical and physical characters are very close between Taxol and Cephalomannine (CPM).

CPM characterized by the same ring structure as Taxol and distinguishes from them only in C-13 ester structure. The present invention disclosed that CPM and related derivative are used to produce HHT.

The following specific examples will provide detailed illustrations of methods of producing relative drugs, according to the present invention and pharmaceutical dosage units containing demonstrates its effectiveness in treatment of cancer cells. These examples are not intended, however, to limit or restrict the scope of the invention in any way, and should not be construed as providing conditions, parameters, reagents, or

EXAMPLE 1

Production of HHT by Culture Cells

So far, HHT is extracted from bark and skins of Cephalotaxus species. However, growth of Cephalotaxus species is very slow and concentration of HHT in plant is extremely low. Furthermore, it is difficult to harvest the plants because of their low propagation rate and the danger of drastic reduced in plant availability. Also, cost of total chemical synthesis of HHT is very expensive and is not available for commerce now. For the reasons given above it is more difficult to obtainCephalotaxus on a large scale for long time. Therefore, Cephalotaxus cell cultures are one of best methods for obtaining HHT. In this present invention, special elicitation is disclosed and it will significantly increase production of HHT.

The methods of cell and tissue culture are disclosed as below.

Parts of bark, stems, leaves, or roots of Cephalotaxus species were surface disinfected by treatment in 70% ethanol for 10 minutes and followed by 0.1 HgCl2for 3 minutes. Plant materials were washed five times for 10 minutes each by sterilized water. Parts of plant were cut into small pieces (0.5-1 mm) and put pieces to Murashige and Skoog’s (MS) medium and supplemented with derivative of new active ingredient of phylum mycota (IPM), precursor of HHT which is a derivative of Cephalotaxus (CEP), tyrosine (TYR) naphthaleneacetic acid (NAA), Kinetin (3 mg/L), and 3% sucrose (w/v). PH of medium was adjusted to 5.7˜5.8. Agar (10 g/L) added to medium. Callus tissues are collected from agar media and suspension cultured cells were harvested by filtration and cultured in MS medium.

The cultures were kept in a culture room at 26° C.±1° C. Friable callus tissues were obtained. The callu was inoculated into 4 L of MS liquid medium containing sucrose, derivative of CEP, PHE, TYR, NAA and Kinetin. Then callus tissues were cultivated 26° C. for 35 days on rotary shaker operated at 120 rpm in the dark. Cells were subcultured into fresh medium of same composition every 2 weeks and maintained at 120 rpm at 26°±1° C. Packed cell volume (PCV), fresh weight (FW), dry weight (DW), concentration of HHT and concentration of sugar were determined every 5th day. The cells were harvested and dried.

In general, callus and suspension cultures of cephalotaxus species grow very slow and no production of free or esterified HHT. However, according to the present invention, addition of IPM to cultures cause a drastic increasing in HHT after 30 days of incubation. For example, in control group (no IPM), HHT in cultured cells is 0.020 mg/g dry weight, but in treatment group (addition of IPM) HHT is about 0.050 mg/g dry weight. Therefore, IPM can increase 250% of content of HHT. It has resulted in plant cell culture systems that producing HHT at concentration higher than those produced by the mother plant. The production of HHT increases significantly after the addition of precursors (CEP). Addition of CEP can increase HHT. Obviously, the present invention provided a new commercial and economic method for producing HHT. The IPM and precursors (CEP) play key role in cultured cells.

EXAMPLE 2

Semi-Synthesis of HHT

HHT shows a significant inhibitory activity against leukemia and other cancer. Concentration of HHT, however, has only 0.01% in natural sources. Cephalotazine (CEP) is major alkaloids present in plant extracts and the concentration ofCephalotaxus has about 1%. Therefore, concentration of CEP is about 100 times higher then HHT in nature plant sources. But CEP is inactive. For the reason given above, semisynthesis of HHT from CEP will increase huge natural sources of HHT.

    • (1) Extraction of CEP

10 kg of dried stems or leaves or roots of Cephalotaxus species were milled, placed in a percolator, along 80 L of 95% of ethanol, and allowed to stand 24 hours. The ethanol was recovered under reduced pressure (below 40° C.). 20 L of 5% tartaric acid was added to concentrated ethanol solution. The ammonia water was added to the acidic solution and adjusted pH to 9. The solution of pH 9 was filtered and yielded a filtrate. The filtrate was extracted with CHCl3. CHClwas recovered under reduced pressure and residue was obtained. The residue was chromatographed packed with alumna and eluted by CHCl3-MeOH (9:1). Eluate was concentrated under reduced pressure. Residue was dried under vacuum. The product is CEP.

    • (2) Semisynthesized HHT from CEP

Materials and Methods

Melting points were determined on a Fisher-Johns apparatus. Infrared spectra were obtained on a Perkin-Elmer 567 infrared spectrophotometer or on a Beckman 4230 IR spectrophotometer. Peak positions were given in cm−1. The IR spectra of solid samples were measured as potassium bromide dispersions, and the spectra of liquids were determined in chloroform or carbon tetrachloride solutions. NMR spectra were measured on a Varian A-60, Perkin-Elmer R-32, Varian EM-390, or Brüker WH-90 NMR spectrometer. Chemical-shift values were given in parts per million downfield from Me4Si as an internal standard. Mass spectra were run on an AE1 MS-12 Finnigan 3300, or CEC21-110B mass spectrometer.

Preparative thin-layer chromatography was accomplished using 750-μm layers of aluminum oxide HF-254 (type E), aluminum oxide 60 PF-254 (type E), silica gel HF-254 (type 60 PF-254), or silica gel GF-254. Visualization was by short-wave ultraviolet light. Grace silica gel, Grade 923, and Woelm neutral aluminum oxide, activity III, were used for column chromatography. Analytical thin-layer chromatography was run on plastic sheets precoated with aluminum oxide F-254 neutral (type T), 200-μm thick, and on Polygram Sil G/UV254 (silica gel), 250 μm on plastic sheets. Visualization was usually by short-wave ultraviolet light, phosphomolybdic acid, or iodoplatinate.

Preparation of α-Ketoester-Harringtonine

1 g of Benzene-α-acetone Na was put into 10 L of benzene. Mixture was stirred at room temperature then was dissolved in 10 L of pyridine and stirred at 0° C. Oxalic chloride was added from a dropping funnel to solution of pyridine. Stirring was continued while the solution warmed to room temperature and stand overnight. Excess reagent was removed. This solution was dissolved in CH2Cl2and cooled to near 0° C. in an ice water bath. 5 g of CEP, 2.5 L of CH2Cland 2.5 L of pyridine were added to cold CH2Clsolution. Manipulations were done in a dry Natmosphere and all glassware heat-dried just before use. The suspension was stirred at room temperature and overnight. The mixture was washed with 10% Na2COand saturated aqueous NaCl, then dried with auhydrous magenesium sulfate, and filtered and the solvents were removed in vacuo. Evaporation provided as an amorphous solid α-ketoester-harringtonine (mp 143˜145° C.).

Semi-Synthesis of HHT

10 L of CH3CHBrCOOEt and activated zin dust and THF were added to the α-ketoester-harringtonine (at −78° C.) for 6 hours followed by slow warming to room temperature with stirred. The reaction mixture was diluted with 10 L CHCland 10 L H2O and solid Na2COwas added. CHClwas evaporated under reduced pressure and residue was obtained.

The residue was purified by chromatography on alumina. The column was flushed with chloroform and followed by chloroform-methanol (9:1). The solvents were recovered under reduced pressure to provide as a solid. Solid was dissolved in pure ethanol and crystallized. The crystals were refined by recrystalization in diethyl ether. The crystals dried under vacuum. The product is HHT, which has the following characters:

[α]−119° (C=0.96),

MSm/e (%): 689 (M+, 3), 314 (3), 299 (20), 298 (100), 282 (3), 266 (4), 20 (3), 150 (8), 131 (12), 73 (18)

EXAMPLE 3

HHT Extracted from Plant Tissue

Extraction of HHT has several major methods which including extraction by organic solvent, chromatograph and adjust pH.

HHT was extracted from plant tissue culture, plant cells or leaves of Cephalotaxusspecies.

1 kg of ground Cephalotaxus fortunei Hook was extracted with 10 liters of water at room temperature for 24 hrs. To filtered the solution to yield a filtrate. Ten liters of 90% ethanol added to filtrate. The mixture was Centrifugalized to yield a sediment. Percolated the sediment with ethanol and filter again to yield filtrate, combined filtrates, and distilled under reduced pressure to recover ethanol and an aqueous residue. To this residue, added 10% of HCl to adjust the pH to 2.5. To separated the solids from the solution by filtration to yield a filtrate (1). Washed the solids once with 2% HCl and filtered to yield a filtrate (2). Combined (1) and (2) and adjusted the pH to 9.5 by adding saturated sodium carbonate solution. Extracted the alkaline filtrate with chloroform and separated the chloroform layer from the aqueous layer. To repeated this extraction process five times. Combined all the chloroform extracts and distilled at reduced pressure to recover chloroform and alkaloid as a solid residue obtained. The solid alkaloid was then dissolved in 6% citric acid in water. The solution was divided into three equal portions. These were adjusted to pH 7, 8 and 9 by adding saturated sodium carbonate solution. The portions having pH 8 and 9 were combined and extracted with chloroform. The chloroform extracts were distilled under reduced pressure, whereby chloroform was removed and recovered and crude HHT was obtained. The crude HHT was dissolved in pure ethanol and crystallized. The crystals were refined by recrystallization in diethyl ether. The crude HHT obtained.

The portion having a pH of 7 passed through a liquid chromatographic column packed with alumina of diameter to height 1:50. The column was finally flushed with chloroform and followed by chloroform-methanol of 9:1 mixture. The resulting alkaloids were mixture crude of HHT. Combined crude HHT and then separated from each other by countercurrent distribution employing chloroform and pH 5 buffers. The first fraction of the countercurrent distribution was HHT. HHT was purified by crystallization in methyl alcohol. The crystallization was purified by recrystallization in methyl alcohol and dried under vacuum.

…………………….

EP1373275A2

Example 1 : Preparation of harringtonine drug substance by purification of commercial natural harringtonine

A. Analytical profile of starting product

By combination of HPLC analysis with UV detection (see Figure 6) and mass spectrometry detection (see figure 7 and 8) a total of 6.5% of related compound (identified as b,c: position isomer of harringtonine = 3.4%; d: homoharringtonine = 3%; e: 4′-demethyl harringtonine = 0.01%; f: drupacine derivative: 0.05%) are found in the starting product.

B. Chromatography of natural harringtonine

Natural harringtonine (5 grams) is injected on a preparative high-pressure liquid chromatography (HPLC) system (Prochrom stainless steel; permanent axial compression; diameter: 80 mm; length: 1000 mm) containing 1000 grams of reverse phase octadecylsilane specially dedicated for basic compounds as stationary phase. Then elution is performed in using a gradient of pH 3 buffered methanol-water solution as mobile phase (pressure 1200 psi). Unwanted fractions are discarded based upon in-line UV spectrophotometric detection. Kept fractions are collected in 16 separate containers which each are individually checked in using an analytical HPLC system exhibiting a different selectivity pattern (octadecylsilane as stationary phase and buffered acetonitrile-water system as mobile phase). During the development phase, a dual in-line UV-MS detection is used. After discarding of the fractions representing more than 0.5 % of the total content of harringtonine, fractions which complied with pre-established specification were gathered, neutralized then evaporated under reduce pressure. Then crude concentrated solution of harringtonine are alkalinized at pH 8.5 with aqueous ammonia and partitioned with dichloromethane. Resulting organic solution is concentrated under high vacuum. In-process HPLC analysis indicated a total of related compound lower than 1.5 %. C. Crystallization of raw harringtonine

Under a laminar flow hood, the above raw harringtonine (4.1 grams) is dissolved in methanol (5ml), at 30°C. The resulting alcoholic solution was filtered on a 0.25 μ sterile Millipore filter to remove microparticules and germs and collected in a sterilized rotary flask. Then, desionized water (50mL) is added and methanol is completely removed under vacuum at 30°C in using a decontaminated rotary evaporator. After removing methanol, heating is stopped and the aqueous solution of harringtonine is kept under vacuum and rotation is continued during appearance of white crystals of pure harringtonine. The stirring is continued until no more crystal occurs. Under a laminar flow hood, the suspension of is poured on a sintered glass filter with house vacuum. The resulting crystalline solid cake is washed two times with cold desionized water (10 mL x 2). The white translucent crystals are then dried using high vacuum at 40°C for 24 hours. Overall yield is 76%. All operations were documented prior to start the process and full current Good Manufacturing Practices were applied. This clinical batch corresponds to 400 therapeutic units dosed at 10mg.

D. Analysis

Routine analytical procedure includes solvent residues, loss on drying, water determination, melting point, IR and NMR spectrum, related compound and assay by HPLC. Figure 7 and 9 compare HPLC chromatogram before and after purification in using this process. Table II shows the comparison of the corresponding related compound content.

 

Figure imgf000011_0001

For the aim of further characterization, more advanced studies were performed including differential scanning calorimetry (DSC) thermogravimetry, 2D NMR, solid NMR and X-ray powder diffractometry.

Infrared Spectrometry:

Identical IR spectra were obtained by either the KBr pellet and/or mineral oil mull preparation technique. Figure 5 shows typical infrared spectrum (KBr) for unambiguous identification at the solid state of the crystalline harringtonine obtained by this process. A series of sharp absorption bands are noted at 615, 654, 674, 689, 709, 722, 750, 761 805, 850, 928, 989, 1022, 1033, 1062, 1083, 1112, 1162, 1205, 1224, 1262, 1277, 1308, 1340, 1364, 1382, 1438 1486, 1508, 1625, 1656, 1725, 1745, 2883, 2936, 2972, 3079, 3353, 3552 and 3647 cm“1

Differential Scanning Calorimetry (DSC) And Thermogravimetry (TG) Measurement of DSC and TG were obtained on a Mettler Toledo STAR System. Approximately 12 mg of harringtonine drug substance were accurately weighed (12.4471 mg) into a DSC pan. The sample was heated from 25°C to 200°C at a rate of 10°C/min. The DSC data were obtained following a standard method in the art. The DSC curve of crystalline harringtonine drug substance ((Figure 4), exhibits a melting endotherm at 79.5 °C . No subsequent decomposition occurred under the upper tested temperature 200°C. Simultaneous TG measurement, indicated a loss on drying of 1.3 % which did not correspond to a lost of structural molecule of solvent or water.

Example 2: Preparation of homoharringtonine drug substance by purification of raw semi- synthetic (hemi-synthetic) homoharringtonine

A. Analytical profile of starting product

Crude reaction mixture of raw homoharringtonine contains a potential of 250 grams of homoharringtonine DS together with process impurities such as catalyst, unchanged starting product (anhydro-homo-harringtonine), and some related side product. HPLC analysis with UV detection (see left-side chromatogram on Figure 10) indicated a total of 9 % of related impurities. B. Chromatography of semi-synthetic homoharringtonine

Raw semi-synthetic homoharringtonine (550 grams) is injected on a preparative high-pressure liquid chromatography (HPLC) system (Prochrom stainless steel; permanent axial compression; diameter: 450 mm; length: 1000 mm) containing 48,000 grams of reverse phase octadecylsilane specially dedicated for basic compounds as stationary phase. Then elution is performed in using a gradient of pH 3 buffered methanol-water solution as mobile phase (pressure 1200 psi, flow-rate 540 L/hour). Unwanted fractions are discarded based upon by- passed in-line UV spectrophotometric detector. Kept fractions are collected in 30 separate stainless steel containers (20 or 50 L each) which are individually checked in using an analytical HPLC system exhibiting a different selectivity pattern (octadecylsilane as stationary phase and buffered acetonitrile-water system as mobile phase) and equipped with a diode array detector. After discarding of the fractions representing more than 0.5 % of the total content of homoharringtonine, fractions which complied with pre-established specification were gathered, neutralized then evaporated under reduce pressure in using a mechanically stirred thin film evaporator. Then crude concentrated solution of homoharringtonine are alkalinized at pH 8.5 with aqueous ammonia and partitioned with dichloromethane. Resulting organic solution is concentrated under high vacuum. In-process HPLC analysis indicated a total of related compound lower than 0.5 % (see rigth-side chromatogram on Figure 10)

C. Crystallization of homoharringtonine DS

In a controlled clean room, under a laminar flow hood, the above raw homoharringtonine DS (210 grams) is dissolved in methanol (240 mL), at 30°C. The resulting alcoholic solution is filtered on a 0.25 μ sterile Millipore filter to remove microparticules and germs and collected in a sterilized pilot rotary flask. Then, desionized water (2400mL) is added and methanol is completely removed under vacuum at 30°C in using a decontaminated pilot rotary evaporator. After removing methanol, heating is stopped and the aqueous solution of homoharringtonine DS is kept under vacuum and rotation is continued during appearance of white crystals of pure homoharringtonine. The stirring is continued until no more crystal occurs. Under a laminar flow hood, the suspension of is poured on a sintered glass filter with house vacuum. The resulting crystalline solid cake is washed two times with cold desionized water (450 mL x 2). The white cryitals are then dried using high vacuum at 60°C for 48 hours. Overall yield is 88% from potential content of homoharringtonine in raw semi-synthetic homoharringtonine. All operations were documented prior to start the process and full current Good Manufacturing Practices were applied. This clinical batch corresponds to 40,000 therapeutic units dosed at 5mg.

D. Analysis

Routine analytical procedure includes solvent residues, loss on drying, water determination, melting point, IR and NMR spectrum, related compound and assay by HPLC. Figure 11 shows HPLC chromatogram before and after crystallization. Total of related impurities of homoharringtonine DS is 0.03%.

For the aim of further characterization, more advanced studies were performed including differential scanning calorimetry (DSC), thermogravimetry (TD), 2D NMR, solid NMR and X-ray powder diffractometry.

Infrared Spectrometry:

Identical IR spectra were obtained by either the KBr pellet and/or mineral oil mull preparation technique. Figure 3 shows typical infrared spectrum (KBr) for unambiguous identification at the solid state of the crystalline homoharringtonine obtained by this process. A series of sharp absorption bands are noted at 612, 703, 771 , 804, 826, 855, 879, 932, 1029, 1082, 1119,

1135, 1161 , 1191 , 1229, 1274, 1344, 1367, 1436, 1457, 1488, 1505, 1653, 1743, 2814, 2911 ,

2958, 3420, and 3552 cm“1

Differential Scanning Calorimetry (DSC) And Thermogravimetry (TG)

Measurement of DSC and TG were obtained on a Mettler Toledo STAR System. Approximately 11 mg of homoharringtonine drug substance were accurately weighed (10.6251 mg) into a DSC pan. The sample was heated from 25°C to 250°C at a rate of 5°C/min. The

DSC data were obtained following a standard method in the art. The DSC curve of crystalline homoharringtonine drug substance (Figure 1), exhibits a melting endotherm at 145.6 °C.

Melting range performed by the capillary method (Bucchi Apparatus) gave 143-145°C. Literature indicated 144-146°C [Anonymous, Acta Bot. Sin. 22, 156 (1980) cited by L. Huang and Z. Xue, Cephalotaxus Alkaloids, in "The Alkaloids", vol. XXIII, pp157, (1988).

Crystallization medium was not published. This is the only literature reference regarding melting point of a crystalline form of HHT] X-Ray Powder Diffraction

X-ray powder diffraction pattern was collected on a INEL microdiffractomer, model

DIFFRACTINEL. Powdered homoharringtonine DS was packed in a glass capillary tube and was analyzed according to a standard method in the art. The X-ray generator was opered at 45 kV and 40 mA, using the copper Kalpha line as the radiation source. The sample was rotated along the chi axis and data was collected between 0 and 120 deg 2-theta. A collection time of 1200 sec was used. As showed on Figure 2, the x-ray powder diffraction for this crystalline form of homoharringtonine shows a typical pattern including major reflection peaks at approximately 7.9, 9.2, 10.9, 14.9 16.0, 17.7, 19.5, 19.7, 21.78, 23.1 , 25.3, 25.4 and 25.7 deg 2-theta.

Example 3: Preparation of homoharringtonine drug substance by purification of a commercial sample of impure homoharringtonine from Chinese source

A. Analytical profile of starting product

Analytical HPLC chromatogram of natural homoharringtonine (China National Pharmaceutical) is displayed on Figure 12 (bottom left).

B. Chromatography of Natural Homoharringtonine

Natural homoharringtonine (25 grams) is injected on a preparative high-pressure liquid chromatography (HPLC) system (Prochrom stainless steel; permanent axial compression; diameter: 200 mm; length: 1000 mm) containing 12,000 grams of reverse phase octadecylsilane specially dedicated for basic compounds as stationary phase. Then elution is performed in using a gradient of pH 3 buffered methanol-water solution as mobile phase (pressure 1200 psi, flow-rate 120 IJhour). Unwanted fractions are discarded based upon bypassed in-line UV spectrophotometric detector. Kept fractions are collected in 22 separate stainless steel containers which are individually checked in using an analytical HPLC system exhibiting a different selectivity pattern (octadecylsilane as stationary phase and buffered acetonitrile-water system as mobile phase) and equipped with a diode array detector. After discarding of the fractions representing more than 0.5 % of the total content of homoharringtonine, fractions which complied with pre-established specification were gathered, neutralized then evaporated under reduce pressure in using a mechanically stirred thin film evaporator. Then crude concentrated solution of homoharringtonine are alkalinized at pH 8.5 with aqueous ammonia and partitioned with dichloromethane. Resulting organic solution is concentrated under high vacuum. In-process HPLC analysis indicated a total of related compound lower than 0.5 %.

C. Crystallization of homoharringtonine DS

In a controlled clean room, under a laminar flow hood, the above chromatographied homoharringtonine DS (18 grams) is dissolved in methanol (35 mL), at 30°C. The resulting alcoholic solution is filtered on a 0.25 μ sterile Millipore filter to remove microparticules and germs and collected in a sterilized pilot rotary flask. Then, desionized water (300 mL) is added and methanol is completely removed under vacuum at 30°C in using a decontaminated pilot rotary evaporator. After removing methanol, heating is stopped and the aqueous solution of homoharringtonine DS is kept under vacuum and rotation is continued during appearance of white crystals of pure homoharringtonine. The stirring is continued until no more crystal occurs.

Under a laminar flow hood, the suspension of is poured on a sintered glass filter with house vacuum. The resulting crystalline solid cake is washed two times with cold desionized water

(50 mL x 2). The white crystals are then dried using high vacuum at 60°C for 48 hours. Overall yield is 84% from potential content of homoharringtonine in raw semi-synthetic homoharringtonine. All operations were documented prior to start the process and full current

Good Manufacturing Practices were applied.

D. Analysis

Routine analytical procedure includes solvent residues, loss on drying, water determination, melting point, IR and NMR spectrum, related compound and assay by HPLC. Figure 12 (bottom right) shows HPLC chromatogram after crystallization. Total of related impurities of homoharringtonine DS is 0.05%.

For the aim of further characterization, more advanced studies were performed including differential scanning calorimetry (DSC), thermogravimetry (TD), 2D NMR, solid NMR and X-ray powder diffractometry. Infrared Spectra, Differential Scanning Calorimetry (DSC) and X-Ray Powder Diffraction gave patterns strictly superimposable to the one of example 2 obtained from semi-synthetic homoharringtonine (Figure 3, 1 , and 2, respectively).

………………………………….

KOREAN PAPER.. LINK

Title: 한국산 개비자(Cephalotaxus koreans)에서의 Harringtonine과 Homoharringtonine의 확인 및 함량 분석
Author: 박호일 ; 이연 (한국생물공학회)
Source: 한국생물공학회지 = Korean journal of biotechnology and bioengineering; ISSN:1225-7117 @ 1225-7117 @ ; VOL.11; NO.6; PAGE.689-695; (1996)
Pub.Country: Korea
Language: Korean
Abstract: Harringtonine and homoharringtonine known as anti-cancer agents were isolated from Korean native plumyew(Cephalotaxus koreana) using column chromatography(CHCl3:MeOH=19:1, Rf=0.28). The structure of the mixture of two compounds was characterized by 1H-NMR. Comparison of our spectra of harringtonine and homoharringtonine with previously reported ones indicated that the two are identical. The contents of harringtonine and homoharringtonine in the needles, stems, and roots of Korean native plumyew were determined by high performance liquid chromatography(HPLC). The contents of both compounds varied with the site of location and the part of plant. The content of harringtonine was higher in needles and roots than in stems, whereas the content of homoharringtonlne was lower than harringtonine. Homoharringtonine contents in needles at Mt. Palgong, Mt. Dukyu, Mt. Baekyang, Mt. Jiri, and Namhae were higher than in stems and roots. But homoharringtonine contents in needles al Mt. Jokye and Jindo were lower than in stems and roots.

http://img.kisti.re.kr/originalView/originalView.jsp

……………………………………………………………………………….

SYNTHESIS OF HOMOHARRINGTONINE AND SEPARATION OF ITS STEREOMERS

WANG YONG-KENG LI YU-LIN PAN XIN-FU LI SHAO-BAI HUANG WEN-KUI (Institute of Organic Chemistry,Lanzhou University)
Ethyl 2-oxo-6-ethylene dioxy-heptanoate(2),an important intermediate in the preparation of homoharringtonine(8a),was prepared by the reaction of Grignard reagent made from 1-bromo-4-ethylene dioxy pentane with ethyl oxalate in THF. Compound 2 was converted into α-keto-acyl-cephalotaxine(5)via sodium carboxylate 3 and acyl chloride 4.Reformatsky reaction of 5 with methyl bromoacetate in the presence of freshly prepared active zinc affords 6.Acid treatment of 6 gave 7.Reaction of 7 with methyl magnesium iodide provided a mixture of homoharringtonine(8a) and its epimer 8b.Their separation is effected by fractional crystallization of their picrates and subsequent recovery of the free alkaloids 8a and 8b.The TLC,IR,~1H NMR and MS data of 8a are identical with those of natural homoharringtonine.The IR and MS of 8a and 8b are quite similar,but their ~1H NMR are markedly different
………………………………………………..
READ
  1. [PDF]

    Chapter 1 Drug Discovery from Plants – Springer

    LC-NMR-MS and LC-SPE-NMR to accelerate their future discovery. Keywords …..Ceflatonine (34), a synthetic version of homoharringtonine produced by.

    …………………………………………………………………………….

References

  1.  “Synribo (omacetaxine) dosing, indications, interactions, adverse effects, and more”Medscape Reference. WebMD. Retrieved 18 February 2014.
  2.  “SYNRIBO (omacetaxine mepesuccinate) injection, powder, lyophilized, for solution [Cephalon, Inc.]“DailyMed. Cephalon, Inc. October 2012. Retrieved 18 February 2014.
  3.  Sweetman, S, ed. (14 November 2012). Omacetaxine Mepesuccinate. “Martindale: The Complete Drug Reference”. Medicines Complete(Pharmaceutical Press).
  4.  Li, Y. F.; Deng, Z. K.; Xuan, H. B.; Zhu, J. B.; Ding, B. H.; Liu, X. N.; Chen, B. A. (2009). “Prolonged chronic phase in chronic myelogenous leukemia after homoharringtonine therapy”. Chinese medical journal122 (12): 1413–1417. PMID 19567163edit
  5.  Quintás-Cardama, A.; Kantarjian, H.; Cortes, J. (2009). “Homoharringtonine, omacetaxine mepesuccinate, and chronic myeloid leukemia circa 2009″. Cancer 115 (23): 5382–5393.doi:10.1002/cncr.24601PMID 19739234edit
  6.  Wu, L.; Li, X.; Su, J.; Chang, C.; He, Q.; Zhang, X.; Xu, L.; Song, L.; Pu, Q. (2009). “Effect of low-dose cytarabine, homoharringtonine and granulocyte colony-stimulating factor priming regimen on patients with advanced myelodysplastic syndrome or acute myeloid leukemia transformed from myelodysplastic syndrome”. Leukemia & Lymphoma50 (9): 1461. doi:10.1080/10428190903096719edit
  7.  Gu, L. F.; Zhang, W. G.; Wang, F. X.; Cao, X. M.; Chen, Y. X.; He, A. L.; Liu, J.; Ma, X. R. (2010). “Low dose of homoharringtonine and cytarabine combined with granulocyte colony-stimulating factor priming on the outcome of relapsed or refractory acute myeloid leukemia”.Journal of Cancer Research and Clinical Oncology 137 (6): 997–1003.doi:10.1007/s00432-010-0947-zPMID 21152934edit
  8.  Kantarjian, H. M.; Talpaz, M.; Santini, V.; Murgo, A.; Cheson, B.; O’Brien, S. M. (2001). “Homoharringtonine”. Cancer 92 (6): 1591–1605.doi:10.1002/1097-0142(20010915)92:6<1591::AID-CNCR1485>3.0.CO;2-UPMID 11745238edit
  9.  Wetzler M, Segal D. Omacetaxine as an Anticancer Therapeutic: What is Old is New Again. Current Pharmaceutical Design 2011;17:59-64
  10. Concise total synthesis of (±)-cephalotaxine via a transannulation strategy: Development of a facile reductive oxy-nazarov cyclization
    Org Lett 2011, 13(13): 3538
  11. The first semi-synthesis of enantiopure homoharringtonine via anhydrohomoharringtonine from a preformed chiral acyl moiety
    Tetrahedron Lett 1999, 40: 2931
  12. Synthesis of homoharringtonine and its derivative by partial esterification of cephalotaxine
    Tetrahedron Lett 1982, 23(34): 3431
  13. Construction of chiral tertiary alcohol stereocenters via the (2,3)-Meisenheimer rearrangement: Enantioselective synthesis of the side-chain acids of homoharringtonine and harringtonine
    J Org Chem 2013, 78(2): 339
  14. Studies in Cephalotaxus alkaloids. Stereospecific total synthesis of homoharringtonine
    J Org Chem 1983, 48(26): 5321
  15. Chemistry – A European Journal, 2008 ,  vol. 14,   14  pg. 4293 – 4306
WO2000040269A2 * Jan 5, 2000 Jul 13, 2000 Clarence C Lee Pharmaceutical compositions for treatment of diseased tissues
WO2002032904A1 * Oct 17, 2000 Apr 25, 2002 Oncopharm Corp New cephalotaxanes, their method of preparation and their use in treatment of cancers, leukemias, parasites including thus resistant to usual chemotherapeutic agents and as reversal agents
EP0393575A1 * Apr 17, 1990 Oct 24, 1990 G.D. Searle &amp; Co. Neoplasia treatment compositions containing antineoplastic agent and side-effect reducing protective agent
USH271 * Dec 18, 1985 May 5, 1987 The United States Of America As Represented By The Secretary Of The Army Treatment of malaria with esters of cephalotaxine
US7169774 Jun 25, 2004 Jan 30, 2007 Stragen Pharma S.A. Cephalotaxane derivatives and their processes of preparation and purification
US7842687 May 25, 2006 Nov 30, 2010 Chemgenex Pharmaceuticals, Inc. Cephalotaxane derivatives and their processes of preparation and purification
US8466142 Mar 3, 2009 Jun 18, 2013 Sloan-Kettering Institute For Cancer Research Cephalotaxus esters, methods of synthesis, and uses thereof
Reference
1 * KANTARJIAN H.M. ET AL: “Chronic myelogenous leukemia – Progress at the M. D. Anderson Cancer Center over the past two decades and future directions: First Emil J Freireich Award Lecture.” CLINICAL CANCER RESEARCH, (1997) 3/12 II (2723-2733). , XP001095529
2 * LEVY, VINCENT (1) ET AL: “Subcutaneous homoharringtonine (SQ HHT ): 1. Pharmacokinetic study in dogs and HHT determination in blood in using LC-MS method.” BLOOD, (NOVEMBER 16, 2001) VOL. 98, NO. 11 PART 2, PP. 179B. HTTP://WWW.BLOODJOURNAL.ORG/. PRINT. MEETING INFO.: 43RD ANNUAL MEETING OF THE AMERICAN SOCIETY OF HEMATOLOGY, PART 2 ORLANDO, FLORIDA, USA DECEMBER 07-11, 2001 , XP001095449
3 * LEVY, VINCENT (1) ET AL: “Subcutaneous homoharringtonine (SQ HHT ): 2. Tolerance in humans and case report of a refractory patient with AML treated by very small dose of SQ HHT.” BLOOD, (NOVEMBER 16, 2001) VOL. 98, NO. 11 PART 2, PP. 202B. HTTP://WWW.BLOODJOURNAL.ORG/. PRINT. MEETING INFO.: 43RD ANNUAL MEETING OF THE AMERICAN SOCIETY OF HEMATOLOGY, PART 2 ORLANDO, FLORIDA, USA DECEMBER 07-11, 2001 , XP001095450
4 * WHAUN J M ET AL: “TREATMENT OF CHLOROQUINE -RESISTANT MALARIA WITH ESTERS OF CEPHALOTAXINE HOMOHARRINGTONINE.” ANN TROP MED PARASITOL(1990) 84(3), 229-237, XP008006193

1H NMR

13 CNMR

HPLC


Filed under: 0rphan drug status, cancer, FDA 2014, NEW DRUGS Tagged: chronic myeloid leukaemia (CML)., Homoharringtonine, omacetaxine mepesuccinate, Synribo, Teva's

The secret of fertile sperm

$
0
0

Originally posted on lyranara.me:

The secret of fertile sperm

Progesterone and other fatty signaling molecules are critical for sperm fertility. Credit: C. Cain

To better understand the causes of male infertility, a team of Bay Area researchers is exploring the factors, both physiological and biochemical, that differentiate fertile sperm from infertile sperm. At the 58th Annual Biophysical Society Meeting, which takes place Feb. 15-19, 2014, in San Francisco, Calif., the team will present its work to identify and characterize proteins known as ion channels, which are crucial for sperm fertility and expressed within a sperm cell’s plasma membrane.

View original


Filed under: Uncategorized

MOXIFLOXACIN, Bay-12-8039

$
0
0

MOXIFLOXACIN

Bay-12-8039

US FDA:link

CAS 354812-41-2

186826-86-8 HYDROCHLORIDE

 1-cyclopropyl-7-[(1S,6S)-2,8-diazabicyclo[4.3.0]non-8-yl]-6-fluoro-8-methoxy-4-oxo- quinoline-3-carboxylic acid
(4aS-Cis) -l-cyclopropyl-7- (2, 8- diazabicyclo [4.3.0] non-8-yl) -6-fluoro-8-methoxy-4-oxo-l, 4-dihydro-3- quinoline carboxylic acid
ALSO AS….
Vigamox, Avelox I.V., 151096-09-2, Moxifloxacin [INN:BAN], MXFX, CHEMBL32, Actira (*Hydrochloride*), Avelox (*Hydrochloride*)
Molecular Formula: C21H24FN3O4
Molecular Weight: 401.431363
Moxifloxacin is a synthetic fluoroquinolone antibiotic agent. Bayer AG developed the drug (initially called BAY 12-8039) and it is marketed worldwide (as the hydrochloride) under the brand name Avelox (in some countries also Avalox) for oral treatment
For the treatment of sinus and lung infections such as sinusitis, pneumonia, and secondary infections in chronic bronchitis. Also for the treatment of bacterial conjunctivitis (pinkeye).
Moxifloxacin and its salts are antibacterial agents, which were disclosed in EP 550,903. Moxifloxacin, chemically 1-Cyclopropyl-6-fluoro-1,4-dihydro-8-methoxy-7-[(4aS, 7aS)-octahydro-6H-pyrrolo[3,4-b]pyridine-6-yl]-4-oxo-3-quinolinecarboxylic acid

Moxifloxacin is a fourth-generation synthetic fluoroquinolone antibacterial agent developed by Bayer AG (initially called BAY 12-8039). It is marketed worldwide (as the hydrochloride) under the brand names AveloxAvalox, and Avelon for oral treatment. In most countries, the drug is also available in parenteral form for intravenous infusion. Moxifloxacin is also sold in an ophthalmic solution (eye drops) under the brand names VigamoxMoxezafor the treatment of conjunctivitis (pink eye).

