New Drug Approvals

Home » Posts tagged 'FDA 2015' (Page 3)

Tag Archives: FDA 2015

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

Blog Stats

  • 4,802,106 hits

Flag and hits

Flag Counter

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

Join 37.9K 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 37.9K 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

Verified Services

View Full Profile →

Archives

Categories

Flag Counter

Amgen receives FDA approval for chronic heart failure medicine Corlanor


 

Amgen receives FDA approval for chronic heart failure medicine Corlanor
Amgen has received approval from the US Food and Drug Administration (FDA) for its Corlanor (ivabradine) to treat patients with chronic heart failure.

read at http://www.pharmaceutical-technology.com/news/newsamgen-receives-fda-approval-chronic-heart-failure-medicine-corlanor-4555383?WT.mc_id=DN_News

keep watching

will be updated………………..

FDA approves first generic Copaxone to treat multiple sclerosis


04/16/2015 01:10 PM EDT

April 16, 2015

The U.S. Food and Drug Administration today approved the first generic version of Copaxone (glatiramer acetate injection), used to treat patients with relapsing forms of multiple sclerosis (MS).

Sandoz has received FDA approval to market generic glatiramer acetate in a 20 mg/1 ml daily injection.

“Health care professionals and patients can be assured that FDA-approved generic drugs have met the same rigorous standards of quality as the brand-name drug,” said Janet Woodcock, M.D., director of the FDA’s Center for Drug Evaluation and Research. “Before approving this generic product, given its complexity, we reviewed additional information to make sure that the generic product is as safe and effective as the brand name product.”

The FDA applies the same rigorous and reliable standards to evaluate all generic drug products. As needed, the agency requires appropriate information to demonstrate sameness for complex active ingredients, such as glatiramer acetate. For this approval, FDA scientists established a thorough scientific approach for demonstrating active ingredient sameness that takes into consideration the complexity of glatiramer acetate.

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 periods may be incomplete, leading to progressive decline in function and increased disability. MS patients often experience muscle weakness and difficulty with coordination and balance. Most people experience their first symptoms of MS between the ages of 20 and 40.

In the clinical trials for Copaxone, the most common adverse reactions reported by those taking Copaxone were skin problems at the injection site (redness, pain, swelling and itching), flushing (vasodilation), rash, shortness of breath and chest pain.

BLOG STATS OF NEW DRUG APPROVALS

Flag Counter
646453 VIEWS

 

MOSCOW


P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.
P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.
P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

COCK WILL TEACH YOU NMR
COCK SAYS MOM CAN TEACH YOU NMR
DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO …..FOR BLOG HOME CLICK HERE
Join me on Linkedin

View Anthony Melvin Crasto Ph.D's profile on LinkedIn

Join me on Facebook FACEBOOK
Join me on twitterFollow amcrasto on Twitter  @amcrasto
Join me on google plus Googleplus

 amcrasto@gmail.com

FDA approves new treatment for diabetic retinopathy in patients with diabetic macular edema


03/25/2015
The U.S. Food and Drug Administration today expanded the approved use for Eylea (aflibercept) injection to treat diabetic retinopathy in patients with diabetic macular edema.

March 25, 2015

Release

The U.S. Food and Drug Administration today expanded the approved use for Eylea (aflibercept) injection to treat diabetic retinopathy in patients with diabetic macular edema.

Diabetic retinopathy (DR) is the most common diabetic eye disease and is a leading cause of blindness in adults in the United States. According to the Centers for Disease Control and Prevention, diabetes (type 1 and type 2) affects more than 29 million people in the United States and is the leading cause of new blindness among people ages 20 to 74 years. In 2008, 33 percent of adults with diabetes aged 40 years or older had some form of DR. In some cases of DR with diabetic macular edema (DME), abnormal new blood vessels grow on the surface of the retina. Severe vision loss or blindness can occur if the new blood vessels break.

“Diabetes is a serious public health crisis, affecting more patients every year,” said Edward Cox, M.D., M.P.H, director of the Office of Antimicrobial Products in the FDA’s Center for Drug Evaluation and Research. “Today’s approval gives patients with diabetic retinopathy and diabetic macular edema another therapy to treat this vision-impairing complication.”

In February, the FDA approved Lucentis (ranibizumab injection) 0.3 mg to treat DR in patients with DME.

Eylea is administered by a physician as an injection into the eye once a month for the first five injections and then once every two months. It is intended to be used along with appropriate interventions to control blood sugar, blood pressure and cholesterol.

The safety and efficacy of Eylea to treat DR in patients with DME were evaluated in 679 participants in two clinical studies where participants were randomly assigned to receive Eylea or macular laser photocoagulation, a laser-based treatment used to burn small areas of the retina. At week 100, participants being treated with Eylea showed significant improvement in the severity of their DR, compared to patients who did not receive Eylea.

The most common side effects associated with Eylea include bleeding of the conjunctiva (the tissue that lines the inside of the eyelids and covers the white part of the eye); eye pain; cataracts; floaters; increased pressure inside the eye (increased intraocular pressure); and separation of the interior jelly of the eye from the retina (vitreous detachment). Serious adverse reactions include infection within the eye (endophthalmitis) and retinal detachments.

The FDA granted breakthrough therapy designation to Eylea for the treatment of DR with DME. The FDA can designate a drug a breakthrough therapy at the request of the sponsor if preliminary clinical evidence indicates the drug may demonstrate a substantial improvement over available therapies for patients with serious or life-threatening conditions. The FDA also reviewed the new use for Eylea under the agency’s priority review program, which provides for an expedited review of drugs that demonstrate the potential to be a significant improvement in safety or effectiveness in the treatment of a serious condition.

The FDA previously approved Eylea to treat wet (neovascular) age-related macular degeneration, a condition in which abnormal blood vessels grow and leak fluid into the macula. Eylea is also approved to treat DME and macular edema secondary to retinal vein occlusions, both of which cause fluid to leak into the macula resulting in blurred vision.

Eylea is marketed by Tarrytown, N.Y.-based Regeneron Pharmaceuticals Inc. Lucentis is marketed by South San Francisco, California-based Genentech, a subsidiary of Roche Pharmaceuticals.

Brexpiprazole ブレクスピプラゾール


Brexpiprazole structure.svg

Brexpiprazole

ブレクスピプラゾール

OPC-34712, UNII-2J3YBM1K8C, OPC34712,
CAS 913611-97-9,
Molecular Formula:C25H27N3O2S
Molecular Weight:433.56578 g/mol
7-[4-[4-(1-benzothiophen-4-yl)piperazin-1-yl]butoxy]-1H-quinolin-2-one
7-[4-[4-(1-Benzothiophen-4-yl)piperazin-1-yl]butoxy]quinolin-2(1H)-one
2(1H)​-​Quinolinone, 7-​[4-​(4-​benzo[b]​thien-​4-​yl-​1-​piperazinyl)​butoxy]​-
7- [ 4- ( 4-benzo[b]thiophen-4- yl-piperazin-l-yl)butoxy] -lH-quinolin-2-one
7-[4-(4-benzo[b]thiophen-4-yl-piperazin-1-yl)butoxy]-1H-quinolin-2-one
Otsuka Pharma Co Ltd,

OTSUKA ……………INNOVATOR

NDA is considered filed as of September 9, 2014 (60 days after submission). The PDUFA date is July 11, 2015.

 UPDATE JULY 2015 ON STATUS OF APPROVAL

Approval Status:

Approved July 2015

Specific Treatments:

depression and schizophrenia

Therapeutic Areas

Brexpiprazole (/brɛksˈpɪprəzl/ breks-pip-rə-zohl; also called OPC-34712) is a novel D2 dopamine partial agonist investigational product currently in clinical trials for the treatment of depression, schizophrenia, and attention deficit hyperactivity disorder(ADHD).[1]Although it failed Stage 2 trials for ADHD, it has been designed to provide improved efficacy and tolerability (e.g., lessakathisia, restlessness and/or insomnia) over established adjunctive treatments for major depressive disorder (MDD).[2]

OPC-34712 is an antidepressant and antipsychotic drug candidate awaiting approval in the U.S. for the treatment of schizophrenia and also as adjunctive treatment of major depressive disorder (MDD). The product is in phase III clinical trials for the treatment of agitation associated with Alzheimer’s disease. Phase III clinical trials are also underway for the treatment of post-traumatic stress disorder (PTSD).

brexpiprazole (pre-registration, as of April 2015), which is being developed by Otsuka and Lundbeck, useful for treating schizophrenia, agitation associated with Alzheimer’s disease, major depressive disorder and attention deficit hyperactivity disorder. Family members of the product case, WO2006112464, hold protection in EU states until 2026 and its US equivalent, US7888362, has US154 extension, expiring in 2027. Suzhou Vigonvita Life Sciences appears to be new to patenting and is the first collaborative filing from the three assignees.

Phase II clinical trials are also ongoing for use as adjunctive therapy in adults with attention deficit hyperactivity disorder (ADHD). The compound is being developed by Otsuka Pharmaceutical. In 2011, a codevelopment and commercialization agreement was signed by Lundbeck and Otsuka Pharmaceutical in Latin and North America, Australia and Europe for the treatment of psychiatric disorders.

The drug is being developed by Otsuka, and is considered to be a successor[3] of its top-selling antipsychotic agent aripiprazole(brand names: Abilify, Aripiprex). Otsuka’s US patent on aripiprazole expired on October 20, 2014;[4] however, due to a pediatric extension, a generic will not become available until at least April 20, 2015.[5]

Brexpiprazole (1) , a serotonin–dopamine activity modulator, is an investigational new drug currently in phase-III clinical trials for the treatment of depression, schizophrenia, and attention deficit hyperactivity disorder.(1A) Brexpiprazole is also considered to be a possible successor to the top-selling antipsychotic agent aripiprazole.(2A)

  1. 1A……….Phase II and Phase III Drugs in U.S. Development for Depression, Anxiety, Sleep Disorders, Psychosis & ADHD, 2011. http://www.neurotransmitter.net/newdrugs.html(accessed Jan 27, 2015).

  2. 2A…………FDA accepts new schizophrenia drug filing, 2014.http://www.pharmafile.com/news/194878/fda-accepts-new-schizophrenia-drug-filing(accessed Jan 27, 2015).
    BREXPIPRAZOLE.png
    Brexpiprazole

    In the clinical program, brexpiprazole demonstrated improvement in symptoms in both schizophrenia and as adjunctive therapy in major depressive disorder (MDD)

    July 2015 is the anticipated completion timing of the FDA’s review (based on PDUFA timeline)Otsuka Pharmaceutical Co., Ltd. (Otsuka) and H. Lundbeck A/S (Lundbeck) today announced that the U.S. Food and Drug Administration (FDA) has determined that the New Drug Application (NDA) for brexpiprazole for monotherapy in adult patients with schizophrenia and for adjunctive treatment of major depressive disorder (MDD) in adult patients is sufficiently complete to allow for a substantive review, and the NDA is considered filed as of September 9, 2014 (60 days after submission). The PDUFA date is July 11, 2015.The NDA is supported by seven completed placebo-controlled clinical phase II or III studies in the proposed indications – three studies in schizophrenia and four studies with brexpiprazole as adjunctive therapy in MDD. The dossier included data from more than 6,000 participants of whom more than 5,000 received brexpiprazole.

    Brexpiprazole in adult patients with schizophreniaOne clinical phase II and two clinical phase III placebo-controlled studies have been completed using brexpiprazole in adult patients suffering from schizophrenia. Across the three studies more than 1,700 patients have been randomized.In the first pivotal phase III study randomizing approximately 625 patients, brexpiprazole 2mg/day and 4 mg/day both demonstrated greater improvement of symptoms relative to placebo as measured by change from baseline in the Positive and Negative Syndrome Scale (PANSS) Total Score at week 6 (p<0.05). Results of the key secondary endpoint supported primary results.In the second pivotal phase III study randomizing approximately 650 patients, brexpiprazole 4 mg/day again demonstrated greater improvement of symptoms relative to placebo (p<0.05) in change from baseline in the PANSS Total Score at Week 6. Brexpiprazole 2 mg/day showed numerical improvement (p>0.05) over placebo at Week 6.The results from the clinical phase II studyi were presented at the 24th Annual US Psychiatric and Mental Health Congress in November 2011. The study showed a clinically meaningful improvement from baseline measured by PANSS total score at week 6, although it did not achieve statistical separation from placeboii.In the placebo-controlled phase II and III studies, the rates of discontinuation due to adverse events were 8.1% for patients receiving brexpiprazole compared to 12.7% of patients receiving placebo; the only adverse event that occurred in more than 5% of brexpiprazole patients and more frequently than placebo was akathisia (5.8% vs. 4.5%).
    Brexpiprazole as adjunctive therapy in major depressive disorder (MDD) Four studies have been included in the dossier using brexpiprazole as adjunctive therapy for adult patients suffering from MDD who had demonstrated a consistent, inadequate response to at least two regimens of prior antidepressant treatment. Patients with MDD and an inadequate response to one to three antidepressants were enrolled and received antidepressants for 8 weeks, single blinded, in the two phase III studies. Patients with an inadequate response during this prospective phase were provided antidepressant therapy and randomized adjunctive treatment with either brexpiprazole or placebo for 6 weeks. The primary efficacy endpoint was the change in MADRS (Montgomery–Åsberg Depression Rating Scale) Total Score from baseline at week 6. MADRS is a commonly used scale to assess the range of symptoms in patients with MDD. Across the four studies, more than 3,900 patients entered the prospective phase and more than 1,800 patients were included in the randomized phase of the studies.The first pivotal phase III results were presented in a poster session at the 22nd European Psychiatry Association Congress (EPA) in March 2014. This two-arm phase III study randomized approximately 380 patients and demonstrated an improvement of symptoms with an antidepressant plus 2 mg brexpiprazole that was greater than an antidepressant plus placebo (p<0.001)The second pivotal phase III study was a three-arm study in which approximately 675 patients were randomized to treatment with an antidepressant plus either placebo, 1 mg brexpiprazole or 3 mg brexpiprazole.v Patients in both brexpiprazole treatment groups showed greater improvement in symptoms as measured by the MADRS compared to placebo (1 mg p>0.05, 3 mg p<0.05). Results of the second pivotal phase III study in MDD have not yet been published.

    The first clinical phase IIvi study randomized approximately 425 patients in four arms and was presented at the 164th Annual Meeting of the American Psychiatric Association in May 2011. Patients exhibited greater improvements than adjunctive placebo in MADRS Total score with the 1.5 (±0.5) mg/day dose of brexpiprazole after six weeks of treatment (p

    About brexpiprazole (OPC-34712)Brexpiprazole is a novel investigational psychotropic compound discovered by Otsuka and under co-development with Lundbeck. Brexpiprazole is a serotonin-dopamine activity modulator (SDAM) that acts as a partial agonist at 5-HT1A and dopamine D2 receptors at similar potency, and an antagonist at 5-HT2A and noradrenaline alpha1B/2C receptors.

Partnership with Lundbeck

In November 2011, Otsuka and Lundbeck have announced a global alliance.[6] Lundbeck has given Otsuka an upfront payment of $200 million, and the deal includes development, regulatory and sales payments, for a potential total of $1.8 billion. Specifically for OPC-34712, Lundbeck will obtain 50% of net sales in Europe and Canada and 45% of net sales in the US from Otsuka.

The partnership has been presented by Otsuka to its investors as a good fit for several reasons:[7]

  • Geographic strategy: Otsuka in Japan, Asia, US; Lundbeck in Europe, South America and emerging markets
  • Research strategy: Otsuka has knowledge in antipsychotics, Lundbeck in anti-depressant and anxiolytic.
  • CNS strategy: Otsuka has a robust portfolio in next-generation CNS drugs, while Lundbeck covers a wide range of CNS conditions from Alzheimer’s to schizophrenia.
  • Similar corporate culture

Clinical trials

OPC-34712 is currently in clinical trials for adjunctive treatment of MDD, adjunctive treatment of adult ADHD and schizophrenia.[8]

Major depression

Phase II

The Phase 2 multicenter, double-blind, placebo-controlled study randomized 429 adult MDD patients who exhibited an inadequate response to one to three ADTs in the current episode. The study was designed to assess the efficacy and safety of OPC-34712 as an adjunctive treatment to standard ADT. The ADTs included in the study were desvenlafaxine, escitalopram, fluoxetine, paroxetine, sertraline, and venlafaxine.[9]

Phase III

A new Phase III study is currently in the recruiting stage: “Study of the Safety and Efficacy of Two Fixed Doses of OPC-34712 as Adjunctive Therapy in the Treatment of Adults With Major Depressive Disorder (the Polaris Trial)”.[10] Its goal is “to compare the effect of OPC-34712 to the effect of placebo (an inactive substance) as add on treatment to an assigned FDA approved antidepressant treatment (ADT) in patients with Major Depressive Disorder who demonstrate an incomplete response to a prospective trial of the same assigned FDA approved ADT”. Estimated enrollment is 1250 volunteers.

Brexpiprazole, code: OPC-34712) is Otsuka Pharmaceutical Co., Ltd. developed a new generation of anti-psychotic drug candidates, and its role in multiple receptors, dopamine D2 receptor partial agonist (improving positive and negative symptoms, cognitive impairment and depressive symptoms), 5-HT2A receptor antagonist (improving negative symptoms, cognitive dysfunction, symptoms of depression, insomnia), α1 adrenoceptor antagonists (improving positive symptoms of schizophrenia), 5 – serotonin uptake / reuptake inhibitors (improve depressive symptoms); at the same time, but also a 5-HT1A partial agonist (anxiolytic and antidepressant activity) and 5-HT7 antagonist (temperature, circadian rhythms, learning and memory, sleep) . Currently, in the United States and Europe as adjuvant treatment of severe depression (MDD) Phase III clinical trial; III clinical trial for the treatment of schizophrenia in the United States, Europe and Japan, meanwhile, is still the United States Phase II adult ADHD Clinical Trials.

Adult ADHD

Phase II

  • Study of the Safety and Efficacy of OPC-34712 as a Complementary Therapy in the Treatment of Adult Attention Deficit/Hyperactivity Disorder (STEP-A)[11] The company did not move the product to Phase III, and it is presumed this drug failed Phase II trials for the disorder.

Schizophrenia

Phase I

  • Trial to Evaluate the Effects of OPC-34712 on QT/QTc in Subjects With Schizophrenia or Schizoaffective Disorder[12]

Phase II

  • A Dose-finding Trial of OPC-34712 in Patients With Schizophrenia[13]

Phase III

  • Efficacy Study of OPC-34712 in Adults With Acute Schizophrenia (BEACON)[14]
  • Safety and Tolerability Study of Oral OPC-34712 as Maintenance Treatment in Adults With Schizophrenia (ZENITH)[15]
  • Study of the Effectiveness of Three Different Doses of OPC-34712 in the Treatment of Adults With Acute Schizophrenia (VECTOR)[16]
  • A Long-term Trial of OPC-34712 in Patients With Schizophrenia[17]

Conferences

  • Phase II results were presented at the American Psychiatric Association’s 2011 annual meeting in May 2011.[18]
  • The drug has been presented at the 2nd Congress of Asian College of Neuropsychopharmacology[19] in September 2011.
  • At the US Psychiatric and Mental Health Congress in November 2011 in Vegas, Robert McQuade presented the Phase II Trial results for Schizophrenia[20]

 Pharmacology

Brexpiprazole acts as a partial agonist of the 5-HT1A, D2, and D3 receptors, and as an antagonist of the 5-HT2A, 5-HT2B, 5-HT7, α1A, α1B, α1D, and α2C-adrenergic, and H1receptors.[22] It has negligible affinity for the mACh receptors.[22]

Patents

  • U.S. Patent 8,071,600
  • WIPO PCT/JP2006/317704
  • Canadian patent: 2620688[24]
  • WO 2013162046
  • WO 2013161830
  •  WO 2013162048
  • WO 2013015456
  • JP 2008115172
  • WO 2006112464
  • WO2015054976 NEW
Patent Submitted Granted
PIPERAZINE-SUBSTITUTED BENZOTHIOPHENES FOR TREATMENT OF MENTAL DISORDERS [US2011152286] 2011-06-23
Piperazine-substituted benzothiophenes for treatment of mental disorders [US7888362] 2010-07-15 2011-02-15
Otsuka Pharmaceutical Co., Ltd. are disclosed in PCT Application WO2006112464A1 in the preparation route see Scheme 1, the difficulty of the route is the first reaction generates byproducts easily separated by column chromatography is not easy to obtain high-purity intermediates, thus affecting the final product Bray prazosin purity and yield.Scheme 1:

Subsequently, Otsuka Pharmaceutical Co., Ltd. are disclosed in PCT Application WO2013015456A1 in the alternative method of preparing the reaction of this step, see Scheme 2, the route the reagents are more expensive, high-cost, environmentally unfriendly and not suitable for industrial production.

Reaction Scheme 2:

Due to the above production process there is a high cost, and difficult to separate impurities and other shortcomings, it is necessary to find an economical, practical, environmental protection, new routes to improve process stability, reduce costs, improve product quality.

Synthesis

WO 2013015456

IN THIS BELOW PIC WE SEE

click on pics below to view

Synthesis of A

1 BROMO 4 CHLORO BUTANE WAS REACTED WITH 7 HYDROXY 1H QUINOLINE -2-ONE TO GIVE A

7 ( 4 CHLORO BUTOXY)-1H -QUINOLINE-2-ONE, WHICH WILL BE USED FOR COUPLING AT LAST STAGE

1 BROMO 4 CHLORO BUTANE

WP_000310

IN THE BELOW PIC  2,6-Dichlorobenzaldehyde AND RHODANINE WERE REACTED TO GIVE 2,6-dichlorobenzylidenerhodanine.

2,6-Dichlorobenzaldehyde

RHODANINE

NEXT WAS
2,6-dichlorobenzylidenerhodanine, GAVE (Z)-3-(2,6-dichlorophenyl)-2-mercapto-2-propenoic acid.

1H-NMR (DMSO-d6) d
ppm; 7.23-7.67 (4H, m), 3.5-5.7 (1H, br.), 11.7-14.5 (1H, br.).

Next was prepration of K salt

(Z)-3-(2,6-dichlorophenyl-2-mercapto-2-propenoic acid and  potassium hydroxide gave ((Z)-3-(2,6-dichlorophenyl-2-mercapto-2-propenoic acid potassium salt).

Next stage

((Z)-3-(2,6-dichlorophenyl-2-mercapto-2-propenoic acid potassium
salt) GAVE  2-carboxy-4-chlorobenzo[b]thiophene.
Yield: 48.8 g. 1H-NMR (DMSO-d6) d ppm; 7.53 (1H, t, J = 7.7 Hz), 7.58 (1H, dd, J = 7.7, 1.3
Hz), 8.03 (1H, d, J = 0.5 Hz), 8.07 (1H, d, J = 7.6 Hz).

NEXT IS DECARBOXYLATION

A mixture of 2-carboxy-4-chlorobenzo[b]thiophene, 1,3-dimethyl-2-imidazolidinone, and 1,8-
diazabicyclo[5.4.0]-undec-7-ene GAVE  compound. 4-chlorobenzo[b]thiophene.  1H-NMR (DMSO-d6) d ppm; 7.38 (1H, t, J = 8.4
Hz), 7.51 (1H, dd, J = 5.5, 0.8 Hz), 7.48 (1H, dd, J = 7.7, 0.9 Hz), 7.94 (1H, dd, J = 5.5, 0.4
Hz), 8.02 (1H, dt, J = 8.0, 0.9 Hz).

WP_000309

BETTER REPRESENTATION OF ABOVE PIC

CLIPS FROM PATENT

Synthesis of 2,6-dichlorobenzylidenerhodanine

2,6-Dichlorobenzaldehyde (77.0 g) , rhodanine (58.6 g) , and acetic acid (539 ml) were suspended with stirring at room temperature. Anhydrous sodium acetate (116 g) was added to the suspension, and the resulting mixture was heated under reflux for 3 hours. The reaction mixture was cooled to 45°C, and ice water (700 ml) was added. After the mixture was stirred for 0.2 hours, the precipitated crystals were collected by filtration, washed with water, and then dried to obtain 2,6- dichlorobenzylidenerhodanine. Even in non-dried form, this product could be subjected to the subsequent step.

Yield: 125.4 g^- MR (CDC13) 6ppm;7.30-7.44 (3H, m) , 7.70 (1H. s), 9.6 (1H, br.).

Reference Example 3

• Synthesis of (Z)-3-(2,6-dichlorophenyl)-2-mercapto-2-propenoic acid

A suspension of 2,6- dichlorobenzylidenerhodanine (160.4 g) and water (800 ml) was stirred at room temperature, and sodium hydroxide (83.0 g) was added over a period of 1 hour. The resulting mixture was heated with stirring for another 0.5 hours. The reaction mixture was cooled with ice (10°C), and concentrated hydrochloric acid (192 ml) was added. After the mixture was stirred while cooling with ice for 0.5 hours, the precipitated crystals were collected by filtration. The crystals obtained by filtration were washed with water and then dried to obtain an equivalent amount of (Z)-3-(2,6-dichlorophenyl)-2-mercapto-2- propenoic acid.

Yield: 138.9 g l-NMR (DMSO-de) δρρπΐ;7.23-7.67 (4H, m) , 3.5-5.7 (1H, br.), 11.7-14.5 (1H, br.).

Reference Example 4

• Synthesis of 2-carboxy-4-chlorobenzo[b] thiophene

A suspension of (Z)-3-(2,6-dichlorophenyl-2-mercapto-2- propenoic acid (72.4 g) and water (362 ml) was stirred at room temperature. Further, potassium hydroxide (40.8 g) was added, and the mixture was heated under reflux for 4 hours . After the mixture was allowed to cool, the mixture was stirred for 1 hour while cooling with ice. The precipitated crystals ((Z)-3-(2,6- dichlorophenyl-2-mercapto-2-propenoic acid potassium salt) were collected by filtration and washed with cold water. After the crystals were suspended in water, 35% concentrated hydrochloric acid (32 ml) was added (pH = 1), and the mixture was stirred at room temperature for 1 hour. The precipitated crystals were collected by filtration and dried to obtain 2-carboxy-4- chlorobenzo[b] thiophene.

Yield of 48.8 g ^- MRiDMSO-de) 6ppm; 7.53 (1H, t, J = 7.7 Hz), 7.58 (1H, dd, J = 7.7, 1.3 Hz), 8.03 (1H, d, J = 0.5 Hz), 8.07 (1H, d, J = 7.6 Hz).

Reference Example 5

• Synthesis of K salt  4-chlorobenzo[b] thiophen-2-carboxylate

Reference Example 6

· Synthesis of 2-carboxy-4-chlorobenzo[b]thiophene

Sodium 4-chlorobenzo[b] thiophen-2-carboxylate (2.40 g) was dissolved in water (33 ml) at 60°C. Concentrated hydrochloric acid (1.3 ml) was added to the solution at the same temperature, and the resulting mixture was stirred. The precipitated crystals were collected by filtration, washed with water, and then dried to obtain 2-carboxy-4-chlorobenzo[b] thiophene.

Yield: 1.61 g ^- MR (DMS0-d6);7.53 (1H, t, J = 7.7 Hz), 7.58 (1H, dd, J = 7.7, 1.3 Hz), 8.03 (1H, d, J = 0.5 Hz), 8.07 (1H, d, J = 7.6 Hz).

e

d

Elaborate description

IN THIS BELOW PIC WE SEE

Synthesis of 4-(1-piperazinyl)benzo[b]thiophene

4-Chlorobenzo[b]thiophene and xylene , Subsequently, piperazine, sodium tert-butoxide, palladium acetate (II), and 2-dicyclohexylphosphino-2′,6′-di-iso-propoxy-1,1′-biphenyl (RuPhos) …… producing 4-(1-piperazinyl)benzo[b]thiophene.

NEXT IS PREPARATION OF HYDROCHLORIDE

4-(1-piperazinyl)benzo[b]thiophene hydrochloride. 1H-NMR (DMSO-d6) d ppm;
3.30 (4H, br.s), 3.61 (4H, br.s), 6.97 (1H, d, J = 7.8 Hz), 7.32 (1H, br. dd, J = 8.4, 7.8 Hz),
7.53 (1H, d, J = 5.6 Hz), 7.70 (1H, d, J = 8.4 Hz), 7.76 (1H, d, J = 5.6 Hz), 9.37 (1H, br.s).

NEXT  IS REACTION WITH A TO GIVE BREXPIPRAZOLE

1-benzo[b]thiophen-4-yl-piperazine hydrochloride, potassium carbonate
and DMF  and  7-(4-chlorobutoxy)-1H-quinolin-2-one A  FROM PIC 1 and potassium iodide  GAVE BREXPIPRAZOLE, ie 7-[4-(4-benzo[b]thiophen-4-yl-piperazin-1-yl)butoxy]-1H-quinolin-2-one.

1H-NMR (DMSO-d6) d ppm; 1.6-1.75 (2H, m), 1.75-1.9 (2H, m), 2.44
(2H, t, J = 7.0 Hz), 2.55-2.70 (4H, m), 3.00-3.15 (4H, m), 4.06 (2H, t, J = 6.3 Hz), 6.30 (1H,
d, J = 9.5 Hz), 6.75-6.85 (2H, m), 6.88 (1H, d, J = 7.5 Hz), 7.27 (1H, dd, J = 8 Hz, 8 Hz),
7.40 (1H, d, J = 5.5 Hz), 7.55 (1H, d, J = 9.5 Hz), 7.61 (1H, d, J = 8 Hz), 7.69 (1H, d, J = 5.5
Hz), 7.80 (1H, d, J = 9.5 Hz), 11.58 (1H, bs).

WP_000308

BETTER REPRESENTATION OF  PIC

Example 2

• Synthesis of 4- (l-piperazinyl)benzo[b]thiophene hydrochloride

4-Chlorobenzo[b] thiophene (5.00 g), piperazine (5.11 g) , palladium acetate (II) (2.7 mg), tri-tert-butylphosphonium

tetraphenylborate (6.2 mg), sodium tert-butoxide (8.548 g), and xylene (70 ml) were stirred at 120 to 130°C for 5 hours. After the reaction mixture was cooled to room temperature, water was added thereto, and the layers were separated. The xylene layer was washed with water, and then with saline. After addition of activated carbon, the mixture was stirred at room temperature for 30 minutes. After filtration of the mixture, concentrated

hydrochloric acid was added to the filtrate, and the resulting mixture was stirred at room temperature for 30 minutes. The precipitated crystals were collected by filtration and dried to obtain 4- ( l-piperazinyl)benzo[b] thiophene hydrochloride.

Yield: 6.94 g !H-NMRiDMSO-de) 6ppm; 3.30 (4H, br.s), 3.61 (4H, br.s), 6.97 (1H, d, J= 7.8 Hz), 7.32 (1H, br.dd, J= 8.4. 7.8 Hz), 7.53 (1H, d, J= 5.6 Hz), 7.70 (1H, d, J= 8.4 Hz), 7.76 (1H, d, J= 5.6 Hz), 9.37 (1H, br.s).

Example 3

• Synthesis of 4- ( 1-piperazinyl)benzo[b] thiophene hydrochloride

4-Chlorobenzo[b] thiophene (10.0 g) and xylene (100 ml) were placed in a reaction vessel. The reaction vessel was

evacuated and then purged with argon. Subsequently, piperazine (15.3 g) , sodium tert-butoxide (17.1 g) , palladium acetate (II) (13.0 mg) , and 2-dicyclohexylphosphino-2′,6′-di-iso-propoxy-1,1′- biphenyl (RuPhos) (69.0 mg) were added. After evacuation and purging with argon, the mixture was refluxed for 2 hours. After the reaction mixture was cooled to about 80°C, water (50 ml) and silica #600H (0.65 g) were added. The mixture was stirred at approximately 60°C for about 10 minutes, and then filtered. After the filtrate was separated into layers, the xylene layer was washed with water. Subsequently, the xylene layer was placed into the reaction vessel again. After addition of water (200 ml) and concentrated hydrochloric acid (8.0 ml) , the mixture was heated with stirring for dissolution. The layers were separated at 75°C or more. After the aqueous layer was collected, toluene (150 ml) and 25% aqueous sodium hydroxide solution (16 ml) were added, and the mixture was stirred. The layers were separated, and the organic layer was collected. The organic layer was washed with water and concentrated with an evaporator. Methanol (150 ml) was added to the concentrated oil to dissolve the oil, thus producing a methanol solution. 2-Propanol (150 ml) and concentrated

hydrochloric acid (7 ml) were placed into another reaction vessel, and the methanol solution was added dropwise over a period of 15 minutes or more. After completion of the dropwise addition, the mixture was cooled and stirred at 10°C or less for about 30 minutes, and then filtered (washed with a mixture of 5 ml of methanol and 5 ml of 2-propanol) . The crystals were collected, and then dried to obtain 4-(l-piperazinyl)benzo[b]thiophene hydrochloride.

