New Drug Approvals

Home » 2017 » March

Monthly Archives: March 2017

DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO .....FOR BLOG HOME CLICK HERE

Blog Stats

  • 4,479,802 hits

Flag and hits

Flag Counter

Enter your email address to follow this blog and receive notifications of new posts by email.

Join 2,841 other subscribers
Follow New Drug Approvals on WordPress.com

Archives

Categories

Recent Posts

Flag Counter

ORGANIC SPECTROSCOPY

Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL 

Enter your email address to follow this blog and receive notifications of new posts by email.

Join 2,841 other subscribers
DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO, Born in Mumbai in 1964 and graduated from Mumbai University, Completed his Ph.D from ICT, 1991,Matunga, Mumbai, India, in Organic Chemistry, The thesis topic was Synthesis of Novel Pyrethroid Analogues, Currently he is working with AFRICURE PHARMA, ROW2TECH, NIPER-G, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Govt. of India as ADVISOR, earlier assignment was with GLENMARK LIFE SCIENCES LTD, as CONSUlTANT, Retired from GLENMARK in Jan2022 Research Centre as Principal Scientist, Process Research (bulk actives) at Mahape, Navi Mumbai, India. Total Industry exp 32 plus yrs, Prior to joining Glenmark, he has worked with major multinationals like Hoechst Marion Roussel, now Sanofi, Searle India Ltd, now RPG lifesciences, etc. He has worked with notable scientists like Dr K Nagarajan, Dr Ralph Stapel, Prof S Seshadri, etc, He did custom synthesis for major multinationals in his career like BASF, Novartis, Sanofi, etc., He has worked in Discovery, Natural products, Bulk drugs, Generics, Intermediates, Fine chemicals, Neutraceuticals, GMP, Scaleups, etc, he is now helping millions, has 9 million plus hits on Google on all Organic chemistry websites. His friends call him Open superstar worlddrugtracker. His New Drug Approvals, Green Chemistry International, All about drugs, Eurekamoments, Organic spectroscopy international, etc in organic chemistry are some most read blogs He has hands on experience in initiation and developing novel routes for drug molecules and implementation them on commercial scale over a 32 PLUS year tenure till date Feb 2023, Around 35 plus products in his career. He has good knowledge of IPM, GMP, Regulatory aspects, he has several International patents published worldwide . He has good proficiency in Technology transfer, Spectroscopy, Stereochemistry, Synthesis, Polymorphism etc., He suffered a paralytic stroke/ Acute Transverse mylitis in Dec 2007 and is 90 %Paralysed, He is bound to a wheelchair, this seems to have injected feul in him to help chemists all around the world, he is more active than before and is pushing boundaries, He has 100 million plus hits on Google, 2.5 lakh plus connections on all networking sites, 100 Lakh plus views on dozen plus blogs, 227 countries, 7 continents, He makes himself available to all, contact him on +91 9323115463, email amcrasto@gmail.com, Twitter, @amcrasto , He lives and will die for his family, 90% paralysis cannot kill his soul., Notably he has 38 lakh plus views on New Drug Approvals Blog in 227 countries......https://newdrugapprovals.wordpress.com/ , He appreciates the help he gets from one and all, Friends, Family, Glenmark, Readers, Wellwishers, Doctors, Drug authorities, His Contacts, Physiotherapist, etc He has total of 32 International and Indian awards

Personal Links

Verified Services

View Full Profile →

Archives

Categories

Flag Counter

FDA approves new drug to treat multiple sclerosis Ocrevus (ocrelizumab)


03/29/2017
On March 28, the U.S. Food and Drug Administration approved Ocrevus (ocrelizumab) to treat adult patients with relapsing forms of multiple sclerosis (MS) and primary progressive multiple sclerosis (PPMS). This is the first drug approved by the FDA for PPMS. Ocrevus is an intravenous infusion given by a health care professional.

On March 28, the U.S. Food and Drug Administration approved Ocrevus (ocrelizumab) to treat adult patients with relapsing forms of multiple sclerosis (MS) and primary progressive multiple sclerosis (PPMS). This is the first drug approved by the FDA for PPMS. Ocrevus is an intravenous infusion given by a health care professional.

“Multiple sclerosis can have a profound impact on a person’s life,” said Billy Dunn, M.D., director of the Division of Neurology Products in the FDA’s Center for Drug Evaluation and Research. “This therapy not only provides another treatment option for those with relapsing MS, but for the first time provides an approved therapy for those with primary progressive MS.”

MS is a chronic, inflammatory, autoimmune disease of the central nervous system that disrupts communication between the brain and other parts of the body. It is among the most common causes of neurological disability in young adults and occurs more frequently in women than men. For most people with MS, episodes of worsening function (relapses) are initially followed by recovery periods (remissions). Over time, recovery may be incomplete, leading to progressive decline in function and increased disability. Most people experience their first symptoms of MS between the ages of 20 and 40.

PPMS is characterized by steadily worsening function from the onset of symptoms, often without early relapses or remissions. The U.S. Centers for Disease Control and Prevention estimates that approximately 15 percent of patients with MS have PPMS.

The efficacy of Ocrevus for the treatment of relapsing forms of MS was shown in two clinical trials in 1,656 participants treated for 96 weeks. Both studies compared Ocrevus to another MS drug, Rebif (interferon beta-1a). In both studies, the patients receiving Ocrevus had reduced relapse rates and reduced worsening of disability compared to Rebif.

In a study of PPMS in 732 participants treated for at least 120 weeks, those receiving Ocrevus showed a longer time to the worsening of disability compared to placebo.

Ocrevus should not be used in patients with hepatitis B infection or a history of life-threatening infusion-related reactions to Ocrevus. Ocrevus must be dispensed with a patient Medication Guide that describes important information about the drug’s uses and risks. Ocrevus can cause infusion-related reactions, which can be serious. These reactions include, but are not limited to, itchy skin, rash, hives, skin redness, flushing, low blood pressure, fever, tiredness, dizziness, headache, throat irritation, shortness of breath, swelling of the throat, nausea, and fast heartbeat. Additionally, Ocrevus may increase the risk for malignancies, particularly breast cancer. Delay Ocrevus treatment for patients with active infections. Vaccination with live or live attenuated vaccines is not recommended in patients receiving Ocrevus.

In addition to the infusion-related reactions, the most common side effect of Ocrevus seen in the clinical trials for relapsing forms of MS was upper respiratory tract infection. The most common side effects in the study of PPMS were upper respiratory tract infection, skin infection, and lower respiratory tract infection.

The FDA granted this application breakthrough therapy designation, fast track designation, and priority review.

The FDA granted approval of Ocrevus to Genentech, Inc.

//////multiple sclerosis, Ocrevus, ocrelizumab, fda 2017, genentech,

FDA approves new eczema drug Dupixent (dupilumab)


03/28/2017 11:14
The U.S. Food and Drug Administration today approved Dupixent (dupilumab) injection to treat adults with moderate-to-severe eczema (atopic dermatitis). Dupixent is intended for patients whose eczema is not controlled adequately by topical therapies, or those for whom topical therapies are not advisable. Dupixent can be used with or without topical corticosteroids.

The U.S. Food and Drug Administration today approved Dupixent (dupilumab) injection to treat adults with moderate-to-severe eczema (atopic dermatitis). Dupixent is intended for patients whose eczema is not controlled adequately by topical therapies, or those for whom topical therapies are not advisable. Dupixent can be used with or without topical corticosteroids.

“FDA’s approval of Dupixent demonstrates our commitment to approving new and innovative therapies for patients with skin disease,” said Julie Beitz, M.D., director of the Office of Drug Evaluation III in the FDA’s Center for Drug Evaluation and Research. “Eczema can cause significant skin irritation and discomfort for patients, so it is important to have a variety of treatment options available to patients, including those patients whose disease is not controlled by topical therapies.”

Atopic dermatitis, a chronic inflammatory skin disease, is often referred to as “eczema,” which is a general term for several types of inflammation of the skin. Atopic dermatitis is the most common of the many types of eczema; onset typically begins in childhood and can last through adulthood. The cause of atopic dermatitis is a combination of genetic, immune and environmental factors. In atopic dermatitis, the skin develops red, scaly and crusted bumps, which are extremely itchy. Scratching leads to swelling, cracking, “weeping” clear fluid, and finally, coarsening and thickening of the skin.

Dupixent is administered as an injection under the skin. Dupixent’s active ingredient is an antibody (dupilumab) that binds to a protein [interleukin-4 (IL-4) receptor alpha subunit (IL-4Ra)], that causes inflammation. By binding to this protein, Dupixent is able to inhibit the inflammatory response that plays a role in the development of atopic dermatitis.

The safety and efficacy of Dupixent were established in three placebo-controlled clinical trials with a total of 2,119 adult participants with moderate-to-severe atopic dermatitis not adequately controlled by topical medication(s). Overall, participants who received Dupixent achieved greater response, defined as clear or almost clear skin, and experienced a reduction in itch after 16 weeks of treatment.

Dupixent can cause side effects such as serious allergic reactions and eye problems, such as pink eye (conjunctivitis) and inflammation of the cornea (keratitis). If patients experience new or worsening eye symptoms such as redness, itching, pain or visual changes, they should consult a health care provider. The most common side effects include injection site reactions; cold sores in the mouth or on the lips; and eye and eyelid inflammation, including redness, swelling and itching.

The safety and efficacy of Dupixent have not been established in the treatment of asthma. Patients who also have asthma should not adjust or stop their asthma treatment without talking to their physicians.

The FDA granted the application for Dupixent Priority Review and Breakthrough Therapy designation.

The FDA granted the approval of Dupixent to Regeneron Pharmaceuticals, Inc.

TRIENTINE HYDROCHLORIDE, 塩酸トリエンチン , 曲恩汀


Skeletal formula of triethylenetetramine

TRIENTINE

  • Molecular Formula C6H18N4
  • Average mass 146.234 Da

112-24-3 CAS

曲恩汀, KD-034, MK-0681, MK-681, TECZA, TETA, TJA-250

1,2-Ethanediamine, N1,N2-bis(2-aminoethyl)-
1,8-diamino-3,6-diazaoctane
Image result for TRIENTINE

TRIENTINE HYDROCHLORIDE

  • Molecular Formula C6H19ClN4
  • Average mass 182.695 Da

38260-01-4 CAS

Launched – 1986 VALEANT, WILSONS DISEASE

Image result for MSD

Image result for VALEANT

塩酸トリエンチン
Trientine Hydrochloride

C6H18N4▪2HCl : 219.16
[38260-01-4]

UPDATE CDSCO INDIA Trientine 08.06.2021 APPROVED

Trientine Tetrahydrochloride bulk and
Trientine Tetrahydrochloride capsules 333 mg
(Each capsule contains Trientine
tetrahydrochloride 333mg equivalent to
Trientine 167mg base)

For the treatment of Wilson’s disease
(hepatolenticular degeneration) in patients
intolerant to Penicillamine. It should be
used when continued treatment with
Penicillamine is no longer possible because
of intolerable or life endangering side
effects.

Aton Pharma, a subsidiary of Valeant Pharmaceuticals, has developed and launched Syprine, a capsule formulation of trientine hydrochloride, for treating Wilson disease.

Image result for TRIENTINE

Triethylenetetramine, abbreviated TETA and trien and also called trientine (INN), is an organic compound with the formula [CH2NHCH2CH2NH2]2. This oily liquid is colorless but, like many amines, assumes a yellowish color due to impurities resulting from air-oxidation. It is soluble in polar solvents. The branched isomer tris(2-aminoethyl)amine and piperazine derivatives may also be present in commercial samples of TETA.[1]

Trientine hydrochloride is a metal antagonist that was first launched by Merck, Sharp & Dohme in the U.S. in 1986 under the brand name Syprine for the oral treatment of Wilson’s disease.

Orphan drug designation has also been assigned in the U.S. for the treatment of patients with Wilson’s disease who are intolerant or inadequately responsive to penicillamine and in the E.U. by Univar for the treatment of Wilson’s disease

 Trientine hydrochloride pk_prod_list.xml_prod_list_card_pr?p_tsearch=A&p_id=90373

By condensation of ethylenediamine (I) with 1,2-dichloroethane (II)

Trientine hydrochloride is N,N’-bis (2-aminoethyl)-1,2-ethanediamine dihydrochloride. It is a white to pale yellow crystalline hygroscopic powder. It is freely soluble in water, soluble in methanol, slightly soluble in ethanol, and insoluble in chloroform and ether.

The empirical formula is C6H18N4·2HCI with a molecular weight of 219.2. The structural formula is:

NH2(CH2)2NH(CH2)2NH(CH2)2NH2•2HCI

Trientine hydrochloride is a chelating compound for removal of excess copper from the body. SYPRINE (Trientine Hydrochloride) is available as 250 mg capsules for oral administration. Capsules SYPRINE contain gelatin, iron oxides, stearic acid, and titanium dioxide as inactive ingredients.

Image result for TRIENTINE

Production

TETA is prepared by heating ethylenediamine or ethanolamine/ammonia mixtures over an oxide catalyst. This process gives a variety of amines, which are separated by distillation and sublimation.[2]

Uses

The reactivity and uses of TETA are similar to those for the related polyamines ethylenediamine and diethylenetriamine. It was primarily used as a crosslinker (“hardener”) in epoxy curing.[2]

The hydrochloride salt of TETA, referred to as trientine hydrochloride, is a chelating agent that is used to bind and remove copper in the body to treat Wilson’s disease, particularly in those who are intolerant to penicillamine. Some recommend trientine as first-line treatment, but experience with penicillamine is more extensive.[3]

Coordination chemistry

TETA is a tetradentate ligand in coordination chemistry, where it is referred to as trien.[4] Octahedral complexes of the type M(trien)Cl3 can adopt several diastereomeric structures, most of which are chiral.[5]

Trientine, chemically known as triethylenetetramine or N,N’-bis(2-aminoethyl)-l,2-ethanediamine belongs to the class of polyethylene polyamines. Trientine dihydrochloride is a chelating agent which is used to bind and remove copper in the body in the treatment of Wilson’s disease.

Image result for TRIENTINE

Trientine dihydrochloride (1)

Trientine dihydrochloride formulation, developed by Aton with the proprietary name SYPRINE, was approved by USFDA on November 8, 1985 for the treatment of patients with Wilson’s disease, who are intolerant to penicillamine. Trientine dihydrochloride, due to its activity on copper homeostasis, is being studied for various potential applications in the treatment of internal organs damage in diabetics, Alzheimer’s disease and cancer.

Various synthetic methods for preparation of triethylenetetramine (TETA) and the corresponding dihydrochloride salt have been disclosed in the prior art.

U.S. 4,806,517 discloses the synthesis of triethylenetetramine from ethylenediamine and monoethanolamine using Titania supported phosphorous catalyst while U.S. 4,550,209 and U.S. 5,225,599 disclose catalytic condensation of ethylenediamine and ethylene glycol for the synthesis of linear triethylenetetramine using catalysts like zirconium trimethylene diphosphonate, or metatungstate composites of titanium dioxide and zirconium dioxide.

U.S. 4,503,253 discloses the preparation of triethylenetetramine by reaction of an alkanolamine compound with ammonia and an alkyleneamine having two primary amino groups in the presence of a catalyst, such as supported phosphoric acid wherein the support is comprised of silica, alumina or carbon.

The methods described above for preparation of triethylenetetramine require high temperatures and pressure. Further, due to the various possible side reactions and consequent associated impurities, it is difficult to control the purity of the desired amine.

CN 102924289 discloses a process for trientine dihydrochloride comprising reduction of Ν,Ν’-dibenzyl-,N,N’-bis[2-(l,3-dioxo-2H-isoindolyl)ethyl]ethanediamine using hydrazine hydrate to give N,N’-dibenzyl-,N,N’-bis(2-aminoethyl)ethanediamine, which, upon condensation with benzyl chloroformate gave N,N’-dibenzyl-,N,N’-bis[2-(Cbz-amino)ethyl]ethanediamine, and further reductive deprotection to give the desired compound.

CS 197,093 discloses a process comprising reaction of triethylenetetramine with concentrated hydrochloric acid to obtain the crystalline tetrahydrochlonde salt. Further reaction of the salt with sodium ethoxide in solvent ethanol, filtration of the solid sodium chloride which is generated in the process, followed by slow cooling and crystallization of the filtrate provided the dihydrochloride salt. Optionally, aqueous solution of the tetrahydrochloride salt was passed through a column of an anion exchanger and the eluate containing free base was treated with a calculated amount of the tetrahydrochloride, evaporated, and the residue was crystallized from aqueous ethanol to yield the dihydrochloride salt.

The process is quite circuitous and cumbersome, requiring use of strong bases, filtration of sodium chloride and results in yields as low as 60%.

US 8,394,992 discloses a method for preparation of triethylenetetramine dihydrochloride wherein tertiary butoxycarbonyl (boc) protected triethylenetetramine is first converted to its tetrahydrochloride salt using large excess of hydrochloric acid in solvent isopropanol, followed by treatment of the resulting tetrahydrochloride salt with a strong base like sodium alkoxide to produce the amine free base (TETA) and sodium chloride salt in anhydrous conditions. The free amine is extracted with tertiary butyl methyl ether (TBME), followed by removal of sodium chloride salt and finally the amine free base TETA is treated with hydrochloric acid in solvent ethanol to give trientine hydrochloride salt.

PATENT

WO-2017046695

str1

EXAMPLES

Example 1: Preparation of 2-([2-[cyanomethyl]-t-butyloxycarbonylamino]ethyl- 1-butyloxy carbonylamino)acetonitrile (5)

Potassium carbonate (481.9 g) was added to a stirred mixture of ethylenediamine (100.0 g) in acetonitrile (800 ml) and cooled to around 10°C. Chloroacetonitrile (263.8 g) was gradually added at same temperature and stirred at 25-30°C, till completion of the reaction, as monitored by HPLC. The mixture was cooled to 5-15°C and Boc-anhydride (762. lg) was added to it, followed by stirring at the same temperature. The temperature was raised to 25-30°C and the mass was stirred till completion of the reaction, as monitored by HPLC.

The reaction mass was filtered and the filtrate was concentrated. Toluene was added to the residue, and the mixture was heated to around 70°C followed by cooling and filtration to give 2-([2-[cyanomethyl)-t-butyloxycarbonylamino]ethyl-t-butyloxycarbonylamino) acetonitrile (5).

Yield: 506.8 g

% Yield: 89.9 %

Example 2: Preparation of t-butyl( N-2-aminoethyl)N-([2-[(2-aminoethyl)t-butyloxy)carbonylamino] ethyl) carbamate (6)

Raney nickel (120.0 g) in isopropanol (100 ml) was charged into an autoclave, followed by a mixture of Compound 5 (200 g) in isopropanol (400 ml). Cooled ammonia solution prepared by purging ammonia gas in 1400 ml isopropanol, equivalent to 125 g ammonia was gradually charged to the autoclave and the reaction was carried out around 15-25°C under hydrogen pressure of 2-5 Kg/cm2.

After completion of the reaction, as monitored by HPLC, the mass was filtered, concentrated, and methyl tertiary butyl ether was added to the residue. The mixture was heated to around 50°C, followed by cooling of the mass, stirring, optional seeding with compound 6 and filtration to give tertiary butyl-(N-2-aminoethyl)N-([2-[(2-aminoethyl)-(tert-butyloxy) carbonylamino] ethyl) carbamate.

Yield: 174 g

%Yield: 85 %

Example 3: Preparation of triethylenetetramine dihydrochloride (1)

Concentrated hydrochloric acid (121.5 g) was gradually added to a stirred mixture of tertiary-butyl-N-(2-aminoethyl)-N-2-[(2-aminoethyl)-(tert-butoxy) carbonyl] amino] ethyl} carbamate (Compound 6, 200.0 g) and water (1400 ml) at 20-30°C. The reaction mixture was heated in the temperature range of 100-105°C till completion of the reaction, as monitored by HPLC, with optionally distilling out water, if so required.

The reaction mass was concentrated and ethanol (600 ml) was added to the residue, followed by heating till a clear solution was obtained. The reaction mixture was gradually cooled with stirring, filtered and dried to provide triethylenetetramine dihydrochloride (1).

Yield: 88.9 g, (70 %)

Purity : > 99%

Patent

https://www.google.com/patents/US8394992

Trientine was said to be used in the synthesis of benzylidene-(2-{3-[2-(benzylidene-amino)-ethyl]-2-phenyl-imidazolidin-1-yl}-ethyl)-amine in French Patent No. FR2810035 to Guilard et al. Cetinkaya, E., et al., “Synthesis and characterization of unusual tetraminoalkenes,” J. Chem. Soc. 5:561-7 (1992), is said to be directed to synthesis of benzylidene-(2-{3-[2-(benzylidene-amino)-ethyl]-2-phenyl-imidazolidin-1-yl}-ethyl)-amine from trientine, as is Araki T., et al., “Site-selective derivatization of oligoethyleneimines using five-membered-ring protection method,” Macromol., 21:1995-2001 (1988). Triethylenetetramine may reportedly also be used in the synthesis of N-methylated triethylenetetramine, as reported in U.S. Pat. No. 2,390,766, to Zellhoefer et al.