A United States patent application was submitted on 30 June 1989, for Avelox (moxifloxacin hydrochloride).[1] In 1999 Avelox was approved by theU.S. Food and Drug Administration (FDA) for use in the United States.[2]

In the United States, moxifloxacin is licensed for the treatment of acute bacterial sinusitis, acute bacterial exacerbation of chronic bronchitis, community acquired pneumonia, complicated and uncomplicated skin and skin structure infections, and complicated intra-abdominal infections.[3]

In the European Union, it is licensed for acute bacterial exacerbations of chronic bronchitis, non-severe community-acquired pneumonia, and acute bacterial sinusitis. Based on its investigation into reports of rare but severe cases of liver toxicity and skin reactions, the European Medicines Agency recommended in 2008 that the use of the oral (but not the IV) form of moxifloxacin be restricted to infections in which other antibacterial agents cannot be used or have failed.[4] In the US, the marketing approval does not contain these restrictions, though the label contains prominent warnings against skin reactions.

MOXIFLOXACIN

Avelox (moxifloxacin) was launched in the United States in 1999 and is currently marketed in more than 80 countries worldwide. In the United States, Avelox is marketed by Bayer’s partner Merck.

In 2011 the FDA added two boxed warnings for this drug in reference to spontaneous tendon ruptures and the fact that moxifloxacin may cause worsening of myasthenia gravis symptoms, including muscle weakness and life-threatening breathing problems.[5]

Moxifloxacin is used to treat a number of infections including: respiratory tract infectionscellulitisanthrax, intraabdominal infections, endocarditis,meningitis, and tuberculosis.[6]

The initial approval by the FDA (December 1999)[7] encompassed the following indications:

  • Acute Exacerbations of Chronic Bronchitis (AECB)
  • Acute Bacterial Sinusitis (ABS)
  • Community Acquired Pneumonia (CAP)

Additional indications were approved by the FDA as follows:

  • April 2001: Uncomplicated Skin and Skin Structure Infections (uSSSI)[8]
  • May 2004: Community Acquired Pneumonia caused by multi-drug resistant Streptococcus pneumoniae.[9]
  • June 2005: Complicated Skin and Skin Structure Infections (cSSSI)[10]
  • November 2005: Complicated Intra-Abdominal Infections (cIAI).[11]

The European Union requires that moxifloxacin only be prescribed when other antibiotics that have been initially recommended for treatment cannot be used or have failed.[12][13]

At the current time,[when?] there are no approved uses within the pediatric population for Oral and I.V. moxifloxacin. A significant number of drugs found within this class, including moxifloxacin, are not licensed by the FDA for use in children due to the risk of permanent injury to the musculoskeletal system.[14][15][16]

In ophthalmology, moxifloxacin is approved for the treatment of conjunctival infections caused by susceptible bacteria.[17]

Note: Moxifloxacin may be licensed for other uses, or restricted, by the various regulatory agencies worldwide

Marketing authorisations for the tablet and injectable forms of Moxifloxacin are held by Bayer, while Alcon (now a subsidiary of Novartis) produces ophthalmic solutions for treating conjunctivitis under the brand names of Moxeza, Vigamox, and Moxivig. Avelox generated sales of USD320 million in the first 9 months of 2013.

Moxifloxacin is available in three distinct administration forms. Formulated as a salt, Moxifloxacin hydrochloride is sold as an oral 400 mg film-coated tablet and as an injectable solution for infusion by Bayer; although in the US it is distributed by Merck Sharp and Dohme under license from Bayer.

Alcon has formulated Moxifloxacin hydrochloride as a 0.5% ophthalmic solution under license from Bayer. Moxifloxacin was first discovered in 1988 and received the first market authorisation eleven years later in 1999 in the US.

Moxifloxacin hydrochloride is a synthetic broad-spectrum antibacterial agent. The active moiety, moxifloxacin has been shown to be clinically active against most strains of microorganisms such as aerobic gram-positive microorganisms including staphylococcus aureus, streptococcus pneumonia (penicillin-susceptible strains) and streptococcus pyogenes, aerobic gram-negative microorganisms including haemophilus influenza hemophilus parainfluenzae, klebisiella pneumonia. Moxifloxacin is commercially available under the brand name of AVELOX® marketed by Bayer pharms.

VIGAMOX® (moxifloxacin hydrochloride ophthalmic solution) 0.5% is a sterile solution for topical ophthalmic use. Moxifloxacin hydrochloride is an 8-methoxy fluoroquinolone anti-infective, with a diazabicyclononyl ring at the C7 position.

VIGAMOX®<br /><br /><br /><br /><br /><br /><br /><br /><br /><br /><br /><br /><br /><br />
  (moxifloxacin hydrochloride) Structural Formula Illustration” src=”<a href=http://images.rxlist.com/images/rxlist/vigamox1.gif&#8221; width=”286″ height=”171″ />

C21H24FN304•HC1       Mol Wt 437.9

Chemical Name: l-Cyclopropyl-6-fluoro-l,4-dihydro-8-methoxy-7-[(4aS,7aS)-octahydro-6H-pyrrolol[3,4-b]pyridin-6-yl]-4-oxo-3-quinolinecarboxylic acid, monohydrochloride. Moxifloxacin hydrochloride is a slightly yellow to yellow crystalline powder. Each mL of VIGAMOX® solution contains 5.45 mg moxifloxacin hydrochloride, equivalent to 5 mg moxifloxacin base.

Contains: Active: Moxifloxacin 0.5% (5 mg/mL); Inactives: Boric acid, sodium chloride, and purified water. May also contain hydrochloric acid/sodium hydroxide to adjust pH to approximately 6.8.

VIGAMOX® solution is an isotonic solution with an osmolality of approximately 290 mOsm/kg.

Market Considerations

Amongst the US approvals, Dr. Reddy’s, Teva, Torrent, and Aurobindo have received tentative approvals for the 400 mg oral tablet formulation. Akorn, Teva and Apotex have received tentative approvals for a Moxifloxacin hydrochloride ophthalmic solution. No 180 day period of exclusivity has been awarded since all patents were found to be valid.

In the UK, Teva, Rivopharm, and Double-E Pharma have received marketing authorisations for the 400 mg Moxifloxacin tablets, while Noridem has received a market authorisation for the equivalent 400mg/250ml solution for infusion. A similar trend of generic competition, for tablets and infusions, following molecule patent expiry is expected throughout Europe. Currently no generic market authorisations for ophthalmic formulations have been granted in major European countries. However, Sandoz and Hexal have gained market authorisations in some European markets for the ophthalmic dosage form following Novartis’ acquisition of Alcon.

In Canada the only generic manufacturer holding a marketing authorisation is Sandoz, however, this was granted as a New Drug Submission rather than as an ANDS. Following patent expiries from mid-2014, the European and North American markets are likely to have significant competition if the numbers of companies filing litigation, ANDS, ANDAs and the like in the northern hemisphere is anything to go by.

MOXIFLOXACIN

History

Moxifloxacin was first patented (United States patent) in 1991 by Bayer A.G., and again in 1997.[47] Avelox was subsequently approved by the U.S. Food and Drug Administration (FDA) for use in the United States in 1999 to treat specific bacterial infections.[2] Ranking 140th within the top 200 prescribed drugs in the United States for 2007[48] moxifloxacin, in the same manner asciprofloxacin, has proven to be a blockbuster drug for Bayer A. G., generating billions of dollars in additional revenue. In 2007 alone, Avelox generated sales of $697.3 million dollars worldwide.[26]

Moxifloxacin is also manufactured by Alcon as Vigamox.[citation needed]

Patent

A United States patent application was made on 30 June 1989, for Avelox (moxifloxacin hydrochloride),(Bayer A.G. being the assignee), which was subsequently approved on 5 February 1991. This patent was scheduled to expire on 30 June 2009. However, this patent was extended for an additional two and one half years on 16 September 2004, and as such is not expected to expire until 2012.[49] Moxifloxacin was subsequently (ten years later) approved by the U.S. Food and Drug Administration (FDA) for use in the United States in 1999. There have been at least four additional United States patents filed regarding moxifloxacin hydrochloride since the 1989 United States application,[47][50] as well as patents outside of the USA.

Additional regulatory history

6/12/2002 Changes made to minimize the impact of warnings concerning adverse reactions.[51]

26 June 2003 New Zealand Pharmacovigilance warns of moxifloxacin induced respiratory insufficiency.[52]

10/6/2003 Changes made to minimize the impact of post marketing reports as well as the risk of tendon injuries.[53]

29 December 2008 Addition of numerous adverse reactions associated with the use of moxifloxacin.[54]

27 April 2009 Issuance of a Medication Guide and revisions to include new safety information including the addition of the Black Box Warning to the Medication Guide. The FDA had determined that Moxifloxacin poses a serious and significant public health concern, requiring the distribution of a Medication Guide.[55]

24 June 2009 Updating of the carton and container labels to include a statement to let dispensers know that a Medication Guide must be dispensed with the product.(emphasis added)[56]

Patent related

Country Patent Number Approved Expires (estimated)
Canada 1340114 1998-11-03 2015-11-03
Canada 2342211 2009-05-26 2019-09-29
United States 4990517 1994-12-08 2011-12-08
United States 6548079 2000-07-25 2020-07-25

As indicated by the Key Patent Indicator (Fig. 2), patents in the families with priority DE3824072A, 15/07/1988 (‘072), and DE4200414A, 10/01/1992, (‘414) provide protection for the Moxifloxacin molecule and are considered to be the main constraint to generic entry. As patents in the ‘414 family have expired or were never granted, the only remaining constraint to generic entry is the ‘072 family. The term of the Australian patent of this family have been extended to 19 June 2014 while the Canadian member will enjoy the longer term of 17 years from grant, expiring in November 2015.

Supplementary protection certificates (SPCs) have been granted in France, Germany, Spain and the UK, and will expire in June 2014. Given that there are less than 2 years until these SPCs expire, and that as yet no applications for paediatric extension of the SPCs have been published in Europe, they are unlikely to be extended by 6 months on the basis of the approved Paediatric Investigation Plans.

The US member, 4,990,517 (‘517), protecting the general structure of the Moxifloxacin molecule, expired in June 2012, after enjoying 6 month paediatric extension on top of a 901 day s156 extension. However, the absence of generics on the US market is due to Bayer securing a divisional patent, 5,607,942 (‘942). This patent claims the Moxifloxacin molecule specifically and is due to expire in September 2014, after being awarded a 6 month paediatric extension.

Members of the ‘072 family from both Canada and the US have been the subject of litigation after generic manufacturers identified these patents in paragraph IV filings, and the equivalent in Canada, as early as 2006. After filing an infringement suit in the US against Teva in relation to US ‘517, US ‘942, Bayer enjoyed a satisfying validification of their patents when Teva agreed that it would be infringing two of the patents, while the third was decided by the court to be equally valid.

In Canada, Novopharm, Cobalt, Apotex, Mylan and Apotex have also all tested the litigation waters relating to the equivalent patent with no success noted so far.

A third patent family that promises to be a constraint for generic ophthalmic formulations is Alcon’s 1998 patent, US10250498P (Fig. 2), which identifies an ophthalmic formulation of Moxifloxacin and its use in the treatment and prevention of eye infections. Patents in this family are set to expire in August 2019. US members 6,716,830 (‘830) and 7,671,070 (‘070) have been awarded a 6 month paediatric extension, extending their expiration until March 2020. The validity of US ‘830 was upheld following Teva filing paragraph IV certifications to manufacture generic Vigamox. Teva has since appealed this ruling.

In addition, Alcon has filed patent infringement suits against Watson, Lupin and Apotex in relation to US ‘070 after they submitted Abbreviated New Drug Applications (ANDAs)with paragraph IV filings in preparation for commercialisation of a Moxeza/Vigamox generic equivalent. There has been no outcome from these suits to date. In addition, applications for Orders of Prohibition against Cobalt, Apotex and Teva have also been noted for the equivalent Canadian patent following the filing of Abbreviated New Drug Submissions (ANDS) by these companies.

The equivalent European patent 1,117,401 was revoked following opposition by Teva filed in the European patent office. Its divisional patents, 1,384,478 (granted) and 2,301,541 (accepted) have restricted claims to the use of Moxifloxacin in the topical treatment of ophthalmic infections caused by P. aeruginosa and H. influenza, respectively. This may provide a prepared generic competitor an opportunity to launch their Moxifloxacin ophthalmic equivalent in Europe soon after the expiry of patents protecting the molecule, subject to legal review of the remaining claims of the patent.

Alcon have secured additional protection for their Moxeza ophthalmic formulation by way of patents in the family with priority US5987708P (09/06/2008). Patent claims specify ratios of Moxifloxacin to inactive ingredients and additional inactive ingredients and therefore generic competitors are likely to circumvent the patent by reformulation. Lupin has filed paragraph IV certifications to US8450311, which is currently subject of a patent infringement suit.

Families with priorities DE19546249A (12/12/1995), DE19751948A (24/11/1997), DE19855758A (10/11/1998) and US36433499A (30/07/1999) are not considered to be a constraint to generic entry because the protected technologies are likely to be circumvented.

Generic equivalents

In 2007, the U.S. District Court for the District of Delaware held that two Bayer patents on Avelox (moxifloxacin hydrochloride) are valid and enforceable, and infringed by Dr. Reddy’s ANDA for a generic version of Avelox.[70][71] The district court sided with Bayer, citing the Federal Circuit’s prior decision in Takeda v. Alphapharm[72] as “affirming the district court’s finding that defendant failed to prove a prima facie case of obviousness where the prior art disclosed a broad selection of compounds, any one of which could have been selected as a lead compound for further investigation, and defendant did not prove that the prior art would have led to the selection of the particular compound singled out by defendant.” According to Bayer’s press release[70] announcing the court’s decision, it was noted that Teva had also challenged the validity of the same Bayer patents at issue in the Dr. Reddy’s case. Within Bayer’s first quarter 2008 stockholder’s newsletter[73]

Bayer stated that they had reached an agreement with Teva Pharmaceuticals USA, Inc., the adverse party, to settle their patent litigation with regard to the two Bayer patents. Under the settlement terms agreed upon, Teva would obtain a license to sell its generic moxifloxacin tablet product in the U.S. shortly before the second of the two Bayer patents expires in March 2014.

  • Economic impact: adverse reactions:

The advocacy group Public Citizen has lobbied for increasing safety warnings and for the removal of some fluoroquinolone drugs from clinical practice.[74][75][76][77][78][79][80][81]

3-5-1997
7-(1-pyrrolidinyl)-3-quinolone- and – naphthyridone-carboxylic acid derivatives as antibacterial agents and feed additives
6-9-2000
INHIBITORS OF MULTIDRUG TRANSPORTERS INHIBITORS OF MULTIDRUG TRANSPORTERS
5-19-2000
PHARMACEUTICAL MOXIFLOXACIN PREPARATION
5-12-2000
AQUEOUS DRUG FORMULATION FOR ORAL APPLICATION AQUEOUS DRUG FORMULATION FOR ORAL APPLICATION
4-7-2000
ANTIBIOTIC COMPOSITIONS FOR TREATMENT OF THE EYE, EAR AND NOSE ANTIBIOTIC COMPOSITIONS FOR TREATMENT OF THE EYE, EAR AND NOSE
4-7-2000
ANTIBIOTIC COMPOSITIONS FOR TREATMENT OF THE EYE, EAR AND NOSE ANTIBIOTIC COMPOSITIONS FOR TREATMENT OF THE EYE, EAR AND NOSE
4-7-2000
ANTIBIOTIC COMPOSITIONS FOR TREATMENT OF THE EYE, EAR AND NOSE
1-20-2000
COMBINATION PREPARATION FOR ORALLY ADMINISTERED ANTIBIOTICS
12-17-1999
NOVEL THERAPEUTIC AGENTS THAT MODULATE ENZYMATIC PROCESSES
4-2-1999
MEDICAMENT FORMULATION WITH A CONTROLLED RELEASE OF AN ACTIVE AGENT
8-28-1998
COMBINATION PREPARATION FOR ORALLY ADMINISTERED ANTIBIOTICS COMBINATION PREPARATION FOR ORALLY ADMINISTERED ANTIBIOTICS
11-10-2006
Amorphous moxifloxacin hydrochloride
10-4-2006
Method for producing 8-methoxy-quinolinecarboxylic acids
7-15-2005
Pharmaceutical composition
7-13-2005
Aqueous pharmaceutical composition containing moxifloxacin or salts thereof
5-25-2005
Method for producing 8-methoxy-quinolinecarboxylic acids
2-6-2004
Medicinal composition
5-21-2003
Method for the enantiomer separation of cis-8-benzyl-7,9-dioxo-2,8-diazabicyclo[4.3.0]nonane
11-31-2000
OPTICALLY ACTIVE QUINOLINE CARBOXYLIC ACID DERIVATIVES HAVING 7-PYRROLIDINE SUBSTITUTES CAUSING OPTICAL ACTIVITY AND A PROCESS FOR PREPARING THEREOF
11-17-2000
ANTIBACTERIAL OPTICALLY PURE BENZOQUINOLIZINE CARBOXYLIC ACIDS, PROCESSES, COMPOSITIONS AND METHODS OF TREATMENT (S)-BENZOQUINOLIZINE CARBOXYLIC ACIDS AND THEIR USE AS ANTIBACTERIAL AGENTS
8-32-2000
COMPOSITIONS AND METHODS FOR IMPROVED DELIVERY OF HYDROPHOBIC THERAPEUTIC AGENTS
6-18-2010
MULTI-ARM POLYMER PRODRUGS
4-9-2010
COMPOSITION COMPRISING AN ANTIBIOTIC AND A CORTICOSTEROID
7-22-2009
Tri-, tetra-substituted-3-aminopyrrolidine derivative
7-3-2009
NOVEL HYDRATE FORM
3-20-2009
Multi-Arm Polymer Prodrugs
1-16-2009
Sulfonamide Derivatives for the Treatment of Bacterial Infections
11-21-2008
Phosphonated Fluoroquinolones, Antibacterial Analogs Thereof, and Methods for the Prevention and Treatment of Bone and Joint Infections
8-15-2008
Multi-arm polymer prodrugs
8-24-2007
PHARMACEUTICAL COMPOSITION
6-29-2007
PHARMACEUTICAL COMPOSITION
               7-4-2012
                                                                 TRI-, TETRA-SUBSTITUTED-3-AMINOPYRROLIDINE DERIVATIVE
6-13-2012
Process for the Synthesis of Moxifloxacin Hydrochloride
12-2-2011
COMPACTED MOXIFLOXACIN
10-12-2011
Treatment of bacterial diseases of the respiratory organs
9-16-2011
Novel Hydrate Form
9-2-2011
NOVEL POLYMORPH OF MOXIFLOXACIN HYDROCHLORIDE
6-31-2011
PHARMACEUTICAL COMPOSITION
6-10-2011
SYNTHESIS OF (4aS,7aS)-OCTAHYDRO-1H-PYRROLO[3,4-b]PYRIDINE
7-30-2010
MULTI-ARM POLYMER PRODRUGS
6-30-2010
Multi-arm polymer prodrugs

DESCRIPTION

  • Moxifloxacin is a therapeutic agent that shows a broad spectrum antibacterial action. Moxifloxacin is the international non-proprietary name (INN) for 1-cyclopropyl-6-fluoro-1,4-dihydro-8-methoxy-7-[(4aS,7aS)-octahydro-6H-pyrrolo[3,4-b]pyridin-6-yl]-4-oxo-3-quinolinecarboxylic acid.
  • The structure of moxifloxacin corresponds to formula (I):
    Figure imgb0001
  • Racemic moxifloxacin was firstly described in EP-A-350733 and, particularly, moxifloxacin having a (S,S)-configuration is described inEP-A-550903 .
  • In experimental example 19 of EP-A-550903 and example Z19 of EP-A-591808 , a method for preparing and isolating moxifloxacin base is described. The same method is described in patent document EP-A-592868 ). These published patent applications neither describe nor suggest the possible existence of a crystalline form of moxifloxacin base. In WO-A-2008059521 it is disclosed that by performing the mentioned examples an acetonitrile solvated form of moxifloxacin with low purity is obtained, and so the obtained solvate form can not be used as such in pharmaceutical formulations. In the above mentioned examples, the obtained moxifloxacin base crude is purified and isolated by chromatography using methylene chloride/methanol/17% aqueous ammonia as the solvent system. The purification process disclosed in said documents has been reproduced by the authors of the present invention, but only an amorphous form of moxifloxacin was obtained. This process for the purification and isolation of moxifloxacin base is complex and difficult to perform on industrial scale due to the need for purifying the product by column chromatography.
  • WO-A-9926940 discloses a process for the preparation of moxifloxacin from a difluoro precursor comprising the step of adjusting the pH to 6.8- 7.0. However, the reproduction of this example shows that at this pH moxifloxacin hydrochloride or a mixture of moxifloxacin hydrochloride and moxifloxacin base is obtained, since the X-ray diffractogram of the isolated compound corresponds with the hydrochloride moxifloxacin. This fact has also been confirmed by the reproduction of the subsequent crystallization described in the international patent application of the previous compound in ethanol/water. The behaviour of this solid was very different regarding its solubility in respect what was described in the international patent application.
  • WO-A-2004091619 and WO-A-2007010555 disclose the preparation of moxifloxacin base as a solid. Nevertheless, they do not describe nor suggest the preparation of a crystalline form of moxifloxacin base. The authors of the present invention have proved that the X-ray diffractogram of the product obtained by reproducing the reference example disclosed in WO-A-2004091619 , based on a pH adjustment to 7.0-7.2, corresponds to the X-ray diffractogram of the monohydrate of moxifloxacin hydrochloride disclosed in US 5849752 . Similarly, by reproducing the reference example of WO-A-2007010555 , also for obtaining moxifloxacin base but based on a pH adjustment to 5.0-6.0, it is neither expected to obtain moxifloxacin base due to the adjustment of the solution within a range of acidic pH values.
  • [0008]
    WO-A-2008059521 discloses a process for the preparation of a crystalline form of moxifloxacin base, designated as Form I, by recrystallization in a ketosolvent, such as acetone. This form is characterized by its X-ray diffraction pattern corresponding to a hemihydrate. The best yield obtained is a 78.2% starting from moxifloxacin hydrochloride in example 18. This crystalline form has a tendency to occlude solvent molecules within the crystalline network in amounts very superiors to the allowed ones by for Guidelines Residual Solvents (CPMP/ICH/283/95) and can be difficult to impossible to remove by drying, what forces to carry out laborious treatments, either physical, or chemical to reach allowed solvent levels. The presence of any non-aqueous solvents in amounts over the allowed ones would not make this crystalline form suitable for the preparation of pharmaceutical formulations.
  • The existence of polymorphs is unpredictable and there is no a priori established procedure to prepare an unknown polymorph. The difference in the physical properties of different morphological forms results from the orientation and intermolecular interactions of adjacent molecules are complexes in the bulk solid. Furthermore, the different solid forms of a pharmaceutically active ingredient can have different characteristics, and offer certain advantages in methods of manufacture and also in pharmacology. Thus, the discovery of new solid forms can contribute to clear improvements in the efficiency of methods of production and/or improvements in the characteristics of the pharmaceutical formulations of the active ingredients, since some forms are more adequate for one type of formulation, and other forms for other different formulations.

Moxifloxacin Hydrochloride namely (4aS-Cis) -l-cyclopropyl-7- (2, 8- diazabicyclo [4.3.0] non-8-yl) -6-fluoro-8-methoxy-4-oxo-l, 4-dihydro-3- quinoline carboxylic acidhydrochloride has the formula

Figure imgf000003_0001

Moxifloxacin Hydrochloride

Moxifloxacin is a fluoroquinolone broad spectrum antibacterial particularly against Gram-positive bacteria significantly better than those of Sparfloxacin and Ciprofloxacin that was disclosed in EP No 350,733 and EP No 550,903. Moxifloxacin has activity against Gram- negative and Gram-positive organisms, including Streptococcus pneumonia, Staphylococcus aureus, Pseudomonas aeruginosa, particularly against the respiratory disease-causing pathogens like Mycoplasma pneumonia, Mycobacterium tuberculosis, Chlamydia pneumoniae and the activity shown to be unaffected by B-lactamases .

US Patent No 5,157,117 discloses (l-cyclopropyl-6, 7-difluoro-8-methoxy- 4-oxo-l, 4-dihydro-3-quinoline carboxylic acid-O3, 04)bis (acyloxy-O) borate and process for its preparation by reacting ethyl-1- cyclopropyl-6, 7-difluoro-8-methoxy-4-oxo-l, -dihydro-3-quinoline carboxylate with Boric acid and acetic anhydride in presence of zinc chloride and its conversion to Gatifloxacin hydrochloride.

WO 2005/012285 discloses the process for the preparation of moxifloxacin hydrochloride using a novel intermediate namely (4aS-Cis)-(1-cyclopropyl-7-(2,8-diazabicyclo[4,3,0]non-8-yl)-6-fluoro-8-methoxy-4-oxo-1,4-dihydro-3-quinoline carboxylic acid-O3,O4)bis(acycloxy-O)borate.

Hydrates of Moxifloxacin hydrochloride known are the anhydrous and monohydrate. US Patent No. 5,849,752 discloses the monohydrate of Moxifloxacin hydrochloride and its preparation by treating the anhydrous crystalline form with ethanol/ water mixtures.

The prior art disclosed in European Patent No’s EP 350,733, EP 550,903 and EP 657,448 discloses the preparation of Moxifloxacin hydrochloride involving the condensation of l-cyclopropyl-6, 7-difluoro-8-methoxy-4- oxo-1, 4-dihydro-3-quinoline carboxylic acid or its esters with (S,S) 2,8-Diaza bicyclo [4.3.0] nonane in presence of a base and its conversion to hydrochloride at higher temperatures leading to the desired Moxifloxacin along with its positional isomer namely (4aS-Cis)-l- cyclopropyl-6- (2, 8-diazabicyclo [4.3.0] non-8-yl) -7-fluoro-8-methoxy-4- oxo-1, 4-dihydro-3-quinoline carboxylic acid as a major impurity. As the impurity and the Moxifloxacin are positional isomers they are difficult to separate. Purification of Moxifloxacin to remove this isomer results in lower yields thereby increasing the product cost. Similarly methods described in the prior art involves the preparation of Moxifloxacin and then its conversion to its hydrochloride thereby incorporating an additional step in the manufacturing process also leading to lowering of yields.

Moxifloxacin and its pharmacologically acceptable salts are disclosed in European patents EP 350733, EP 550903 and EP 657,448. The disclosed process for the preparation of moxifloxacin hydrochloride comprises of condensing l-cyclopropyl-6,7- difluoro-8-methoxy-4-oxo-l,4-dihydro-3-quinoline carboxylic acid or its esters with (S,S)2,8-diazobicyclo[4.3.0]nonane, in presence of a base at high temperature followed by conversion into hydrochloride salt .

This process not only produces desired moxifloxacin hydrochloride but also its positional isomer namely l-cyclopropyl-7-fluoro- 1,4- dihydro -8- methoxy -6- (4aS,7aS)- octahydro- 6H -pyrrolo [3,4-b] pyridine-6-yl] -4- oxo-quinolinecarboxylic acid as a major impurity which is difficult to separate. The purification of moxifloaxcin to remove this isomer results in lower yields thereby increasing the product cost.

The International publication WO 2005/012285 discloses an improved process for the preparation of moxifloxacin hydrochloride incorporated herein by reference. The disclosed process involves the preparation of moxifloxacin hydrochloride from the ethyl 1 -cyclopropyl-6,7-difluoro-8-methoxy-4-oxo- 1 ,4-dihydro-3-quinolme carboxylate through a novel intermediate (4aS-cis)-l-cyclopropyl-7-(2,8-diazabicyclo[4.3.0]non-8- yl)-6-fluoro-8-methoxy-4-oxo- 1 ,4-dihydro-3 -quinolinecarboxylicacid-03,04)bis(acyloxy -0)-borate.

US patent application 6897315 discloses a process for the preparation of 8-methoxy-3-quinoline carboxylic acid especially moxifloxacin incorporated herein by reference. The disclosed process involves the preparation of moxifloxacin from 8-halo moxifloxacin derivative using methanol and potassium tertiary butoxide. US patent 5639886 discloses one-pot process for the preparation of 3-quinoline carboxylic acid derivatives including moxifloxacin.

WO 2004 091619 claims anhydrous crystalline form-Ill of moxifloxacin hydrochloride and WO 2004/039804 claims amorphous form of moxifloxacin hydrochloride. US Pat.No.5, 849,752 discloses specific crystalline forms of anhydrous moxifloxacin mono hydrochloride and monohydrated moxifloxacin mono hydrochloride. Anhydrous moxifloxacin mono hydrochloride disclosed in US Pat. No.5, 849,752 has been designated as “Form-I” and the hydrated form as “Form-II” in US Pat. No.7,230,006. It also discloses a novel crystalline Form-Ill of anhydrous moxifloxacin mono hydrochloride.

US patent US 5,480,879 discloses the melting range of moxifloxacin in example part as 203-208°C and does not speaks about polymorphism of moxifloxacin. Experiment executed as per the procedure given in example Zl 9 of US 5,480,879 and resulted in acetonitrile solvated form of moxifloxacin with low purity and the obtained solvated form can not used for formulations

U.S. Pat. No. 5,849, 752 (“the ‘752 patent”), incorporated by reference, described two crystalline forms of moxifloxacin hydrochloride namely, anhydrous moxifloxacin hydrochloride and monohydrated moxifloxacin hydrochloride. For convenience, the anhydrous crystalline form described in the 752 patent is designated as “Form I”, and the hydrated form as “Form II”. According to U.S. Pat. No. ‘752’, moxifloxacin hydrochloride monohydrate Form II was obtained by stirring a suspension of the anhydrous moxifloxacin hydrochloride in aqueous media until hydration. Moxifloxacin hydrochloride monohydrate of ‘752’ was also prepared by crystallizing moxifloxacin hydrochloride from a media having a water content which is stoichiometrically sufficient but limited to 10%.

WO patent application publication No. 04/091619 disclosed anhydrous Form III of moxifloxacin hydrochloride.

WO patent application publication No. 04/039804 disclosed amorphous form of moxifloxacin hydrochloride.

WO 2005/054240 disclosed two novel crystalline forms which were designated as Form A and Form B of moxifloxacin hydrochloride.

WO patent application publication No. 07/010555 disclosed two crystalline forms which were Form X and Form Y of moxifloxacin hydrochloride. According to WO Publication No. 2007/010555, Form Y was obtained by crystallization of moxifloxacin hydrochloride from the mixture of methanol and water in the ratio of about 8:1 by volume.

WO patent application publication No. 07/148137 disclosed hydrate form of moxifloxacin hydrochloride. According to WO Publication No. 2007/148137, moxifloxacin hydrochloride monohydrate was obtained by crystallization moxifloxacin hydrochloride by humidification of moxifloxacin hydrochloride at 50-90% relative humidity at 25-60° C. for 8 to 24 hours.

WO patent application publication No. 08/028959 disclosed crystalline form of moxifloxacin hydrochloride. According to WO Publication No. 2008/028959, moxifloxacin hydrochloride was obtained by dissolving moxifloxacin hydrochloride in a mixture of methanol and water and adding acetone and recovering moxifloxacin hydrochloride crystalline form.

WO patent application publication No. 08/059521 disclosed process for the preparation of anhydrous crystalline form I of moxifloxacin hydrochloride.

WO patent application publication No. 08/095964 disclosed crystalline form of moxifloxacin base.

……………………

SYNTHESIS

Drugs Fut 1997,22(2),109

36th Intersci Conf Antimicrob Agents Chemother (Sept 15-18, New Orleans) 1996,Abst. F1.

The anhydrization of pyridine-2,3-dicarboxylic acid (I) with acetic anhydride gives the corresponding anhydride (II), which by treatment with benzylamine (III) is converted into the benzylimide (IV). The hydrogenation of (IV) with H2 over Pd/C yields 8-benzyl-2,8-diazabicyclo[4.3.0]nonane-7,9-dione (V), which is further hydrogenated with LiAlH4, affording (?-cis-8-benzyl-2,8-diazabicyclo[4.3.0]nonane (VI) (1). The optical resolution of (VI) by separation of the cis-(R,R)-isomer as crystalline L-(+)-tartrate and further purification of the cis-(S,S)-isomer (VII) as the D-(-)-tartrate affords enantiomerically pure (S,S)-8-benzyl-2,8-diazabicyclo[4.3.0]nonane (VII). The debenzylation of (VII) by hydrogenolysis with H2 over Pd/C gives (S,S)-2,8-diazabicyclo[4.3.0]nonane (VIII), which is condensed with 1-cyclopropyl-6,7-difluoro-8-methoxy-4-oxo-1,4-dihydroquinoline-3-carboxylic acid (IX) in basic medium and finally salified with HCl. The 1-cyclopropyl-6,7-difluoro-8-methoxy-4-oxo-1,4-dihydroquinoline-3-carboxylic acid (IX) has been obtained as follows: The reaction of 2,4,5-trifluoro-3-methoxybenzoyl chloride (X) with malonic acid monoethyl ester monopotassium salt (XI) by means of triethylamine gives 2-(2,4,5-trifluoro-3-methoxybenzoyl)acetic acid ethyl ester (XII), which is condensed with triethyl orthoformate yielding the corresponding ethoxymethylene derivative (XIII). The reaction of (XIII) with cyclopropylamine affords the cyclopropylaminomethylene derivative (XIV), which is finally cyclized to (IX) by means of NaF in DMF.

………………………..

J Label Compd Radiopharm 2000,43(8),795

The condensation of 2,4,5-trifluoro-3-methoxybenzoyl chloride (I) with 14C-labeled diethyl malonate (II) by means of MgCl2 and TEA gives the benzoylmalonate (III), which is monodecarboxylated with TsOH in refluxing water, yielding the benzoylacetate (IV). The reaction of (IV) with triethyl orthoformate and Ac2O at 140 C affords the benzoylacrylate (V), which is treated with cyclopropylamine (VI) in cyclohexane to provide ethyl 3-(cyclopropylamino)-2-(2,4,5-trifluoro-3-methoxybenzoyl)acrylate (VII). The cyclization of (VII) by means of K2CO3 in hot N-methylpyrrolidone gives the quinolone carboxylate (VIII), which is hydrolyzed with NaOH in hot methanol, affording the carboxylic acid (IX). Finally, this compound is condensed with (S,S)-2,8-diazabicyclo[4.3.0]octane (X) by means of 1,4-diazabicyclo[2.2.2]octane (DABCO) in refluxing acetonitrile.