Yield: 11.61 g

^-NMRfDMSO-de) oppm;

3.25-3.40 (8H, br.s), 6.96 (1H, d, J = 7.5 Hz), 7.32 (1H, dd, J = 8.0, 7.5 Hz), 7.52 (1H, d, J = 5.5 Hz ) . 7.70 (1H, d, J = 8.0 Hz), 7.75 (1H, d, J = 5.5 Hz), 9.35 (1H, br.s).

Reference Example 9

· Synthesis of 7- ( 4-chlorobutoxy) -lH-quinolin-2-one

After 7-hydroxy-lH-quinolin-2-one (10 g) and DMF (50 ml) were heated to approximately 30°C, an aqueous potassium carbonate solution (potassium carbonate: 8.6 g, water: 10 ml) was added. After the mixture was stirred at 30 to 40°C for about 15 minutes, l-bromo-4-chlorobutane (14.3 ml) was added and stirred at approximately 40°C for 5 hours. Water (100 ml) was added dropwise over a period of 30 minutes or more while the

temperature was maintained at 30°C or more. After the mixture was stirred at approximately 30°C for 30 minutes, stirring was continued at 10°C or less for 1 hour, after which the precipitated crystals were collected by filtration. After methanol (100 ml) was added to the precipitated crystals, the mixture was stirred under reflux to ensure dissolution. This solution was cooled and stirred at 30 to 40°C for 30 minutes and then at 5°C or less for about 1 hour, after which the precipitated crystals were

collected by filtration. The crystals were dried at 60°C to obtain 7- (4-chlorobutoxy) -lH-quinolin-2-one as white powder.

Yield: 12.3 g

^I-NMR (300 MHz; CDC13) oppm; 1.95-2.05 (4H, m) , 3.64 (2H, t, J = 6.0Hz), 4.10 (2H, t. J = 5.5 Hz), 6.56 (1H, d, J = 9.5 Hz), 6.80 (1H. dd, J = 9.0 Hz, 2.5 Hz), 6.84 (1H, d, J = 2.5 Hz), 7.45 (1H, d, J = 9.0 Hz), 7.73 (1H, d, J = 9.5 Hz), 12.45 (1H, brs).

Example 4

· Synthesis of 7- [4- (4-benzo[b]thiophen-4-yl-piperazin-l- yl)butoxy] -lH-quinolin-2-one

After 1-benzo[b] thiophen-4-yl-piperazine hydrochloride (10.6 g), potassium carbonate (5.8 g) , and DMF (50 ml) were stirred at 30 to 40°C for about 30 minutes, 7-(4-chlorobutoxy) -1H- quinolin-2-one (10.0 g) and potassium iodide (6.9 g) were added. The mixture was stirred at 90 to 100°C for 2 hours. While the temperature of the mixture was maintained at 60°C or more, water (150 ml) was added dropwise over a period of 10 minutes or more.

After the mixture was cooled to 10°C or less, the precipitated crystals were collected by filtration, and washed with water and then with ethanol.

After ethanol (325 ml) and acetic acid (25 ml) were added to the precipitated crystals, the mixture was stirred under reflux for dissolution. Concentrated hydrochloric acid (3.6 ml) was added at around 70°C, and the mixture was cooled. After confirming the precipitation of crystals, the mixture was heated again and stirred under reflux for 1 hour. After the mixture was cooled to 10°C or less, the precipitated crystals were collected by filtration and washed with ethanol.

After ethanol (191 ml) and water (127 ml) were added to the precipitated crystals, the mixture was stirred under reflux for dissolution. After activated carbon (0.89 g) was added, the mixture was stirred under reflux for 30 minutes and then hot filtered. After activated carbon was removed, the mixture was heated again for dissolution. After 25% aqueous sodium hydroxide solution (5.8 ml) was added at approximately 70°C, the mixture was stirred under reflux for 30 minutes, after which water (64 ml) was added at approximately 70°C. After the mixture was stirred at 40°C for 30 minutes, the precipitated crystals were collected by filtration at 40°C or less, then washed with water, and dried to obtain 7- [4-(4-benzo[b]thiophen-4-yl-piperazin-l-yl)butoxy] -1H- quinolin-2-one as white crystals.

Yield: 14.30 g ^-NMRfDMSO-de) 6ppm; 1.6-1.75 (2H, m) . 1.75-1.9 (2H, m) , 2.44 (2H, t, J = 7.0 Hz),2.55-2.70 (4H, m) , 3.00-3.15 (4H, m) , 4.06 (2H, t, J = 6.3 Hz), 6.30 (1H, d, J = 9.5 Hz), 6.75-6.85 (2H, m) , 6.88 (1H, d, J = 7.5 Hz), 7.27 (1H, dd, J = 8 Hz, 8 Hz), 7.40 (1H, d, J = 5.5 Hz), 7.55 (1H, d, J = 9.5 Hz), 7.61 (1H, d, J = 8 Hz), 7.69 (1H, d, J = 5.5 Hz), 7.80 (1H, d, J = 9.5 Hz), 11.58 (1H, bs) .

c

  logo

SEE  http://www.molbase.com/en/index.html

IH NMR PREDICT

7-[4-[4-(1-benzothiophen-4-yl)piperazin-1-yl]butoxy]-1H-quinolin-2-one NMR spectra analysis, Chemical CAS NO. 913611-97-9 NMR spectral analysis, 7-[4-[4-(1-benzothiophen-4-yl)piperazin-1-yl]butoxy]-1H-quinolin-2-one H-NMR spectrum

  logo

13 C NMR PREDICT

7-[4-[4-(1-benzothiophen-4-yl)piperazin-1-yl]butoxy]-1H-quinolin-2-one NMR spectra analysis, Chemical CAS NO. 913611-97-9 NMR spectral analysis, 7-[4-[4-(1-benzothiophen-4-yl)piperazin-1-yl]butoxy]-1H-quinolin-2-one C-NMR spectrum

Patent

Reaction Scheme 3:

https://patentscope.wipo.int/search/en/detail.jsf;jsessionid=D842B4D68D66F641E505E9690CF876D0.wapp2nB?docId=WO2015054976&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=FullText

 

Wherein, X is halogen, such as fluorine, chlorine, bromine, iodine; R and R 1 as defined above in the definition of the compounds of formula I the same;
Scheme 4:

Wherein, X is fluorine, chlorine, bromine or iodine; R 1 as defined above, with a compound of formula I as defined for the same;
Reaction Scheme 5:
Wherein, X is fluorine, chlorine, bromine or iodine; R 1 is the same as defined in the compounds shown above, and R are as defined for formula I. The present invention also provides processes for preparing key intermediates Bray prazosin/  Brexpiprazole or a salt thereof, the method as shown in Scheme 6:
Scheme 6:
Example 26
7- [4- (benzothiazol-4-yl-1-piperazinyl) butoxy] -3,4-dihydro -2 (1H) – quinolinone
Preparation of

The product (400mg, 0.83mmol) of Example 25 will be implemented, silver carbonate (46mg, 0.16mmol) was dissolved in DMSO (5mL) and the acetic acid was heated to 120 ℃ overnight. Cooling, water was added, extracted with ethyl acetate, ethyl acetate layer was washed with saturated sodium bicarbonate and brine each wash again, dried over anhydrous sodium sulfate, and silica gel column chromatography, to give a solid (80mg, yield 22%).
1 HNMR (400 MHz, DMSO-d 6 ): δ10.00 (s, 1H), 7.69 (d, 1H), 7.61 (d, 1H), 7.40 (d, 1H), 7.27 (t, 1H), 7.04 ( d, 1H), 6.89 (d, 1H), 6.50 (dd, 1H), 6.45 (d, 1H), 3.93 (t, 2H), 3.06 (br, 4H), 2.78 (t, 2H), 2.60 (br , 4H), 2.41 (t, 4H), 1.74 (t, 2H), 1.60 (t, 2H) ESI: [M + 1] + = 436.3.

Example 27
7- [4- (2-carboxy-benzothiophen-4-yl-1-piperazinyl) butoxy] -2 (1H) – quinolinone
Preparation of

A mixture of 2-chloro-6- (4- (4 – ((2-oxo-1,2-dihydro-quinolin-7-yl) oxy) butyl) piperazin-1-yl) benzaldehyde (80mg , 0.18mmol) was dissolved in DMF (5mL) was added sodium hydroxide (29mg, 0.73mmol) and thioglycolic acid (0.025mL, 0.36mmol), 120 ℃ stirred for 16 hours. Cooling, water was added, adjusted with 1N HCl aqueous solution is about pH = 5, extracted with ethyl acetate, the ethyl acetate layer was washed with saturated brine, dried over anhydrous sodium sulfate, and silica gel column chromatography, to give a solid (40mg, yield 46 %).
ESI: [M + 1] + = 478.0.

Piperazine hydrochloride – (2-carboxy-benzothiophen-4-yl)
Example 28 1-

The product of Example 17 (100mg, 0.25mmol) was dissolved in acetic acid (3mL) and concentrated hydrochloric acid (0.5 mL) in, 100 ℃ stirred for 10 hours. The reaction solution was poured into ice water, stirred for 10min after filtration, to obtain the target substance (38mg, 50% yield).

1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 8.04 (s, 1H), 7.69 (d, 1H), 7.43 (t, 1H), 7.00 (d, 1H), 3.30 ( bs, 8H) ESI: [M + 1] + = 262.9.
Preparation of tert-butyl piperazine-1 – Example 224- (2-carboxy-benzothiophen-4-yl)

Under nitrogen, to N, at room temperature was added N- dimethylformamide (5mL) within the reference product (200g, 0.62mmol) of Example 1, thioglycolic acid (114mg, 1.23mmol), sodium methoxide (133mg, 2.45mmol ), and the mixture was stirred at 105 ℃ 18 hours. Cooling, water was added, extracted with ethyl acetate, separated and the aqueous phase was adjusted pH = 5 or so, the precipitated solid was filtered and dried to obtain the target substance (130mg, 58% yield).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 7.98 (s, 1H), 7.64 (d, 1H), 7.42 (t, 1H), 6.95 (d, 1H), 3.53 (bs, 4H), 3.035 ( bs, 4H) ESI: [M-1] = 361.1.
Preparation of piperazine-1-carboxylic acid tert-butyl ester – (2-carboxy-benzothiophen-4-yl) Example 234-

Under nitrogen, to N, at room temperature was added N- dimethylformamide (5mL) within the reference product (200g, 0.62mmol) of Example 1, thioglycolic acid (114mg, 1.23mmol), sodium hydroxide (99mg, 2.45 mmol), the mixture was stirred at 105 ℃ 18 hours. Cooling, water was added, extracted with ethyl acetate, separated and the aqueous phase was adjusted pH = 5 or so, the precipitated solid was filtered and dried to obtain the target substance (180mg, yield 81%).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 7.98 (s, 1H), 7.64 (d, 1H), 7.42 (t, 1H), 6.95 (d, 1H), 3.53 (bs, 4H), 3.035 ( bs, 4H) ESI: [M-1] = 361.1.

Example 24 7- [4- (2-ethoxycarbonyl-4-phenyl and thienyl-1-piperazinyl) butoxy] -2 (1H) – quinolinone Preparation of

A mixture of 2-chloro-6- (4- (4 – ((2-oxo-1,2-dihydro-quinolin-7-yl) oxy) butyl) piperazin-1-yl) benzaldehyde (80mg , 0.18mmol) was dissolved in DMF (5mL) was added DIPEA (94mg, 0.73mmol) and ethyl mercaptoacetate (0.024mL, 0.22mmol), 110 ℃ stirred for 16 hours. Cooling, water was added, extracted with ethyl acetate, the ethyl acetate layer was washed with saturated brine, dried over anhydrous sodium sulfate, and silica gel column chromatography, to give a solid (40mg, 46% yield).
Preparation of piperazine-1-carboxylic acid tert-butyl ester – Example 184- (2-ethoxycarbonyl phenyl and thien-4-yl)

Under nitrogen, was added at room temperature to DMF (5mL) within the reference product (200mg, 0.62mmol) of Example 1, ethyl mercaptoacetate (0.081ml, 0.74mmol), DIPEA (342mg, 2.48mmol), the mixture was at 105 ℃ stirred for 18 hours, 1N HCl aqueous solution was added adjust pH = 7, and extracted with methyl tert-butyl ether, the ether layer was washed three times with saturated brine, dried over anhydrous sodium sulfate, the drying agent was filtered off, and concentrated by column chromatography to obtain the target (170mg, yield 71%).
1 HNMR (400 MHz, CDCl 3 ): [delta] 8.40 (s, 1H), 7.58 (d, 1H), 7.37 (t, 1H), 6.95 (d, 1H), 4.44 (q, 2H), 3.64 (m, 4H), 3.15 (m, 4H) ESI: [M + 1] + = 391.1.

Preparation of piperazine-1-carboxylic acid tert-butyl ester – Example 194- (2-ethoxycarbonyl phenyl and thien-4-yl)

Under nitrogen at room temperature was added the product of Reference Example 1 to ethanol (5mL) inside (200mg, 0.62mmol), ethyl mercaptoacetate (0.081ml, 0.74mmol), sodium hydroxide (100mg, 2.48mmol), the mixture 85 ℃ stirred for 6 hours, and concentrated by column chromatography to obtain the target substance (70mg, 30% yield).
1 HNMR (400 MHz, CDCl 3 ): [delta] 8.40 (s, 1H), 7.58 (d, 1H), 7.37 (t, 1H), 6.95 (d, 1H), 4.44 (q, 2H), 3.64 (m, 4H), 3.15 (m, 4H) ESI: [M + 1] + = 391.1.

Piperazine hydrochloride – (2-carboxy-benzothiophen-4-yl) Example 201-

The product of Example 2 (200mg, 0.55mmol), was dissolved in THF (5mL) was added concentrated hydrochloric acid (0.5mL), 50 ℃ heated 6h.Cooling, methyl tert-butyl ether (5mL), filtered to give the target (130mg, yield 79%).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 8.04 (s, 1H), 7.69 (d, 1H), 7.43 (t, 1H), 7.00 (d, 1H), 3.30 ( bs, 8H) ESI: [M + 1] + = 262.9.

Piperazine hydrochloride – (benzothiophen-4-yl) Example 211-

The product of Example 20 (130mg, 0.43mmol) was added to diphenyl ether (3mL) in, 260 ℃ heating 0.5h. Cooling, filtration object (60mg, 55% yield).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 7.75 (d, 1H), 7.69 (d, 1H), 7.53 (t, 1H), 7.31 (t, 1H), 6.97 ( t, 1H), 3.30 (bs, 8H) ESI: [M + 1] + = 219.2.

PAPER

Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.5b00027
Figure

Figure 1. Brexpiprazole (1) and intermediate 18.

Abstract Image

2-Chloro-6-fluorobenzaldehyde was converted to 4-(1-piperazinyl)benzo[b]thiophene dihydrochloride (18), an intermediate in the synthesis of brexpiprazole, via a five-step sequence in 54% overall yield. This procedure requires no expensive catalyst and avoids the side products produced in the coupling step in the reported process. Several kilograms of compound 18 were prepared using this economical and scalable process.

1-(Benzo[b]thiophen-4-yl)piperazine Dihydrochloride (18)

Compound 10 (1.5 kg, 4.71 mol) was dissolved in ………………..DELETED…………………, and then dried to give compound 18 (1.17 kg, 85% yield). HPLC for compound 18 (tR = 6.3 min, identical to authentic sample) 99.8% purity; HPLC method B.
18:
1H NMR (400 MHz, DMSO-d6) δ 11.86 (s, 1H), 9.65 (s, 2H), 7.75 (d, J = 5.5 Hz, 1H), 7.69 (d, J = 8.1 Hz, 1H), 7.53 (d, J = 5.5 Hz, 1H), 7.30 (t, J = 7.9 Hz, 1H), 6.96 (d, J = 7.6 Hz, 1H), 3.30 (s, 8H).
13C NMR (100 MHz, DMSO-d6): δ 146.92, 140.62, 133.40, 126.50, 125.06, 121.91, 117.73, 112.56, 48.52, 43.00.
MS (ESI, eV): m/z = 219.1 [M + H]+.

 ………..

PATENT

http://www.google.com/patents/US20140187782

A 4-(1-piperazinyl)benzo[b]thiophene compound represented by Formula (1):

Figure US20140187782A1-20140703-C00002

is useful for various medicines such as antipsychotic drugs. Moreover, a 4-(1-piperazinyl)benzo[b]thiophene compound represented by Formula (4):

Figure US20140187782A1-20140703-C00003

wherein R1 is a hydrogen atom or a protecting group, is useful as an intermediate for synthesizing the compound represented by Formula (1).

Reference Example 30 and Example 1 of PTL 1 specifically disclose a method for producing a benzo[b]thiophene compound (the reaction scheme shown below). In Reference Example 30, 4-(1-piperazinyl)benzo[b]thiophene is produced by heating under reflux a mixture comprising 14.4 g of 4-bromobenzo[b]thiophene, 29.8 g of anhydrous piperazine, 9.3 g of sodium tert-butoxide, 0.65 g of (R)-(+)-2,2′-bis(diphenylphosphino)-1,1′-binaphthyl (BINAP), 0.63 g of tris(dibenzylideneacetone)dipalladium (0), and 250 ml of toluene (step X).

Figure US20140187782A1-20140703-C00004

However, the reaction of the step X produces a relatively large amount of by-products that can hardly be separated, and the purity of the compound (4a) is thus inevitably reduced. Moreover, although column purification is performed to increase the purity of the compound (4a), it is very difficult to completely remove by-products, even by column chromatography purification. For this reason, there is a demand for the development of a novel method for producing the compound (4a) with high yield and high purity.

Furthermore, by-products contained in the compound (4a) inevitably reduce the purity of the compound (1) in the subsequent step Y. Since the method described in PTL 1 requires purification by column chromatography to obtain the target compound (1) with high purity, the method is not suitable for the industrial process of mass production. In addition, according to the method, incorporation of by-products that can hardly be separated is inevitable, and high-purity products usable as active pharmaceutical ingredients cannot be produced without purification by column chromatography.

CITATION LISTPatent Literature

  • PTL 1: Japanese Unexamined Patent Publication No. 2006-316052 Non Patent Literature
  • NPL 1: Tetrahedron Lett., 2004, 45, 9645

Figure US20140187782A1-20140703-C00020

Figure US20140187782A1-20140703-C00021

Figure US20140187782A1-20140703-C00022

Example 4

Synthesis of 7-[4-(4-benzo[b]thiophen-4-yl-piperazin-1-yl)butoxy]-1H-quinolin-2-one

After 1-benzo[b]thiophen-4-yl-piperazine hydrochloride (10.6 g), potassium carbonate (5.8 g), and DMF (50 ml) were stirred at 30 to 40° C. for about 30 minutes, 7-(4-chlorobutoxy)-1H-quinolin-2-one (10.0 g) and potassium iodide (6.9 g) were added. The mixture was stirred at 90 to 100° C. for 2 hours. While the temperature of the mixture was maintained at 60° C. or more, water (150 ml) was added dropwise over a period of 10 minutes or more. After the mixture was cooled to 10° C. or less, the precipitated crystals were collected by filtration, and washed with water and then with ethanol.

After ethanol (325 ml) and acetic acid (25 ml) were added to the precipitated crystals, the mixture was stirred under reflux for dissolution. Concentrated hydrochloric acid (3.6 ml) was added at around 70° C., and the mixture was cooled. After confirming the precipitation of crystals, the mixture was heated again and stirred under reflux for 1 hour. After the mixture was cooled to 10° C. or less, the precipitated crystals were collected by filtration and washed with ethanol.

After ethanol (191 ml) and water (127 ml) were added to the precipitated crystals, the mixture was stirred under reflux for dissolution. After activated carbon (0.89 g) was added, the mixture was stirred under reflux for 30 minutes and then hot filtered. After activated carbon was removed, the mixture was heated again for dissolution. After 25% aqueous sodium hydroxide solution (5.8 ml) was added at approximately 70° C., the mixture was stirred under reflux for 30 minutes, after which water (64 ml) was added at approximately 70° C. After the mixture was stirred at 40° C. for 30 minutes, the precipitated crystals were collected by filtration at 40° C. or less, then washed with water, and dried to obtain 7-[4-(4-benzo[b]thiophen-4-yl-piperazin-1-yl)butoxy]-1H-quinolin-2-one as white crystals.

Yield: 14.30 g

1H-NMR (DMSO-d6) δ ppm;

1.6-1.75 (2H, m), 1.75-1.9 (2H, m), 2.44 (2H, t, J=7.0 Hz), 2.55-2.70 (4H, m), 3.00-3.15 (4H, m), 4.06 (2H, t, J=6.3 Hz), 6.30 (1H, d, J=9.5 Hz), 6.75-6.85 (2H, m), 6.88 (1H, d, J=7.5 Hz), 7.27 (1H, dd, J=8 Hz, 8 Hz), 7.40 (1H, d, J=5.5 Hz), 7.55 (1H, d, J=9.5 Hz), 7.61 (1H, d, J=8 Hz), 7.69 (1H, d, J=5.5 Hz), 7.80 (1H, d, J=9.5 Hz), 11.58 (1H, bs).

………………………

PATENT

http://www.google.com/patents/WO2006112464A1?cl=en

Example 1

Preparation of 7- [4- (4-benzo [b] thiophen-4-yl- piperazin-1-yl) butoxy] -lH-quinolin-2-one

A mixture of 9.0 g of 7- ( 4-chlorobutoxy) -IH- quinolin-2-one, 10 g of 1-benzo [b] thiophene-4-yl- piperazine hydrochloride, 14 g of potassium carbonate, 6 g of sodium iodide and 90 ml of dimethylformamide was stirred for 2 hours at 😯0C. Water was added to the reaction solution and precipitated crystals were separated by filtration. The crystals were dissolved in a mixed solvent of dichloromethane and methanol, dried over magnesium sulfate, and the solvent was evaporated under reduced pressure. The residue was purified by silica gel column chromatography (dichloromethane .-methanol = 100:3). Recrystallized from ethanol, 13.6 g of 7- [4- (4-benzo [b] thiophen-4-yl- piperazin-1-yl) butoxy] -lH-quinolin-2-one in the form of a white powder was obtained.

Melting point 183.5-184.50C

1H-NMR ( DMSO-dg) δppm:

1.6-1.75 (2H, m) , 1.75-1.9(2H, m) , 2.44(2H, t, J=7Hz) , 2.5-2.8(4H, m) , 2.9-3.2(4H, m) , 4.06(2H, t, J=6.5Hz), 6.3O(1H, d, J=9.5Hz), 6.75-6.85 (2H, m) , 6.88(1H, d, J=7.5Hz), 7.27 (IH, dd, J=8Hz, 8Hz), 7.40 (IH, d, J=5.5Hz), 7.55 (IH, d, J=9.5Hz), 7.61(1H, d, J=8Hz) , 7.69(1H, d, J=5.5Hz), 7.8O(1H, d, J=9.5Hz), 11.59(1H, bs) .

……………….

PATENT

Figure imgf000006_0001 7- [ 4- ( 4-benzo[b]thiophen-4- yl-piperazin-l-yl)butoxy] -lH-quinolin-2-one

The dihydrate of the benzothiophene compound represented by Formula (I) or of a salt thereof according to the present invention can be produced from an anhydride of the benzothiophene compound or of a salt thereof.

The benzothiophene compound (in the form of an

anhydride) of Formula (I), from which the dihydrate of the present invention is produced, is a known compound, and can be obtained by the production method disclosed in Example 1 of

JP2006-316052A or according to Reference Examples 1 and 2

Fig. 1 shows the ^-NMR spectrum of the dihydrate of the benzothiophene compound represented by Formula (I) prepared in Example 1.

Fig. 2 shows the X-ray powder diffraction pattern of the dihydrate of the benzothiophene compound represented by

Formula (I) prepared in Example 1.

Fig. 3 shows the infrared absorption spectrum of the dihydrate of the benzothiophene compound represented by Formula (I) prepared in Example 1.

Fig. 4 shows the Raman spectrum of the dihydrate of the benzothiophene compound represented by Formula (I) prepared in Example 1.

Fig. 5 shows the XH- MR spectrum of the benzothiophene compound represented by Formula (I) prepared in Example 2.

Reference Example 1: Synthesis of 7-(4-chlorobutoxy)-lH-quinolin- 2-one Methanol (149 L) , 7-hydroxy-lH-quinolin-2-one (14.87 kg), and potassium hydroxide (6.21 kg) were mixed and stirred. After dissolution, l-bromo-4-chlorobutane (47.46 kg) was further added thereto and the resulting mixture was stirred under reflux for seven hours. Thereafter, the mixture was stirred at 10° C for one hour. The precipitated crystal was centrifuged and washed with methanol (15 L). The wet crystal was collected and placed in a tank. Water (149 L) was added thereto, followed by stirring at room temperature. After centrifugation, the resulting solid was washed with water (30 L). The wet crystal was collected and placed in a tank. After adding methanol (74 L), the mixture was stirred under reflux for one hour, cooled to 10° C, and then stirred. The precipitated crystal was centrifuged and washed with methanol (15 L). The separated crystal was dried at 60° C to obtain 7- (4-chlorobutoxy) -lH-quinolin-2-one (15.07 kg).

Reference Example 2: Synthesis of 7- [ 4- ( 4-benzo[b] thiophen-4-yl- piperazin-l-yl)butoxy] -lH-quinolin-2-one

Water (20 L), potassium carbonate (1.84 kg), 1- benzo[b] thiophen-4-yl-piperazine hydrochloride (3.12 kg), and ethanol (8 L) were mixed and stirred at 50° C. 7- ( 4-Chlorobutoxy) – lH-quinolin-2-one (2.80 kg) obtained in Reference Example 1 was added to the mixture and stirred under reflux for nine hours.

After concentrating the solvent (8 L) under ordinary pressure, the mixture was stirred at 90° C for one hour and then cooled to 9° C . The precipitated crystal was centrifuged and then

sequentially washed with water (8 L) and ethanol (6 L). The separated crystal was dried at 60° C to obtain a crude product. The crude product (4.82 kg) and ethanol (96 L) were mixed in a reaction vessel, and acetic acid (4.8 L) was introduced into the reaction vessel. The mixture was stirred under reflux for one hour to dissolve the crude product. After introducing

hydrochloric acid (1.29 kg), the mixture was cooled to 10° C. The mixture was heated again, refluxed for one hour, and cooled to 7° C. The precipitated crystal was centrifuged and washed with ethanol (4.8 L). The separated crystal was dried at 60° C to obtain 7- [4- (4-benzo[b] thiophen-4-yl-piperazin-l-yl)butoxy] -1H- quinolin-2-one hydrochloride (5.09 kg). The resulting 7- [4- (4- benzo[b]thiophen-4-yl-piperazin-1-yl)butoxy] -lH-quinolin-2-one hydrochloride (5.00 kg), ethanol (45 L), and water (30 L) were mixed in a reaction vessel. The mixture was stirred under reflux to dissolve the 7-[4-(4-benzo[b]thiophen-4-yl-piperazin-l- yl)butoxy] -lH-quinolin-2-one hydrochloride. Activated carbon (500 g) and water (5 L) were added thereto, and an activated carbon treatment was conducted under reflux for 30 minutes. After performing hot filtration, a solution containing sodium hydroxide (511 g) dissolved in water (1.5 L) was flowed into the reaction vessel while stirring the filtrate under reflux. After stirring under reflux for 30 minutes, water (10 L) was introduced thereto and the mixture was cooled to approximately 40° C. The

precipitated crystal was centrifuged and washed with water (125 L). The separated crystal was dried at 80° C to obtain 7- [4- (4- benzo[b]thiophen-4-yl-piperazin-1-yl)butoxy] – lH-quinolin-2-one (3.76 kg).

Example 1: Preparation of 7- [ 4- ( 4-benzo[b]thiophen-4-yl- piperazin-l-yl)butoxy] -lH-quinolin-2-one dihydrate

The 7- [4- (4-benzo[b] thiophen-4-yl-piperazin-1- yl)butoxy] -lH-quinolin-2-one (3.2 kg) obtained in Reference

Example 2, ethanol (64 L) , water (74 L) , and acetic acid (1.77 kg) were mixed in a reaction vessel to prepare an acidic liquid mixture. The mixture was stirred under reflux to dissolve the 7- [ 4- ( 4-benzo[b] thiophen-4-yl-piperazin-1-yl)butoxy] -1H-quinolin-2- one (reflux temperature: 84° C). After cooling to -5°C, the solution obtained above was introduced, over a period of 30 minutes, into a solution containing 25% sodium hydroxide (5.9 kg) and water (54 L) that was cooled to 0°C, to prepare a liquid mixture with pHlO. After being stirred at 5° C or below for one hour, the mixture was heated to 20 to 30° C and further stirred for-seven hours . The precipitated crystal was filtered and washing with water (320 L) was performed, until alkali in the solid component disappeared (i.e.. until the pH value of the filtrate became 7 ) . The solid component was then air-dried until its weight became constant to obtain a white solid 7-[4-(4- benzofb] thiophen-4-yl-piperazin-l-yl)butoxy] -lH-quinolin-2-one dihydrate (unground, 3.21 kg).

Fig. 1 shows the XH-NMR spectrum (D SO-d6, TMS) of the dihydrate prepared by the aforesaid method. As shown in Fig. 1, in the ^- MR spectrum (DMSO-d6, TMS) , peaks were observed at 1.64 ppm (tt, J = 7.4 Hz, J = 7.4 Hz, 2H) , 1.80 ppm (tt, J = 7.0 Hz, J = 7.0 Hz, 2H), 2.44 ppm (t, J = 7.5 Hz, 2H) , 2.62 ppm (br, 4H) , 3.06 ppm (br, 4H) , 3.32 ppm (s, 4H + H20) , 4.06 ppm (t, J = 6.5 Hz, 2H), 6.29 ppm (d, J = 9.5 Hz, 1H), 6.80 ppm (d, J = 2.5 Hz, 1H) , 6.80 ppm (dd, J = 2.5 Hz, J = 9.0 Hz, 1H) , 6.88 ppm (d, J = 7.5 Hz, 1H), 7.27 ppm (dd, J = 7.8 Hz, J = 7.8 Hz, 1H) , 7.40 ppm (dd, J = 0.5 Hz, J = 5.5 Hz, 1H), 7.55 ppm (d, J = 9.0 Hz, 1H) , 7.61 ppm (d, J = 8.0 Hz, 1H) , 7.69 ppm (d, J = 5.5 Hz, 1H) , 7.80 ppm (d, J = 9.5 Hz, 1H), and 11.57 ppm (s, 1H) .

The X-ray powder diffraction spectrum of the dihydrate prepared by the aforesaid method was measured using an X-ray diffractometer (D8 ADVANCE, available from Bruker AXS). Fig. 2 shows the X-ray powder diffraction spectrum. As shown in Fig. 2, in the X-ray powder diffraction spectrum, diffraction peaks were observed at 2Θ = 8.1° , 8.9° , 15.1° , 15.6° , and 24.4° . Other than those mentioned above, the diffraction peaks were also observed at 2Θ = 11.6°.. 12.2°, 14.0°, 16.3°, 18.1°, 18.4°, 18.9°, 19.5°, 20.5°, 21.5°, 22.6°, 23.3°, 25.0°, 26.1°, 26.4°, 27.1°. 28.1°, 28.5°, 28.9°, 29.8°, 30.4°, 30.7°, 31.6°, 32.9°, 33.9°, 34.4°, 35.2°, 36.0°, 36.7°, 37.4° , and 38.3°.

The IR (KBr) spectrum of the dihydrate prepared by the aforesaid method was measured. Fig. 3 shows the IR (KBr) spectrum. As shown in Fig. 3, in the IR (KBr) spectrum, absorption bands were observed in the vicinity of wavenumbers 3509 cm“1, 2934 cm“1, 2812 cm“1, 1651 cm“1, 1626 cm“1, 1447 cm“1, 1223 cm“1 and 839 cm“1.