Synthesis of polyethylenepolyamines, including triethylenetetramines, from ethylenediamine and monoethanolamine using pelleted group IVb metal oxide-phosphate type catalysts was reported by Vanderpool et al. in U.S. Pat. No. 4,806,517. Synthesis of triethylenetetramine from ethylenediamine and ethanolamine was also proposed in U.S. Pat. No. 4,550,209, to Unvert et al. U.S. Pat. No. 5,225,599, to King et al. is said to be directed to the synthesis of linear triethylene tetramine by condensation of ethylenediamine and ethylene glycol in the presence of a catalyst. Joint production of triethylenetetramine and 1-(2-aminoethyl)-aminoethyl-piperazine was proposed by Borisenko et al. in U.S.S.R. Patent No. SU1541204. U.S. Pat. No. 4,766,247 and European Patent No. EP262562, both to Ford et al., reported the preparation of triethylenetetramine by reaction of an alkanolamine compound, an alkaline amine and optionally either a primary or secondary amine in the presence of a phosphorous containing catalyst, for example phosphoric acid on silica-alumina or Group IIIB metal acid phosphate, at a temperature from about 175° C. to 400° C. under pressure. These patents indicate that the synthetic method used therein was as set forth in U.S. Pat. No. 4,463,193, to Johnson. The Ford et al. ‘247 patent is also said to be directed to color reduction of polyamines by reaction at elevated temperature and pressure in the presence of a hydrogenation catalyst and a hydrogen atmosphere. European Patent No. EP450709 to King et al. is said to be directed to a process for the preparation of triethylenetetramine and N-(2-aminoethyl)ethanolamine by condensation of an alkylenamine and an alkylene glycol in the presence of a condensation catalyst and a catalyst promoter at a temperature in excess of 260° C.

Russian Patent No. RU2186761, to Zagidullin, proposed synthesis of diethylenetriamine by reaction of dichloroethane with ethylenediamine. Ethylenediamine has previously been said to have been used in the synthesis of N-carboxylic acid esters as reported in U.S. Pat. No. 1,527,868, to Hartmann et al.

Japanese Patent No. 06065161 to Hara et al. is said to be directed to the synthesis of polyethylenepolyamines by reacting ethylenediamine with ethanolamine in the presence of silica-treated Nb205 supported on a carrier. Japanese Patent No. JP03047154 to Watanabe et al., is said to be directed to production of noncyclic polyethylenepolyamines by reaction of ammonia with monoethanolamine and ethylenediamine. Production of non-cyclic polyethylenepolyamines by reaction of ethylenediamine and monoethanolamine in the presence of hydrogen or a phosphorous-containing substance was said to be reported in Japanese Patent No. JP03048644. Regenerative preparation of linear polyethylenepolyamines using a phosphorous-bonded catalyst was proposed in European Patent No. EP115,138, to Larkin et al.

A process for preparation of alkyleneamines in the presence of a niobium catalyst was said to be provided in European Patent No. 256,516, to Tsutsumi et al. U.S. Pat. No. 4,584,405, to Vanderpool, reported the continuous synthesis of essentially noncyclic polyethylenepolyamines by reaction of monoethanolamine with ethylenediamine in the presence of an activated carbon catalyst under a pressure between about 500 to about 3000 psig., and at a temperature of between about 200° C. to about 400° C. Templeton, et al., reported on the preparation of linear polyethylenepolyamides asserted to result from reactions employing silica-alumina catalysts in European Patent No. EP150,558.

Production of triethylenetetramine dihydrochloride was said to have been reported in Kuhr et al., Czech Patent No. 197,093, via conversion of triethylenetetramine to crystalline tetrahydrochloride and subsequently to triethylenetetramine dihydrochloride. “A study of efficient preparation of triethylenetetramine dihydrochloride for the treatment of Wilson’s disease and hygroscopicity of its capsule,” Fujito, et al., Yakuzaigaku, 50:402-8 (1990), is also said to be directed to production of triethylenetetramine.

Preparation of triethylenetetramine salts used for the treatment of Wilson’s disease was said to be reported in “Treatment of Wilson’s Disease with Triethylene Tetramine Hydrochloride (Trientine),” Dubois, et al., J. Pediatric Gastro. & Nutrition, 10:77-81 (1990); “Preparation of Triethylenetetramine Dihydrochloride for the Treatment of Wilson’s Disease,” Dixon, et al., Lancet, 1(1775):853 (1972); “Determination of Triethylenetetramine in Plasma of Patients by High-Performance Liquid Chromatography,” Miyazaki, et al., Chem. Pharm. Bull., 38(4):1035-1038 (1990); “Preparation of and Clinical Experiences with Trien for the Treatment of Wilson’s Disease in Absolute Intolerance of D-penicillamine,” Harders, et al., Proc. Roy. Soc. Med., 70:10-12 (1977); “Tetramine cupruretic agents: A comparison in dogs,” Allen, et al., Am. J. Vet. Res., 48(1):28-30 (1987); and “Potentiometric and Spectroscopic Study of the Equilibria in the Aqueous Copper(II)-3,6-Diazaoctane-1,8-diamine System,” Laurie, et al., J.C.S. Dalton, 1882 (1976).

Preparation of Triethylenetetramine Salts by Reaction of Alcohol Solutions of Amines and acids was said to be reported in Polish Patent No. 105793, to Witek. Preparation of triethylenetetramine salts was also asserted in “Polycondensation of polyethylene polyamines with aliphatic dicarboxylic acids,” Witek, et al., Polimery, 20(3):118-119 (1975).

Baganz, H., and Peissker, H., Chem. Ber., 1957; 90:2944-2949; Haydock, D. B., and Mulholland, T. P. C., J. Chem. Soc., 1971; 2389-2395; and Rehse, K., et al., Arch. Pharm., 1994; 393-398, report on Strecker syntheses. Use of Boc and other protecting groups has been described. See, for example, Spicer, J. A. et al., Bioorganic & Medicinal Chemistry, 2002; 10: 19-29; Klenke, B. and Gilbert, I. H., J. Org. Chem., 2001; 66: 2480-2483.

FIG. 6 shows an 1H-NMR spectrum of a triethylenetetramine hydrochloride salt in D2O, as synthesized in Example 3. NMR values include a frequency of 400.13 Mhz, a 1H nucleus, number of transients is 16, points count of 32768, pulse sequence of zg30, and sweep width of 8278.15 H

Image result for TRIENTINE

CLIP

Click to access JP17e_1.pdf

Method of purification: Dissolve Trientine Hydrochloride in water while warming, and recrystallize by addition of ethanol (99.5). Or dissolve Trientine Hydrochloride in water while warming, allow to stand after addition of activated charcoal in a cool and dark place for one night, and filter. To the filtrate add ethanol (99.5), allow to stand in a cool and dark place, and recrystallize. Dry the crystals under reduced pressure not exceeding 0.67 kPa at 409C until ethanol odor disappears.

References

  1.  “Ethyleneamines” (PDF). Huntsman. 2007.
  2. ^ Jump up to:a b Eller, K.; Henkes, E.; Rossbacher, R.; Höke, H. (2005). “Amines, Aliphatic”. Ullmann’s Encyclopedia of Industrial Chemistry. Weinheim: Wiley-VCH. doi:10.1002/14356007.a02_001.
  3. Jump up^ Roberts, E. A.; Schilsky, M. L. (2003). “A practice guideline on Wilson disease” (pdf). Hepatology. 37 (6): 1475–1492. doi:10.1053/jhep.2003.50252. PMID 12774027.
  4. Jump up^ von Zelewsky, A. (1995). Stereochemistry of Coordination Compounds. Chichester: John Wiley. ISBN 047195599X.
  5.  Utsuno, S.; Sakai, Y.; Yoshikawa, Y.; Yamatera, H. (1985). “Three Isomers of the Trans-Diammine-[N,N′-bis(2-Aminoethyl)-1,2-Ethanediamine]-Cobalt(III) Complex Cation”. Inorganic Syntheses. 23: 79–82. doi:10.1002/9780470132548.ch16.
Triethylenetetramine
Skeletal formula of triethylenetetramine
Ball and stick model of triethylenetetramine
Spacefill model of triethylenetetramine
Names
Other names

N,N’-Bis(2-aminoethyl)ethane-1,2-diamine; TETA; Trien; Trientine (INN); Syprine (brand name)
Identifiers
3D model (Jmol)
605448
ChEBI
ChemSpider
ECHA InfoCard 100.003.591
EC Number 203-950-6
27008
KEGG
MeSH Trientine
RTECS number YE6650000
UNII
UN number 2259
Properties
C6H18N4
Molar mass 146.24 g·mol−1
Appearance Colorless liquid
Odor Fishy, ammoniacal
Density 982 mg mL−1
Melting point −34.6 °C; −30.4 °F; 238.5 K
Boiling point 266.6 °C; 511.8 °F; 539.7 K
Miscible
log P 1.985
Vapor pressure <1 Pa (at 20 °C)
1.496
Thermochemistry
376 J K−1 mol−1 (at 60 °C)
Pharmacology
A16AX12 (WHO)
Hazards
GHS pictograms The corrosion pictogram in the Globally Harmonized System of Classification and Labelling of Chemicals (GHS) The exclamation-mark pictogram in the Globally Harmonized System of Classification and Labelling of Chemicals (GHS)
GHS signal word DANGER
H312, H314, H317, H412
P273, P280, P305+351+338, P310
Corrosive C
R-phrases R21, R34, R43, R52/53
S-phrases (S1/2), S26, S36/37/39, S45
Flash point 129 °C (264 °F; 402 K)
Lethal dose or concentration (LD, LC):
  • 550 mg kg−1 (dermal, rabbit)
  • 2.5 g kg−1 (oral, rat)
Related compounds
Related amines
Related compounds
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).

///////////////TRIENTINE, 112-24-3, 曲恩汀 , KD-034 , MK-0681, MK-681, TECZA, TETA, TJA-250, Orphan drug

NCCNCCNCCN

SEN 826


figure

SEN 826
CAS 1160833-51-1
C25 H31 N5 O, 417.55
Methanone, [1-[3-(1-methyl-1H-benzimidazol-2-yl)phenyl]-4-piperidinyl](4-methyl-1-piperazinyl)-
CAS HBr SALT 1612250-71-1

WO2009074300 product patent

Russell John Thomas, Mohr Gal.La Pericot, Giacomo Minetto, Annette Cornelia Bekker, Pietro Ferruzzi
Applicant Siena Biotech S.P.A.
Image result for Siena Biotech S.P.A.
Siena Biotech S.p.A. operates as a drug discovery and development company which develops a portfolio of disease modifying small molecule therapeutics for oncology and neurodegenerative diseases. Its products include blood-brain barrier penetrant compounds, which are in pipeline, for the treatment of brain cancers and peripheral tumors capable of metastasizing to the brain; clinical candidates for Alzheimer’s disease; and SEN196, a Sirtuin 1 inhibitor against Huntington disease. The company also provides contract research services, drug discovery, integrated chemistry, in-vitro technologies, and preclinical technologies. Siena Biotech S.p.A. has a strategic partnership with Aptuit Inc. The company was founded in 2000 and is based in Siena, Italy. Siena Biotech S.p.A operates as a subsidiary of THERAMetrics holding AG
Russell Thomas

Russell Thomas

https://www.linkedin.com/in/russell-thomas-0317464/

PLEASE MAIL ME AT amcrasto@gmail.com if picture is a mistake or cal +919323115463

The SMO receptor mediates Hedgehog (Hh) signaling critical to development, differentiation, growth, and cell migration. In normal conditions, activation of the pathway is induced by binding of specific endogenous ligands (i.e., Sonic Hh) to its receptor Patched (Ptch), which in turns reverts the Ptch inhibitory effect on SMO. SMO activation ultimately determines specific target genes activation through a family of three transcription factors, Gli1, Gli2 and Gli3.
Although Hh signaling is significantly curtailed in adults, it retains functional roles in stem cell maintenance, and aberrant Hh signaling has been described in a range of tumours.
Mutational inactivation of the inhibitory pathway components results in a constitutive ligand-independent activation seen in tumours such as basal cell carcinoma (BCC) and medulloblastoma. Ligand-dependent activation is seen in tumours such as prostate cancer, pancreatic cancer, gastrointestinal malignancies, melanoma, gliomas, breast cancer, ovarian cancer, leukemia, and B-cell lymphomas. A significant body of evidence supports the conclusion that SMO receptor antagonism will block the downstream signaling events.
As part of a program to address unmet medical need with regard to tumours in the CNS, Siena Biotech has designed and investigated selective antagonists of the SMO receptor. The newly designed API development candidate SEN826 1  is part of a group of potent antagonists of the Hedgehog pathway.
SYNTHESIS

PATENT

WO 2009074300

Figure imgf000025_0001

Figure imgf000019_0002

Figure

The synthesis starts with the formation of the 2-arylbenzimidazole derivative 6 which can be carried out starting from N-methylphenylenediamine 2 (Method A; blue path in Scheme 1) or employing o-phenylenediamine 4 in the ring closure reaction followed by N-methylation (Method B; orange path in Scheme 1). Sodium hydrogen sulfite is used to promote the condensation of the corresponding o-phenylenediamine with the Br-aromatic aldehyde 3.(6b) The next step is the coupling of the aryl bromide with isonipecotic ethyl ester in Buchwald conditions. After acidic hydrolysis with HCl under microwave irradiation, the final amide 1 was synthesized with CDI as coupling agent.

PAPER

A Scalable Route to the SMO Receptor Antagonist SEN826: Benzimidazole Synthesis via Enhanced in Situ Formation of the Bisulfite–Aldehyde Complex

Process Chemistry Unit, Siena Biotech SpA, 53100 Siena, Italy
Compound Management & Analysis Unit, Siena Biotech SpA, 53100 Siena, Italy
Org. Process Res. Dev., 2014, 18 (6), pp 699–708
Abstract Image

A practical and scalable route to the SMO antagonist SEN826 1 is described herein, including the discussion of an alternative approach to the synthesis of the target molecule. The optimized route consists of five chemical steps. A new and efficient access to the key intermediate 6 via the bisulfite–aldehyde complex was developed, significantly enhancing the yields and reducing costs. As a result, a synthetic procedure for preparation of multihundred gram quantities of the final product has been developed.

1 as hydrobromide salt. Yield: 71%.
UPLC–MS: tR = 1.24 min; m/z = 418 [M + 1]+.
HRMS calcd for C25H33N5O [M + 1]+ 418.26069, found 418.26075.
HPLC: tR = 5.99 min; purity 99.1%.
1H NMR (400 MHz DMSO-d6): δ 9.80 (broad, 1H), 7.89 (m, 1H), 7.77 (m, 1H), 7.55–7.45 (m, 3H), 7.38 (s, 1H), 7.24 (m, 2H), 4.48–4.15 (m, 2H), 3.96 (s, 3H), 3.86 (m, 2H), 3.55–3.15 (m, 3H), 3.10–2.82 (m, 6H), 2.81 (s, 3H), 1.76–1.57 (m, 4H).
13C NMR (100 MHz DMSO-d6): δ 173.5, 152.3, 151.5, 135.1, 135.0, 130.5, 126.2, 125.6, 125.3, 119.9, 119.1, 117.1, 116.5, 113.0, 53.2, 48.2, 42.7, 38.8, 37.4, 33.1, 28.2.
Water content (KF): 3.5 wt %.
Pd content (ICP-MS): 128 ppm.
Bromine content (ionic exchange LC): 20 wt % (1.2 equiv).
str1 str2
/////////////////

FDA approves first treatment Bavencio (avelumab)for rare form of skin cancer


 Image result for avelumab
str1
03/23/2017
The U.S. Food and Drug Administration today granted accelerated approval to Bavencio (avelumab) for the treatment of adults and pediatric patients 12 years and older with metastatic Merkel cell carcinoma (MCC), including those who have not received prior chemotherapy. This is the first FDA-approved treatment for metastatic MCC, a rare, aggressive form of skin cancer.

March 23, 2017

Release

The U.S. Food and Drug Administration today granted accelerated approval to Bavencio (avelumab) for the treatment of adults and pediatric patients 12 years and older with metastatic Merkel cell carcinoma (MCC), including those who have not received prior chemotherapy. This is the first FDA-approved treatment for metastatic MCC, a rare, aggressive form of skin cancer.

“While skin cancer is one of the most common cancers, patients with a rare form called Merkel cell cancer have not had an approved treatment option until now,” said Richard Pazdur, M.D., acting director of the Office of Hematology and Oncology Products in the FDA’s Center for Drug Evaluation and Research and director of the FDA’s Oncology Center of Excellence. “The scientific community continues to make advances targeting the body’s immune system mechanisms for the treatment of various types of cancer. These advancements are leading to new therapies—even in rare forms of cancer where treatment options are limited or non-existent.”

According to the National Cancer Institute, approximately 1,600 people in the United States are diagnosed with MCC every year. While the majority of patients present with localized tumors that can be treated with surgical resection, approximately half of all patients will experience recurrence, and more than 30 percent will eventually develop metastatic disease. In patients with metastatic MCC, the cancer has spread beyond the skin into other parts of the body.

Bavencio targets the PD-1/PD-L1 pathway (proteins found on the body’s immune cells and some cancer cells). By blocking these interactions, Bavencio may help the body’s immune system attack cancer cells.

Bavencio received an Accelerated Approval, which enables the FDA to approve drugs for serious conditions to fill an unmet medical need using clinical trial data that is thought to predict a clinical benefit to patients. Further clinical trials are required to confirm Bavencio’s clinical benefit and the sponsor is currently conducting these studies.

Today’s approval of Bavencio was based on data from a single-arm trial of 88 patients with metastatic MCC who had been previously treated with at least one prior chemotherapy regimen. The trial measured the percentage of patients who experienced complete or partial shrinkage of their tumors (overall response rate) and, for patients with a response, the length of time the tumor was controlled (duration of response). Of the 88 patients who received Bavencio in the trial, 33 percent experienced complete or partial shrinkage of their tumors. The response lasted for more than six months in 86 percent of responding patients and more than 12 months in 45 percent of responding patients.

Common side effects of Bavencio include fatigue, musculoskeletal pain, diarrhea, nausea, infusion-related reactions, rash, decreased appetite and swelling of the limbs (peripheral edema). The most common serious risks of Bavencio are immune-mediated, where the body’s immune system attacks healthy cells or organs, such as the lungs (pneumonitis), liver (hepatitis), colon (colitis), hormone-producing glands (endocrinopathies) and kidneys (nephritis). In addition, there is a risk of serious infusion-related reactions. Patients who experience severe or life-threatening infusion-related reactions should stop using Bavencio. Women who are pregnant or breastfeeding should not take Bavencio because it may cause harm to a developing fetus or a newborn baby.

The FDA granted this application Priority Review and Breakthrough Therapydesignation. Bavencio also received Orphan Drug designation, which provides incentives to assist and encourage the development of drugs for rare diseases.

The FDA granted accelerated approval of Bavencio to EMD Serono Inc.

Image result for avelumab

Image result for avelumab

Avelumab
Monoclonal antibody
Type ?
Source Human
Legal status
Legal status
  • Investigational
Identifiers
CAS Number
ChemSpider
  • none
UNII
KEGG

Avelumab (MSB0010718C) is a fully human monoclonal PD-L1antibody of isotypeIgG1, currently in development by Merck KGaA, Darmstadt, Germany & Pfizer for use in immunotherapy, especially for treatment of Non-small-cell lung carcinoma (NSCLC) .[1]

Mechanism of action

Avelumab binds to the PD ligand 1 and therefore inhibits binding to its receptor programmed cell death 1 (PD-1). Formation of a PD-1/PD-L1 receptor/ligand complex leads to inhibition of CD8+ T cells, and therefore inhibition of an immune reaction. Immunotherapy aims at ceasing this immune blockage by blocking those receptor ligand pairs. In the case of avelumab, the formation of PD-1/PDL1 ligand pairs is blocked and CD8+ T cell immune response should be increased. PD-1 itself has also been a target for immunotherapy.[2] Therefore, avelumab belongs to the group of Immune checkpoint blockade cancer therapies.

Clinical trials

As of May 2015, according to Merck KGaA, Darmstadt, Germany & Pfizer, avelumab has been in Phase Iclinical trials for bladder cancer, gastric cancer, head and neck cancer, mesothelioma, NSCLC, ovarian cancer and renal cancer. For Merkel-cell carcinoma, Phase II has been reached and for NSCLC there is also a study already in Phase III.[1]

Merkel-cell carcinoma

On March 23, 2017, the U.S. Food and Drug Administration granted accelerated approval to avelumab (BAVENCIO, EMD Serono, Inc.) for the treatment of adults and pediatric patients 12 years and older with metastatic Merkel cell carcinoma (MCC).

Approval was based on data from an open-label, single-arm, multi-center clinical trial (JAVELIN Merkel 200 trial) demonstrating a clinically meaningful and durable overall response rate (ORR). All patients had histologically confirmed metastatic MCC with disease progression on or after chemotherapy administered for metastatic disease.

ORR was assessed by an independent review committee according to Response Evaluation Criteria in Solid Tumors (RECIST) 1.1. The ORR was 33% (95% confidence interval [CI]: 23.3, 43.8), with 11% complete and 22% partial response rates. Among the 29 responding patients, the response duration ranged from 2.8 to 23.3+ months with 86% of responses durable for 6 months or longer. Responses were observed in patients regardless of PD-L1 tumor expression or presence of Merkel cell polyomavirus.

Safety data were evaluated in 1738 patients who received avelumab, 10 mg/kg, every 2 weeks. The most common serious adverse reactions to avelumab are immune-mediated adverse reactions (pneumonitis, hepatitis, colitis, adrenal insufficiency, hypo- and hyperthyroidism, diabetes mellitus, and nephritis) and life-threatening infusion reactions. Among the 88 patients enrolled in the JAVELIN Merkel 200 trial, the most common adverse reactions were fatigue, musculoskeletal pain, diarrhea, nausea, infusion-related reaction, rash, decreased appetite, and peripheral edema. Serious adverse reactions that occurred in more than one patient in the trial were acute kidney injury, anemia, abdominal pain, ileus, asthenia, and cellulitis.