…………………….

……………………

EP1651630A1

The reaction scheme is given below: Stage-I

Acetic anhydride

Figure imgf000006_0002
Figure imgf000006_0001

Ethyl-l-cyclopropyl-6, 7- ( l-cyclopropyl-6, 7-difluoro-l, 4- difluoro-1, 4-dihydro-8- dihydro-8- methoxy-4-oxo-3- methoxy-4-oxo-3-quinoline quinoline carboxylic acid-03, 04) carboxylate . Bis ( acetate-O) -borate (Borate complex)

Stage-II

Triet yl amine

Acetonitrile

Figure imgf000006_0003
Figure imgf000006_0004

l-cyclo propyl-6, 7-difluoro- [S, S] -2, 8-diazabicyclo- (1- cyclo propyl-6, fluoro-7 (2, 8- 1, 4-dihydro-8- methoxy-4-oxo [4 ,3.0]nonane Diazabicyclo-nonane) 1,4- -3-quinoline carboxylic acid- dihydro-8-methoxy-4-oxo-3 03,04)Bis ( acetate-O) -borate- quinoline carboxylic acid- (Borate complex) (03,04) bis (acetate-O) -borate itage-III

Figure imgf000007_0001

(1- cyclo propyl-6, fluoro- (2, 8- Moxifloxacin HCI pseudohydrate Diazabicyclo-nonane) 1,4- dihydro-8-methoxy-4-oxo-3 -quinolinecarboxylicacid- (O^O4) bis (acetate-O) -borate

Stage-TV

Figure imgf000007_0002

Moxifloxacin HCI pseudohydrate Moxifloxacin HCI monohydrate

EXAMPLE – I

Stage-1: Preparation of l-cyclopropyl-6, 7-difluoro-8-methoxy-4-oxo- l,4-dihydro-3-quinoline carboxylic acid-O3,O*)bis (acyloxy-O)borate

Acetic anhydride (175 g) is heated to 70°C and boric acid (30 g) is slowly added lot wise in a temperature range of 70°C to 90°C. The temperature is then raised, maintained under reflux for 1 hr followed by cooling to about 70°C. Ethyl-l-cyclopropyl-6, 7-difluoro-8-methoxy-4- oxo-1, 4-dihydro-3-quinoline carboxylate (100 g) is added under stirring. The temperature is then raised and maintained for 1 hr in the range of 100°C to 105°C. The reaction mass is cooled to 0°C, chilled water (400 ml) is added slowly followed by cold water (600 ml) at temperature 0°C to 5°C and maintained for 2 hrs at 0°C to 5°C. The product which is a boron acetate complex is filtered, washed with water (500 ml) and dried at 55°C to 60°C under vacuum to constant weight. The dry wt is 130.0 g corresponding to yield of 95.2%.

Stage-2: Preparation of (4aS-Cis) -l-Cyclopropyl-7- (2, 8-diazabicyclo [4.3.0]non-8-yl) -6-fluoro-8-methoxy-4-oxo-l , 4-dihydro-3-quinoline carboxylicacid-03,0*)bis (acyloxy-O)borate

The boron acetate complex (130 g) prepared in stage 1 is suspended in acetonitrile (650 ml), and [S, S] -2, 8-diazabicyclo [4.3.0] nonane (47 g) and triethyl amine (72.9 g) are added. The temperature is raised to reflux and maintained for 1 hr. at reflux, followed by cooling to about 40°C. The solvent is removed under vacuum at temperature below 40°C, and n-hexane (200 ml) is added. After maintaining the reaction mass for 1 hr at room temperature the product is isolated by filtration followed by washing of the wet cake with n-hexane . The product is dried at about 45°C to about 50°C to constant weight.

Dry wt of the novel intermediate is 117.0 g corresponding to yield of 71.5%.

Elemental analysis: C: 56.42%, H: 5.62%, N: 7.76% and the calculated values for the intermediate, formula C25H29BFN308C: 56.6%, H: 5.47%, N: 7.92%

IR Spectrum (KBr, cm-1) : 3415, 3332, 2936, 1718, 1630, 1573, 1526, 1445, 1273, 1042, 935, 860, 798, 682

^ NMR (200 MHz, CDC13, ppm) : 9.00 (1H), 7.82 (1H), 4.12 (4H), 3.57 (3H), 3.43 (4H), 3.07 (2H) , 2.75 (2H), 2.4 (1H),’ 2.1 (6H), 1.84 (2H) , 1.6 (1H), 1.31 (2H)

Mass Spectrum (MJ : 530.3 [M+H] , 470.2 [M+ - CH3COOH] , 428.2 [M+- (CH3CO)20, 100%], 402.2, 388.2

Stage -3: Preparation of Moxifloxacin Hydrochloride pseudohydrate

The intermediate (117 g) prepared stage-2 is dissolved in ethanol (600 ml) by stirring for about 30 min. at room temperature and the insolubles if any are filtered off. pH of the filtrate is adjusted to about 0.5 by addition of hydrochloric acid at room temperature and maintained for 2 hrs. The reaction mass is cooled, and maintained for two hrs, at about 0°C to about 5°C. The product is filtered, washed with chilled ethanol (50 ml) and dried at about 50°C to about 55°C till constant weight.

The dry weight of the Moxifloxacin hydrochloride pseudohydrate is 87.5g corresponding to yield of 91.0%. Water content of the product by KF is 0.64% w/w.

X-ray diffraction pattern data are given in Table-1 EXAMPLE – II

Stage- 2 : Preparation of Moxifloxacin pseudohydrate with out isolating (4aS-Cis) -l-Cyclopropyl-7- (2 , 8-diazabicyclo [4.3.0] on-8-yl) -6-fluoro- 8-methoxy-4-oxo-l,4-dihydro-3-quinolinecarboxylicacid-03,04)bis (acyloxy-O) borate

The boron acetate complex (130 g) prepared in stage-1 of Example-1 is suspended in acetonitrile (650ml) and [S, S] -2, 8-Diazabicyclo [4.3.0]nonane (47 g) & triethyl amine (72.9 g) are added. Temperature of the reaction mass is raised to reflux, maintained for 1 hr. at reflux and cooled to room temperature. Methanol (600 ml) is added and maintained for 30 min at room temperature to obtain a clear solution. The solution is filtered to remove insolubles if any and pH of the filtrate is adjusted to about 0.5 with hydrochloric acid (57.5 g) . The reaction mass is maintained for 2 hrs at temperature in the range of about 20°C to about 25°C, cooled to 0°C followed by maintaining the reaction mass at about 0°C to about 5°C for 2 hrs. The product is filtered, washed with methanol (50 ml) and dried at about 50°C to 55°C until constant weight.

Dry wt of the Moxifloxacin hydrochloride pseudohydrate is 88g corresponding to yield of 68.7%.

EXAMPLE – III : Preparation of Moxifloxacin Hydrochloride monohydrate

Moxifloxacin hydrochloride (50 g) prepared as above is suspended in a mixture of ethanol (250 ml) and hydrochloric acid (25 ml) . Raised the temperature, maintained for two hrs at 40°C to 45°C followed by cooling to about 25°C. The product is filtered and dried under vacuum at 50-55°C until become constant weight.

Dry wt of Moxifloxacin hydrochloride monohydrate is 46 g corresponding to yield of 90.5%.

The IR spectral data and XRD pattern are identical with available Moxifloxacin hydrochloride monohydrate.

Figure imgf000014_0001
……………………….
Synthesis

WO2008059521A2

The preferred embodiments of the present invention is represented as follows

Figure imgf000020_0001

Formula-2a Formula-2b

Figure imgf000020_0002

Formula-3a Formula-3b Formula-3c Formula-3d

Figure imgf000020_0003

Formula-4b

Figure imgf000020_0004

Formula-4c

Figure imgf000020_0005

SCHEME-5:

Figure imgf000015_0003

SCHEME-6:

Figure imgf000019_0001

 Example-1: Preparation of (S,S)-2-benzyl-8-trityI-2,8-diazabicyclo (4.3.0) nonane:

A mixture of (S5S)-2-benzyl-2,8-diazabicyclo-(4.3.0) nonane 50 grams, dichloromethane 300 ml and triethylamine 28 ml was stirred at 25-35°C. Trityl chloride 71 grams was added to the reaction mixture. The reaction mixture was stirred at 25-350C for 5 hrs. The organic layer was washed with 5% sodium bicarbonate solution followed by water. The organic layer was distilled off completely to provide the title compound as a residue.

Yield: 96 grams

Example-2:

Preparation of (S,S)-8-trityI-2,8-diazabicyclo (4.3.0) nonane:

A mixture of (S,S)-2-benzyl-8-trityl-2,8-diazabicyclo (4.3.0) nonane 130 grams,

2- butanol 1600 ml and 5% Pd/C was taken in an autoclave and heated to 45-50°C at 4.0-4.5 Kg/cm2 of hydrogen pressure. The reaction mixture was stirred at this condition for 45-50 hrs. The reaction mixture was cooled to 25-300C and filtered through hyflow bed. The solvent was distilled off completely to provide the title compound as a residue.

Yield: 95 grams.

Example-3:

Preparation of condensed compouad of formula-4a:

A mixture of 20 grams of l-cyclopropyl-NjN-diethyl-όjT-difluoro-l^-dihydro-δ- methoxy-4-oxoquinoline-3-carboxamide, 11.8 grams of (S,S)2,8-diazabicyclo (4.3.0) nonane, 100 ml of acetonitrile and 2 grams of l,8-Diazabicyclo[5.4.0]undec-7-ene (DBU) was heated to 800C. The reaction mixture was stirred for 35 hours at 8O 0C. The reaction mixture was cooled to 320C and stirred for 45 minutes at 32°C. The solvent was distilled off under reduced pressure. Water (100 ml) was added to the obtained residue. The reaction mixture was cooled to 25-350C. The reaction mixture was extracted with ethyl acetate. The solvent was distilled off to get the title compound as a residue. Yield: 12 grams.

Example-4:

Preparation of condensed compound of formula-4b:

A mixture of 5 grams of l-cyclopropyl-NjN-diethyl-όJ-difluoro-M-dihydro-δ- methoxy-4-oxoquinoline-3-carboxamide, 2.9 grams of (S,S)-8-phenyloxy carbonyl-2,8- diazabicyclo (4.3.0) nonane, 25 ml of acetonitrile and 0.5 grams of diazabicyclo[5.4.0]undec-7-ene (DBU) was heated to 800C. The reaction mixture was stirred for 35 hours at 75-8O0C. The reaction mixture was cooled to 25-35°C. The reaction mixture was stirred for 45 minutes at 25-35°C. The solvent was distilled off under reduced pressure. Water (100 ml) was added to the obtained residue. The reaction mixture was cooled to 25-35°C. The reaction mixture was extracted with ethyl acetate. The solvent was distilled off to get the title compound as a residue. Yield: 1 gram.

Example-5: Preparation of condensed compound of formula-4c:

A mixture of 5 grams of l-cyclopropyl-NjN-diethyl-βJ-difluoro-l^-dihydro-S- methoxy-4-oxoquinoline-3-carboxamide, 2.9 grams of (S,S)-8-tertiary butyloxycarbonyl- 2,8-diazabicyclo (4.3.0) nonane, 25 ml of acetonitrile and 0.5 grams of diazabicyclo[5.4.0]undec-7-ene (DBU) was heated to 800C. The reaction mixture was stirred for 35 hours at 75-800C. The reaction mixture was cooled to 25-350C and stirred for 45 minutes at 25-35°C. The solvent was distilled off under reduced pressure. Water (100 ml) was added to the obtained residue. The reaction mixture was cooled to 25-350C. The reaction mixture was extracted with ethyl acetate. The solvent was distilled off to get the title compound as a residue Yield: lgram. Example-6:

Preparation of condensed compound of formula-4d:

A mixture of 20 grams of l-cyclopropyl-6,7-difluoro-8-methoxy-l,4-dihydro-4- oxoquinoline-3-carboxamide, 2.9 grams of (S,S)-2,8-diazabicyclo (4.3.0) nonane , 100 ml of acetonitrile and 2 grams of diazabicyclo[5.4.0]undec-7-ene (DBU) was heated to 75-80°C. The reaction mixture was stirred for 35 hours at 75-80°C. The reaction mixture was cooled to 25-350C. The reaction mixture was stirred for 45 minutes at 25-350C. The solid obtained was filtered and washed with acetonitrile. The material was dried at 40-450C to get the title compound. Yield: 12 grams; M.R: 210-2120C.

Example-7:

Preparation of condensed compound of formula-4e:

A mixture of (S,S)-8-trityl-2,8-diazabicyclo (4.3.0) nonane (42 grams), l-cyclopropyl-N,N-diethyl-6,7-difluoro-l,4-dihydro-8-methoxy-4-oxoquinoline-3- carboxamide (10 grams), potassium carbonate ( 7.9 grams) and dimethyl formamide (80 ml) was heated to 110-1200C and stirred for 20 hours at 110-1200C. The reaction mixture was cooled to 800C. The solvent was distilled off under reduced pressure. Water (100 ml) was added to the obtained residue. The reaction mixture was cooled to 25-35°C. The reaction mixture was extracted with ethyl acetate. The solvent was distilled off to obtain the title compound as a residue. Yield: 14.5 grams

Example-8: Preparation of moxifloxacin from 4d:

A mixture of 165 ml of water, 35 grams of sodium hydroxide, 150 ml of ethylene glycol and 12 grams of condensed compound of formula-4d was heated to 115°C. The reaction mixture was stirred for 15 hours at 115°C. The reaction mixture was cooled to 5°C. The pH of the reaction mixture was adjusted to 5.5 using hydrochloric acid and stirred for 30 minutes at 5°C. The obtained solid was filtered and washed with water. The solid was dried at 45°C to get the title compound. Yield: 8 grams; M.R: 203-2050C Example-9:

Preparation of moxifloxacin from 4b:

Moxifloxacin can be prepared from 4b (3grams), by a method which is analogous to the method illustrated in Example-8 Yield: 0.85 grams; M.R: 203-205°C

Example-10:

Preparation of moxifloxacin from 4c:

Moxifloxacin can be prepared from 4c (3 grams), by a method which is analogous to the method illustrated in Example-8 Yield: 1.0 grams; M.R: 203-205°C

Example-11:

Preparation of moxifloxacin from 4a: Moxifloxacin can be prepared from 4a (10 grams), by a method which is analogous to the method illustrated in Example-8 Yield: 7.5 grams; M.R: 203-2050C

Example-12: Preparation of moxifloxacin from 4e:

The condensed compound of formula-4e (14.5 grams) was dissolved in ethyl acetate 100ml. Aqueous hydrochloric acid (5 ml in 20 ml of water) was added to the above reaction mixture. The reaction mixture was stirred at 25-30°C for 30-45 min. 20ml of water was added to the reaction mixture. The two layers were separated. The aqueous layer was washed twice with ethyl acetate. The pH of the aqueous layer was adjusted to 10.8 using sodium hydroxide solution. The reaction mixture was extracted with methylene chloride. The solvent distilled off to get a residue. The residue was suspended in 165 ml of water, 35 grams of sodium hydroxide; 150 ml of ethylene glycol was heated to 115°C. The reaction mixture was stirred for 15 hours at 115°C. The reaction mixture was cooled to 5°C. The pH of the reaction mixture was adjusted to 5.5 using hydrochloric acid and stirred for 30 minutes at 5°C.The obtained solid was filtered and washed with water and dried at 450C to get the title compound. Yield: 4.5 grams; M.R: 203-2050C

Example-13:

Preparation of moxifloxacin hydrochloride compound of formula-1:

A mixture of 8 grams of moxifloxacin, 16 ml of water and 64 ml of methanol was heated to reflux temperature of 700C. The reaction mixture was filtered to remove undissolved material. Filtrate was cooled to 35°C. The pH of the filtrate was adjusted to 1.6 using hydrochloric acid. The reaction mixture was cooled to 150C. The reaction mixture was stirred for 45 minutes at 150C. The solid obtained was filtered and washed with methanol and dried at 40-450C to get crystalline Form-Y of moxifloxacin hydrochloride monohydrate. Yield: 5.5 grams

Example- 14:

Preparation of moxifloxacin hydrochloride monohydrate:

A mixture of 25 grams of moxifloxacin, 16 ml of water and 64 ml of methanol was heated to reflux temperature of 700C. The reaction mixture was filtered to remove undissolved material. Filtrate was cooled to 35°C. The pH of the filtrate was adjusted to 1.6 using hydrochloric acid. The reaction mixture was cooled to 150C. The reaction mixture was stirred for 45 minutes at 150C. The obtained solid was taken into a mixture of 22 ml of water and 0.5ml of hydrochloric acid and stirred 30 min at 5°C. The compound was filtered and washed with water, dried at 40-450C to get moxifloxacin hydrochloride monohydrate particles having oval shape. Yield: 20 grams

Example-15:

Preparation of anhydrous crystalline Form-I of moxifloxacin hydrochloride: Moxifloxacin hydrochloride (Form-Y, 10 grams) suspended in 50 ml of methanol and the reaction mixture was heated to 45-5O0C. 18ml of dichloromethane was added slowly to the reaction mixture. The reaction mixture was stirred at 45-50°C for 15-20 minutes. The reaction mixture was cooled slowly to around 0-5°C and stirred for 1-1.5 hours. The precipitated solid was filtered under nitrogen atmosphere and washed with 5 ml of methanol. The solid obtained was dried at 110-120°C till the moisture content reached below 0.5%. Yield: 8.0 grams.

Bulk density: 0.28 g/ml; Tapped density: 0.52 g/ml Particle size distribution : D( v,0.1) :1.6 μm ; D( vs0.5) : 5.5 μm ; D( v,0.9) :25.0 μm

……………………….
SYNTHESIS

 Preparation of Moxifloxacin

A solution of lOOg of 1-Cyclopropyl -6,7difluoro-l,4-dihydro-4-oxo-methoxyquinolone- 3-carboxilic acid), 52gr of (S,S)2,8 diazabicyclo[4,3,0]nonane, lOgr of 1,8- diazabycyclo[5,4,0]undec-7-ene and 300ml of acetonitrile is heated to 65 -70°C temperature and stirred till the reaction get completed and evaporated the solvent. Added 5 %aqueous isopropylalcohol to the reaction mass and adjusted the pH to basic with caustic solution and then filtered the reaction mass. Combined the total filtrate and adjusted the pH to 5.0 to 6.0 with aqueous HCl.and isolated the separated solid at a temperature of 10-150C to afford Moxifloxacin.

………………….

SYNTHESIS REVIEW

http://www.beilstein-journals.org/bjoc/single/articleFullText.htm?publicId=1860-5397-9-265

Beilstein J. Org. Chem. 2013, 9, 2265–2319.

While isolated pyridines are a vital component in numerous drugs the related quinoline/quinolone scaffolds are becoming increasingly common. One such example is nalidixic acid (1.101, in fact a naphthyridone, Figure 2). Nalidixic acid is a prototype quinolone antibiotic that has been used extensively as an effective treatment against both gram positive and gram negative bacteria. In general, quinolone antibiotics act by interfering with the enzymes DNA-gyrase and/or topoisomerase of bacteria [55]. Moxifloxacin (1.102, Avelox) and levofloxacin (1.103, Levaquin) are two third-generation fluoroquinolone antibiotics which also appear amongst the top selling drugs. These compounds display similar SAR data [56]. For instance, in the case of moxifloxacin the fluorine atom in the C6 position enhances microbial activity while a methoxy group in the C8 position is reported to increase potency and decrease toxicity. Furthermore, it was found that a cyclopropyl group was beneficial for the enzyme–DNA binding complex while the bulky nitrogen-based appendage at C7 helps to bind to DNA gyrase and hinders the efflux of the drug from the bacterial cell.

[1860-5397-9-265-2]
Figure 2: Structures of nalidixic acid, levofloxacin and moxifloxacin.

Due to the elaborate substitution pattern of the parent quinolone ring systems these compounds are usually prepared via a linear consecutive sequence. In the case of moxifloxacin, an intramolecular base catalysed nucleophilic aromatic substitution is used to prepare the bicyclic ring system of the highly substituted aromatic1.104 (Scheme 19). A SNAr reaction is then used to introduce the saturated piperidinopyrrolidine appendage 1.105to furnish the desired structure [57-60]. In order to obtain a high yield for the substitution reaction a one-pot procedure was developed, initial masking of the acid (1.104) is achieved by silylation with subsequent borane chelate formation. Addition of the amine nucleophile 1.105 under basic conditions then renders the desired product in high yield. The available patent literature however does not comment on regioselectivity issues of the SNAr reaction due to the presence of the second fluoride substituent in the substrate, although not necessarily as electronically favourable for displacement it is certainly more accessible.

[1860-5397-9-265-i19]
Scheme 19: Synthesis of moxifloxacin.

The saturated (S,S)-2,8-diazabicyclo[4.3.0]nonane (1.105) used in the final step can be prepared by a double nucleophilic substitution between tosylamine and 2,3-bis-chloromethylpyridine (1.112) followed by catalytic reduction of the resulting bicycle using palladium on carbon in acetic acid (Scheme 20). As the corresponding sulfonamide 1.113 was found to be a crystalline solid a resolution using (D)-(+)-O,O-dibenzoyltartaric acid was reported to separate the enantiomers [Petersen, U.; Schenke, T.; Krebs, A.; Schenke, T.; Philipps, T.; Grohe, K.; Bremm, K.-D.; Endermann, R.; Metzger, K. G. New Quinoline and Naphthyridinonecarboxylic Acid Derivatives. Ger. Patent DE 4 208 792 A1, March 23, 1993.].

[1860-5397-9-265-i20]
Scheme 20: Synthesis of (S,S)-2,8-diazabicyclo[4.3.0]nonane 1.105.
…………………………………
PAPER
First way of enantioselective synthesis of moxifloxacin intermediate
LI GuangXun, WU Lei, FU QingQuan, TANG Zhuo, ZHANG XiaoMei

A new method of enantioselective synthesis of (S,S)-2,8-diazobicyclo [4.3.0] nonane was found by using (R)-2-amino-2-phenyl-ethanol as chiral induction reagent. The entire synthetic process included 8 steps which were easy to operate with high yield. The purification method was only simple recrystallization or even used directly in the next step without further purification. The total yield was 29%.

2013 Vol. 56 (3): 307-311 [Abstract] ( 22 ) [ PDF (518 KB)   ] ( 92 )  [Supporting Information]  DOI10.1007/s11426-012-4803-7
  1. Drugwatch. “Avelox Patent Family”. Retrieved 16 September 2012.
  2.  “Details for NDA:021085″. DrugPatentWatch. Retrieved 17 July 2009.
  3.  “www.accessdata.fda.gov”.
  4.  “www.emea.europa.eu”.
  5. http://www.accessdata.fda.gov/drugsatfda_docs/label/2011/021085s047,021277s041lbl.pdf
  6.  “Avelox”The American Society of Health-System Pharmacists. Retrieved 3 April 2011.
  7. http://www.accessdata.fda.gov/drugsatfda_docs/appletter/1999/21085ltr.pdf
  8. http://www.accessdata.fda.gov/drugsatfda_docs/nda/2001/21-085S010_Avelox_Approv.pdf.
  9.  http://www.accessdata.fda.gov/drugsatfda_docs/appletter/2004/21085se1-022,21277se1-017ltr.pdf.
  10. http://www.accessdata.fda.gov/drugsatfda_docs/appletter/2005/021085s026,021277s022ltr.pdf.
  11. http://www.accessdata.fda.gov/drugsatfda_docs/appletter/2005/021085s027,029,021277s024,025ltr.pdf.
  12.  “Moxifloxacin: restricted use : MHRA”.
  13.  European Medicines Agency (24 July 2008). “European Medicines Agency recommends restricting the use of oral moxifloxacin-containing medicines”. Retrieved 20 July 2009.
  14.  “SYNOPSIS”. Retrieved 29 January 2009.
  15.  Karande SC, Kshirsagar NA (February 1992). “Adverse drug reaction monitoring of ciprofloxacin in pediatric practice”. Indian Pediatr 29 (2): 181–8. PMID 1592498.
  16.  Dolui SK, Das M, Hazra A (2007). “Ofloxacin-induced reversible arthropathy in a child”. Journal of Postgraduate Medicine 53 (2): 144–5. doi:10.4103/0022-3859.32220 .PMID 17495385.
  17.  “Center for drug evaluation and research Application number 21-598″Food and Drug Administration (FDA). 15 April 2005. Retrieved 21 July 2009.
  18. Jump up^ Babar, S. (October 2013). “SIADH Associated With Ciprofloxacin.”The Annals of Pharmacotherapy (Sage Publiahing) 47 (10): 1359–1363.doi:10.1177/1060028013502457 . ISSN 1060-0280.PMID 24259701. Retrieved November 139,2013.
  19.  Renata Albrecht (28 July 2004). “NDA 21-085/S-024, NDA 21-277/S-019″Center for Drug Evaluation and ResearchFood and Drug Administration (FDA). Retrieved 31 July 2009.
  20.  Renata Albrecht (31 May 2007). “NDA 21-085/S-036, NDA 21-277/S-030″Center for Drug Evaluation and ResearchFood and Drug Administration (FDA). Retrieved 31 July 2009.
  21.  Renata Albrecht (15 February 2008). “NDA 21-085/S-038, NDA 21-277/S-031″Division of Special Pathogen and Transplant ProductsFood and Drug Administration (FDA). Retrieved 31 July 2009.
  22.  DEPARTMENT OF HEALTH & HUMAN SERVICES (28 February 2008). “NDA 21-085/S-014, S-015, S-017″Food and Drug Administration (FDA). Retrieved 17 July 2009.
  23.  Nea Zealand Government (26 June 2003). “Adverse Reaction Reporting and IMMP”. New Zealand: Medsafe. Retrieved 30 January 2009.
  24. http://www.mhra.gov.uk/Safetyinformation/DrugSafetyUpdate/CON087781, retrieved 2012-11-10 Missing or empty|title= (help)
  25. http://www.akdae.de/Arzneimittelsicherheit/RHB/Archiv/2009/20090119.pdf, retrieved 2012-11-10 Missing or empty|title= (help)
  26.  “EU agency recommends restricting moxifloxacin use”. Reuters. 24 July 2008. Retrieved 21 July 2009.
  27.  Renata Albrecht (3 October 2008). “NDA 21-085/S-040, NDA 21-277/S-034″Center for Drug Evaluation and ResearchFood and Drug Administration (FDA). Retrieved 31 July 2009.
  28.  Updated Labeling for Antibiotic Avelox (Moxifloxacin) Regarding Risk of Severe Liver Injury / Information Update: 2010-42 For immediate release 22 March 2010http://www.hc-sc.gc.ca/ahc-asc/media/advisories-avis/_2010/2010_42-eng.php
  29.  Saint F, Gueguen G, Biserte J, Fontaine C, Mazeman E (September 2000). “[Rupture of the patellar ligament one month after treatment with fluoroquinolone]“Rev Chir Orthop Reparatrice Appar Mot (in French) 86 (5): 495–7.PMID 10970974.
  30. http://www.akdae.de/Arzneimittelsicherheit/RHB/Archiv/2009/20090119.pdf, retrieved 2012-11-10 Missing or empty|title= (help)
  31.  Bayer (December 2008). “AVELOX (moxifloxacin hydrochloride) Tablets AVELOX I.V. (moxifloxacin hydrochloride in sodium chloride injection)”Food and Drug Administration (FDA). p. 19. Retrieved 2 November 2010. Unknown parameter |line= ignored (help)
  32.  “Moxifloxacin”. University of Maryland Medical Center. 2009. Retrieved 22 July 2009.
  33. http://www.akdae.de/Arzneimittelsicherheit/RHB/Archiv/2009/20090119.pdf
  34.  Shin HC, Kim JC, Chung MK (September 2003). “Fetal and maternal tissue distribution of the new fluoroquinolone DW-116 in pregnant rats”. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 136 (1): 95–102.doi:10.1016/j.cca.2003.08.004 . PMID 14522602.
  35.  Dan M, Weidekamm E, Sagiv R, Portmann R, Zakut H (February 1993). “Penetration of fleroxacin into breast milk and pharmacokinetics in lactating women” (PDF).Antimicrob. Agents Chemother. 37 (2): 293–6.doi:10.1128/AAC.37.2.293 . PMC 187655.PMID 8452360.
  36.  Noel GJ, Bradley JS, Kauffman RE (October 2007). “Comparative safety profile of levofloxacin in 2523 children with a focus on four specific musculoskeletal disorders”.Pediatr. Infect. Dis. J. 26 (10): 879–91.doi:10.1097/INF.0b013e3180cbd382 . PMID 17901792.
  37.  Joette Meyer; Renata Albrecht (16 March 2004).“Division of Special Pathogen and Immunologic Drug Products Summary of Clinical Review of Studies Submitted in Response to a Pediatric Written Request”Food and Drug Administration (FDA). Retrieved 22 July 2009.
  38.  Elbe DH, Chang SW (February 2005). “Moxifloxacin-warfarin interaction: a series of five case reports”Ann Pharmacother 39 (2): 361–4. doi:10.1345/aph.1E179 .PMID 15632222.
  39.  Karen E. Brown. “Top Ten Dangerous Drug Interactions in Long-Term Care”. Medication Management Project. Retrieved 1 August 2009.
  40.  Stass HH (29 June – 3 July 1997). “Bay 12-8039 (BA) does not interact with theophylline (TH)”. Presented at the 20th International Congress of Chemotherapy.
  41.  Medscape: Medscape Access
  42.  http://69.20.19.211/cder/warn/dec99/wl121599.pdf
  43.  Renata Albrecht (16 May 2002). “NDA 21-085/S-012″.Food and Drug Administration (FDA). Retrieved 17 July 2009.
  44.  Drlica K, Zhao X (1 September 1997). “DNA gyrase, topoisomerase IV, and the 4-quinolones”Microbiol Mol Biol Rev. 61 (3): 377–92. PMC 232616PMID 9293187.
  45.  “Drug card for Moxifloxacin (DB00218)”. Canada: DrugBank. 19 February 2009. Retrieved 3 August 2009.
  46. Alffenaar J. W. C., van Altena R., Bökkerink H. J (2009). “Pharmacokinetics of moxifloxacin in cerebrospinal fluid and plasma in patients with tuberculous meningitis”. Clinical Infectious Diseases 49 (7): 1080–2. doi:10.1086/605576 .PMID 19712035.
  47.  “Inventors/Applicants”. patentlens.net. 3 October 2006. Retrieved 17 July 2009.
  48.  Ed Lamb (1 May 2008). “Top 200 Prescription Drugs of 2007″. Pharmacy Times. Retrieved 21 July 2009.
  49. http://www.uspto.gov/web/offices/pac/dapp/opla/term/certs/4990517.pdf
  50.  US 4990517 A (Feb 1991) Petersen et al. Seehttp://www.patentlens.net/patentlens/search_ajax.cgi?patnum=US+4990517#list US 5051509 A (Sep 1991) Nagano et al. Seehttp://www.patentlens.net/patentlens/search_ajax.cgi?patnum=US+5051509#list US 5059597 A (Oct 1991) Petersen et al. Seehttp://www.patentlens.net/patentlens/search_ajax.cgi?patnum=US+5059597#list US 5395944 A (Mar 1995) Petersen et al. Seehttp://www.patentlens.net/patentlens/search_ajax.cgi?patnum=US+5395944#list US 5416096 A (May 1995) Petersen et al. Seehttp://www.patentlens.net/patentlens/search_ajax.cgi?patnum=US+5416096#list
  51.  Renata Albrecht (12 June 2002). “NDA 21-085/S-006, S-007 – NDA 21-277/S-002″Food and Drug Administration(FDA). Retrieved 12 August 2009.
  52.  The 114Th Medicines Adverse Reactions Committee Meeting; Professor P. Ellis, Professor D.C.G. Skegg, Dr M. Rademaker, Dr F. McClure, Dr N. Rafter, Dr M. Tatley (26 June 2003). “Adverse Reaction Reporting and IMMP”. New Zealand: Medsafe. Retrieved 12 August 2009.
  53.  Renata Albrecht, (6 October 2003). “NDA 21-085/S-019 – NDA 21-277/S-011″Food and Drug Administration(FDA). Retrieved 12 August 2009.
  54.  Renata Albrecht (6 October 2003). “NDA 21-085/S-039 NDA 21-277/S-033″Food and Drug Administration(FDA). Retrieved 12 August 2009.
  55.  Bayer HealthCare Pharmaceuticals Inc. (August 2008).“MEDICATION GUIDE AVELOX (AV-eh-locks) (moxifloxacin hydrochloride) Tablets AVELOX I.V. (AV-eh-locks) (moxifloxacin hydrochloride in sodium chloride injection)”Food and Drug Administration (FDA). Retrieved 12 August 2009.
  56.  Ozlem Belen (24 June 2009). “NDA 21-085/S-041 NDA 21-277/S-035″Food and Drug Administration (FDA). Retrieved 12 August 2009.
  57.  Bailey RR, Natale R, Linton AL (October 1972). “Nalidixic acid arthralgia”Can Med Assoc J 107 (7): 604 passim.PMC 1940945PMID 4541768.
  58.  Bailey RR, Kirk JA, Peddie BA (July 1983). “Norfloxacin-induced rheumatic disease”. N Z Med J 96 (736): 590.PMID 6223241.
  59.  Szarfman A, Chen M, Blum MD (January 1995). “More on fluoroquinolone antibiotics and tendon rupture” (PDF). N Engl J Med 332 (3): 193.doi:10.1056/NEJM199501193320319 . PMID 7800023.
  60.  “Petition to Require a Warning on All Fluoroquinolone Antibiotics (HRG Publication #1399)”Public Citizen. 1 August 1996. Retrieved on 27 December 2008.
  61.  “Reports of adverse events with fluoroquinolones”FDA Medical Bulletin 26 (3). October 1996.[dead link] Retrieved on 27 December 2008.
  62.  “Madigan, Public Citizen, petition FDA for “black box” warning regarding potential adverse effects of certain popular antibiotics” (Press release). Office of the Illinois Attorney General. 29 August 2006. Retrieved 27 December 2008.
  63.  “Public Citizen Petitions the FDA to Include a Black Box Warning on Fluoroquinolone Antibiotics (HRG Publication #1781)”. Public Citizen. 29 August 2006. Retrieved 27 December 2008.
  64.  “Public Citizen v. Food and Drug Administration (FDA) (Fluoroquinolone)”. Public Citizen. 3 January 2008. Retrieved 27 December 2008.
  65.  Ravn, Karen (18 August 2008). “Behind the FDA’s ‘black box’ warnings”Los Angeles Times. Retrieved 27 December 2008.
  66.  “FDA Requests Boxed Warnings on Fluoroquinolone Antimicrobial Drugs” (Press release). U.S. Food and Drug Administration. 8 July 2008. Retrieved 11 October 2008.
  67.  “Drugs@FDA”Food and Drug Administration (FDA). Retrieved 12 August 2009.
  68.  “MedWatch: The FDA Safety Information and Adverse Event Reporting Program”Food and Drug Administration (FDA). Retrieved 12 August 2009.
  69.  MacCarthy, Paul (22 October 2008). “Important Change in the Avelox (moxifloxacin hydrochloride) and Cipro (ciprofloxacin) Complete Prescribing Information – Addition of Boxed Warning and Medication Guide Regarding Tendinitis and Tendon Rupture”. Bayer HealthCare Pharmaceuticals. Retrieved 27 December 2008.
  70.  Bayer AG (6 November 2007). “Ruling in Bayer’s favor over Avelox patents”. Bayer. Retrieved 29 August 2009.
  71. http://www.orangebookblog.com/Bayer_20v._20Dr._20Reddy_27s.pdf
  72.  “United States Court of Appeals for the Federal Circuit”. uscourts.gov. 28 June 2007. Retrieved 29 August 2009.
  73.  Bayer AG (24 April 2008). “Risk Report”. Bayer. Retrieved 29 August 2009.
  74.  In The United States District Court For The District Of Columbia Public Citizen, Inc. VS. Food And Drug Administration 3 January 2008
  75.  Office of the Attorney General State of Illinois Lisa Madigan Citizen Petition to Include a Black Box Warning on Fluoroquinolone Antibiotics 18 May 2005
  76.  Public Citizen’s Petition to Include a Black Box Warning on Fluoroquinolone Antibiotics (HRG Publication #1781) 29 August 2006
  77.  Public Citizen’s Petition to Require a Warning on All Fluoroquinolone Antibiotics (HRG Publication #1399) 1 August 1996
  78.  Public Citizen’s Petition to Ban the Antibiotic Gatifloxacin (Tequin) (HRG Publication #1768)
  79. Public Citizen’s Petition to immediately ban the antibiotic Trovafloxacin (Trovan). (HRG Publication #1485) Date: 3 June 1999
  80.  Public Citizen’s Petition to immediately stop the distribution of dangerous, misleading prescription drug information to the public. HRG Publication #1442 Date: 9 June 1998
  81.  June 2004, A petition To the United States Congress to immediately take action to protect consumers from the reckless and negligent abuses of the FDA and the following Pharmaceutical Companies: Bayer, Ortho-McNeill, Pfizer, Merck, Bristol-Myers Squibb, Sanofi Winthrop, Bertek Pharmaceuticals – Rhone-Poulenc Rorer and Barr. These companies manufacture and distribute fluoroquinolone antibiotics in the United States in a manner that fails to warn of serious adverse event risks, and downplays and fails to warn physicians of the serious risks associated with fluoroquinolone therapy.
EP0350733A2 Jun 30, 1989 Jan 17, 1990 Bayer Ag 7-(1-Pyrrolidinyl)-3-quinolone- and -naphthyridone-carboxylic-acid derivatives, method for their preparation and for substituted mono- and bi-cyclic pyrrolidine intermediates, and their antibacterial and feed additive compositions
EP0550903A1 Dec 28, 1992 Jul 14, 1993 Bayer Ag Quinolone- and naphthyridone carboxylic acid derivatives as antibacterial agents
EP0591808A1 Sep 27, 1993 Apr 13, 1994 Bayer Ag Quinolonecarboxylic acids
EP0592868A1 Sep 29, 1993 Apr 20, 1994 Bayer Ag Quinolonecarboxylic acids
US5849752 Dec 5, 1996 Dec 15, 1998 Bayer Aktiengesellschaft Crystal modification of CDCH a process for its preparation and pharmaceutical formulations comprising this modification
WO1999026940A2 Nov 12, 1998 Jun 3, 1999 Bayer Ag Method for producing 8-methoxy-quinoline carboxylic acids
WO2004091619A1 Apr 9, 2004 Oct 28, 2004 Reddys Lab Ltd Dr A crystalline form iii of anhydrous moxifloxacin hydrochloride and a process for preparation thereof
WO2007010555A2 Jul 13, 2006 Jan 25, 2007 Ramprasad Achampeta Kodanda Novel crystalline forms of moxifloxacin hydrochloride and process for preparation thereof
WO2008059521A2 Sep 27, 2007 May 22, 2008 Ramprasad Achampeta Kodanda Novel process for the preparation of moxifloxacin hydrochloride and a novel polymorph of moxifloxacin
CN102030751B Dec 1, 2010 Nov 21, 2012 上虞京新药业有限公司 Process for crystallizing moxifloxacin hydrochloride
EP2154137A1 Aug 4, 2008 Feb 17, 2010 Chemo Ibérica, S.A. Crystalline form of moxifloxacin base
US20110212990 * Nov 6, 2008 Sep 1, 2011 Hetero Research Foundation Novel polymorph of moxifloxacin hydrochloride
WO2005012285A1 * Aug 5, 2004 Feb 10, 2005 Satyanarayana Chava An improved process for the preparation of moxifloxacin hydrochloride
EP0443498A1 * Feb 18, 1991 Aug 28, 1991 Kyorin Pharmaceutical Co., Ltd. Isoindoline derivatives
EP0550903A1 * Dec 28, 1992 Jul 14, 1993 Bayer Ag Quinolone- and naphthyridone carboxylic acid derivatives as antibacterial agents
US6323213 * Feb 12, 1997 Nov 27, 2001 Bayer Aktiengesellschaft Possibly substituted 8-cyano-1-cyclopropyl-7-(2,8-diazabicyclo-[4.3.0]-nonan-8-yl)-6-fluoro-1,4-dihydro-4-oxo-3-quinolin carboxylic acids and their derivatives
US5849752 * Dec 5, 1996 Dec 15, 1998 Bayer Aktiengesellschaft Crystal modification of CDCH a process for its preparation and pharmaceutical formulations comprising this modification
WO2007010555A2 * Jul 13, 2006 Jan 25, 2007 Ramprasad Achampeta Kodanda Novel crystalline forms of moxifloxacin hydrochloride and process for preparation thereof
WO2008059521A2 * Sep 27, 2007 May 22, 2008 Ramprasad Achampeta Kodanda Novel process for the preparation of moxifloxacin hydrochloride and a novel polymorph of moxifloxacin
WO2008138759A1 * Apr 30, 2008 Nov 20, 2008 Sandoz Ag Process for the preparation of moxifloxacin hydrochloride
WO2007148137A1 * Jun 22, 2007 Dec 27, 2007 Generics Uk Ltd Novel hydrate form of moxifloxacin monohydrochloride
WO2008028959A1 * Sep 7, 2007 Mar 13, 2008 Sint Quimica Sa Crystalline form of moxifloxacin hydrochloride
WO2008059223A2 * Nov 13, 2007 May 22, 2008 Cipla Ltd Process for the synthesis of moxifloxacin hydrochloride
WO2008095964A1 * Feb 6, 2008 Aug 14, 2008 Sint Quimica Sa Crystalline form of moxifloxacin base
WO2009087151A1 * Jan 7, 2009 Jul 16, 2009 Chemo Iberica Sa Polymorphic forms of moxifloxacin hydrochloride and processes for preparation thereof
CN102603738B Feb 24, 2012 Dec 11, 2013 天津市汉康医药生物技术有限公司 一种稳定的盐酸莫西沙星化合物
EP2083010A1 Jan 8, 2008 Jul 29, 2009 Chemo Ibérica, S.A. Polymorphic Forms of Moxifloxacin hydrochloride and processes for preparation thereof
EP2154137A1 Aug 4, 2008 Feb 17, 2010 Chemo Ibérica, S.A. Crystalline form of moxifloxacin base
US8198451 Nov 13, 2007 Jun 12, 2012 Cipla Limited Process for the synthesis of moxifloxacin hydrochloride
US20110212990 * Nov 6, 2008 Sep 1, 2011 Hetero Research Foundation Novel polymorph of moxifloxacin hydrochloride