The Raman spectrum of the dihydrate prepared by the aforesaid method was measured. Fig. 4 shows the Raman spectrum. As shown in Fig. 4, in the Raman spectrum, absorption bands were observed in the vicinity of wavenumbers 1497 cm“1, 1376 cm“1, 1323 cm“1, 1311 cm“1, 1287 cm“1, 1223 cm“1, and 781 cm“1.

Other than those mentioned above, absorption was also observed in the vicinity of wavenumbers 1656 cm“1, 1613 cm“1, 1563 cm“1, 1512 cm“1, 1468 cm“1, 1446 cm“1, 1241 cm“1, 1203 cm“1, 1145 cm“1, 1096 cm“1, 1070 cm“1, 971 cm“1, and 822 cm“1.

The water content of the dihydrate prepared by the aforesaid method was measured using a moisture meter (CA-100, available from Mitsubishi Chemical Analytech Co., Ltd.) by the Karl Fischer method. As a result, the dihydrate had a water content of 7.79% by weight.

Example 2; Preparation of finely ground dihydrate

Dihydrate crystal (2.73 kg) obtained in Example 1 was ground using a jet mill. Here, the air pressure was set at 5 kgf/cm2, and the rotational speed of the feeder was set at 20 rpm. As a result, finely ground 7-[4-(4-benzo[b]thiophen-4-yl- piperazin-1-yl)butoxy] -1H-quinoli -2-one dihydrate (2.61 kg,

95.6%) was obtained.

The dihydrate (finely ground product) thus obtained had a mean particle diameter of 5.5 um. The mean particle diameter was measured using a Microtrack HRA, manufactured by Nikkiso Co., Ltd.

Fig. 5 shows the ^-NMR spectrum (DMSO-d6, TMS) of the dihydrate prepared by the above method. As shown in Fig. 5, in the ^- MR spectrum (DMSO-d6, TMS), peaks were observed at 1.64 ppm (tt, J = 7.3 Hz, J = 7.3 Hz, 2H), 1.80 ppm (tt, J = 6.9 Hz, J = 6.9 Hz, 2H), 2.44 ppm (t, J = 7.3 Hz, 2H) , 2.62 ppm (br, 4H) , 3.06 ppm (br, 4H) , 3.32 ppm (s, 4H + H20) , 4.06 ppm (t, J = 6.5 Hz, 2H), 6.29 ppm (d, J = 9.5 Hz, 1H) , 6.80 ppm (d, J = 2.5 Hz , 1H) , 6.80 ppm (dd, J = 2.3 Hz, J = 9.3 Hz, 1H) , 6.88 ppm (d, J = 7.5 Hz, 1H), 7.27 ppm (dd, J = 8.0 Hz, J = 8.0 Hz, 1H) , 7.40 ppm (d, J = 5.5 Hz, 1H), 7.55 ppm (d, J = 9.5 Hz , 1H) , 7.61 ppm (d, J = 8.0 Hz, 1H), 7.69 ppm (d, J = 5.5 Hz, 1H) , 7.80 ppm (d, J = 9.5

Hz, 1H), and 11.57 ppm (s, 1H) .

The X-ray powder diffraction spectrum of the dihydrate prepared by the aforesaid method was measured in the same manner as in Example 1. Fig. 6 shows the X-ray powder diffraction spectrum. As shown in Fig. 6, in the X-ray powder diffraction spectrum, diffraction peaks were observed at 2Θ = 8.2° , 8.9° ,

15.2° , 15.7° and 24.4° .

Other than those mentioned above, the diffraction peaks were also observed at 2Θ = 6.8°, 12.2°, 14.0°, 14.5″, 17.4°,

18.1°, 18.5°, 19.0°, 19.2°, 19.6°, 20.3°, 20.6°, 21.5°, 22.7°,

23.4°, 25.0°, 26.1°, 27.1°, 28.6°, 29.0°, 30.4°, 34.0°, 34.5°,

35.3° , and 36.7° .

The IR (KBr) spectrum of the dihydrate prepared by the aforesaid method was measured in the same manner as in Example 1.

Fig. 7 shows the IR (KBr) spectrum. As shown in Fig. 7, in the IR

(KBr) spectrum, absorption bands were observed in the vicinity of wavenumbers 3507 cm“1, 2936 cm“1, 2812 cm“1, 1651 cm“1, 1626 cm“1,

1447 cm“1, 1223 cm“1 and 839 cm“1.

The Raman spectrum of the dihydrate prepared by the aforesaid method was measured. Fig. 8 shows the Raman spectrum.

As shown in Fig. 8, in the Raman spectrum, absorption bands were observed in the vicinity of wavenumbers 1496 cm‘1, 1376 cm“1, 1323 cm‘1, 1311 cm“1, 1286 cm“1, 1223 cm“1, and 781cm“1.

Other than those mentioned above, absorption was also observed in the vicinity of wavenumbers 1656 cm“1, 1614 cm“1, 1563 cm“1, 1512 cm“1, 1467 cm“1, 1446 cm“1, 1241 cm“1, 1203 cm“1, 1145 cm“1,

1095 cm“1, 1069 cm“1, 971 cm“1, and 822 cm“1.

The water content of the dihydrate prepared by the aforesaid method was measured using a moisture meter (CA-100, available from Mitsubishi Chemical Analytech Co., Ltd.) by the

Karl Fischer method. As a result, the dihydrate had a water content of 6.74% by weight . Example 3 : Preparation of 7- [ 4- ( 4-benzo[b] thiophen-4-yl- piperazin-l-yl)butoxy] -lH-quinolin-2-one dihydrate

7- [ 4- ( 4-Benzo[ ] thiophen-4-yl-piperazin-1-yl)butoxy] – lH-quinolin-2-one (5.0 kg), ethanol (100 L), water (115 L), and DL-lactic acid (2.29 kg) were mixed to prepare an acidic liquid mixture. The liquid mixture was stirred under reflux to dissolve the 7- [4- (4-benzo[b] thiophen-4-yl-piperazin-l-yl)butoxy] -1H- quinolin-2-one (reflux temperature: 82° C). After cooling to -5°C, the solution obtained above was introduced, over a period of about 15 minutes, into a solution containing sodium hydroxide (1.48 kg) and water (135 L) that was cooled to 1°C, to prepare a liquid mixture with pHll. After being stirred at approximately 2 to 5° C for three hours, the mixture was heated to 45° C and

further stirred at 45 to 50° C for two hours. The precipitated crystal was filtered and washing with water (200 L) was performed until alkali in the solid component disappeared (i.e., until the pH value of the filtrate became 7). The solid component was further washed with a liquid mixture of ethanol (15 L) and water (20 L). The solid component was then dried at room temperature until its weight became constant to obtain a white solid 7- [4- (4- benzo[b] thiophen-4-yl-piperazin-1-yl)butoxy] -1H-quinolin-2-one dihydrate (unground, 5.11 kg).

The dihydrate thus obtained was the same as that obtained in Example 1.

The Raman spectrum of the dihydrate prepared by the aforesaid method was measured. Fig. 9 shows the Raman spectrum. As shown in Fig. 9, in the Raman spectrum, absorption bands were observed in the vicinity of wavenumbers 1497 cm“1, 1376 cm“1, 1323 cm“1, 1311 cm“1, 1287 cm“1, 1223 cm“1, and 782 cm“1.

Other than those mentioned above, absorption was also observed in the vicinity of wavenumbers 1656 cm“1, 1614 cm“1, 1563 cm“1, 1512 cm“1, 1468 cm“1, 1446 cm“1, 1241 cm“1, 1203 cm“1, 1145 cm“1, 1126 cm“1, 1096 cm“1, 1070 cm“1, 972 cm“1, and 822 cm“1.

…………………….

PATENT

http://www.google.com/patents/WO2006112464A1?cl=en

…………………..

PATENT

http://www.google.com/patents/US20110152286

References

  1.  “Phase II and Phase III Drugs in U.S. Development for Depression, Anxiety, Sleep Disorders, Psychosis, & ADHD”. Retrieved 9 February 2012.
  2.  “Otsuka Pharmaceutical Development & Commercialization, Inc.”. Bloomberg Businessweek. Retrieved 10 February 2012.
  3.  “Otsuka HD places top priority on development of OPC-34712.”. Chemical Business Newsbase. January 3, 2011. Retrieved 10 February 2012.
  4. Patent 5006528, Oshiro, Yasuo; Seiji Sato & Nobuyuki Kurahashi, “Carbostyril derivatives”, published October 20, 1989
  5. “Patent and Exclusivity Search Results”. Electronic Orange Book. US Food and Drug Administration. Retrieved 8 December 2008.
  6.  “Lundbeck and Otsuka Pharmaceutical sign historic agreement to deliver innovative medicines targeting psychiatric disorders worldwide”. Lundbeck. Retrieved 10 February2012.
  7.  “Otsuka Holdings Financial Results Presentation Q2 FY2011”. Retrieved 10 February2012.
  8.  “OPC-34712 search results”. Retrieved 10 February 2012.
  9.  “Study of the Safety and Efficacy of OPC-34712 as Adjunctive Therapy in the Treatment of Patients With Major”. Retrieved 15 February 2012.
  10.  “Study of the Safety and Efficacy of Two Fixed Doses of OPC-34712 as Adjunctive Therapy in the Treatment of Adults With Major Depressive Disorder (the Polaris Trial)”. Retrieved 10 February 2012.
  11. ^ Jump up to:a b “Study of the Safety and Efficacy of OPC-34712 as a Complementary Therapy in the Treatment of Adult Attention Deficit/Hyperactivity Disorder (STEP-A)”. Retrieved10 February 2012.
  12.  “Trial to Evaluate the Effects of OPC-34712 on QT/QTc in Subjects With Schizophrenia or Schizoaffective Disorder”. Retrieved 10 February 2012.
  13.  “A Dose-finding Trial of OPC-34712 in Patients With Schizophrenia”. Retrieved10 February 2012.
  14.  “Efficacy Study of OPC-34712 in Adults With Acute Schizophrenia (BEACON)”. Retrieved 10 February 2012.
  15. Jump up^ “Safety and Tolerability Study of Oral OPC-34712 as Maintenance Treatment in Adults With Schizophrenia (ZENITH)”. Retrieved 10 February 2012.
  16.  “Study of the Effectiveness of Three Different Doses of OPC-34712 in the Treatment of Adults With Acute Schizophrenia (VECTOR)”. Retrieved 10 February 2012.
  17.  “A Long-term Trial of OPC-34712 in Patients With Schizophrenia”. Retrieved10 February 2012.
  18.  “Otsuka Pharmaceutical Co., Ltd. Announces Results from a Phase 2 Study of Investigational Product OPC-34712 as Adjunctive Therapy in Adults with Major Depressive Disorder”. Retrieved 16 February 2012.
  19.  “Preclinical Pharmacology of Brexpiprazole (Opc-34712): A Novel Compound with Dopamine D2 Receptor Partial Agonist Activity”. Retrieved 16 February 2012.
  20.  “2011 U.S. Psych Congress Poster Session Abstracts”. Retrieved 16 February 2012.
  21.  “Otsuka Pharmaceutical reports OPC-34712 Phase 2 trial results in major depressive disorder”. News-Medical.Net. Retrieved 10 February 2012.
  22.  Maeda K, Sugino H, Akazawa H et al. (September 2014). “Brexpiprazole I: in vitro and in vivo characterization of a novel serotonin-dopamine activity modulator”. J. Pharmacol. Exp. Ther. 350 (3): 589–604. doi:10.1124/jpet.114.213793.PMID 24947465.
  23. “Trial to Evaluate the Effects of OPC-34712 on QT/QTc in Subjects With Schizophrenia or Schizoaffective Disorder”. Retrieved 10 February 2012.
  24.  “Canadian Patents Database 2620688”. Retrieved 16 February 2012.
  25. http://zliming2004.blog.163.com/blog/static/6456372120149963644472/

 ………

JP2006316052A Title not available
US20110152286 * Dec 16, 2010 Jun 23, 2011 Otsuka Pharmaceutical Co., Ltd. Piperazine-substituted benzothiophenes for treatment of mental disorders
 UPDATED
SUZHOU VIGONVITA LIFE SCIENCES CO., LTD. [CN/CN]; 398, Ruoshui Road, Suzhou Industrial Park Suzhou, Jiangsu 215123 (CN).
TOPHARMAN SHANGHAI CO., LTD. [CN/CN]; 1088, Chuansha Road, Pudong Shanghai 201209 (CN).
SHANGHAI INSTITUTE OF MATERIA MEDICA, CHINESE ACADEMY OF SCIENCES [CN/CN]; 555, Zuchongzhi Road, Zhangjiang, Pudong Shanghai 201203 (CN)
(EN)The present invention relates to methods of preparing brexpiprazole, analogs thereof, key intermediates and salts thereof. Specifically, the present invention relates to new methods of preparing brexpiprazole, analogs thereof, key intermediates and salts thereof, and to the key intermediates and salts thereof used in the methods. The methods involve mild reaction conditions, stable intermediates, easy operations, and widely available reagents, thereby allowing for reduced synthesis costs, a shortened production cycle, high yield, and high product quality. The methods are suited for use in large-scale production.
Bray prazosin (Brexpiprazole, code: OPC-34712) is Otsuka Pharmaceutical Co., Ltd. developed a new generation of anti-psychotic drug candidates that act on more than one receptor, dopamine D2 receptor partial agonist (improving positive and negative symptoms, cognitive impairment and depressive symptoms), 5-HT2A receptor antagonist (improving negative symptoms, cognitive dysfunction, symptoms of depression, insomnia), α1 adrenoceptor antagonists (improving positive symptoms of schizophrenia), 5 – serotonin uptake / reuptake inhibitors (symptoms of depression); at the same time, but also a 5-HT1A partial agonist (anxiolytic and antidepressant activity) and 5-HT7 antagonist (temperature, circadian rhythms, learning and memory, sleep) . Currently, in the United States and Europe as an adjunctive treatment of severe depression (MDD) Phase III clinical trials; III clinical trials the treatment of schizophrenia in the United States, Europe and Japan, meanwhile, is still the United States II Adult ADHD clinical trials.
Otsuka Pharmaceutical Co., Ltd. are disclosed in PCT Application WO2006112464A1 in the preparation route Bray prazosin, see Scheme 1, the difficulty of the route is the first step in the reaction by-products easily separated by column chromatography is not easy to obtain high-purity intermediates, thus affecting the final product Bray prazosin purity and yield.
Scheme 1:
Subsequently, Otsuka Pharmaceutical Co., Ltd. are disclosed in PCT Application WO2013015456A1 in the alternative method of preparing the reaction of this step, see Scheme 2, along the route of the reagents are more expensive, high-cost, environmentally unfriendly and not suitable for industrial production.
Reaction Scheme 2:
Due to the above production process there is a high cost, and difficult to separate impurities and other shortcomings, it is necessary to find an economical, practical, environmental protection, new routes to improve process stability, reduce costs, improve product quality.
DISCLOSURE
In response to these deficiencies, an object of the present invention is to provide a new, simple operation, high yield, low cost, environmentally friendly and suitable for industrial mass production Bray prazosin and the like, key intermediates and preparing a salt thereof.
Another object of the present invention is to provide novel compounds and salts thereof of the manufacturing process.
To achieve the above objects, the present invention provides compounds of formula I, the structure is as follows:
Wherein, R is C1 ~ C6 straight or branched chain alkyl, benzyl; preferably, R is C1 ~ C4 straight or branched chain alkyl group; most preferably, R is methyl, ethyl, t-butyl group;
R 1 is acyl amino-protecting groups (e.g. formyl ( ), an acetyl group, a propionyl group, a benzoyl group, haloacetyl group, phthaloyl) or class alkoxycarbonyl amino-protecting group (e.g. t-butoxycarbonyl , benzyloxycarbonyl, 9-fluorenyl methoxy carbonyl); said haloacetyl group is a fluorinated acetyl, bromoacetyl, chloroacetyl or iodoacetyl group; preferably, R 1 is formyl, acetyl and tert-butoxycarbonyl groups;
The present invention also provides a method for preparing a compound of formula I, the compound of formula II with a thioglycolate as shown in the reaction, the compound, the method shown in Formula I as shown in Scheme 3,
Reaction Scheme 3:
Wherein, X is halogen, such as fluorine, chlorine, bromine, iodine; R and R 1 are as defined above, the compound of formula I as defined the same;
The above reaction in the presence of a base, in particular, is an inorganic base (e.g. sodium hydroxide, potassium hydroxide, strontium hydroxide, lithium hydroxide, barium hydroxide, calcium hydroxide, cesium hydroxide, sodium hydrogen carbonate, potassium bicarbonate, potassium carbonate, sodium carbonate, strontium carbonate, cesium carbonate, sodium sulfide, sodium hydroxide, etc.) or an organic base (e.g. sodium alkoxide, potassium alkoxide, butyl lithium, 1,8-diazabicyclo [5,4 0] undecene -7 (DBU), pyridine, quinoline, 4-dimethylaminopyridine (DMAP) or an organic amine, etc.) performed in the presence of, wherein the sodium alkoxide may be sodium methoxide, sodium ethoxide, propoxy sodium alkoxide, sodium isopropoxide, n-butoxide, sodium tert-butoxide and the like; may be the potassium alkoxide, potassium methoxide, potassium ethoxide, potassium-propanol, potassium isopropoxide, n-butoxide, potassium tert-butoxide , the organic amine may be triethylamine, diethylamine, n-butylamine, tripropylamine, diisopropylamine, diisopropylethylamine, etc., preferably, the base may be an inorganic alkali sodium hydroxide, potassium hydroxide, lithium hydroxide, sodium hydrogencarbonate, potassium hydrogencarbonate, potassium carbonate, sodium carbonate, strontium carbonate, sodium sulfide, sodium hydroxide, or organic bases as sodium methoxide, sodium ethoxide, tert-butoxide potassium, triethylamine, diethylamine, diisopropylamine or diisopropylethylamine;
The above reaction is carried out in a suitable solvent, the solvent is water, C 1 ~ C 5 lower alcohol (such as methanol, ethanol, n-propanol, isopropanol, n-butanol, isobutanol, tert-butanol, n-amyl alcohol, amyl alcohol, ethylene glycol, propylene glycol, glycerol), N, N- dimethylformamide (DMF), dimethyl sulfoxide (DMSO), tetrahydrofuran (THF), acetonitrile, dioxane, N- methylpyrrolidone, methylene chloride, chloroform, ethylene glycol dimethyl ether, diethylene glycol dimethyl ether or ethylene glycol monomethyl ether, and the like, one or more, preferably, the solvent is water , methanol, ethanol, N, N- dimethylformamide (DMF), dimethylsulfoxide (DMSO), tetrahydrofuran (THF), acetonitrile, dioxane or ethylene glycol dimethyl ether or a species; the reaction time from 1 hour to 24 hours, preferably 2 hours to 12 hours. The reaction temperature is 0 ℃ ~ 150 ℃, preferably room temperature ~ 100 ℃.
To achieve the above object, the present invention also provides a compound of formula III, is structured as follows:
Wherein, R 1 is acyl amino protecting groups (e.g. formyl, acetyl, propionyl, benzoyl, halo acetyl, phthaloyl) or class alkoxycarbonyl amino-protecting group (e.g. t-butoxycarbonyl , benzyloxycarbonyl, 9-fluorenyl methoxycarbonyl), said haloacetyl group is a fluorinated acetyl, bromoacetyl, chloroacetyl or iodoacetyl;
Preferably, R 1 is formyl, acetyl or t-butoxycarbonyl;
The present invention also provides a method of preparing compounds of Formula III are shown, thioglycolic acid compound and reacting compound of formula III as shown in the method shown by the formula II as shown in Scheme 4,
Scheme 4:
Wherein, X is fluorine, chlorine, bromine or iodine; R 1 as defined above, with a compound of formula I as defined the same;
Bray prazosin (Brexpiprazole, code: OPC-34712) is Otsuka Pharmaceutical Co., Ltd. developed a new generation of anti-psychotic drug candidates that act on more than one receptor, dopamine D2 receptor partial agonist (improving positive and negative symptoms, cognitive impairment and depressive symptoms), 5-HT2A receptor antagonist (improving negative symptoms, cognitive dysfunction, symptoms of depression, insomnia), α1 adrenoceptor antagonists (improving positive symptoms of schizophrenia), 5 – serotonin uptake / reuptake inhibitors (symptoms of depression); at the same time, but also a 5-HT1A partial agonist (anxiolytic and antidepressant activity) and 5-HT7 antagonist (temperature, circadian rhythms, learning and memory, sleep) . Currently, in the United States and Europe as an adjunctive treatment of severe depression (MDD) Phase III clinical trials; III clinical trials the treatment of schizophrenia in the United States, Europe and Japan, meanwhile, is still the United States II Adult ADHD clinical trials.
Scheme 5:

 

Wherein, X is fluorine, chlorine, bromine or iodine; R 1 are the same as defined in the compounds illustrated and R are as defined above for formula I. The present invention also provides processes for preparing key intermediates Bray prazosin or a salt thereof, the method as shown in Scheme 6:
Scheme 6:
14- (3-chloro-2-carboxaldehyde-phenyl-1 -) – Reference Example Synthesis of piperazine-1-carboxylic acid tert-butyl ester
A mixture of 2-chloro-6-fluorobenzaldehyde (500mg, 3.15mmol), piperazine-1-carboxylate (646mg, 3.47mmol) was dissolved in N, N- dimethylformamide (5mL), and nitrogen at, at room temperature was added potassium carbonate (2.18g, 15.77mmol), the mixture was stirred for 4 hours at 80 ℃, cooled and filtered, water (20mL), ethyl acetate (3 × 5mL) was extracted, dried over anhydrous sodium sulfate, filtered The desiccant was concentrated to give a solid, after with petroleum ether (50mL) beating 1h, filtered to give a pale yellow solid (750mg, 75% yield).
1 HNMR (400 MHz, CDCl 3 ): δ10.37 (s, 1H), 7.40 (t, 1H), 7.01 (d, 1H), 6.99 (d, 1H), 3.20 (m, 4H), 3.00 (s, 4H), 1.47 (s, 9H) ESI: [M + 1] + = 325.8.
Reference Example 21- carboxylic acid (3-chloro-2-carboxaldehyde-phenyl-1 -) – -4- piperazine

 

A mixture of 2-chloro-6-fluorobenzaldehyde (500mg, 3.15mmol), 1- formyl piperazine (396mg, 3.47mmol) was dissolved in DMF (5mL), and under nitrogen at room temperature was added potassium carbonate (2.18g, 15.77mmol). The mixture was stirred for 4 hours at 80 ℃, cooled water (20mL), ethyl acetate (3 × 5mL) was extracted, dried over anhydrous sodium sulfate, and concentrated to give a solid with petroleum ether (50mL) After beating 1h, filtered to give a pale yellow solid (588mg, yield 70%).
1 HNMR (400 MHz, CDCl 3 ): [delta] 10.45 (s, 1H), 8.13 (s, 1H), 7.44 (t, 1H), 7.18 (d, 1H), 7.02 (d, 1H), 3.80 (s, 2H), 36.4 (s, 2H), 3.10 (m, 4H) ESI: [M + 1] + = 253.1.
Acetyl-31- (3-chloro-2-carboxaldehyde-phenyl-1 -) – 4- Reference piperazine
A mixture of 2-chloro-6-fluorobenzaldehyde (500mg, 3.15mmol), 1- acetyl-piperazine (444mg, 3.47mmol) was dissolved in DMF (5mL), and under nitrogen at room temperature was added potassium carbonate (2.18g, 15.77 mmol), the mixture was stirred at 80 ℃ 4 hours, cooled and filtered, water (20mL), ethyl acetate (3 × 5mL) was extracted, dried over anhydrous sodium sulfate, and concentrated to give a solid, after with petroleum ether (50mL) beating 1h, filtered to give a pale yellow solid (588mg, yield 70%).
1 HNMR (400 MHz, CDCl 3 ): [delta] 10.44 (s, 1H), 7.44 (t, 1H), 7.17 (d, 1H), 7.03 (d, 1H), 3.79 (bs, 4H), 3.10 (m, 4H), 2.18 (s, 3H) ESI: [M + 1] + = 267.1.
Piperazine-1-carboxylic acid tert-butyl ester – 14- (2-ethoxycarbonyl phenyl and thien-4-yl) Example
Under nitrogen, to N, was added the product (1.0g, 3.08mmol) of Reference Example 1 N- dimethylformamide (5mL) at room temperature within, ethyl thioglycolate (388mg, 3.20mmol), potassium carbonate (1.38 g, 10mmol), the mixture was stirred for 4 hours at 80 ℃, cooled and filtered, water (20mL), ethyl acetate (3x5mL) was extracted, dried over anhydrous sodium sulfate, the drying agent filtered, and concentrated to give a solid with petroleum ether (50mL ) after beating 1h, filtered to give a pale yellow solid (900mg, 75% yield).
1 HNMR (400 MHz, CDCl 3 ): [delta] 8.40 (s, 1H), 7.58 (d, 1H), 7.37 (t, 1H), 6.95 (d, 1H), 4.44 (q, 2H), 3.64 (m, 4H), 3.15 (m, 4H) ESI: [M + 1] + = 391.1.
Example 24- (2-carboxy-benzothiophen-4-yl) – piperazine-1-carboxylate Synthesis of
Of the product (1.0g, 2.5mmol) in Example 1 was dissolved into 1,4-dioxane (5mL), was added 4N aqueous sodium hydroxide solution (1.8mL, 7.2mmol), the mixture was stirred for 3h at 80 ℃, cooled to room temperature, water (5mL) and ethyl acetate (10mL), separated and the aqueous phase with 1N HCl at 0 ℃ pH was adjusted to about 4.0, the resulting solid was filtered, dried to give a pale yellow solid.
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 7.98 (s, 1H), 7.64 (d, 1H), 7.42 (t, 1H), 6.95 (d, 1H), 3.53 (bs, 4H), 3.035 ( bs, 4H) ESI: [M-1] = 361.1.
34- (benzothiophen-4-yl) Example – Synthesis of piperazine-1-carboxylic acid tert-butyl ester
The product of Example 2 (20g, 54mmol) will be implemented, cuprous oxide (1g, 7mmol) was dissolved in quinoline (50mL) inside, heated to 140 ℃ overnight. After cooling and filtration, the filtrate was added water, extracted with ethyl acetate, the organic phase was washed with 1N HCl to slightly acidic, saturated aqueous sodium bicarbonate solution, purified by silica gel column chromatography, the concentrated solid slurried with petroleum ether to give an off-white solid (13g, yield 70%).
1 HNMR (400 MHz, CDCl 3 ): [delta] 7.57 (d, 1H), 7.41 (s, 2H), 7.27 (t, 1H), 6.88 (d, 1H), 3.66 (m, 4H), 3.01 (m, 4H), 1.50 (s, 9H) ESI: [M + 1] + = 319.1.
44- (benzothiophene-4-yl) Example – Synthesis of piperazine-1-carboxylic acid tert-butyl ester
The product of Example 2 (500mg, 1.35mmol) will be implemented, silver carbonate (40mg, 0.135mmol) and acetic acid (8mg) was dissolved in dimethylsulfoxide (5mL) inside, heated to 120 ℃, the reaction overnight, cooled and filtered, and the filtrate Water was added, extracted with ethyl acetate, and concentrated by column chromatography to obtain the target substance.
1 HNMR (400 MHz, CDCl 3 ): [delta] 7.57 (d, 1H), 7.41 (s, 2H), 7.27 (t, 1H), 6.88 (d, 1H), 3.66 (m, 4H), 3.01 (m, 4H), 1.50 (s, 9H) ESI: [M + 1] + = 319.1.
Piperazine hydrochloride – 51- (benzothiophen-4-yl) Example
At room temperature, the product of Example 3 will be implemented (2g, 6.2mmol) was dissolved in dioxane (6mL) was added 4N HCl / dioxane (6mL), stirred 3h, concentrated to dryness, the residue was beating ethyl acetate, filtered to obtain the target substance (1.3g, 95% yield).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 7.75 (d, 1H), 7.69 (d, 1H), 7.53 (t, 1H), 7.31 (t, 1H), 6.97 ( t, 1H), 3.30 (bs, 8H) ESI: [M + 1] + = 219.2.
Example 61- formyl-4- (2-ethoxycarbonyl phenyl and thien-4-yl) – piperazine Synthesis
In N 2 protected, at room temperature was added the product of Reference Example 2 to DMF (5mL) inside (1.0g, 3.7mmol), ethyl thioglycolate (410mg, 3.80mmol), potassium carbonate (1.38g, 10mmol), the mixture 80 ℃ stirred for 4 hours. Cooling water was added (20mL), ethyl acetate (3 × 5mL) was extracted, dried over anhydrous sodium sulfate, and concentrated to give a solid with petroleum ether (50mL) After beating 1h, filtered to give a pale yellow solid (1.0 g, yield 83%).
1 HNMR (400 MHz, CDCl3): [delta] 8.15 (d, 2H), 7.59 (d, 1H), 7.41 (t, 1H), 6.94 (d, 1H), 4.44 (q, 2H), 3.85 (t, 2H ), 3.68 (t, 2H), 3.21-3.15 (m, 4H), 1.44 (t, 3H) ESI: [M + 1] + = 319.1.

Example 71- formyl-4- (2-carboxy-benzothiophen-4-yl) – piperazine

The product (1.0g, 3.1mmol) of Example 6 was dissolved in methanol (5mL) and water (2mL) the addition of lithium hydroxide (420mg, 10mmol), the mixture was stirred at room temperature for 5h, was added water (5mL) and acetic acid ethyl ester (10mL), extracted, the aqueous phase was collected, the pH was adjusted to about 4.0 at 0 ℃ with 1N HCl solution, the precipitated solid was filtered and dried to give a pale yellow solid (510mg, 56% yield).
ESI: [M-1] = 289.1.
Example 81- formyl (benzothiophen-4-yl) -4 – piperazine
The product (1.0g, 3.4mmol) Example 7 will be implemented, cuprous oxide (50mg) was dissolved in quinoline (5mL) inside, heated to 140 ℃ overnight. After cooling and filtration, water was added, extracted with ethyl acetate, the organic phase was washed with aqueous 1N HCl to slightly acidic, then with saturated aqueous sodium bicarbonate solution, and concentrated by silica gel column chromatography, the resulting solid was slurried with petroleum ether to give white solid (520mg, 62% yield).
1 HNMR (400 MHz, CDCl 3 ): [delta] 8.15 (s, 1H), 7.62 (d, 1H), 7.42 (m, 2H), 7.31 (t, 1H), 6.04 (d, 1H), 3.82 (t, 2H), 3.63 (t, 2H), 3.19-3.12 (m, 4H) ESI: [M + 1] + = 247.1.
Example 91- (benzothiophen-4-yl) – piperazine hydrochloride
A mixture of the product of Example 8 (500mg) was dissolved in dioxane (2mL) was added 4N HCl / dioxane (3mL), stirred 3h, concentrated to dryness, slurried with ethyl acetate, filtered to give the target (470mg, yield 90%).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 7.75 (d, 1H), 7.69 (d, 1H), 7.53 (t, 1H), 7.31 (t, 1H), 6.97 ( t, 1H), 3.30 (bs, 8H) ESI: [M + 1] + = 219.2.
Example 10 1-Acetyl-4- (2-ethoxycarbonyl phenyl and thien-4-yl) – piperazine Synthesis
Under the protection of N2, at room temperature was added the product of Reference Example 3 (1.0g, 3.74mmol) to DMF (5mL) inside, ethyl thioglycolate (388mg, 3.20mmol), potassium carbonate (1.38g, 10mmol), the mixture was 80 ℃ stirred for 4 hours, cooled water was added (20mL), ethyl acetate (3 × 5mL) was extracted, dried over anhydrous sodium sulfate, and concentrated to give a solid with petroleum ether (50mL) beating 1h, filtered to give a pale yellow solid (863mg, yield 70%).
1 HNMR (400 MHz, CDCl 3 ): [delta] 8.17 (s, 1H), 7.60 (d, 1H), 7.42 (t, 1H), 7.01 (d, 1H), 4.44 (q, 2H), 3.94 (br, 2H), 3.80 (br, 2H), 3.21 (br, 4H), 2.19 (s, 3H), 1.44 (t, 3H) ESI: [M + 1] + = 333.3.
Example 11 1-Acetyl-(2-carboxy-benzothiophen-4-yl) -4 – piperazine
The product (1.0g, 3.0mmol) from Example 10 was dissolved in methanol (5mL) and water (2mL) the addition of lithium hydroxide (300mg, 7.2mmol), the mixture was stirred at rt for 3h, water was added (5mL) and ethyl acetate ester (10mL), separated and the aqueous phase was collected, the pH adjusted with aqueous 1N HCl at 0 ℃ to about 4.0, and the precipitated solid was filtered, dried to give a pale yellow solid (820mg, yield 90%).
ESI: [M-1] = 303.1.
Example 12 1-Acetyl-4- (benzothiazol-4-yl) – piperazine
The product of Example 11 (1.0g, 3.2mmol), cuprous oxide (50mg) was dissolved in quinoline (5mL) inside, heated to 140 ℃ overnight. After cooling and filtration, water was added, extracted with ethyl acetate, washed with 1N HCl solution was added to a weak acid, a saturated aqueous solution of sodium bicarbonate, silica gel column chromatography, and concentrated to give a solid slurried with petroleum ether to give a white solid (600mg , yield 70%).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 7.95 (s, 1H), 7.65 (d, 1H), 7.41 (t, 1H) 6.95 (d, 1H), 3.69 (q, 4H), 3.10 (t , 2H), 3.02 (t, 2H), 2.06 (s, 3H) ESI: [M + 1] + = 261.1.
EXAMPLE 131- (benzothiophen-4-yl) – piperazine hydrochloride
A mixture of the product of Example 12 (1g, 3.8mmol) was dissolved in dioxane (6mL) was added 4N HCl / dioxane (6mL), stirred 3h, concentrated dry, with ethyl beating, filtered to give the product (870mg, yield 90%).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 7.75 (d, 1H), 7.69 (d, 1H), 7.53 (t, 1H), 7.31 (t, 1H), 6.97 ( t, 1H), 3.30 (bs, 8H) ESI: [M + 1] + = 219.2.
141- (benzothiophen-4-yl) Example – piperazine

The product of (500mg, 1.38mmol) of Example 2 was dissolved in quinoline (3mL) the addition of cuprous oxide (20mg), the reaction temperature was raised to 140 ℃ After 2h, the reaction continues to heat up to 240 ℃ 3h, cooled to room temperature, filtered , water was added, extracted with ethyl acetate, washed with saturated aqueous sodium bicarbonate, silica gel column chromatography, and concentrated to give the desired product. 1 HNMR (300 MHz, DMSO-d 6 ): [delta] 8.74 (bs, 1H), 7.75 (d, 1H), 7.69 (d, 1H), 7.51 (d, 1H), 7.31 (t, 1H), 6.95 ( d, 1H), 3.24 (m, 8H) ESI: [M + 1] + = 219.2.