The recommended dose and schedule of avelumab is 10 mg/kg as an intravenous infusion over 60 minutes every 2 weeks. All patients should receive premedication with an antihistamine and acetaminophen prior to the first four infusions of avelumab.

Full prescribing information for avelumab is available at: http://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761049s000lbl.pdf

References

  1. ^ Jump up to:a b Merck-Pfizer Alliance. “Merck-Pfizer Alliance Avelumab Fact Sheet” (PDF). Retrieved 2 December 2015.
  2. Jump up^ Hamid, O; Robert, C; Daud, A; Hodi, F. S.; Hwu, W. J.; Kefford, R; Wolchok, J. D.; Hersey, P; Joseph, R. W.; Weber, J. S.; Dronca, R; Gangadhar, T. C.; Patnaik, A; Zarour, H; Joshua, A. M.; Gergich, K; Elassaiss-Schaap, J; Algazi, A; Mateus, C; Boasberg, P; Tumeh, P. C.; Chmielowski, B; Ebbinghaus, S. W.; Li, X. N.; Kang, S. P.; Ribas, A (2013). “Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma”. New England Journal of Medicine. 369 (2): 134–44. doi:10.1056/NEJMoa1305133. PMC 4126516Freely accessible. PMID 23724846.

//////////fda 2017, Bavencio, avelumab, EMD Serono Inc., Priority Review,  Breakthrough Therapy designation.  Orphan Drug designation, skin cancer

 

 

UPDATE ON EMA

Bavencio : EPAR – Summary for the public EN = English 13/10/2017

 http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Summary_for_the_public/human/004338/WC500236650.pdf

Product details

Name Bavencio
Agency product number EMEA/H/C/004338
Active substance avelumab
International non-proprietary name(INN) or common name avelumab
Therapeutic area Neuroendocrine Tumors
Anatomical therapeutic chemical (ATC) code L01XC31
Additional monitoring This medicine is under additional monitoring. This means that it is being monitored even more intensively than other medicines. For more information, see medicines under additional monitoring.
Treatment of rare diseases This medicine has an “orphan designation” which means that it is used to treat life-threatening or chronically debilitating conditions that affect no more than five in 10,000 people in the European Union, or are medicines which, for economic reasons, would be unlikely to be developed without incentives.
Conditional Approval Sometimes, the CHMP recommends that a medicine be given ‘conditional approval’. This happens when the Committee has based its positive opinion on data which, while not yet comprehensive, indicate that the medicine’s benefits outweigh its risks.

The company is given obligations to fulfil, such as the performance of further studies. The approval is renewed on a yearly basis until all obligations have been fulfilled, and is then converted from a conditional approval into a normal approval. Conditional approvals can only be granted for medicines that satisfy an ‘unmet medical need’, meaning the medicine is intended to be used for a disease or condition for which no treatment is readily available, and it is therefore important that patients have early access to the medicine concerned.

Publication details

Marketing-authorisation holder Merck Serono Europe Limited
Revision 1
Date of issue of marketing authorisation valid throughout the European Union 18/09/2017

Contact address:

Merck Serono Europe Limited
56 Marsh Wall
London E14 9TP
United Kingdom

FDA approves drug Xadago (Safinamide, сафинамид , سافيناميد , 沙非胺 , ) to treat Parkinson’s disease


ChemSpider 2D Image | Safinamide | C17H19FN2O2

Safinamide

  • Molecular Formula C17H19FN2O2
  • Average mass 302.343 Da
(2S)-2-[[[4-[(3-Fluorophenyl)methoxy]phenyl]methyl]amino]propanamide
133865-89-1 ,
сафинамид ,
سافيناميد 
沙非胺 
EMD-1195686, ZP-034, FCE-28073(R-isomer), PNU-151774E, NW-1015, FCE-26743
CAS   202825-46-5 (mesylate) SEE BELOW

str1

(+)-(S)-2-[[p-[(m-fluorobenzyl)oxy]benzyl]amino]propionamide monomethanesulfonate

Propanamide, 2-[[[4-[(3-fluorophenyl)methoxy]phenyl]methyl]amino]-, (2S)-, methanesulfonate

Molecular Weight 398.45
Formula C17H19FN2O2 ● CH4O3S

CAS 202825-46-5 (Safinamide Mesylate)

Safinamide is a white to off-white, non-hygroscopic crystalline solid. It shows pH dependent solubility in aqueous buffers due to the secondary amine moiety, being soluble at acidic pH and practically insoluble at neutral pH.

It is freely soluble in de-ionized water, methanol and DMSO but practically insoluble in non-polar organic solvents.

Safinamide is chiral and possesses a single stereogenic centre.

Three crystalline forms are known. The anhydrous form selected for commercialisation is the most thermodynamically stable form, whilst the others are either not physiologically relevant or have very similar dissolution profiles. SOURCE EMA

Safinamide methanesulfonate was approved by European Medicine Agency (EMA) on Feb 22, 2015. It was developed by Newron and Zambon, then marketed as Xadago® by Zambon in EU.

FDA approved March 21, 2017,

STR1

Safinamide is a unique molecule with a novel dual mechanism of action based on the enhancement of the dopaminergic function (through potent reversible inhibition of MAO-B and of dopamine uptake) and inhibition of the excessive release of glutamate. It is indicated for the treatment of Parkinson’s disease (PD).

Xadago® is available as film-coated tablet for oral use, containing Eq. 50 mg/100 mg of free Safinamide. The recommended dose is 50 mg or 100 mg once daily.

SYNTHESIS WILL BE UPDATED…………..
03/21/2017
The U.S. Food and Drug Administration today approved Xadago (safinamide) tablets as an add-on treatment for patients with Parkinson’s disease who are currently taking levodopa/carbidopa and experiencing “off” episodes. An “off” episode is a time when a patient’s medications are not working well, causing an increase in Parkinson’s symptoms, such as tremor and difficulty walking.

March 21, 2017, Release

The U.S. Food and Drug Administration today approved Xadago (safinamide) tablets as an add-on treatment for patients with Parkinson’s disease who are currently taking levodopa/carbidopa and experiencing “off” episodes. An “off” episode is a time when a patient’s medications are not working well, causing an increase in Parkinson’s symptoms, such as tremor and difficulty walking.

“Parkinson’s is a relentless disease without a cure,” said Eric Bastings, M.D., deputy director of the Division of Neurology Products in the FDA’s Center for Drug Evaluation and Research. “We are committed to helping make additional treatments for Parkinson’s disease available to patients.”

An estimated 50,000 Americans are diagnosed with Parkinson’s disease each year, according to the National Institutes of Health, and about one million Americans have the condition. The neurological disorder typically occurs in people over age 60, though it can occur earlier, when cells in the brain that produce a chemical called dopamine become impaired or die. Dopamine helps transmit signals between the areas of the brain that produce smooth, purposeful movement – such as eating, writing, and shaving. Early symptoms of the disease are subtle and occur gradually. In some people, Parkinson’s disease progresses more quickly than in others.

The efficacy of Xadago in treating Parkinson’s disease was shown in a clinical trial of 645 participants who were also taking levodopa and were experiencing “off” time. Those receiving Xadago experienced more beneficial “on” time, a time when Parkinson’s symptoms are reduced, without troublesome uncontrolled involuntary movement (dyskinesia), compared to those receiving a placebo. The increase in “on” time was accompanied by a reduction in “off” time and better scores on a measure of motor function assessed during “on” time than before treatment.

In another clinical trial of 549 participants, the participants adding Xadago to their levodopa treatment had more “on” time without troublesome uncontrolled involuntary movement compared to those taking a placebo, and also had better scores on a measure of motor function assessed during “on” time than before treatment.

Certain patients should not take Xadago. These include patients who have severe liver problems, or who take a medicine used to treat a cough or cold called dextromethorphan. It also should not be taken by patients who take another medicine called a monoamine oxidase inhibitor (MAOI) because it may cause a sudden severe increase in blood pressure, or by those who take an opioid drug, St. John’s wort, certain antidepressants (such as serotonin-norepinephrine reuptake inhibitors, tricyclics, tetracyclics, and triazolopyridines), or cyclobenzaprine, because it may cause a life-threatening reaction called serotonin syndrome.

The most common adverse reactions observed in patients taking Xadago were uncontrolled involuntary movement, falls, nausea, and trouble sleeping or falling asleep (insomnia).

Serious, but less common, risks include the following: exacerbated high blood pressure (hypertension); serotonin syndrome when used with MAOIs, antidepressants, or opioid drugs; falling asleep during activities of daily living; hallucinations and psychotic behavior; problems with impulse control/compulsive behaviors; withdrawal-emergent hyperpyrexia (fever) and confusion; and retinal pathology.

The FDA granted approval of Xadago to Newron Pharmaceuticals.

Safinamide (INN; brand name Xadago) is a drug indicated for the treatment of Parkinson’s disease with monoamine oxidase B inhibiting and other methods of action.[2] It was approved in Europe in February 2015,[3] and in the United States on March 21, 2017[4]. It has also been tested for the use in patients with restless legs syndrome (RLS), but no study results have been published.

Image result for SAFINAMIDE SYNTHESIS

Medical uses

Safinamide has been approved by the European Medicines Agency for the treatment of adult patients with idiopathic Parkinson’s disease as add-on therapy to a stable dose of levodopa (L-dopa) alone or in combination with other Parkinson drugs in patients with mid-to-late-stage fluctuating disease.[5]

Contraindications

Safinamide is contraindicated in patients with severe liver impairment, with albinism, retinitis pigmentosa, severe diabetic neuropathy, uveitis and other disorders of the retina. Combination with other monoamine oxidase (MAO) inhibitors and pethidine is also contraindicated.[6]

Adverse effects

Common adverse events in clinical trials (in more than 1% of patients) included nausea, dizziness, tiredness, sleeplessness, orthostatic hypotension (low blood pressure), and headache. There was no significant difference in the occurrence of these effects between safinamide and placebo treated patients.[6][7]

In experiments with rats (but not in those with monkeys), retinopathies have been observed.[1][8]

Overdose

Expected overdose effects are hypertension (high blood pressure), orthostatic hypotension, hallucinations, psychomotor agitation, nausea, vomiting, and dyskinesia. In studies, a singe patient was suspected to have overdosed for a month; symptoms were confusion, drowsiness and mydriasis (dilation of the pupils) and subsided completely after the drug was discontinued. No specific antidote is available.[6]

Interactions

As a MAO inhibitor, safinamide can theoretically cause hypertensive crises, serotonin syndrome and other severe side effects when combined with other MAO inhibitors or with drugs that are known to interact with MAO inhibitors, such as pethidine, dextromethorphan, selective serotonin reuptake inhibitors (SSRIs), serotonin–noradrenaline reuptake inhibitors (SNRIs), tricyclic and tetracyclic antidepressants. An interaction with tyramine, a substance found in various foods, could be expected by the same reasoning but has been excluded in studies.[6]

Another theoretical interaction is with drugs with affinity to the transporter protein ABCG2 (also known as BCRP), such as pitavastatin, pravastatin, ciprofloxacin, methotrexat, and diclofenac; a study with the latter has shown no clinical relevance.[9] A study testing possible interactions with amidase inhibitors is part of the post-authorisation development plan.[1] There are no relevant interactions related to cytochrome P450 (CYP) liver enzymes, although one inactivation pathway of safinamide seems to be mediated by CYP3A4.[6]

Pharmacology

Mechanisms of action

Like the older antiparkinson drugs selegiline and rasagiline, safinamide is a selective monoamine oxidase B inhibitor, reducing degradation of dopamine; in contrast to the other two, its action is reversible. Safinamide also inhibits glutamate release[7][10] and dopamine reuptake.[11] Additionally, it blocks sodium and calcium channels,[10][12] the relevance of which for its antiparkinson action is however unknown.[6]

Pharmacokinetics

Safinamide is absorbed quickly and nearly completely from the gut and reaches highest blood plasma concentrations after 1.8 to 2.8 hours. There is no relevant first-pass metabolism; total bioavailability is 95%. The substance is bound to plasma proteins to 88–90%.[6]

The metabolism is not well understood. The principal step is mediated by amidases which have not been identified, and produces safinamide acid (NW-1153). Other relevant metabolites are O-debenzylated safinamide (NW-1199),[9] the N-dealkylated amine which is then oxidized to a carboxylic acid (NW-1689), and the glucuronide of the latter.[6][13] In tests with liver microsomes, dealkylation seemed to be mediated by CYP3A4, but other CYP enzymes appear to be involved as well. Safinamide acid binds to the organic anion transporter 3 (OAT3), but this has probably no clinical relevance. Safinamide itself transiently binds to ABCG2. No other transporter affinities have been found in preliminary studies.[6]

Safinamide is eliminated, mainly (>90%) in form of its metabolites, via the kidney, with an elimination half-life of 20 to 30 hours. Only 1.5% are found in the stool.[6]

Metabolism pathways of safinamide.[9][13] Enzymes: CYP = cytochrome P450, MAO-A = monoamine oxidase A, ALDH = aldehyde dehydrogenases, UGT = UDP-glucuronosyltransferases. Gluc = acyl glucuronide.

History

The compound was originally discovered at Farmitalia-Carlo Erba, which was acquired by Pharmacia in 1993. In 1995, Pharmacia merged with Upjohn. Safinamide was first disclosed in 1998.[14] In the course of a major restructuring in the same year, all rights for safinamide were transferred to the newly formed company Newron Pharmaceuticals, which developed the drug until it was sold to Merck KGaA in 2006.[15]

In 2007, a Phase III clinical trial was started, scheduled to run until 2011.[16] In October 2011 Merck, now Merck-Serono, announced that they would give all rights to develop the compound back to Newron because they wanted to prioritise other projects and had corrected their estimates for safinamide’s market potential downwards.[17]

The US Food and Drug Administration (FDA) refused to file Newron’s application in 2014 on formal grounds.[18] Newron re-applied in December 2014.[19] In spring 2015, the European Medicines Agency (EMA) approved the drug. Safinamide is the first antiparkinson medication to be approved for ten years.[8]

Research

Potential additional uses might be restless legs syndrome (RLS) and epilepsy.[20] They were being tested in Phase II trials in 2008, but no results are available.

str1

(+)-(S)-2-[[p-[(m-fluorobenzyl)oxy]benzyl]amino]propionamide monomethanesulfonate

Propanamide, 2-[[[4-[(3-fluorophenyl)methoxy]phenyl]methyl]amino]-, (2S)-, methanesulfonate

Molecular Weight 398.45
Formula C17H19FN2O2 ● CH4O3S

CAS 202825-46-5 (Safinamide Mesylate)

Safinamide is a white to off-white, non-hygroscopic crystalline solid. It shows pH dependent solubility in aqueous buffers due to the secondary amine moiety, being soluble at acidic pH and practically insoluble at neutral pH.

It is freely soluble in de-ionized water, methanol and DMSO but practically insoluble in non-polar organic solvents.

Safinamide is chiral and possesses a single stereogenic centre.

Three crystalline forms are known. The anhydrous form selected for commercialisation is the most thermodynamically stable form, whilst the others are either not physiologically relevant or have very similar dissolution profiles.SOURCE EMA

Safinamide methanesulfonate was approved by European Medicine Agency (EMA) on Feb 22, 2015. It was developed by Newron and Zambon, then marketed as Xadago® by Zambon in EU.

FDA approved March 21, 2017

Safinamide is a unique molecule with a novel dual mechanism of action based on the enhancement of the dopaminergic function (through potent reversible inhibition of MAO-B and of dopamine uptake) and inhibition of the excessive release of glutamate. It is indicated for the treatment of Parkinson’s disease (PD).

Xadago® is available as film-coated tablet for oral use, containing Eq. 50 mg/100 mg of free Safinamide. The recommended dose is 50 mg or 100 mg once daily.

SYNTHESIS

Safinamide has been obtained by reductocondensation of 4-(3-fluorobenzyloxy)benzaldehyde (I) with L-alaninamide (II) by means of sodium cyanoborohydride in methanol.EP 0400495; EP 0426816; JP 1992500215; US 5236957; US 5391577; US 5502079; WO 9014334

CLIP

http://pubs.rsc.org/en/content/articlehtml/2016/sc/c6sc00197aImage result for SAFINAMIDE SYNTHESIS

image file: c6sc00197a-s2.tif

Scheme 2 Synthesis and isolation of [18F]safinamide, [18F]FMT, and [18F]mFBG.

PATENT

WO2009074478A1

Safinamide (NW- 1015, FCE-26743A, PNU- 151774E) is a sodium channel blocker, a calcium channel modulator, a monoamino oxidase B (MAO-B) inhibitor, a glutamate release inhibitor and a dopamine metabolism modulator. Safinamide is useful in the treatment of CNS disorders, in particular of epilepsy, Parkinson’s disease, Alzheimer’s disease, depression, restless legs syndrome and migraine (WO 90/ 14334, WO 2004/089353, WO 2005/ 102300 and WO 2004/062655). Ralfinamide (NW- 1029, FCE-26742A, PNU-0154339E) is a sodium channel blocker useful in the treatment of pain conditions, including chronic pain and neuropathic pain, migraine, bipolar disorders, depressions, cardiovascular, inflammatory, urogenital, metabolic and gastrointestinal disorders (WO 99/35125, WO 03/020273, WO 2004/062655, WO 2005/018627, WO 2005/070405, WO 2005/ 102300).

In particular, safinamide is specifically described in WO 90/ 14334. Safinamide, its R-enantiomer, their racemic mixture and their salts with pharmaceutically acceptable acids and the use thereof for the preparation of pharmaceutical compositions active as anti-epileptic, anti-Parkinson, neuroprotective, antidepressant, antispastic and/or hypnotic agents are specifically claimed in WO 90/ 14334. Ralfinamide is specifically described in WO 90/ 14334. Ralfinamide, its R- enantiomer, their racemic mixture and their salts with pharmaceutically acceptable acids and their use thereof for the preparation of pharmaceutical compositions active as anti-epileptic, anti-Parkinson, neuroprotective, antidepressant, antispastic and/or hypnotic agent are comprised by the claims of WO 90/ 14334.

Moreover, the use as analgesics of safinamide, ralfinamide, the respective R-enantiomers, the respective racemic mixtures and their salts with pharmaceutically acceptable acids is claimed in WO 99/035125. WO 2006/027052 A2 specifically discloses and claims the use of the single R-enantiomer of ralfinamide i.e., (R)-2-[4-(2- fluorobenzyloxy)benzylamino]propanamide (I’b), and its salts with pharmaceutically acceptable acids as a selective sodium and calcium channel modulator for the selective treatment of pathological affections wherein sodium or calcium channel mechanism(s) play(s) a pathological role, including pain, migraine, inflammatory processes affecting all body systems, disorders affecting skin and related tissue, disorders of the respiratory system, disorders of the immune and endocrinological systems, gastrointestinal, and urogenital disorders, wherein the therapeutical activity of said compound is substantially free from any MAO inhibitory side effect or exhibits significantly reduced MAO inhibitory side effect.

It has now been discovered that the large scale preparations of safinamide and ralfinamide according to the methods described in the prior art, contain two undesired impurities, i.e., respectively, (S)-2-[3-(3- fluorobenzyl)-4-(3-fluorobenzyloxy)-benzylamino]propanamide (Ha) and (S)- 2-[3-(2-fluorobenzyl)-4-(2-fluorobenzyloxy)-benzylamino]propanamide (lib), and their salt, in particular the respective methanesulfonates (lie) and (Hd)

Figure imgf000004_0001

(Ha) (lib)

The same situation occurs with the preparation according the prior art methods for the R-enantiomers (I’a) and (I’b) of, respectively, safinamide and ralfinamide, the respective racemic mixtures (Ia, I’a) and (Ib, I’b), and the salts thereof with pharmaceutically acceptable acids, (I’c), (I’d) and the respective racemic mixtures (Ic, I’c) and (Id, I’d) in particular the methanesulfonates, which result to be contaminated by the respective R isomers (Il’a), (Il’b), (II’c), and (Il’d) of the above identified impurities (Ha), (lib), (lie) and (Hd) or the respective racemic mixtures (Ha, Il’a), (lib, Il’b), (Hc, II’c) and (Hd, Il’d).

PATENT

WO2014178083A1.

Parkinson’s disease (PD) is a progressive neurodegenerative disease characterized by bradykinesia, rigidity, resting tremor, and ataxia. These symptoms are caused by decreased dopamine release in the striatum. Clinically, PD is defined by presence of Lewy bodies, intracellular neuronal inclusions in the substantia nigra and at other sites in the brain. Estimated prevalence of this disease is 100 to 200 per 100,000 population including males and females across the entire age group. Current treatment for PD comprises dopaminergic medications that include levodopa, dopamine agonists (DAs), monoamine oxidase-B (MAO-B) inhibitors. Figure 1 provides few examples of pharmaceutically important benzyloxy-benzylamine derivatives. Many of these benzyl oxy-benzylamines with various amine functions were studied and has been patented as sodium channel blockers. Among them, safinamide ((5)-N2– {4-[3- fluorobenzyl)oxy] benzyl}- alaninamide methanesulfonate) is a noted example which is under phase III clinical trials for treatment of Parkinson’s disease. Its mechanism of action is manifold which comprise MAO-B and dopamine uptake inhibition. Further, safinamide is believed to block voltage-dependent sodium channels, modulates calcium channels and reduction of glutamate release in the central nervous system. WOl 998003472 discloses serinamide, glycinamide, alaninamide and phenylalaninamide derivatives of a compound (I). These compounds (I) are useful for the treatment of neurological diseases.