Filed under: GENERIC DRUG, Uncategorized Tagged: MOXIFLOXACIN

FDA Breakthrough Therapy Designation: Fourth Drug Receives FDA Approval

$
0
0

Originally posted on Orphan Druganaut Blog:

.

The FDA approves on February 12, the fourth drug to have the coveted Breakthrough Therapy Designation (BTD). The approval is for orphan drug Imbruvica (Ibrutinib) as a single agent for the treatment of patients with Chronic Lymphocytic Leukemia (CLL) who have received at least one prior therapy. Imbruvica, a once-daily, oral kinase inhibitor, is developed and commercialized by Pharmacyclics and Janssen Biotech. Imbruvica is the :

•   1st FDA BTD drug to receive approval in 2014

•   1st FDA BTD drug to receive approval for a 2nd indication – Mantle Cell Lymphoma (MCL) on 11.13.13

View original


Filed under: Uncategorized

Sonidegib/Erismodegib..Novartis Cancer Drug LDE225 Meets Primary Endpoint in Phase 2

$
0
0

Sonidegib/Erismodegib

CODE DESIGNATION ..LDE225, NVP-LDE-225

Treatment of medulloblastoma PHASE3 2014 FDA FILING

Treatment of advanced basal cell carcinoma PHASE3 2014 FDA FILING

Treatment of SOLID TUMORS..PHASE1 2017 FDA FILING

READMalignant Solid Tumors of Childhood

THERAPEUTIC CLAIM Oncology, Antineoplastics & Adjunctive Therapies

CHEMICAL NAMES

1. [1,1'-Biphenyl]-3-carboxamide, N-[6-[(2R,6S)-2,6-dimethyl-4-morpholinyl]-3-pyridinyl]-2-
methyl-4′-(trifluoromethoxy)-, rel-

2. N-{6-[(2R,6S)-2,6-dimethylmorpholin-4-yl]pyridin-3-yl}-2-methyl-4′-
(trifluoromethoxy)biphenyl-3-carboxamide

N-[6-[(2S,6R)-2,6-dimethylmorpholin-4-yl]pyridin-3-yl]-2-methyl-3-[4-(trifluoromethoxy)phenyl]benzamide

N-(6-((2S,6R)-2,6-dimethylmorpholino)pyridin-3-yl)-2-methyl-4′-(trifluoromethoxy)biphenyl-3-carboxamide

MOLECULAR FORMULA C26H26F3N3O3

MOLECULAR WEIGHT 485.5

SPONSOR Novartis Pharma AG

CAS REGISTRY NUMBER 956697-53-3  free form

NOTE… DIPHOSPHATE SALT IS THE DRUG WITH CAS 1218778-77-8

sonidegib - European Medicines Agency READ THIS..

Summary EudraCT Number: 2012-004022-21 Sponsor’s Protocol  READ THIS

Novartis announced that the pivotal trial of the investigational oral compound LDE225 (sonidegib) in advanced basal cell carcinoma met its primary endpoint of demonstrating an objective response rate among patients within six months of treatment. Objective response included complete response (clinically significant tumor response with complete absence of disease) and partial response (clinically significant tumor shrinkage).
Basal cell carcinoma is the most common form of skin cancer, accounting for more than 80% of non-melanoma skin cancers, and can be highly disfiguring and life-threatening if it grows. Worldwide incidence of basal cell carcinoma is rising by 10% each year due to factors such as an aging population and increased ultraviolet exposure. Although basal cell carcinoma rarely metastasizes, once it does, it can be associated with significant morbidity.
“For people living with advanced basal cell carcinoma there are currently limited treatment options,” said Alessandro Riva, president, Novartis Oncology ad interim and global head, Oncology Development and Medical Affairs. “These results demonstrate the potential for LDE225 to offer a treatment option for this patient population, and we look forward to sharing these data with regulatory authorities worldwide.”
Full study results will be presented at a future scientific meeting.

About the Study

The Phase II, randomized, double-blind BOLT (Basal cell carcinoma Outcomes in LDE225 Trial) study was designed to assess the safety and efficacy of two oral dose levels of LDE225 (200 mg and 800 mg) in patients with locally advanced or metastatic basal cell carcinoma[4], which are subtypes of advanced basal cell carcinoma.

The primary endpoint was the proportion of patients achieving an objective response rate, defined as a confirmed complete response and partial response as their best overall response per modified RECIST criteria, within six months of starting treatment with LDE225. Key secondary endpoints of the study included assessing the duration of tumor responseand the rate of complete response. Other secondary endpoints included progression-free survival, time to tumor response and overall surviva

Date: February 19, 2013
Source: Novartis
Links
MORE ABOUT SONIDEGIB

Sonidegib (INN) or Erismodegib (USAN), also known as LDE225 is a Hedgehog signalling pathway inhibitor (via smoothened antagonism) being developed as an anticancer agent by Novartis.[1][2] It has been investigated as a potential treatment for:

NVP-LDE-225, a product candidate developed by Novartis, is in phase III clinical trials for the treatment of medulloblastoma and basal cell carcinoma. Phase II trials are in progress for the treatment of adult patients with relapsed or refractory or untreated elderly patients with acute leukemia.

Early clinical trials are ongoing for the oral treatment of advanced solid tumors, for the treatment of myelofibrosis in combination with ruxolitinib and for the treatment of small cell lung cancer. A phase II clinical trial for the treatment of basal cell carcinomas in Gorlin’s syndrome patients with a cream formulation of NVP-LDE-225 was discontinued in 2011 since the formulation did not demonstrate tumor clearance rate sufficient to support further development.

Dana-Farber Cancer Institute and the Massachusetts General Hospital are conducting phase I clinical trials for the treatment of locally advanced or metastatic pancreatic cancer in combination with chemotherapy. In 2009, orphan drug designation was assigned in the E.U. for the treatment of Gorlin syndrome.

It has demonstrated significant efficacy against melanoma in vitro and in vivo.[21] It also demonstrated efficacy in a mouse model of pancreatic cancer.[22]

NVP-LDE225 Diphosphate salt (Erismodegib, Sonidegib) 

Formula Image

Synonym:Erismodegib, Sonidegib
CAS Number:1218778-77-8
Mol. Formula:C26H26F3N3O3 ∙ 2H3PO4
MW:681.5
nmr.http://www.chemietek.com/Files/Line2/Chemietek,%20NVP-LDE225%20[02],%20NMR.pdf
hplc–http://www.chemietek.com/Files/Line3/Chemietek,%20NVP-LDE225%20[02],%20HPLC.pdf

Brief Description:

A potent, selective, and orally bioavailable Smoothened (Hedgehog Signaling Pathway) antagonist, currently in clinical trials. Diphosphate salt offers a much better bioavailability than free base (Ref. a)
a. Pan, S., et al, Discovery of NVP-LDE225, a Potent and Selective Smoothened Antagonist, ACS Med. Chem. Lett., 2010, 1 (3), pp 130–134.

About LDE225

LDE225 (sonidegib) is an oral, investigational, selective smoothened inhibitor being studied in a variety of cancers. Smoothened (SMO) is a molecule that regulates the hedgehog (Hh) signaling pathway, which plays a critical role in stem cell maintenance and tissue repair. LDE225 is currently in clinical development for a variety of diseases including myelofibrosis, leukemia and solid tumors.

Given that LDE225 is an investigational compound, the safety and efficacy profile has not yet been fully established. Access to this investigational compound is available only through carefully controlled and monitored clinical trials. These trials are designed to better understand the potential benefits and risks of the compound. Given the uncertainty of clinical trials, there is no guarantee that LDE225 will ever be commercially available anywhere in the world.

Possibility (LDE225) is effective in medulloblastoma relapsed or refractory hedgehog pathway inhibitor sonidegib has been revealed. That the anti-tumor effect was observed in some patients and tolerability in 1/2 test phase.

4th Quadrennial Meeting of the World Federation of Neuro-Oncology in conjunction with the 18th Annual Meeting of the Society for Neuro-Oncology, which was held in San Francisco November 21 to 24 in (WFNO-SNO2013), rice Dana-Farber It was announced by Mark Kieran Mr. Children’s Hospital Cancer Center.

The research group, announced the final results of the Phase 1 trial that target advanced solid cancer in children of sonidegib.  1 dose increased multi-test phase, was initiated from 372mg/m2 once-daily dosing to target children under the age of 18 more than 12 months. (233mg/m2 group 11 people, 16 people 372mg/m2 group, 11 people group 425mg/m2, 680mg/m2 group 21 women) who participated 59 people, including medulloblastoma 38 patients. 12 median age was (2-17).

Creatine phosphokinase elevation of grade 4 only were seen at 372mg/m2 as dose-limiting toxicity only, and became two recommended dose phase and 680mg/m2.  Nausea muscle pain creatine kinase rise malaise (22.0%) (15.3%) (15.3%), (13.6%), vomiting side effects were many, was (13.6%). Hypersensitivity vomiting creatine kinase increased (3.4%) (1.7%) (1.7%), rhabdomyolysis side effects of grade 3/4 was (1.7%).  (One group 372mg/m2, 425mg/m2 group one) complete response was obtained in two people, a strong correlation was found between the activation of the hedgehog pathway and effect.

Phase III clinical trials that target medulloblastoma the activated hedgehog pathway currently are underway.

About Novartis

Novartis provides innovative healthcare solutions that address the evolving needs of patients and societies. Headquartered in Basel, Switzerland, Novartis offers a diversified portfolio to best meet these needs: innovative medicines, eye care, cost-saving generic pharmaceuticals, preventive vaccines and diagnostic tools, over-the-counter and animal health products. Novartis is the only global company with leading positions in these areas. In 2013, the Group achieved net sales of USD 57.9 billion, while R&D throughout the Group amounted to approximately USD 9.9 billion (USD 9.6 billion excluding impairment and amortization charges). Novartis Group companies employ approximately 136,000 full-time-equivalent associates and operate in more than 140 countries around the world.

Increased levels of Hedgehog signaling are sufficient to initiate cancer formation and are required for tumor survival.
These cancers include, but are not limited to, prostate cancer (“Hedgehog signalling in prostate regeneration, neoplasia and metastasis”, Karhadkar S S, Bova G S, Abdallah N, Dhara S, Gardner D, Maitra A, Isaacs J T, Berman D M, Beachy P A., Nature. 2004 Oct. 7; 431(7009):707-12;
“Inhibition of prostate cancer proliferation by interference with SONIC HEDGEHOG-GLI1 signaling”, Sanchez P, Hernandez A M, Stecca B, Kahler A J, DeGueme A M, Barrett A, Beyna M, Datta M W, Datta S, Ruiz i Altaba A., Proc Natl Acad Sci USA. 2004 Aug. 24; 101(34):12561-6),
breast cancer (“Hedgehog signaling pathway is a new therapeutic target for patients with breast cancer”, Kubo M, Nakamura M, Tasaki A, Yamanaka N, Nakashima H, Nomura M, Kuroki S, Katano M., Cancer Res. 2004 Sep. 1; 64(17):6071-4),
medulloblastoma (“Medulloblastoma growth inhibition by hedgehog pathway blockade”, Berman D M, Karhadkar S S, Hallahan A R, Pritchard J I, Eberhart C G, Watkins D N, Chen J K, Cooper M K, Taipale J, Olson J M, Beachy P A., Science. 2002 Aug. 30; 297(5586):1559-61),
basal cell carcinoma (“Identification of a small molecule inhibitor of the hedgehog signaling pathway: effects on basal cell carcinoma-like lesions”, Williams J A, Guicherit O M, Zaharian B I, Xu Y, Chai L, Wichterle H, Kon C, Gatchalian C, Porter J A, Rubin L L, Wang F Y., Proc Natl Acad Sci USA. 2003 Apr. 15; 100(8):4616-21;
“Activating Smoothened mutations in sporadic basal-cell carcinoma”, Xie J, Murone M, Luoh S M, Ryan A, Gu Q, Zhang C, Bonifas J M, Lam C W, Hynes M, Goddard A, Rosenthal A, Epstein E H Jr, de Sauvage F J., Nature. 1998 Jan. 1; 391(6662):90-2),
pancreatic cancer (“Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis”, Thayer S P, di Magliano M P, Heiser P W, Nielsen C M, Roberts D J, Lauwers G Y, Qi Y P, Gysin S, Fernandez-del Castillo C, Yajnik V, Antoniu B, McMahon M, Warshaw A L, Hebrok M., Nature. 2003 Oct. 23; 425(6960):851-6;
“Widespread requirement for Hedgehog ligand stimulation in growth of digestive tract tumours”, Berman D M, Karhadkar S S, Maitra A, Montes De Oca R, Gerstenblith M R, Briggs K, Parker A R, Shimada Y, Eshleman J R, Watkins D N, Beachy P A., Nature. 2003 Oct. 23; 425(6960):846-51),
and small-cell lung cancer (“Hedgehog signalling within airway epithelial progenitors and in small-cell lung cancer”, Watkins D N, Berman D M, Burkholder S G, Wang B, Beachy P A, Baylin S B., Nature. 2003 Mar. 20; 422(6929):313-7).
Links
PATENTS
2 WO 2008154259
3 WO 2010033481
4 WO 2011009852
5 WO 2011062939
………………………………………
Links
SYNTHESIS
2-Methyl-4′-tr{fluoromethoxy-biphenyl-3-carboxylic acid {6-(cis-2,6-dimethyl- morpholin-4-yl)-pyrid»n-3-yl|-amide:
Figure imgf000003_0001

The following Examples serve to illustrate the invention without limiting the scope thereof, it is understood that the invention is not limited to the embodiments set forth herein, but embraces ali such forms thereof as come within the scope of the disclosure,

Figure imgf000013_0001

Step 1:

To a solution of 2-chloro-5-nitro-pyridine 1 (5.58 g, 35.2 mmoL) and c/s-2,6- dimethylmorpholine (4.05 g, 35.2 mmoL) in anhydrous DMF (30 mi.) was added K2CO3 (9.71 g, 70.4 mnrtoL). The mixture was heated at 50ºC overnight. After concentration, the residue is partitioned between EtOAc and water. The EtOAc layer is dried over anhydrous Na2SO4 and concentrated to give crude product 3 as a yellow solid, after purification by silica gel chromatography, obtained pure product (7.80 g, 93.2%). LC-MS m/z: 238.2 (M+ 1).

Step 2:

The above material 3 (7.3Og. 30.8 mmoL) was hydrogenated in the presence of 10% Pd-C (1.0 g) in MeOH (120 ml) under hydrogen overnight. The suspension was filtered through celite and the filtrate was concentrated to give the crude product 4 (5.92 g) as a dark brown oil which was used directly in the next step without further purification. LC-MS m/z. 208.2 (M+1).

Step 3:

To a solution of 3-bromo-2-methyl benzoic acid (2.71 g, 12.6 mmoL), 6-((2S,6R)-2,6- dimethylmorpholino)pyridin-3-arnine 4 (2.61 g, 12.6 mmoL), and HATU (4.80 g, 12.6 mmoL) in anhydrous DMF (30 mL) was added diisopropylethylamine (6.58 mL, 37.8 mmoL) dropwise. The resulting mixture was stirred overnight at room temperature. The reaction mixture was diluted with water (50 mL), and then extracted with EtOAc (3×120 mL). The organic layer was dried and concentrated to give the crude product. This crude product was then purified by flash column chromatography using 30% EtOAc in hexane as eiuent to give 5 as a white solid (4.23 g, 83.0%). LC-MS m/z: 404.1 (M+1).

Step 4:

A mixture of 4-(trif!uoromethoxy)phenylboronic acid (254 mg, 1.24 mmol), 3-bromo- N-[6-(2,6-dimethyl-morpholin-4-yl)-pyridin-3-ylJ-4-methyl-benzamide 5 (250 mg, 0.62mmol), Pd(PPh3)4 (36 mg, 0.03 mmol), Na2CO3 (2.0M aqueous solution, 1.23 mL, 2.4 mmol) and DME (4.5 mL) in a sealed tube was heated at 130ºC overnight. The reaction mixture was diluted with EtOAc and water. The aqueous layer was extracted with EtOAc. The combined organic layer was washed with brine and concentrated to give the crude product which was then purified by preparative mass triggered HPLC (C18 column, etuted with CH3CN-H2O containing 0.05% TFA) to give N-(6-((2S,6R)-2,6-dimethyfmorpholino)pyridin-3-yl)-2-rnethyl- 4'-(trifluoromethoxy)biphenyi-3-carboxamide (183.5 mg, 61.1% yield). LC-MS m/z: 486.2 (M+1).

The resultant crystalline product (Form A) was converted to the amorphous form by dissolving in 3% w/w aqueous ethanol, and the resultant solution spray dried at about 150ºC.

Form B was prepared by heating the amorphous form in an oven at 110ºC for 2 hours. In a further embodiment, the invention relates to a process step or steps, or an intermediate as described herein.

........................
Links
PAPER
ChemMedChem, 2013 ,  vol. 8,   8  p. 1261 - 1265
Thumbnail image of graphical abstract
Continued optimization provided a novel type of Smoothened (Smo) antagonist based on a pyridazine core. The compound, NVP-LEQ506, currently in phase I clinical trials, combines high intrinsic potency and good pharmacokinetic properties resulting in excellent efficacy in rodent tumor models of medulloblastoma. Activity against a Smo mutant conferring resistance observed in a previous clinical trial with a competitor compound suggests additional therapeutic potential.

................................

Links

SYNTHESIS

US20120196849,  ENTRY.....95
Figure US20120196849A1-20120802-C00097
LC-MS m/z 486.2 (M + 1)
USE SIMILAR METHODOLOGY
EXAMPLESThe present invention is further exemplified, but not limited, by the following example that illustrates the preparation of compounds of Formula I according to the invention.Example 1 4′-cyano-6-methyl-biphenyl-3-carboxylic acid [4-(morpholine-4-sulfonyl)-phenyl]-amide
Figure US20120196849A1-20120802-C00003

Step 1: To a solution of 3-iodo-4-methyl-benzoic acid (10.0 g, 38.2 mmol) in methanol (70 ml) is added concentrated sulfuric acid (0.5 ml). The reaction mixture is heated at 70° C. for 48 hours, cooled to room ambient temperature and then concentrated. After that, ethyl acetate (100 ml) and aqueous NaHCO(saturated, 100 ml) solution are added to the residue. The organic layer is separated and washed again with aqueous NaHCO(saturated, 100 ml) solution. The organic layer is separated, dried over anhydrous Na2SOand concentrated to yield 3-iodo-4-methyl-benzoic acid methyl ester 1. It is used without further purification in the next step. 1H NMR (400 MHz, DMSO-d6) δ 8.31 (s, 1H), 7.87 (d, 1H, J=8.4 Hz), 7.48 (d, 1H, J=8.4 Hz), 3.85 (s, 3H), 3.35 (s, 3H); LC-MS m/z: 277.0 (M+1).

Step 2: To a round-bottom flask containing 3-iodo-4-methyl-benzoic acid methyl ester (1.38 g, 5.00 mmol), 4-cyanophenylboronic acid (1.10 g, 7.48 mmol), palladium acetate (168 mg, 0.748 mmol), 2-(dicyclohexylphosphino)biphenyl (0.526 g, 1.50 mmol) and potassium fluoride (0.870 g, 15.0 mmol) is added anhydrous 1,4-dioxane (15 ml). The flask is purged with argon and sealed. The mixture is stirred at 130° C. for 18 hours, cooled to ambient temperature and then water (20 ml) and ethyl acetate (20 ml) are added. Solid is removed under vacuum filtration. The filtrate is extracted with EtOAc (20 ml×2). The organic layers are combined, washed with aqueous HCl (5%, 20 ml) and saturated NaHCO(20 ml). It is dried over MgSO4, and concentrated. The residue is purified by silica gel column chromatography (EtOAc/Hexane, gradient) to give 4′-cyano-6-methyl-biphenyl-3-carboxylic acid methyl ester 2; LC-MS m/z: 252.1 (M+1).

Step 3: To a solution of 4′-cyano-6-methyl-biphenyl-3-carboxylic acid methyl ester 2 (2.56 g, 10.3 mmol) in 1,4-dioxane-H2O (1:1 mixture, 20 ml) is added NaOH (1.22 g, 30.2 mmol)). The reaction is stirred at ambient temperature for 24 hours. To this mixture is added aqueous HCl (1 N, 36 ml) and it is then extracted with ethyl acetate (40 ml×3). The organic layers are combined, dried over anhydrous Na2SO4. The solver is removed. The solid obtained is washed with small amount of acetonitrile and air dried to give 4′-cyano-6-methyl-biphenyl-3-carboxylic acid 3: 1H NMR (DMSO-d6) δ 7.94 (d, 2H, J=8.0 Hz), 7.84 (dd, 1H, J1=8.4 Hz, J2=1.2 Hz), 7.75 (d, 1H, J=1.2 Hz), 7.61 (d, 2H, J=8.0 Hz), 7.48 (d, 1H, J=8.4 Hz), 2.29 (s, 3 H); LC-MS m/z 238.1 (M+1).

Step 4: To a suspension of 4′-cyano-6-methyl-biphenyl-3-carboxylic acid 3 (40 mg, 0.17 mmol) in anhydrous methylene chloride (5 ml) is added 2 drops of DMF. Then oxalyl chloride (32 mg, 22 μl, 0.25 mmol) is added. The mixture is stirred at ambient temperature until it turns clear. After that, it is concentrated, re-dissolved in anhydrous methylene chloride (3 ml), and added to a solution of 4-(morpholine-4-sulfonyl)-phenylamine (61 mg, 0.25 mmol) and triethylamine (34 mg, 47 μl, 0.33 mmol) in methylene chloride (2 ml). The mixture is stirred for 2 hours, concentrated and the residue is purified by preparative mass triggered HPLC (C18 column, eluted with CH3CN—H2O containing 0.05% TFA) to give 4′-cyano-6-methyl-biphenyl-3-carboxylic acid [4-(morpholine-4-sulfonyl)-phenyl]-amide: 1H NMR (DMSO-d6) δ 10.64 (s, 1H), 8.07 (d, 2H, J=8.8 Hz), 7.97 (d, 2H, J=8.4 Hz), 7.95 (d, 1H, J=8.8 Hz), 7.89 (s, 1H), 7.43 (d, 2H, J=8.4 Hz), 7.67 (d, 2H, J=8.8 Hz), 7.53 (d, 2H, J=8.8 Hz), 3.63 (m, 4H), 2.84 (m, 4H) 2.32 (s, 3H); LC-MS m/z: 462.1 (M+1).

Example 2 4′-cyano-6-methyl-biphenyl-3-carboxylic acid [6-(2,6-dimethyl-morpholin-4-yl)-pyridin-3-yl]-amide

Figure US20120196849A1-20120802-C00004

Step 1: To a solution of 2-chloro-5-nitro-pyridine 4 (2.38 g, 15 mmol.) and cis-2,6-dimethylmorpholine (1.73 g, 15 mmol.) is added K2CO(4.14 g, 30 mmol.). The mixture was heated at 50° C. overnight. After concentration, the residue is partitioned between EtOAc and water. The EtOAc layer is dried over anhydrous Na2SOand concentrated to give crude product 6 as a yellow solid. The crude product is used directly in next step without further purification. LC-MS m/z: 238.1 (M+1).

Step 2: The above crude material 6 is hydrogenated in the presence of Pd—C (0.2 g) in MeOH (100 mL) under hydrogen over 10 h. The suspension is filtered through celite and the filtrate is concentrated to give the crude product 7 as a dark brown oil which is used directly in the next step without further purification. LC-MS m/z: 208.1 (M+1).

Step 3: To a solution of 3-bromo-4-methyl benzoic acid (108 mg, 0.5 mmol.), 6-(2,6-Dimethyl-morpholin-4-yl)-pyridin-3-ylamine 7 (104 mg, 0.5 mmol.), amd HATU (190 mg, 0.5 mmol.) in dry DMF (5 mL) is added triethylamine (139 uL, 1.0 mmol.) dropwise. The resulting mixture is stirred at room temperature for 2 h. After concentration, the residue is partitioned between EtOAc and water. The organic layer is dried and concentrated to give the crude product. The final compound is purified by flash column chromatography using 50% EtOAc in hexane as eluent to give 8 as a white solid. LC-MS m/z: 404.1 (M+1).

Step 4: A mixture of 4-cyanophenyl boronic acid (18 mg, 0.12 mmol), 3-bromo-N-[6-(2,6-dimethyl-morpholin-4-yl)-pyridin-3-yl]-4-methyl-benzamide 8 (40 mg, 0.1 mmol), Pd(PPh3)(11 mg, 0.01 mmol), and Na2CO(42 mg, 0.4 mmol) in a combined solvent system of toluene (0.2 mL) and water (0.2 mL) and ethanol (0.05 mL) is heated at 140° C. under microwave irradiation for 30 min. The reaction mixture is diluted with EtOAc and water. The aqueous layer is extracted with EtOAc. The combined organic layer is washed with brine and concentrated to give the crude product which is purified by preparative mass triggered HPLC (C18 column, eluted with CH3CN—H2O containing 0.05% TFA) to give 4′-cyano-6-methyl-biphenyl-3-carboxylic acid [6-(2,6-dimethyl-morpholin-4-yl)-pyridin-3-yl]-amide. LC-MS m/z: 427.2 (M+1).