Example 15 7- [4- (2-carboxy-4-yl-benzothiophene-1-piperazinyl) butoxy] -3,4-dihydro -2 (1H) – quinolinone Preparation of

7- [4- (2-ethoxycarbonyl-4-phenyl and thienyl-1-piperazinyl) butoxy] -3,4-dihydro -2 (1H) – quinolinone (300mg, 0.59 mmol) was dissolved in methanol (3mL) and water (1mL) was added lithium hydroxide (76mg, 1.8mmol), stirred at rt for 3h, ethyl acetate was added, the aqueous phase was adjusted with 1N dilute hydrochloric acid to about pH 4.0, using bis dichloromethane: methanol (10: 1) extraction, concentration did a white solid (210mg, 46% yield).

1 HNMR (400 MHz, DMSO-d 6 ): [delta] 10.01 (s, 1H), 7.88 (s, 1H), 7.61 (d, 1H), 7.38 (t, 1H), 7.03 (q, 1H), 6.93 ( d, 1H), 6.48 (m, 2H), 3.92 (m, 4H), 3.35 (s, 4H), 2.84 (s, 4H), 2.77 (s, 2H), 2.62 (s, 2H), 1.72 (m , 4H), ESI: [M-1] = 478.3.

EXAMPLE 167- [4- (benzothiazol-4-yl-1-piperazinyl) butoxy] -3,4-dihydro -2 (1H) – quinolinone Preparation of

The product (500mg, 1.04mmol) of Example 15 will be implemented, cuprous oxide (50mg) was dissolved in quinoline (5mL) inside, heated to 140 ℃ overnight. After cooling and filtration, water was added, extracted with ethyl acetate, washed with 1N HCl solution was added until pH = 4.0, dichloromethane: methanol (10: 1) extracted, dried over anhydrous sodium sulfate, and silica gel column chromatography to give a solid (320mg , yield 70%).

1 HNMR (400 MHz, DMSO-d 6 ): δ10.00 (s, 1H), 7.69 (d, 1H), 7.61 (d, 1H), 7.40 (d, 1H), 7.27 (t, 1H), 7.04 ( d, 1H), 6.89 (d, 1H), 6.50 (dd, 1H), 6.45 (d, 1H), 3.93 (t, 2H), 3.06 (br, 4H), 2.78 (t, 2H), 2.60 (br , 4H), 2.41 (t, 4H), 1.74 (t, 2H), 1.60 (t, 2H) ESI: [M + 1]+ = 436.3.

Preparation of piperazine-1-carboxylic acid tert-butyl ester – Example 17 4- (2-ethoxycarbonyl phenyl and thien-4-yl)

Under nitrogen at room temperature was added to ethanol (5mL) within the reference product (200mg, 0.62mmol) of Example 1, ethyl mercaptoacetate (0.081ml, 0.74mmol), potassium carbonate (342mg, 2.48mmol), the mixture was 85 ℃ stirred for 18 hours, concentrated, and column chromatography to obtain the target substance (100mg, 42% yield).
1 HNMR (400 MHz, CDCl 3 ): [delta] 8.40 (s, 1H), 7.58 (d, 1H), 7.37 (t, 1H), 6.95 (d, 1H), 4.44 (q, 2H), 3.64 (m, 4H), 3.15 (m, 4H) ESI: [M + 1] + = 391.1.
Preparation of piperazine-1-carboxylic acid tert-butyl ester – Example 18 4- (2-ethoxycarbonyl phenyl and thien-4-yl)
Under nitrogen, was added at room temperature to DMF (5mL) within the reference product (200mg, 0.62mmol) of Example 1, ethyl mercaptoacetate (0.081ml, 0.74mmol), DIPEA (342mg, 2.48mmol), the mixture was at 105 ℃ stirred for 18 hours, 1N HCl solution was added adjust the pH = 7, and extracted with methyl tert-butyl ether, the ether layer was then washed three times with saturated brine, dried over anhydrous sodium sulfate, the drying agent filtered, and concentrated by column chromatography to obtain the target (170mg, yield 71%).
1 HNMR (400 MHz, CDCl 3 ): [delta] 8.40 (s, 1H), 7.58 (d, 1H), 7.37 (t, 1H), 6.95 (d, 1H), 4.44 (q, 2H), 3.64 (m, 4H), 3.15 (m, 4H) ESI: [M + 1] + = 391.1.
Preparation of piperazine-1-carboxylic acid tert-butyl ester – Example 19 4- (2-ethoxycarbonyl phenyl and thien-4-yl)

Under nitrogen at room temperature was added the product of Reference Example 1 to ethanol (5mL) inside (200mg, 0.62mmol), ethyl mercaptoacetate (0.081ml, 0.74mmol), sodium hydroxide (100mg, 2.48mmol), the mixture 85 ℃ stirred for 6 hours, concentrated to column chromatography to obtain the target substance (70mg, 30% yield).

1 HNMR (400 MHz, CDCl 3 ): [delta] 8.40 (s, 1H), 7.58 (d, 1H), 7.37 (t, 1H), 6.95 (d, 1H), 4.44 (q, 2H), 3.64 (m, 4H), 3.15 (m, 4H) ESI: [M + 1] + = 391.1.
Piperazine hydrochloride – (2-carboxy-benzothiophen-4-yl) Example 201-
The product of Example 2 (200mg, 0.55mmol), was dissolved in THF (5mL), concentrated hydrochloric acid (0.5mL), 50 ℃ heated 6h. Cooling, methyl tert-butyl ether (5mL), filtered to give the target (130mg, yield 79%).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 8.04 (s, 1H), 7.69 (d, 1H), 7.43 (t, 1H), 7.00 (d, 1H), 3.30 ( bs, 8H) ESI: [M + 1] + = 262.9.
Piperazine hydrochloride – (benzothiophene-4-yl) Example 211-
The product of Example 20 (130mg, 0.43mmol) was added to diphenyl ether (3mL) in, 260 ℃ heating 0.5h. Cooled and filtered to give the object (60mg, 55% yield).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 7.75 (d, 1H), 7.69 (d, 1H), 7.53 (t, 1H), 7.31 (t, 1H), 6.97 ( t, 1H), 3.30 (bs, 8H) ESI: [M + 1] + = 219.2.
Preparation of tert-butyl piperazine-1 – Example 224- (2-carboxy-benzothiophen-4-yl)
Under nitrogen, to N, at room temperature was added N- dimethylformamide (5mL) within the reference product (200g, 0.62mmol) of Example 1, thioglycolic acid (114mg, 1.23mmol), sodium methoxide (133mg, 2.45mmol ), and the mixture was stirred at 105 ℃ 18 hours. Cooling, water was added, extracted with ethyl acetate, separated and the aqueous phase was adjusted pH = 5 or so, the precipitated solid was filtered and dried to obtain the target substance (130mg, 58% yield).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 7.98 (s, 1H), 7.64 (d, 1H), 7.42 (t, 1H), 6.95 (d, 1H), 3.53 (bs, 4H), 3.035 ( bs, 4H) ESI: [M-1] = 361.1.
Preparation of piperazine-1-carboxylic acid tert-butyl ester – (2-carboxy-benzothiophen-4-yl) Example 234-
Under nitrogen, to N, at room temperature was added N- dimethylformamide (5mL) within the reference product (200g, 0.62mmol) of Example 1, thioglycolic acid (114mg, 1.23mmol), sodium hydroxide (99mg, 2.45 mmol), the mixture was stirred at 105 ℃ 18 hours. Cooling, water was added, extracted with ethyl acetate, separated and the aqueous phase was adjusted pH = 5 or so, the precipitated solid was filtered and dried to obtain the target substance (180mg, yield 81%).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 7.98 (s, 1H), 7.64 (d, 1H), 7.42 (t, 1H), 6.95 (d, 1H), 3.53 (bs, 4H), 3.035 ( bs, 4H) ESI: [M-1] = 361.1.
Example 24 7- [4- (2-ethoxycarbonyl-4-phenyl and thienyl-1-piperazinyl) butoxy] -2 (1H) – quinolinone Preparation of
2-chloro-6- (4- (4 – ((2-oxo-1,2-dihydro-quinolin-7-yl) oxy) butyl) piperazin-1-yl) benzaldehyde (80mg , 0.18mmol) was dissolved in DMF (5mL) was added DIPEA (94mg, 0.73mmol) and ethyl mercaptoacetate (0.024mL, 0.22mmol), 110 ℃ stirred for 16 hours.Cooling, water was added, extracted with ethyl acetate, the ethyl acetate layer was washed with saturated brine, dried over anhydrous sodium sulfate, and silica gel column chromatography to give a solid (40mg, 46% yield).
1 HNMR (400 MHz, DMSO-d 6 ): δ11.69 (s, 1H), 11.24 (s, 1H), 8.09 (s, 1H), 7.81 (d, 1H), 7.74 (d, 1H), 7.57 ( d, 1H), 7.48 (t, 1H), 7.04 (d, 1H), 6.82 (m, 2H), 6.30 (d, 1H), 4.32 (m, 4H), 4.06 (t, 2H), 3.67-3.16 (m, 8H), 1.96 (m, 2H), 1.84 (m, 2H), 1.32 (t, 3H) ESI: [M + 1] + = 506.4.
Example 25 7- [4- (2-carboxy-4-yl-benzothiophene-1-piperazinyl) butoxy] -3,4-dihydro -2 (1H) – quinolinone Preparation of
7- [4- (2-ethoxycarbonyl-4-phenyl and thienyl-1-piperazinyl) butoxy] -3,4-dihydro -2 (1H) – quinolinone (100mg, 0.19 mmol) was dissolved in acetic acid (3mL) and concentrated hydrochloric acid (0.5mL), 100 ℃ stirred for 10 hours. The reaction mixture was poured into ice water, stirred for 10min after filtration, to obtain the target substance (40mg, 43% yield).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 10.01 (s, 1H), 7.88 (s, 1H), 7.61 (d, 1H), 7.38 (t, 1H), 7.03 (q, 1H), 6.93 ( d, 1H), 6.48 (m, 2H), 3.92 (m, 4H), 3.35 (s, 4H), 2.84 (s, 4H), 2.77 (s, 2H), 2.62 (s, 2H), 1.72 (m , 4H), ESI: [M-1] = 478.3.
Example 26 7- [4- (benzothiazol-4-yl-1-piperazinyl) butoxy] -3,4-dihydro -2 (1H) – quinolinone Preparation of
The product (400mg, 0.83mmol) of Example 25 will be implemented, silver carbonate (46mg, 0.16mmol) was dissolved in DMSO (5mL) and the acetic acid was heated to 120 ℃ overnight. Cooling, water was added, extracted with ethyl acetate, the ethyl acetate layer was washed with saturated aqueous sodium bicarbonate and brine again each wash, dried over anhydrous sodium sulfate, and silica gel column chromatography to give a solid (80mg, yield 22%).
1 HNMR (400 MHz, DMSO-d 6 ): δ10.00 (s, 1H), 7.69 (d, 1H), 7.61 (d, 1H), 7.40 (d, 1H), 7.27 (t, 1H), 7.04 ( d, 1H), 6.89 (d, 1H), 6.50 (dd, 1H), 6.45 (d, 1H), 3.93 (t, 2H), 3.06 (br, 4H), 2.78 (t, 2H), 2.60 (br , 4H), 2.41 (t, 4H), 1.74 (t, 2H), 1.60 (t, 2H) ESI: [M + 1]+ = 436.3.
Example 27 7- [4- (2-carboxy-4-yl-benzothiophene-1-piperazinyl) butoxy] -2 (1H) – quinolinone Preparation of
2-chloro-6- (4- (4 – ((2-oxo-1,2-dihydro-quinolin-7-yl) oxy) butyl) piperazin-1-yl) benzaldehyde (80mg , 0.18mmol) was dissolved in DMF (5mL) was added sodium hydroxide (29mg, 0.73mmol) and thioglycolic acid (0.025mL, 0.36mmol), 120 ℃ stirred for 16 hours. Cooling, water was added, adjusted with 1N HCl aqueous solution is about pH = 5, extracted with ethyl acetate, the ethyl acetate layer was washed with saturated brine, dried over anhydrous sodium sulfate, and silica gel column chromatography to give a solid (40mg, yield 46 %).

ESI: [M + 1] + = 478.0.

Piperazine hydrochloride – (2-carboxy-benzothiophen-4-yl) Example 28 1-
The product of Example 17 (100mg, 0.25mmol) was dissolved in acetic acid (3mL) and concentrated hydrochloric acid (0.5 mL) in, 100 ℃ stirred for 10 hours. The reaction mixture was poured into ice water, stirred for 10min after suction filtration to give the object (38mg, 50% yield).
1 HNMR (400 MHz, DMSO-d 6 ): [delta] 9.46 (bs, 2H), 8.04 (s, 1H), 7.69 (d, 1H), 7.43 (t, 1H), 7.00 (d, 1H), 3.30 ( bs, 8H) ESI: [M + 1] + = 262.9.
uUpdate july 2015

On July 10, the U.S. Food and Drug Administration approved Rexulti (brexpiprazole) tablets to treat adults with schizophrenia and as an add-on treatment to an antidepressant medication to treat adults with major depressive disorder (MDD).

Schizophrenia is a chronic, severe, and disabling brain disorder affecting about one percent of Americans. Typically, symptoms are first seen in adults younger than 30 years of age and include hearing voices; believing other people are reading their minds or controlling their thoughts; and being suspicious or withdrawn.

MDD, commonly referred to as depression, is also a severe and disabling brain disorder characterized by mood changes and other symptoms that interfere with a person’s ability to work, sleep, study, eat, and enjoy once-pleasurable activities. Episodes of depression often recur throughout a person’s lifetime, although some may experience a single occurrence. Other signs and symptoms of MDD include loss of interest in usual activities; significant change in weight or appetite; insomnia or excessive sleeping (hypersomnia); restlessness/pacing (psychomotor agitation); increased fatigue; feelings of guilt or worthlessness; slowed thinking or impaired concentration; and suicide attempts or thoughts of suicide. Not all people with MDD experience the same symptoms.

“Schizophrenia and major depressive disorder can be disabling and can greatly disrupt day-to-day activities,” said Mitchell Mathis, M.D., director of the Division of Psychiatry Products in the FDA’s Center for Drug Evaluation and Research. “Medications affect everyone differently so it is important to have a variety of treatment options available for patients with mental illnesses.”

The effectiveness of Rexulti in treating schizophrenia was evaluated in 1,310 participants in two 6-week clinical trials. Rexulti was shown to reduce the occurrence of symptoms of schizophrenia compared to placebo (inactive tablet).

The effectiveness of Rexulti as an add-on treatment for MDD was evaluated in two 6-week trials that compared Rexulti plus an antidepressant to placebo plus an antidepressant in 1,046 participants for whom an antidepressant alone did not adequately treat their symptoms. The participants taking Rexulti reported fewer symptoms of depression than those taking the placebo.

Rexulti and other drugs used to treat schizophrenia have a Boxed Warning alerting health care professionals about an increased risk of death associated with the off-label use of these drugs to treat behavioral problems in older people with dementia-related psychosis. No drug in this class is approved to treat patients with dementia-related psychosis.

The Boxed Warning also alerts health care professionals and patients to an increased risk of suicidal thinking and behavior in children, adolescents, and young adults taking antidepressants. Patients should be monitored for worsening and emergence of suicidal thoughts and behaviors. Rexulti must be dispensed with a patient Medication Guide that describes important information about the drug’s uses and risks.

The most common side effects reported by participants taking Rexulti in clinical trials included weight gain and an inner sense of restlessness, such as feeling the need to move.

Rexulti is manufactured by Tokyo-based Otsuka Pharmaceutical Company Ltd.

 

update………….

4-Chlorobenzo[b]thiophene a key intermediate in brexpiprazole synthesis

Abstract Image
We established an improved synthetic route to 4-chlorobenzo[b]thiophene, a key intermediate in brexpiprazole synthesis, via a practical decarboxylation process in three steps. Thermal analysis demonstrated that the coexistence of the decarboxylated product with DBU should be avoided and that removal of the product outside the reactor was vital. Our process yields the target compound by distillation under reduced pressure and is safe, highly batch efficient, cost-effective, and high yielding. Furthermore, manufacturing on a pilot scale was also accomplished through our approach.

Figure

4-Chlorobenzo[b]thiophene-2-carboxylic Acid (4)

 4 as a white solid
mp 260 °C.
1H NMR (300 MHz, DMSO-d6) δ 7.54 (d, 1H, J = 11.6, 7.7 Hz), 7.56 (dd, 1H, J = 17.8, 7.7 Hz), 8.03 (d, 1H, J = 0.7 Hz), 8.07 (td, 1H, J = 7.6, 0.9 Hz), 13.19 (brs, 1H).

13C NMR (75 MHz, DMSO-d6) δ 122.21, 125.01, 126.84, 127.99, 128.96, 136.50, 136.57, 142.55, 163.10.

Elemental analysis calcd for C: 50.83%, H: 2.37%, found C: 50.84%, H: 2.21%.

2,3,4,6,7,8,9,10-Octahydropyrimido[1,2-a]azepin-1-ium 4-Chlorobenzo[b]thiophene-2-carboxylate 5

5 as a white solid
Mp 182.5 °C.
1H NMR (300 MHz, CDCl3) δ 1.66 (m, 6H), 1.80–1.75 (m, 2H), 2.98–2.94 (m, 2H), 3.45–3.39 (m, 4H), 3.55–3.51 (m, 2H), 7.31–7.20 (m, 2H), 7.69 (dd, 1H, J = 3.9, 0.6 Hz), 7.97 (s, 1H), 13.19 (brs, 1H).

13C NMR (75 MHz, CDCl3) δ 19.51, 24.03, 26.70, 28.88, 31.99, 38.01, 48.36, 53.94, 121.04, 123.00, 125.17, 126.61, 128.98, 138.21, 142.43, 146.85, 165.91, 167.20.

Elemental analysis calcd for C: 59.25%, H: 5.80%, N: 7.68%, found C: 59.10%, H: 5.44%, N: 7.53%.

4-Chlorobenzo[b]thiophene (2)

1H NMR (300 MHz, CDCl3) δ 7.26 (t, 1H, J = 7.8 Hz), 7.36 (dd, 1H, J = 7.8, 0.9 Hz), 7.50 (d, 1H, J = 5.7 Hz), 7.52 (d, 1H, J = 5.7 Hz), 7.76 (d, 1H, J = 7.8 Hz).

13C NMR (75 MHz, CDCl3) δ 121.12, 122.40, 125.02, 127.43, 128.93, 138.06, 141.07.

Elemental analysis calcd for C: 56.98%, H: 2.99%, found C: 56.76%, H: 2.94%.

SEE

http://pubs.acs.org/doi/abs/10.1021/acs.oprd.5b00340

http://pubs.acs.org/doi/suppl/10.1021/acs.oprd.5b00340/suppl_file/op5b00340_si_001.pdf

Safe and Efficient Decarboxylation Process: A Practical Synthetic Route to 4-Chlorobenzo[b]thiophene

Bulk Pharmaceutical Chemicals Department, Second Tokushima Factory, Production Headquarters, Otsuka Pharmaceutical Co., Ltd., 224-18, Hiraishi Ebisuno, Kawauchi-cho, Tokushima 771-0182, Japan
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.5b00340

 

FDA Approves Farydak (panobinostat) for Multiple Myeloma


Panobinostat

syn……….https://newdrugapprovals.org/2014/01/23/panobinostat/

HDAC inhibitors, orphan drug

cas 404950-80-7 

2E)-N-hydroxy-3-[4-({[2-(2-methyl-1H-indol-3-yl)ethyl]amino}methyl)phenyl]acrylamide

N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2E-2-propenamide (alternatively, N-hydroxy-3-(4-{[2-(2-methyl-1H-indol-3-yl)-ethylamino]-methyl}-phenyl)-acrylamide)

Molecular Formula: C21H23N3O2   Molecular Weight: 349.42622

  • Faridak
  • LBH 589
  • LBH589
  • Panobinostat
  • UNII-9647FM7Y3Z

A hydroxamic acid analog histone deacetylase inhibitor from Novartis.

NOVARTIS, innovator

Histone deacetylase inhibitors

syn……….https://newdrugapprovals.org/2014/01/23/panobinostat/

FDA Approves Farydak (panobinostat) for Multiple Myeloma

February 23, 2015 — The U.S. Food and Drug Administration today approved Farydak (panobinostat) for the treatment of patients with multiple myeloma.

Multiple myeloma is a form of blood cancer that arises from plasma cells, a type of white blood cell, found in bone marrow. According to the National Cancer Institute, approximately 21,700 Americans are diagnosed with multiple myeloma and 10,710 die from the disease annually

read at

http://www.drugs.com/newdrugs/fda-approves-farydak-panobinostat-multiple-myeloma-4170.html?utm_source=ddc&utm_medium=email&utm_campaign=Today%27s+news+summary+-+February+23%2C+2015&utm_content=FDA+Approves+Farydak+%28panobinostat%29+for+Multiple+Myeloma

AND

FDA approves Farydak for treatment of multiple myeloma [press release].http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm435296.htmPublished February 23, 2015. Accessed february 23, 2015

syn……….https://newdrugapprovals.org/2014/01/23/panobinostat/

syn……….https://newdrugapprovals.org/2014/01/23/panobinostat/

syn……….https://newdrugapprovals.org/2014/01/23/panobinostat/

syn……….https://newdrugapprovals.org/2014/01/23/panobinostat/

 

FDA approves Farydak for treatment of multiple myeloma [press release].http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm435296.htmPublished February 23, 2015. Accessed february 23, 2015

FDA approves Pfizer’s Ibrance (palbociclib) for postmenopausal women with advanced breast cancer


PALBOCICLIB

Mechanism of action: selective inhibitor of the cyclin-dependent kinases CDK4 and CDK6
Indication: Estrogen receptor-positive (ER+), HER2-negative (HER2 -) breast cancer

FDA approves Ibrance for postmenopausal women with advanced breast cancer

February 3, 2015

syn……….https://newdrugapprovals.org/2014/01/05/palbociclib/

The U.S. Food and Drug Administration today granted accelerated approval to Ibrance (palbociclib) to treat advanced (metastatic) breast cancer.

Breast cancer in women is the second most common type of cancer in the United States. It forms in the breast tissue and in advanced cases, spreads to surrounding normal tissue. The National Cancer Institute estimates that 232,670 American women were diagnosed with breast cancer and 40,000 died from the disease in 2014.

Ibrance works by inhibiting molecules, known as cyclin-dependent kinases (CDKs) 4 and 6, involved in promoting the growth of cancer cells. Ibrance is intended for postmenopausal women with estrogen receptor (ER)-positive, human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer who have not yet received an endocrine-based therapy. It is to be used in combination with letrozole, another FDA-approved product used to treat certain kinds of breast cancer in postmenopausal women.

“The addition of palbociclib to letrozole provides a novel treatment option to women diagnosed with metastatic breast cancer,” said Richard Pazdur, M.D., director of the Office of Hematology and Oncology Products in the FDA’s Center for Drug Evaluation and Research. “The FDA is committed to expediting marketing approval of cancer drugs through our accelerated approval regulations.”

syn……….https://newdrugapprovals.org/2014/01/05/palbociclib/

The FDA granted Ibrance breakthrough therapy designation because the sponsor demonstrated through preliminary clinical evidence that the drug may offer a substantial improvement over available therapies. It also received a priority review, which provides for an expedited review of drugs intended to provide a significant improvement in safety or effectiveness in the treatment of a serious condition or meet an unmet medical need. Ibrance is being approved more than two months ahead of the prescription drug user fee goal date of April 13, 2015, the date when the agency was scheduled to complete its review of the application.

Ibrance is being approved under the FDA’s accelerated approval program, which allows approval of a drug to treat a serious or life-threatening disease based on clinical data showing the drug has an effect on a surrogate endpoint reasonably likely to predict clinical benefit to patients. This program provides earlier patient access to promising new drugs while the company conducts confirmatory clinical trials.

The drug’s efficacy was demonstrated in 165 postmenopausal women with ER-positive, HER2-negative advanced breast cancer who had not received previous treatment for advanced disease. Clinical study participants were randomly assigned to receive Ibrance in combination with letrozole or letrozole alone. Participants treated with Ibrance plus letrozole lived about 20.2 months without their disease progressing (progression-free survival), compared to about 10.2 months seen in participants receiving only letrozole. Information on overall survival is not available at this time.

The most common side effects of the drug were a decrease in infection-fighting white blood cells called neutrophils (neutropenia), low levels of white blood cells (leukopenia), fatigue, low red blood cell counts (anemia), upper respiratory infection, nausea, inflammation of the lining of the mouth (stomatitis), hair loss (alopecia), diarrhea, low blood platelet counts (thrombocytopenia), decreased appetite, vomiting, lack of energy and strength (asthenia), damage to the peripheral nerves (peripheral neuropathy) and nosebleed (epistaxis). Healthcare professionals should inform patients of these risks.

It is recommended that treatment begin with a 125 milligram dose for 21 days, followed by seven days without treatment. Healthcare professionals are advised to monitor complete blood count prior to start of therapy and at the beginning of each cycle, as well as on Day 14 of the first two cycles, and as clinically indicated.

Ibrance is marketed by New York City-based Pfizer, Inc.

see synthesis……….https://newdrugapprovals.org/2014/01/05/palbociclib/

 

New York City-based Pfizer, Inc.

 

Pfizer World Headquarters building in New York City. Zoetis, based in Madison, N.J., traces its roots back to 1952 as a Pfizer unit and has made at least 10 …

Pfizer’s NYC headquarters

 

Daiichi Sankyo receives FDA approval for anti-clotting drug Savaysa , EDOXABAN


Edoxaban, DU-176b

Edoxaban (DU-176b, trade names Savaysa, Lixiana) is an anticoagulant drug which acts as a direct factor Xa inhibitor. It was developed by Daiichi Sankyo and approved in July 2011 in Japan for prevention of venous thromboembolisms (VTE) following lower-limb orthopedic surgery.[1] It was also approved by the FDA in January 2015 for the prevention of stroke and non–central-nervous-system systemic embolism.[2]

Daiichi Sankyo receives FDA approval for anti-clotting drug Savaysa
Japanese drug-maker Daiichi Sankyo has obtained approval from US Food and Drug Administration (FDA) for its anti-clotting drug Savaysa (edoxaban tablets)..8 JAN 2015

Daiichi Sankyo, APPROVED IN JAPAN as tosylate monohydrate salt in 2011 for the prevention of venous embolism in patients undergoing total hip replacement surgery

for synthesis see….http://www.sciencedirect.com/science/article/pii/S0968089613002642  Bioorganic & Medicinal Chemistry 21 (2013) 2795–2825,  see s[pecific page 2808 for description  ie 14/31 of pdf

WO 2010071121, http://www.google.com/patents/WO2010071121A1

WO 2007032498

N’-(5-chloropyridin-2-yl)-N-[(1S,2R,4S)-4-(dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[1,3]thiazolo[5,4-c]pyridine-2-carbonyl)amino]cyclohexyl]oxamide

    N1-(5-Chloropyridin-2-yl)-N2-((1S,2R,4S)-4-[(dimethylamino)carbonyl]-2-{[(5-methyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridin-2-yl)carbonyl]amino}cyclohexyl)ethanediamide

Edoxaban (INN, codenamed DU-176b, trade name Lixiana) is an anticoagulant drug which acts as a direct factor Xa inhibitor. It is being developed by Daiichi Sankyo. It was approved in July 2011 in Japan for prevention of venous thromboembolisms (VTE) following lower-limb orthopedic surgery.[1]

 

In animal studies, edoxaban is potent, selective for factor Xa and has good oral bioavailability.[2]

Daichi Sankyo’s edoxaban tosilate is an orally administered
coagulation factor Xa inhibitor that was approved and launched
in Japan for the preventive treatment of venous thromboembolic
events (VTE) in patients undergoing total knee arthroplasty, total
hip arthroplasty, or hip fracture surgery. Edoxaban has been
shown to have a rapid onset of anticoagulant effect due to short
Tmax (1–2 h) after dosing and sustained for up to 24 h post-dose.
Marketed under the brand name Lixiana, it is currently in phase
III studies in the US for the prevention of stroke and systemic embolic
events in patients with atrial fibrillation (AF) and venous
thromboembolism (VTE).

Several Phase II clinical trials have been conducted, for example for thromboprophylaxis after total hip replacement[3] (phase III early results compare well to enoxaparin[4]), and for stroke prevention in patients with atrial fibrillation[5][6].Those papers follow similar recent major trials showing similar results for the other new factor Xa inhibitorsrivaroxaban and apixaban.

A large phase III trial showed that edoxaban was non inferior to warfarin in preventing recurrent venous thromboembolic events with fewer episodes of major bleeding.[7]

……………..

PATENT

http://www.google.com/patents/WO2014081047A1?cl=en

Chemically, edoxaban is

N1– (5-chloropyridin-2-yl) -N2– ( (IS, 2R/4S) -4- [ (dimethylamino) carbo nyl] -2- { [ ( 5-methyl-4 , 5,6, 7-tetrahydrothiazolo [5 , 4-c] pyridin-2-yl ) carbonyl] amino}eyelohexyl) ethanediamide , represented by the following formula (A) :

Figure imgf000003_0002

(A) The p-toluenesulfonic acid monohydrate salt of compound A is represented b the following formula (B) :

Figure imgf000004_0001

(B)

Edoxaban is known as a compound that exhibits an inhibitory effect on activated blood coagulation factor X (also referred to as activated factor X or FXa) , and is useful as a preventive and/or therapeutic drug for thrombotic diseases.

Several processes are known in the literature for preparing edoxaban for example, U.S. Patent No. 7365205; U.S. Publication No . 20090105491.

U.S. Patent No. 7365205 provides a process for the preparation of edoxaban, wherein the process involves the use of

(IS, 4S, 5S) -4-iodo-6-oxabicyclo [3.2.1] octan-7-one, represented by the following formula (C) :

Figure imgf000004_0002

(C)

as an intermediate.