EP2474521 discloses high purity degree (S)-2-[4-(3-fluorobenzyloxy)- benzylamino]propanamide (safinamide) or (S)-2-[4-(2-fluorobenzyloxy)- benzylamino]propanamide (ralfinamide) or a salt thereof with a pharmaceutically acceptable acid with a content of the respective impurity (S)-2-[3-(3-fluorobenzyl)-4-(3- fluorobenzyloxy)-benzylamino]propanamide or (S)-2-[3-(2-fluorobenzyl)-4-(2- fluorobenzyloxy)-benzylamino]propanamide.

US2009149544 relates to novel alpha- aminoamide derivatives, their pharmaceutically acceptable salts, solvates, and hydrates thereof. The application also provides compositions comprising a compound and the use of such compositions in methods of treating diseases and conditions that are beneficially treated by administering an inhibitor of monoamine oxidase type B (MAO-B) and/or a sodium (Na.sup.+) channel blocker, and/or a calcium (Ca.sup.2+) channel modulator.

The strategy employed in the art to prepare benzyloxy-benzylamine derivatives including safinamide or its analogue ralfinamide is chiral pool approach starting from L-alaniriamide and reductively aminating with 4-(3-fluorobenzyloxy) benzaldehyde. Although this method is very simple and straightforward, it suffers from several serious drawbacks, such as need to use toxic reagents such as sodium cyanoborohydride and further formation of toxic by-products such as hydrogen cyanide and sodium cyanide and other toxic impurities in large-scale production Importantly, the possibility of generating a range of safinamide analogues by means of the chiral-pool approach is limited in terms of the structure and stereochemistry of the products because of inadequacies in the availability of D-alaninamide and its analogues

Hence, the developments of newer methods for the preparation of compounds of formula (I) comprising safinamide and related analogues are highly desirable

Example 2: Synthesis of (R)-l-(benzyIoxy)propan-2-ol [(R)-compound 3]

To a solution of (7? benzyl glycidyl ether [fR)-compound 2] (4 g, 24.4 mmol) in dry THF (10 mL) at 0 °C, a pre-cooled solution of lithium aluminium hydride (1.4 g, 36.6 mmol) in anhydrous THF (10 mL) was added slowly with stirring under nitrogen. After 60 min, the reaction mixture was quenched with 1 ml of water and 1 ml of 15 % NaOH solution and the content was stirred for 15 min. The inorganic precipitate was filtered, washed with ethyl acetate and the solvent evaporated under reduced pressure. The residue was purified by a short filtration column to afford (-fl)-compound 3 as a colorless oil (3.8 g, 95%); [a]22D = -14.5 (c 2, CHC13); IR (CHC13): vmax3418, 3087, 3063, 3030, 2963, 2924, 1952, 1873, 1600, 1495, 1454, 1363, 1244, 1099, 1028, 918, 808, 698 cm“1; Ή NMR (200 MHz, CDC13): δΗ 1.13 (d, J = 6.3 Hz, 3H), 2.5 (bs, 1H), 3.23-3.32 (dd, J = 9.8, 1.3 Hz, 1H), 3.43-3.49 (dd, J = 9.45, 3.2 Hz, 1H), 3.91-4.03 (m, 1H), 4.55 (s, 2H), 7.25-7.37 (m, 5H); I3C NMR (50 MHz, CDC13): 5C 137.8 (C), 128.3 (CH, 2 carbons), 127.7 (CH, 3 carbons), 75.7 (CH2), 73.2 (CH2), 66.4 (CH), 18.6 (CH3); MS: m/z 189 [M+Na]+.

Example 3: Synthesis of (S)-((2-azidopropoxy)methyl)benzene [(S)- compound 4]

To a stirred solution of secondary alcohol ( )-compound 3 (3 g, 18.1 mmol) in dry dichloromethane (25 mL), Et3N (3.1 mL, 21.7 mmol) at 0 °C was added, followed by drop wise addition of mesyl chloride (1.8 mL, 21.7 mmol). The reaction mixture was stirred at 0°C for 2 hours, subsequently at room temperature for 3 hours under a nitrogen atmosphere. After completion of the reaction (indicated by TLC), the reaction mixture was diluted with dichloromethane and washed with a saturated solution of sodium bicarbonate (30 mL) and water (2 x 10 mL). The organic layer was separated, dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure to give the O-mesyl compound (4.3 g; crude).

To a solution of the crude 0-mesyl compound (4 g, 16.37 mmol) in dry DMF (10 mL), sodium azide (1.6 g, 24.55 mmol) was added and the reaction mixture was heated at 60°C for 6 hours under nitrogen atmosphere. After completion of the reaction (indicated by TLC), water (10 mL) was added to the reaction mixture, then extracted with ethyl acetate (2 x 15 mL). The combined organic layers were washed with brine solution, dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure. Purification of the crude residue was done by column chromatography (silica gel, petroleum ether/EtOAc, 95:5) to yield (¾)-compound 4 as a colorless oil. (2.8 g; 89%); [a]22D = +6.1 (c 1.3, CHC13); IR (CHC13): vmax 3394, 3032, 2977, 2864, 2500, 2104, 1724, 1641 , 1496, 1454, 1363, 1269, 1 101 , 913, 698 αη ‘,Ή NMR (200 MHz, CDC13): δΗ 1.20 (d, J = 6.7 Hz, 3H), 3.39-3.54 (m, 2H), 3.61-3.77 (m, 1H), 4.57 (s, 2H), 7.25-7.39 (m, 5H); 13C NMR (50 MHz, CDC13): 5C 137.8 (C), 128.4 (CH, 2 carbons), 127.7 (CH), 127.5 (CH, 2 carbons), 73.7 (CH2), 73.2 (CH2), 56.9 (CH), 16.1 (CH3);MS: m/z 214 [M+Na]+.

Example 4: Synthesis of (S)-N-(l-hydroxypropan-2-yl)-2-nitrobenzenesulfonamide [(S)- compound 5]

To a solution of ^-compound 4 (2.5 g, 13.1 mmol) in methanol (15 mL), trifluoroacetic acid (2 mL) and palladium hydroxide on activated carbon (0.05 g, 10-20 wt %) were added and the reaction mixture was stirred under hydrogen (60 psi) for 8 hours. After completion of the reaction (indicated by TLC), the catalyst was filtered over a plug of celite and the solvent was evaporated under reduced pressure to half of its volume which was basified with 2.5 M methanolic NaOH. Evaporation of the remaining solvent under reduced pressure was done followed by filtration of the residue through a short bed of basic alumina (eluent; MeOH) to obtain the amino alcohol as a pale brown oil (0.94 g, crude) which was subjected to the next reaction without further purification.

To a solution of amino alcohol (0.9 g, 1 1.98 mmol) in dry dichloromethane (5 mL), 2-nitrobenzenesulfonylchloride (3.2 g, 14.37 mmol) in dichloromethane (8 mL) and triethylamine (2.6 mL, 17.97 mmol) at 0 °C were slowly added under nitrogen atmosphere. The solution was stirred for 2 hours. After completion of the reaction (indicated by TLC), water (10 mL) was added to the reaction mixture, then extracted with dichloromethane (2 x 15 mL). The combined organic layers were washed with brine solution, dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure. Purification of the crude residue was done by column chromatography (silica gel, petroleum ether/EtOAc, 60:40) to yield (S)- compound 5 as a pale yellow oil (2.33 g, 75% ); [a]22D = +80.2 (c 2.1, CHClj); IR (CHC13): vmax 3546, 3367, 3022, 2883, 2401, 1594, 1542, 1412, 1362, 1216, 1170, 1 125, 1059, 971, 854, 668 cm“1; ]H NMR (200 MHz, CDC13): δΗ 1.13 (d, J = 6.5 Hz, 3H), 2.16 (bs, 1H), 3.45-3.70 (m, 3H), 5.61 (d, J = 6.6 Hz, 1H), 7.73-7.80 (m, 2H), 7.86-7.91 (m, 1H), 8.13-8.22 (m, 1H); 13C NMR (50 MHz, CDC13): 5C 147.8 (C), 134.4 (C), 133.7 (CH), 133.0 (CH), 130.9 (CH), 125.5 (CH), 66.2 (CH2), 52.5 (CH), 17.8 (CH3); MS: m/z 283 [M+Na]+.

Example 5: Synthesis of l-fluoro-3-(iodomethyl)benzene ( compound 7)

To a stirred solution of triphenyl phosphine (4.15 g, 15.85 mmol), imidazole (1.1 g, 15.85 mmol) in dry dichloromethane (20 mL), iodine (4.8 g, 19.02 mmol) at 0°C was added and the solution was stirred for 5 min. To this, 3-fluoro benzyl alcohol (compound 6) (2 g, 15.85 mmol) dissolved in dichloromethane (5 mL) was added drop wise over 10 min and the stirring was continued for 1 hour with exclusion of light. After completion of the reaction (indicated by TLC), the reaction mixture was quenched by addition of an aqueous Na2S203 solution (15 mL), then extracted with dichloromethane (2 x 20 mL). The combined organic layers were washed with brine solution, dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure. Purification of the crude residue was done by column chromatography (silica gel, petroleum ether/EtOAc, 95:5) to yield compound 7 as a colorless oil (3.5 g, 95% ); (IR (CHC13): vmax 3460, 3060, 2965, 1695, 1613, 1593, 1482, 1446, 1259, 1 156, 1068, 944, 871, 782, 736, 686 cm“1 ; Ή NMR (200 MHz, CDC13): δΗ 4.42 (s, 2H), 6.89-6.99 (m, 1H), 7.05-7.17 (m, 2H), 7.21-7,29 (m, 1H); 13C NMR (50 MHz, CDC13): 6C 165.0 (C), 141.6 (C), 130.2 (CH), 124.4 (CH), 1 15.9 (CH), 1 14.7 (CH), 3.9 (C¾).

Example 6: Synthesis of (4-((3-flurobenzyl)oxy)phenyl)methanol (compound 8)

To a stirred solution of 4-(hydroxymethyl)phenol (1.57 g, 12.7 mmol) and K2C03 (8.8 g, 63.55 mmol) in dry acetonitrile (25 mL), compound 7 (3 g, 12.7 mmol) in acetonitrile was slowly added and the reaction mixture was heated at 70°C for 6 hours. After completion of the reaction (indicated by TLC), water (20 mL) was added to the reaction mixture, then extracted with ethylacetate (3 x 20 mL). The combined organic layers were washed with brine solution, dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure. Purification of the crude residue was done by column chromatography (silica gel, petroleum ether/EtOAc, 70:30) to yield compound 8 as a colorless solid (2.7 g, 91% ); mp 63-65 °C; IR (CHC13): vmax 3422, 3017, 1612, 1512, 1489, 1381, 1216, 1 174, 1020, 829, 668 cm“1; Ή NMR (200 MHz, CDC13): δΗ 4.61 (s, 2H), 5.06 (s, 2H), 6.91-6.98 (m, 2H), 7.00-7.06 (m, 1H), 7.12-7.20 (m, 2H), 7.25-7.37 (m, 3H); 13C NMR (50 MHz, CDC13): 5C 165.4 (C), 160.5 (C), 158.0 (C), 139.6 (C), 133.5 (CH), 130.2 (CH), 128.7 (CH, 2 carbons), 122.7 (CH), 1 14.8 (CH, 2 carbons), 1 13.9 (CH), 69.1 (CH2), 64.9 (CH2); MS: m/z 255 [M+Na]+.

Example 7: Synthesis of l-fluoro-3-((4-(iodomethyl)phenoxy)methyI)benzene (compound 9)

To a stirred solution of triphenyl phosphine (2.82 g, 10.8 mmol), imidazole (0.73 g, 10.76 mmol) in dry dichloromethane (20 mL), iodine (3.27 g, 12.9 mmol) at 0 °C was added and the solution was stirred for 5 min. To this, compound 8 (2.5 g, 10.8 mmol) dissolved in dichloromethane (5 mL) was added drop wise over 10 min and the stirring was continued for 1 hour with exclusion of light. After completion of the reaction (indicated by TLC), the reaction mixture was quenched by addition of an aqueous Na2S203 solution (15 mL), then extracted with dichloromethane (2 x 20 mL). The combined organic layers were washed with brine solution, dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure. Purification of the crude residue was done by column chromatography (silica gel, petroleum ether/EtOAc, 95:5) to yield compound 9 as a colorless oil (3.4 g, 93%); IR (CHC13): vmax 3503, 3033, 2925, 2089, 1607, 1509, 1488, 1381, 1301, 1250, 1 155, 1079, 944, 869, 776, 684 cm“1; 1H NMR (200 MHz, CDC13): δΗ 4.47 (s, 2H), 5.04 (s, 2H), 6.85-6.91 (m, 2H), 6.96-7.02 (m, 1H), 7.05-7.12 (m, 1H), 7.16-7.20 (m, 1H), 7.29-7.40 (m, 3H).

,3C NMR (50 MHz, CDC13): 6C 165.4 (C), 160.5 (C), 158.1 (C), 131.9 (C), 130.2 (CH), 130.1 (CH, 2 carbons), 122.7 (CH), 1 15.1 (CH, 2 carbons), 1 14.7 (CH), 1 13.9 (CH), 69.2 (CH2), 6.33 (CH2).

Example 8: Synthesis of (S)-N-(4-((3-flurobenzyl)oxy)benzyl)-N-(l-hydroxypropan-2-yl)-2-nitrobenzenesulfonamide [(S)-compound 10]

To a stirred solution of (^-compound 5 (1 g, 3.8 mmol) and K2C03 (2.65 g, 19.2 mmol) in dry acetonitrile (25 mL), compound 9 (1.84 g, 5.4 mmol) in acetonitrile was slowly added and the reaction mixture was heated at 70°C for 72 hours. After completion of the reaction (indicated by TLC), water (20 mL) was added to the reaction mixture, then extracted with ethylacetate (3 15 mL). The combined organic layers were washed with brine solution, dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure. Purification of the crude residue was done by column chromatography (silica gel, petroleum ether/EtOAc, 80:20) to yield (¾)-compound 10 as a colorless oil (1.46 g, 80% ); [a]22D = +5.4 (c 1.5, CHC13); IR (CHC13): vmax 3445, 3020, 2928, 2400, 1613, 1544, 1512, 1453, 1371, 1216, 1 162, 1029, 852, 668 cm“1; 1H NMR (200 MHz, CDC13): δΗ 1.07 (d, J = 6.9 Hz, 3H), 1.91 (t, J = 5.2 Hz, 1H), 3.41-3.53 (m, 2H), 4.05-4.22 (m, 1H), 4.37-4.57 (m, 2H), 5.02 (m, 2H), 6.87 (d, J = 8.53 Hz, 2H), 6.97-7.12 (m, 2H), 7.20 (d, J = 7.2 Hz, 2H), 7.32 (d, J = 8.7 Hz, 2H), 7.47-7.67 (m, 3H), 7.89 (d, J = 8.09 Hz, 1H); 13C NMR (50 MHz, CDC13): 6C 165.5 (C), 160.6 (C), 158.4 (C), 147.7 (C), 139.6 (C), 134.1 (C), 133.4 (CH), 131.6 (CH), 131.4 (CH), 130.3 (CH), 129.7 (CH, 2 carbons), 124.1 (CH), 122.8 (CH), 115.1 (CH), 114. 9 (CH, 2 carbons), 114.0 (CH), 69.2 (CH2), 64.3 (CH2), 56.2 (CH), 46.9 (CH2), 15.4 (CH3); MS: m/z 497 [M+Na]+.

Example 9: Synthesis of (S)-2-(N-(4-((3-fluorobenzyl)oxy)benzyl)-2-nitrophenylsulfonamido) propanoic acid [(S)-compound 11]

A mixture of (S compound 10 (1.25 g, 2.6 mmol), TEMPO (0.028 g, 0.18 mmol), acetonitrile (20 mL), and sodium phosphate buffer (16 mL, 0.67 M, pH 6.7) was heated to 35°C. Next, sodium chlorite (0.47 g dissolved in 2 mL water, 7.9 mmol) and diluted bleach (4-6%, 0.09 mL diluted in 1 mL water) were added simultaneously over 1 hour. The reaction mixture was stirred at 35°C until the reaction was complete (3 hours, TLC), then cooled to room temperature. Water (30 mL) was added and the pH adjusted to 8 with 2 M NaOH. The reaction was quenched by pouring it into ice cold Na2S03 solution maintained at <20°C. After stirring for 30 min at room temperature, ethyl acetate (20 mL) was added and the stirring was continued for an additional 15 min. The organic layer was separated and discarded. More ethyl acetate (20 mL) was added, and the aqueous layer was acidified with 1 M HC1 to pH 3-4. The organic layer was separated, washed with water (2 x 15 mL), brine and concentrated under reduced pressure to afford the carboxylic acid (S -compound 1 1 (1.1 g, 85%); [ ]22ο = -20.4 (c 1.1, CHC13); IR (CHC13): vmax 3398, 3095, 1718, 1612, 1591, 1543, 1512, 1489, 1457, 1371, 1303, 1251, 1163, 1059, 900, 852, 831 , 778, 684 cm“1; 1H NMR (200 MHz, CDC13): 8H 1.44 (d, J = 7.3 Hz, 3H), 4.23 (d, J = 15.6 Hz, 1H), 4.64 (d, J = 15.6 Hz, 1H), 4.82-4.90 (q, J = 7.4 Hz, 1H), 4.92 (s, 2H), 6.68 (d, J = 8.6 Hz, 2H), 6.89-7.01 (m, 2H), 7.07-7.13 (m, 3H), 7.18-7.33 (m, 2H), 7.43-7.55 (m, 3H), 8.81 (bs, 1H); 13C NMR (50 MHz, CDC13): 5C 176.5 (CO), 165. 0 (C), 158.0 (C), 147.4 (C), 139.4 (C), 134.1 (C), 133.2 (CH), 131.4 (CH), 130.3 (CH), 129.9 (CH, 2 carbons), 128.4 (C), 124.1

(CH), 122.6 (CH), 1 15.0 (CH), 114.6 (CH, 2 carbons), 1 14.3 (CH), 1 13.8 (CH) 69.1 (CH2), 56.1 (CH), 49.0 (CH2), 16.8 (CH3); MS: m/z 51 1 [M+Na .

Example 10: Synthesis of (S)-2-(N-(4-((3-fluorobenzyI)oxy)benzyl)-2-nitrophenylsulfonamido) propanamide [(S)- compound 12]

To a solution of carboxylic acid (¾)-compound 1 1 (1 g, 2.04 mmol) and triethyl amine (0.34 mL, 2.4 mmol) in dry THF (20 mL), ethyl chloroformate (0.21 mL, 2.2 mmol) at 0 °C was added under nitrogen atmosphere. After 1 hour, ammonium hydroxide (25% w/v aqueous solution, 1.4 mL, 10.2 mmol) was added and the resulting reaction mixture was stirred at room temperature for 16 hours. After completion of the reaction, potassium carbonate (0.29 g, 2.1 mmol) was added and the reaction mixture was filtered, and washed with ethylacetate. The solvent was removed under reduced pressure and the crude product was subjected to column chromatography (silica gel, petroleum ether/EtOAc, 50:50) to obtain sulfonamide (Sj-compound 12 as a colorless oil (0.9 g, 91%); [a]22D = -32.1 (c 1.2, CHC13); IR (CHC13): vmax 3472, 1961 , 161 1, 1592, 1542, 1511, 1449, 1371, 1304, 1243, 1 163, 1060, 1029, 895, 852, 684 cm“1; Ή NMR (200 MHz, CDC13): δΗ 1.43 (d, J = 7.1 Hz, 3H), 4.44 (d, J = 15.4 Hz, 1H), 4.59 (d, J = 15.5 Hz, 1H), 4.60-4.71 (q, J= 7.0 Hz, 1 H), 5.01 (s, 2H), 5.50 (bs, 1H), 6.31 (bs, 1H), 6.78 (d, J = 8.71 Hz, 2H), 6.98-7.1 1 (m, 2H), 7.15-7.22 (m, 3H), 7.31-7.45 (m, 2H), 7.59-7.64 (m, 3H);13C NMR (50 MHz, CDC13): 5C 172.3 (CO), 165.5 (C), 158.2 (C), 147.5 (C), 139.6 (C), 139.4 (C), 133.6 (CH), 131.7 (CH), 130.5 (CH, 2 carbons),130.3 (CH), 128.1 (C), 124.2 (CH), 122.7 (CH), 1 15.1 (CH), 1 14.7 (CH, 2 carbons),1 14.4 (CH), 1 13.9 (CH), 69.0 (CH2), 55.7 (CH), 48.3 (CH2), 14.9 (CH3); MS: m/z 510 [M+Na]+.