USE THIS COMPD IN ABOPVE  AND YOU WILL GET SONIDEGIB

4-(Trifluoromethoxy)phenylboronic acid

  • CAS Number 139301-27-2 
  • Linear Formula CF3OC6H4B(OH)2 
  • Molecular Weight 205.93

CONDENSE WITH …3-bromo-N-[6-(2,6-dimethyl-morpholin-4-yl)-pyridin-3-yl]-4-methyl-benzamideACS Medicinal Chemistry Letters, 2010 ,  vol. 1,   3  p. 130 – 134

……………………………………………….
Links
PAPER
ACS Medicinal Chemistry Letters, 2010 ,  vol. 1,   3  p. 130 – 134
Figure
ENTRY 5m

A mixture of 4-(trifluoromethoxy)phenylboronic acid (254 mg, 1.24 mmol), 3-bromo-N-[6-(2,6-
dimethyl-morpholin-4-yl)-pyridin-3-yl]-4-methyl-benzamide E (250 mg, 0.62mmol), Pd(PPh3)4
(36 mg, 0.03 mmol), Na2CO3 (2.0M aqueous solution, 1.23 mL, 2.4 mmol) and DME (4.5 mL)
in a sealed tube was heated at 1300C overnight. The reaction mixture was diluted with EtOAc
and water. The aqueous layer was extracted with EtOAc. The combined organic layer was
washed with brine and concentrated to give the crude product which was then purified by
preparative mass triggered HPLC (C18 column, eluted with CH3CN-H2O containing 0.05% TFA)
to give N-(6-((2S,6R)-2,6-dimethylmorpholino)pyridin-3-yl)-2-methyl-4′-
(trifluoromethoxy)biphenyl-3-carboxamide (5m, 183.5 mg, 61.1% yield). LC-MS m/z: 486.2 (M+1).
HRMS (m/z): [M+H]+
calcd for C26H27N3O3F3 486.2005; found 486.1986,
1H-NMR (500 MHz, DMSO-d6): δ (ppm) 10.15 (s, 1H), 8.43 (d, 1H), 7.94 (dd, 1H), 7.52-7.43
(m, 5H), 7.38 (m, 1H), 7.33 (m, 1H), 6.86 (d, 1H), 4.06 (d, 2H), 3.62 (m, 2H), 2,34 (m, 2H), 2.22
(s, 3H), 1.16 (d, 6H).

http://pubs.acs.org/doi/suppl/10.1021/ml1000307/suppl_file/ml1000307_si_001.pdf

Links

Reference

  1.  “LDE225 – PubChem”PubChem. National Institutes of Health. Retrieved 16 February 2014.
  2.  Pan, S; Wu, X; Jiang, J; Gao, W; Wan, Y; Cheng, D; Han, D; Liu, J; Englund, NP; Wang, Y; Peukert, S; Miller-Moslin, K; Yuan, J; Guo, R; Matsumoto, M; Vattay, A; Jiang, Y; Tsao, J; Sun, F; Pferdekamper, AC; Dodd, S; Tuntland, T; Maniara, W; Kelleher, JF; Yao, Y; Warmuth, M; Williams, J; Dorsch, M (10 June 2010). “Discovery of NVP-LDE225, a Potent and Selective Smoothened Antagonist”. ACS Medicinal Chemistry Letters 1 (3): 130–134. doi:10.1021/ml1000307.
  3.  “A Biomarker Study to Identify Predictive Signatures of Response to LDE225 (Hedgehog Inhibitor) In Patients With Resectable Pancreatic Cancer”ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  4.  “Gemcitabine + Nab-paclitaxel With LDE-225 (Hedgehog Inhibitor) as Neoadjuvant Therapy for Pancreatic Adenocarcinoma”.ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  5.  “Dose-escalation, and Safety Study of LDE225 and Gemcitabine in Locally Advanced or Metastatic Pancreatic Cancer Patients”.ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  6.  “A Pilot Study of a Hedgehog Pathway Inhibitor (LDE-225) in Surgically Resectable Pancreas Cancer”ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  7.  “Study With LDE225 in Combination With Docetaxel in Triple Negative (TN) Advanced Breast Cancer (ABC) Patients (EDALINE)”.ClinicalTrials.gov. National Institutes of Health. 13 February 2014.
  8.  “LDE225 in Treating Patients With Stage II-III Estrogen Receptor- and HER2-Negative Breast Cancer”ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  9.  “A Phase II Study of Efficacy and Safety in Patients With Locally Advanced or Metastatic Basal Cell Carcinoma (BOLT)”.ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  10.  “To Evaluate the Safety, Local Tolerability, PK and PD of LDE225 on Sporadic Superficial and Nodular Skin Basal Cell Carcinomas(sBCC)”ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  11.  “A Trial to Evaluate the Safety, Local Tolerability, Pharmacokinetics and Pharmacodynamics of LDE225 on Skin Basal Cell Carcinomas in Gorlin Syndrome Patients”ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  12.  “Combination of the Hedgehog Inhibitor, LDE225, With Etoposide and Cisplatin in the First-Line Treatment of Patients With Extensive Stage Small Cell Lung Cancer (ES-SCLC)”ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  13.  “A Phase III Study of Oral LDE225 Versus (vs) Temozolomide (TMZ) in Patients With Hedge-Hog (Hh)-Pathway Activated Relapsed Medulloblastoma (MB)”ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  14.  “A Phase I Dose Finding and Safety Study of Oral LDE225 in Children and a Phase II Portion to Assess Preliminary Efficacy in Recurrent or Refractory MB”ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  15.  “Phase Ib, Dose Escalation Study of Oral LDE225 in Combination With BKM120 in Patients With Advanced Solid Tumors”.ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  16.  “Dose Finding and Safety of Oral LDE225 in Patients With Advanced Solid Tumors”ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  17.  “LDE225 and Paclitaxel in Solid Tumors”ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  18.  “Study of Efficacy and Safety of LDE225 in Adult Patients With Relapsed/Refractory Acute Leukemia”ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  19.  “Nilotinib and LDE225 in the Treatment of Chronic or Accelerated Phase Myeloid Leukemia in Patients Who Developed Resistance to Prior Therapy”ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  20.  “A Phase Ib/II Dose-finding Study to Assess the Safety and Efficacy of LDE225 + INC424 in Patients With MF”ClinicalTrials.gov. National Institutes of Health. 13 February 2014. Retrieved 16 February 2014.
  21.  Jalili, A; Mertz, KD; Romanov, J; Wagner, C; Kalthoff, F; Stuetz, A; Pathria, G; Gschaider, M; Stingl, G; Wagner, SN (30 July 2013). “NVP-LDE225, a potent and selective SMOOTHENED antagonist reduces melanoma growth in vitro and in vivo.” (PDF). PloS one 8 (7): e69064. doi:10.1371/journal.pone.0069064PMC 3728309.PMID 23935925.
  22.  Fendrich, V; Wiese, D; Waldmann, J; Lauth, M; Heverhagen, AE; Rehm, J; Bartsch, DK (November 2011). “Hedgehog inhibition with the orally bioavailable Smo antagonist LDE225 represses tumor growth and prolongs survival in a transgenic mouse model of islet cell neoplasms.”. Annals of Surgery 254 (5): 818–23.doi:10.1097/SLA.0b013e318236bc0fPMID 22042473.
  23. ChemMedChem, 2013 ,  vol. 8,   8  p. 1261 – 1265
  24. ACS Med. Chem. Lett., 2010, 1 (3), pp 130–134.
  25. MORE REF

sonidegib

Skin Cancer Foundation. “Skin Cancer Facts.” Available at:http://www.skincancer.org/skin-cancer-information/skin-cancer-facts . Accessed on February 14, 2014.

Rubin AI, Chen EH, Ratner D (2005). Current Concepts: Basal-Cell Carcinoma. N Engl J Med; 353:2262-9.

ClinicalTrials.gov. “A Phase II Study of Efficacy and Safety in Patients With Locally Advanced or Metastatic Basal Cell Carcinoma (BOLT)” Available at:http://clinicaltrials.gov/ct2/show/NCT01327053?term=%22LDE225%22+and+%22BOLT%22&rank=1. Accessed on February 14, 2014.

National Cancer Institute Dictionary of Cancer Terms. “Complete Response.” Available at: http://www.cancer.gov/dictionary?CdrID=45652 . Accessed on February 14, 2014.

 National Cancer Institute Dictionary of Cancer Terms. “Partial Response.” Available at: http://www.cancer.gov/dictionary?CdrID=45819 . Accessed on February 14, 2014.

Wong C S M, Strange R C, Lear J T (2003). Basal cell carcinoma. BMJ; 327:794-798.

 Copcu E, Aktas A. Simultaneous two organ metastases of the giant basal cell carcinoma of the skin. Int Semin Surg Oncol. 2005;2:1-6. Available at:http://www.ncbi.nlm.nih.gov/pmc/articles/PMC544837/ . Accessed on February 14, 2014.

 Skin Cancer Foundation. “Basal Cell Carcinoma Treatment Options.” Available athttp://www.skincancer.org/skin-cancer-information/basal-cell-carcinoma/bcc-treatment-options . Accessed on February 14, 2014.

Stuetz A, et al. LDE225, a specific smoothened inhibitor, for the topical treatment of nevoid basal cell carcinoma syndrome (Gorlin’s syndrome). Melanoma Research. 2010; 20:e40. Available at:http://journals.lww.com/melanomaresearch/Fulltext/2010/06001/FC24_LDE225,_a_specific_smoothened_inhibitor,_for.87.aspx#FC24_LDE225%2C_a_specific_smoothened_inhibitor%2C_for.87.aspx?s=2&_suid=139234380607909969110518506816.

Novartis.com. “The Pipeline of Novartis Oncology: LDE225.” Available at:http://www.novartisoncology.com/research-innovation/pipeline.jsp #. Accessed on February 14, 2014.

 Children’s Medical Research Center, Children’s Memorial Hospital/Northwestern University Feinberg School of Medicine. “The Sonic hedgehog/patched/gli signal transduction pathway.” Available at http://www.childrensmrc.org/iannaccone/gli/ . Accessed on February 14, 2014.

 Gupta S, Takebe N, LoRusso P. Targeting the Hedgehog pathway in cancer. Ther Adv Med Oncol. 2010 July; 2(4): 237-250. Available at:http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3126020/ . Accessed on February 14, 2014.

SONIDEGIB

Links

WO2004078163A2 Feb 26, 2004 Sep 16, 2004 Oern Almarsson Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
WO2007113120A1 Mar 22, 2007 Oct 11, 2007 Frank Hoffmann Stamping apparatus with feed device
WO2007131201A2 * May 4, 2007 Nov 15, 2007 Irm Llc Compounds and compositions as hedgehog pathway modulators
WO2008154259A1 Jun 4, 2008 Dec 18, 2008 Irm Llc Biphenylcarboxamide derivatives as hedgehog pathway modulators

 

 

ANTHONY MELVIN CRASTO

THANKS AND REGARD’S
DR ANTHONY MELVIN CRASTO Ph.D

amcrasto@gmail.com

MOBILE-+91 9323115463
GLENMARK SCIENTIST ,  INDIA
web link
http://anthonycrasto.jimdo.com/
Congratulations! Your presentation titled “Anthony Crasto Glenmark scientist, helping millions with websites” has just crossed MILLION views.
アンソニー     安东尼   Энтони    안토니     أنتوني
join my process development group on google
you can post articles and will be administered by me on the google group which is very popular across the world

Filed under: Phase2 drugs, Phase3 drugs Tagged: Erismodegib, LDE225, novartis, NVP-LDE225, phase 2, PHASE 3, sonidegib

Cinnamon cuts blood glucose levels in diabetes patients

$
0
0

Originally posted on lyranara.me:

Consumption of cinnamon is associated with favorable reductions in plasma glucose and lipid levels, according to research published in the September/October issue of the  Annals of Family Medicine .

Robert W. Allen, Pharm.D., of the Western University of Health Sciences in Pomona, Calif., and colleagues used data from 10 randomized, controlled trials involving 543 patients with  to conduct an update of a previous systematic review and meta-analysis examining the effect of cinnamon consumption on glucose and lipid levels.

The researchers found that cinnamon, in daily doses of 120 mg/d to 6 g/d for four to 18 weeks, was associated with a significant reduction in levels of fasting plasma glucose (?24.59 mg/dL), but no significant effect on glycosylated hemoglobin. Cinnamon intake also was linked to significant changes in , including decreases in levels of total cholesterol (?15.60 mg/dL), low-density lipoprotein cholesterol (LDL-C) (?9.42 mg/dL), and triglycerides (?29.59 mg/dL), and increases in levels of high-density lipoprotein cholesterol (HDL-C) (1.66 mg/dL). High degrees of statistical heterogeneity were detected in the analysis of all parameters except HDL-C.

View original


Filed under: Uncategorized

FDA Accepts Filing of NDA for IV Antibiotic Oritavancin with Priority Review

$
0
0

File:Oritavancin.svg

Oritavancin
(4R)-22-O-(3-Amino-2,3,6-trideoxy-3-C-methyl-alpha-L-arabinohexopyranosyl)-N3-(p-(p-chlorophenyl)benzyl)vancomycin

(3S, 6R, 7R, 22R, 23S, 26S, 36R, 38aR) -22 – (3-Amino-2 ,3,6-trideoxy-3-C-methyl-alpha-L-mannopyranosyloxy) -3 – (carbamoylmethyl ) -10,19-dichloro-44-[2-O-[3 - (4'-chlorobiphenyl-4-ylmethylamino) -2,3,6-trideoxy-3-C-methyl-alpha-L-mannopyranosyl] – beta-D-glucopyranosyloxy] -

CAS No. 171099-57-3
CBNumber: CB92451283
Molecular Formula: C86H97Cl3N10O26
Formula Weight: 1793.12

Also known as NDISACC-(4-(4-chlorophenyl)benzyl)A82846B and LY333328,N-(4-(4-chlorophenyl)benzyl)A82846B

Abbott (Supplier), Lilly (Originator), InterMune (Licensee)

The medicines company—

  1. the Oritavancin Program Results.pdf

    phx.corporate-ir.net/External.File?item…t=1

    Jul 2, 2013 - Inhibits two key steps of cell wall synthesis: – Transglycosylation. – Transpeptidation. • Disrupts bacterial membrane integrity. Differentiated from  


FDA Accepts Filing of NDA for IV Antibiotic Oritavancin with Priority Review

PARSIPPANY, NJ — (Marketwired) — 02/19/14 — The Medicines Company (NASDAQ: MDCO) today announced that the U.S. Food and Drug Administration (FDA) has accepted the filing of a new drug application (NDA) for oritavancin, an investigational intravenous antibiotic, with priority review. The Medicines Company is seeking approval of oritavancin for the treatment of acute bacterial skin and skin structure infections (ABSSSI) caused by susceptible gram-positive bacteria, including methicillin-resistant Staphylococcus aureus (MRSA), administered as a single dose.

In December 2013, the FDA designated oritavancin as a Qualified Infectious Disease Product (QIDP). The QIDP designation provides oritavancin priority review, and an additional five years of exclusivity upon approval of the product for the treatment of ABSSSI. Priority review means the FDA’s goal is to take action on the application within six months, compared to 10 months under standard review. The FDA action date (PDUFA date) for oritavancin is August 6, 2014.
Oritavancin (INN, also known as LY333328) is a novel semi-synthetic glycopeptide antibiotic being developed for the treatment of serious Gram-positive infections. Originally discovered and developed by Eli Lilly, oritavancin was acquired by InterMune in 2001 and then by Targanta Therapeuticsin late 2005.[1]

In Dec 2008 the FDA declined to approve it, and an EU application was withdrawn.

In 2009 the development rights were acquired by The Medicine Co. who are running clinical trials for a possible new FDA application in 2013.[2]

Its structure is similar to vancomycin[3] It is a lipoglycopeptide

About Oritavancin

Oritavancin is an investigational intravenous antibiotic for which The Medicines Company is seeking approval in the treatment of ABSSSI caused by susceptible gram-positive bacteria, including MRSA. In clinical trials, the most frequently reported adverse events associated with oritavancin were nausea, headache, vomiting and diarrhea. Hypersensitivity reactions have been reported with the use of antibacterial agents including oritavancin.

 

Oritavancin shares certain properties with other members of the glycopeptide class of antibiotics, which includes vancomycin, the current standard of care for serious Gram-positive infections in the United States and Europe.[4] Data presented at the 47th Annual Interscience Conference on Antimicrobial Agents and Chemotherapy (ICAAC) in September 2007 demonstrated that oritavancin possesses potent and rapid bactericidal activity in vitro against a broad spectrum of both resistant and susceptible Gram positive bacteria, including Staphylococcus aureusmethicillin-resistant Staphylococcus aureusEnterococci, and Streptococci.[5] Two posters presented at the meeting also demonstrated that oritavancin was more active than either metronidazole or vancomycin against strains of Clostridium difficile tested.[6]

Anthrax : Research presented at the American Society for Microbiology (ASM) 107th Annual General Meeting in May 2007, suggested oritavancin’s potential utility as a therapy for exposure to Bacillus anthracis, the gram-positive bacterium that causes anthrax, having demonstrated efficacy in a mouse model both pre- and post-exposure to the bacterium[7]

oritavancin

The 4′-chlorobiphenylmethyl group disrupts the cell membrane of gram positive bacteria.[8] It also acts by inhibition of transglycosylation and inhibition of transpeptidation.[9]

Results have been presented (in 2003) but possibly not yet published from two pivotal Phase 3 clinical trials testing the efficacy of daily intravenous oritavancin for the treatment of complicated skin and skin-structure infections (cSSSI) caused by Gram-positive bacteria. The primary endpoints of both studies were successfully met, with oritavancin achieving efficacy with fewer days of therapy than the comparator agents (vancomycin followed by cephalexin). In addition, oritavancin showed a significantly improved safety profile with a 19.2 percent relative reduction in the overall incidence of adverse events versus vancomycin/cephalexin (p<0.001) in the second and larger pivotal trial.[10]

A Phase 2 clinical study was planned to run until May 2008 entitled “Single or Infrequent Doses for the Treatment of Complicated Skin and Skin Structure Infections (SIMPLIFI),” evaluating the efficacy and safety of either a single dose of oritavancin or an infrequent dose of oritavancin compared to the previously studied dosing regimen of 200 mg oritavancin given once daily for 3 to 7 days.[11] Results published May 2011.[12]

Regulatory submissions

USA

On February 11, 2008, Targanta submitted a New Drug Application (NDA) to the US FDA seeking approval of oritavancin;[13] in April 2008, the FDA accepted the NDA submission for standard review.[14] On 9 Dec 2008 the FDA said insufficient data for approval of oritavancin had been provided and they requested a further phase 3 clinical study to include more patients with MRSA.[15]

Europe

June 2008, Targanta’s Marketing Authorization Application (MAA) for oritavancin was submitted and accepted for review by the European Medicines Agency (EMEA),[16] but the company later withdrew the application in Aug 2009.[17]

About The Medicines Company

The Medicines Company’s purpose is to save lives, alleviate suffering, and contribute to the economics of healthcare by focusing on 3,000 leading acute/intensive care hospitals worldwide. Its vision is to be a leading provider of solutions in three areas: acute cardiovascular care, surgery and perioperative care, and serious infectious disease care. The company operates in the Americas, Europe and the Middle East, and Asia Pacific regions with global centers today in Parsippany, NJ, USA and Zurich, Switzerland.

“We look forward to working with the FDA during the review process, and sharing the knowledge we have gained in our studies of oritavancin,” said Matthew Wikler, MD, Vice President and Medical Director, Infectious Disease Care for The Medicines Company. “We believe that upon approval, oritavancin, administered as a single dose for the treatment of ABSSSI, will offer new options for both physicians and their patients for the treatment of these infections.”

The oritavancin NDA is based on data from two Phase 3 clinical trials, SOLO I and SOLO II, which were conducted under a Special Protocol Assessment (SPA) agreement with the FDA. These Phase 3 trials evaluated the efficacy and safety of a single 1200mg dose of oritavancin compared to 7 to 10 days of twice-daily vancomycin in adults with ABSSSI, including infections caused by MRSA. The combined SOLO studies were conducted in 1,959 patients (modified intent-to -treat population, or mITT), with 405 of the patients suffering from an ABSSSI with a documented MRSA infection.

Figure US20130172237A1-20130704-C00001oritavancin

Drug substance

Oritavancin diphosphate

CLINICAL TRIALS..http://clinicaltrials.gov/search/intervention=oritavancinLinks

  • LY 333328 diphosphate
  • LY333328 diphosphate
  • Oritavancin diphosphate
  • UNII-VL1P93MKZN
  • 192564-14-0 CAS NO

INTRODUCTION

Oritavancin

Oritavancin inhibits cell wall synthesis by complexing with the terminal D-Ala-D-Ala of a nascent peptidoglycan chain and also to the pentaglycine bridge, thus inhibiting transglyco- sylation and transpeptidation. Unlike other glycopeptides, oritavancin is able to bind to depsipeptides including D-Ala-D-Lac, which fa- cilitates its inhibition of cell wall synthesis even in organisms exhibiting VanA-type resistance. Oritavancin forms homodimers prior to binding to D-Ala-D-Ala or D-Ala-D-Lac, which increases its binding affinity for the target site.The p-chloro-phenylbenzyl side chain of oritavancin interacts with the cell membrane, exerting two beneficial effects. This binding acts to main- tain the antibacterial in a prime position for peptidoglycan interactions and it also imparts oritavancin with the ability to disrupt the bac- terial membrane potential and thus increase membrane permeability.[22,23] Oritavancin has been shown to dissipate membrane potential in both stationary and exponential phase growing bacteria, which is rare and may carry clinical implications in terms of its activity against slowly growing organisms and biofilms. The dual mechanism of action could also theoretically increase effectiveness and reduce the risk of resist- ance selection. In addition to the aforemen- tioned mechanisms, it has also been hypothesized that oritavancin inhibits RNA synthesis.

vancomycin, desmethylvancomycin, eremomycin, teicoplanin (complex of five compounds), dalbavancin, oritavancin, telavancin, and A82846B (LY264826) having structures A, B, C, D, E, F, G and H:

Figure imgf000002_0001

R = B-2-Acetylamido-glucopyraπosyl- Attorney Docket No 33746-704 602

Figure imgf000003_0001
Figure imgf000003_0002

Dalbavancin, oritavancin and telavancin are semisynthetic lipoglycopeptides that demonstrate promise for the treatment of patients with infections caused by multi-drug-resistant Gram-positive pathogens. Each of these agents contains a heptapeptide core, common to all glycopeptides, which enables them to inhibit transglycosylation and transpeptidation (cell wall synthesis). Modifications to the heptapeptide core result in different in vitro activities for the three semisynthetic lipoglycopeptides. All three lipoglycopeptides contain lipophilic side chains, which prolong their half-life, help to anchor the agents to the cell membrane and increase their activity against Gram-positive cocci. In addition to inhibiting cell wall synthesis, telavancin and oritavancin are also able to disrupt bacterial membrane integrity and increase membrane permeability; oritavancin also inhibits RNA synthesis. Enterococci exhibiting the VanA phenotype (resistance to both vancomycin and teicoplanin) are resistant to both dalbavancin and telavancin, whileoritavancin retains activity. Dalbavancin, oritavancin and telavancin exhibit activity against VanB vancomycin-resistant enterococci.

All three lipoglycopeptides demonstrate potent in vitro activity against Staphylococcus aureus and Staphylococcus epidermidis regardless of their susceptibility to meticillin, as well as Streptococcus spp. Both dalbavancin and telavancin are active against vancomycin-intermediate S. aureus (VISA), but display poor activity versus vancomycin-resistant S. aureus (VRSA). Oritavancin is active against both VISA and VRSA. Telavancin displays greater activity against Clostridium spp. than dalbavancin, oritavancin or vancomycin. The half-life of dalbavancin ranges from 147 to 258 hours, which allows for once-weekly dosing, the half-life of oritavancin of 393 hours may allow for one dose per treatment course, while telavancin requires daily administration. Dalbavancin and telavancin exhibit concentration-dependent activity and AUC/MIC (area under the concentration-time curve to minimum inhibitory concentration ratio) is the pharmacodynamic parameter that best describes their activities.Links

Oritavancin’s activity is also considered concentration-dependent in vitro, while in vivo its activity has been described by both concentration and time-dependent models; however, AUC/MIC is the pharmacodynamic parameter that best describes its activity. Clinical trials involving patients with complicated skin and skin structure infections (cSSSIs) have demonstrated that all three agents are as efficacious as comparators. The most common adverse effects reported with dalbavancin use included nausea, diarrhoea and constipation, while injection site reactions, fever and diarrhoea were commonly observed withoritavancin therapy. Patients administered telavancin frequently reported nausea, taste disturbance and insomnia. To date, no drug-drug interactions have been identified for dalbavancin, oritavancin or telavancin. All three of these agents are promising alternatives for the treatment of cSSSIs in cases where more economical options such as vancomycin have been ineffective, in cases of reduced vancomycin susceptibility or resistance, or where vancomycin use has been associated with adverse events.

Oritavancin diphosphate (oritavancin) is a semi-synthetic lipoglycopeptide derivative of a naturally occurring glycopeptide. Its structure confers potent antibacterial activity against gram-positive bacteria, including vancomycin-resistant enterococci (VRE), methicillin- and vancomycin-resistant staphylococci, and penicillin-resistant streptococci. The rapidity of its bactericidal activity against exponentially-growing S. aureus (≧3-log reduction within 15 minutes to 2 hours against MSSA, MRSA, and VRSA) is one of the features that distinguishes it from the prototypic glycopeptide vancomycin (McKay et al., J Antimicrob Chemother. 63(6):1191-9 (2009), Epub 2009 Apr. 15).

Oritavancin inhibits the synthesis of peptidoglycan, the major structural component of the bacterial cell wall by a mechanism that is shared with glycopeptides, such as vancomycin (Allen et al., Antimicrob Agents Chemother 41(1):66-71 (1997); Cegelski et al., J Mol Biol 357:1253-1262 (2006); Arhin et al., Poster C1-1471: Mechanisms of action of oritavancin in Staphylococcus aureus [poster]. 47th Intersci Conf Antimicro Agents Chemo, Sep. 17-20, 2007; Chicago, Ill.). Oritavancin, like vancomycin, binds to the Acyl-D-Alanyl-D-Alanine terminus of the peptidoglycan precursor, lipid-bound N-acetyl-glucosamine-N-acetyl-muramic acid-pentapeptide (Reynolds, Eur J Clin Microbiol Infect Dis 8(11):943-950 (1989); Nicas and Allen, Resistance and mechanism of action.

In: Nagarajan R, editor. Glycopeptide antibiotics. New York: Marcel Dekker 195-215 (1994); Allen et al., Antimicrob Agents Chemother 40(10):2356-2362 (1996); Allen and Nicas, FEMS Microbiology Reviews 26:511-532 (2003); Kim et al., Biochemistry 45:5235-5250 (2006)). However, oritavancin inhibits cell wall biosynthesis even when the substrate is the altered peptidoglycan precursor that is present in VRE and vancomycin-resistant S. aureus (VRSA). Thus, the spectrum of oritavancin antibacterial activity extends beyond that of vancomycin to include glycopeptide-resistant enterococci and staphylococci (Ward et al., Expert Opin Investig Drugs 15:417-429 (2006); Scheinfeld, J Drugs Dermatol 6:97-103 (2007)). Oritavancin may inhibit resistant bacteria by interacting directly with bacterial proteins in the transglycosylation step of cell wall biosynthesis (Goldman and Gange, Curr Med Chem 7(8):801-820 (2000); Halliday et al., Biochem Pharmacol 71(7):957-967 (2006); Wang et al., Poster C1-1474: Probing the mechanism of inhibition of bacterial peptidoglycan glycotransferases by glycopeptide analogs. 47th Intersci Conf Antimicro Agents Chemo, Sep. 17-20, 2007). Oritavancin also collapses transmembrane potential in gram positive bacteria, leading to rapid killing (McKay et al., Poster C1-682: Oritavancin disrupts transmembrane potential and membrane integrity concomitantly with cell killing in Staphylococcus aureus and vancomycin-resistant Enterococci. 46th Intersci Conf Antimicro Agents Chemo, San Francisco, Calif., Sep. 27-30, 2006). These multiple effects contribute to the rapid bactericidal activity of oritavancin.

Vancomycin (U.S. Patent 3,067,099); A82846A, A82846B, and A82846C (U.S. Patent 5,312,738, European Patent Publication 256,071 A1); PA-42867 factors A, C, and D (U.S. Patent4,946,941 and European Patent Publication 231,111 A2); A83850 (U.S. Patent No. 5,187,082); avoparcm (U.S. Patent 3,338,786 and U.S. Patent 4,322,343); actmoidin, also known as K288 (J. Antibiotics Series A 14:141 (1961); helevecardin (Chem. Abstracts 110:17188 (1989) and Japanese Patent Application 86/157,397); galacardin (Chem. Abstracts 110:17188 (1989) and Japanese Patent Application 89/221,320); and M47767 (European Patent Publication 339,982).

Oritavancin is in clinical development against serious gram-positive infections, where administration of the drug is via intravenous infusion using several dosages administered over a series of days. The development of alternative dosing regimens for the drug could expand treatment options available to physicians. The present invention is directed to novel dosing regimens.

Means for the preparation of the glycopeptide antibiotics, including oritavancin and analogs thereof, may be found, for example, in U.S. Pat. No. 5,840,684,

ORITAVANCIN DIPHOSPHATE

LinksSYNTHESIS

LY-333328 was synthesized by reductocondensation of the glycopeptide antibiotic A82846B (I) with 4′-chlorobiphenyl-4-carboxaldehyde (II) by means of sodium cyanoborohydride in refluxing methanol.

J Antibiot1996, 49, (6) :575-81

(3S,6R,7R,22R,23S,26S,36R,38aR)-3-(Carbamoylmethyl)-10,19-dichloro-7,28,30,32-tetrahydroxy-6-(N-methyl-D-leucylamido)-2,5,24,38,39-pentaoxo-22-(L-vancosaminyloxy)-44-[2-O-(L-vancosaminyl)-beta-D-glucopyranosyloxy]-2,3,4,5,6,7,23,24,25,26,36,37,38,38a-tetradecahydro-1H,22H-8,11:18,21-dietheno-23,36-(iminomethano)-13,16:31,36-dimetheno-[1,6,9]oxadiazacyclohexadecino[4,5-m][10,2,16]benzoxadiazacyclotetracosine-26-carboxylic acid; A82846B (I)
4′-chloro[1,1'-biphenyl]-4-carbaldehyde (II)

LY-333328 was synthesized by reductocondensation of the glycopeptide antibiotic A82846B (I) with 4′-chlorobiphenyl-4-carboxaldehyde (II) by means of sodium cyanoborohydride in refluxing methanol.

…………………..Links

WO1996030401A1

EXAMPLE 4

Preparation of Compound 229

A three liter 3-necked flask was fitted with a

condenser, nitrogen inlet and overhead mechanical stirring apparatus. The flask was charged with pulverized A82846B acetate salt (20.0 g, 1.21 × 10-3 mol) and methanol (1000 mL) under a nitrogen atmosphere. 4′-chlorobiphenylcarboxaldehyde (2.88 g, 1.33 × 10-2 mol, 1.1 eq.) was added to this stirred mixture, followed by methanol (500 mL). Finally, sodium cyanoborohydride (0.84 g, 1.33 × 10-2 mol, 1.1 eq.) was added followed by methanol (500 mL). The resulting mixture was heated to reflux (about 65°C).

After 1 hour at reflux, the reaction mixture attained homogeneity. After 25 hours ac reflux, the heat source was removed and the clear reaction mixture was measured with a pH meter (6.97 at 58.0°C). 1 N NaOH (22.8 mL) was added

dropwise to adjust the pH to 9.0 (at 54.7°C). The flask was equipped with a distillation head and the mixture was concentrated under partial vacuum to a weight of 322.3 grams while maintaining the pot temperature between 40-45°C.

The distillation head was replaced with an addition funnel containing 500 mL of isopropanol (IPA). The IPA was added dropwise to the room temperature solution over 1 hour. After approximately 1/3 of the IPA was added, a granular precipitate formed. The remaining IPA was added at a faster rate after precipitation had commenced. The flask was weighed and found to hold 714.4 grams of the IPA/methanol slurry.

The flask was re-equipped with a still-head and

distilled under partial vacuum to remove the remaining methanol. The resulting slurry (377.8 g) was allowed to chill in the freezer overnight. The crude product was filtered through a polypropylene pad and rinsed twice with 25 mL of cold IPA. After pulling dry on the funnel for 5 minutes, the material was placed in the vacuum oven to dry at 40°C. A light pink solid (22.87 g (theory = 22.43 g) ) was recovered. HPLC analysis versus a standard indicated 68.0% weight percent of Compound 229 (4- [4-chlorophenyl] benzyl-A82846B] in the crude solid, which translated into a

corrected crude yield of 69.3%.

The products of the reaction were analyzed by reverse-phase HPLC utilizing a Zorbax SB-C18 column with ultraviolet light (UV; 230 nm) detection. A 20 minute gradient solvent system consisting of 95% aqueous buffer/5% CH3CN at time=0 minutes to 40% aqueous buffer/60% CH3CN at time=20 minutes was used, where the aqueous buffer was TEAP (5 ml CH3CN, 3 ml phosphoric acid in 1000 ml water).

………………….

WO2008097364A2

Oritavancin (also termed N-(4-(4-chlorophenyl)benzyl)A82846B and LY333328) has the following Formula III:

Figure imgf000029_0001

ReferencesLinks

  1.  Targanta Revives Oritavancin: Next Weapon Against cSSSI? BioWorld Today, November 26, 2007
  2.  “Biotechs pick up slack in antibiotics development”. 17 May 2011.
  3.  http://www.farm.ucl.ac.be/Full-texts-FARM/Domenech-2009-1.pdf ”Interactions of oritavancin, a new lipoglycopeptide derived from vancomycin, with phospholipid bilayers: Effect on membrane permeability and nanoscale lipid membrane organization” 2009
  4.  Scheinfeld, N (2007). “A comparison of available and investigational antibiotics for complicated skin infections and treatment-resistant Staphylococcus aureus and enterococcus“.J Drugs Dermatol. 6 (4): 97–103. PMID 17373167.
  5.  2007 ICAAC Posters: E-1612 “In Vitro Activity Profile of Oritavancin against a Broad Spectrum of Aerobic and Anaerobic Bacterial Pathogens”/E -1613 “In Vitro Activity Profile of Oritavancin (ORI) Against Organisms Demonstrating Key Resistance Profiles to Other Antimicrobial Agents”/E-1614 “In vitro Time Kill Studies of Oritavancin against Drug-resistant Isolates ofStaphylococcus aureus and Enterococci”/E-1615 “Anti-Enterococcal Activity Profile of Oritavancin, a Potent Lipoglycopeptide under Development for Use Against Gram-Positive Infections”/E-1616 “Anti-Streptococcal Activity Profile of Oritavancin, a Potent Lipoglycopeptide under Development for Use Against Gram-Positive Infections”/E-1617 “In Vitro Activity Profile of Oritavancin (ORI) Against Resistant Staphylococcal Populations From a Recent Surveillance Initiative”/E-1620 “Pharmacokinetic Concentrations of Oritavancin Kill Stationary-Phase and Biofilm Staphylococcus aureus In Vitro.” / Targanta Press Release September 19, 2007
  6.  ICAAC 2007 Posters: “In Vitro Susceptibility of Genotypically Distinct Clostridium difficileStrains to Oritavancin” and “Activity of Metronidazole, Vancomycin and Oritavancin Against Epidemic Clostridium difficile Spores” / Targanta Press Release September 19, 2007
  7.  ASM 2007 Poster: “Efficacy of Oritavancin in a Murine Model of Bacillus anthracis Spore Inhalation Anthrax” / Targanta Press Release May 24, 2007
  8.  Belley; McKay, GA; Arhin, FF; Sarmiento, I; Beaulieu, S; Fadhil, I; Parr Jr, TR; Moeck, G (2010).“Oritavancin Disrupts Membrane Integrity of Staphylococcus aureus and Vancomycin-Resistant Enterococci To Effect Rapid Bacterial Killing”Antimicrobial agents and chemotherapy 54(12): 5369–71. doi:10.1128/AAC.00760-10PMC 2981232PMID 20876372.
  9.  Zhanel et al. (2012). “Oritavancin: Mechanism of Action”Clin Infect Dis.doi:10.1093/cid/cir920.
  10. ICAAC 2003 Late-breaker poster: “Phase III Trial Comparing 3-7 days of Oritavancin vs. 10-14 days of Vancomycin/Cephalexin in the Treatment of Patients with Complicated Skin and Skin Structure Infections (cSSSI)” / InterMune Press Release September 15, 2003
  11.  ClinicalTrials.gov NCT00514527
  12.  Comparison of the Efficacy and Safety of Oritavancin Front-Loaded Dosing Regimens to Daily Dosing: An Analysis of the SIMPLIFI Trial. May 2011. doi:10.1128/AAC.00029-11.
  13.  “Drugs.com, Targanta Submits Oritavancin New Drug Application”. Retrieved 2008-02-12.
  14.  “FDA News, Targanta to Get FDA Decision by December”. Retrieved 2008-04-10.
  15.  http://www.fiercebiotech.com/press-releases/fda-issues-complete-response-letter-oritavancin Dec 2008.
  16.  “Pharmaceutical Business Review, EMEA accepts Targanta’s oritavancin MAA for review”. Retrieved 2008-06-26.
  17.  http://www.nelm.nhs.uk/en/NeLM-Area/News/2009—August/24/European-application-for-investigational-antibiotic-oritavancin-withdrawn-/
  18. http://onlinelibrary.wiley.com/doi/10.1111/j.1574-6976.2003.tb00628.x/pdf
  19. http://www.pjps.pk/wp-content/uploads/pdfs/26/5/Paper-30.pdf
  20. Antimicrobial Agents and Chemotherapy, 2003 ,  vol. 47,   5  p. 1700 – 1706
  21. Antimicrobial Agents and Chemotherapy, 1999 ,  vol. 43,   1  p. 115 – 120
  22. Antimicrobial Agents and Chemotherapy, 1997 ,  vol. 41,   10  p. 2165 – 2172
  23. Tetrahedron, 2004 ,  vol. 60,   47  p. 10611 – 10618………… NMRhttp://www.sciencedirect.com/science/article/pii/S0040402004015108

LinksCooper, R.D.G.; Snyder, N.J.; Zweifel, M.J.; et al.; Reductive alkylation of glycopeptide antibiotics: Synthesis and antibacterial activity. J Antibiot 1996, 49, 6, 575-81.