The present inventors have identified that

(IS, 4S, 5S) -4-bromo-6-oxabicyclo [3.2.1] octan-7-one, represented by the following formula (I) :

Figure imgf000005_0001

( I )

could also be used as an intermediate for the preparation of FXa inhibitory compounds like edoxaban. The present inventors have found that replacement of

(IS, 4S, 5S) -4-iodo-6-oxabicyclo [3.2.1] octan-7-one (C) with

(IS, 4S, 5S) -4-bromo-6-oxabicyclo [3.2.1] octan-7-one (I) has a better atom economy and also an impact on cost.

A method for the synthesis of the

(IS, 4S, 5S) -4 -bromo- 6 -oxabicyclo [3.2.1] octan-7-one (I) was reported in Tetrahedron Letters, 51, (2010) Pages 3433-3435 which involves the reaction of ( IS) -cyclohex-3 -ene- 1-carboxylic acid represented by the following formula (II) :

Figure imgf000005_0002

( Π )

with N-bromosuccinimide in the presence of molecular sieves using dichloromethane as a solvent. However, this reaction is carried out in dark over a period of 7 hours and does not provide a pure product .

Tetrahedron, Vol. 28, Pages 3393 -3399 , 1972 provides a process for the preparation of 4 -bromo- 6 -oxabicyclo [3.2.1] octan-7-one which involves the addition of 20% excess of a 2M solution of bromine in chloroform to a stirred solution of cyclohex- 3 -ene- 1-carboxylic acid (0.04 mol) in chloroform (250 mL) in the absence of a base . Extraction with aqueous sodium bicarbonate followed by acidification gave, after extraction with ether and evaporation of the extract, a mixture of cis & trans 3 , 4-dibromocyclohexanecarboxylic acid (6.7 g) and evaporation of the chloroform layer afforded the bromolactone (0.59 g) . It further provides a process for the preparation of

4 -bromo-6 -oxabicyclo [3.2.1] octan-7-one which involves the treating of cyclohex-3-ene-l-carboxylic acid (0.08 mol) dissolved in chloroform (450 mL) with 20% excess bromine in the presence of an equimolar amount of triethylamine (8.1 g) . After extraction of the amine with 2N hydrochloric acid, and work-up, bromolactone (10.7 g) and a mixture of cis & trans 3 , 4 -dibromocyclohexanecarboxylic acid (6.6 g) were obtained.

Tetrahedron Vol. 48, No. 3, Pages 539-544, 1992 provides a process for the preparation of

(IS, 4S, 5S) -4-bromo-6-oxabicyclo [3.2.1] octan-7-one (I) which involves the addition of 1M solution of bromine in chloroform (30 mL) at 0°C to a solution of ( IS) -cyclohex-3 -ene- 1-carboxylic acid (0.024 mol) of formula (II) in chloroform (600 mL) in the presence of an equimolar amount of triethylamine (3.33 mL) . After work-up, the crude bromolactone obtained was recrystallized from petroleum ether.

However, bromination using bromine does not provide a pure product in good yield.

Heterocycles, Vol. 23, No. 8, Pages 2035-2039, 1985 provides a process for the 4-bromo-6-oxabicyclo [3.2.1] octan-7-one which involves the addition of cyclohex-3-ene-l-carboxylic acid (1.0 mM) in 1 , 2 -dimethoxyethane (2 mL) to a stirred solution of 90% Lead (IV) acetate (1.1 or 2.2 mM) in 1 , 2 -dimethoxyethane (4 mL) followed by the addition of Zinc bromide (2.2 mM) in 1 , 2 -dimethoxyethane (4 mL) and continuing the stirring for 10-30 minutes at 0°C . The reaction mixture was poured into a solution of ice-cold water (30 mL) and 10% hydrochloric acid (10 mL) , and extracted with ether (50 mL X 3) . The combined ether extract was washed successively with saturated sodium hydrogen carbonate solution (20 mL) , 10% sodium thiosulphate solution (5 mL) , and brine (10 mL) , and dried over sodium sulphate. Evaporation of the solvent gave crude lactone which were separated and purified (42% yield) . However, this reaction does not provide a pure product in good yield.

Heterocycles, Vol. 31, No. 6, Pages 987-991, 1990 provides a method for bromolactonization using a

dimethylsulfoxide-trimethylsilyl bromide-amine system. The bromolactonization is carried out for 10 to 72 hours using different solvents and triethylamine or diisopropylethyl amine as base. However, this process does not provide a product in high yield. Further the process afforded the cis isomer exclusively. Journal of the Chemical Society, Perkin Transactions 1:

Organic and Bio-Organic Chemistry (1972-1999) (1994) , (7) , Pages 847-851 provides a method for bromolactonization using a

dimethylsulfoxide-trimethylsilyl bromide-amine system. The bromolactonization is carried out for 12 hours using

dimethylsulfoxide and chloroform solvent system and triethylamine or diisopropylethyl amine as base. However, this process resulted in a low yield of about 55%. Citation List

Patent Literature

PTLl: U.S. Patent No. 7365205

PTL2: U.S. Publication No. 20090105491.

Non Patent Reference

NPLl: Feng Chen et al . , Tetrahedron Letters, 51, (2010) Pages 3433-3435.

NPL2 : G. Belluci et al . , Tetrahedron, Vol. 28, No. 13, Pages 3393-3399, 1972.

NPL3 : Marco Chini et al ., Tetrahedron Vol .48, No. 3, Pages 539-544 , 1992.

NPL4 : Y. Fujimoto et al . , Heterocycles , Vol. 23, No. 8, Pages 2035-2039, 1985.

NPL5: C. Iwata et al . , Heterocycles, Vol. 31, No. 6, Pages 987-991, 1990. –

NPL6 : K. Miyashita et al . , Journal of the Chemical Society, Perkin Transactions 1: Organic and Bio-Organic Chemistry (1972-1999) (1994) , (7) , Pages 847-851.

Summary of Invention

Technical Problem

It is an object of the present invention to solve the problems associated with the prior art, and to provide an improved and efficient method for the preparation of

(IS, 4S, 5S) -4-bromo-6-oxabicyclo [3.2.1] octan-7-one of formula (I).

Solution to Problem As a result of conducting diligent studies to attain the object, the present inventors have found that: surprisingly, the use of N-bromosuccinimide or bromohydantoin (representative is

1, 3-dibromo-5, 5-dimethylhydantoin) as brominating agent in the presence of a base selected from calcium oxide or calcium hydroxide, in specific mole ratios in a solvent selected from the group consisting of dichloromethane , toluene, tetrahydrofuran, ethyl acetate, hexanes, cyclopentyl methyl ether (CPME) or a mixture thereof can efficiently produce a pure

( IS , 4S , 5S) -4 -bromo- 6 -oxabicyclo [3.2.1] octan- 7 -one (I) in better yields. The process provides obvious benefits with respect to economics, convenience to operate at a commercial scale.

…………………………..

SEE

http://www.google.co.ug/patents/US20090105491

………………………….

PATENT

http://www.google.com/patents/EP2589590A1?cl=en

FREE BASE

      (Reference Example 6) N1-(5-Chloropyridin-2-yl)-N2-((1S,2R,4S)-4-[(dimethylamino)carbonyl]-2-{[(5-methyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridin-2-yl)carbonyl]amino}cyclohexyl)ethanediamide (X) (production method described in the pamphlet of International Publication No. WO 2007/032498)
  • Figure imgb0052
  • Methanesulfonic acid (66 ml) was added to a suspension of tert-butyl [(1R,2,S,5S)-2-({[(5-chloropyridin-2-yl)amino](oxo)acetyl}amino)-5-(dimethylaminocarbonyl)cyclohexyl]carbamate (5) (95.1 g) in acetonitrile (1900 ml) at room temperature, and the mixture was stirred at this temperature for 2 hours. To the reaction solution, triethylamine (155 ml), 5-methyl-4,5,6,7-tetrahydro[1,3]thiazolo[5,4-c]pyrzdine-2-carboxylic acid hydrochloride (8) (52.5 g), 1-hydroxybenzotriazole (33.0 g), and 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (46.8 g) were added under ice cooling, and the mixture was stirred at room temperature for 16 hours. Triethylamine and water were added thereto, and the mixture was stirred for 1 hour under ice cooling. Then, crystals were collected by filtration to obtain the title compound (X) (103.2 g). 1H-NMR (CDCl3) δ : 1.60-1.98 (3H, m), 2.00-2.16 (3H, m), 2.52 (3H, s), 2.78-2.90 (3H, m), 2.92-2.98 (2H, m), 2.95 (3H, s), 3.06 (3H, s), 3.69 (1H, d, J = 15.4 Hz), 3.75 (1H, d, J = 15.4 Hz), 4.07-4.15 (1H, m), 4.66-4.72 (1H, m), 7.40 (1H, dd, J = 8.8, 0.6 Hz), 7. 68 (1H, dd, J = 8.8, 2.4 Hz), 8.03 (1H, d, J = 7.8 Hz), 8.16 (1H, dd, J = 8.8, 0.6 Hz), 8.30 (1H, dd, J = 2. 4, 0.6 Hz), 9.72 (1H, s). MS (ESI) m/z: 548 (M+H)+.

TOSYLATE

      (Reference Example 7) N1-(5-Chloropyridin-2-yl)-N2-((1S,2R,4S)-4-[(dimethylamino)carbonyl]-2-{[(5-methyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridin-2-yl)carbonyl]amino}cyclohexyl)ethanediamide mono-p-toluenesulfonate monohydrate (X-a) (production method described in the pamphlet of International Publication No. WO 2007/032498)
  • Figure imgb0053
  • N1-(5-Chloropyridin-2-yl)-N2-((1S,2R,4S)-4-[(dimethylamino)carbonyl]-2-{[(5-methyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridin-2-yl)carbonyl]amino}cyclohexyl)ethanediamide (X) (6.2 g) was dissolved in methylene chloride (120 ml). To the solution, a 1 mol/L solution of p-toluenesulfonic acid in ethanol (11.28 ml) was added, and the solvent was distilled off. To the residue, 15% hydrous ethanol (95 ml) was added, and the mixture was dissolved by stirring at 60°C. Then, the mixture was cooled to room temperature and stirred for 1 day. The precipitated crystals were collected by filtration, washed with ethanol, and then dried under reduced pressure at room temperature for 2 hours to obtain the title compound (X-a) (7.4 g).
    1H-NMR (DMSO-d6) δ : 1. 45-1. 54 (1H, m), 1.66-1.78 (3H, m), 2.03-2.10 (2H, m), 2.28 (3H, s), 2.79 (3H, s), 2.91-3.02 (1H, m), 2.93 (3H, s), 2.99 (3H, s), 3.13-3.24 (2H, m), 3.46-3.82 (2H, m), 3.98-4.04 (1H, m), 4.43-4.80 (3H, m), 7.11 (2H, d, J = 7.8 Hz), 7.46 (2H, d, J = 8.2 Hz), 8.01 (2H, d, J = 1.8 Hz), 8.46 (1H, t, J = 1.8 Hz), 8.75 (1H, d, J = 6.9 Hz), 9.10-9.28 (1H, br), 10.18 (1H, br), 10.29 (1H, s).
    MS (ESI) m/z: 548 (M+H)+.
    Anal.: C24H30ClN7O4S·C7H8O3S·H2O
    Theoretical: C; 50.43, H; 5.46, N; 13.28, Cl; 4.80, S; 8.69.
    Found: C; 50.25, H; 5.36, N; 13.32, Cl; 4.93, S; 8.79. mp (dec.): 245-248°C.

……………………………………………..

PATENT

http://www.google.com/patents/EP2589590A1?cl=en

    • A compound represented by the following formula (X) [hereinafter, also referred to as compound (X)] or a pharmacologically acceptable salt thereof, or a hydrate thereof is a compound that exhibits an FXa inhibitory effect, as disclosed in Patent Literatures 1 to 3, and is useful as a preventive and/or therapeutic drug for thrombotic and/or embolic diseases:
    • Figure imgb0001
    • The pamphlet of International Publication No. WO 2007/032498discloses a process for preparing an FXa inhibitor compound (X) or a pharmacologically acceptable salt thereof, or a hydrate thereof. The process for producing compound (X) disclosed therein involves, as shown in [Scheme A] below, azidifying compound (2) to produce azide compound (3), subsequently reducing compound (3) into amino compound (1a), subsequently treating compound (1a) with anhydrous oxalic acid to obtain compound (1), which is then treated with compound (4) (ethyl[5-chloropyridin-2-yl]amino](oxo)acetate hydrochloride) in the presence of a base to produce compound (5), followed by several steps from compound (5). This pamphlet also discloses crystals of the oxalate of compound (1) as a production intermediate.
    • Figure imgb0002
    • wherein Boc represents a tert-butoxycarbonyl group.

Citation ListPatent Literatures

  1.  Furugohri T, Isobe K, Honda Y, Kamisato-Matsumoto C, Sugiyama N, Nagahara T, Morishima Y, Shibano T (September 2008). “DU-176b, a potent and orally active factor Xa inhibitor: in vitro and in vivo pharmacological profiles”. J. Thromb. Haemost.6 (9): 1542–9. doi:10.1111/j.1538-7836.2008.03064.xPMID18624979.
  2.  Raskob, G.; Cohen, A. T.; Eriksson, B. I.; Puskas, D.; Shi, M.; Bocanegra, T.; Weitz, J. I. (2010). “Oral direct factor Xa inhibition with edoxaban for thromboprophylaxis after elective total hip replacement”. Thrombosis and Haemostasis104 (3): 642–649. doi:10.1160/TH10-02-0142.PMID20589317edit
  3. “Phase III Trial Finds Edoxaban Outclasses Enoxaparin in Preventing Venous Thromboembolic Events”. 8 Dec 2010.
  4.  Weitz JI, Connolly SJ, Patel I, Salazar D, Rohatagi S, Mendell J, Kastrissios H, Jin J, Kunitada S (September 2010). “Randomised, parallel-group, multicentre, multinational phase 2 study comparing edoxaban, an oral factor Xa inhibitor, with warfarin for stroke prevention in patients with atrial fibrillation”. Thromb. Haemost.104 (3): 633–41. doi:10.1160/TH10-01-0066.
  5.  Edoxaban versus Warfarin in Patients with Atrial Fibrillation Robert P. Giugliano, M.D., Christian T. Ruff, M.D., M.P.H., Eugene Braunwald, M.D., Sabina A. Murphy, M.P.H., Stephen D. Wiviott, M.D., Jonathan L. Halperin, M.D., Albert L. Waldo, M.D., Michael D. Ezekowitz, M.D., D.Phil., Jeffrey I. Weitz, M.D., Jindřich Špinar, M.D., Witold Ruzyllo, M.D., Mikhail Ruda, M.D., Yukihiro Koretsune, M.D., Joshua Betcher, Ph.D., Minggao Shi, Ph.D., Laura T. Grip, A.B., Shirali P. Patel, B.S., Indravadan Patel, M.D., James J. Hanyok, Pharm.D., Michele Mercuri, M.D., and Elliott M. Antman, M.D. for the ENGAGE AF-TIMI 48 InvestigatorsDOI: 10.1056/NEJMoa1310907
  6.  “Edoxaban versus Warfarin for the Treatment of Symptomatic Venous Thromboembolism”. N. Engl. J. Med. August 2013. doi:10.1056/NEJMoa1306638PMID23991658.
  7. WO 03/000657 pamphlet WO 03/000680 pamphlet WO 03/016302 pamphlet WO 04/058715 pamphlet WO 05/047296 pamphlet WO 07/032498 pamphlet WO 08/129846 pamphlet WO 08/156159 pamphlet
  8. J Am Chem Soc 1978, 100(16): 5199
[1] 王利华, 赵丽嘉, 李文利, 等. 直接抑制凝血因子Xa 的口服抗凝药物Edoxaban Tosilate Hydrate [J]. 药物评价研究, 2011, 34(6): 478-481.
[2] Ohta T, Komoriya S, Yoshino T, et al. Preparation of N,N’-bis( heterocyclicacyl) cycloalkanediamine and heterocyclediamine derivatives as inhibitors of activated blood coagulation factor X (factor Xa): WO, 2003 000657 [P]. 2003-01-03. (CA 2003, 138: 73271)
[3] Ohta T, Komoriya S, Yoshino T, et al. Preparation of heterocyclic moiety-containing diamine derivatives as FXa inhibitors: WO, 2003 000680 [P]. 2003-01-03. (CA 2003, 138: 89801)
[4] Mochizuki A, Nagata T. Triamine derivative: WO, 2006106963 [P]. 2005-03-31. (CA 2006, 145: 419128)
[5] Kawanami K, Ishikawa H, Shoji M. Process for preparation of optically active (1S,3R,4R)-3-amino-4-hydroxy-N,Ndimethylcyclohexanecarboxamide derivative salt: WO, 2012002538 [P]. 2012-01-05. (CA 2012, 156: 122056)
[6] Sato K, Kubota K. Process for producing optically active carboxylic acid: WO, 2010067824 [P]. 2010-06-17. (CA 2010, 153: 36882)
[7] Yoshikawa K, Yokomizo A, Naito H, et al. Design, synthesis, and SAR of cis-1,2-diaminocyclohexane derivatives as potent factor Xa inhibitors. Part I: Exploration of 5-6fused rings
[8] as alternative S1 moieties [J]. Bioorg Med Chem, 2009, 17(24): 8206-8220.
[9] Sato K, Kawanami K, Yagi T. Process for the preparation of optically active cyclohexane-1,2-diamine derivative from 7-oxabicyclo[4.1.0]heptane compound: WO, 2007032498
[10] 2007-03-22. (CA 2007, 146: 358502)
[11] Kawanami K. Method for the preparation of optically active diamine derivative: WO, 2010104106 [P]. 2010-09-16. (CA 2010, 153: 406061)
[12] Koyama T, Kondo S. Process for the preparation of diamine derivative: WO, 2010104078 [P]. 2010-09-16. (CA 2010, 153: 382938)
[13] Suzuki T, Ono M. Crystal of diamine derivative and method of producing same: WO, 2011115066 [P]. 2011-09-22. (CA 2011, 155: 467954)

 

US8357808 9 Sep 2011 22 Jan 2013 Daiichi Sankyo Company, Limited Process for producing diamine derivative
US8394821 13 Jul 2011 12 Mar 2013 Daiichi Sankyo Company, Limited Activated blood coagulation factor inhibitor
US8404847 17 Jun 2011 26 Mar 2013 Daiichi Sankyo Company, Limited Method for producing diamine derivative
US8449896 16 Dec 2011 28 May 2013 Daiichi Sankyo Company, Limited Pharmaceutical composition having improved solubility
US8541443 19 Sep 2012 24 Sep 2013 Daiichi Sankyo Company, Limited Crystal of diamine derivative and method of producing same
US20130004550 * 22 Aug 2012 3 Jan 2013 Daiichi Sankyo Company, Limited Sustained-release solid preparation for oral use
WO2014081047A1 22 Nov 2013 30 May 2014 Daiichi Sankyo Company,Limited Process for the preparation of (1s,4s,5s)-4-bromo-6-oxabicyclo[3.2.1] octan-7-one

Molecular Formula C24H30ClN7O4S.C7H7HSO3
Molecular Weight 720.26
CAS Registry Number 480449-71-6 (912273-65-5)

Drug formulation , lixiana, edoxaban tosylate monohydrate, CAS 912273-65-5, C24 H30 Cl N7 O4 S . C7 H8 O3 S . H2 O, 738.274

    • N1-(5-chloropyridin-2-yl)-N2-((1S,2R,4S)-4-[(dimethylamino)carbonyl]-2-{[(5-methyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridin-2-yl)carbonyl]amino}cyclohexyl)ethanediamide p-toluenesulfonic acid monohydrate represented by the following formula (A) (hereinafter, also referred to as compound A) :
    • Figure imgb0001
      Figure imgb0002
    • is known as a compound that exhibits an inhibitory effect on activated blood coagulation factor X (FXa), and is useful as a preventive and/or therapeutic drug for thrombotic diseases (Patent Literature 1 to 8).
    • For example, a method comprising mixing the free form of compound A represented by the following formula (B) (hereinafter, also referred to as compound B):
    • Figure imgb0003
    • with p-toluenesulfonic acid or p-toluenesulfonic acid monohydrate, followed by crystallization from aqueous ethanol, is known as a method for obtaining compound A (Patent Literature 1 to 8). These literature documents do not make any mention about adding p-toluenesulfonic acid or p-toluenesulfonic acid monohydrate in a stepwise manner in the step of obtaining compound A from compound B.

Citation ListPatent Literature

    • Patent Literature 1: International Publication No. WO 03/000657
    • Patent Literature 2: International Publication No. WO 03/000680
    • Patent Literature 3: International Publication No. WO 03/016302
    • Patent Literature 4: International Publication No. WO 04/058715
    • Patent Literature 5: International Publication No. WO 05/047296
    • Patent Literature 6: International Publication No. WO 07/032498
    • Patent Literature 7: International Publication No. WO 08/129846
    • Patent Literature 8: International Publication No. WO 08/156159

SIMILAR

OTHER SALTS

Edoxaban hydrochloride
CAS Number: 480448-29-1
Molecular Formula: C24H30ClN7O4S · HCl
Molecular Weight: 584.52 g.mol-1

Edoxaban is reported to be a member of the so-called “Xaban-group” and as such to be a low molecular inhibitor of the enzyme factor Xa, participating in the blood coagulation system. Therefore, edoxaban is classified as an antithrombotic drug and its possible medical indications are reported to be treatment of thrombosis and thrombosis prophylaxis after orthopaedic operations, such as total hip replacement, as well as for stroke prevention in patients with atrial fibrillation, the prophylaxis of the acute coronary syndrome and the prophylaxis after thrombosis and pulmonary embolism.

The IUPAC name for edoxaban is N’-(5-chloropyridin-2-yl)-N-[(15,2^,4S)-4- (dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[l ,3]thiazolo[5,4-c]pyridine-2- carbonyl)amino]cyclohexyl]oxamide. The chemical structure of edoxaban is shown in the formula (1) below:

Figure imgf000002_0001

formula ( 1 ) While Edoxaban is reported to be soluble in strongly acidic aqueous solutions, its solubility is considered to be very low in neutral or alkaline aqueous media. EP 2 140 867 A 1 claims an edoxaban-containing pharmaceutical composition comprising a water-swelling additive and/or a sugar alcohol. Further, it is alleged that compositions comprising lactose or cornstarch do not have good dissolution properties. The claimed pharmaceutical compositions in EP 2 140 867 Al are considered to show good dissolution properties in a neutral aqueous medium as well. Tablets comprising said composition were produced by wet granulation. However, it turned out that prior art pharmaceutical formulations comprising edoxaban being suitable for oral administration are still improvable with regards to dissolution rate and bioavailability. Further, stability and content uniformity of the known formulations could be improved. Further, due to the intolerance of many people to sugar alcohol(s), such as sorbitol, the use of sugar alcohol(s) should be avoided.

UPDATE

2-amino-5-methyl-4,5,6,7-tetrahydro thiazolone [5,4-c] pyridine 

WO2015125710

(Reference Example 1) 2-amino-5-methyl-4,5,6,7-tetrahydro thiazolone [5,4-c] pyridine (1-n) (The method described in WO 2005/047296 Pamphlet )
[0091]
[Of 35]  in 2-PrOH (1.44L) solution was heated to 50 ℃ 1- methyl-4-piperidone (180.0g), 2-PrOH (360mL) solution of cyanamide (67.0g), and sulfur powder (51.0 g) it was added. Pyrrolidine (13.3mL) was added to the reaction mixture, after stirring for 2 hours at 50 ℃, followed by stirring overnight and allowed to cool to room temperature.  The reaction mixture was cooled to 10 ℃ less in an ice water bath and stirred for 1 hour at the same temperature. Is filtered and the precipitated crystals were washed with 2-PrOH (540mL), the title compound was dried under reduced pressure at 40 ℃ (209.9g, 78%) was obtained.

[0092]
1 H-NMR (CDCl 3 ) ppm: 4.86 (Br, 2H), 3.47-3.46 (t, 2H, J = 1.9 Hz), 2.78-2.71 (M, 2H), 2.71-2.65 (M, 2H), 2.47 . (s, 3H)
MS (FAB) M / z: 170 (M + H) +
elemental analysis: C 7 H 11 N 3 as S,
theoretical value: C, 49.68; H, 6.55; N, 24.83; S, 18.95
measured value: C, 49.70; H, 6.39; N, 24.91; S, 19.00.

WO2015125710

 (Example 11) 1 – (5-Chloro-2-yl) -N 2 – [(1S, 2R, 4S)-4-(dimethylcarbamoyl) -2 – {[(5-methyl-4,5 , 6,7-tetrahydro [1,3] thiazolo [5,4-c] pyridin-2-yl) carbonyl] amino} cyclohexyl] Etanjiamido (X) [Production method via Compound (1-p2)]
[0137]
 In 10 mL test tube, compound (5-ms) (the compound of Reference Example 8) (100 mg, 0.216 mmol), Compound (1-p2) (81.4 mg, 0.216 mmol), K 3 PO 4 (91.7 mg, 0.432 mmol) and DMF (1 mL) was added, and the mixture was stirred at room temperature conditions for 3 hours. H To the reaction mixture 2 O (2 mL) was added and the resulting slurry was stirred at room temperature overnight, the solid was filtered. The resulting solid H 2 was washed with O (1 mL), was obtained by drying under reduced pressure the title compound (110.0 mg, 92.9%) as a solid.
[0138]
1 H-NMR (500 Hz, CDCl 3 ) delta: 9.72 (s, 1H), 8.30 (dd, 1H, J = 2.5, 0.5 Hz), 8.17 (dd, 1H, J = 9.0, 0.5 Hz), 8.03 (D , 1H, J = 8.5 Hz), 7.68 (dd, 1H, J = 9.0, 2.5 Hz), 7.39 (d, 1H, J = 8.5 Hz), 4.70-4.67 (m, 1H), 4.13-4.09 (m, 1H), 3.73 (d, 1H, J = 16.0 Hz), 3.70 (d, 1H, J = 16.0 Hz), 3.06 (s, 3H), 2.96-2.93 (m, 2H), 2.95 (s, 3H), 2.89-2.79 (m, 3H), 2.52 (s, 3H), 2.14-2.06 (m, 3H), 1.96-1.90 (m, 1H), 1.84-1.78 (m, 1H), 1.69-1.62 (m, 1H ).

UPDATE

Edoxaban, DU-176b

1H NMR PREDICTION

edoxaban NMR spectra analysis, Chemical CAS NO. 480449-70-5 NMR spectral analysis, edoxaban H-NMR spectrum

………….

13 C NMR

edoxaban NMR spectra analysis, Chemical CAS NO. 480449-70-5 NMR spectral analysis, edoxaban C-NMR spectrum

FREE BASE

      (Reference Example 6) N1-(5-Chloropyridin-2-yl)-N2-((1S,2R,4S)-4-[(dimethylamino)carbonyl]-2-{[(5-methyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridin-2-yl)carbonyl]amino}cyclohexyl)ethanediamide (X) (production method described in the pamphlet of International Publication No. WO 2007/032498)
  • Figure imgb0052

  • Methanesulfonic acid (66 ml) was added to a suspension of tert-butyl [(1R,2,S,5S)-2-({[(5-chloropyridin-2-yl)amino](oxo)acetyl}amino)-5-(dimethylaminocarbonyl)cyclohexyl]carbamate (5) (95.1 g) in acetonitrile (1900 ml) at room temperature, and the mixture was stirred at this temperature for 2 hours. To the reaction solution, triethylamine (155 ml), 5-methyl-4,5,6,7-tetrahydro[1,3]thiazolo[5,4-c]pyrzdine-2-carboxylic acid hydrochloride (8) (52.5 g), 1-hydroxybenzotriazole (33.0 g), and 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (46.8 g) were added under ice cooling, and the mixture was stirred at room temperature for 16 hours. Triethylamine and water were added thereto, and the mixture was stirred for 1 hour under ice cooling. Then, crystals were collected by filtration to obtain the title compound (X) (103.2 g).

  • 1H-NMR (CDCl3) δ : 1.60-1.98 (3H, m), 2.00-2.16 (3H, m), 2.52 (3H, s), 2.78-2.90 (3H, m), 2.92-2.98 (2H, m), 2.95 (3H, s), 3.06 (3H, s), 3.69 (1H, d, J = 15.4 Hz), 3.75 (1H, d, J = 15.4 Hz), 4.07-4.15 (1H, m), 4.66-4.72 (1H, m), 7.40 (1H, dd, J = 8.8, 0.6 Hz), 7. 68 (1H, dd, J = 8.8, 2.4 Hz), 8.03 (1H, d, J = 7.8 Hz), 8.16 (1H, dd, J = 8.8, 0.6 Hz), 8.30 (1H, dd, J = 2. 4, 0.6 Hz), 9.72 (1H, s).

  • MS (ESI) m/z: 548 (M+H)+.


Molecular Formula C24H30ClN7O4S.C7H7HSO3
Molecular Weight 720.26
CAS Registry Number 480449-71-6 (912273-65-5)

TOSYLATE

      (Reference Example 7) N1-(5-Chloropyridin-2-yl)-N2-((1S,2R,4S)-4-[(dimethylamino)carbonyl]-2-{[(5-methyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridin-2-yl)carbonyl]amino}cyclohexyl)ethanediamide mono-p-toluenesulfonate monohydrate (X-a) (production method described in the pamphlet of International Publication No. WO 2007/032498)
  • Figure imgb0053

  • N1-(5-Chloropyridin-2-yl)-N2-((1S,2R,4S)-4-[(dimethylamino)carbonyl]-2-{[(5-methyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridin-2-yl)carbonyl]amino}cyclohexyl)ethanediamide (X) (6.2 g) was dissolved in methylene chloride (120 ml). To the solution, a 1 mol/L solution of p-toluenesulfonic acid in ethanol (11.28 ml) was added, and the solvent was distilled off. To the residue, 15% hydrous ethanol (95 ml) was added, and the mixture was dissolved by stirring at 60°C. Then, the mixture was cooled to room temperature and stirred for 1 day. The precipitated crystals were collected by filtration, washed with ethanol, and then dried under reduced pressure at room temperature for 2 hours to obtain the title compound (X-a) (7.4 g).

  • 1H-NMR (DMSO-d6) δ : 1. 45-1. 54 (1H, m), 1.66-1.78 (3H, m), 2.03-2.10 (2H, m), 2.28 (3H, s), 2.79 (3H, s), 2.91-3.02 (1H, m), 2.93 (3H, s), 2.99 (3H, s), 3.13-3.24 (2H, m), 3.46-3.82 (2H, m), 3.98-4.04 (1H, m), 4.43-4.80 (3H, m), 7.11 (2H, d, J = 7.8 Hz), 7.46 (2H, d, J = 8.2 Hz), 8.01 (2H, d, J = 1.8 Hz), 8.46 (1H, t, J = 1.8 Hz), 8.75 (1H, d, J = 6.9 Hz), 9.10-9.28 (1H, br), 10.18 (1H, br), 10.29 (1H, s).

    MS (ESI) m/z: 548 (M+H)+.

    Anal.: C24H30ClN7O4S·C7H8O3S·H2O

    Theoretical: C; 50.43, H; 5.46, N; 13.28, Cl; 4.80, S; 8.69.

    Found: C; 50.25, H; 5.36, N; 13.32, Cl; 4.93, S; 8.79. mp (dec.): 245-248°C.