Example 11: Synthesis of (S)-2-((4-((3-fluorobenzyl)oxy) benzyl) amino) propanamide [(S)-compound of formula I]

To a solution of sulfonamide (S)- compound 12 (0.8 g, 1.64 mmol), potassium carbonate (0.56 g, 4.9 mmol) in dry DMF (10 mL), thiophenol (0.2 mL, 1.9 mmol) was added. The reaction mixture was vigorously stirred for 6 hours. After completion of the reaction (indicated by TLC), water (10 mL) was added to the reaction mixture, then extracted with ethylacetate (2 x 20 mL). The combined organic layers were washed with brine solution, dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure. Purification of the crude residue was done by column chromatography (silica gel, petroleum ether/EtOAc, 60:40) to yield (S) -compound of formula I as a colorless solid (0.43 g, 86% ); mp 207-09 °C; [a]22D = +3.89 (c 1.55, CHC13); IR (CHC13): vmax 3341, 2970, 2927, 2853, 1648, 1592, 1512, 1489, 1445, 1406, 1384, 1254, 1176, 1 137, 1030, 953, 928, 829, 680 cm“1; Ή NMR (200 MHz, CDC13): δΗ 1.34 (d, J = 6.9 Hz, 3H), 2.49 (bs, 2H), 3.19-3.30 (q, J = 6.8 Hz, 1H), 3.63-3.78 (dd, J = 19.4, 3.9 Hz, 2H), 5.05 (s, 2H), 5.85 (bs, 1H), 6.95 (d, J = 8.7 Hz, 2H), 7.00-7.06 (m, 1H), 7.13-7.24 (m, 4H), 7.29-7.40 (m, 1H). 13C NMR (50 MHz, CDC13): 8C 178.3 (CO), 165.4 (C), 157.7 (C), 139.6 (C), 132.1 (C), 130.2 (CH), 129.3 (CH, 2 carbons), 122.7 (CH), 1 14.9 (CH, 2 carbons), 1 14.6 (CH), 1 13.9 (CH), 69.2 (CH2), 57.5 (CH), 51.9 (CH2), 19.6 (CH3); MS: m/z 302 [M]+, 325 [M+Na]+.

Example 12: Synthesis of (S)-Safinamide mesylate

To a stirred solution of (^-compound of formula I (0.1 g, 0.33 mmol) in ethylacetate (3 mL) at 70°C, methanesulfonic acid (0.02 mL, 0.33 mmol) was added and the reaction mixture was stirred for 2 hours. Subsequently, the temperature was lowered to 35°C and the stirring was continued for additional 1 hour. The solvent was evaporated under reduced pressure and the residue was filtered through a short bed of basic alumina [eluent: EtOAc/MeOH; (95:5)] to obtain safinamide mesylate as a white solid (0.11 g, 90%); mp 209-10 °C [lit.7mp 210]; [a]22D = +9.6 (c 1.1, AcOH); {lit.7 [a] D = +12.9 (c 1.1, AcOH)} ee >98% [The ee of safinamide mesylate was determined by chiral HPLC analysis; Chiralcel OD-RH (150 x 4.6 mm) column; eluent:

Methanol/ Acetonitrile/Buffer-TEAP, pH 3 (20: 10:70); flow rate 0.5 mL/min (780 psi); detector: 224 nm] [f¾)-isomer tR = 1 1.55 min, (SJ-isomer tR = 12.94 min].

PAPERS

Synthesis2014, 46, 1751-1756.

N2-{4-[(3-Fluorobenzyl)oxy]benzyl}-L-alaninamide [(S)-14] BASE FORM
PhSH (0.2 mL, 1.9 mmol) was added to a solution of sulfonamide (S)-13 (0.8 g, 1.64 mmol) and K2CO3 (0.56 g, 4.9 mmol) in anhyd DMF (10 mL), and the mixture was vigorously stirred for 6 h. When the reaction was complete (TLC), H2O (10 mL) was added and the mixture was extracted with EtOAc (2 × 20 mL). The organic layers were combined, washed with brine (2 × 10), dried (Na2SO4), filtered, and concentrated under reduced pressure. The crude residue was purified by column chromatography [silica gel, PE–EtOAc(60:40)] to give a colorless solid; yield: 0.43 g (86%); mp 207–09 °C;

[α]D22 +3.89 (c 1.55, CHCl3).
IR (CHCl3): 3341, 2970, 2927, 2853, 1648, 1592, 1512, 1489, 1445,1406, 1384, 1254, 1176, 1137, 1030, 953, 928, 829, 680 cm–1.

1H NMR (200 MHz, CDCl3): δH = 1.34 (d, J = 6.9 Hz, 3 H), 2.49 (brs, 2 H), 3.19–3.30 (q, J = 6.8 Hz, 1 H), 3.71 (dd, J = 19.4, 3.9 Hz, 2H), 5.05 (s, 2 H), 5.85 (br s, 1 H), 6.95 (d, J = 8.7 Hz, 2 H), 7.00–7.06 (m, 1 H), 7.13–7.24 (m, 4 H), 7.29–7.40 (m, 1 H).

13C NMR (50 MHz, CDCl3): δC = 178.3 (CO), 165.4 (C), 157.7 (C),139.6 (C), 132.1 (C), 130.2 (CH), 129.3 (CH, 2 C), 122.7 (CH), 114.9 (CH, 2 C), 114.6 (CH), 113.9 (CH), 69.2 (CH2), 57.5 (CH),51.9 (CH2), 19.6 (CH3).

MS: m/z = 302 [M]+, 325 [M + Na]+.

(S)-Safinamide Mesylate (1)
MsOH (0.02 mL, 0.33 mmol) was added to a stirred solution of sulfonamide (S)-14 (0.1 g, 0.33 mmol) in EtOAc (3 mL) at 70 °C, and the mixture was stirred for 2 h. The temperature was then lowered to 35 °C, and the mixture was stirred for an additional 1 h. The solvent was evaporated under reduced pressure and the residue was filtered
through a short bed of basic alumina with elution by EtOAc–MeOH; (95:5) to give a white solid; yield: 0.11 g (90%);

mp 209–210 °C [Lit.7a 210 °C];

[α]D22 +9.6 (c 1.1, AcOH); {Lit.7 [α]D22+12.9 (c 1.1, AcOH)}.
Chiral HPLC: column: Chiralcel OD-RH (150 × 4.6 mm); eluent:MeOH–MeCN–buffer-TEAP (pH 3) (20:10:70); flow rate: 0.5mL/min (780 psi); detector: 224 nm [(R)-isomer: tR = 11.55 min;
(S)-isomer: tR = 12.94 min]; ee >98%.

7a) Pevarello, P.; Bonsignori, A.; Dostert, P.;
Heidempergher, F.; Pinciroli, V.; Colombo, M.; McArthur,
R. A.; Salvati, P.; Post, C.; Fariello, R. G.; Varasi, M. J. Med.
Chem. 1998, 41, 579.

PAPER

Chin. J. Pharmas.2012, 43, 161-163.

…………….BASE

…………MESYLATE

PAPER

J. Med. Chem. 2007, 50, 4909-4916.

(S)-2-[6-(3-Fluorobenzyloxy)-3,4-dihydro-1H-isoquinolin-2-yl]-propionamide (21). The title compound was obtained using the same procedure described for the synthesis of (R)-2-[6-(3-fluorobenzyloxy)-3,4-dihydro-1H-isoquinolin-2-yl]propionamide, starting from 6-(3-fluorobenzyloxy)-1,2,3,4-tetrahydroisoquinoline (0.24 g, 0.95 mmol) and (R)-2-amino-1-methyl-2-oxoethyl-2-nitrobenzenesulfonate (0.52 g, 1.9 mmol). After column chromatography
purification using 99:1 DCM/MeOH as eluent, 0.075 g (24% yield) of the title compound was obtained as a pure white solid. Mp 153- 154 °C. 1H NMR (CDCl3) ä 1.35 (d, 3H, J ) 7.0), 2.67-2.97 (m, 4H), 3.28 (q, 1H, J ) 7.0), 3.64 (d, 1H, J ) 14.2), 3.77 (d, 1H, J ) 14.2), 5.05 (s, 2H), 5.36 (br, 1H), 6.74 (d, 1H, J ) 2.5), 6.79 (dd, 1H, J ) 8.5, 2.5), 6.97 (d, 1H, J ) 8.5), 6.99-7.06 (m, 1H), 7.06-7.24 (m, 3H), 7.30-7.40 (m, 1H).

J. Med. Chem.1998, 41, 579-590.

Molecules 21 00793 g001 1024

References

  1. “Summary of the risk management plan (RMP) for Xadago (safinamide)” (PDF). European Medicines Agency. January 2015.
  2.  Fariello, RG (2007). “Safinamide”. Neurotherapeutics. 4 (1): 110–116. doi:10.1016/j.nurt.2006.11.011. PMID 17199024.
  3.  “EPAR Summary for the Public for Xadago” (PDF). European Medicines Agency. February 2015.
  4.  “After an odyssey of setbacks, FDA finally green-lights Newron’s Parkinson’s drug Xadago”. endpts.com. Retrieved 2017-03-21.
  5.  Lawrence, Janna (2015-01-19). “Safinamide recommended for approval as Parkinson’s disease therapy”. The Pharmaceutical Journal. Royal Pharmaceutical Society. Retrieved 2015-01-19.
  6.  Haberfeld, H, ed. (2015). Austria-Codex (in German). Vienna: Österreichischer Apothekerverlag.
  7.  H. Spreitzer (14 April 2014). “Neue Wirkstoffe – Safinamid”. Österreichische Apothekerzeitung (in German) (8/2014): 30.
  8. Klement, A (18 July 2016). “Xadago”. Österreichische Apothekerzeitung (in German) (15/2016): 10.
  9.  “Summary of Product Characteristics for Xadago” (PDF). European Medicines Agency. 24 February 2015.
  10. ^ Jump up to:a b Caccia, C; Maj, R; Calabresi, M; Maestroni, S; Faravelli, L; Curatolo, L; Salvati, P; Fariello, RG (2006). “Safinamide: From molecular targets to a new anti-Parkinson drug”. Neurology. 67 (7 Suppl 2): S18–23. doi:10.1212/wnl.67.7_suppl_2.s18. PMID 17030736.
  11.  Merck Serono: Vielversprechende Daten zur kognitiven Wirkung von Safinamid bei Parkinson im Frühstadium. (German) 8 June 2007.
  12.  Pevarello, P; Bonsignori, A; Caccia, C; Amici, R; Salvati, P; Fariello, RG; McArthur, RA; Varasi, M (1999). “Sodium channel activity and sigma binding of 2-aminopropanamide anticonvulsants”. Bioorganic & Medicinal Chemistry Letters. 9 (17): 2521–2524. doi:10.1016/s0960-894x(99)00415-1.
  13. ^ Jump up to:a b Krösser, Sonja; Marquet, Anne; Gallemann, Dieter; Wolna, Peter; Fauchoux, Nicolas; Hermann, Robert; Johne, Andreas (2012). “Effects of ketoconazole treatment on the pharmacokinetics of safinamide and its plasma metabolites in healthy adult subjects”. Biopharmaceutics & Drug Disposition. 33 (9): 550. doi:10.1002/bdd.1822. PMID 23097240.
  14. Jump up^ Pevarello, P; Bonsignori, A; Dostert, P; Heidempergher, F; Pinciroli, V; Colombo, M; McArthur, RA; Varasi, M (1998). “Synthesis and Anticonvulsant Activity of a New Class of 2-[(Arylalkyl)amino]alkanamide Derivatives”. Journal of Medicinal Chemistry. 41 (4): 579–590. doi:10.1021/jm970599m. PMID 9484507.
  15. Jump up^ “Wichtigste Ergebnisse der Langzeitstudie mit Safinamid als Begleittherapie zu Levodopa bei Parkinson im fortgeschrittenen Stadium” [Major results from the long-term study of safinamide as add-on to levodopa for late-stage Parkinson] (in German). Merck KGaA. 4 November 2010.
  16. Jump up^ Study of Safinamide in Early Parkinson’s Disease as Add-on to Dopamine Agonist (MOTION)
  17. Jump up^ Merck Returns Rights for Safinamide to Newron, 21 October 2011.
  18. Jump up^ “Information about FDA Refusal to File” (PDF). Newron. 29 July 2014.
  19.  “Information about FDA re-application” (PDF). Newron. 29 December 2014.
  20.  Chazot, PL (2007). “Drug evaluation: Safinamide for the treatment of Parkinson’s disease, epilepsy and restless legs syndrome”. Current Opinion in Investigational Drugs. 8 (7): 570–579. PMID 17659477.
Safinamide
Safinamide.svg
Clinical data
Trade names Xadago
AHFS/Drugs.com UK Drug Information
Pregnancy
category
  • Fetal malformations in animal studies[1]
Routes of
administration
Oral
ATC code
Legal status
Legal status
  • UK:POM (Prescription only)
Pharmacokinetic data
Bioavailability 95%
Protein binding 88–90%
Metabolism Amidases, glucuronidation
Biological half-life 20–30 hrs
Excretion 76% renal, 1.5% faeces
Identifiers
Synonyms EMD-1195686, PNU-15774E;
(2S)-2-[[4-[(3-fluorophenyl)methoxy]phenyl] methylamino]propanamide
CAS Number
PubChemCID
ChemSpider
UNII
KEGG
ChEMBL
ECHA InfoCard 100.120.167
Chemical and physical data
Formula C17H19FN2O2
Molar mass 302.34 g/mol
3D model (Jmol)

//////////Xadago, safinamide,  Newron Pharmaceuticals, FDA 2017, Parkinson’s disease, 133865-89-1 , сафинамид , سافيناميد沙非胺, EMD-1195686, ZP-034, FCE-28073(R-isomer), PNU-151774E, NW-1015, FCE-26743

C[C@H](NCC1=CC=C(OCC2=CC=CC(F)=C2)C=C1)C(N)=O

New paper on Trelagliptin succinate


Trelagliptin succinate, a novel once-weekly oral dipeptidyl peptidase-4 (DPP-4) inhibitor, was approved for the Japanese market on March 26, 2015

Trelagliptin exhibited a better potency against human DPP-4 than alogliptin and sitagliptin, along with its excellent selectivity and slow-binding properties that may partially contribute to its sustained efficacy. In phase III clinical studies, once-weekly oral trelagliptin provided long-term safety and efficacy in both monotherapy and combination with other antidiabetic medicines and was proved to be noninferior to its analogue alogliptin used once daily.

2-({6-[(3R)-3-Aminopiperidin-1-yl]-3-methyl-2,4-dioxo-3,4-dihydropyrimidin-1(2H)-yl}methyl)-4-fluorobenzonitrile Monosuccinate (1)

white solid
Mp: 186–188 °C (187.1 °C in literature(x)),
[α]D20 = 16.4 (c 1, DMSO, 16.7 in literature(x)),
1H NMR (400 MHz, CD3OD) δ (ppm):7.79–7.82 (m, 1H), 7.15–7.25 (m, 2H), 5.46 (s, 1H), 5.20–5.32 (m, 2H), 3.35–3.37 (m, 2H), 3.22 (s, 3H), 3.03–3.06 (m, 1H), 2.74–2.81 (m, 2H), 2.49 (s, 4H), 2.07–2.11 (m, 1H), 1.82–1.89 (m, 1H), 1.65–1.76 (m, 1H), 1.55–1.59 (m, 1H).
MS (ESI+): m/z, 358.24 ([M + H]+).
Anal. (C22H26FN5O6) calcd: C, 55.57; H, 5.51; N, 14.73; found: C, 55.32; H, 5.46; N, 14.62.

Synthesis of Trelagliptin Succinate

State Key Lab of New Drug & Pharmaceutical Process, Shanghai Key Lab of Anti-Infectives, Shanghai Institute of Pharmaceutical Industry, State Institute of Pharmaceutical Industry, Shanghai 201203, China
Abstract Image

An improved process for the synthesis of antidiabetic drug trelagliptin succinate through unprotected (R)-3-aminopiperidine was described. The impurity profile with different conditions of the key substitution was illustrated, and then the best reaction condition was identified. The optimizations also included the bromination of 4-fluoro-2-methylbenzonitrile so that the process became efficient and concise.

  • 1.
    Zhang, Z.; Wallace, M. B.; Feng, J.; Stafford, J. A.; Skene, R. J.; Shi, L.; Lee, B.; Aertgeerts, K.; Jennings, A.; Xu, R.; Kassel, D. B.; Kaldor, S. W.; Navre, M.; Webb, D. R.; Gwaltney, S. L.J. Med. Chem. 2011, 54, 510524, DOI: 10.1021/jm101016w
  • 2.
    Feng, J.; Gwaltney, S. L.; Dipeptidyl Peptidase Inhibitors. PCT Int. Appl. WO 2005095381, October 13, 2005.
  • 3.

    Grimshaw, C. E.; Jennings, A.; Kamran, R.; Ueno, H.; Nishigaki, N.; Kosaka, T.; Tani, A.; Sano, H.; Kinugawa, Y.; Koumura, E.; Shi, L.; Takeuchi, K. PLoS One 2016, 11, e0157509, DOI: 10.1371/journal.pone.0157509

4.

Once-weekly trelagliptin versus daily alogliptin in Japanese patients with type 2 diabetes: a randomised, double-blind, phase 3, non-inferiority study
Inagaki, Nobuya; Onouchi, Hitoshi; Maezawa, Hideaki; Kuroda, Shingo; Kaku, Kohei
Lancet Diabetes & Endocrinology (2015), 3 (3), 191-197 CODEN: LDEAAS; ISSN:2213-8587. (Elsevier Ltd.)
Trelagliptin is a novel once-weekly oral DPP-4 inhibitor. We assessed the efficacy and safety of trelagliptin vs. the daily oral DPP-4 inhibitor alogliptin in Japanese patients with type 2 diabetes. We did a randomised, double-blind, active-controlled, parallel-group, phase 3, non-inferiority study at 26 sites in Japan.
We included individuals with type 2 diabetes inadequately controlled by diet and exercise. We randomly assigned patients (2:2:1) to receive trelagliptin (100 mg) once per wk, alogliptin (25 mg) once per day, or placebo for 24 wk. Randomisation was done electronically and independently from the study with permuted blocks of ten patients. Patients and clinicians were masked to group assignment.
Patients in the trelagliptin group were given trelagliptin once a week and oral alogliptin placebo every day, whereas patients in the alogliptin group were given oral trelagliptin placebo once a week and oral alogliptin every day (double-dummy design). Patients in the placebo group were given an oral alogliptin placebo once a day and an oral trelagliptin placebo once a week. Our primary outcome was between-groups difference in change in HbA1c concn. from baseline to the end of treatment. The non-inferiority margin was 0·4%. Our anal. included all patients who were randomised and received at least one dose of study drug. The study is registered with ClinicalTrials.gov, no. NCT01632007.
Between May 26, 2012, and Nov 20, 2012, we enrolled 357 patients. 243 patients were included in the anal. (101 for trelagliptin, 92 for alogliptin, and 50 for placebo). In the primary anal., the least squares mean change in HbA1c concn. was -0·33% in the trelagliptin group (SE 0·059) and -0·45% in the alogliptin group (0·061) based on the ANCOVA model. The least squares mean difference (trelagliptin minus alogliptin) of change from baseline in HbA1c concn. was 0·11% (95% CI -0·054 to 0·281). Trelagliptin was non-inferior to alogliptin. Both active groups had significantly reduced mean HbA1c concns. at end of treatment compared with placebo (p<0·0001). The frequency of adverse events was similar between groups. No hypoglycemia was reported with trelagliptin and the drug was well tolerated.
The once-weekly DPP-4 inhibitor trelagliptin showed similar efficacy and safety to alogliptin once daily in Japanese patients with type 2 diabetes. Trelagliptin could be a useful new antidiabetes drug that needs to be given once a week.Takeda Pharmaceutical Company.
  • 4.

    Inagaki, N.; Onouchi, H.; Maezawa, H.; Kuroda, S.; Kaku, K. Lancet Diabetes Endocrinol.2015, 3, 191197, DOI: 10.1016/S2213-8587(14)70251-7

  • 5.

    Inagaki, N.; Sano, H.; Seki, Y.; Kuroda, S.; Kaku, K. J.Diabetes Investig. 2016, 7, 718726, DOI: 10.1111/jdi.12499

//////////

NEW DRUG APPROVALS BLOG HITS 16 LAKH VIEWS IN 213 COUNTRIES


STR0

NEW DRUG APPROVALS BLOG HITS 16 LAKH VIEWS IN 213 COUNTRIES

Novartis Kisqali® (ribociclib, LEE011) receives FDA approval as first-line treatment for HR+/HER2- metastatic breast cancer in combination with any aromatase inhibitor


Novartis logo: a global healthcare company

  • Approved based on a first-line Phase III trial that met its primary endpoint of progression-free survival (PFS) at interim analysis due to superior efficacy compared to letrozole alone[1]
  • At this interim analysis, Kisqali plus letrozole reduced risk of disease progression or death by 44% over letrozole alone, and demonstrated tumor burden reduction with a 53% overall response rate[1]
  • Kisqali plus letrozole showed treatment benefit across all patient subgroups regardless of disease burden or tumor location[1]
  • At a subsequent analysis with additional follow-up and progression events, a median PFS of 25.3 months for Kisqali plus letrozole and 16.0 months for letrozole alone was observed[2]

Basel, March 13, 2017 The US Food and Drug Administration (FDA) has approved Kisqali®(ribociclib, formerly known as LEE011) in combination with an aromatase inhibitor as initial endocrine-based therapy for treatment of postmenopausal women with hormone receptor positive, human epidermal growth factor receptor-2 negative (HR+/HER2-) advanced or metastatic breast cancer.

Kisqali is a CDK4/6 inhibitor approved based on a first-line Phase III trial that met its primary endpoint early, demonstrating statistically significant improvement in progression-free survival (PFS) compared to letrozole alone at the first pre-planned interim analysis[1]. Kisqali was reviewed and approved under the FDA Breakthrough Therapy designation and Priority Review programs.

“Kisqali is emblematic of the innovation that Novartis continues to bring forward for people with HR+/HER2- metastatic breast cancer,” said Bruno Strigini, CEO, Novartis Oncology. “We at Novartis are proud of the comprehensive clinical program for Kisqali that has led to today’s approval and the new hope this medicine represents for patients and their families.”