 Fromtling, R.A.; Castaer, J.; LY-333328. Drugs Fut 1998, 23, 1, 17.
Cooper, R.D.G.; Huff, B.E.; Nicas, T.I.; Quatroche, J.T.; Rodriguez, M.J.; Snyder, N.J.; Staszak, M.A.; Thompson, R.C.; Wilkie, S.C.; Zweifel, M.J. (Eli Lilly and Company); Glycopeptide antibiotic derivs. EP 0817797; JP 1999502534; WO 9630401 .

Cooper, R.D.G.; Huff, B.E.; Nicas, T.I.; Quatroche, J.T.; Rodriguez, M.J.; Snyder, N.J.; Staszak, M.A.; Thompson, R.C.; Wilkie, S.C.; Zweifel, M.J. (Eli Lilly and Company); Glycopeptide antibiotic derivs. EP 0667353; EP 1016670; EP 1031576 .

EP0435503A1 * Dec 11, 1990 Jul 3, 1991 Eli Lilly And Company Improvements in or relating to gylcopeptide derivatives
US4639433 * Aug 14, 1985 Jan 27, 1987 Eli Lilly And Company Glycopeptide derivatives
US4698327 * Apr 18, 1986 Oct 6, 1987 Eli Lilly And Company Novel glycopeptide derivatives
US20040106590 * Aug 29, 2003 Jun 3, 2004 Barry Eisenstein Methods and reagents for treating infections of clostridium difficile and diseases associated therewith
US20050197333 Dec 22, 2004 Sep 8, 2005 Van Duzer John H. Rifamycin analogs and uses thereof
US20070014849 Sep 20, 2006 Jan 18, 2007 Daniela Jabes Use of ramoplanin to treat diseases associated with the use of antibiotics
US20030176327 * Oct 18, 2002 Sep 18, 2003 Cassell Gail Houston Antibiotics for treating biohazardous bacterial agents
US20040147441 Aug 25, 2003 Jul 29, 2004 Leach Timothy S. Methods and reagents for preventing bacteremias
WO1999010006A1 Aug 18, 1998 Mar 4, 1999 Lilly Co Eli Therapy for staphylococcus aureus
WO2000066144A2 Apr 19, 2000 Nov 9, 2000 Lilly Co Eli Monthly doses of glycopeptide antibiotics for treatment of streptococcus pneumoniae infections
WO2008097364A2 Sep 24, 2007 Aug 14, 2008 Targanta Therapeutics Corp Use of oritavancin for prevention and treatment of anthrax
WO1998052592A1 * May 5, 1998 Nov 26, 1998 Lilly Co Eli Urea and thiourea derivatives of glycopeptides
WO2002036612A1 * Nov 2, 2001 May 10, 2002 Univ Cambridge Tech Antibacterial agents comprising conjugates of glycopeptides and peptidic membrane-associating elements
WO2007138999A1 May 25, 2007 Dec 6, 2007 Shionogi & Co Glycopeptide antibiotic derivative
WO2009081958A1 Dec 25, 2008 Jul 2, 2009 Shionogi & Co Glycosylated glycopeptide antibiotic derivative
EP2314599A1 Nov 24, 2005 Apr 27, 2011 National University Corporation Nagoya University Glycopeptide antibiotic monomer derivatives
US5919756 * May 1, 1997 Jul 6, 1999 Eli Lilly And Company Amides
US5919771 * Apr 30, 1998 Jul 6, 1999 Eli Lilly And Company Urea and thiourea derivatives of glycopeptides
US7078380 Nov 2, 2001 Jul 18, 2006 Cambridge University Technical Services Limited Antibacterial agents comprising conjugates of glycopeptides and peptidic membrane associating elements
US8481696 Dec 25, 2008 Jul 9, 2013 Shionogi & Co., Ltd. Glycosylated glycopeptide antibiotic derivatives

Links


Filed under: NDA, Priority review Tagged: Oritavancin

KW-4490 A PDE4 inhibitor from Kyowa Hakko Kirin

$
0
0
KW 4490
cis-4-Cyano-4-(2,3-dihydro-8-methoxy-1,4-benzodioxin-5-yl)cyclohexanecarboxylic Acid 
Cyclohexanecarboxyli​c acid, 4-​cyano-​4-​(2,​3-​dihydro-​8-​methoxy-​1,​4-​benzodioxin-​5-​yl)​-​, cis-
cis-​4-​Cyano-​4-​(2,​3-​dihydro-​8-​methoxy-​1,​4-​benzodioxin-​5-​yl)​cyclohexane-​1-​carboxylic acid;
cis-​4-​Cyano-​4-​(8-​methoxy-​1,​4-​benzodioxan-​5-​yl)​cyclohexanecarboxyli​c acid

KF 66490; KW 4490;

MF C17 H19 N O5

phosphodiesterase type 4 inhibitor, commonly referred to as a PDE4 inhibitor, is a drug used to block the degradative action ofphosphodiesterase 4 (PDE4) on cyclic adenosine monophosphate (cAMP). It is a member of the larger family of PDE inhibitors. The PDE4 family of enzymes are the most prevalent PDE in immune cells. They are predominantly responsible for hydrolyzing cAMP within both immune cells and cells in the central nervous system

PDE4 hydrolyzes cyclic adenosine monophosphate (cAMP) to inactive adenosine monophosphate (AMP). Inhibition of PDE4 blocks hydrolysis of cAMP, thereby increasing levels of cAMP within cells.

Practical synthesis of the PDE4 inhibitor, KW-4490

ORGN 699

Arata Yanagisawa, arata.yanagisawa@kyowa.co.jp1, Koichiro Nishimura2, Tetsuya Nezu2, Kyoji Ando2, Ayako Maki2, Eiichiro Imai2, and Shin-ichiro Mohri2. (1) Pharmaceutical Research Center, Medicinal Chemistry Research Laboratories, Kyowa Hakko Kogyo Co., Ltd, 1188 Shimotogari, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan, (2) Pharmaceutical Research Center, Sakai Research Laboratories, Kyowa Hakko Kogyo Co., Ltd, 1-1-53 Takasu-cyo, Sakai-ku, Sakai, Osaka, Japan
A practical and scaleable synthesis of the PDE4 inhibitor, KW-4490 (1), was developed for the multi-kilogram preparation. This improved synthesis features construction of the 1-arylcyclohexene by Diels-Alder reaction, followed by a newly established acid-mediated hydrocyanation. The synthesis was achieved in 7 steps in 38% overall yield. Efforts toward increasing the regioselectivity in the Diels-Alder reaction, optimization of crystallization-induced dynamic resolution of the hydrocyanation product, and investigation of other synthetic routes will be presented.

A team at Kyowa Hakko Kirin in Japan has used a crystallisation-induced dynamic resolution in the synthesis of KW-4490, a PDE-4 inhibitor being developed for asthma and chronic obstructive pulmonary disease.6 Towards the end of the synthesis, they were faced with a mixture of cis and trans diastereomers of an intermediate derived from a hydrocyanation reaction, which was about 62:38 cis:trans; altering the conditions of the reaction did not give a selective process. The desired isomer was the cis, so they wanted to convert the unwanted trans isomer to cis to improve the yield (Scheme 2).

They first tried using a base-induced isomerisation using a base such as potassium t-butoxide, but although this worked to a degree the best ratio of products obtained was 75:25. The same result was obtained when they tested the system on both pure cis and trans isomers, indicating that this ratio represented the thermodynamic equilibrium. However, they realised that the cis isomer was less soluble in ethanol, so they thought the answer might lie in crystallisation-induced dynamic resolution.

They therefore suspended a crude mixture of the two isomers in ethanol and added a catalytic amount of potassium t-butoxide to effect the isomerisation. It was stirred and warmed, and hexane added portion-wise to crash the cis isomer out of solution. The group managed to increase the ratio of isomers to 99:1 by continuous isomerisation, with a 90% isolated yield.

Scheme 2: Kyowa Hakko Kirin found a way to improve the yield of the cis isomer

A Practical Synthesis of the PDE4 Inhibitor, KW-4490

http://pubs.acs.org/doi/abs/10.1021/op1001287?prevSearch=KW%2B4490&searchHistoryKey=

Arata Yanagisawa, Koichiro Nishimura, Kyoji Ando, Tetsuya Nezu, Ayako Maki, Sachiko Kato, Wakako Tamaki, Eiichiro Imai, and Shin-ichiro Mohri
Org. Process Res. Dev.201014 (5), pp 1182–1187
Figure
A practical and scalable synthesis of a PDE4 inhibitor KW-4490 (1) was developed. This improved synthesis features the construction of the 1-arylcyclohexene (9) by the Diels−Alder reaction followed by a newly established Brønsted acid-promoted hydrocyanation. Subsequent crystallization-induced dynamic resolution enabled the high-yield production of the desired cis-isomer (cis-8). The synthesis was achieved in seven steps in 37% overall yield.
Phosphodiesterase 4 (PDE4) is a cyclic adenosine monophosphate (cAMP)-specific phosphodiesterase which is located predominantly in inflammatory cells. High levels of cAMP inhibit the production of cytokines and other molecules that modulate the inflammatory response.(1) Therefore, PDE4 inhibitors have emerged as potential therapeutic agents in the treatment of asthma and chronic obstructive pulmonary disease (COPD).(2) Since cis-4-cyano-4-(2,3-dihydro-8-methoxy-1,4-benzodioxin-5-yl)cyclohexanecarboxylic acid (KW-4490, 1) was identified in Kyowa Hakko Kirin as a potent PDE4 inhibitor,(3) multikilogram quantities were thus required in order to carry out both pharmacological profiling and clinical trials. Structurally, the compound presents the interesting synthetic challenges of constructing a tetra-substituted electron-rich benzene, a tertiary benzylic nitrile, and cis stereochemistry of a carboxylic acid on a 1,4-disubstituted cyclohexane.
In general, tertiary benzylic nitriles has been prepared by the double alkylation of benzylic nitrile,(4) arylations of secondary nitrile anions with aryl halides,(5) or the displacement of tertiary benzylic alcohol with cyanide.(6) The latest approach was applied to our medicinal chemistry synthesis of 1, which was suitable for the delivery of multigram quantities .(3b)
Figure
cis-4-Cyano-4-(2,3-dihydro-8-methoxy-1,4-benzodioxin-5-yl)cyclohexanecarboxylic Acid (1)
A mixture of ester cis-8 (20.0 g, 57.9 mmol), ethanol (100 mL), and 6 mol/L KOH (19 mL) was stirred at room temperature for 4 h. The resultant mixture was diluted with water (102 mL), cooled to 5 °C, and neutralized with 6 mol/L HCl. The precipitate was collected by filtration and dried to give crude carboxylic acid 1 (18.2 g, 57.4 mmol). The crude 1 (18.0 g, 56.7 mmol) was dissolved in acetone (170 mL) and water (30 mL) under reflux. The solution was filtered hot and maintaining 55 °C during addition of water (180 mL) to form precipitation. The suspension was cooled to 5 °C, stirred 3 h, and filtered to obtain 1 (17.2 g, cis/trans = >99.99/<0.01, 54.2 mmol, 95% yield) as a white solid: mp 245 °C;
1H NMR (DMSO-d6) δ 12.24 (s, 1H), 6.79 (d, J = 8.8 Hz, 1H), 6.60 (d, J = 8.8 Hz, 1H), 4.33−4.23 (m, 4H), 3.75 (s, 3H), 2.36−2.25 (m, 3H), 2.06−1.98 (m, 2H), 1.86−1.64 (m, 4H);
13C NMR (DMSO-d6) δ 175.9, 148.6, 141.9, 133.5, 121.6, 120.5, 116.3, 103.9, 63.7, 63.4, 55.4, 41.0, 38.9, 32.9, 25.6;
IR (KBr) 3288, 2930, 2232, 1730, 1508, 1456, 804 cm−1;
HRMS ESI(−) calcd for C17H18NO5 [M − H]− 316.1185, found 316.1195.
……………….
Figure US20040054197A1-20040318-C00002

Compound (XIII) is disclosed in WO00/14085 as being useful as a PDE-IV inhibitor. A method for the preparation of a typical compound among compounds (XIII) disclosed in WO00/14085 is as follows:

Figure US20040054197A1-20040318-C00003
Figure US20040054197A1-20040318-C00004

However, this method is not practically satisfactory as a industrially applicable preparation method, because of (1) requiring multiple steps, (2) low overall yield, (3) requiring purification by silica-gel column chromatography, and the like.

REFERENCE EXAMPLE 1

Synthesis of cis-4-cyano-4-(2,3-dihydro-8-methoxy-1,4-benzodioxine-5-yl)cyclohexanecarboxylic acid

(1) Synthesis of cis-4-cyano-4-(2,3-dihydro-8-methoxy-1,4-benzodioxine-5-yl)cyclohexanecarboxylic acid ethyl ester

Under a nitrogen atmosphere, trifluoromethanesulfonic acid (2.25 g) and trimethylsilylcyanide (1.57 mL) were dissolved in benzotrifluoride (10 mL), and a solution of 4-(2,3-dihydro-8-methoxy-1,4-benzodioxine-5-yl)-3-cyclohexenecarboxylic acid ethyl ester (0.79 g) prepared according to the method described in EXAMPLE 1 in benzotrifluoride (10 mL) was added dropwise at −25° C. After being stirred for for one hour at −20° C., an aqueous saturated sodium hydrogen carbonate was added and the mixture was extracted with ethyl acetate. The organic layer was washed with brine and dried over anhydrous magnesium sulfate, and the solvent was evaporated under reduced pressure. The residue was crystallized from ethanol (1 mL) to give a solid substance (0.64 g). The solid substance (0.030 g) was crystallized from a mixed solvent of diisopropyl ether and ethyl acetate (0.36 mL, diisopropyl ether/ethyl acetate=4/1) to give cis-4-cyano-4-(2,3-dihydro-8-methoxy-1,4-benzodioxine-5-yl)cyclohexanecarboxylic acid ethyl ester (0.019 g, 47.3%) as a solid.

Melting point 131° C.

1H-NMR (CDCl3, δ ppm) 6.84 (d, J=8.9 Hz, 1H), 6.49 (d, J=8.9 Hz, 1H), 4.39−4.33 (m, 4H), 4.17 (q, J=7.1 Hz, 2H), 3.88 (s, 3H), 2.44 (brd, J=12.6 Hz, 2H), 2.32 (tt, J=11.8, 3.8 Hz, 1H), 2.18−1.95 (m, 4H), 1.86 (dt, J=3.6, 12.6 Hz, 2H), 1.28 (t, J=7.1 Hz, 3H).

[0184] IR (KBr, cm−1) 2953, 2228, 1722, 1607, 1504, 1460, 1381, 1325, 1281, 1117, 1043, 953, 787.

MS (m/z) 346(M+H)+.

 (2) Synthesis of cis-4-cyano-4-(2,3-dihydro-8-methoxy-1,4-benzodioxine-5-yl)cyclohexanecarboxylic acid

To a suspension of cis-4-cyano-4-(2,3-dihydro-8-methoxy-1,4-benzodioxine-5-yl)cyclohexanecarboxylic acid ethyl ester (397 g) prepared according to the method described (1) of REFERENCE EXAMPLE 1 in ethanol (1.99 L) was added a 6 ml/L aqueous potassium hydroxide (377 mL), and the mixture was stirred for 4 hours at room temperature. After water (2.03 L) was added to the reaction mixture, a 6 mol/L aqueous hydrochloric acid (576 mL) was added to crystallize and to give cis-4-cyano-4-(2,3-dihydro-8-methoxy-1,4-benzodioxine-5-yl)cyclohexanecarboxylic acid (366 g, 98.1%) as a solid.

Melting point 245° C.

1H-NMR (DMSO-d6, δ ppm) 12.26 (brs, 1H), 6.79 (d, J=8.9 Hz, 1H), 6.59 (d, J=8.9 Hz, 1H), 4.27 (dd, J=11.9, 5.0 Hz, 4H), 3.75 (s, 3H), 2.34−2.26 (m, 3H), 2.05−2.00 (m, 2H), 1.86−1.63 (m, 4H).

IR (KBr, cm−1) 3287, 2932,1728, 1609, 1508, 1454, 1285, 1119, 953, 802, 764.

MS (m/z) 318(M+H)+.

…………………
Table 1.
Figure 00400001
      Example 1.
      4-Cyano-4-(8-methoxy-1,4-benzodioxan-5-yl) cyclohexanone (Compound 1)(Step A)
      Synthesis of 2-(8-methoxy-1,4-benzodioxan-5-yl)acetonitrile (Compound 1a)
    • To a solution of 12 g (62 mmol) of 8-methoxy-1,4-benzodioxane-5-carbaldehyde in 140 ml of acetonitrile was added 12 g (110 mmol) of lithium bromide, and then 12 ml (95 mmol) of trimethylsilyl chloride was dropwise added thereto. After 15 minutes, the mixture was ice-cooled, and 19 ml (110 mmol) of 1,1,3,3-tetramethyldisiloxane was dropwise added thereto, followed by stirring at room temperature for 2 hours. The mixture was diluted with methylene chloride, and then was filtered through Celite. The solvent was evaporated in vacuo from the filtrate to give a pale yellow oily substance. To a solution of the obtained crude 5-bromomethyl-8-methoxy-1,4-benzodioxane in 180 ml of DMF was added 9.2 g (190 mmol) of sodium cyanide, followed by stirring at room temperature for 60 hours. To the mixture was added water under ice-cooling, and a solid separated out therefrom was collected by filtration to give 6.8 g (53%) of Compound 1a as an ash-colored solid.
      Melting Point: 121 – 125 °C
      1H-NMR (CDCl3, δ, ppm) 3.60 (s, 2H), 3.88 (s, 3H), 4.33 (s, 4H), 6.50 (d, J = 8 Hz, 1H), 6.86 (d, J = 8 Hz, 1H).
      MASS (m/z) 205 (M+).
      (Step B) Synthesis of dimethyl 4-cyano-4-(8-methoxy-1,4-benzodioxan-5-yl)pimelate (Compound 1b)
    • To a solution of 6.2 g (30 mmol) of Compound 1a obtained in Step A in 94 ml of acetonitrile were added 1.4 ml (3.0 mmol) of a 40% methanolic solution of Triton B and 27 ml (300 mmol) of methyl acrylate, followed by heating under reflux for 5 hours. The mixture was allowed to stand for cooling, and then poured into water, followed by extraction with ethyl acetate. The organic layer was washed with brine and dried over sodium sulfate, and the solvent was evaporated in vacuo. The residue was purified by silica gel column chromatography (eluted with hexane/ethyl acetate = 2/1) to give 6.4 g (56%) of Compound 1b as a pale yellow oily substance.
      1H-NMR (CDCl3, δ, ppm) 2.05-2.37 (m, 4H), 2.39-2.59 (m, 2H), 2.62-2.82 (m, 2H), 3.60 (s, 6H), 3.87 (s, 3H), 4.20-4.40 (m, 4H), 6.48 (d, J = 9 Hz, 1H), 7.01 (d, J = 9 Hz, 1H).
      MASS (m/z) 377 (M+).
      (Step C) Synthesis of 4-cyano-4-(8-methoxy-1,4-benzodioxan-5-yl)-2-methoxycarbonylcyclohexanone (Compound 1c)
    • To a solution of 6.4 g (17 mmol) of Compound 1b obtained in Step B in 96 ml of 1,2-dimethoxyethane was added 2.0 g (50 mmol) of 60% sodium hydride. After heating under reflux for 3 hours, the mixture was allowed to stand for cooling, poured into ice water, acidified with a 6 mol/liter aqueous hydrochloric acid and extracted with ethyl acetate. The organic layer was washed with brine and dried over sodium sulfate, and the solvent was evaporated. The residue was purified by silica gel column chromatography (eluted with hexane/ethyl acetate = 2/1) to give 5.0 g (86%) of Compound 1c as a white solid.
      Melting Point: 129 – 132 °C
      1H-NMR (CDCl3, δ, ppm) 2.21-2.50 (m, 3H), 2.61-2.89 (m, 2H), 3.11(d, J = 15 Hz, 1H), 3.79 (s, 3H), 3.89 (s, 3H), 4.37 (s, 4H), 6.49 (d, J = 9 Hz, 1H), 6.84 (d, J = 9 Hz, 1H), 12.2 (s, 1H).
      MASS (m/z) 345 (M+).
      (Step D) Synthesis of Compound 1
    • A mixture of 5.0 g (15 mmol) of Compound 1c obtained in Step C, 50 ml of DMSO, 5 ml of water, and 5.0 g of sodium chloride was stirred at 150°C for 5 hours. The mixture was allowed to stand for cooling, and water was added thereto, followed by extraction with ethyl acetate. The organic layer was washed with brine and dried over sodium sulfate, and the solvent was evaporated in vacuo. The residue was purified by silica gel column chromatography (eluted with hexane/ethyl acetate = 3/1) to give 3.6 g (86%) of Compound 1 as a white solid.
      Melting Point: 157 – 161 °C
      1H-NMR (CDCl3, δ, ppm) 2.21-2.41 (m, 2H), 2.45-2.72 (m, 4H), 2.81-3.00 (m, 2H), 3.89 (s, 3H), 4.37 (s, 4H), 6.51 (d, J = 9 Hz, 1H), 6.88 (d, J = 9 Hz, 1H).
      MASS (m/z) 287 (M+).
      Example 2. 4-Cyano-4-(8-methoxy-1,4-benzodioxan-5-yl)cyclohexanone ethyleneketal (Compound 2)
      (Step A)Synthesis of 4-hydroxy-4-(8-methoxy-1,4-benzodioxan-5-yl)cyclohexanone ethyleneketal (Compound 2a)
    • In 65 ml of THF was dissolved 10 g (41 mmol) of 5-bromo-8-methoxy-1,4-benzodioxane, and 28 ml (45 mmol) of a 1.59 mol/liter solution of n-butyl lithium in hexane was dropwise added thereto at -78°C. After 15 minutes, a solution of 9.6 g (61 mmol) of 1,4-cyclohexadione monoethyleneketal in 50 ml of THF was dropwise added thereto. The mixture was stirred for 1 hour, followed by stirring at room temperature for 20 minutes. Water was added thereto, the mixture was extracted with ethyl acetate, and the extract was washed with brine and dried over sodium sulfate. The solvent was evaporated therefrom, and the residue was purified by silica gel column chromatography (eluted with hexane/ethyl acetate = 1/1) to give 9.0 g (68%) of Compound 2a as a white solid.
      Melting Point: 94 – 96 °C
      1H-NMR (CDCl3, δ, ppm) 1.58-1.72 (m, 2H), 1.88-2.28 (m, 6H), 3.57 (s, 1H), 3.86 (s, 3H), 3.90-4.07 (m, 4H), 4.35 (s, 4H), 6.46 (d, J = 9 Hz, 1H), 6.82 (d, J = 9 Hz, 1H).
      MASS (m/z) 322 (M+).
    • (Step B) Synthesis of Compound 2
    • In 4.9 ml of methylene chloride was dissolved 0.49 g (1.5 mmol) of Compound 2a obtained in Step A, 0.26 ml (1.9 mmol) of trimethylsilyl cyanide was added thereto at -78°C, then 0.20 ml (1.6 mmol) of a boron trifluoride-ethyl ether complex was dropwise added thereto, and the mixture was stirred for 10 minutes, followed by stirring at room temperature for 10 minutes. A saturated aqueous solution of sodium bicarbonate was added thereto and the mixture was extracted with ethyl acetate. The extract was washed with brine and dried over sodium sulfate, and the solvent was evaporated. The residue was purified by silica gel column chromatography (eluted with hexane/ethyl acetate = 2/1) to give 0.30 g (61%) of Compound 2 as a colorless oily substance.
      1H-NMR (CDCl3, δ, ppm) 1.79-1.95 (m, 2H), 2.06-2.20 (m, 4H), 2.30-2.46 (m, 2H), 3.87 (s, 3H), 3.90-4.07 (m, 4H), 4.36 (s, 4H), 6.48 (d, J = 9 Hz, 1H), 6.82 (d, J = 9 Hz, 1H).
      MASS (m/z) 331 (M+).
          Example 3. Compound 1
        • In 2.9 ml of acetone was dissolved 0.29 g (0.87 mmol) of Compound 2 obtained in Example 2, 1.2 ml (7.2 mmol) of a 6 mol/liter aqueous hydrochloric acid was added thereto, and the mixture was heated under reflux for 3 hours. The mixture was allowed to stand for cooling and poured into a saturated aqueous solution of sodium bicarbonate, the mixture was extracted with ethyl acetate, and the extract was washed with brine. The mixture was dried over sodium sulfate, and the solvent was evaporated to give 0.23 g (92%) of Compound 1 as a white solid.
          Example 4. Methyl

cis-4-cyano-4-(8-methoxy-1,4-benzodioxan-5-yl)cyclohexanecarboxylate (Compound 3) and methyltrans-4-cyano-4-(8-methoxy-1,4-benzodioxan-5-yl)cyclohexanecarboxylate (Compound 4)(Step A) Synthesis of 2-[4-cyano-4-(8-methoxy-1,4-benzodioxan-5-yl)cyclohexylidene]-1,3-dithiane (Compound 3a)

    • To a solution of 5.0 ml (26 mmol) of 2-trimethylsilyl-1,3-dithiane in 50 ml of THF was added dropwise 17 ml (26 mmol) of a 1.54 mol/liter solution of n-butyl lithium in hexane under ice-cooling. After 10 minutes, the mixture was cooled to -78°C, and a solution of 3.6 g (13 mmol) of Compound 1 obtained in Example 1 in 40 ml of THF was dropwise added thereto. After 10 minutes, to the mixture was added brine, followed by addition of water at room temperature. The mixture was extracted with ethyl acetate, the extract was dried over sodium sulfate, and the solvent was evaporated. The residue was purified by silica gel column chromatography (eluted with hexane/ethyl acetate = 4/1) to give 3.9 g (79%) of Compound 3a as a white solid.
      Melting Point: 164 – 166 °C
      1H-NMR (CDCl3, δ, ppm) 1.70-1.92 (m, 2H), 2.05-2.24 (m, 2H), 2.28-2.53 (m, 4H), 2.89 (t, J = 6 Hz, 4H), 3.18-3.38 (m, 2H), 3.87 (s, 3H), 4.36 (s, 4H), 6.47 (d, J = 9 Hz, 1H), 6.79 (d, J = 9 Hz, 1H).
      MASS (m/z) 389 (M+).
          (Step B) Synthesis of Compound 3 and Compound 4
        • In 120 ml of methanol was suspended 3.9 g (10 mmol) of Compound 3a obtained in Step A, 1.7 ml (20 mmol) of 70% perchloric acid, and 4.3 g (16 mmol) of mercury chloride (HgCl2) were added thereto, and the mixture was stirred for 4 hours. The mixture was diluted with methylene chloride and was filtered through Celite, the filtrate was poured into a saturated aqueous solution of sodium bicarbonate, and the mixture was extracted with methylene chloride. The organic layer was washed with brine and dried over sodium sulfate, and the solvent was evaporated. The residue was purified by silica gel column chromatography (eluted with hexane/ethyl acetate = 1/1) to give the crude Compound 3 as a white solid and also to give 0.18 g (5.5%) of Compound 4 as a colorless transparent oily substance. Compound 3 was further recrystallized from ethyl acetate to give 0.57 g (17%) of white crystals.
          Compound 3
          Melting Point: 123 – 124 °C
          1H-NMR (CDCl3, δ, ppm) 1.75-2.22 (m, 6H), 2.27-2.51 (m, 3H), 3.71 (s, 3H), 3.88 (s, 3H), 4.36 (s, 4H), 6.48 (d, J = 9 Hz, 1H), 6.84 (d, J = 9 Hz, 1H).
          MASS (m/z) 331 (M+).
          Compound 4
          1H-NMR (CDCl3, δ, ppm) 1.92-2.38 (m, 8H), 2.70-2.88 (m, 1H), 3.69 (s, 3H), 3.87 (s, 3H), 4.36 (s, 4H), 6.48 (d, J = 9 Hz, 1H), 6.81 (d, J = 9 Hz, 1H).
          MASS (m/z) 331 (M+).

Example 5.

cis-4-Cyano-4-(8-methoxy-1,4-benzodioxan-5-yl)cyclohexanecarboxylic acid (Compound 5)

  • To a mixture of 0.55 g (1.7 mmol) of Compound 3 obtained in Example 4 and 3.3 ml of methanol was added 3.3 ml of THF to dissolve them. To the mixture was dropwise added 2.6 ml of a 1.3 mol/liter aqueous solution of potassium hydroxide, followed by stirring at room temperature for 1 hour. The mixture was poured into water, ethyl acetate was added thereto, and an aqueous layer was extracted. The aqueous layer was acidified with a 1 mol/liter aqueous hydrochloric acid, and the precipitated solid was collected by filtration and re-slurried with ethanol to give 0.45 g (86%) of Compound 5 as a white solid.
    Melting Point: 228 – 230 °C
    1H-NMR (DMSO-d6 , δ, ppm) 1.59-1.90 (m, 4H), 1.94-2.10 (m, 2H), 2.20-2.45 (m, 3H), 3.75 (s, 3H), 4.27 (dd, J = 5, 12 Hz, 4H), 6.60 (d, J = 9 Hz, 1H), 6.79 (d, J = 9 Hz, 1H), 12.2 (br s, 1H).
    MASS (m/z) 317 (M+).
    Elemental analysis: C17H19NO5
    Found (%) C 64.09, H : 6.01, N : 4.51
    Calcd. (%) C 64.34, H : 6.03, N : 4.41
………….
  1. Teixeira, M. M.; Gristwood, R. W.; Cooper, N.; Hellewell, P. G. Trends Pharmacol. Sci.1997, 18, 164– 170, [PubMed],
  2. Dyke, H. J.; Montana, J. G. Exp. Opin. Investig. Drugs 2002, 11, 1– 13
    Lipworth, B. J. Lancet 2005, 365 ( 9454) 167– 175
    Kroegel, C.; Foerster, M. Exp. Opin. Investig. Drugs 2007, 16, 109– 124
  3. Yanagawa, K. The 26th Medicinal Chemistry Symposium, 2007, 2P-29.
    Ohshima, E.;Yanagawa, K.; Manabe, H.; Miki, I.; Masuda, Y. PCT Int. Appl. WO0164666 A1, 2001.
  4. Christensen, S. B.; Guider, A.; Forster, C. J.; Gleason, J. G.; Bender, P. E.; Karpinski, J. M.; DeWolf, W. E., Jr.; Barnette, M. S.; Underwood, D. C.; Griswold, D. E.; Cieslinski, L. B.; Burman, M.; Bochnowicz, S.; Osborn, R. R.; Manning, C. D.; Grous, M.; Hillegas, L. M.; O’Leary-Bartus, J.; Ryan, M. D.; Eggleston, D. S.; Haltiwanger, R. C.; Torphy, T. J. J. Med. Chem. 1998, 41, 821– 835
  5. Caron, S.; Vazquez, E.; Wojcik, J. M. J. Am. Chem. Soc. 2000, 122, 712– 713
    Culkin, D. A.; Hartwig, J. F. Acc. Chem. Res. 2003, 36, 234– 245
    You, J.; Verkade, J. G. Angew. Chem., Int. Ed. 2003, 42, 5051–5053
  6. Muratake, H.; Natsume, M. Tetrahedron 1990, 46, 6331– 6342
    Caron, S.; Vazquez, E. Org. Process Res. Dev. 2001, 5, 587– 592
  7. North, M. In Comprehensive Organic Functional Group Transformations; Katritzky, A. R.;Meth-Cohn, O.; Rees, C. W., Eds.; Pergamon: Oxford, 1995; Vol. 3, p 614.
  8. Lemaire, M.; Doussot, J.; Guy, A. Chem. Lett. 1988, 1581– 1584
    Guy, A.; Doussot, J.; Guette, J.-P.; Garreau, R.; Lemaire, M. Synlett 1992, 821– 822
    Kurti, L.; Czako, B.; Corey, E. J. Org. Lett. 2008, 10, 5247– 5250
  9. Yanagisawa, A.; Nezu, T.; Mohri, S. Org. Lett. 2009, 11, 5286– 5289
  10. Brown, H. C.; Cole, T. E. Organometallics 1983, 2, 1316– 1319
  11. Kuivila, H. G.; Nahabedian, K. V. J. Am. Chem. Soc. 1961, 83, 2159– 2163
    Kuivila, H. G.; Nahabedian, K. V. J. Am. Chem. Soc. 1961, 83, 2164– 2166
    Nahabedian, K. V.; Kuivila, H. G. J. Am. Chem. Soc. 1961, 83, 2167–2174
  12. Stang, P. J.; Fisk, T. E. Synthesis 1979, 438– 440
    Stang, P. J.; Treptow,W. Synthesis 1980, 283– 284
    Saulnier, M. G.; Kadow, J. F.; Tun, M. M.; Langley, D. R.; Vyas, D. M. J. Am. Chem. Soc. 1989, 111, 8320– 8321
  13. Kato, S.; Chujo, I.; Suzuki, K. PCT Int. Appl. WO04000795 A1, 2004.
  14. Miyaura, N.; Suzuki, A. Chem. Rev. 1995, 95, 2457– 2483
    O’Keefe,D. F.; Dannock, M. C.; Marcuccio, S. M. Tetrahedron Lett. 1992, 33, 6679– 6680
    Segelstein, B. E.; Bulter, T. W.; Chenard, B. L. J. Org. Chem. 1995, 60, 12– 13
    Kong, K.-C.; Cheng, C.-H. J. Am. Chem. Soc. 1991, 113, 6313– 6315
  15. Brands, K. M. J.; Davies, A. J. Chem. Rev. 2006, 106, 2711– 2733
  16. Baker, W.; Jukes, E. H. T.; Subrahmanyam, C. A. J. Chem. Soc. 1934, 1681–1684
    Dallacker, F.; Van Wersh, J. Chem. Ber. 1972, 105, 3301–3305
WO1998022455A1 * Nov 19, 1997 May 28, 1998 Michio Ichimura Oxygenic heterocyclic compounds
JP10147585A * Title not available
WO2001064666A1 * Mar 2, 2001 Sep 7, 2001 Kyowa Hakko Kogyo Kk Oxygen-containing heterocyclic compounds
WO2002059105A1 * Jan 25, 2002 Aug 1, 2002 Kyowa Hakko Kogyo Kk Styrene derivatives and process for production thereof
WO2006041120A1 * Oct 13, 2005 Apr 20, 2006 Daisuke Harada Pharmaceutical composition
WO2006041121A1 * Oct 13, 2005 Apr 20, 2006 Daisuke Harada Remedies/preventives for chronic skin disease
WO2006123726A1 * May 18, 2006 Nov 23, 2006 Daisuke Harada Pharmaceutical composition
WO2011134468A1 Apr 28, 2011 Nov 3, 2011 Leo Pharma A/S Biaryl phosphodiesterase inhibitors
EP1362853A1 * Jan 25, 2002 Nov 19, 2003 Kyowa Hakko Kogyo Co., Ltd Styrene derivatives and process for production thereof

Filed under: Uncategorized

Immune cells regulate blood stem cells

$
0
0

Originally posted on lyranara.me:

Immune cells regulate blood stem cells

Blood stem cell cultures: Blood stem cells from colonies (cell clusters) in vitro consisting of different blood cells. Nine blood stem cell colonies are illustrated in the image, which have developed into differentiated cell types, particularly into white blood cells (leukocytes).Credit: Department of Clinical Research of the University of Bern, Tumor-Immunology research group

Researchers in Bern,  Switzerland have discovered that, during a viral infection, immune cells control the blood stem cells in the bone marrow and therefore also the body’s own defences. The findings could allow for new forms of therapy, such as for bone marrow diseases like leukaemia.