1H NMR PREDICTION, TOSYLATE

CAS NO. 1229194-11-9, N’-(5-chloropyridin-2-yl)-N-[(1S,2R,4S)-4-(dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[1,3]thiazolo[5,4-c]pyridine-2-carbonyl)amino]cyclohexyl]oxamide,4-methylbenzenesulfonic acid,hydrate H-NMR spectral analysis

N'-(5-chloropyridin-2-yl)-N-[(1S,2R,4S)-4-(dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[1,3]thiazolo[5,4-c]pyridine-2-carbonyl)amino]cyclohexyl]oxamide,4-methylbenzenesulfonic acid,hydrate NMR spectra analysis, Chemical CAS NO. 1229194-11-9 NMR spectral analysis, N'-(5-chloropyridin-2-yl)-N-[(1S,2R,4S)-4-(dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[1,3]thiazolo[5,4-c]pyridine-2-carbonyl)amino]cyclohexyl]oxamide,4-methylbenzenesulfonic acid,hydrate H-NMR spectrum

13 CNMR PREDICTION, TOSYLATE

CAS NO. 1229194-11-9, N’-(5-chloropyridin-2-yl)-N-[(1S,2R,4S)-4-(dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[1,3]thiazolo[5,4-c]pyridine-2-carbonyl)amino]cyclohexyl]oxamide,4-methylbenzenesulfonic acid,hydrate C-NMR spectral analysis

N'-(5-chloropyridin-2-yl)-N-[(1S,2R,4S)-4-(dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[1,3]thiazolo[5,4-c]pyridine-2-carbonyl)amino]cyclohexyl]oxamide,4-methylbenzenesulfonic acid,hydrate NMR spectra analysis, Chemical CAS NO. 1229194-11-9 NMR spectral analysis, N'-(5-chloropyridin-2-yl)-N-[(1S,2R,4S)-4-(dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[1,3]thiazolo[5,4-c]pyridine-2-carbonyl)amino]cyclohexyl]oxamide,4-methylbenzenesulfonic acid,hydrate C-NMR spectrum

………………

WO2015125710

 (Example 11) 1 – (5-Chloro-2-yl) -N 2 – [(1S, 2R, 4S)-4-(dimethylcarbamoyl) -2 – {[(5-methyl-4,5 , 6,7-tetrahydro [1,3] thiazolo [5,4-c] pyridin-2-yl) carbonyl] amino} cyclohexyl] Etanjiamido (X) [Production method via Compound (1-p2)]

[0137]

 In 10 mL test tube, compound (5-ms) (the compound of Reference Example 8) (100 mg, 0.216 mmol), Compound (1-p2) (81.4 mg, 0.216 mmol), K 3 PO 4 (91.7 mg, 0.432 mmol) and DMF (1 mL) was added, and the mixture was stirred at room temperature conditions for 3 hours. H To the reaction mixture 2 O (2 mL) was added and the resulting slurry was stirred at room temperature overnight, the solid was filtered. The resulting solid H 2 was washed with O (1 mL), was obtained by drying under reduced pressure the title compound (110.0 mg, 92.9%) as a solid.

[0138]

1 H-NMR (500 Hz, CDCl 3 ) delta: 9.72 (s, 1H), 8.30 (dd, 1H, J = 2.5, 0.5 Hz), 8.17 (dd, 1H, J = 9.0, 0.5 Hz), 8.03 (D , 1H, J = 8.5 Hz), 7.68 (dd, 1H, J = 9.0, 2.5 Hz), 7.39 (d, 1H, J = 8.5 Hz), 4.70-4.67 (m, 1H), 4.13-4.09 (m, 1H), 3.73 (d, 1H, J = 16.0 Hz), 3.70 (d, 1H, J = 16.0 Hz), 3.06 (s, 3H), 2.96-2.93 (m, 2H), 2.95 (s, 3H), 2.89-2.79 (m, 3H), 2.52 (s, 3H), 2.14-2.06 (m, 3H), 1.96-1.90 (m, 1H), 1.84-1.78 (m, 1H), 1.69-1.62 (m, 1H ).

References

  1. “First market approval in Japan for LIXIANA (Edoxaban)”. Press Release. Daiichi Sankyo Europe GmbH. 2011-04-22.
  2. O’Riordan, Michael (9 January 2015). “FDA Approves Edoxaban for Stroke Prevention in AF and DVT/PE Prevention”. Medscape. Retrieved 10 January 2015.
  3. http://www.accessdata.fda.gov/drugsatfda_docs/label/2015/206316lbl.pdf
  4. lexicomp.com
  5. Savaysa (edoxaban) [prescribing information]. Parsippany, NJ: Daiichi Sankyo; January 2015.
  6. http://www.drugs.com/cons/edoxaban.html
  7. Yoshiyuki, I., et al. “Biochemical and pharmalogical profile of darexaban, an oral direct Xa inhibitor.” European Journal of Pharmacology (2011): 49-55
  8. Katsung, B., S. Masters and A. Trevor. Basic and Clinical Pharmacology 11th Edition. United States of America: McGraw-Hill, 2009
  9. Turpie AG (January 2008). “New oral anticoagulants in atrial fibrillation”. European Heart Journal 29 (2): 155–65. doi:10.1093/eurheartj/ehm575. PMID 18096568.

Edoxaban, a factor Xa inhibitor, is supplied as edoxaban tosylate monohydrate. The chemical name is N-(5-Chloropyridin-2-yl)-N’-[(1S,2R,4S)-4-(N,N-dimethylcarbamoyl)-2-(5-methyl- 4,5,6,7-tetrahydro[1,3]thiazolo[5,4-c]pyridine-2-carboxamido)cyclohexyl] oxamide mono (4- methylbenzenesulfonate) monohydrate. Edoxaban tosylate monohydrate has the empirical formula C24H30ClN7O4S•C7H8O3S•H2O representing a molecular weight of 738.27. The chemical structure of edoxaban tosylate monohydrate is:

SAVAYSA (edoxaban) Structural Formula Illustration

It is a white to pale yellowish-white crystalline powder. The solubility of edoxaban tosylate (pKa 6.7) decreases with increasing pH. It is slightly soluble in water, pH 3 to 5 buffer, very slightly soluble at pH 6 to 7; and practically insoluble at pH 8 to 9.

SAVAYSA is available for oral administration as a 60 mg, 30 mg, or 15 mg round shaped, film-coated tablet, debossed with product identification markings. Each 60 mg tablet contains 80.82 mg edoxaban tosylate monohydrate equivalent to 60 mg of edoxaban. Each 30 mg tablet contains 40.41 mg edoxaban tosylate monohydrate equivalent to 30 mg of edoxaban. Each 15 tablet contains 20.20 mg edoxaban tosylate monohydrate equivalent to 15 mg of edoxaban.

The inactive ingredients are: mannitol, pregelatinized starch, crospovidone, hydroxypropyl cellulose, magnesium stearate, talc, and carnauba wax. The color coatings contain hypromellose, titanium dioxide, talc, polyethylene glycol 8000, iron oxide yellow (60 mg tablets and 15 mg tablets), and iron oxide red (30 mg tablets and 15 mg tablets).

Edoxaban
Edoxaban.svg
Systematic (IUPAC) name
N’-(5-chloropyridin-2-yl)-N-[(1S,2R,4S)-4-(dimethylcarbamoyl)-2-[(5-methyl-6,7-dihydro-4H-[1,3]thiazolo[5,4-c]pyridine-2-carbonyl)amino]cyclohexyl]oxamide
Clinical data
Trade names Lixiana, Savaysa
AHFS/Drugs.com Monograph
Pregnancy
category
  • US: C (Risk not ruled out)
Legal status
Routes of
administration
Oral
Pharmacokinetic data
Bioavailability 62%; Tmax 1–2 hours
Protein binding 55%
Metabolism Minimal hepatic
Biological half-life 10–14 hours
Excretion 50% renal; <50% bile
Identifiers
CAS Registry Number 912273-65-5 Yes
ATC code None
PubChem CID: 25022378
IUPHAR/BPS 7575
ChemSpider 8456212 
UNII NDU3J18APO 
KEGG D09710 
ChEBI CHEBI:85973 Yes
Chemical data
Formula C24H30ClN7O4S
Molecular mass 548.056 g/mol

/////////

SEE ABAN SERIES AT…………http://organicsynthesisinternational.blogspot.in/p/aban-series.html

 

 

Eisai’s lenvatinib 兰伐替尼 レンバチニブ to get speedy review in Europe


Lenvatinib skeletal.svg

Lenvatinib

For the treatment of patients with progressive radioiodine-refractory, differentiated thyroid cancer (RR-DTC).

CAS  417716-92-8,
 CAS 857890-39-2 (lenvatinib mesylate)
E 7080, ER-203492-00, E7080, E 7080,
4-[3-Chloro-4-(cyclopropylaminocarbonyl)aminophenoxy]-7-methoxy-6-quinolinecarboxamide
Molecular Formula: C21H19ClN4O4
Molecular Weight: 426.85296
Eisai Co., Ltd INNOVATOR

European regulators have agreed to undertake an accelerated assessment of Eisai’s lenvatinib as a treatment for progressive radioiodine-refractory, differentiated thyroid cancer.

The drug, which carries Orphan Status in the EU, is to be filed “imminently” and could become the first in a new class of tyrosine kinase inhibitors, the drugmaker said.

Read more at: http://www.pharmatimes.com/Article/14-07-31/Eisai_s_lenvatinib_to_get_speedy_review_in_Europe.aspx#ixzz39OGhRHas

Lenvatinib was granted Orphan Drug Designation for thyroid cancer by the health authorities in Japan in 2012, and in Europe and the U.S in 2013. The first application for marketing authorization of lenvatinib in the world was submitted in Japan on June 2014. Eisai is planning to submit applications for marketing authorization in Europe and the U.S. in the second quarter of fiscal 2014.

Lenvatinib is an oral multiple receptor tyrosine kinase (RTK) inhibitor with a novel binding mode that selectively inhibits the kinase activities of vascular endothelial growth factor receptors (VEGFR), in addition to other proangiogenic and oncogenic pathway-related RTKs including fibroblast growth factor receptors (FGFR), the platelet-derived growth factor (PDGF) receptor PDGFRalpha, KIT and RET that are involved in tumor proliferation. This potentially makes lenvatinib a first-in-class treatment, especially given that it simultaneously inhibits the kinase activities of FGFR as well as VEGFR.

Eisai's lenvatinib to get speedy review in Europe

LENVATINIB BASE

COSY PREDICT

COSY  LENVA BASE

Systematic (IUPAC) name
4-[3-chloro-4-(cyclopropylcarbamoylamino)phenoxy]-7-methoxy-quinoline-6-carboxamide
Clinical data
Legal status Prescription only
Identifiers
CAS number
ATC code None
PubChem CID 9823820
ChemSpider 7999567 Yes
UNII EE083865G2 Yes
Chemical data
Formula C21H19ClN4O4 
Mol. mass 426.853 g/mol

Lenvatinib (E7080) is a multi-kinase inhibitor that is being investigated for the treatment of various types of cancer by Eisai Co. It inhibits both VEGFR2 and VEGFR3 kinases.[1]

The substence was granted orphan drug status for the treatment of various types of thyroid cancer that do not respond toradioiodine; in the US and Japan in 2012 and in Europe in 2013[2] and is now approved for this use.

Clinical trials

Lenvatinib has had promising results from a phase I clinical trial in 2006[3] and is being tested in several phase II trials as of October 2011, for example against hepatocellular carcinoma.[4] After a phase II trial testing the treatment of thyroid cancer has been completed with modestly encouraging results,[5] the manufacturer launched a phase III trial in March 2011.[6]

Chemical structure for Lenvatinib

Lenvatinib Mesilate

Molecular formula: C21H19ClN4O4,CH4O3S =523.0.

CAS: 857890-39-2.

UNII code: 3J78384F61.

About the Lenvatinib (E7080) Phase II Study
The open-label, global, single-arm Phase II study of multi-targeted kinase inhibitor lenvatinib (E7080) in advanced radioiodine (RAI)-refractory differentiated thyroid cancer involved 58 patients with advanced RAI refractory DTC (papillary, follicular or Hurthle Cell) whose disease had progressed during the prior 12 months. (Disease progression was measured using Response Evaluation Criteria in Solid Tumors (RECIST).) The starting dose of lenvatinib was 24 mg once daily in repeated 28 day cycles until disease progression or development of unmanageable toxicities.

2.   About Thyroid Cancer
Thyroid cancer refers to cancer that forms in the tissues of the thyroid gland, located at the base of the throat or near the trachea. It affects more women than men and usually occurs between the ages of 25 and 65.
The most common types of thyroid cancer, papillary and follicular (including Hurthle Cell), are classified as differentiated thyroid cancer and account for 95 percent of all cases. While most of these are curable with surgery and radioactive iodine treatment, a small percentage of patients do not respond to therapy.

3.   About Lenvatinib (E7080)
Lenvatinib is multi-targeted kinase inhibitor with a unique receptor tyrosine kinase inhibitory profile that was discovered and developed by the Discovery Research team of Eisai’s Oncology Unit using medicinal chemistry technology. As an anti-angiogenic agent, it inhibits tyrosine kinase of the VEGF (Vascular Endothelial Growth Factor) receptor, VEGFR2, and a number of other types of kinase involved in angiogenesis and tumor proliferation in balanced manner. It is a small molecular targeting drug that is currently being studied in a wide array of cancer types.

4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (additional name: 4-[3-chloro-4-(N′-cyclopropylureido)phenoxy]-7-methoxyquinoline-6-carboxamide) is known to exhibit an excellent angiogenesis inhibition as a free-form product, as described in Example 368 of Patent Document 1. 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide is also known to exhibit a strong inhibitory action for c-Kit kinase (Non-Patent Document 1, Patent Document 2).

However, there has been a long-felt need for the provision of a c-Kit kinase inhibitor or angiogenesis inhibitor that has high usability as a medicament and superior characteristics in terms of physical properties and pharmacokinetics in comparison with the free-form product of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide.

[Patent Document 1] WO 02/32872

[Patent Document 2] WO 2004/080462

[Non-Patent Document 1] 95th Annual Meeting Proceedings, AACR (American Association for Cancer Research), Volume 45, Page 1070-1071, 2004

………………………..

PATENT

http://www.google.co.in/patents/US8058474

EXAMPLES

Examples will now be described to facilitate understanding of the invention, but the invention is not limited to these examples.

Example 1Phenyl N-(2-chloro-4-hydroxyphenyl)carbamate

After suspending 4-amino-3-chlorophenol (23.7 g) in N,N-dimethylformamide (100 mL) and adding pyridine (23.4 mL) while cooling on ice, phenyl chloroformate (23.2 ml) was added dropwise below 20° C. Stirring was performed at room temperature for 30 minutes, and then water (400 mL), ethyl acetate (300 mL) and 6N HCl (48 mL) were added, the mixture was stirred and the organic layer was separated. The organic layer was washed twice with 10% brine (200 mL), and dried over magnesium sulfate. The solvent was removed to give 46 g of the title compound as a solid.

1H-NMR (CDCl3): 5.12 (1h, br s), 6.75 (1H, dd, J=9.2, 2.8 Hz), 6.92 (1H, d, J=2.8 Hz), 7.18-7.28 (4H, m), 7.37-7.43 (2H, m), 7.94 (1H, br s)

Example 21-(2-chloro-4-hydroxyphenyl)-3-cyclopropylurea

After dissolving phenyl N-(2-chloro-4-hydroxyphenyl)carbamate in N,N-dimethylformamide (100 mL), cyclopropylamine (22.7 mL) was added while cooling on ice and the mixture was stirred overnight at room temperature. Water (400 mL), ethyl acetate (300 mL) and 6N HCl (55 mL) were then added, the mixture was stirred and the organic layer was separated. The organic layer was washed twice with 10% brine (200 mL), and dried over magnesium sulfate. Prism crystals obtained by concentrating the solvent were filtered and washed with heptane to give 22.8 g of the title compound (77% yield from 4-amino-3-chlorophenol).

1H-NMR (CDCl3): 0.72-0.77 (2H, m), 0.87-0.95 (2H, m), 2.60-2.65 (1H, m), 4.89 (1H, br s), 5.60 (1H, br s), 6.71 (1H, dd, J=8.8, 2.8 Hz), 6.88 (1H, d, J=2.8 Hz), 7.24-7.30 (1H, br s), 7.90 (1H, d, J=8.8H)

Example 34-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide

To dimethylsulfoxide (20 mL) were added 7-methoxy-4-chloro-quinoline-6-carboxamide (0.983 g), 1-(2-chloro-4-hydroxyphenyl)-3-cyclopropylurea (1.13 g) and cesium carbonate (2.71 g), followed by heating and stirring at 70° C. for 23 hours. After the reaction mixture was allowed to cool down to room temperature, water (50 mL) was added, and the produced crystals were collected by filtration to give 1.56 g of the title compound (88% yield).

1H-NMR (d6-DMSO): 0.41 (2H, m), 0.66 (2H, m), 2.56 (1H, m), 4.01 (3H, s), 6.51 (1H, d, J=5.6 Hz), 7.18 (1H, d, J=2.8 Hz), 7.23 (1H, dd, J=2.8, 8.8 Hz), 7.48 (1H, d, J=2.8 Hz), 7.50 (1H, s), 7.72 (1H, s), 7.84 (1H, s), 7.97 (1H, s), 8.25 (1H, d, J=8.8 Hz), 8.64 (1H, s), 8.65 (1H, d, J=5.6 Hz)

Example 44-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide

In a reaction vessel were placed 7-methoxy-4-chloro-quinoline-6-carboxamide (5.00 kg, 21.13 mol), dimethylsulfoxide (55.05 kg), 1-(2-chloro-4-hydroxyphenyl)-3-cyclopropylurea (5.75 kg, 25.35 mol) and potassium t-butoxide (2.85 kg, 25.35 mol) in that order, under a nitrogen atmosphere. After stirring at 20° C. for 30 minutes, the temperature was raised to 65° C. over a period of 2.5 hours. After stirring at the same temperature for 19 hours, 33% (v/v) acetone water (5.0 L) and water (10.0 L) were added dropwise over a period of 3.5 hours. Upon completion of the dropwise addition, the mixture was stirred at 60° C. for 2 hours, and 33% (v/v) acetone water (20.0 L) and water (40.0 L) were added dropwise at 55° C. or higher over a period of 1 hour. After then stirring at 40° C. for 16 hours, the precipitated crystals were collected by filtration using a nitrogen pressure filter, and the crystals were washed with 33% (v/v) acetone water (33.3 L), water (66.7 L) and acetone (50.0 L) in that order. The obtained crystals were dried at 60° C. for 22 hours using a conical vacuum drier to give 7.78 kg of the title compound (96.3% yield).

…………………………

SYNTHESIS

SYN YAOPHA

1H NMR PREDICT

NMR 1H GRAPH NMR 1H VAL

13 C NMR PREDICT

NMR 13C GRAPH NMR 13C VAL

…………………..

PATENT

http://www.google.co.in/patents/US7253286

EX 368

Figure US07253286-20070807-C00838

Example 368

4-(3-Chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide

The title compound (22.4 mg, 0.052 mmol, 34.8%) was obtained as white crystals from phenyl N-(4-(6-carbamoyl-7-methoxy-4-quinolyl)oxy-2-chlorophenyl)carbamate (70 mg, 0.15 mmol) and cyclopropylamine, by the same procedure as in Example 11.

1H-NMR Spectrum (DMSO-d6) δ (ppm): 0.41 (2H, m), 0.66 (2H, m), 2.56 (1H, m), 4.01 (3H, s), 6.51 (1H, d, J=5.6 Hz), 7.18 (1H, d, J=2.8 Hz), 7.23 (1H, dd, J=2.8, 8.8 Hz), 7.48 (1H, d, J=2.8 Hz), 7.50 (1H, s), 7.72 (1H, s), 7.84 (1H, s), 7.97 (1H, s), 8.25 (1H, d, J=8.8 Hz), 8.64 (1H, s), 8.65 (1H, d, J=5.6 Hz).

The starting material was synthesized in the following manner.

Production Example 368-1Phenyl N-(4-(6-carbamoyl-7-methoxy-4-quinolyl)oxy-2-chlorophenyl)carbamate

The title compound (708 mg, 1.526 mmol, 87.4%) was obtained as light brown crystals from 4-(4-amino-3-chlorophenoxy)-7-methoxy-6-quinolinecarboxamide (600 mg, 1.745 mmol), by the same procedure as in Production Example 17.

1H-NMR Spectrum (CDCl3) δ (ppm): 4.14 (3H, s), 5.89 (1H, br), 6.50 (1H, d, J=5.6 Hz), 7.16 (2H, dd, J=2.4, 8.8 Hz), 7.22–7.30 (4H, m), 7.44 (2H, m), 7.55 (1H, s), 7.81 (1H, br), 8.31 (1H, d, J=8.8 Hz), 8.68 (1H, d, J=5.6 Hz), 9.27 (1H, s).

……………………

CRYSTALLINE FORM

http://www.google.co.in/patents/US7612208

Preparation Example 1

Preparation of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (1)

Phenyl N-(4-(6-carbamoyl-7-methoxy-4-quinolyl)oxy-2-chlorophenyl)carbamate (17.5 g, 37.7 mmol) disclosed in WO 02/32872 was dissolved in N,N-dimethylformamide (350 mL), and then cyclopropylamine (6.53 mL, 94.25 mmol) was added to the reaction mixture under a nitrogen atmosphere, followed by stirring overnight at room temperature. To the mixture was added water (1.75 L), and the mixture was stirred. Precipitated crude crystals were filtered off, washed with water, and dried at 70° C. for 50 min. To the obtained crude crystals was added ethanol (300 mL), and then the mixture was heated under reflux for 30 min to dissolve, followed by stirring overnight to cool slowly down to room temperature. Precipitated crystals was filtered off and dried under vacuum, and then further dried at 70° C. for 8 hours to give the titled crystals (12.91 g; 80.2%).

Preparation Example 2Preparation of 4-(3-cloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (2)

(1) Preparation of phenyl N-(2-chloro-4-hydroxyphenyl)carbamate

To a suspension of 4-amino-3-chlorophenol (23.7 g) in N,N-dimethylformamide (100 mL) was added pyridine (23.4 mL) while cooling in an ice bath, and phenyl chloroformate (23.2 mL) was added dropwise below 20° C. After stirring at room temperature for 30 min, water (400 mL), ethyl acetate (300 mL), and 6N-HCl (48 mL) were added and stirred. The organic layer was separated off, washed twice with a 10% aqueous sodium chloride solution (200 mL), and dried over magnesium sulfate. The solvent was evaporated to give 46 g of the titled compound as a solid.

  • 1H-NMR Spectrum (CDCl3) δ(ppm): 5.12 (1H, br s), 6.75 (1H, dd, J=9.2, 2.8 Hz), 6.92 (1H, d, J=2.8 Hz), 7.18-7.28 (4H, m), 7.37-7.43 (2H, m), 7.94 (1H, br s).
    (2) Preparation of 1-(2-chloro-4-hydroxyphenyl)-3-cyclopropylurea

To a solution of phenyl N-(2-chloro-4-hydroxyphenyl)carbamate in N,N-dimethylformamide (100 mL) was added cyclopropylamine (22.7 mL) while cooling in an ice bath, and the stirring was continued at room temperature overnight. Water (400 mL), ethyl acetate (300 mL), and 6N-HCl (55 mL) were added thereto, and the mixture was stirred. The organic layer was then separated off, washed twice with a 10% aqueous sodium chloride solution (200 mL), and dried over magnesium sulfate. The solvent was evaporated to give prism crystals, which were filtered off and washed with heptane to give 22.8 g of the titled compound (yield from 4-amino-3-chlorophenol: 77%).

  • 1H-NMR Spectrum (CDCl3) δ(ppm): 0.72-0.77 (2H, m), 0.87-0.95 (2H, m), 2.60-2.65 (1H, m), 4.89 (1H, br s), 5.60 (1H, br s), 6.71 (1H, dd, J=8.8, 2.8 Hz), 6.88 (1H, d, J=2.8 Hz), 7.24-7.30 (1H, br s), 7.90 (1H, d, J=8.8 Hz)
    (3) Preparation of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide

To dimethyl sulfoxide (20 mL) were added 7-methoxy-4-chloroquinoline-6-carboxamide (0.983 g), 1-(2-chloro-4-hydroxyphenyl)-3-cyclopropylurea (1.13 g) and cesium carbonate (2.71 g), and the mixture was heated and stirred at 70° C. for 23 hours. The reaction mixture was cooled to room temperature, and water (50 mL) was added, and the resultant crystals were then filtered off to give 1.56 g of the titled compound (yield: 88%).

Preparation Example 3Preparation of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (3)

7-Methoxy-4-chloroquinoline-6-carboxamide (5.00 kg, 21.13 mol), dimethyl sulfoxide (55.05 kg), 1-(2-chloro-4-hydroxyphenyl)-3-cyclopropylurea 5.75 kg, 25.35 mol) and potassium t-butoxide (2.85 kg, 25.35 mol) were introduced in this order into a reaction vessel under a nitrogen atmosphere. The mixture was stirred for 30 min at 20° C., and the temperature was raised to 65° C. over 2.5 hours. The mixture was stirred at the same temperature for 19 hours. 33% (v/v) acetone-water (5.0 L) and water (10.0 L) were added dropwise over 3.5 hours. After the addition was completed, the mixture was stirred at 60° C. for 2 hours. 33% (v/v) acetone-water (20.0 L) and water (40.0 L) were added dropwise at 55° C. or more over 1 hour. After stirring at 40° C. for 16 hours, precipitated crystals were filtered off using a nitrogen pressure filter, and was washed with 33% (v/v) acetone-water (33.3 L), water (66.7 L), and acetone (50.0 L) in that order. The obtained crystals were dried at 60° C. for 22 hours using a conical vacuum dryer to give 7.78 kg of the titled compound (yield: 96.3%).

1H-NMR chemical, shift values for 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamides obtained in Preparation Examples 1 to 3 corresponded to those for 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide disclosed in WO 02/32872.

Example 5

A Crystalline Form of the Methanesulfonate of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (Form A)

(Preparation Method 1)

In a mixed solution of methanol (14 mL) and methanesulfonic acid (143 μL, 1.97 mmol) was dissolved 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (700 mg, 1.64 mmol) at 70° C. After confirming the dissolution of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide, the reaction mixture was cooled to room temperature over 5.5 hours, further stirred at room temperature for 18.5 hours, and crystals were filtered off. The resultant crystals were dried at 60° C. to give the titled crystals (647 mg).

(Preparation Method 2)

In a mixed solution of acetic acid (6 mL) and methanesulfonic acid (200 μL, 3.08 mmol) was dissolved 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (600 mg, 1.41 mmol) at 50° C. After confirming the dissolution of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide, ethanol (7.2 mL) and seed crystals of a crystalline form of the methanesulfonate of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (Form A) (12 mg) were added in this order to the reaction mixture, and ethanol (4.8 mL) was further added dropwise over 2 hours. After the addition was completed, the reaction mixture was stirred at 40° C. for 1 hour then at room temperature for 9 hours, and crystals were filtered off. The resultant crystals were dried at 60° C. to give the titled crystals (545 mg).

Example 6A Crystalline Form of the Methanesulfonate of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (Form B)

A crystalline form of the acetic acid solvate of the methanesulfonate of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-quinolinecarboxamide (Form I) (250 mg) obtained in Example 10 was dried under aeration at 30° C. for 3 hours and at 40° C. for 16 hours to give the titled crystals (240 mg)…………MORE IN PATENT

……………………………..

PATENT

https://www.google.com/patents/WO2014098176A1?cl=en

According to the present invention 4- (3-chloro-4- (cyclopropylamino-carbonyl) aminophenoxy) -7-methoxy-6-quinolinecarboxamide amorphous is excellent in solubility in water.

Example 1 4- (3-chloro-4- (cyclopropylamino-carbonyl) aminophenoxy) -7-methoxy-6-quinolinecarboxamide manufacture of amorphous amide
4- (3-chloro-4- (cyclopropylamino-carbonyl) amino phenoxy) -7-methoxy-6-quinolinecarboxamide B-type crystals (Patent Document 2) were weighed to 300mg, is placed in a beaker of 200mL volume, it was added tert- butyl alcohol (tBA) 40mL. This was heated to boiling on a hot plate, an appropriate amount of tBA to Compound A is dissolved, water was added 10mL. Then, the weakened heated to the extent that the solution does not boil, to obtain a sample solution. It should be noted, finally the solvent amount I was 60mL. 200mL capacity eggplant type flask (egg-plant shaped flask), and rotated in a state of being immersed in ethanol which had been cooled with dry ice. It was added dropwise a sample solution into the interior of the flask and frozen. After freezing the sample solution total volume, to cover the opening of the flask in wiping cloth, and freeze-dried. We got an amorphous A of 290mg.

Patent Document 2: US Patent Application Publication No. 2007/0117842 Patent specification

Amorphous A 13 C-solid state NMR spectrum in Figure 2, the chemical shifts and I are shown in Table 3.
[Table 3] *: peak of t- butyl alcohol

………………………..

Paper

ACS Medicinal Chemistry Letters (2015), 6(1), 89-94

http://pubs.acs.org/doi/full/10.1021/ml500394m

……………..

Paper

Journal of Pharmaceutical and Biomedical Analysis (2015), 114, 82-87

http://www.sciencedirect.com/science/article/pii/S0731708515002940

KEEP WATCHING WILL BE UPDATED………….most of my posts are updated regularly

References

  1. Matsui, J.; Funahashi, Y.; Uenaka, T.; Watanabe, T.; Tsuruoka, A.; Asada, M. (2008). “Multi-Kinase Inhibitor E7080 Suppresses Lymph Node and Lung Metastases of Human Mammary Breast Tumor MDA-MB-231 via Inhibition of Vascular Endothelial Growth Factor-Receptor (VEGF-R) 2 and VEGF-R3 Kinase”. Clinical Cancer Research 14 (17): 5459–65.doi:10.1158/1078-0432.CCR-07-5270. PMID 18765537.
  2. “Phae III trial shows lenvatinib meets primary endpoint of progression free surival benefit in treatment of radioiodine-refactory differentiated thyroid cancer”. Eisai. 3 February 2014.
  3. Glen, H; D. Boss; T. R. Evans; M. Roelvink; J. M. Saro; P. Bezodis; W. Copalu; A. Das; G. Crosswell; J. H. Schellens (2007). “A phase I dose finding study of E7080 in patients (pts) with advanced malignancies”. Journal of Clinical Oncology, ASCO Annual Meeting Proceedings Part I 25 (18S): 14073.
  4. ClinicalTrials.gov NCT00946153 Study of E7080 in Patients With Advanced Hepatocellular Carcinoma (HCC)
  5. Gild, M. L.; Bullock, M.; Robinson, B. G.; Clifton-Bligh, R. (2011). “Multikinase inhibitors: A new option for the treatment of thyroid cancer”. Nature Reviews Endocrinology 7 (10): 617–624.doi:10.1038/nrendo.2011.141. PMID 21862995. edit
  6. ClinicalTrials.gov NCT01321554 A Trial of E7080 in 131I-Refractory Differentiated Thyroid Cancer

UPDATES

EXTRAS……………

Martin Schlumberger et al. A phase 3, multicenter, double-blind, placebo-controlled trial of lenvatinib(E7080) in patients with 131I-refractory differentiated thyroid cancer (SELECT). 2014 ASCO Annual Meeting. Abstract Number:LBA6008. Presented June 2, 2014. Citation: J Clin Oncol 32:5s, 2014 (suppl; abstr LBA6008). Clinical trial information: NCT01321554.

Bando, Masashi. Quinoline derivative-​containing pharmaceutical composition. PCT Int. Appl. (2011), WO 2011021597 A1

Tomohiro Matsushima, four Nakamura, Kazuhiro Murakami, Atsushi Hoteido, Yusuke Ayat, Naoko Suzuki, Itaru Arimoto, Pinche Hirose, Masaharu Gotoda.Has excellent characteristics in terms of physical properties (particularly, dissolution rate) and pharmacokinetics (particularly, bioavailability), and is extremely useful as an angiogenesis inhibitor or c-Kit kinase inhibitor. US patent number US7612208  Also published as: CA2426461A1, CA2426461C, CN1308310C, CN1478078A, CN101024627A, DE60126997D1, DE60126997T2, DE60134679D1, DE60137273D1, EP1415987A1, EP1415987A4, EP1415987B1, EP1506962A2, EP1506962A3, EP1506962B1, EP1777218A1, EP1777218B1 , US7612092, US7973160, US8372981, US20040053908, US20060160832, US20060247259, US20100197911, US20110118470, WO2002032872A1, WO2002032872A8.Publication date: Aug 7, 2007 Original Assignee: Eisai Co., Ltd

Funahashi, Yasuhiro et al.Preparation of urea derivatives containing nitrogenous aromatic ring compounds as inhibitors of angiogenesis. US patent number US7253286, Also published as:CA2426461A1, CA2426461C, CN1308310C, CN1478078A, CN101024627A, DE60126997D1, DE60126997T2, DE60134679D1, DE60137273D1, EP1415987A1, EP1415987A4, EP1415987B1, EP1506962A2, EP1506962A3, EP1506962B1, EP1777218A1, EP1777218B1, US7612092, US7973160, US8372981, US20040053908, US20060160832, US20060247259, US20100197911, US20110118470, WO2002032872A1, WO2002032872A8.Publication date:Aug 7, 2007. Original Assignee:Eisai Co., Ltd

Sakaguchi, Takahisa; Tsuruoka, Akihiko. Preparation of amorphous salts of 4-​[3-​chloro-​4-​[(cyclopropylaminocarbonyl)​amino]​phenoxy]​-​7-​methoxy-​6-​quinolinecarboxamide as antitumor agents.  PCT Int. Appl. (2006), WO2006137474 A1 20061228.