The FDA approval is based on the superior efficacy and demonstrated safety of Kisqali plus letrozole versus letrozole alone in the pivotal Phase III MONALEESA-2 trial. The trial, which enrolled 668 postmenopausal women with HR+/HER2- advanced or metastatic breast cancer who received no prior systemic therapy for their advanced breast cancer, showed that Kisqali plus an aromatase inhibitor, letrozole, reduced the risk of progression or death by 44 percent over letrozole alone (median PFS not reached (95% CI: 19.3 months-not reached) vs. 14.7 months (95% CI: 13.0-16.5 months); HR=0.556 (95% CI: 0.429-0.720); p<0.0001)[1].

More than half of patients taking Kisqali plus letrozole remained alive and progression free at the time of interim analysis, therefore median PFS could not be determined[1]. At a subsequent analysis with additional 11-month follow-up and progression events, a median PFS of 25.3 months for Kisqali plus letrozole and 16.0 months for letrozole alone was observed[2]. Overall survival data is not yet mature and will be available at a later date.

“In the MONALEESA-2 trial, ribociclib plus letrozole reduced the risk of disease progression or death by 44 percent over letrozole alone, and more than half of patients (53%) with measurable disease taking ribociclib plus letrozole experienced a tumor burden reduction of at least 30 percent. This is a significant result for women with this serious form of breast cancer,” said Gabriel N. Hortobagyi, MD, Professor of Medicine, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center and MONALEESA-2 Principal Investigator. “These results affirm that combination therapy with a CDK4/6 inhibitor like ribociclib and an aromatase inhibitor should be a new standard of care for initial treatment of HR+ advanced breast cancer.”

Kisqali is taken with or without food as a once-daily oral dose of 600 mg (three 200 mg tablets) for three weeks, followed by one week off treatment. Kisqali is taken in combination with four weeks of any aromatase inhibitor[1].

Breast cancer is the second most common cancer in American women[3]. The American Cancer Society estimates more than 250,000 women will be diagnosed with invasive breast cancer in 2017[3]. Up to one-third of patients with early-stage breast cancer will subsequently develop metastatic disease[4].

Novartis is committed to providing patients with access to medicines, as well as resources and support to address a range of needs. The Kisqali patient support program is available to help guide eligible patients through the various aspects of getting started on treatment, from providing educational information to helping them understand their insurance coverage and identify potential financial assistance options. For more information, patients and healthcare professionals can call 1-800-282-7630.

The full prescribing information for Kisqali can be found at https://www.pharma.us.novartis.com/sites/www.pharma.us.novartis.com/files/kisqali.pdf(link is external).

About Kisqali® (ribociclib)
Kisqali (ribociclib) is a selective cyclin-dependent kinase inhibitor, a class of drugs that help slow the progression of cancer by inhibiting two proteins called cyclin-dependent kinase 4 and 6 (CDK4/6). These proteins, when over-activated, can enable cancer cells to grow and divide too quickly. Targeting CDK4/6 with enhanced precision may play a role in ensuring that cancer cells do not continue to replicate uncontrollably.

Kisqali was developed by the Novartis Institutes for BioMedical Research (NIBR) under a research collaboration with Astex Pharmaceuticals.

About the MONALEESA Clinical Trial Program
Novartis is continuing to assess Kisqali through the robust MONALEESA clinical trial program, which includes two additional Phase III trials, MONALEESA-3 and MONALEESA-7, that are evaluating Kisqali in multiple endocrine therapy combinations across a broad range of patients, including premenopausal women. MONALEESA-3 is evaluating Kisqali in combination with fulvestrant compared to fulvestrant alone in postmenopausal women with HR+/HER2- advanced breast cancer who have received no or a maximum of one prior endocrine therapy. MONALEESA-7 is investigating Kisqali in combination with endocrine therapy and goserelin compared to endocrine therapy and goserelin alone in premenopausal women with HR+/HER2- advanced breast cancer who have not previously received endocrine therapy.

About Novartis in Advanced Breast Cancer
For more than 25 years, Novartis has been at the forefront of driving scientific advancements for breast cancer patients and improving clinical practice in collaboration with the global community. With one of the most diverse breast cancer pipelines and the largest number of breast cancer compounds in development, Novartis leads the industry in discovery of new therapies and combinations, especially in HR+ advanced breast cancer, the most common form of the disease.

Kisqali® (ribociclib) Important Safety Information
Kisqali® (ribociclib) can cause a heart problem known as QT prolongation. This condition can cause an abnormal heartbeat and may lead to death. Patients should tell their healthcare provider right away if they have a change in their heartbeat (a fast or irregular heartbeat), or if they feel dizzy or faint. Kisqali can cause serious liver problems. Patients should tell their healthcare provider right away if they get any of the following signs and symptoms of liver problems: yellowing of the skin or the whites of the eyes (jaundice), dark or brown (tea-colored) urine, feeling very tired, loss of appetite, pain on the upper right side of the stomach area (abdomen), and bleeding or bruising more easily than normal. Low white blood cell counts are very common when taking Kisqali and may result in infections that may be severe. Patients should tell their healthcare provider right away if they have signs and symptoms of low white blood cell counts or infections such as fever and chills. Before taking Kisqali, patients should tell their healthcare provider if they are pregnant, or plan to become pregnant as Kisqali can harm an unborn baby. Females who are able to become pregnant and who take Kisqali should use effective birth control during treatment and for at least 3 weeks after the last dose of Kisqali. Do not breastfeed during treatment with Kisqali and for at least 3 weeks after the last dose of Kisqali. Patients should tell their healthcare provider about all of the medicines they take, including prescription and over-the-counter medicines, vitamins, and herbal supplements since they may interact with Kisqali. Patients should avoid pomegranate or pomegranate juice, and grapefruit or grapefruit juice while taking Kisqali. The most common side effects (incidence >=20%) of Kisqali when used with letrozole include white blood cell count decreases, nausea, tiredness, diarrhea, hair thinning or hair loss, vomiting, constipation, headache, and back pain. The most common grade 3/4 side effects in the Kisqali + letrozole arm (incidence >2%) were low neutrophils, low leukocytes, abnormal liver function tests, low lymphocytes, and vomiting. Abnormalities were observed in hematology and clinical chemistry laboratory tests.

Please see the Full Prescribing Information for Kisqali, available at https://www.pharma.us.novartis.com/sites/www.pharma.us.novartis.com/files/kisqali.pdf(link is external).

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, cost-saving generic and biosimilar pharmaceuticals and eye care. Novartis has leading positions globally in each of these areas. In 2016, the Group achieved net sales of USD 48.5 billion, while R&D throughout the Group amounted to approximately USD 9.0 billion. Novartis Group companies employ approximately 118,000 full-time-equivalent associates. Novartis products are sold in approximately 155 countries around the world. For more information, please visit http://www.novartis.com.

Novartis is on Twitter. Sign up to follow @Novartis and @NovartisCancer at http://twitter.com/novartis(link is external) and http://twitter.com/novartiscancer (link is external)
For Novartis multimedia content, please visit www.novartis.com/news/media-library
For questions about the site or required registration, please contact media.relations@novartis.com

References
[1] Kisqali (ribociclib) Prescribing information. East Hanover, New Jersey, USA: Novartis Pharmaceuticals Corporation; March 2016.
[2] Novartis Data on File
[3] American Cancer Society. How Common Is Breast Cancer? Available at https://www.cancer.org/cancer/breast-cancer/about/how-common-is-breast-cancer.html(link is external). Accessed January 23, 2017.
[4] O’Shaughnessy J. Extending survival with chemotherapy in metastatic breast cancer. The Oncologist. 2005;10(Suppl 3):20-29.

Ribociclib skeletal.svg

рибоциклиб ريبوسيكليب 瑞波西利

Ribociclib « New Drug Approvals

////////////////Novartis,  Kisqali®, ribociclib, LEE011,  FDA 2017, HR+/HER2- metastatic breast cancer, рибоциклиб ريبوسيكليب 瑞波西利

GDC 0994, Ravoxertinib


GDC-0994.png

GDC 0994

GDC-0994; Ravoxertinib; 1453848-26-4; GDC0994; UNII-R6AXV96CRH; R6AXV96CRH, RG7842; RG-7842; RG 7842

CAS 1453848-26-4

1-[(1S)-1-(4-chloro-3-fluorophenyl)-2-hydroxyethyl]-4-[2-[(2-methylpyrazol-3-yl)amino]pyrimidin-4-yl]pyridin-2-one

Molecular Formula: C21H18ClFN6O2
Molecular Weight: 440.863 g/mol

PHASE 1

Ravoxertinib also known as GDC-0994 and RG7842, is an orally available inhibitor of extracellular signal-regulated kinase (ERK), with potential antineoplastic activity. Upon oral administration, GDC-0994 inhibits both ERK phosphorylation and activation of ERK-mediated signal transduction pathways. This prevents ERK-dependent tumor cell proliferation and survival. The mitogen-activated protein kinase (MAPK)/ERK pathway is upregulated in a variety of tumor cell types and plays a key role in tumor cell proliferation, differentiation and survival.

GDC-0994 is an ERK inhibitor invented by Array under a collaboration agreement with Genentech. Array has received certain clinical milestones and is entitled to additional potential clinical and commercial milestones and royalties on product sales under the agreement. ERK is a key protein kinase in the RAS/RAF/MEK/ERK pathway, which regulates several key cellular activities including proliferation, differentiation, migration, survival and angiogenesis. Inappropriate activation of this pathway has been shown to occur in many cancers. GDC-0994 is currently advancing in a Phase 1 trial in patients with solid tumors.

Image result for ARRAY BIOPHARMA INC.

Image result for Genentech

Applicants: ARRAY BIOPHARMA INC. [US/US]; 3200 Walnut Street Boulder, Colorado 80301 (US).
GENENTECH, INC. [US/US]; 1 DNA Way South San Francisco, California 94080-4990 (US)
Inventors: BLAKE, James F.; (US).
CHICARELLI, Mark Joseph; (US).
GARREY, Rustam Ferdinand; (US).
GAUDINO, John; (US).
GRINA, Jonas; (US).
MORENO, David A.; (US).
MOHR, Peter J.; (US).
REN, Li; (US).
SCHWARZ, Jacob; (US).
CHEN, Huifen; (US).
ROBARGE, Kirk; (US).
ZHOU, Aihe; (US)

WO2013130976

  • OriginatorArray BioPharma
  • DeveloperGenentech
  • ClassAntineoplastics; Small molecules
  • Mechanism of ActionExtracellular signal-regulated MAP kinase inhibitors; Mitogen activated protein kinase 3 inhibitors; Mitogen-activated protein kinase 1 inhibitors
  • Phase ISolid tumours

Most Recent Events

  • 29 Nov 2016Pharmacodynamics data from a preclinical trial in Solid tumours presented at the 28th EORTC-NCI-AACR Symposium on Molecular Targets and Cancer Therapeutics (EORTC-NCI-AACR-2016)
  • 29 Nov 2016Adverse events, efficacy, pharmacokinetics and pharmacodynamics data from a phase I trial in Solid tumours presented at the 28th EORTC-NCI-AACR Symposium on Molecular Targets and Cancer Therapeutics
  • 16 Jul 2016No recent reports of development identified for phase-I development in Solid-tumours(Late-stage disease, Monotherapy, Second-line therapy or greater) in USA

FREE FORM

Abstract Image

(S)-1-(1-(4-Chloro-3-fluorophenyl)-2-hydroxyethyl)-4-(2-((1-methyl-1H-pyrazol-5-yl)amino)pyrimidin-4-yl)pyridin-2(1H)-one,

mp = 304.4 °C,
[α]D23 +113.8 (c 0.29, MeOH);
IR 1660, 1582, 1557 cm–1;
1H NMR (500 MHz, DMSO-d6) δ 9.69–9.52 (s, 1H), 8.69–8.49 (d, J = 5.1 Hz, 1H), 8.09–7.77 (d, J = 7.3 Hz, 1H), 7.61–7.56 (t, J = 8.1 Hz, 1H), 7.50–7.47 (d, J = 5.1 Hz, 1H), 7.46–7.42 (dd, J = 10.6, 2.0 Hz, 1H), 7.38–7.36 (d, J = 1.9 Hz, 1H), 7.22–7.08 (m, 2H), 6.98–6.79 (dd, J = 7.3, 2.1 Hz, 1H), 6.35–6.23 (d, J = 1.9 Hz, 1H), 6.06–5.90 (dd, J = 8.1, 5.4 Hz, 1H), 5.40–5.23 (d, J = 5.2 Hz, 1H), 4.24–3.97 (m, 2H), 3.80–3.56 (s, 3H);
13C NMR (101 MHz, DMSO-d6) δ 162.18, 161.53, 160.88, 160.55, 157.59 (d, JCF = 246.83 Hz), 147.19, 140.09 (d, JCF = 6.47 Hz), 138.30, 137.75, 137.42, 131.20, 125.53 (d, JCF = 3.45 Hz), 119.29 (d, JCF = 17.45 Hz), 117.74, 116.61 (d, JCF = 21.68 Hz), 109.68, 103.34, 99.36, 61.24, 59.20, 35.93.
HRMS (ESI): m/z [M + H]+ calcd for C21H18ClFN6O2, 441.1242; found, 441.1230.

GDC-0994 benzenesulfonate salt

figure

CAS 1817728-45-2, C21 H18 Cl F N6 O2 . C6 H6 O3 S

GDC-0994 as a light yellow solid,

mp 197.7 °C;

1H NMR (600 MHz, DMSO-d6): 9.93, (s, 1H), 8.65 (d, J = 5.2 Hz, 1H), 7.95 (d, J = 7.27 Hz, 1H), 7.63 (m, 2H), 7.62 (d, J = 1.5 Hz, 1H), 7.58 (t, J = 8.2 Hz, 1H), 7.55 (d, J = 5.2 Hz, 1H), 7.44 (dd, J = 10.6, 1.9 Hz, 1H), 7.33 (m, 3H), 7.18 (d, J = 2.0 Hz, 1H), 7.17 (d, J = 2.1 Hz, 1H), 6.90 (dd, J = 7.3, 2.1 Hz, 1H), 6.48 (d, J = 2.2 Hz, 1H), 5.99 (dd, J = 8.1, 5.5 Hz, 1H), 4.17 (dd, J = 11.9, 8.2 Hz, 1H), 4.05 (dd, J = 11.9, 5.5 Hz, 1H), 3.78 (s, 3H).

13C NMR (150 MHz, DMSO-d6): 161.60, 161.14, 160.02, 159.79, 157.02 (d, J = 245 Hz), 148.0, 146.49, 139.53 (d, J = 6.0 Hz), 139.04, 136.96, 136.39, 130.66, 128.42, 127.59, 125.38, 124.99 (d, J = 3.0 Hz), 118.72 (d, J = 18.0 Hz), 117.29, 116.05 (d, J = 22.5 Hz), 109.75, 102.79, 98.77, 60.64, 58.68, 35.29.

19F NMR (282 MHz, DMSO-d6) −115.86 (dd, J = 10.6, 7.8).

HRMS calcd for C21H18ClFN6O2 [M + H] 441.1242, found 441.1245.

PATENT

WO 2013130976

Example 39

(S)-1-(1-(4-chloro-3-fluorophenyl)-2-hydroxyethyl)-4-(2-((1-methyl-1H-pyrazol-5- yl)amino)pyrimidin-4-yl)pyridin-2(1H)-one

[00398] Step A: (S)-1-(2-(tert-Butyldimethylsiloxy)-1-(4-chloro-3-fluorophenyl)ethyl)-4-(2-(methylsulfonyl)pyrimidin-4-yl)pyridine-2(1H)-one (47 mg, 0.087 mmol), 2-methyl pyrazole-3 -amine (0.175 mmol, 2.0 equivalents) and anhydrous DMF (3.0 mL) were added to a 25 mL round bottomed flask equipped with a stirring bar. The flask was capped with a rubber septum and flushed with nitrogen. Under a blanket of nitrogen, sodium hydride (8.5 mg, 60% dispersion in mineral oil) was added in one portion. The flask was flushed with

nitrogen, capped and stirred at room temperature. The reaction progress was monitored by LCMS, and after 30 minutes, the starting material was consumed. The reaction mixture was quenched by the addition of water (0.5 mL) and ethyl acetate (15 mL). The contents of the round bottomed flask were transferred to a 125 mL separatory funnel, and the reaction flask was rinsed several times with additional ethyl acetate. Crude (S)-1-(2-((tert-butyldimethylsilyl)oxy)-1-(4-chloro-3-fluorophenyl)ethyl)-4-(2-((1-methyl-1H-pyrazol-5-yl)amino)pyrimidin-4-yl)pyridin-2(1H)-one was partitioned between ethyl acetate and water (80 mL/30 mL). The ethyl acetate layer was washed once with brine, dried (MgSO4), filtered and concentrated to give crude (S)-1-(2-((tert-butyldimethylsilyl)oxy)-1-(4-chloro-3-fluorophenyl)ethyl)-4-(2-((1-methyl-1H-pyrazol-5-yl)amino)pyrimidin-4-yl)pyridin-2(1H)-one. The crude was taken directly into the deprotection step.

[00399] Step B: Crude (S)-1-(2-((tert-butyldimethylsilyl)oxy)-1-(4-chloro-3-fluorophenyl)ethyl)-4-(2-((1-methyl-1H-pyrazol-5-yl)amino)pyrimidin-4-yl)pyridin-2(1H)-one (48 mg) was dissolved in ethyl acetate (4 mL) and treated dropwise slowly (over 2 minutes) with an ethyl acetate solution (1.0 mL, which had been saturated with HCl gas). The reaction stirred at room temperature for 15 minutes, after which time LCMS indicated complete consumption of the starting material. The reaction mixture was concentrated to an oily residue and purified by prep RP HPLC to yield (S)-1-(1-(4-chloro-3-fluorophenyl)-2-hydroxyethyl)-4-(2-((1-methyl-1H-pyrazol-5-yl)amino)pyrimidin-4-yl)pyridin-2(1H)-one (20.8 mg, 54.6% yield) as a lyophilized powder. 1H NMR (400 MHz, (CD3)2SO) δ 9.58 (s, 1H), 8.60 (d, J = 5.1 Hz, 1H), 7.91 (t, J = 9.0 Hz, 1H),7.58 (t, J = 8.1 Hz, 1H), 7.52-7.41 (m, 2H), 7.37 (d, J = 1.8 Hz, 1H), 7.14 (dd, J = 10.7,5.1 Hz 2H), 6.86 (dd, J = 7.3, 1.8 Hz, 1H), 6.27(d, J = 1.7 Hz, 1H), 5.97 (dd, J = 7.7, 5.7 Hz, 1H), 5.31(t, J = 5.2 Hz, 1H), 4.15 (m, 1H), 4.10-3.95 (m,1H), 3.69 (s, 3H); LCMS m/z 441 (M+H)+.

PAPER

Discovery of (S)-1-(1-(4-Chloro-3-fluorophenyl)-2-hydroxyethyl)-4-(2-((1-methyl-1H-pyrazol-5-yl)amino)pyrimidin-4-yl)pyridin-2(1H)-one (GDC-0994), an Extracellular Signal-Regulated Kinase 1/2 (ERK1/2) Inhibitor in Early Clinical Development

Array BioPharma Inc., 3200 Walnut Street, Boulder, Colorado 80301, United States
Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
J. Med. Chem., 2016, 59 (12), pp 5650–5660
DOI: 10.1021/acs.jmedchem.6b00389
Abstract Image

The extracellular signal-regulated kinases ERK1/2 represent an essential node within the RAS/RAF/MEK/ERK signaling cascade that is commonly activated by oncogenic mutations in BRAF or RAS or by upstream oncogenic signaling. While targeting upstream nodes with RAF and MEK inhibitors has proven effective clinically, resistance frequently develops through reactivation of the pathway. Simultaneous targeting of multiple nodes in the pathway, such as MEK and ERK, offers the prospect of enhanced efficacy as well as reduced potential for acquired resistance. Described herein is the discovery and characterization of GDC-0994 (22), an orally bioavailable small molecule inhibitor selective for ERK kinase activity.

PATENT

WO 2015154674

https://www.google.com/patents/WO2015154674A1?cl=pt

The present invention provides processes for the manufacture of I which is a useful intermediate that can be used in the manufacture VIII. (WO2013/130976) Compound VIII is an ERK inhibitor and a useful medicament for treating hyperproliferative disorders. The process provides an efficient route to VIII and to the useful intermediates VI and VII. Alkylation of VII with VI affords I, which ultimately is condensed with 1-methyl-1H-pyrazol-5-amine (XIV) . (SCHEME A)
(i) i-PrMgCl, LiCl, THF; (ii) HCO2Na, HCO2H, H2O, EtOH; (iii) GDH-105, morpholineethanesulfonic acid, MgCl2, PEG6000, heptane, 1wt% KRED-NADH-112, NAD, glucose (iv) TBSCl, DMAP, TEA, DCM, 20-25℃ , 15h; (v) MsCl, DCM, 20-25℃ , 3h
Scheme 1. Original Synthetic Process
Scheme 2. Improved Process To I

[0170]
Example 1

[0173]
2- ( (tert-Butyldimethylsilyl) oxy) -1- (4-chloro-3-fluorophenyl) ethyl methanesulfonate

[0174]

[0175]
Step 1: 4-bromo-1-chloro-2-fluorobenzene (64 kg) and dry toluene (170kg) were charged to the 2000 L steel reaction vessel under nitrogen. The reactor was evacuated and backfilled with N2 for three times, and cooled to between-10 and 5 ℃ under nitrogen atmosphere. To the solution was added dropwise i-PrMgCl. LiCl (280kg, 1.3M in THF) at between-10 and 10 ℃ . The reaction was stirred for a further 15 to 30min at between-10 and 10 ℃ and then warmed to about 20 to 25 ℃ over 1h. The reaction mixture was stirred for another 6 h stir to complete the exchange. The resulting solution was cooled to between-50 and-40 ℃ . A solution of 2-chloro-N-methoxy-N-methylacetamide (44.5kg) in dry toluene (289kg) was added dropwise to the above solution at while maintaining the temperature between-50 and-30 ℃ . The reaction mixture was warmed to between 20 and 25 ℃ over 1h and then stirred for 3h to complete the reaction. The reaction was quenched by addition of 1N aq. HCl (808l g) at a temperature between-5 and 15 ℃ . The aqueous layer was separated and organic layer was filtered through a pad of diatomaceous earth. The organic layer was washed with 10%aq. NaCl solution (320kg) twice, then concentrated to about 300L to obtain 1- (4-chloro-3-fluorophenyl) -2-chloroethanone (51.8kg, 81.9%yield) as product in toluene.