View original


Filed under: Uncategorized

Vertex Pharmaceuticals: Another Step Forward For Kalydeco

$
0
0

Originally posted on Orphan Druganaut Blog:

.

On February 21st, Vertex Pharmaceuticals announces that the FDA approves a supplemental New Drug Application (sNDA) for orphan drug Kalydeco (Ivacaftor) for people with Cystic Fibrosis (CF), ages 6 and older, who have one of the 8 additional mutations in the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) gene :

•   G178R
•   S549N
•   S549R
•   G551S
•   G1244E
•   S1251N
•   S1255P
•   G1349D.

Kalydeco receives approval from the FDA in January 2012 for CF patients, ages 6 and older who have at least one copy of the G551D mutation. Thus, Kalydeco is currently approved for 9 mutations. The new approval affects approximately 150 in the United States.

View original


Filed under: Uncategorized

RAMELTEON, TAK 375 ..Melatonin MT1/MT2 receptor agonist

$
0
0

RAMELTEON

ACN-S001714, ZINC00007031 
  • HSDB 7787
  • Ramelteon
  • Rozerem
  • TAK-375
  • UNII-901AS54I69
Molecular Formula: C16H21NO2   Molecular Weight: 259.34344
CAS number 196597-26-9
 (S)-N-[2-(1,6,7,8-tetrahydro-2H-indeno-[5,4-b]furan-8-yl)ethyl]propionamide

(5)-N-[2-(l,6,7,8-tetrahydro-2H-indeno-[5,4-ό]furan-8- yl)ethyl]propionamide

United States US 6034239 1999-07-22 expiry 2019-07-22

EP885210B1 , EP1792899A1 and J. Med Chem. 2002, 45, 4222-4239

read all at

http://www.allfordrugs.com/2014/02/23/ramelteon-tak-375-melatonin-mt1mt2-receptor-agonist/


Filed under: GENERIC DRUG Tagged: ramelteon

Regulatory Considerations for Biosimilars

$
0
0

Biological medicines are already becoming an increasingly important part of health care. With patent expiries on originator biological products, biosimilars are also increasingly become a part of this future. In fact, by 2020 twelve of the top-selling biologicals will have lost patent protection, opening up an estimated US$24 billion in EU sales and US$30 billion in US sales.

Biologicals have potential to reach up to 50% share in global pharmaceutical market in the next few years.

India is one of the leading contributors in the world biosimilar market and is the third-largest in the Asia-Pacific region, after Australia and China. India has demonstrated high acceptance of biosimilars, which is reflected in the 40 biologicals marketed in India, of which 25 are biosimilars The Indian biotechnology industry is also gaining momentum, with revenues of over US$4 billion in 2011, and which are projected to reach up to US$580 million by 2012.

While small molecule drugs are ideal for generics replication, biological drugs are not so simple. Biological drugs are usually large, complex molecular structures derived from or produced through a living organism, making them very difficult to replicate

Currently there is considerable interest in the legislative debate around generic biological drugs or “biosimilars” in the EU and US due to the large, lucrative market that it offers to the industry. While some countries have issued a few regulatory guidelines as well as product specific requirements, there is no general consensus as to a single, simple mechanism similar to the bioequivalence determination that leads to approval of generic small molecules all over the world. The inherent complex nature of the molecules, along with complicated manufacturing and analytical techniques to characterize them make it difficult to rely on a single human pharmacokinetic study for assurance of safety and efficacy.

In general, the concept of comparability has been used for evaluation of the currently approved “similar” biological where a step by step assessment on the quality, preclinical and clinical aspects is made. In India, the focus is primarily on the availability and affordability of life-saving drugs. In this context every product needs to be evaluated on its own merit irrespective of the innovator brand. The formation of the National Biotechnology Regulatory Authority may provide a step in the right direction for regulation of these complex molecules. However, in order to have an efficient machinery for initial approval and ongoing oversight with a country-specific focus, cooperation with international authorities for granting approvals and continuous risk-benefit review is essential. Several steps are still needed for India to be perceived as a country that leads the world in providing quality biological products.

We are now in the twenty-fifth anniversary year of the Drug Price Competition and Patent Term Restoration Act of 1984 (better known as the Hatch Waxman Act), the landmark US regulation that jump-started the generic pharmaceutical industry. The legislation provided the required impetus to make not just cheaper price alternative medicines available to US consumers but stimulated the emergence of the Indian pharmaceutical industry which is now the dominant supplier of generic drugs to the USA.

The regulatory pathway for bringing generic drugs to market is the abbreviated new drug application (ANDA) process which relies on proving bioequivalence to the listed reference product and showing equivalent product quality. Since duplication of proof of safety and efficacy in the preclinical and clinical setting is not required, there are significant cost savings in bringing a copy of a small chemical molecule to market. This model has been so successful in economic terms that almost 7 out of 10 prescriptions in the US are now generic and for the vast majority of products there is no concern in substitution of a generic equivalent for a brand-name prescription.1

The success story of generic small molecule drugs has stimulated interest in the pharmaceutical and biotech industry for applying an analogous approach towards the highly lucrative biologics business. But biologic drugs are very different from small molecules both in their final form and in the process required to produce and control their quality. It is therefore difficult to find a simple, precise “regulatory” definition of biologics. However, biologics are generally understood to be drugs derived from an organic source. Thus, biologics may be obtained or created from living organisms, either naturally or via genetic manipulation or are manufactured from building blocks of living organisms. They demonstrate considerable molecular complexity and may comprise a diversity of molecular forms. Their larger size and heterogeneity make it difficult for complete characterization via physicochemical analysis which is possible for synthetic chemical entities. In general, biologic drugs are more expensive and the cost of a yearly treatment may run into thousands of dollars for some. They are therefore ideal targets for developing cheaper alternatives.

US FDA definition – A “biological product” means a virus, therapeutic serum, toxin, antitoxin, vaccine, blood, blood component or derivative, allergenic product, or analogous product, or arsphenamine or derivative of arsphenamine (or any other trivalent organic arsenic compound), applicable to the prevention, treatment, or cure of a disease or condition of human beings (Public Health Service Act Sec. 351(i)).

Given the complexity of the final biologic product, it is clear that the nature of the manufacturing process is also complicated. In addition to aspects that are disclosed in regulatory applications, there may still be several aspects which might be held as trade secrets, thereby making it practically impossible for another company to make an identical copy of a biologic drug. While changing a host cell line or vector will definitely impact the product, effects of minor changes like temperature used in the manufacturing process may have an effect on the final characteristics of the biologic drug, including its safety and efficacy. It has been stated often that for a biologic, “the process defines the product”. Thus, while it may be possible to make a similar product, it may not be truly bioequivalent. As a result, even the term used to describe these similar biologic drugs has not been standardized globally. While the parallel term for a biologic generic may intuitively be “biogeneric”, the accepted term in Europe and Canada is “biosimilar” and the preferred term in the US is “follow-on biologic”.

Given these differences among innovator biologics and their “similar” counterparts, there is considerable hesitation on the part of the regulatory agencies to follow an abbreviated approval path similar to one widely used for generic small molecules.

Table 1

Table 1
Some Approved biosimilars

Status of biosimilar regulation in Europe

EMEA Guidelines for Similar Biological Medical Products ( CHMP/437/04, 30 October 2005).2

EMEA’s Guideline on Similar Biological Medicinal Products Containing Biotechnology-derived Proteins as Active Substance: Nonclinical and Clinical Issues (EMEA/ CHMP/BMWP/42832/2005).3

EMEA’s Guideline on Similar Biological Medicinal Products Containing Biotechnology-derived Proteins as Active Substance: Quality Issues, EMEA/CHMP/49348/05.4

In Europe, the Committee for Medicinal Products for Human Use (CHMP), the European Medicines Agency (EMEA) led the way for biosimilars, by issuing its first specific regulatory guidance in October 2005. Two general guidance documents addressing quality and nonclinical and clinical perspectives (June 2006), five product-specific annexes on nonclinical and clinical issues (June-July 2006) and a manufacturing change comparability guideline (November 2007) are now available.

Biosimilars
Testing the bioequivalence of biosimilars differs from that of standard generics. Bioequivalence testing procedures for biosimilars are to be performed against the originator product as a control (reference) and include preclinical and clinical testing [2].

In the Biologics Price Competition and Innovation (BPCI) Act, a biosimilar product is defined as a product that is ‘highly similar’ to the reference product, notwithstanding minor differences in clinically inactive components and there are no clinically meaningful differences in terms of safety, purity and potency. However, little or no discussion regarding how similar is considered ‘highly similar’ is given in the BPCI Act.

For biosimilars, most of which have long half-lives, crossover study would be ineffective and unethical. This is due to the fact that a crossover study requires a wash-out period (which would be long for biosimilars with long half-lives) where the patient is not allowed to take the drug and therefore will have no treatment for their condition. On the other hand, parallel-group studies are required, but these studies do not provide an estimate of within-subject variation. For a parallel-group study, each drug is administered to a different group of subjects. Thus, we can only estimate total variance (between and within-subject variances), not individual variance components. This makes an evaluation of interchangeability difficult.

Statistical tests that may be used to asses biosimilarity are Shuirmann’s two one-sided tests procedure or the confidence interval approach.

Status of Biosimilar Regulation in US

In US, in March 2009, Representative Henry Waxman introduced H.R. 1427 to the Congress “Promoting Innovation and Access to Life-saving Medicines Act”, which authorizes FDA to approve follow-on biologics in an abbreviated manner. It has market exclusivity clauses with time frames similar to ones used currently for drugs. Other bills are expected to follow in the 2009 legislative agenda in order to establish a pathway for approval of these follow-on biologics. The contentious issues as expected, are focused around the duration of exclusivity benefits granted to innovators. The issue of substitutability of followon biologics for reimbursement is also an important one as the legislators debate the merits of each bill.

Korea and Singapore have released draft guidelines on biosimilars in 2009. The Singapore guideline is derived mainly from the EMEA guidelines and defines a similar biological/ biosimilar product as “a biological medicinal product referring to an existing registered product, submitted for medicinal product registration by an independent applicant, and is subject to all applicable data protection periods and/or intellectual property rights for the original product”. In addition to specifying the requirements for biosimilars, the guidance requires that the product have prior approval in countries such as Australia, Canada, EMEA or US.

Indian scenario

The Indian biotech industry is a thriving industry which got its start from vaccine manufacturing. In addition to meeting domestic demands, the Indian vaccine industry also fulfils export requirements to a large extent. Therefore it is evident that manufacturing expertise in producing biologic products of required export quality already exists in the country. What is not readily evident is whether these products can prove to be “comparable” to innovator products when we look into all categories of biologics.

The evolution of regulations governing pharmaceuticals in India has historically been driven by the need to make essential medicines accessible to patients. Access encompasses availability and affordability. It applies to medicines for all indications, acute and chronic illness, small molecules and biologics alike. The absence of product patent regulations for drugs marked a period in the country’s history where it was imperative to make inexpensive medicinal products available to the masses – it did not matter whether these products were innovator-made or copies thereof. In the post-TRIPS era however, there is need to offer and enforce adequate protections for patentable drugs, particularly biologics that inherently involve huge investments in R & D, manufacture and clinical development.

Today, several biologics have been approved in India , including recombinant human insulin, recombinant human erythropoietin (EPO), interferon (IFN), granulocyte colony stimulating factor (GCSF). The versions of biologics available in India are typically products whose patents have expired or do not exist in India. Therefore, from a technical standpoint, there is no concern about patent infringement regarding these (there are no patents in India for these products). If a biosimilar results in a price drop of 30%, it is a significant improvement to patients who may now be able to afford this generic version of a life-saving drug. In many ways, the debate about biosimilars that rages across the developed world and regulated markets is irrelevant to India where the central concern revolves around access.

Partly due to the dearth of appropriate resources and experience, Indian regulators have sought to mimic regulations already in use in the developed world without much customization. A host of agencies have been created to address the issues brought forth by biologics.

Basic facts about biosimilars.

Biotechnological drugs have become an essential part of modern pharmacotherapy and are expected to reach a 50% share in the pharmaceutical market in the next few years. The expiry of patent protection for many original biotechnological medicines has led to the development of what are called biosimilars or follow-on biologics. Biosimilars attempt to copy the original technology leading to the production of innovative biotechnological medicines to obtain a product which is similar to the original one. The first two biosimilars have recently been approved in the European Union and one application was rejected. Many more biosimilars will likely see approval in the near future. Our experience with biosimilars has been very limited to date and long-term safety data including immunogenicity are not available. Although biosimilars will likely lower the cost of modern therapies there are issues which have to be discussed at this stage among physicians regarding in particular the differences between biosimilars and generics of the classical chemical drugs, need for appropriate regulations as well as identification of potential problems with biosimilars. Other specific problems which will also be addressed in this review are safety of biosimilars, pharmacovigilance, automatic substitution, naming and labeling/prescription rules. 7

List of agencies

  • Indian Council for Medical Research (ICMR)
  • Central Drugs Standard Control Organisation (CDSCO)
  • Department of Biotechnology (DBT)
  • Genetic Engineering Approval Committee (GEAC)
  • Recombinant DNA advisory Committee (RDAC)
  • Review Committee on Genetic Manipulation (RCGM)
  • Institutional Biosafety Committee (IBSC)
  • National Centre for Biological Sciences
  • National Control Laboratory for Biologicals

Notwithstanding the above, there is clarity on the fact that biologics and drugs need to be scrutinized differently. With this in mind, the DBT has been given the mandate to set up the National Biotechnology Regulatory Authority (NBRA). This is envisaged as an independent, autonomous and professionally led body to provide a single window mechanism for biosafety clearance of genetically modified products and processes.

Before such an organization can be effectively implemented, it will be necessary to put in place appropriate new legislation, namely the “National Biotechnology Regulatory Act” or the NBR Act. Draft establishment plan and “Draft National Biotechnology Regulatory Bill, 2008” are currently available on the DBT website for comments. The responsibility of consolidating the feedback has been entrusted to Biotech Consortium India Limited (BCIL). The draft bill envisions the scope of this authority to encompass research, manufacture, import and use of genetically engineered organisms and products derived thereof.

Biosimilars: how similar or dissimilar are they?

The imminent expiry of patents on biological medicinal products, such as epoetin alfa in 2006, has significant implications for nephrology in Australia. The purpose of this review is to examine the differences between biosimilars (similar biological medicinal products) and generic low molecular weight (chemical) drugs. The approach that regulatory agencies, including the European Medicines Agency (EMEA) and the Therapeutic Goods Administration (TGA), are taking towards biosimilars is also discussed. Biosimilars differ from generic chemical drugs in many important ways, including the size and complexity of the active substance, the nature of the starting materials (cell banks, tissues and other biological products), and the complexity of the manufacturing processes. Therefore, it has been acknowledged by the EMEA that established legal and regulatory principles of ‘essential similarity’ that are applied to standard chemical generics cannot be readily applied to biosimilars. One of the key areas of concern with the introduction of biosimilars into the field of nephrology will be guaranteeing the safety and efficacy of biosimilars. New manufacturers will need to ensure that their biopharmaceutical has a similar efficacy and safety profile to the innovator product through more extensive clinical trials than the limited testing done for generic versions of low molecular weight chemical medicines. 6

Safety
The primary importance of the manufacturing process was highlighted when a slight change in the production process of an originator recombinant erythropoietin resulted in patients developing pure red cell aplasia.

To try to address this possible safety issue, guidelines from EMA on comparability of biosimilars state that preclinical data must be insufficient to demonstrate the immunological safety of some biosimilars. This means that safety must be demonstrated in cohorts of patients enrolled in clinical trials and using post marketing surveillance.

The challenge of biosimilars.

The purpose of this report was to review issues associated with the introduction of alternative versions of biosimilars used in the oncology setting.

Data were obtained by searches of MEDLINE, PubMed, references from relevant English-language articles, and guidelines from the European Medicines Agency.

When biosimilars are approved in EU, they will be considered ‘comparable’ to the reference product, but this does not ensure therapeutic equivalence. Inherent differences between biosimilars may produce dissimilarities in clinical efficacy, safety, and immunogenicity. Switching biosimilars should be considered a change in clinical management. Regulatory guidelines have been established for some biosimilar categories but, because of the limited clinical experience with biosimilars at approval, pharmacovigilance programs will be important to establish clinical databases. Guidelines also provide a mechanism for the extrapolation of clinical indications (approved indications for which the biosimilar has not been studied). This may be of concern where differences in biological activity can result in adverse outcomes or when safety is paramount (e.g. stem cell mobilization in healthy donors). These issues should be addressed in biosimilar labeling.

Biosimilars should provide cost savings and greater accessibility to biopharmaceuticals. A thorough knowledge surrounding biosimilars will ensure the appropriate use of biopharmaceuticals.

Pharmacovigilance
Due to the limited clinical database at the time of approval of a biosimilar, vigorous pharmacovigilance is required. EMA guidelines require pharmacovigilance programmes to monitor the safety of biosimilar products post-approval.

Substitution
For small molecule generics the issue of substitution is easy, since they are considered identical to the originator molecule. This, however, is not the case for biosimilars, which are large complex molecules prone to heterogeneity.

In the US, the BPCI Act gives FDA the authority to designate a biosimilar as interchangeable with its reference product. This means that the biosimilar may be substituted for the originator product by the pharmacist without reference to the prescribing physician. This is not the case, however, in the EU, where decisions on interchangeability are not made by EMA, but at a national level.

Global concerns regarding product safety and quality

Every drug/biologic manufacturer needs to own the responsibility for putting a high quality, safe drug on the market, after appropriate review and approval by the concerned regulatory authority. While the safety of original biologics products is assured by the innovator by adherence to rigorous standards required for approval, the resistance towards biosimilars on the part of regulators, stems from the concern that an abbreviated approval process may not be adequate to ensure safe performance of the product in the market. For a manufacturer looking to get into the biosimilar market, he needs to overcome major challenges in making a complex product, getting regulatory approval by satisfying stringent criteria and then selling it in the market. Typically, facilities required for manufacture of biologicals are very expensive and the kind of infrastructure required to meet high regulatory expectations is limited to only a few companies. Clinical trial expenditure and ongoing analysis requires compliance to pharmacopoeial monographs when available and access to reference standards, which are not always available.

The cornerstone of generic drug approvals has been the concept of bioequivalence, using equivalence of pharmacokinetic parameters as surrogates for clinical efficacy. But in the context of biosimilars, the concept of comparability is the one used to make such an evaluation. Comparability protocols are used for chemistry, manufacturing and controls (CMC) sections to make the case on the quality aspects of the product. Preclinical testing requires knowledge of study designs used by innovator in order to truly compare performance of the biosimilar. For clinical evaluation, at least one clinical comparability trial is required to demonstrate comparability (non-inferiority in terms of efficacy to innovator and comparable safety profile). But long term safety issues remain unaddressed for biosimilars, requiring thorough postmarketing studies and pharmacovigilance and adequate risk management plans.

In terms of preclinical studies, for biologics, pharmacodynamic endpoints are more relevant than pharmacokinetics, which is the key measure with small molecules. For animal safety studies, choice of appropriate animal species and duration of studies are important criteria for proving comparability. Clinically, comparative PK/PD study is required to compare the reference and biosimilar product. However, clinical trial design selection and a thorough understanding and a priori statement of margins chosen for comparability must be stated for meaningful evaluation of data.

Follow-on biologics: challenges of the “next generation”.

The imminent patent expiration of many biopharmaceutical products will produce the possibility for generic versions of these therapeutic agents (i.e. biosimilars). However, there are a number of issues that will make approval of biosimilars much more complicated than the approval of generic equivalents of conventional pharmaceuticals. These issues centre on the intrinsic complexity of biopharmaceutical agents, which are recombinant proteins in most cases, and the heterogeneity of proteins produced by different manufacturing processes (i.e. differences in host cells, purification and processing, formulation and packaging). The increased occurrence of antibody (Ab)-mediated pure red cell aplasia (PRCA) associated with a change in the formulation of one particular epoetin-alpha product highlights the potential for increased immunogenicity of recombinant proteins with different formulations, or those manufactured by different processes. Thus, verification of the similarity to or substitutability of biosimilars with reference innovator biopharmaceutical products will require much more than a demonstration of pharmacokinetic similarity, which is sufficient for conventional, small molecule generic agents. Regulatory requirements for the approval of biosimilars have not yet been fully established, but preliminary guidelines from the European Agency for the Evaluation of Medicinal Products (EMEA) state that the complexity of the product, the types of changes in the manufacturing process, and differences in quality, safety and efficacy must be taken into account when evaluating biosimilars. For most products, results of clinical trials demonstrating safety and efficacy are likely to be required. In addition, because of the unpredictability of the onset and incidence of immunogenicity, extended post-marketing surveillance is also important and may be required. 10

Statistical assessment of biosimilar products.

Biological products or medicines are therapeutic agents that are produced using a living system or organism. Access to these life-saving biological products is limited because of their expensive costs. Patents on the early biological products will soon expire in the next few years. This allows other biopharmaceutical/biotech companies to manufacture the generic versions of the biological products, which are referred to as follow-on biological products by the U.S. Food and Drug Administration (FDA) or as biosimilar medicinal products by the European Medicine Agency (EMEA) of the European Union (EU). Competition of cost-effective follow-on biological products with equivalent efficacy and safety can cut down the costs and hence increase patients’ access to the much-needed biological pharmaceuticals. Unlike for the conventional pharmaceuticals of small molecules, the complexity and heterogeneity of the molecular structure, complicated manufacturing process, different analytical methods, and possibility of severe immunogenicity reactions make evaluation of equivalence (similarity) between the biosimilar products and their corresponding innovator product a great challenge for both the scientific community and regulatory agencies. In this paper, we provide an overview of the current regulatory requirements for approval of biosimilar products. A review of current criteria for evaluation of bioequivalence for the traditional chemical generic products is provided. A detailed description of the differences between the biosimilar and chemical generic products is given with respect to size and structure, immunogenicity, product quality attributed, and manufacturing processes. In addition, statistical considerations including design criteria, fundamental biosimilar assumptions, and statistical methods are proposed. The possibility of using genomic data in evaluation of biosimilar products is also explored.15

A way forward for India

In today’s scenario, India needs to focus on quality of each biological product per se, whether that is demonstrated through comparability or by its own merit; and assurance of safety through appropriate regulatory review and approval of available data.

Irrespective of the authority entrusted to oversight of biologics, the debate on appropriate level of regulatory scrutiny for biologics will continue to focus on requiring adequate characterization while balancing cost, with the overall goal of having a much needed product on the market with reasonable assurance of efficacy and safety. Intense discussion on publication of appropriate monographs in the Indian Pharmacopeia and availability of reference standards continues amidst regulatory circles. Indian manufacturers have always sought to enter new markets and have voluntarily raised the bar in order to secure approvals for their products in the regulated markets where profit margins are high. From a facility infrastructure and systems point of view, most companies eyeing the regulated markets for their products will most likely fulfil expectations. State-of-the art analytical techniques are available within the industry. Therefore from a quality standpoint, biologic products made in India should not have any trouble in meeting market expectations. However, physicochemical characterization of a biologic product and compliant facilities form only one part of the evaluation required to demonstrate product comparability.

The practical way forward for approval of biosimilar products in India would have to be unique to the Indian context while staying rooted to scientific basics and keeping in mind the needs and limitations of the country. The large majority of biosimilars introduced in India would be products whose patents have expired and where the “original innovator” product may not be approved in the country. It is also possible that no patent exists in India for some products and therefore , originator and similars coexist. For all products, the question of available reference standards and monographs would continue to remain. The next wave of biologics of commercial interest to the industry will become a burning issue where the regulator cannot expect to wait to see how the legislation is crafted in the US or elsewhere before making a move.

In my opinion, it seems that India, having the benefit of in-house (in-country) expertise in the area, should utilize the various agencies currently entrusted with splintered tasks and responsibilities to come up with working group or taskforce whose goal is to develop product-specific guidelines for approval. These can be developed using available worldwide regulatory knowledge by signing appropriate MOUs if necessary, studying the scientific literature and current industry standards and practice with respect to characterization, focusing on specific areas of unique concern for each product and proposing an approval path. These guidelines can be widely disseminated in the community. There will still be grey areas that need clarification and in such cases, a system for formal meeting with members of the working group/taskforce can be instituted, similar to the scientific advice that is currently available through the EMEA or individual European country competent authorities.

As a nation that takes pride in being the “exporter to the world” in the arena of pharmaceuticals, it behooves not just the regulators but all those in the regulatory affairs profession in India to support such initiatives to make life-saving products available to our countrymen that are unquestionably of the highest standards in terms of quality, safety and efficacy such that we become the supplier of choice when it comes to exporting biosimilars to markets in every corner of the world.

Biosimilar therapeutics-what do we need to consider?

Patents for the first generation of approved biopharmaceuticals have either expired or are about to expire. Thus the market is opening for generic versions, referred to as ‘biosimilars’ (European Union) or ‘follow-on protein products’ (United States). Healthcare professionals need to understand the critical issues surrounding the use of biosimilars to make informed treatment decisions.The complex high-molecular-weight three-dimensional structures of biopharmaceuticals, their heterogeneity and dependence on production in living cells makes them different from classical chemical drugs. Current analytical methods cannot characterize these complex molecules sufficiently to confirm structural equivalence with reference molecules. Verification of the similarity of biosimilars to innovator biopharmaceuticals remains a key challenge. Furthermore, a critical safety issue, the immunogenicity of biopharmaceuticals, has been highlighted in recent years, confirming a need for comprehensive immunogenicity testing prior to approval and extended post-marketing surveillance.Biosimilars present a new set of challenges for regulatory authorities when compared with conventional generics. While the demonstration of a pharmacokinetic similarity is sufficient for conventional, small-molecule generic agents, a number of issues will make the approval of biosimilars more complicated. Documents recently published by the European Medicines Agency (EMEA) outlining requirements for the market approval of biosimilars provide much-needed guidance. The EMEA has approved a number of biosimilar products in a scientifically rigorous and balanced process. Outstanding issues include the interchangeability of biosimilars and innovator products, the possible need for unique naming to differentiate the various biopharmaceutical products, and more comprehensive labelling for biosimilars to include relevant clinical data. 5

Biosimilars: policy, clinical, and regulatory considerations.

The regulatory background surrounding biosimilars (biopharmaceuticals that are considered similar in composition to an innovator product, but not necessarily clinically interchangeable); equivalence, interchangeability, and unique considerations associated with biopharmaceuticals; the biopharmaceutical protein production process; scientific facts for use in the policy discussion about biosimilars; the European Union system for biosimilars; and the current status of biosimilars legislation in the United States are described.

An abbreviated regulatory pathway for the approval of biosimilars, and a process for safely demonstrating the therapeutic interchangeability of these proteins, has the potential to provide meaningful cost savings. This economic advantage to patients can translate into important public health benefits. But to date, no formal regulatory process exists in the United States for bringing these drugs to market. In addition, the current tools for fully characterizing biopharmaceuticals are not–in certain cases–well developed, especially for proteins that have complex structures or are heavily glycosylated. In addition, using “similar” but not completely “identical” proteins interchangeably raises concerns about potentiating immunogenicity. The bottom line is that demonstrating therapeutic equivalence and interchangeability for biosimilars is not a straightforward matter–it cannot be based on the same criteria as for conventional small-molecule drugs. The science, while obtainable, is more complex. For example, it is assumed that showing that a biosimilar protein can be safely used interchangeably with an innovator protein would require, at the least, some limited clinical data and interchangeability studies. Notwithstanding the more complex scientific and clinical issues particular to protein products, most believe that a process for enabling the approval of safe and effective biosimilar proteins is not only possible, but an important public health goal. The European Union system for biosimilars may provide a model for anticipating and resolving the scientific and policy issues related to biosimilars in the U.S. 

The legal and regulatory status of biosimilars remains to be resolved in the United States as policymakers address the scientific and policy issues surrounding product manufacturing, patent terms, and clinical use.

Biosimilars: it’s not as simple as cost alone.

Biosimilars or follow-on biologics (FoB) are biopharmaceuticals that, unlike small molecule generic products, are copies of larger, much more complex proteins. As such, data generated from one biopharmaceutical cannot be extrapolated to another. Unlike small molecule generics, FoB require a full developmental programme, albeit smaller than for an originator product. This has been recognized by European regulatory authorities and it is becoming clear that accelerated processes for FoB marketing approval are not feasible.

To determine the balance between costs surrounding FoB (including relatively extensive developmental programmes and subsequent price to the market) and the necessity to ensure efficacy and safety.

It is important that FoB are sufficiently tested to ensure patient safety is not compromised. Conducting such a development programme followed by sound pharmacovigilance is very challenging and costly.

Cost-savings associated with FoB may be limited. 10

 
Recommendations regarding technical standards for follow-on biologics: comparability, similarity, interchangeability.

Policy makers around the world are currently considering the creation of a regulatory pathway for follow-on biologics (FOB), which will have to account for the substantial technical challenges associated with FOB development. These challenges will likely involve more complexity than comparability assessments of process changes made by the same manufacturer. The history of industry-regulator comparability discussions helps explain why the same degree of testing and flexibility now applied to change-control within a manufacturer’s own process, at this time, cannot be extrapolated to the observed and possibly unknown differences between two manufacturing processes that are independently developed by different (non-collaborating) parties.

This commentary provides recommendations on the technical aspects that should be considered in the creation of an approval pathway for FOB products.

In the authors’ view, analytical methodology in its current state cannot alone provide full assurance that the FOB is sufficiently similar to the innovator product. Moreover, the FOB manufacturer will not have access to the extensive knowledge accumulated by the innovator manufacturer from early development through marketing. Thus, extensive clinical evaluation will likely be necessary to provide assurance that the FOB is safe and efficacious. If such testing demonstrates the FOB is safe and efficacious per existing regulatory standards, the product should receive marketing approval as a ‘similar’ product. Since ‘similarity’ is a fundamentally different determination than establishing interchangeability between the two products, an interchangeability determination must be based on additional testing and market experience to ensure patient safety. Post-marketing surveillance of the FOB should be conducted to ensure that the approved molecule has similar clinical safety and efficacy as the innovator product, prior to any consideration of interchangeability 11

European regulatory guidelines for biosimilars.

The impending arrival en masse of biosimilars on Western markets is placing drug regulatory agencies under pressure to realign their policies. Biosimilars require more rigorous assessments than traditional chemical generics. This is because of the molecular complexity of recombinant proteins, and the complexity of biological manufacturing processes. Small differences can arise in a recombinant protein product which are hard or impossible to detect with even state-of-the-art analytical techniques. Yet, these differences can have significant impact on the safety and efficacy of the drug. The European Medicines Agency (EMEA) has taken the lead in issuing guidelines, most of which are still under review. The guidelines advocate pre-clinical and clinical testing of biosimilars prior to market authorization, complemented by tailored pharmacovigilance plans. These guidelines provide a valuable base from which to develop in this evolving regulatory environment.12
Legislative initiatives in Europe, Canada and the US for market authorization of follow-on biologics.

The formulation and application of legal and regulatory requirements for the market authorization of follow-on versions of biological drugs present challenges. This review discusses relevant regulatory guidelines and legislative initiatives related to market authorization for follow-on biologics in Europe, Canada and the US. The respective positions of these three markets is analyzed with regard to several factors: criteria for the choice of reference products; requirements for the comparability exercise between a candidate follow-on biologic and the selected reference product, with an emphasis on considerations of quality, safety and efficacy data; the interchangeability of a reference product with related follow-on drugs; data exclusivity provisions; and the application of specialized patent enforcement mechanisms to follow-on biologics.13

Quality, safety and efficacy of follow-on biologics in Japan.

Recently, WHO, EU, Japan and Canada have published guidelines on biosimilar/follow-on biologics. While there seems to be no significant difference in the general concept in these guidelines, the data to be submitted for product approval are partially different. Differences have been noted in the requirements for comparability studies on stability, prerequisites for reference product, or for the need of comparability exercise for determination of process-related impurities. In Japan, there have been many discussions about the amount and extent of data for approval of follow-on biologics. We try to clarify the scientific background and rational for regulatory pathway of biosimilar/follow-on biologics in Japan in comparison with the guidelines available from WHO, EU and Canada. In this article, we address and discuss the scientific background underlying these differences to facilitate the harmonization of follow-on biologic principles in the guidelines in future.14

References


Filed under: Biosimilar drugs, Monoclonal antibody Tagged: Biologics, biosimilar, Comparability, Equivalence, Guidelines, Regulations

FDA Approves BMS Drug for Rare Fat Disorder

$
0
0
CHEMICAL NAMES

1. Leptin (human), N-methionyl-

2. N-methionylleptin (human)

STRUCTURAL FORMULA

MVPIQKVQDD TKTLIKTIVT RINDISHTQS VSSKQKVTGL DFIPGLHPIL 50
TLSKMDQTLA VYQQILTSMP SRNVIQISND LENLRDLLHV LAFSKSCHLP 100
WASGLETLDS LGGVLEASGY STEVVALSRL QGSLQDMLWQ LDLSPGC 147

Disulfide bridge location
97-147
http://www.ama-assn.org/resources/doc/usan/metreleptin.pdf
MOLECULAR FORMULA C714H1167N191O221S6

MOLECULAR WEIGHT 16.16 kDa

MANUFACTURER Amylin Pharmaceuticals, Inc.

CODE DESIGNATION r-metHuLeptin

metreleptin

An analog of human leptin, metreleptin, has been approved in Japan and is currently under review by the FDA in the US for the treatment of diabetes and/or hypertriglyceridemia, in patients with rare forms of lipodystrophy, syndromes characterized by abnormalities in adipose tissue distribution, and severe metabolic abnormalities. Bristol-Myers Squibb has submitted a New Drug Approval (NDA) for metreleptin to the US Food and Drug Administration (FDA) Office of Orphan Products Development. In a three-year study of metreleptin in patients with lipodystrophy organized by the National Institute of Diabetes and Digestive and Kidney Diseases at the National Institutes of Health, metreleptin treatment was associated with a significant decrease in blood glucose (A1c decreased from 9.4% at baseline to 7.0% at study end) and triglyceride concentration (from 500 mg/dl at baseline to 200 mg/dl at study end). The Juvenile Diabetes Research Foundation has also partnered with Amylin Pharmaceuticals and researchers at the University of Texas Southwestern Medical Center to study whether metreleptin can be used to improve the treatment of type 1 diabetes.