Naito, Toshihiko and Yoshizawa, Kazuhiro. Preparation of urea moiety-containing quinolinecarboxamide derivatives. PCT Int. Appl., WO2005044788, 19 May 2005

Itaru Arimoto et al. Crystal of salt of 4-​[3-​chloro-​4-​(cyclopropylaminocarbonyl)​amino-​phenoxy]​-​7-​methoxy-​6-​quinolinecarboxamide or solvate thereof and processes for producing these. PCT Int. Appl. (2005), WO2005063713 A1 20050714.

10-23-2009
ANTITUMOR AGENT FOR UNDIFFERENTIATED GASTRIC CANCER
10-2-2009
ANTI-TUMOR AGENT FOR MULTIPLE MYELOMA
8-21-2009
ANTITUMOR AGENT FOR THYROID CANCER
8-14-2009
THERAPEUTIC AGENT FOR LIVER FIBROSIS
2-27-2009
USE OF COMBINATION OF ANTI-ANGIOGENIC SUBSTANCE AND c-kit KINASE INHIBITOR
9-5-2008
Medicinal Composition
8-8-2007
Nitrogen-containing aromatic derivatives
5-25-2007
Polymorph of 4-[3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy]-7-methoxy-6- quinolinecarboxamide and a process for the preparation of the same
7-21-2006
Nitrogen-containing aromatic derivatives
6-23-2006
Use of sulfonamide-including compounds in combination with angiogenesis inhibitors
11-16-2011
UREA DERIVATIVE AND PROCESS FOR PREPARING THE SAME
8-10-2011
c-Kit kinase inhibitor
7-6-2011
Nitrogen-Containing Aromatic Derivatives
12-24-2010
COMBINED USE OF ANGIOGENESIS INHIBITOR AND TAXANE
9-24-2010
COMBINATION OF ANTI-ANGIOGENIC SUBSTANCE AND ANTI-TUMOR PLATINUM COMPLEX
4-30-2010
METHOD FOR PREDICTION OF THE EFFICACY OF VASCULARIZATION INHIBITOR
4-16-2010
METHOD FOR ASSAY ON THE EFFECT OF VASCULARIZATION INHIBITOR
3-24-2010
Urea derivative and process for preparing the same
2-26-2010
COMPOSITION FOR TREATMENT OF PANCREATIC CANCER
2-26-2010
COMPOSITION FOR TREATMENT OF UNDIFFERENTIATED GASTRIC CANCER
US7253286 * 18 Apr 2003 7 Aug 2007 Eisai Co., Ltd Nitrogen-containing aromatic derivatives
US20040053908 18 Apr 2003 18 Mar 2004 Yasuhiro Funahashi Nitrogen-containing aromatic derivatives
US20040242506 9 Aug 2002 2 Dec 2004 Barges Causeret Nathalie Claude Marianne Formed from paroxetine hydrochloride and ammonium glycyrrhyzinate by precipitation, spray, vacuum or freeze drying, or evaporation to glass; solid or oil; masks the bitter taste of paroxetine and has a distinctive licorice flavor; antidepressants; Parkinson’s disease
US20040253205 10 Mar 2004 16 Dec 2004 Yuji Yamamoto c-Kit kinase inhibitor
US20070004773 * 22 Jun 2006 4 Jan 2007 Eisai R&D Management Co., Ltd. Amorphous salt of 4-(3-chiloro-4-(cycloproplylaminocarbonyl)aminophenoxy)-7-method-6-quinolinecarboxamide and process for preparing the same
US20070078159 22 Dec 2004 5 Apr 2007 Tomohiro Matsushima Has excellent characteristics in terms of physical properties (particularly, dissolution rate) and pharmacokinetics (particularly, bioavailability), and is extremely useful as an angiogenesis inhibitor or c-Kit kinase inhibitor
US20070117842 * 22 Apr 2004 24 May 2007 Itaru Arimoto Polymorph of 4-[3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy]-7-methoxy-6- quinolinecarboxamide and a process for the preparation of the same
EP0297580A1 30 Jun 1988 4 Jan 1989 E.R. SQUIBB & SONS, INC. Amorphous form of aztreonam
JP2001131071A Title not available
JP2005501074A Title not available
JPS6422874U Title not available
WO2002032872A1 19 Oct 2001 25 Apr 2002 Itaru Arimoto Nitrogenous aromatic ring compounds
WO2003013529A1 9 Aug 2002 20 Feb 2003 Barges Causeret Nathalie Claud Paroxetine glycyrrhizinate
WO2004039782A1 29 Oct 2003 13 May 2004 Hirai Naoko QUINOLINE DERIVATIVES AND QUINAZOLINE DERIVATIVES INHIBITING AUTOPHOSPHORYLATION OF Flt3 AND MEDICINAL COMPOSITIONS CONTAINING THE SAME
WO2004080462A1 10 Mar 2004 23 Sep 2004 Eisai Co Ltd c-Kit KINASE INHIBITOR
WO2004101526A1 22 Apr 2004 25 Nov 2004 Itaru Arimoto Polymorphous crystal of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7-methoxy-6-qunolinecarboxamide and method for preparation thereof
WO2005044788A1 8 Nov 2004 19 May 2005 Eisai Co Ltd Urea derivative and process for producing the same
WO2005063713A1 22 Dec 2004 14 Jul 2005 Itaru Arimoto Crystal of salt of 4-(3-chloro-4-(cyclopropylaminocarbonyl)amino-phenoxy)-7-methoxy-6-quinolinecarboxamide or of solvate thereof and processes for producing these
WO2006030826A1 14 Sep 2005 23 Mar 2006 Eisai Co Ltd Medicinal composition

UPDATE………….

1H NMR PREDICT OF LENVATINIB BASE

LEN BASE NMR GRAPH 1H LEN BASE NMR VALUES 1H

hplc real len real nmr

MASS NMR ABMOLE NMR SHIFT

AZD 9291, Osimertinib, Third-generation, oral, irreversible, selective epidermal growth factor receptor (EGFR) inhibitor for Non-small cell lung cancer (NSCLC)


Osimertinib.svg

str1

AZD 9291, Osimertinib

2-Propenamide, N-[2-[[2-(dimethylamino)ethyl]methylamino]-4-methoxy-5-[[4-(1-methyl-1H-indol-3-yl)-2-pyrimidinyl]amino]phenyl]-

 N-(2-((2-(dimethylamino)ethyl)(methyl)amino)-4-methoxy-5-((4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl)amino)phenyl)acrylamide. 
cas :1421373-65-0, 1421373-66-1(mesylate salt)

UPDATE…………FDA APPROVED NOV2015 

EU …… 3 FEB 2016 APPROVED

03 February 2016

AstraZeneca today announced that the European Commission (EC) has granted conditional marketing authorisation for TAGRISSO™ (AZD9291, osimertinib) 80mg once-daily tablets for the treatment of adult patients with locally advanced or metastatic epidermal growth factor receptor (EGFR) T790M mutation-positive non-small cell lung cancer (NSCLC).

Osimertinib is indicated for patients with T790M mutation-positive NSCLC, irrespective of previous treatment with a

ASTRAZENECA

Astrazeneca AbAstrazeneca Uk Limited


Mechanism of Action: Third-generation, oral, irreversible, selective  epidermal growth factor receptor (EGFR) inhibitor
Non-small cell lung cancer (NSCLC)

AZD-9291 M. Wt: 499.61 
AZD-9291 Formula: C28H33N7O2 

AZD9291, a third-generation orally irreversible epidermal growth factor receptor (EGFR) inhibitor, is under development by British drug maker AstraZeneca for the treatment of patients with metastatic EGFR T790M mutation-positive non-small cell lung cancer (NSCLC).
Lung cancer is the major cause of cancer death in the world while non small cell lung cancer (NSCLC) accounts approx. 85% of all lung cancer diagnosis. Approximately 50% of non–small cell lung cancer (NSCLC) patients who develop resistance to inhibitors of the epidermal growth factor receptor (EGFR) have acquired a second mutation, T790M. There are currently no approved treatments for patients who develop a T790 mutation.

Chemical structure for AZD9291 mesylate

AZD9291 mesylate

Also known as: AZD 9291 mesylate; AZD-9291 mesylate; HY-15772A; 1421373-66-1
AZD-9291 is a potent and selective mutated forms EGFR inhibitor(Exon 19 deletion EGFR IC50=12.92 nM, L858R/T790M EGFR IC50= 11.44 nM, wild type EGFR IC50= 493.8 nM).

Osimertinib (previously known as mereletinib[2] and AZD9291; trade name Tagrisso) is a third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) drug[3][4] developed by AstraZeneca Pharmaceuticals – for mutated EGFR cancers.

Approvals and indications

In November 2015, after a Priority Review, the US FDA granted accelerated approval to osimertinib for the treatment of metastaticepidermal growth factor receptor (EGFR) T790M mutation-positive non-small cell lung cancer (NSCLC), as detected by an FDA-approved test, which has progressed on or after EGFR tyrosine kinase inhibitor (TKI) therapy.[5][6]

The FDA approval made reference to two clinical trials, in which an EGFR T790M mutation was confirmed by a Cobas EGFR mutation test; osimertinib was given as 80 mg once daily.[1][7]

AZD-9291 is a third-generation EGFR inhibitor, showed promise in preclinical studies and provides hope for patients with advanced lung cancers that have become resistant to existing EGFR inhibitors. AZD9291 is highly active in preclinical models and is well tolerated in animal models. It inhibits both activating and resistant EGFR mutations while sparing the normal form of EGFR that is present in normal skin and gut cells, thereby reducing the side effects encountered with currently available medicines

Synthesis of AZD9291,

Synthesis-of-AZD9291

 

CLICK ON IMAGES FOR CLEAR VIEW

WO 2013014448 

http://www.google.com/patents/WO2013014448A1?cl=en

Example 27: V-f5-{[5-Cvano-4-flH-indol-3-yl)pyrimidin-2-yllamino}-4-methoxy-2-{4- methylpiperazin-l-yl}phenyl)prop-2-enamide

Acryloyl chloride (0.100 mL, 1M in THF, 0.1 mmol) was added dropwise to a fine slurry of 2- {[5-amino-2-methoxy-4-(4-methylpiperazin- 1 -yl)phenyl] amino} -4-(lH-indol-3- yl)pyrimidine-5-carbonitrile (Intermediate 99, 47 mg, 0.10 mmol) and DIPEA (0.027 mL, 0.16 mmol) in THF (2 mL) at -10°C over a period of 2 minutes under N2.The mixture was then stirred at 0°C for 10 minutes then allowed to warm to r.t. over 20 minutes. The mixture was then cooled again to -10°C and further acryloyl chloride (0.06 mL, 1M in THF, 0.06 mmol) was added dropwise. The mixture was stirred at 0°C for a further 10 minutes, then allowed to warm to r.t. over 20 minutes. The mixture was then concentrated in vacuo and the resulting reside was dissolved in CH2C12 (2 mL). This solution was washed with sat. NaHC03 (1 mL), dried (MgS04) and concentrated in vacuo. Purification by FCC, eluting with 1.5-7% 7N methanolic ammonia in CH2C12 gave a residue that was washed with CH3OH (0.1 mL) and dried in air to give the title compound (1 lmg, 20%) as a cream crystalline solid; 1H NMR: 2.28 (3H, s), 2.54-2.65 (4H, m), 2.93 (4H, s), 3.75 (3H, s), 5.71 (1H, d), 6.18 (1H, d), 6.64 (1H, dd), 6.91 (2H, m), 7.18 (1H, s), 7.47 (1H, d), 8.02 (1H, s), 8.52 (1H, s), 8.67 (1H, s), 9.04 (1H, s), 9.40 (1H, s), 11.99 (1H, s); m/z: ES+ MH+ 509. Example 28: V-f2-{2-Dimethylaminoethyl-methylamino}-4-methoxy-5-{[4-fl- methylindol-3-yl)pyrimidin-2-yllamino}phenyl)prop-2-enamide

Figure imgf000067_0001

A solution of acryloyl chloride (34.5 mg, 0.38 mmol) in CH2C12 (1 mL) was added dropwise to a stirred mixture of N;-(2-dimethylaminoethyl)-5-methoxy-N;-methyl-N¥-[4- (l-methylindol-3-yl)pyrimidin-2-yl]benzene-l,2,4-triamine (Intermediate 100, 170 mg, 0.38 mmol) and DIPEA (0.073 mL, 0.42 mmol) in CH2C12 (5 mL), which was cooled in an ice/water bath. The mixture was stirred for 1.5h and then diluted with CH2C12 (25 mL) and washed with sat.NaHCOs (50 mL). The aqueous washes were extracted with CH2C12 (2 x 25 mL). The combined organic solutions were dried (MgSC^) and concentrated in vacuo. Purification by FCC, eluting with 0-4% 7N methanolic ammonia in CH2C12 gave the title compound (75 mg, 39%) as a cream solid after trituration with diethyl ether; 1H NMR: 2.21 (6H, s), 2.29 (2H, t), 2.72 (3H, s), 2.89 (2H, t), 3.86 (3H, s), 3.92 (3H, s), 5.77 (1H, dd), 6.27 (1H, dd), 6.43 (1H, dd), 7.04 (1H, s), 7.15 (1H, t), 7.20-7.27 (2H, m), 7.53 (1H, d), 7.91 (1H, s), 8.24 (1H, d), 8.33 (1H, d), 8.68 (1H, s), 9.14 (1H, s), 10.22 (1H, s); m/z: ES+ MH+ 500.42.

Example 28 (Alternative synthesis 1): V-f2-{2-Dimethylaminoethyl-methylamino}-4- methoxy-5-{[4-(l-methylindol-3-yl)pyrimidin-2-yllamino}phenyl)prop-2-enamide

Figure imgf000067_0002

To a stirred solution of 3-chloro-N-[2-[2-dimethylaminoethyl(methyl)amino]-4-methoxy- 5-[[4-(l-methylindol-3-yl)pyrimidin-2-yl]amino]phenyl]propanamide (Intermediate 174, 31.5 g, 58.76 mmol) in acetonitrile (310 mL) was added triethylamine (17.84 g, 176.28 mmol) at r.t. The resulting mixture was heated to 80°C for 6h then cooled to r.t.. Water (130 mL) was then added and the mixture stirred for 12h. The mixture was then filtered, washed with a mixture of water and acetonitrile (160 mL, 1 : 1) and dried at 50°C for overnight to give the title compound (19.2 g, 94%) as a solid form identified herein as polymorphic form D. 1H NMR: 2.69 (3H, s), 2.83 (6H, d), 3.35 (4H, s), 3.84 (3H, s), 3.91 (3H, s), 5.75 (IH, d), 6.28 (IH, d), 6.67 (IH, dd), 7.05-7.23 (2H, m), 7.29 (IH, t), 7.43 (IH, d), 7.56 (IH, d), 8.21 (2H, s), 8.81 (IH, s), 9.47 (IH, s), 9.52 (IH, s), m/z: ES+ MH+ 500.26.

Example 28 (Alternative synthesis 2): V-f2-{2-Dimethylaminoethyl-methylamino}-4- methoxy-5-{[4-(l-methylindol-3-yl)pyrimidin-2-yllamino}phenyl)prop-2-enamide

Figure imgf000068_0001

To a stirred solution of N1-(2-dimethylaminoethyl)-5-methoxy-N1-methyl-N4-[4-(l- methylindol-3-yl)pyrimidin-2-yl]benzene-l,2,4-triamine (Intermediate 100, 10 g, 21.32 mmol) in THF (95 mL) and water (9.5 mL) at 0°C was added the 3-chloropropanoyl chloride (3.28 g, 25.59 mmol). The mixture was stirred at r.t. for 15 minutes then NaOH (3.48 g, 85.28 mmol) was added. The resulting mixture was heated to 65°C for lOh. The mixture was then cooled to r.t. and CH3OH (40 mL) and water (70 mL) were added. The resulting mixture was stirred overnight. The resulting solid was collected by filtration, washed with water (25 mL) and dried at 50°C for 12h to give the title compound (7.0 g, 94%) as a solid form identified herein as polymorphic Form D.1H NMR: 2.69 (3H, s) 2.83 (6H, d) 3.35 (4H, s) 3.84 (3H, s) 3.91 (3H, s) 5.75 (IH, d) 6.28 (IH, d) 6.67 (IH, dd) 7.05-7.23 (2H, m) 7.29 (IH, t) 7.43 (IH, d) 7.56 (IH, d) 8.21 (2H, s) 8.81 (IH, s) 9.47 (IH, s) 9.52 (IH, s) ES+ MH+ 500.26.

Example 28A: V-f2-{2-Dimethylaminoethyl-methylamino}-4-methoxy-5-{[4-fl- methylindol-3-yl)pyrimidin-2-yll amino}phenyl)prop-2-enamide mesylate salt

Figure imgf000069_0001
AZD 9291

Procedure 1: To a stirred solution of N-[2-[2-dimethylaminoethyl(methyl)amino]-4- methoxy-5 – [[4-( 1 -methylindol-3 -yl)pyrimidin-2-yl] amino]phenyl]prop-2-enamide

(Example 28, 20 g, 36.63 mmol) in ethanol (120 mL) and EtOAc (80 mL) at 70°C was added methane sulfonic acid (3.59 g, 36.63 mmol) as a solution in EtOAc (40 mL). The resulting mixture was stirred for 1.5h. The resulting solid was collected by filtration and dried at 80°C under vacuum overnight to give the title salt (20.5 g, 94%) in a solid form defined herein as polymorphic Form B for this salt.

Procedure 2: To a stirred solution of N-[2-[2-dimethylaminoethyl(methyl)amino]-4- methoxy-5 – [[4-( 1 -methylindol-3 -yl)pyrimidin-2-yl] amino]phenyl]prop-2-enamide

(Example 28, 5 g, 9.11 mmol) in acetone (45.5 mL) and water (4.55 mL) at 50°C was added methane sulfonic acid (0.893 g, 9.11 mmol) as a solution in acetone (4.55 mL). The resulting mixture was stirred for 1.5h. The resulting solid was collected by filtration and dried at 80°C under vacuum overnight to give the title salt (4.9 g, 94%) in a solid form defined herein as polymorphic Form B for this salt; ΧΗ NMR (acetone-ii6): 2.72 (3H, s), 2.96 (3H, s), 3.01 (6H, s), 3.58 (3H, t), 3.87-3.90 (7H, m), 5.76 (1H, dd), 6.38-6.53 (2H, m), 7.12 (1H, t), 7.20 (1H, t), 7.29 (1H, s), 7.40 (2H, t), 8.07-8.16 (3H, m), 8.56 (1H, s), 9.30 (1H, s), 9.60 (1H, s), 9.66 (1H, s ); m/z: ES+ MH+ 500.26.

Procedure 3: Polymorphic Form A of N-(2-{2-dimethylaminoethyl-methylamino}-4- methoxy-5 – { [4-( 1 -methylindol-3 -yl)pyrimidin-2-yl] amino } phenyl)prop-2-enamide mesylate salt was prepared in a similar manner as described above on a ~50 mg scale, except that acetonitrile was used as the solvent. Specifically, ~9.6mg methanesulfonic acid was dissolved into a minimum volumn of acetonitrile. ~50 mg N-(2- {2-dimethylamino- ethyl-methylamino } -4-methoxy-5 – { [4-( 1 -methylindol-3 -yl)pyrimidin-2-yl] amino } phenyl)- prop-2-enamide was also dissolved into a minimum volume of acetonitrile and then the resulting solution was added to the methanesulfonic acid solution. Formation of a solid resulted upon addition. This solid was collected by filtration and was air-dried and then analysed. The particular solid form produced in this experiment was designated as

Polymorphic Form A for this salt.

Osimertinib
Osimertinib.svg
Systematic (IUPAC) name
N-(2-{2-dimethylaminoethyl-methylamino}-4-methoxy-5-{[4-(1-methylindol-3-yl)pyrimidin-2-yl]amino}phenyl)prop-2-enamide
Clinical data
Trade names Tagrisso
Routes of
administration
Oral tablets
Pharmacokinetic data
Protein binding probably high[1]
Metabolism oxidation (CYP3A)
Biological half-life 48 hours
Excretion feces (68%), urine (14%)
Identifiers
CAS Number 1421373-65-0
PubChem CID 71496458
ChemSpider 31042598
UNII 3C06JJ0Z2O
ChEBI CHEBI:90943 Yes
Chemical data
Formula C28H33N7O2
Molar mass 499.6 g/mol

References

  1.  “Tagrisso (osimertinib) Tablet, for Oral Use. Full Prescribing Information” (PDF). AstraZeneca Pharmaceuticals LP, Wilmington, DE 19850. Retrieved 16 November 2015.
  2.  “Proposed INN: List 113” (PDF). International Nonproprietary Names for Pharmaceutical Substances (INN) 29 (2): 285. 2015. Retrieved16 November 2015.
  3.  Ayeni D, Politi K, Goldberg SB (2015). “Emerging Agents and New Mutations in EGFR-Mutant Lung Cancer”. Clin. Cancer Res. 21 (17): 3818–20. doi:10.1158/1078-0432.CCR-15-1211. PMID 26169963.
  4.  Tan CS, Gilligan D, Pacey S (2015). “Treatment approaches for EGFR-inhibitor-resistant patients with non-small-cell lung cancer”. Lancet Oncol. 16 (9): e447–59. doi:10.1016/S1470-2045(15)00246-6. PMID 26370354.
  5.  U.S. Food and Drug Administration. Hematology/Oncology (Cancer) Approvals & Safety Notifications. [1] Xu M, Xie Y, Ni S, Liu H (2015). “The latest therapeutic strategies after resistance to first generation epidermal growth factor receptor tyrosine kinase inhibitors (EGFR TKIs) in patients with non-small cell lung cancer (NSCLC)”. Ann Transl Med 3 (7): 96.doi:10.3978/j.issn.2305-5839.2015.03.60. PMC 4430733. PMID 26015938.
  6. U.S. Food and Drug Administration. “Osimertinib”. [2]

 

AstraZeneca R&D Facility, Alderley Park, Cheshire, United Kingdom

Cheshire Map Showing Location of Alderley Park

District: Macclesfield
Easting: 384439 Northing: 374467
Latitude: 53.27 Longitude: -2.23
= Alderley Park

 

INN or syn URL CID InChIKey
AZ11657312
AZ876 AZ12260493 CID 11655079 IVANYIPLGFVBGR-UHFFFAOYSA-N
AZ12419304
AZ12609721
AZ12823138
AZ12971554 CID 44537923 GMKHQRCPNMGCIX-ZWKOTPCHSA-N
AZ13483342
AZD0328 CID 9794392 OCKIPDMKGPYYJS-ZDUSSCGKSA-N
Saracatinib AZD0530 CID 10302451 OUKYUETWWIPKQR-UHFFFAOYSA-N
Barasertib AZD1152 CID 11497983 GBJVVSCPOBPEIT-UHFFFAOYSA-N
AZD1208 CID 58423153 MCUJKPPARUPFJM-UWCCDQBKSA-N
AZD1236
AZD1332 CID 49831044 LBVKEEFIPBQIMD-UHFFFAOYSA-N
AZD1480 CID 16659841 PDOQBOJDRPLBQU-QMMMGPOBSA-N
AZD1656 CID 16039797 FJEJHJINOKKDCW-INIZCTEOSA-N
MK175 AZD1775 CID 24856436 BKWJAKQVGHWELA-UHFFFAOYSA-N
AZD1981 CID 11292191 JWYIGNODXSRKGP-UHFFFAOYSA-N
AZD2014 CID 25262792 JUSFANSTBFGBAF-IRXDYDNUSA-N
Olaparib AZD2281 CID 23725625 FDLYAMZZIXQODN-UHFFFAOYSA-N
AZD2624 CID 23649245 QYTBBBAHNIWFOD-NRFANRHFSA-N
AZD2927 CID 57345449 GAHPWXLXWUVMIV-MRXNPFEDSA-N
Lesogaberan AZD3355 CID 9833984 WVTGPBOMAQLPCP-GSVOUGTGSA-O
AZD3463 CID 56599293 GCYIGMXOIWJGBU-UHFFFAOYSA-N
AZD3857
AZD4017 CID 24946280 NCDZABJPWMBMIQ-INIZCTEOSA-N
AZD4320
AZD4547 CID 51039095 NCDZABJPWMBMIQ-INIZCTEOSA-N
AZD4877 CID 10368812 SMFXSYMLJDHGIE-UHFFFAOYSA-N
AZD5363 CID 25227436 JDUBGYFRJFOXQC-KRWDZBQOSA-N
AZD5582 CID 49847690 WLMCRYCCYXHPQF-ZVMUOSSASA-N
TX4 AZD5904 CID 10264211 RSPDBEVKURKEII-ZCFIWIBFSA-N
Selumetinib AZD6244 CID 10127622 IAYGCINLNONXHY-LBPRGKRZSA-N
AZD6495
AZD6738
Lanicemine AZD6765 CID 3038485 FWUQWDCOOWEXRY-UHFFFAOYSA-N
AZD7325 CID 23581869 KYDURMHFWXCKMW-UHFFFAOYSA-N
AZD7762 CID 11152667 IAYGCINLNONXHY-LBPRGKRZSA-N
AZD8055 CID 25262965 KVLFRAWTRWDEDF-IRXDYDNUSA-N
AZD8186 CID 52913929 LMJFJIDLEAWOQJ-UHFFFAOYSA-N
AZD8329 CID 25006684 XWBXJBSVYVJAMZ-UHFFFAOYSA-N
AZD8529 CID 25125217 IPCYZQQFECEHLI-UHFFFAOYSA-N
AZD8542 CID 53344810 SMQVBAGSZVHCJP-UHFFFAOYSA-N
AZD8931 CID 11488320 DFJSJLGUIXFDJP-UHFFFAOYSA-N
 STAT3Rx AZD9150
AZD9291 CID 71496458 DUYJMQONPNNFPI-UHFFFAOYSA-N
Alvelestat AZD9668 CID 46861623 QNQZWEGMKJBHEM-UHFFFAOYSA-N
Zibotentan ZD4054 CID 9910224 FJHHZXWJVIEFGJ-UHFFFAOYSA-N
Vandetanib ZD6474 CID 3081361 UHTHHESEBZOYNR-UHFFFAOYSA-N

INNs or synonyms are in the first column, the 2nd column is the AZ OI links, 3rd column the PubChem CID, followed by the InChIKey as the 4th column.

UPDATE

13 November 2015

One of fastest development programmes – from start of clinical trials to approval in just over two and a half years to meet unmet patient need

With objective response rate of 59% and duration of response of 12.4 months, TAGRISSO provides important new option for patients

AstraZeneca today announced that the US Food and Drug Administration (FDA) has approved TAGRISSO™ (AZD9291) 80mg once-daily tablets for the treatment of patients with metastatic epidermal growth factor receptor (EGFR) T790M mutation-positive non-small cell lung cancer (NSCLC), as detected by an FDA-approved test, who have progressed on or after EGFR tyrosine kinase inhibitor (TKI) therapy.

AZD9291 is the only approved medicine indicated for patients with metastatic EGFR T790M mutation-positive non-small cell lung cancer. This indication is approved under the FDA’s accelerated approval process based on tumour response rate and duration of response (DoR).

AZD9291 is an EGFR-TKI, a targeted cancer therapy, designed to inhibit both the activating, sensitising mutations (EGFRm), and T790M, a genetic mutation responsible for EGFR-TKI treatment resistance. Nearly two-thirds of NSCLC patients who are EGFR mutation-positive and experience disease progression after being treated with an EGFR-TKI develop the T790M resistance mutation, for which there have been limited treatment options.

Pasi A Jänne MD, PhD, Director, Lowe Center for Thoracic Oncology at Dana-Farber Cancer Institute, Scientific Director, Belfer Center for Applied Cancer Science and Professor of Medicine, Harvard Medical School, said: “In the AURA clinical studies, AZD9291 has demonstrated compelling early efficacy and tolerability in patients with EGFRm T790M metastatic non-small cell lung cancer. This treatment has the potential to become the standard of care for patients living with EGFRm T790M non-small cell lung cancer. The accelerated approval of AZD9291 highlights its clinical promise for a targeted group of patients and gives healthcare providers an important new option.”

Pascal Soriot, Chief Executive Officer, AstraZeneca, said: “The FDA approval of TAGRISSO marks an important milestone for lung cancer patients who urgently need new treatment options. We have built on our heritage in this area and acted on the breakthrough clinical evidence to ensure this next-generation medicine reaches patients in record time. As we advance our comprehensive lung cancer portfolio, we have the opportunity to treat greater numbers of patients across all stages of this disease through precision medicines, immunotherapies and novel combinations.”

AstraZeneca has collaborated with Roche to develop the cobas® EGFR Mutation Test v2 as the companion diagnostic for AZD9291. The cobas® EGFR Mutation Test v2 is intended to identify a range of EGFR mutations in patients with non-small cell lung cancer, including T790M.

AZD9291 was granted Fast Track, Breakthrough Therapy, Priority Review and Accelerated Approval status by the FDA. In Europe and Japan, AZD9291 was granted Accelerated Assessment and Priority Review status respectively. Interactions with regulatory authorities in the rest of the world are ongoing.

The FDA approval of AZD9291 is based on data from the two AURA Phase II studies (AURA extension and AURA2) which demonstrated efficacy in 411 EGFRm T790M NSCLC patients that had progressed on or after an EGFR TKI. In those trials, overall objective response rate ((ORR) a measurement of tumor shrinkage) was 59% (95% CI: 54% to 64%). In a supportive Phase I study in 63 patients, ORR was 51% and median duration of response was 12.4 months.

The AZD9291 tolerability profile showed that no individual severe grade 3+ adverse events occurred at ≥ 3.5%.The most common adverse events were generally mild to moderate and included diarrhoea (42% all grades; 1.0% Grade 3/4),  rash (41% all grades; 0.5% Grade 3/4), dry skin (31% all grades; 0% Grade 3/4), and nail toxicity (25% all grades; 0% Grade 3/4). There are no contraindications for AZD9291. Warnings and precautions include interstitial lung disease, QT interval prolongation, cardiomyopathy and embryofoetal toxicity.

AZD9291 Development Programme

AZD9291 is being studied in the confirmatory trial, AURA3, an open label, randomised Phase III study designed to assess the efficacy and safety of AZD9291 versus platinum-based doublet chemotherapy in patients with EGFR T790M positive, locally advanced, or metastatic NSCLC who have progressed following prior therapy with an EGFR-TKI. AZD9291 is also being investigated in the adjuvant setting and in the metastatic first-line setting, including in patients with brain metastases, as well as in combination with other compounds.

NOTES TO EDITORS

About Non-Small Cell Lung Cancer

Lung cancer is the leading cause of cancer death among both men and women, accounting for about one-third of all cancer deaths, more than breast, prostate and colorectal cancers combined. Lung cancer has a five-year survival rate that is less than 20%. Approximately 85% of all lung cancers in the US are NSCLC; 10% to 15% of these are EGFR mutation-positive. Approximately two-thirds of patients treated with EGFR TKI therapy will acquire resistance related to the T790M mutation.

About AZD9291

AZD9291 80mg once-daily tablet is the first medicine indicated for the treatment of patients with metastatic epidermal growth factor receptor (EGFR) T790M mutation-positive non-small-cell lung cancer (NSCLC), as detected by an FDA-approved test, who have progressed on or after EGFR tyrosine kinase inhibitor (TKI) therapy. Non-clinical in vitro studies have demonstrated that AZD9291 has high potency and inhibitory activity against mutant EGFR phosphorylation across the range of clinically relevant EGFRm and T790M mutant NSCLC cell lines with significantly less activity against EGFR in wild-type cell lines.

Osimertinib has recently been published by the World Health Organisation (WHO) as the proposed International Non-proprietary Name (INN) for AZD9291, and may become formally adopted during November 2015. In the US, the American Medical Association accepted osimertinib as the United States Adopted Name (USAN).