[0176]
Step 2: The solution of II (51.7kg) in toluene was concentrated and solvent exchanged to EtOH to afford a suspension of II in EtOH (326kg) . A solution of HCOONa·2H2O (54.8kg) and HCOOH (44.5kg) in water (414kg) was added at a temperature between 15 and 35 ℃ under a nitrogen atmosphere. The resulting mixture was heated to reflux and stirred for 4 to 5 h. The solution was cooled to between 20 and 30 ℃ after over 95%conversion occurred. Water (450kg) was added dropwise at between 10 and 30℃ for over 2 h. The resulting suspension was cooled to between-10 and-3 ℃ and the cooled solution stirred for 1 to 2 h. The solid was filtered and the filter cake washed with water (400 kg) to remove the residual HCOONa and HCOOH. The 1- (4-chloro-3-fluorophenyl) -2-hydroxyethanone obtained was suspended in EtOAc (41kg) and n-heptane (64kg) , then warmed to between 45 and 50 ℃ , stirred for 2h, then cooled to between-2 and 5 ℃ for over 2h and stirred at this temperature for 2h. The solids were filtered and dried in vacuo at between 40 and 50 ℃ for 12 h to afford the product as white solid (40.0kg, 99.3%purity, 84.5%yield) .

[0177]
Step 3: A 500 L reactor under nitrogen was charged with purified water (150 kg) , 4-morpholineethanesulfonic acid (0.90kg) , anhydrous MgCl2(0.030kg) , n-heptane (37kg) , 1- (4-chloro-3-fluorophenyl) -2-hydroxyethanone (30kg) , D- (+) -glucose monohydrate (34.8kg) and PEG 6000 (30.0kg) . The pH of the solution was adjusted to between 6.5 and 7.0 with 1N aq. NaOH at between 28 and 32 ℃ . The cofactor recycling enzyme, glucose dehydrogenase GDH-105 (0.300kg) (Codexis Inc., Redwood City, CA, USA) , the cofactor nicotinamide adenine dinucleotide NAD (0.300kg) (Roche) and the oxidoreductase KRED-NADH-112 (0.300kg) (Codexis Inc., Redwood City, CA, USA) were added. The resulting suspension was stirred at between 29 and 31 ℃ for 10 to 12 h while adjusting the pH to maintain the reaction mixture pH between 6.5 and 7.0 by addition of 1N aq. NaOH (160kg) . The pH of the reaction mixture was adjusted to between 1 and 2 by addition of 49%H2SO4 (20kg) to quench the reaction. EtOAc (271kg) was added and the mixture was stirred at between 20 and 30 ℃for 10-15min then filtered through a pad of diatomaceous earth. The filter cake was washed with EtOAc (122kg) . The combined organic layers were separated and aqueous layer was extracted with EtOAc (150kg) . Water (237kg) was added to the combined organic layers. The pH of the mixture was adjusted to between 7.0 and 8.0 by addition of solid NaHCO3. The organic layer was separated, concentrated and then diluted with DCM to afford (R) -1- (4-chloro-3-fluorophenyl) ethane-1, 2-diol (30.9kg, yield 100%) as product in DCM.

[0178]
Step 4: A 1000 L reactor under nitrogen was charged with (R) -1- (4-chloro-3-fluorophenyl) ethane-1, 2-diol (29.5kg) and dry DCM (390kg) . The solution was cooled to between-5 and 0 ℃ . tert-Butylchlorodimethylsilane (25.1 kg) was added in portions while maintaining the temperature between-5 and 2 ℃ . A solution of DMAP (0.95kg) and TEA (41.0kg) in dry DCM (122kg ) was added dropwise to above solution at between-5 and 2 ℃ . The reaction solution was stirred for 1 h, then warmed to between 20 and 25 ℃ and stirred for 16 h. The solution of (R) -2- ( (tert-butyldimethylsilyl) oxy) -1- (4-chloro-3-fluorophenyl) ethanol was recooled to between-10 and-5 ℃ . A solution of methanesulfonyl chloride (19.55 kg) in dry DCM (122kg) was added dropwise to the above solution of while maintaining the temperature between-10 and 0 ℃ . The reaction solution was stirred at between-10 and 0 ℃ for 20 to 30 min, and then warmed to between 0 and 5℃ for over 1h, and stirred. The reaction solution was washed with water (210kg) , followed by 5%aq. citric acid (210kg) , 2%aq. NaHCO3 (210kg) and finally water (2 x 210kg) . The resulting DCM solution was dried (Na2SO4) , filtered and concentrated in vacuo below 15℃ (jacket temperature below 35℃) to afford (R) -2- ( (tert-butyldimethylsilyl) oxy) -1- (4-chloro-3-fluorophenyl) ethyl methanesulfonate (49.5kg, 83.5%yield, KarlFischer=0.01%) as product in DCM.

[0179]
Example 2

[0180]
4- (2- (methylsulfonyl) pyrimidin-4-yl) pyridin-2 (1H) -one

[0181]

[0182]
Step 1: A 1000 L reactor was charged with 2-fluoro-4-iodopyridine (82.2kg) and dry THF (205 kg) . The reactor was evacuated and backfilled with N2three times then cooled to between-30 and-20 ℃ . To the solution was added dropwise i-PrMgCl·LiCl (319 kg, 1.3M in THF) . The reaction was warmed to between-20 and-10℃ and stirred for 1.5 h to complete the transmetallation.

[0183]
A 2000 L reactor was charged with 4-chloro-2-methylthiopyrimidine (45.6kg) , dry THF (205kg) and [1, 3-bis (2, 6-diisopropylphenyl) imidazol-2-ylidene] (3-chloropyridyl) palladium (II) dichloride (PEPPSITM-IPr, 1.850kg) . The 2000 L reactor was evacuated and backfilled with N2 three times and heated to between 55 and 57 ℃ . To the reactor was added over 0.5 to 1 h, the solution of (2-fluoropyridin-4-yl) magnesium chloride while maintaining the temperature between 50 and 62℃ . The resulting reaction mixture was stirred at between 50 and 62 ℃ for a further 2h. The reaction mixture was cooled to between 5 and 25℃ while the reaction was quenched with water (273kg) . The pH of the mixture was adjusted to 8 to 9 by adding solid citric acid monohydrate (7.3kg) . The organic layer was separated, washed with 12.5%aqNaCl (228kg) and concentrated in vacuo below 50℃ to afford 4- (2-fluoropyridin-4-yl) -2- (methylthio) pyrimidine (38.3kg, 61%yield) as product in THF.

[0184]
Step 2: The solution of 4- (2-fluoropyridin-4-yl) -2- (methylthio) pyrimidine (38.2kg) in THF was concentrated and co-evaporated with THF to remove residual water. The suspension was filtered through a pad of diatomaceous earth to remove inorganic salts. To the resulting solution in THF (510kg) was added tert-BuOK (39.7kg) in portions while maintaining the temperature between 15 and 25 ℃ . The mixture was warmed to between 20 and 25 ℃ and stirred for 5h. NaHCO3 (14.9kg) added charged and then a citric acid solution (5kg) in THF (15kg) was added to adjust the pH to between 8 and 9. Water (230kg) was added. The mixture was filtered and the filter cake was washed with THF (100kg) . The combined THF solutions were washed with 12.5%aqueous NaCl (320kg) and concentrated to about 380L to afford a solution of 4- (2- (tert-butoxy) pyridin-4-yl) -2- (methylthio) pyrimidine in THF.

[0185]
To the THF solution cooled to between 15 and 30 ℃ was added1N H2SO4 aq. solution (311kg) . The mixture was stirred at this temperature for 4h. MTBE (280kg) was charged and the pH of reaction solution was adjusted to 14 with 30%aqueous NaOH (120kg) . The aqueous layer was separated and the organic phase filtered to remove inorganic salts. The obtained aqueous layer was washed with MTBE (2 x 280kg) . 2-MeTHF (1630kg) and i-PrOH (180kg) were added to the aqueous solution. The pH was then adjusted to 8 slowly with conc. HCl (19kg) . An organic layer separated and aqueous layer was extracted with 2-MeTHF (305kg) . The combined 2-MeTHF extracts were washed with water (300kg) and concentrated to about 100L. MTBE (230kg) was added and stirred at 20-30 ℃ for 0.5h. The solid was filtered and slurried in a mixture solvent of 2-MeTHF (68kg) and MTBE (230kg) . The suspension was stirred at 35-50 ℃ for 3h, and then cooled to 0 to 10 ℃ and stirred at a further 2h.

[0186]
The solid was filtered and dried in vacuo at between 50 and 62 ℃ for 20 h to afford product 4- (2- (methylthio) pyrimidin-4-yl) pyridin-2 (1H) -one as brown solid (33.55kg, 89.6%assay, 79.4%yield) .

[0187]
Example 3

[0188]
(S) -1- (2- ( (tert-Butyldimethylsilyl) oxy) -1- (4-chloro-3-fluorophenyl) ethyl) -4- (2- (methylthio) pyrimidin-4-yl) pyridin-2 (1H) -one (XI)

[0189]

[0190]
Step 1: The THF was co-evaporated from the THF solution of 4- (2- (methylthio) pyrimidin-4-yl) pyridin-2 (1H) -one (25.5kg) to remove residual water. Dry bis- (2-methoxyethyl) ether (75kg) was added. A solution of KHMDS (131kg, 1M in THF) was added dropwise while maintaining the temperature between 25 and 40 ℃ . The mixture was heated to between 75 and 80℃ and stirred for 30 to 40 min. The resulting mixture was cooled to between 20 and 30℃ under nitrogen atmosphere. A solution of (R) -2- ( (tert-butyldimethylsilyl) oxy) -1- (4-chloro-3-fluorophenyl) ethyl methanesulfonate (47.6kg) in THF (50kg) was added over 30 to 60 min while maintaining the temperature between 20 and 40℃ . The reaction solution was warmed to between 80 and 85 ℃ and stirred for 7 h. The solution was cooled to between 5 and 15 ℃ and water (155 kg) was added. The pH of the solution was adjusted to 7.5 with 30%aqueous citric acid (30 kg) . EtOAc (460kg) was added and the mixture was stirred for 20 min. The organic layer was separated and washed with 12.5%aqueous NaCl (510kg) . The combined aqueous layers were extracted with EtOAc (115kg) . The ethyl acetate layers were concentrated to about 360L to afford (S) -1- (2- ( (tert-butyldimethylsilyl) oxy) -1- (4-chloro-3-fluorophenyl) ethyl) -4- (2- (methylthio) pyrimidin-4-yl) pyridin-2 (1H) -one (44.6kg, 75.7%yield) as product in EtOAc.

[0191]
Step 2: To a solution of (S) -1- (2- ( (tert-butyldimethylsilyl) oxy) -1- (4-chloro-3-fluorophenyl) ethyl) -4- (2- (methylthio) pyrimidin-4-yl) pyridin-2 (1H) -one (44.6kg) in EtOAc (401kg, 10vol) cooled to between 5 and 10 ℃ was added in portions MCPBA (58kg) . The reaction mixture was added to a solution of NaHCO3 (48.7kg) in water (304kg) at a temperature between10 and-20℃ . A solution of Na2S2O3 (15kg) in water (150 kg) was added dropwise to consume residual MCBPA. The organic layer was separated and aqueous layer was extracted with EtOAc (130kg) . The combined organic layers were washed with water (301 kg) , concentrated and solvent exchanged to DCM to afford (S) -1- (2- ( (tert-butyldimethylsilyl) oxy) -1- (4-chloro-3-fluorophenyl) ethyl) -4- (2- (methylsulfonyl) pyrimidin- 4-yl) pyridin-2 (1H) -one (45.0kg, 94.9%yield) as product in DCM. The DCM solution was concentrated to about 100 L, filtered through a pad of SiO2 (60kg) and eluted with an EtOAc/DCM gradient (0, 25 and 50%EtOAc) . The fractions were combined and concentrated to get the product which was re-slurried with (acetone: n-heptane=1: 3 v/v) four times to afford the final product (31.94kg, 71%yield) .

[0192]
Example 4

[0193]
(S) -1- (1- (4-Chloro-3-fluorophenyl) -2-hydroxyethyl) -4- (2- ( (1-methyl-1H-pyrazol-5-yl) amino) pyrimidin-4-yl) pyridin-2 (1H) -one, benzenesulfonate salt (VIIIb)

[0194]

[0195]
Step 1: A clean 100 L cylindrical reaction vessel was charged with THF (13 kg) then (S) -1- (2- ( (tert-butyldimethylsilyl) oxy) -1- (4-chloro-3-fluorophenyl) ethyl) -4- (2- (methylsulfonyl) pyrimidin-4-yl) pyridin-2 (1H) -one (I, 5 kg) and 1-methyl-1H-pyrazol-5-amine (1.1 kg) were added sequentially with medium agitation followed by THF (18 kg) . The mixture was cooled to-35 ℃ and to the resulting thin slurry was added slowly a THF solution of LiHMDS (17.4 kg, 1.0 M) at a rate that maintained the internal temperature below-25 ℃ . After the addition was completed, the reaction was held between-35 and-25 ℃ for 20 min and monitored by HPLC. If the HPLC result indicated ≤ 98.5%conversion, additional LiHMDS (0.34 kg, 1.0 M, 0.05 mol%) was added slowly at-35 ℃ . The reaction was quenched slowly at the same temperature with H3PO4 solution (4.4 kg of 85%H3PO4and 15 kg of water) and the internal temperature was kept below 30 ℃ . The reaction was diluted with EtOAc (18 kg) and the phases separated, the organic layer was washed with H3PO4 solution (1.1 kg of 85%H3PO4 and 12 kg of water) followed by a second H3PO4wash (0.55 kg of 85%H3PO4and 12 kg of water) . If 1-methyl-1H-pyrazol-5-remained, the organic layer was washed again with H3PO4 solution (0.55 kg of 85%H3PO4 and 12 kg of water) . Finally the organic layer was washed sequentially with water (20 kg) and a NaCl and NaHCO3 solution (2 kg of NaCl, 0.35 kg of NaHCO3and 10 kg of water) . After the phase separation, residue water in organic solution was removed through an azeotropic distillation with EtOAc to ≤ 0.5% (by KF) and then solution was concentrated to 20-30 L under a vacuum below 50 ℃ . The solvent was then swapped to MeOH using 35 kg of MeOH and then concentrated to between 20 and 30 L for the next step.

[0196]
Step 2: To the methanolic (S) -1- (2- ( (tert-butyldimethylsilyl) oxy) -1- (4-chloro-3-fluorophenyl) ethyl) -4- (2- ( (1-methyl-1H-pyrazol-5-yl) amino) pyrimidin-4-yl) pyridin-2 (1H) -one (IX) solution in MeOH was added HCl (10.7 kg, 1.25 M in MeOH) at RT. It was slightly exothermic. After the addition was completed, the reaction was heated to 45 ℃ . If the reaction was incomplete after 14 to 16 h, additional HCl (1 kg, 1.25 M in MeOH) was added and agitation at 45 ℃ was continued for 2 h. The reaction was equipped with a distillation setup with acid scrubber. The reaction was concentrated to between 20 and 30 L under a vacuum below 50 ℃ . To the resulting solution was added MeOH (35 kg) and the reaction was concentrated to 20 to 30 L again under a vacuum below 50 ℃ . The solvent was then switched to EtOAc using 40 kg of EtOAc. The solvent ratio was monitored by Headspace GC and the solvent swap continued until it was less than 1/5. The solution was concentrated to between 20 and 30 L under a vacuum below 50 ℃ . After the solution was cooled below 30 ℃ , aqueous NaHCO3 (1.2 kg of NaHCO3 and 20 kg of water) was added slowly with a medium agitation and followed by EtOAc (40 kg) . The organic layer was washed with water (2 x 10 kg) then concentrated to 20-30 L under a vacuum below 50 ℃ . The solvent was then switched to MEK using 35 kg of MEK. The residue MeOH was monitored by Headspace GC and the solvent swap continued until the MeOH was < 0.3%. The solution containing (S) -1- (1- (4-chloro-3-fluorophenyl) -2-hydroxyethyl) -4- (2- ( (1-methyl-1H-pyrazol-5-yl) amino) pyrimidin-4-yl) pyridin-2 (1H) -one (VIII) was concentrated to 20 to 30 L under a vacuum below 50 ℃ for the next step.

[0197]
Step 3: The solution of VIII in MEK was transferred to a second 100 L cylindrical reaction vessel through a 1μm line filter. In a separate container was prepared benezenesulfonic acid solution (1.3 kg of benzenesulfonic acid, 1.4 kg of water and 4.4 kg of MEK) . The filtered VIII solution was heated to 75 ℃ and to the resulting solution was added 0.7 kg of the benzenesulfonic acid solution through a 1μm line filter. The clear solution was seeded with crystalline benzenesulfonic acid salt of VIII (0.425 kg) as a slurry in MEK (0.025 kg of VIIIb crystalline seed and 0.4 kg of MEK) which produced a thin slurry. The remaining benzenesulfonic acid solution was then added through a 1μm line filter in 2 h. After addition, the slurry was heated at 75 ℃ for additional 1 h and then cooled to 18 ℃ in a minimum of 3 h. The resulting thick slurry was agitated at 20 ℃ for 14 to 16 h. The solid was filtered using an Aurora dryer. The mother liquor was assayed by HPLC (about . 3%loss) . The solid was then washed with 1μm line filtered 15.8 kg of MEK and water solution (0.8 kg of water and 15 kg of MEK) and followed by 1μm line filtered 30 kg of MEK. Washes were assayed by HPLC (<1%loss) . The wet cake was dried under a vacuum and a nitrogen sweep at a jacket temperature of 45 ℃ for a minimum 12 h to afford the benzenesulfonic acid salt of VIII, which is labeled VIIIb.

[0198]
Additional Examples

[0199]
Step 1:

[0200]

[0201]
To a clean 100 L cylindrical reaction vessel was charged 13 kg of THF first. With a medium agitation, 5.0 kg of I and 1.1 kg of 1-methyl-1H-pyrazol-5-amine was charged sequentially and followed by the rest of THF (18 kg) . At-35 ℃ to the resulting thin slurry was added 17.4 kg of LiHMDS (1.0 mol/L) in THF slowly and the internal temperature was remained below-25 ℃ . After addition, the reaction was held between-35 and-25 ℃ for 20 min. The reaction was monitored by HPLC. If the HPLC result indicated ≤ 98.5%conversion, additional 0.34 kg (0.05 mol%) of LiHMDS (1.0 mol/L) in THF was charged slowly at-35 ℃ . Otherwise, the reaction was quenched at the same temperature with 19.4 kg of H3PO4 solution (4.4 kg of 85%H3PO4and 15 kg of water) slowly and the internal temperature was remained below 30 ℃ . The reaction was diluted with 18 kg of EtOAc. After the phase separation, the organic layer was washed with 13.1 kg of H3PO4 solution (1.1 kg of 85%H3PO4 and 12 kg of water) and then with 12.6 kg of H3PO4solution (0.55 kg of 85%H3PO4 and 12 kg of water) . The organic layer was assayed for the 1-methyl-1H-pyrazol-5-amine level by HPLC. If the HPLC result indicated ≥ 20 μg/mL of 1-methyl-1H-pyrazol-5-amine, the organic layer needed an additional wash with 12.6 kg of H3PO4 solution (0.55 kg of 85%H3PO4 and 12 kg of water) . Otherwise, the organic layer was washed with 20 kg of water. The organic layer was assayed again for the 1-methyl-1H-pyrazol-5-amine level. If the HPLC result indicated ≥ 2 μg/mL of 1-methyl-1H-pyrazol-5-amine, the organic layer needed an additional wash with 20 kg of water. Otherwise, the organic layer was washed with 12.4 kg of NaCl and NaHCO3 solution (2 kg of NaCl, 0.35 kg of NaHCO3 and 10 kg of water) . After the phase separation, residue water in organic solution was removed through an azeotropic distillation with EtOAc to ≤ 0.5% (by KF) and then the solution was concentrated to 20 to 30 L under a vacuum below 50 ℃ . The solvent was then swapped to MeOH using 35 kg of MeOH and then concentrated to 20 to 30 L for the next step.