N-Methionylleptin (human)
Recombinant human OB protein, purified to homogenicity as a 16-kDa monomer

Treatment of obesity and related disorders (metabolic homeostasis regulator)

LAUNCHED 2013 IN JAPAN BI SHINOGI
186018-45-1 CAS NO
BLA STN125390
  • Brand name: Myalept
  • Generic name: metreleptin
  • Company: Amylin Pharmaceuticals, Inc.
  • Treatment for: Lipodystrophy
Feb 25, 2014  FDA Approves Myalept to Treat Generalized Lipodystrophy
Dec 12, 2013 FDA Advisory Committee Votes on Investigational Medicine Metreleptin
Apr  3, 2012 Amylin Completes Biologics License Application for Metreleptin to Treat Diabetes and/or Hypertriglyceridemia in Patients With Rare Forms of Lipodystrophy
Dec 20, 2010 Amylin Submits Clinical and Nonclinical Sections of Rolling Biologics License Application for Metreleptin to Treat Rare Forms of Lipodystrophy

http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/EndocrinologicandMetabolicDrugsAdvisoryCommittee/UCM377929.pdf

Leptin
PDB 1ax8 EBI.jpg
Structure of the obese protein leptin-E100.
FDA approves Myalept to treat rare metabolic disease
On Feb. 24, 2014, the U.S. Food and Drug Administration approved Myalept (metreleptin for injection) as replacement therapy to treat the complications of leptin deficiency, in addition to diet, in patients with congenital generalized or acquired generalized lipodystrophy.Generalized lipodystrophy is a condition associated with a lack of fat tissue. Patients with congenital generalized lipodystrophy are born with little or no fat tissue. Patients with acquired generalized lipodystrophy generally lose fat tissue over time. Because the hormone leptin is made by fat tissue, patients with generalized lipodystrophy have very low leptin levels. Leptin regulates food intake and other hormones, such as insulin.Patients with both types of generalized lipodystrophy often develop severe insulin resistance at a young age and may have diabetes mellitus that is difficult to control or very high levels of triglycerides in the blood (hypertriglyceridemia) that can lead to inflammation of the pancreas.

“Myalept is the first approved therapy indicated for treating the complications associated with congenital or acquired generalized lipodystrophy and provides a needed treatment option for patients with this orphan disease,” said Mary Parks, M.D., deputy director of the Office of Drug Evaluation II in the FDA’s Center for Drug Evaluation and Research.

The safety and effectiveness of Myalept, an analog of leptin made through recombinant DNA technology, were evaluated in an open-label, single-arm study that included 48 patients with congenital or acquired generalized lipodystrophy who also had diabetes mellitus, hypertriglyceridemia, and/or elevated levels of fasting insulin. The trial showed reductions in HbA1c (a measure of blood sugar control), fasting glucose, and triglycerides.

Anti-drug antibodies with neutralizing activity to leptin and/or Myalept may develop, which could result in severe infections or loss of treatment effectiveness. T-cell lymphoma has been reported in patients with acquired generalized lipodystrophy, both treated and not treated with Myalept, so healthcare professionals should carefully consider the benefits and risks of treatment with Myalept in patients with significant hematologic abnormalities and/or acquired generalized lipodystrophy. Myalept is contraindicated in patients with general obesity. Myalept is not approved for use in patients with HIV-related lipodystrophy or in patients with metabolic disease, including diabetes mellitus and hypertriglyceridemia, without concurrent evidence of generalized lipodystrophy.

Because of the risks associated with the development of neutralizing antibodies and lymphoma, Myalept is available only through the Myalept Risk Evaluation and Mitigation Strategy (REMS) Program. Under this REMS program, prescribers must be certified with the program by enrolling in and completing training. Pharmacies must be certified with the program and only dispense Myalept after receipt of the Myalept REMS Prescription Authorization Form for each new prescription.

Myalept is also approved with a Medication Guide and instructions for use that provides patients with important information about the medication. The guide will be distributed each time a patient fills a prescription.

The FDA is requiring seven studies (post-marketing requirements) for Myalept, including a long-term prospective observational study (product exposure registry) of patients treated with Myalept, a study to assess for the immunogenicity (antibody formation) of Myalept, and an assessment and analysis of spontaneous reports of potential serious risks related to the use of Myalept. Eight additional studies are being requested as post-marketing commitments.

In clinical trials, the most common side effects observed in patients treated with Myalept were low blood sugar (hypoglycemia), headache, decreased weight, and abdominal pain.

Myalept is marketed by San Diego-based Amylin Pharmaceuticals, L.L.C.

For more information:

Metreleptin is an analogue of the human hormone leptin being developed by Amylin Pharmaceuticals (a subsidiary of Bristol-Myers Squibb) for the subcutaneous treatment of metabolic disorders including lipodystrophy. The compound is expected to improve insulin sensitivity, hypertriglyceridaemia and hyperglycaemia in patients with lipodystrophy who are unresponsive to conventional treatment.

Metreleptin has been approved in Japan as a leptin therapy for the treatment of lipodystrophy. Amylin has also completed a submission for regulatory approval to the US FDA for metreleptin in the treatment of diabetes mellitus and/or hypertriglyceridaemia in patients with rare forms of lipodystrophy.

Clinical development of the drug is also underway in the USA for the treatment of type 1 diabetes. Amgen was previously assessing the use of metreleptin as a treatment for amenorrhoea; however, it appears that development in this indication has been discontinued. This article summarizes the milestones in the development of metreleptin leading to this first approval for lipodystrophy.

Metreleptin is a leptin replacement therapy first launched in Japan in 2013 for the treatment of congenital lipodystrophy. Amylin filed for approval in the U.S. in 2010 for the treatment of diabetes and/or hypertriglyceridemia in patients with rare forms of lipodystrophy. In 2013, the Endocrinologic and Metabolic Drugs Advisory Committee (EMDAC) recommended the approval for the treatment of pediatric and adult patients with generalized lipodystrophy , but not for partial lipodystrophy.

Phase II clinical studies are also under way at Beth Israel Deaconess Medical Center for the treatment of lipodystrophy syndrome associated with AIDS. The National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) is conducting phase II clinical trials for the treatment of nonalcoholic steatohepatitis. Phase II are ongoing at the National Institute for Diabetes and Digestive and Kidney Diseases for the treatment of non-alcoholic fatty liver disease (NAFLD) associated with lipodystropy. Early clinical studies had also been ongoing for the treatment of leptin deficiencies.

The University Texas Southwestern Medical Center at Dallas is evaluating metreleptin for the treatment of type 1 diabetes. Beth Israel Deaconess Medical Center is conducting phase II clinical trials for the treatment of amenorrhea. Amgen had been conducting clinical trials for this indication and for the treatment of type 1 diabetes and depression; however no recent development has been reported for this research.

In 2011, Amylin and Takeda put on hold their clinical trials with metreleptin in combination with pramlintide for the treatment of obesity in order to investigate an antibody-related laboratory finding. Amylin is currently evaluating the compound as monotherapy for the treatment of obesity. The companies had been conducting phase II clinical trials of metreleptin not in combination with pramlintide for the treatment of obesity; however, no recent development has been reported for this research.

Originally developed at the Rockefeller University, an exclusive license to metreleptin was granted to Amgen in 1995. In 2009, the drug candidate was licensed to Takeda by Amylin worldwide for the treatment of obesity. In 2010, orphan drug designation was assigned in the U.S. for the treatment of metabolic disorders secondary to lipodystrophy and for the treatment of leptin deficiency secondary to generalized lipodystrophy and partial familial lipodystrophy.

In 2012, orphan drug designation was assigned in Japan for the treatment of diabetes or hyperlipidemia due to lipoatrophy. In 2012, orphan drug designation was assigned in the E.U. for the treatment of Barraquer-Simons syndrome, Berardinelli-Seip syndrome, familial partial lipodystrophy and Lawrence syndrome. In 2014, AstraZeneca acquired the global rigths for development, manufacture and commercialization of the product.

……………

EP2195034A2

Other exemplary leptins for use in the methods and compositions described herein include, but are not limited to, the amino acid sequence for mature, recombinant methionyl human leptin (herein called rmetHu-Leptin 1-146 or Metreleptin) having the amino acid sequence:

MVPIQKVQDDTKTLIKTΓVTRINDISHTQSVSSKQKVTGLDFIPGLHPILTLSKMDQTLA VYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHLPWASGLETLDSLGGVLEASG YSTEWALSRLQGSLQDMLWQLDLSPGC (SEQ ID NO:274).


Filed under: FDA 2014, Uncategorized Tagged: FDA2014, metreleptin, MYALEPT

Bhasma : The ancient Indian nanomedicine

$
0
0

Figure 1: Standardization of <i>Bhasma</i>

Bhasma : The ancient Indian nanomedicine

Dilipkumar pal

Department of Pharmaceutical Sciences, Guru Ghasidash Vishwavidyalya (A Central University), Koni, Bilaspur – 495 009, Chhattisgarh 

India

Ayurveda is the science made up of Veda (knowledge) and Ayush (life) i.e. knowledge of life. An Ayurvedic system adopts a holistic approach towards health care by balancing the physical, mental and spiritual functions of the human body. Rasa-Shastra (vedic-chemistry) is one of the parts of Ayurveda, which deals with herbo-mineral/metals/non-metals preparations called Bhasmas. Rasayana (immunomodulation and anti-aging quality) and yogavahi (ability to target drugs to the site) are characteristics of a properly made herbo-mineral/metals/non-metals preparation, which is also nontoxic, gently absorbable, adaptable and digestible in the body

read at http://www.japtr.org/article.asp?issn=2231-4040;year=2014;volume=5;issue=1;spage=4;epage=12;aulast=Pal

Pal D, Sahu CK, Haldar A. Bhasma : The ancient Indian nanomedicine. J Adv Pharm Technol Res [serial online] 2014 [cited 2014 Feb 26];5:4-12. Available from: http://www.japtr.org/text.asp?2014/5/1/4/126980

DOI: 10.4103/2231-4040.126980
more info

Bhasma[1] in Ayurveda has been defined as a substance obtained by calcination.

Use of both bhasma (Residue after incineration – calcined preparation) as well as in pishti (powdered gem or metal) form along with appropriate herbs for treatment of critical ailments is a medicinal preparation in Ayurveda and to some extent Unani (both Indian branches of medical science using natural curative methods. The procedures for preparing these medicines are time-consuming and complicated.

Bhasma is a calcined preparation in which the gem or metal is converted into ash. Gems or metals are purified to remove impurities and treated by triturating and macerating in herbal extracts. The dough so obtained is calcinated to obtain the ashes.[2]^

Bhasma or vibhooti is the sacred ash from the dhuni or fire of a yogi or avadhoota, or from the sacrificial fire or yajna, where special wood, ghee, herbs, grains and other auspicious and purifying items are offered in worship along with mantras. It is believed that bhasma destroys sins (paap), and that it links us with the divine. It is called ‘bhasma’ because it has the power to consume all evils. Any matter, broken up through the process of fire is reduced to its ‘bhasmic’ form, which is infinitely more refined and pure than the original matter, devoid as it is of all impurities niranjan. The grossness of matter obscures the subtle essence inherent within it, just as wood hides fire and milk conceals butter and cheese, but when it is burnt (or churned in the case of milk) only the pure essence remains. Similarly, the great heat of tapasya and the churning of the mind in meditation reveals the underlying subtle spirit or atman.

Vibhuti

In certain circumstances Bhasma, ‘Vibhuti‘ (Sanskrit) and ‘Thiruneeru’ (Tamil) are synonymous.

Bhasmikaran

Bhasmikaran is a process by which a substance which is otherwise bioincompatible is made biocompatible by certain samskaras or processes (Puranik and Dhamankar, 1964e). The objectives of samskara are :- a) elimination of harmful matters from the drug b) modification of undesirable physical properties of the drug c)conversion of some of the characteristics of the drug d) enhancement of the therapeutic action(Puranik and Dhamankar, 1964e). Various steps involved in the preparation of bhasma(or bhasmikaran) are:- 1) Shodhan -Purification, 2) Maran – Powdering, 3) Chalan- Stirring, 4) Dhavan – Washing, 5) Galan- Filtering, 6) Putan- Heating, 7) Mardan- Triturating, 8) Bhavan- Coating with herbal extract, 9) Amrutikaran – Detoxification and 10) Sandharan- Preservation (Puranik and Dhamankar, 1964e). Selection of these steps depends on the specific metal. Sometimes there is an overlapping of the steps e.g. maran is achieved by puttan. Since the present thesis work is on bhasma, Bhasmikaran process is elaborated in details in the following paragraphs.

Steps of bhasmikaran

1. Shodhan: The principle objective of shodhan is to remove unwanted part from the raw material and separate out impurities( Vaiday and Dole 1996b). Metals obtained from ores may contain several impurities, which are removed by subjecting them to Shodhan process. In context of bhasma, shodhan means purifying and making the product suitable for the next step i.e. Maran. Ayurveda classifies shodhan into a) General process and b) Specific process.

General process for shodhan:

“The sheets of metals are heated till red hot and are successively dipped into liquids like oil, buttermilk, cow’s urine etc. The procedure is repeated seven times”.

b. Specific process for shodhan For some metals a specific process is described for shodhan e.g. for purification of Jasad, the molten mass is poured in cow’s milk 21 times (Shastri K,1979b).

2. Maran : Maran literally means killing. As the name suggests in maran process, a change is brought about in the chemical form or state of the metal. This makes it to lose its metallic characteristics and physical nature. In short, after maran, metal can be converted into powder or other form suitable for administration. To convert various metals into a form appropriate for human consumption, several techniques have been employed which ultimately gave birth to concept: “Bhasma prepared by using Rasa i.e. mercury is the best, whereas the one prepared using herbs are of better quality and those prepared using Gandhak (sulfur) are of inferior quality. Thus there are 3 methods given for maran. It is carried out by heating the metal in presence of 1) mercury 2) plants and 3 ) sulfur.

When various maran procedures for different metals were reviewed, it was found that mercury is mainly used. The unique property of mercury to amalgamate with many metals must have been the reason behind its maximum use in the process of Bhasmikaran. Ancient practitioners might have found it as the most suitable chemical and therefore probably have mentioned that bhasmas using mercury are superior. Plants used in maran process may be serving as catalyst in the process or the minerals in the plants may be forming complexes with the metals. However, no such explanation can be obtained for the use of sulfur.

3. Chalan: Process of stirring during heating the metal is chalan. Stirring is carried out either with iron rod or stick made from a specific plant. As we know today, iron serves as catalyst in many chemical reactions. The phytoconstituents of plant stick may be enhancing the therapeutic effect. For example, stick of Neem is used for chalan process of Jasad bhasma, which is used topically for ophthalmic diseases. We can interpret the significance of this process now. Neem is an antiseptic (Puranik and Dhamankar, 1964h). Zinc is antiseptic, astringent and has ulcer healing property (Block et al., 1982b). These effects of both the constituents may impart the final product better therapeutic activity.

4. Dhavan: In this process, several water washes are given to the product obtained in the previous stage. Perhaps this is to remove the excess amounts of agents used in shodhan or maran stage. Such agents may adversely affect the quality of final product. Hence intermediates are washed with water, thereby water soluble constituents are removed (Puranik and Dhamankar, 1964h).

5. Galan: The product is then sifted either through a fine cloth or through sieves of suitable mesh so as to separate residual material larger in size (Puranik and Dhamankar, 1964h).

6. Puttan: The term puttan means ignition. The general term used for heating in the process of Bhasmikaran is Puta. A special earthen pot, Sharav is generally used for the process. It has two parts, each having a shape of soccer. Sharav is used for direct heating of the material. Its shallowness is useful in heating the material faster and uniformly. After keeping the material on the shallow surface, other part is used as a lid, by placing it in an inverted position. This Puttan process can be looked upon as the key step in manufacturing of bhasma. The classification of putta is primarily done on the basic nature of the process and is as under :- (Puranik and Dhamankar, 1964f) 1)Chandraputta 2) Dhanyarashiputta 3) Suryaputta 4)Bhugarbhaputta 5) Agniptuta.

Toxicity

Modern medical science finds that mercury is inherently toxic, and that its toxicity is not due to the presence of impurities. While mercury does have anti-microbial properties, and formerly waswidely used in Western medicine, its toxicity does not warrant the risk of using it as a health product in most circumstances.[3][4] The Centers for Disease Control and Prevention have also reported a number of cases of lead and mercury poisoning associated with rasa shastra containing Ayurvedic medicines.[5]

Literal and symbolic meaning of bhasma

The Sanskrit word bhasma literally means ‘disintegration’. Bha implies bhartsanam (to destroy), while sma implies smaranam (to remember). Bhasma is thus a reminder to us of the ephemeral nature of life. Also, if we wish to unite with God (or the ‘supreme self’) and remember him constantly, our ego or ‘little self’ has first to be disintegrated or burnt to ashes. Bhasma is a symbol of this process. It is also called raksha because it protects one from all fears. When applied to the forehead before sleep, it is said to keep away spirits or ghosts, whether external or those which manifest from the depths of the mind in the form of nightmares.

Bhasma symbolises the burning of our false identification with the mortal body, and freedom from the limitations of the painfully illusive cycle of birth and death. It also reminds us of the perishable quality of the body, which will one day be reduced to mere ashes. As it says in the Bible, “Ashes to ashes; soul to soul” – the body will return to dust but the soul will continue its journey until it unites with God. All the saints and sages beseech us to remember the ephemeral nature of our earthly existence. In the Rubayyat of Omar Khyyam the poet tells us to, “ . . . make the most of what we yet may spend, before we too into the dust descend, dust into dust, and under dust to lie.” Here he calls for us to seek the eternal, not the temporal. Ash or dust, on the other hand, can be said to represent permanency (or the soul itself), because the ash, just like imperishable truth, does not itself decay. The realised soul is said to rise from the ashes (of the individual self) as the mythical phoenix. The Sufis say, “To reach the goal we have to be burned with the fire of love, so that nothing remains but ashes, and from the ashes will resurrect the new being. Only then can there be real creation!”

The power of bhasma

Bhasma is also called ‘vibhooti’, because it gives spiritual power. The Sanskrit word, vibhooti means ‘glory’, as it gives glory to one who applies it, protection (raksha) from ill health and negative forces, and attracts the higher forces of nature. Another meaning of vibhooti is ‘healing power’, and it is widely used as a medicinal treatment in both Ayurveda and Chinese and Tibetan medicine, which are all ancient and profound systems for the rejuvenation of life. Gold, silver, copper, pearls, mica and other precious stones and metals have curative properties which can quite safely and most effectively be taken into the body after being reduced to ash using great heat.

In Indian villages you will find tantric healers called ojhas who say certain mantras over the ash, which the sick person then applies to the body or eats. These healers can take some earth in their hands, hold it up to the sun, repeat some mantras, and the earth turns into the most beautifully scented ash for curative purposes. Vibhooti is also the name given to siddhis (perfections or psychic powers), as it acts as a vehicle for them. Patanjali’s Yoga Sutras devotes an entire chapter to yogic siddhis. Vibhooti also means ‘dominion’, and is the subtle power lying behind creation, from which all things manifest. From vibhooti or bhasma, anything can be created by a tantric and aghora, because the potential of creation lies within it, and he has penetrated the law and controlled the elements.

Maha Yogi Shiva, father of tantra, is usually depicted naked in sadhana, his whole body covered in bhasma. The first verse of the Shiva Panchakshara Stotram gives the following description: Naagendrahaaraaya trilochanaaya, bhasmaangaraagaaya maheshwaraaya. Nityaaya shuddhaaya digambaraaya – ‘Salutations to the mighty three-eyed Shiva, eternal and pure, wearing the king of snakes as his garland, naked and besmeared with sacred ash.’ Some other names given to Lord Shiva are Bhasmashayaaya (abode of bhasma) and Bhasmabhootaaya (covered with bhasma). Covering the body with ash is considered to be an auspicious act for discovering one’s Shiva nature. Shiva is said to be responsible for mahapralaya, the dissolution of the universe at the end of each kalpa. At this time he dances his tandava nritya, the dance of destruction.

The great tantric siddha Avadhoota Dattatreya was referred to as Bhasma Nishta – one who loves bhasma. Bhasma is generally applied on the forehead, while many sadhus also apply it on the arms, chest and stomach. Some ascetics, especially nagas (naked ascetics) rub it all over the body. While applying it, many devotees also consume a pinch. Shaivites use only bhasma from cremated bodies, which is believed to be very powerful. Bhasma has the power of fire. Agni, the inner fire, scorches and reduces all impurities in the body. It is said that one who smears ash on the body is purified as if bathed in fire. This is known as ‘the bath of fire’. After smearing the body with ash, one should reflect on and realise the highest truth.

Tripundra

Sannyasins wear three lines of bhasma on the forehead. These three lines (tripundra), with a red dot of kumkum underneath, between the eyebrows, symbolise Shiva-Shakti (the unity of energy and matter that creates the entire seen and unseen universe). The lower line represents tamoguna (the state of inertia and darkness), the middle line represents rajas (activity and dynamism) and the top line represents sattwa (balance and illumination). The red dot or tika represents the power of shakti through sadhana, which can take the sadhaka beyond the three gunas or qualities to the state of turiya, the fourth dimension of existence. This is the state of trigunatita – beyond the three gunas.

Swami Niranjanananda says, “The three stripes represent the tradition of the paramahamsas. Jignasus are one stripe sannyasins, representing the drive and motivation to overcome the tamasic tendency. Karma sannyasins are given two stripes, representing their drive to overcome the rajasic along with the tamasic tendencies. Poorna sannyasins are given three stripes, which represent their motivation to transcend the three gunas and attain inner sublimation. The red dot represents the spiritual power or energy that gives us the strength to control the three gunas. It is the awakening of that shakti which is the real aim of sannyasa.”

Bhasma and tattwa shuddhi

Consciousness manifests as energy, which then condenses into matter. In the tantric practice of tattwa shuddhi, in order to experience consciousness free from matter, we reverse the process of evolution back through more and more subtle dimensions to its original cause. Bhasma is an integral part of tattwa shuddhi sadhana, as a symbol of purification on the physical, subtle and causal realms of consciousness. The process of disintegration undergone in tattwa shuddhi is the breaking down of conscious awareness. Just as we reduce matter to its bhasmic form, the ‘fire’ of this practice leads us to the realisation of our essential essence. The stages of pratyahara (sense withdrawal) and dharana (concentration) take us through the more subtle states of consciousness, culminating in samadhi, the ultimate experience or ‘Shiva consciousness’. The journey is from gross matter to pure consciousness.

At the end of the practice of tattwa shuddhi, bhasma is applied to the forehead with the repetition of mantras. It is taken on the middle and ring fingers and wiped slowly on the forehead from left to right, repeating the mantra Om Hraum Namah Shivaya. Sannyasins use the index, middle and ring fingers, and repeat the mantra Om Hamsa. The bhasma used in tattwa shuddhi is prepared from gobar or cow dung. The word gobar literally means ‘gift from the cow’; it is also known as go-maya. The cow is a pure and sacred animal, full of auspicious qualities. It is even said to contain all the devas and devatas within it. Not only does gobar have mystical qualities, but it also contains useful hormones with germicidal properties. The word go also means ‘senses’. So bhasma is also symbolic of the disintegration of the senses which keep us trapped and bound in the gross material world. The transformation of gobar to bhasma is parallel to the transformation from the material world to cosmic consciousness that we find in tattwa shuddhi.

Panchagni bhasma

During the Sat Chandi Mahayajna, and on other auspicious occasions at Rikhia Dham, devotees receive the precious prasadam of panchagni bhasma. This is much prized by sadhakas, because as it has the power of Swami Satyananda’s sadhana behind it, it quickly helps to raise the consciousness at the time of mantra japa and other sadhana when applied to the forehead. Just keeping it in the pooja room is auspicious. This bhasma given is from the Maha Kaal Chita Dhuni, where the previously fierce fires of Sri Swamiji’s panchagni tapasya now lie smouldering quietly under ashes in their shanta roopa or peaceful form. Dhuni is the yogi’s fire, which is the witness or sakshi to his sadhana. It is also where he cooks, takes warmth, and chants the name of God. (Maha means ‘great’, kaal is ‘time’ and chita is ‘consciousness’).

This akhanda dhuni, eternal fire, has been burning in Sri Swamiji’s pooja area ever since he first came to Rikhia in 1989 and devotees come daily for its darshan. It was the centre and support of his life during his austerity, and is the very heart of the Rikhia Ashram (next to Sri Swamiji himself). Although Sri Swamiji no longer goes to this area, the fire is still tended daily. The ashes are moved to the side and the burning embers taken out. Balls of dried cow dung mixed with purifying herbs (vanaspati) are then placed inside along with fresh wood. The embers are then replaced, and the whole area is covered over once more with the ashes. From time to time the ash is removed, carefully sieved through fine cloth, and given as prasadam (that which has been blessed by a divine power.

For the panchagni sadhana itself, Sri Swamiji prepared his own bhasma to protect his body from the great heat, according to the formula prescribed in the Devi Bhagavat Purana. This special bhasma is called mahabhasma and is made from pure cow dung cakes, reeds and ghee. It is treated eleven times with many herbs, honey and other ingredients, being re-burnt each time. Bhasma is smeared on the body only during the first few days of the panchagni sadhana, and is applied in the morning. Sri Swamiji’s dog-cum-companion Bholenath, in whom he manifested the spirit of Bhairava, also took part in the tapasya. “Alsatian dogs can’t bear the heat,” commented Sri Swamiji. “I would put bhasma on him in the morning and he would sit with me.”

Tantric siddhas like Maha Yogi Shiva, Avadhoota Dattatreya and Sri Swamiji are extremely rare beings, and a gift to us beyond our understanding. They belong to a great tradition and leave behind for us a great spiritual legacy. The parampara, the line of avadhootas (those who have become immortal), continues, just as the Mahakaal Chita Dhuni continues to smoulder, unseen beneath the symbol of their glory, their bhasma – the sacred ash.

 

Abhrak Bhasma

Strengthens body, ligaments & Saptadhatu, effective in raktapitta, vat diseases and joint pain. Relieves problems related to chronic hyperacidity like stomache, headache etc.

 

Details

The Indian ayurved philosophy is founded on three basic classifications of human body known as doshasand its well-being:

Vat - related to the central nervous system and gastric tract,

Pitta -digestion and metabolism, governs movement of heat in the body and

Kapha - concerned with structure, stability and fluid balance in the body.

Indian herbal medicines are designed for the specific maintenance of these respective doshas or the aspects of the human body.

Abhrak Bhasma or Sahastraputi is an ancient Indian ayurvedic medicine which is trusted worldwide for healing Vat related issues of the human body. It is also found extremely effective for treating heart related issues.

Abhrak is known to have a high penetrative property which spreads in the body at a faster rate and impacts micro tissues quickly.That is why this herbal medicine is supremely effective in cell regeneration. When used as an alterative, it strengthens ligaments. It is known for rapidly increasing the production of T-cell phagocytes, antibacterial components, which are responsible for a strong immune system.

Kesar Herbals has been in the ayurvedic medicine manufacturing business for about 25 years. This company is one of the foremost and trusted ayurvedic medicine suppliers in India.The concept was let entire mankind benefit from the ancient herbal wealth of India.

Composition:

Purified Mica, Magnesium, Iron, Potassium, Calcium and Aluminium.

Major Benefits of Abhrak Bhasma

  • It finds its application in curing diseases like Hepatitis, Tuberculosis, Asthma and Plague.
  • Abhrak Bhasma is also recommended for breathlessness due to chronic Bronchitis, Asthma, and even Chest Congestion. When taken in the initial stage of TB, it can completely cure it.
  • It is beneficial to all the seven dhatus (energy elements) in the body.
  • It rejuvenates the human mind.
  • It improves blood circulation, improves the general tone of tissues and conductivity.
  • It cures erectile dysfunction and impotency in men.
  • Abhrak is hematinic, which means increases the count of red blood cells. This also increases oxygen carrying ability of the cells.
  • It helps in curing jaundice and anemia.
  • It restores damaged tissues and maintains their health.
  • It is highly effective in cases of bone marrow depletion and hepatic dysfunction.
  • It is effective in curing respiratory tract infections.
  • It is beneficial for curing Bells Palsy and dysentery.
  • It is beneficial in curing anemia, pernicious and azoospermia.
  • Also, helps in controlling many types of veneral diseases like EBV, HIV, Lupus bronchitis and pneumonia.
  • It cures breast cancer.
  • It is also known to cure chronic hyperacidity like stomach-ache and vomiting with blood.

 

Suvarna Bhasma

Boosts immunity, Acts as a nervine tonic , Indicated in old age debility, urinal disorders, anemia, Shwas, Helps to improve complexion, Kas & chronic fever etc.

 

Details

Swarna Bhasma is an ancient Indian ayurvedic medicine that enjoys wide popularity and application.Gold has been accepted as most useful ingredient in Indian ayurvedic medicine system. SwarnaBhasma is used for the effective treatment of Gonorrhoea and Syphilis.

Fortified with potent ayurvedic herbs it is effective for maintaining a healthy life-style and relieves stress.Bhasma denotes the metal based medicine prepared from metals after scientific process to harness the hidden richness of raw metals for therapeutic effects. Swarna Bhasmaor Gold Ash is a therapeutic form of gold metal of nano-sized particles that is one of the most treasured ayurvedic medicines in the world.

It has multi-purpose ayurvedic medicine: it can be used as an antacid, haematinic, and alterative. It is highly effective as a cardiac tonic, maintaining the vitality of the human heart. It is a general vitality booster.

Kesar Herbals firmly believes in the power of nature in healing various ailments. Ayurvedic medicines from India are a gift from nature itself, without any side-effects, and hence they are trusted world-wide.Through scientific processes, nature’s best healing powers from herbs are obtained and used for medicinal purposes, helping million people worldwide. Kesar Herbals is involved in consulting, manufacturing and supplying FDA approved ayurvedic medicines for over three decades.

Major Benefits of Swarna Bhasma

  • Swarna Bhasma improves resistance power in human body.
  • It is responsible for boosting immunity and strengthening the human body.
  • It can be used on a regular basis as an alterative and acts as a nerve tonic.
  • Swarna Bhasma is excellent in treating Anemia, and chronic fever.
  • Due to its gold base, it is helpful in improving complexion.
  • Swarna Bhasma eradicates all chronic disorders, when used consistently.
  • It is indicated in chronic fever, cough, Asthma, urinary disorders, sleeplessness and weak digestion.
  • It strengthens weak muscles, debilities associated with old-age.
  • It is excellent for treating Tuberculosis.
  • It increases sexual power.
  • Swarna Bhasma slows down the process of degeneration of tissues thus helps to prevent premature ageing.
  • It helps prevent symptoms of ageing like wrinkles, dullness of skin, dark circles around the eyes, debility, fatigue, etc.
  • Swarna Bhasma strengthens tissues and ligaments of the human body.
  • It is highly effective in maintaining vigor, vitality and stamina.

Hirak Bhasma

effective against immunity disorders, Bone marrow depression and arthritis, increases stamina, vitality and strength

Ayurvedic medicinal science has been at the forefront of solving most diseases and disorders of the human body because of the purity of its ingredients- this forms the core of the ancient Indian healing system. Ayurvedic medicines and herbal products are effective because of the composition or the formulation of herbs. One such effective ayurvedic medicine is the Hirak Bhasma, which contains diamond ash.

According to Ayurveda, diamond ash is very useful in cancers, immunity disorders, Crippling Rheumatoid arthritis, and even bone marrow depression. Hirak Bhasma is known for toning heart muscles and avoiding cardiovascular syndromes and diseases. It is highly effective as a tonic and alterative, which means, it can be used regularly to strengthen immunity, aid stamina and overall vitality or the functionality of the human body.

Hirak Bhasma is made with diamond- the most precious as well as the hardest gemstone known to mankind. In its healing formulation, diamond is known since ages to make the human body stronger. It is also known for sharpening the human mind by strengthening memory power.

The effectiveness of ayurvedic medicines depends on the company. So be careful in selecting the brand for these medicines and products. Kesar Herbals has been in the ayurvedic consulting space and even manufacturing and supplying of ayurvedic medicines and herbal products for over 25 years. The trust and good will it represents in the field of Ayurveda is affirmed by its customers the world over. It is important to ascertain the brand equity of the company before purchasing these medicines.

Composition:

Hirak Bhasma (diamond ash), Kumari Rasa (Aloe Vera Juice), Gulab Jala (Rose Water), Sunthi Kwath (Dry Ginger Dicoction)

Benefits:

  • Hirak Bhasma is known the world over to make the human body stronger and the mind sharper.
  • It is also recommended for the well-being of the human heart.
  • It helps in toning the muscles of the heart.
  • Its regular consumption as a tonic or an alterative, under medical prescription or supervision is known for toning the heart muscles.
  • Hirak Bhasma also increases stamina, vitality and strength.
  • It is highly effective against immunity disorders.
  • It is known for curing Crippling Rheumatoid
  • It is effective against Bone marrow depression and arthritis.
  • Hirak Bhasma is also known to cure one of the most dangerous and lethal diseases known to mankind: cancer. When used in the initial stages, under medical supervision, it can cure cancerous cells.

Filed under: AYURVEDA Tagged: AYURVEDA, bhasma

What Does 100% of Your Daily Value of Cholesterol Look Like?

$
0
0

Healthline just published an interesting infograph that gives a visualization of what your daily value of cholesterol looks like.  In the graphic, you can see what 300 mg of cholesterol looks like for 20 high cholesterol foods: http://www.healthline.com/health/high-cholesterol/daily-value

This is a very informative resource as it helps us visualize what their cholesterol intake look like

What Does 100% of Your Daily Value of Cholesterol Look Like?

It’s no secret that eating fatty foods raises your bad cholesterol level, also known as LDL. An elevated LDL clogs up your arteries and makes it difficult for your heart to do its job. Potentially, it could lead to heart disease.

The USDA recommends consuming no more than 300 mg of cholesterol a day. While a deep-fried Twinkie at the county fair is an obvious no-no, other high cholesterol culprits may be sneaking into your diet. Check out what that number looks like in terms of everyday food items.

Warning: you may need to revise your grocery list—and your eating habits!

Image

Fried Chicken:

4 pieces=300mg cholesterol

Image

Croissants:

6 2/3 rolls=300mg cholesterol

Image

Cheddar Cheese:

12 3/4 slices=300mg cholesterol

Image

Prosciutto:

28 slices=300mg cholesterol

Image

Corned Beef:

14 thin slices=300mg cholesterol

Image

Butter:

1 1/5 sticks=300mg cholesterol

read at

http://www.healthline.com/health/high-cholesterol/daily-value


Filed under: Uncategorized Tagged: cholesterol
Viewing all 2025 articles
Browse latest View live