[0202]
Step 2:

[0203]

[0204]
To the IX solution in MeOH from the last step was charged 10.7 kg of HCl (1.25 M in MeOH) at the ambient temperature. It was observed slightly exothermic. After addition, the reaction was heated to 45 ℃ . After 14-16 h, the reaction was monitored by HPLC. If the HPLC result indicated the conversion was ≤ 98%, an additional 1 kg of HCl (1.25 M in MeOH) was charged and the reaction was agitated at 45 ℃ for additional 2 h. Otherwise, the reaction was equipped with a distillation setup with acid scrubber. The reaction was concentrated to 20 to 30 L under a vacuum below 50 ℃ . To the resulting solution was charged 35 kg of MeOH and the reaction was concentrated to 20 to 30 L again under a vacuum below 50 ℃ . The solvent was then switched to EtOAc using 40 kg of EtOAc. The solvent ratio was monitored by Headspace GC. If the ratio of MeOH/EtOAc was greater than 1/5, the solvent swap should be continued. Otherwise, the solution was concentrated to 20 to 30 L under a vacuum below 50 ℃ . After the solution was cooled below 30 ℃ , 21.2 kg of NaHCO3 solution (1.2 kg of NaHCO3 and 20 kg of water) was charged slowly with a medium agitation and followed by 40 kg of EtOAc. After the phase separation, the organic layer was washed with 2 X 10 kg of water. The organic layer was concentrated to 20 to 30 L under a vacuum below 50 ℃ . The solvent was then switched to MEK using 35 kg of MEK. The residue MeOH was monitored by Headspace GC. If the level of MeOH was ≥ 0.3%, the solvent swap should be continued. Otherwise, the solution was concentrated to 20 to 30 L under a vacuum below 50 ℃ for the next step.

[0205]
Step 3:

[0206]

[0207]
The VIII solution in MEK from the last step was transferred to a second 100 L cylindrical reaction vessel through a 3 μm line filter. In a separated container was prepared 7.1 kg of benzenesulfonic acid solution (1.3 kg of benzenesulfonic acid, 1.4 kg of water and 4.4 kg of MEK) . The filtered G02584994 solution was heated to 75 ℃ and to the resulting solution was charged 0.7 kg of benzenesulfonic acid solution (10%) through a 3 μm line filter. To the clear solution was charged 0.425 kg of VIIIb crystalline seed slurry in MEK (0.025 kg of VIIIb crystalline seed and 0.4 kg of MEK) . This resulted in a thin slurry. The rest of benzenesulfonic acid solution was then charged through a 3 μm line filter in 2 h. After addition, the slurry was heated at 75 ℃ for additional 1 h and then cooled to 20 ℃ in a minimum of 3 h. The resulting thick slurry was agitated at 20 ℃ for 14-16 h. Solid was filtered using a filter dryer. Mother liquor was assayed by HPLC (about 3%loss) . Solid was then washed with 3 μm line filtered 15.8 kg of MEK and water solution (0.8 kg of water and 15 kg of MEK) and followed by 3 μm line filtered 30 kg of MEK. Washes were assayed by HPLC (<1%loss) . The wet cake was dried under a vacuum and the nitrogen sweep at a jacket temperature of 45 ℃ for a minimum 12 h.

[0208]
Recrystallization

[0209]

[0210]
To a clean 100 L cylindrical reaction vessel was charged 16 kg of EtOH first. With a medium agitation, 3.5 kg of VIIIb was charged and then followed by the rest of EtOH (8.5 kg) . The thick slurry was heated to 78 ℃ and water (~1.1 kg) was charge until a clear solution was obtained. The hot solution was filtered through a 3 μm line filter to a second clean 100 L cylindrical reaction vessel. The temperature dropped to 55-60 ℃ and the solution remained clear. To the resulting solution was charged with 0.298 kg of VIIIb crystalline seed slurry in EtOH (0.018 kg of VIIIb crystalline seed and 0.28 kg of EtOH) . The thick slurry was concentrated to 20 to 30 L at 60 ℃ under a vacuum and then cooled 20 ℃ in 3 h. The resulting slurry was agitated at 20 ℃ for 14 to 16 h. Solid was filtered using a filter dryer. The mother liquor was assayed by HPLC (about 10%loss) . Solid was then washed with 3 μm line filtered 11.1 kg of EtOH and water solution (0.56 kg of water and 11 kg of EtOH) and followed by 3 μm line filtered 21 kg of MEK. Washes were assayed by HPLC (3%loss) . The wet cake was dried under a vacuum and the nitrogen sweep at a jacket temperature of 45 ℃ for a minimum 12 h.

[0211]
An additional synthetic process is set forth below.

[0212]
Step 1:

[0213]

[0214]
To a clean 100 L cylindrical reaction vessel was charged 18 kg of THF first. With a medium agitation, 4.2 kg of I and 0.91 kg of 1-methyl-1H-pyrazol-5-amine was charged sequentially and followed by the rest of THF (21 kg) . At-40 ℃ to the resulting thin slurry was added 14.9 kg of LiHMDS (1.0 mol/L) in THF slowly and the internal temperature was remained below-30 ℃ . After addition, the reaction was held between-35 and-40 ℃ for 20 min. The reaction was monitored by HPLC. The HPLC result indicated 99.1%conversion. The reaction was quenched at the same temperature with 16.7 kg of H3PO4 solution (3.7 kg of 85%H3PO4 and 13 kg of water) slowly and the internal temperature was remained below 30 ℃ . The reaction was diluted with 17 kg of EtOAc. After the phase separation, the organic layer was washed with 13.1 kg of H3PO4 solution (1.1 kg of 85%H3PO4 and 12 kg of water) and then with 10.5 kg of H3PO4 solution (0.46 kg of 85%H3PO4 and 10 kg of water) . The organic layer was assayed for the 1-methyl-1H-pyrazol-5-amine level by HPLC. The HPLC result indicated 2 μg/mL of 1-methyl-1H-pyrazol-5-amine. The organic layer was washed with 15.8 kg of NaCl solution (0.3 kg of NaCl and 15.5 kg of water) . The organic layer was assayed again for the G02586778 level. The HPLC result indicated 0.5 μg/mL of 1-methyl-1H-pyrazol-5-amine. The organic layer was washed with 10.3 kg of NaCl and NaHCO3 solution (1.7 kg of NaCl, 0.6 kg of NaHCO3and 8 kg of water) . After the phase separation, residue water in organic solution was removed through an azeotropic distillation with EtOAc to ≤ 0.5% (by KF) and then the solution was concentrated to 20 to 30 L under a vacuum below 50 ℃ . The solvent was then swapped to MeOH using 30 kg of MeOH and then concentrated to 20 to 30 L for the next step.

[0215]
Step 2:

[0216]

[0217]
To the IX solution in MeOH from the last step was charged 9.0 kg of HCl (1.25 M in MeOH) at the ambient temperature. It was observed slightly exothermic. After addition, the reaction was heated to 45 ℃ . After 16 h, the reaction was monitored by HPLC. The HPLC result indicated the conversion was 99.4%. The reaction was equipped with a distillation setup. The reaction was concentrated to 20 L under a vacuum below 50 ℃ . To the resulting solution was charged 35 kg of MeOH and the reaction was concentrated to 20 L again under a vacuum below 50 ℃ . The solvent was then switched to EtOAc using 40 kg of EtOAc. The solvent ratio was monitored by Headspace GC. If the ratio of MeOH/EtOAc was greater than 1/5, the solvent swap should be continued. Otherwise, the solution was concentrated to 20 L under a vacuum below 50 ℃ . After the solution was cooled below 30 ℃ , 18 kg of NaHCO3 solution (1 kg of NaHCO3 and 17 kg of water) was charged slowly with a medium agitation and followed by 34 kg of EtOAc. After the phase separation, the organic layer was washed with 2 X 8 kg of water. The organic layer was concentrated to 20 L under a vacuum below 50 ℃ . The solvent was then switched to MEK using 35 kg of MEK. The residue MeOH was monitored by Headspace GC. If the level of MeOH was ≥ 0.3%, the solvent swap should be continued. Otherwise, the solution was concentrated to 20 L under a vacuum below 50 ℃ for the next step.

[0218]
Step 3:
The VIII solution in MEK from the last step was transferred to a second 100 L cylindrical reaction vessel through a 1 μm polish filter. In a separated container was prepared 6.0 kg of benzenesulfonic acid solution (1.1 kg of benzenesulfonic acid, 1.2 kg of water and 3.7 kg of MEK) . The filtered solution was heated to 75 ℃ and to the resulting solution was charged 0.6 kg of benzenesulfonic acid solution (10%) through a 1 μm line filter. To the clear solution was charged 0.36 kg of VIIIb crystalline seed slurry in MEK (0.021 kg of VIIIb crystalline seed and 0.34 kg of MEK) . This resulted in a thin slurry. The rest of benzenesulfonic acid solution was then charged through a 1 μm line filter in 2 h. After addition, the slurry was heated at 75 ℃ for additional 1 h and then cooled to 18 ℃ in a minimum of 3 h. The resulting thick slurry was agitated at 18 ℃ for 14-16 h. Solid was filtered using an Aurora dryer. Solid was then washed with 1 μm line filtered 8.15 kg of MEK and water solution (0.35 kg of water and 7.8 kg of MEK) and followed by 1 μm line filtered 12 kg of MEK.
Recrystallization
To a clean 100 L cylindrical reaction vessel was charged 21 kg of EtOH first. With a medium agitation, 3.5 kg of VIIIb was charged and then followed by the rest of EtOH (9 kg) . The thick slurry was heated to 78 ℃ and water (1.2 kg) was charge until a clear solution was obtained. The hot solution was filtered through a 1 μm line filter to a second clean 100 L cylindrical reaction vessel. The temperature dropped to 69 ℃ and the solution remained clear. To the resulting solution was charged with 0.37 kg of VIIIb crystalline seed slurry in EtOH (0.018 kg of VIIIb crystalline seed and 0.35 kg of EtOH) . The thin slurry was concentrated to 20 L at 60-70 ℃ under a vacuum and then cooled 18 ℃ in 3 h. The resulting slurry was agitated at 18 ℃ for 14-16 h. Solid was filtered using a filter dryer. Solid was then washed with 1 μm line filtered 8.6 kg of EtOH and water solution (0.4 kg of water and 8.2 kg of EtOH) . The solution was introduced in two equal portions. The solid was then washed by 1 μm line filtered 6.7 kg of MEK. The wet cake was dried under a vacuum and the nitrogen sweep at a jacket temperature of 35-40 ℃ for a minimum 12 h.
Alternative Synthetic Route (Steps 1 to 10 below)

Step 1:

[0227]

[0228]
Procedure:

[0229]
1. Charge compound 1 and MeBrPPh3 to a four-necked jacketed flask with a paddle stirrer under N2

[0230]
2. Charge THF (5.0V., KF<0.02%) to the flask (Note: V is the volume of solution to mass of limited reagent or L/Kg)

[0231]
3. Stir the suspension at 0 ℃

[0232]
4. Add the NaH (60%suspended in mineral oil) portionwise to the flask at 0 ℃

[0233]
5. Stir at 0 ℃ for 30min

[0234]
6. Heat to 30 ℃ and stir for 6 hrs

[0235]
7. Cool to 0 ℃

[0236]
8. Charge PE (petroleum ether) (5.0V. ) to the flask

[0237]
9. Add the crystal seed of TPPO (triphenylphospine oxide) (1 to about 5%wt of total TPPO) to the flask

[0238]
10. Stir at-10 ℃ for 2hrs

[0239]
11. Filter, and wash the cake with PE (5.0V. )

[0240]
12. Concentrate the filtrate to dryness

[0241]
13. Purification of the product by distillation under reduced pressure affords 2 as colorless oil

[0242]
Step 2:

[0243]

[0244]
Procedure:

[0245]
1. Add (DHQD) 2PHAL, Na2CO3, K2Fe (CN) 6, K2OsO2 (OH) 4 into a flask under N2 (Ad-mix beta, Aldrich, St. Louis, MO) .

[0246]
2. Cool to 0 ℃

[0247]
3. Add tBuOH (5V) and H2O (5V)

[0248]
4. Add 2

[0249]
5. Stir the mixture at 0 ℃ for 6h

[0250]
6. Cool to 0 ℃

[0251]
7. Add Na2SO3 to quench the reaction

[0252]
8. Stir at 0 ℃ for 2h

[0253]
9. Filter and wash the cake with EA (ethyl acetate)

[0254]
10. Separate the organic layer

[0255]
11. Filter and concentrate to dryness

[0256]
Step 3:

[0257]

[0258]
Procedure:

[0259]
1. Add IV (1 eq. ) and DCM (5V) to a flask under N2

[0260]
2. Cool to 0 ℃

[0261]
3. Add DMAP (0.1 eq. ) , then TEA (1.5 eq. )

[0262]
4. Add TBSCl (1.05 eq. ) dropwise at 0 ℃

[0263]
5. Stir the mixture at 0 ℃ for 1h

[0264]
6. Add water to quench the reaction

[0265]
7. Separate the layers

[0266]
8. Dry the organic layer over Na2SO4

[0267]
9. Filter

[0268]
10. Concentrate the filtrate to dryness

[0269]
11. Use for next step directly

[0270]
Step 4:

[0271]

[0272]
Procedure:

[0273]
1. Add V (1.0 eq. ) and DCM (5V) into a flask under N2.

[0274]
2. Cool to 0 ℃

[0275]
3. Add TEA (1.51 eq. )

[0276]
4. Add MsCl (1.05 eq. ) dropwise at 0 ℃

[0277]
5. Stir the mixture at rt for 1h

[0278]
6. Add DCM to dilute the mixture for better stirring

[0279]
7. Add water to quench the reaction

[0280]
8. Separate the layers

[0281]
9. Wash the organic layer with NaHCO3

[0282]
10. Dry over Na2SO4

[0283]
11. Filter and concentrate the filtrate to dryness

[0284]
12. Used for next step directly

[0285]
Step 5:

[0286]

[0287]
Procedure:

[0288]
1. Add VII (1eq. ) and DGME (20V) into flask under N2

[0289]
2. Cool to 0 ℃

[0290]
3. Add KHMDS (1M in THF, 1 eq. )

[0291]
4. Add VI (1.2-1.5 eq. ) in DGME solution

[0292]
5. Stir at 0 ℃ for 5min

[0293]
6. Heat to reflux (jacket 120 ℃) and stir for over 4h

[0294]
7. Cool down

[0295]
8. Quench with water and extraction with MTBE

[0296]
9. Wash with 20%NaCl

[0297]
10. Dry over Na2SO4

[0298]
11. Concentrate to dryness and use to next step directly

[0299]
Step 6:

[0300]

[0301]
Procedure:

[0302]
1. Charge XI (1eq. ) , DCM (8V) into flask under N2

[0303]
2. Add mCPBA by portions

[0304]
3. Stir at room temperature for 2h

[0305]
4. Add 7%NaHCO3 aq. to wash

[0306]
5. Quench with Na2S2O4 aq.

[0307]
6. Wash with 20%NaCl aq.

[0308]
7. Dry over Na2SO4

[0309]
8. Filter and concentrate to dryness

[0310]
9. Slurry the result in MTBE (3V) to afford I

[0311]
Step 7:

[0312]

[0313]
Procedure:

[0314]
1. Add I (1eq. ) , 1-methyl-1H-pyrazol-5-amine (4 eq. ) , Cs2CO3, DMF (4V) into a flask under N2

[0315]
2. Stir at room temperature for 3h

[0316]
3. Work-up to afford product.

[0317]
Step 8:

[0318]

[0319]
Procedure:

[0320]
1. IX was dissolved in MeOH

[0321]
2. HCl (1.25 M in MeOH) was charged at the ambient temperature.

[0322]
3. After addition, the reaction was heated to 45 ℃ for 16 h.

[0323]
4. The reaction was cooled to rt and quenched with aqueous NaHCO3 and diluted with EtOAc

[0324]
5. After the phase separation, the organic layer was washed with water. The organic layer was concentrated to afford the crude VIII

[0325]
Step 9:

[0326]

[0327]
Procedure:

[0328]
1. Charge compound 6-2, XIII, Pd-catalyst and sodium bicarbonate to a four-necked jacketed flask with paddle stirrer under N2

[0329]
2. Charge water and 1, 4-dioxane (5.0V., KF<0.02%) to the flask

[0330]
3. Stir the suspension at 85 ℃ for 16hrs

[0331]
4. Filter through the silica-gel (2.0 X) and diatomaceous earth (0.5X)

[0332]
5. Remove the 1, 4-dioxane by distillation under a vacuum

[0333]
6. Partition between water (2.0V) and EtOAc (5.0V)

[0334]
7. Separate the organic phase and concentrate

[0335]
8. Purify by re-crystallization from PE and EtOAc

[0336]
Step 10:

[0337]

[0338]
Procedure:

[0339]
● Add X into a flask

[0340]
● Add 2M HCl (10-15V)

[0341]
● Heat to 100 ℃ and stir for 3h

[0342]
● Cool down

[0343]
● Neutralize pH to 7 to 8 with 30%NaOH aq.

[0344]
● Extract with THF

[0345]
● Wash with 20%NaCl aq.

[0346]
● Dry over Na2SO4

[0347]
● Filter and concentrate to dryness

[0348]
Synthesis of Crystalline (S) -1- (1- (4-chloro-3-fluorophenyl) -2-hydroxyethyl) -4- (2- ( (1-methyl-1H-pyrazol-5-yl) amino) pyrimidin-4-yl) pyridin-2 (1H) -one benzenesulfonate salt

[0349]
(S) -1- (1- (4-chloro-3-fluorophenyl) -2-hydroxyethyl) -4- (2- ( (1-methyl-1H-pyrazol-5-yl) amino) pyrimidin-4-yl) pyridin-2 (1H) -one (21.1 mg, 0.048 mmol) was dissolved in MEK (0.5 mL) . Benzenesulfonic acid (Fluka, 98%, 7.8 mg, 0.049 mmol) was dissolved in MEK (0.5 mL) and the resulting solution added drop wise to the free base solution with stirring. Precipitation occurred and the precipitate slowly dissolved as more benzenesulfonic acid solution was added. A small amount of sticky solid remained on the bottom of the vial. The vial contents were sonicated for 10 minutes during which further precipitation occurred. The solid was isolated after centrifugation and vacuum dried at 40 ℃ using house vacuum.
A process for the preparation of a compound of formula VIII, the process comprising the steps of:

(a) contacting 4-bromo-1-chloro-2-fluorobenzene with a metallating agent in an aprotic organic solvent to afford an organomagnesium compound, which is reacted with 2-chloro-N-methoxy-N-methylacetamide to afford 2-chloro-1- (4-chloro-3-fluorophenyl) ethanone (II) ;

(b) contacting II with sodium formate and formic acid in aqueous ethanol to afford 1- (4-chloro-3-fluorophenyl) -2-hydroxyethanone (III)

(c) contacting III with a ketoreductase to afford (R) -1- (4-chloro-3-fluorophenyl) ethane-1, 2-diol (IV) ;

(d) contacting IV with a silyl chloride (Ra) 3SiCl and at least one base in a non-polar aprotic solvent to afford (V) , and subsequently adding sulfonylchloride RbS (O) 2Cl to afford VI, wherein Ra is independently in each occurrence C1-6 alkyl or phenyl and Rb is selected from C1-4 alkyl or phenyl, optionally substituted with 1 to 3 groups independently selected from C1-3 alkyl, halogen, nitro, cyano, or C1-3 alkoxy;

(e) contacting 4- (2- (methylsulfonyl) pyrimidin-4-yl) pyridin-2 (1H) -one (VII) with a strong base in an organic solvent and subsequently adding VI to afford XI;

(f) treating XI with an oxidizing agent to afford I;

(g) treating 1-methyl-1H-pyrazol-5-amine with a strong base in an aprotic solvent at reduced temperature and adding the compound of formula I to afford IX; and,

(h) contacting IX with a de-silylating agent to afford VIII.

PAPER

Development of a Practical Synthesis of ERK Inhibitor GDC-0994

Small Molecule Process Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
Process Research, F. Hoffmann-La Roche AG, Grenzacherstrasse 124, CH-4070 Basel, Switzerland
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.7b00006
Abstract Image

The process development of a synthetic route to manufacture ERK inhibitor GDC-0994 on multikilogram scale is reported herein. The API was prepared as the corresponding benzenesulfonate salt in 7 steps and 41% overall yield. The synthetic route features a biocatalytic asymmetric ketone reduction, a regioselective pyridone SN2 reaction, and a safe and scalable tungstate-catalyzed sulfide oxidation. The end-game process involves a telescoped SNAr/desilylation/benzenesulfonate salt formation sequence. Finally, the development of the API crystallization allowed purging of process-related impurities, obtaining >99.5A% HPLC and >99% ee of the target molecule.

1 to 6 of 6
Patent ID Patent Title Submitted Date Granted Date
US2016136150 COMPOUNDS AND COMPOSITIONS AS INHIBITORS OF MEK 2015-11-13 2016-05-19
US2016122316 SERINE/THREONINE KINASE INHIBITORS 2016-01-12 2016-05-05
US2015111869 USE OF A COMBINATION OF A MEK INHIBITOR AND AN ERK INHIBITOR FOR TREATMENT OF HYPERPROLIFERATIVE DISEASES 2014-08-29 2015-04-23
US2015051209 COMPOUNDS AND COMPOSITIONS AS INHIBITORS OF MEK 2014-08-05 2015-02-19
US2014249127 SERINE/THREONINE KINASE INHIBITORS 2014-02-14 2014-09-04
US8697715 Serine/threonine kinase inhibitors 2013-03-01 2014-04-15

///////////GDC 0994, Ravoxertinib, 1453848-26-4, GDC0994, UNII-R6AXV96CRH, R6AXV96CRH, RG7842, RG-7842, RG 7842, PHASE 1

CN1C(=CC=N1)NC2=NC=CC(=N2)C3=CC(=O)N(C=C3)C(CO)C4=CC(=C(C=C4)Cl)F