About AstraZeneca in Oncology

Oncology is a therapeutic area in which AstraZeneca has deep-rooted heritage. It will be potentially transformational for the company’s future, becoming the sixth growth platform. Our vision is to help patients by redefining the cancer treatment paradigm and one day eliminate cancer as cause of death. By 2020, we are aiming to bring six new cancer medicines to patients.

Our broad pipeline of next-generation medicines is focused on four main disease areas – lung, ovarian, breast, and hematological cancers. These are being targeted through four key platforms – immuno-oncology, the genetic drivers of cancer and resistance, DNA damage repair and antibody drug conjugates.

About Roche

Headquartered in Basel, Switzerland, Roche is a leader in research-focused healthcare with combined strengths in pharmaceuticals and diagnostics. Roche is the world’s largest biotech company, with truly differentiated medicines in oncology, immunology, infectious diseases, ophthalmology and neuroscience. Roche is also the world leader in in vitro diagnostics and tissue-based cancer diagnostics, and a frontrunner in diabetes management. Roche’s personalised healthcare strategy aims at providing medicines and diagnostics that enable tangible improvements in the health, quality of life and survival of patients. Founded in 1896, Roche has been making important contributions to global health for more than a century. Twenty-nine medicines developed by Roche are included in the World Health Organization Model Lists of Essential Medicines, among them life-saving antibiotics, antimalarials and chemotherapy.

About AstraZeneca

AstraZeneca is a global, innovation-driven biopharmaceutical business that focuses on the discovery, development and commercialisation of prescription medicines, primarily for the treatment of cardiovascular, metabolic, respiratory, inflammation, autoimmune, oncology, infection and neuroscience diseases. AstraZeneca operates in over 100 countries and its innovative medicines are used by millions of patients worldwide. For more information please visit www.astrazeneca.com.

SEE………http://apisynthesisint.blogspot.in/2016/02/azd-9291-osimertinib-third-generation_4.html

/////

CN1C=C(C2=CC=CC=C21)C3=NC(=NC=C3)NC4=C(C=C(C(=C4)NC(=O)C=C)N(C)CCN(C)C)OC

Eluxadoline …Diarrhea-predominant irritable bowel syndrome


Eluxadoline

5 JAN 2014

Furiex Pharmaceuticals Inc.  more than doubled in its best day of trading after its experimental drug alleviated diarrhea and abdominal pain caused by irritable bowel syndrome in two studies.

The drug eluxadoline met targets for improvements in stool consistency and abdominal pain that were developed in conjunction with U.S. and European regulators, the company said today. Furiex will apply for approval in June, Chairman Fred Eshelman said in an investor call today. He estimated annual sales of $750 million to $1 billion.

“By our math, it looks like a pretty doggone good market,” Eshelman said on the call, noting that there is only one currently approved drug available in the U.S. for the condition.

Diarrhea-predominant irritable bowel syndrome is a chronic disorder that affects about 28 million patients in the U.S. and Europe, Furiex said in the statement.Furiex said it would apply by mid-year for U.S. approval of the drug, eluxadoline, to treat diarrhea-predominant irritable bowel syndrome (IBS-d), a debilitating bowel disorder that affects about 28 million people in the United States and major European markets.

Furiex said it expected to seek European approval in early 2015.

“We believe that there are a lot of patients out there who need this drug. There is a huge unmet need,” Furiex Chief Medical Officer June Almenoff said in a telephone interview.

Currently approved drugs for IBS address constipation associated with the disorder, but there are few options for diarrhea predominant IBS.

Furiex founder and chairman Fred Eshelman said he believes the drug has the potential for blockbuster sales, which he defined as annual sales of between $750 million and $1 billion.

Eluxadoline was tested at two doses against a placebo over the course of 12 weeks to meet requirements by the U.S. Food and Drug Administration, and for 26 weeks for European health regulators, in Phase III studies involving 2,428 patients, Furiex said.

For the combined goal of improvement in abdominal pain and stool consistency for at least half the days in the study, eluxadoline achieved a statistically significant improvement at the 100 milligram and 75 mg doses through 12 weeks in both studies.

On the 26-week measure, the higher dose succeeded in both studies but the lower dose missed statistical significance in one of the two trials, according to initial results released by the company.

The success appeared to be driven by the percentage of patients reporting improvements in diarrhea, which ranged from 30 percent to 37 percent versus 22 percent and 20.9 percent for the placebo groups.

When the composite goal was broken into its two components, researchers found a numerical improvement in pain response rates that did not achieve statistical significance.

The drug appeared to be safe and well-tolerated in both studies, Furiex said. The most commonly reported side effects were constipation and nausea.

The company plans to present a far more detailed analysis of the late stage studies at an upcoming medical meeting.

“We’re very excited about the path ahead and about how this can transform patients’ lives,” Almenoff said.

Eluxadoline 

5-({[(2S)-2-amino-3-(4-carbamoyl-2,6-dimethylphenyl)propanoyl][(1S)-1-(4-phenyl-1H-imidazol-2-yl)ethyl]amino}methyl)-2-methoxybenzoic acid

5-({[2-Amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid

864821-90-9 CAS

JNJ-27018966

Molecular Formula: C32H35N5O5

Molecular Weight: 569.6508

Agents for Irritable Bowel Syndrome, mu-Opioid Agonists, delta-Opioid Antagonists

Mu Delta is a locally active mu opioid receptor agonist and delta opioid receptor antagonist in phase III clinical evaluation at Furiex Pharmaceuticals for the oral treatment of diarrheal predominant irritable bowel syndrome (d-IBS).

The product candidate holds an advantage over currently marketed products for this indication because it acts locally on the enteric nervous system, possibly decreasing adverse effects on the central nervous system. In 2011, fast track designation was assigned in the U.S. for the treatment of d-IBS. In 2011, Mu Delta was licensed to Furiex Pharmaceuticals by Janssen for the treatment of d-IBS, granting an option to Furiex to continue development and commercialization following phase II proof of concept studies.

The opioid receptors were identified in the mid-1970’s, and were quickly categorized into three sub-sets of receptors (mu, delta and kappa). More recently the original three types of receptors have been further divided into sub-types. Also known is that the family of opioid receptors are members of the G-protein coupled receptor (GPCR) super-family. More physiologically pertinent are the well established facts that opioid receptors are found throughout the central and peripheral nervous system of many mammalian species, including humans, and that modulation of the respective receptors can elicit numerous, albeit different, biological effects, both desirable and undesirable (D. S. Fries, “Analgesics”, inPrinciples of Medicinal Chemistry, 4th ed.; W. O. Foye, T. L. Lemke, and D. A. Williams, Eds.; Williams and Wilkins: Baltimore, Md., 1995; pp. 247-269; J. V. Aldrich, “Analgesics”, Burger’s Medicinal Chemistry and Drug Discovery, 5thEdition, Volume 3: Therapeutic Agents, John Wiley & Sons, Inc., 1996, pp. 321-441). In the most current literature, the likelihood of heterodimerization of the sub-classes of opioid receptors has been reported, with respective physiological responses yet undetermined (Pierre J. M. Riviere and Jean-Louis Junien, “Opioid receptors: Targets for new gastrointestinal drug development”, Drug Development 2000, pp. 203-238).

A couple biological effects identified for opioid modulators have led to many useful medicinal agents. Most significant are the many centrally acting mu opioid agonist modulators marketed as analgesic agents to attenuate pain (e.g., morphine), as well as peripherally acting mu agonists to regulate motility (e.g., loperamide). Currently, clinical studies are continuing to evaluate medicinal utility of selective delta, mu, and kappa modulators, as well as compounds possessing combined sub-type modulation. It is envisioned such explorations may lead to agents with new utilities, or agents with minimized adverse side effects relative to currently available agents (examples of side effects for morphine includes constipation, respiratory depression, and addiction potential). Some new GI areas where selective or mixed opioid modulators are currently being evaluated includes potential treatment for various diarrheic syndromes, motility disorders (post-operative ileus, constipation), and visceral pain (post operative pain, irritable bowel syndrome, and inflammatory bowel disorders) (Pierre J. M. Riviere and Jean-Louis Junien, “Opioid receptors: Targets for new gastrointestinal drug development” Drug Development, 2000, pp. 203-238).

Around the same time the opioid receptors were identified, the enkephalins were identified as a set of endogenous opioid ligands (D. S. Fries, “Analgesics”, inPrinciples of Medicinal Chemistry, 4th ed.; W. O. Foye; T. L. Lemke, and D. A. Williams, Eds.; Williams and Wilkins: Baltimore, Md., 1995; pp. 247-269). Schiller discovered that truncating the original pentapeptide enkephalins to simplified dipeptides yielded a series of compounds that maintained opioid activity (Schiller, P. WO 96/06855). However one potential drawback cited for such compounds is the likelihood of their inherent instability (P. W. Schiller et al., Int. J. Pept. Protein Res. 1993, 41 (3), pp. 313-316).

More recently, a series of opioid pseudopeptides containing heteroaromatic or heteroaliphatic nuclei were disclosed, however this series is reported showing a different functional profile than that described in the Schiller works. (L. H. Lazarus et al., Peptides 2000, 21, pp. 1663-1671).

Most recently, works around morphine related structures were reported by Wentland, et al, where carboxamido morphine derivatives and it’s analogs were prepared (M. P. Wentland et al., Biorg. Med. Chem. Letters 2001, 11, pp. 1717-1721; M. P. Wentland et al., Biorg. Med. Chem. Letters 2001, 11, pp. 623-626). Wentland found that substitution for the phenol moiety of the morphine related structures with a primary carboxamide led anywhere from equal activities up to 40 fold reduced activities, depending on the opioid receptor and the carboxamide. It was also revealed that any additional N-substitutions on the carboxamide significantly diminished the desired binding activity.

Compounds of the present invention have not been previously disclosed and are believed to provide advantages over related compounds by providing improved pharmacological profiles.

Opioid receptor modulators, agonists or antagonists are useful in the treatment and prevention of various mammalian disease states, for example pain and gastrointestinal disorders such as diarrheic syndromes, motility disorders including post-operative ileus and constipation, and visceral pain including post-operative pain, irritable bowel syndrome and inflammatory bowel disorders.

It is an object of the present invention to provide opioid receptor modulators. It is a further object of the invention to provide opioid receptor agonists and opioid receptor antagonists. It is an object of the present invention to provide opioid receptor ligands that are selective for each type of opioid receptor, mu, delta and kappa. It is a further object of the present invention to provide opioid receptor ligands that modulate two or three opioid receptor types, mu, delta and kappa, simultaneously.

It is an object of the invention to provide certain instant compounds that are also useful as intermediates in preparing new opioid receptor modulators. It is also an object of the invention to provide a method of treating or ameliorating a condition mediated by an opioid receptor. And, it is an object of the invention to provide a useful pharmaceutical composition comprising a compound of the present invention useful as an opioid receptor modulator.

5-({[2-Amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1 h-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid is an opoid receptor modulator (mu receptor agonist and delta receptor antagonist) and may be useful for treating irritable bowel syndrome, pain or other opioid receptor disorders.

5-({[2-Amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1h-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid and methods of making this molecule are disclosed in

US application 2005/02033143. Example 9 of US application 2005/02033143 makes the hydrochloride salt of 5-({[2-amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1h-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid.

Applicants have discovered a process of making the zwitterion of 5-({[2-amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1h-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid and two novel crystals of this zwitterion. In Applicant’s hands, these novel crystals provide improved properties and can be purified at higher purity. Applicant’s new process results in improved and less costly process manufacturing conditions than the procedure disclosed in US application 2005/02033143.

………………..

FIG. 6 is the molecular structure of the zwitterion 5-({[2-amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1h-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid.

US7994206

…………………..

SYNTHESIS OF 5-formyl-2- methoxy-benzoic acid methyl ester

WO2002022612A1

Example 8: 2-Methoxy-5-formylbenzoic acid

Figure imgf000023_0001

Lithium hydroxide (1.04g, 0.043mol, 3eq) in water (lOmL) was added to a stirred solution of methyl 2-methoxy-5-formylbenzoate (2.8g, 0.014mol, leq) in a mixture of tetrahydrofuran (30mL) and methanol (20mL). The solution was stirred overnight, acidified to pH 1 with 10% HCl and the organic solvents removed in vacuo. The aqueous solution was extracted with ethyl acetate (lOOmL) and the organic solution washed with brine (lOOmL), then extracted with saturated aqueous sodium bicarbonate (3 x lOOmL). The basic solution was washed with ethyl acetate (lOOmL), then acidified to pH 1 with 10% HCl and back extracted with dichloromethane (3 x lOOmL). The organic solution was dried over sodium sulfate and evaporated in vacuo to give a cream coloured powder (2.01g, 77%). 1H NMR (CDC13) δ 9.99 (s, IH, O=C- H), 4.14 (s, 3H, CH3).

………………

ANALOGOUS METHOD TO PREPARE..2-methoxy-5-{[1 -(4-phenyl-1 H-imidazol-2-yl)- ethylamino]-methyl}-benzoic acid methyl ester

USE 5-formyl-2- methoxy-benzoic acid methyl ester  for 3,4- dimethoxybenzaldehyde, TO GET 2-methoxy-5-{[1 -(4-phenyl-1 H-imidazol-2-yl)- ethylamino]-methyl}-benzoic acid methyl ester 

Example 4

(3,4-Dimethoxy-benzyl)-[1-(4-phenyl-1 H-imidazol-2-yl)-ethyl]-amine

Figure imgf000076_0001
NOTE THIS IS NOT THE COMPD….IT IS REF FOR AN ANALOGOUS PROCEDURE

A solution of 1-(4-phenyl-1 W-imidazol-2-yl)-ethylamine (0.061 g, 0.33 mmol) of Example 3, and 0.55 g (0.33 mmol) of 3,4-dimethoxybenzaldehyde in 5 ml_ of anhydrous methanol was stirred at room temperature for 1 h and then cooled to about 0-100C in an ice bath for 1 h. The reaction was treated carefully with 0.019 g (0.49 mmol) of sodium borohydride in one portion and maintained at about 0-100C for 21 h. Cold 2M aqueous HCI was added dropwise (30 drops), the mixture was stirred for 5 min, and then partially concentrated in vacuo unheated. The residual material was taken up in EtOAc to yield a suspension that was treated with 5 ml_ of cold 3M aqueous NaOH and stirred vigorously until clear. The phases were separated and the aqueous layer was extracted three times additional with EtOAc. The combined extracts were dried over MgSO4, filtered, and concentrated to yield (3,4-dimethoxy- benzyl)-[1-(4-phenyl-1 H-imidazol-2-yl)-ethyl]-amine as a light yellow oil (HPLC: 87% @ 254nm and 66% @ 214 nm).

MS (ES+) (relative intensity): 338.1 (100) (M+1)

This sample was of sufficient quality to use in the next reaction without further purification.

…………………..

SYNTHESIS

WO2006099060A2

In an embodiment, the present invention is directed to processes for the preparation of the compound of formula (IV)

Figure imgf000016_0001

also known as, 5-({[2-amino-3-(4-carbamoyl-2,5-dimethyl-phenyl)- propionyl]-[1 -(4-phenyl-1 H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy- benzoic acid

Example 1

(S)-2-ferf-Butoxycarbonylamino-3-(4-carbamoyl-2.6-dimethyl-phenyl)- propionic acid

Figure imgf000067_0001
Figure imgf000068_0001

STEP A: Trifluoromethanesulfonic acid 4-bromo-3,5-dimethyl-phenyl ester

To a cooled (0°C) solution of 4-bromo-3,5-dimethylphenol (3.05 g, 15.2 mmol) in pyridine (8 ml_) was added trifluoromethanesulfonic anhydride (5.0 g, 17.7 mmol) dropwise. After completion of addition, the resulting mixture was stirred at 0°C for 15 min, and then at room temperature overnight. The reaction was quenched by addition of water, and then extracted with EtOAc. The organic extracts were washed sequentially with water, 2N HCI (2x ), brine, and then dried over MgSO4. Filtration and evaporation to dryness yielded compound 1 b as a colorless oil.

1H NMR (300 MHz, CDCI3): δ 2.45 (6H, s), 7.00 (2H, s).

Step B: 4-Bromo-3,5-dimethylbenzoic acid

Into a solution of compound 1 b (6.57 g, 19.7 mmol) in DMF (65 ml_) were added K2CO3 (13.1 g, 94.7 mmol), Pd(OAc)2 (0.44 g, 1.97 mmol) and 1 ,1′-bis(diphenylphosphino)ferrocene (2.29 g, 4.14 mmol). The resulting mixture was bubbled in gaseous CO for 10 min and was heated to 60°C for 7.5h with a CO(9) balloon. The cooled mixture was partitioned between aqueous NaHCO3 and EtOAc, and filtered. The aqueous phase was separated, acidified with aqueous 6N HCI, extracted with EtOAc, and then dried over Na2SO4. Filtration and concentration of the filtrate yielded crude compound 1c as a brown residue, which was used in the next step without further purification. STEP C: Method A: 4-Bromo-3,5-dimethyl-benzamide

Into a suspension of compound 1c in DCM (40 ml_) was added SOCI2 (3.1 rnL, 42 mmol) and the mixture was heated at reflux for 2 h. Upon removal of the solvent by evaporation, the residue was dissolved in DCM (40 ml_) and then ammonium hydroxide (28% NH3 in water, 2.8 ml_) was added. The reaction mixture was heated at 5O0C for 2 h and concentrated. The residue was diluted with H2O, extracted with EtOAc, and the organic portion was dried over Na2SO4. After filtration and evaporation, the residue was purified by flash column chramotagraphy (eluent: EtOAc) to yield compound 1 d as an off-white solid.

1H NMR (300 MHz, CD3CN): δ 2.45 (6H, s), 5.94 (1 H, br s), 6.71 (1 H, br s), 7.57 (2H, s)

MS(ES+)(relative intensity): 228.0 (100%) (M+1).

Step C: Method B: 4-Bromo-3,5-dimethyl-benzamide

A mixture of compound 1 b (3.33 g, 10 mmol), PdCI2 (0.053 g, 0.3 mmol), hexamethyldisilazane (HMDS, 8.4 ml_, 40 mmol), and DPPP (0.12 g, 0.3 mmol) was bubbled with a gaseous CO for 5 min and then stirred in a CO balloon at 80°C for 4 h. To the reaction mixture was added MeOH (5 ml_). The reaction mixture was stirred for 10 min, diluted with 2N H2SO4 (200 ml_), and then extracted with EtOAc. The EtOAc extract was washed with saturated aqueous NaHCO3, brine, and then dried over Na2SO4. Filtration and evaporation of the resultant filtrate yielded a residue, which was purified by flash column chromatography (eluent: EtOAc) to yield compound 1d as a white solid.

Step D: 2-terf-Butoxycarbonylaminoacrylic acid methyl ester

To a suspension of /V-Boc-serine methyl ester (Compound 1e, 2.19 g, 10 mmol) and EDCI (2.01 g, 10.5 mmol) in DCM (70 ml_) was added CuCI (1.04 g, 10.5 mmol). The reaction mixture was stirred at room temperature for 72 h. Upon removal of the solvent, the residue was diluted with EtOAc, washed sequentially with water and brine and then dried over MgSO4. The crude product was purified by flash column chromatography (eluent: EtOAc:hexane ~1 :4) to yield compound 1f as a colorless oil.

1H NMR (300 MHz, CDCI3): δ 1.49 (9H, s), 3.83 (3H, s), 5.73 (1 H, d, J = 1.5 Hz), 6.16 (1 H1 S), 7.02 (1 H, s).

STEP E: (2)-2-fert-Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl- phenyl)acrylic acid methyl ester

A flask charged with compound 1d (0.46 g, 2.0 mmol), compound 1f (0.80 g, 4.0 mmol), tri-o-tolylphosphine (0.098 g, 0.32 mmol) and DMF (8 ml_) was purged with N2(g) 3 times. After the addition of tris(dibenzylideneacetone)dipalladium (0) (0.074 g, 0.08 mmol) and TEA (0.31 ml_, 2.2 mol), the reaction mixture was heated at 110°C for 24 h. At that time, the reaction was quenched by addition of water, and then extracted with EtOAc. The organic phase was washed with 1 N HCI, saturated aqueous NaHCO3, brine, and dried over MgSO4. The mixture was concentrated to a residue, which was purified by flash column chromatography (eluent: EtOAc:hexane~1 :1 to EtOAc only) to yield compound 1g as a white solid.

1H NMR (300 MHz, CD3OD): δ 1.36 (9H, s), 2.26 (6H, s), 3.83 (3H, s), 7.10 (1 H, s), 7.56 (2H, s); 13C NMR (75 MHz, DMSO-d6): δ 17.6, 25.7, 50.2, 78.7, 124.9, 126.4,

128.3, 131.2, 135.2, 135.5, 152.8, 164.3, 169.6;

MS (ES+) (relative intensity): 349.1 (38%)(M+1).

STEP F: (S)-2-ferf-Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl- phenyl)propionic acid methyl ester

Into a reactor charged with a solution of compound 1g (0.56 g, 1.6 mmol) in degassed MeOH (80 mL) was added [Rh(COd)(H1R-DIPAMP)J+BF4  under a stream of argon. The reactor was sealed and flushed with H2, stirred at 6O0C under 1000 psi of H2 for 14 days. The crude product was purified by flash column chromatography (eluent: EtOAc:hexane ~1 :1) to yield compound 1 h as a white solid. ee: >99%; 1H NMR (300 MHz, CDCI3): δ 1.36 (9H, s), 2.39 (6H, s), 3.11 (2H, J = 7.2 Hz), 3.65 (3H, s), 4.53-4.56 (1 H, m), 5.12 (1 H, d, J = 8.7 Hz), 5.65 (1 H, br s), 6.09 (1 H, br s), 7.46 (2H, s);

MS(ES+) (relative intensity): 250.9 (100) (M-BoC)+.

STEP G: (S)-2-tert-Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl- phenyl)propionic acid

Into an ice-cooled solution of compound “I h (0.22 g, 0.63 mmol) in THF (3.5 ml_) was added an aqueous LiOH solution (1 N, 3.5 ml_) and the reaction mixture stirred at 0°C. Upon completion of the reaction, the reaction mixture was concentrated and the aqueous phase was neutralized with cooled aqueous 1 N HCI at 0°C, and then extracted with EtOAc. The combined extracts were dried over Na2SO4 overnight. Filtration and evaporation of the filtrate to dryness yielded compound 1j as a white solid. 1H NMR (300 MHz, DMSO-cfe): δ 1.30 (9H, s), 2.32 (6H, s), 2.95(1 H, dd,

J= 8.8, 13.9 Hz), 3.10 (1 H, dd, J= 6.2, 14.0 Hz), 4.02-4.12 (1 H, m), 7.18-7.23 (2H, m), 7.48 (2H1 s), 7.80 (1 H, s);

MS(ES+) (relative intensity): 236.9 (6) (M-BoC)+.

Example 5

5-((r2-Amino-3-(4-carbamoyl-2.6-dimethyl-phenyl)-propionvn-n-(4-phenyl- 1 H-imidazol-2-yl)-ethvπ-aminol-methyl)-2-methoxy-benzoic acid

Figure imgf000076_0002
Figure imgf000077_0001

STEP A. 2-Methoxy-5-{[1-(4-phenyl-1 W-imidazol-2-yl)-ethylamino]-methyl}- benzoic acid methyl ester

Using the procedures described for Example 4, substituting 5-formyl-2- methoxy-benzoic acid methyl ester (WO 02/22612) for 3,4- dimethoxybenzaldehyde, 2-methoxy-5-{[1 -(4-phenyl-1 H-imidazol-2-yl)- ethylamino]-methyl}-benzoic acid methyl ester was prepared.

STEP B. 5-({[2-ferf-ButoxycarbonylmethyI-3-(4-carbamoyl-2,6-dimethyl- phenyl)-propionyl]-[1 -(4-phenyl-1 H-imidazoI-2-yl)-ethyl]-amino}-methyl)-2- methoxy-benzoic acid methyl ester

Using the procedure of Example 3 for the conversion of Cpd 3d to Cpd 3e, substituting 2-methoxy-5-{[1-(4-phenyl-1 /-/-imidazol-2-yl)-ethylamino]- methylj-benzoic acid methyl ester for Cpd 3d and substituting 2-tert- Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionic acid for 2- tø/t-Butoxycarbonylamino-3-(4-hydroxy-2,6-dimethyl-phenyl)-propionic acid, Cpd 5a was prepared.

STEP C. 5-({[2-tert-butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl- phenyl)-propionyl]-[1 -(4-phenyl-1 W-imidazol-2-yl)-ethyl]-amino}-methyl)-2- methoxy-benzoic acid

5-({[2-tørf-Butoxycarbonylmethyl-3-(4-carbamoyl-2,6-dimethyl-phenyl)- propionyl]-[1-(4-phenyl-1 H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy- benzoic acid methyl ester was dissolved in an ice-chilled (0-10°C), mixed solvent system of THF (10 ml_) and MeOH (5 ml_). A LiOH H2O/water suspension (2.48 M; 3.77 ml_) was added dropwise, then the reaction was allowed to warm to room temperature and stirred overnight. The resulting mixture was cooled in an ice bath and the basic solution was neutralized with 2N citric acid until slightly acidic. The mixture was concentrated under reduced pressure to remove the volatile materials, after which time the remaining aqueous phase was extracted with EtOAc (3 x 26 ml_). These combined organic phases were dried over MgSO4, filtered, and concentrated under reduced pressure to yield a pale yellowish white solid. This crude material was dissolved in a 10% MeOH/CH2CI2 solution and adsorbed onto 30 g of silica. The adsorbed material was divided and chromatographed on an ISCO normal phase column over two runs, using a 40 g Redi-Sep column for both runs. The solvent system was a gradient MeOHZCH2CI2 system as follows: Initial 100% CH2CI2, 98%-92% over 40 min; 90% over 12 min, and then 88% over 13 min. The desired product eluted cleanly between 44-61 min. The desired fractions were combined and concentrated under reduced pressure to yield 5-({[2-terf- butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4- phenyl-1 /-/-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid, Cpd 5b, as a white solid.

STEP D. 5-({[2-Amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1 – (4-phenyl-1 W-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid

A portion of Cpd 5b (0.27g, 0.41 mmol) was dissolved in EtOAc (39 ml_)/THF (5 ml_), filtered, and subsequently treated with gaseous HCI for 15 min. After completion of the HCI addition, the reaction was slowly warmed to room temperature and a solid precipitate formed. After 5 h the reaction appeared >97% complete by LC (@214nm; 2.56 min.). The stirring was continued over 3 d, then the solid was collected and rinsed with a small amount of EtOAc. The resulting solid was dried under high vacuum under refluxing toluene for 2.5 h to yield Cpd 5c as a white solid di-HCI salt.

Example 2

Racemic 2-terf-Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethvl- phenvD-propionic acid

Figure imgf000071_0001

STEP A: Racemic 2-tert-butoxycarbonylamino-3-(4-carbamoyl-2,6- dimethyl-phenyl)propionic acid methyl ester

To a reactor charged with a solution of compound 1g (0.68 g, 1.95 mmol) in MeOH (80 mL) was added 10% Pd-C (0.5 g). The reactor was connected to a hydrogenator and shaken under 51 psi of H2 overnight. The mixture was filtered through a pad of Celite and the filtrate was concentrated to dryness to yield compound 2a as a white solid.

The 1H NMR spectrum was identical to that of (S)-2-tert- butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl-phenyl)propionic acid methyl ester, compound 1 h.

STEP B: Racemic 2-terf-butoxycarbonylamino-3-(4-carbamoyl-2,6- dimethyl-phenyl)propionic acid

Following the procedure described for Example 1 , STEP G (preparation of (S)-2-teAt-Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl- phenyl)propionic acid), compound 2b – racemic 2-te/?-butoxycarbonylamino-3- (4-carbamoyl-2,6-dimethyl-phenyl)propionic acid – was prepared.

…………….

POLYMORPHS

US8609865

Example 1 Preparation of the zwitterion of 5-({[2-Amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid

A 1 L three-necked round-bottomed flask equipped with a mechanical stirrer, addition funnel and a thermocouple was charged without agitation. 34.2 g of 5-({[2-tert-butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid (see Example 9 of US 2005/0203143), 340 ml of acetone, and 17 ml of 204 mmolar concentrated HCl were combined in the flask. The stirring was started and the resulting slurry formed a clear solution. This solution was heated to 45° C. under vigorous stirring and aged at this temperature for a period of two hours. After the completion, the reaction mass was cooled to ambient temperature and the supernatant was removed by suction. The vessel along with the residue was rinsed with 20 ml of acetone and then removed as previously. 170 ml of water was added and the reaction mass and was aged under stirring until a homogeneus solution resulted. This solution was then added over a period of ˜½ hr to a solution of 90 ml of 1N NaOH and water. The pH was adjusted to 6.5-7.0 accordingly. The resulting slurry was aged for about 2 hrs at ambient temperature, cooled to 10-15° C., aged at that temperature for about 1 hr, and then filtered. The solid was washed with 10 ml water, air-dried for a period of 4 to 5 hrs, and then placed in a vacuum oven at 50-55° C. until the water content was less than 3%.

Example 2 Preparation of the Form α Crystal

The Form α crystal can be prepared by storing the zwitterion of 5-({[2-amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid at 0-25% relative humidity for 3 days. Representative PXRD, TGA, and DSC data are shown in FIGS. 1-3 respectively.

Example 3 Preparation of the Form β crystal

The Form β crystal can be prepared by storing the zwitterion of 5-({[2-amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid at greater than 60% relative humidity for 3 days. Representative PXRD, TGA, and DSC data are shown in FIGS. 1, 4, and 5 respectively.

…………….

SYNTHESIS

US20050203143

Example 9 5-({[2-Amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid

Figure US20050203143A1-20050915-C00035

A. 2-Methoxy-5{[1-(4-phenyl-1 H-imidazol-2-yl)-ethylamino]-methyl}-benzoic acid methyl ester.

Using the procedures described for Example 3, substituting 5-formyl-2-methoxy-benzoic acid methyl ester (WO 02/22612) for 3,4-dimethoxybenzaldehyde, 2-methoxy-5-{[1-(4-phenyl-1H-imidazol-2-yl)-ethylamino]-methyl}-benzoic acid methyl ester was prepared.

B. 5-({[2-tert-Butoxycarbonyl methyl-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid methyl ester.

Using the procedure of Example 1 for the conversion of Cpd 1d to Cpd 1e, substituting 2-methoxy-5-{[1-(4-phenyl-1H-imidazol-2-yl)-ethylamino]-methyl}-benzoic acid methyl ester for Cpd 1 d and substituting 2-tert-Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl-phenyl-propionic acid of Example 8 for 2-tert-Butoxycarbonylamino-3-(4-hydroxy-2,6-dimethyl-phenyl)-propionic acid, Cpd 9a was prepared.

C. 5-({[2-tert-butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[11-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid.

5-({[2-tert-Butoxycarbonyl methyl-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid methyl ester was dissolved in an ice-chilled (0-10° C.), mixed solvent system of THF (10 mL) and MeOH (5 mL). A LiOH.H2O/water suspension (2.48 M; 3.77 mL) was added dropwise, then the reaction was allowed to warm to room temperature and stirred overnight. The resulting mixture was cooled in an ice bath and the basic solution was neutralized with 2N citric acid until slightly acidic. The mixture was concentrated under reduced pressure to remove the volatile materials, after which time the remaining aqueous phase was extracted with EtOAc (3×26 mL). These combined organic phases were dried over MgSO4, filtered, and concentrated under reduced pressure to give 2.26 g (146% of theory) of pale yellowish white solid. This crude material was dissolved in a 10% MeOH/CH2Clsolution and adsorbed onto 30 g of silica. The adsorbed material was divided and chromatographed on an ISCO normal phase column over two runs, using a 40 g Redi-Sep column for both runs. The solvent system was a gradient MeOH/CH2Clsystem as follows: Initial 100% CH2Cl2, 98%-92% over 40 min; 90% over 12 min, and then 88% over 13 min. The desired product eluted cleanly between 44-61 min. The desired fractions were combined and concentrated under reduced pressure to yield 1.74 g (113% of theory) of 5-({[2-tert-butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid, Cpd 9b, as a white solid.

D. 5-({[2-Amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid.

A portion of Cpd 9b (0.27g, 0.41 mmol) was dissolved in EtOAc (39 mL)/THF (5 mL), filtered, and subsequently treated with gaseous HCl for 15 min. After completion of the HCl addition, the reaction was slowly warmed to room temperature and a solid precipitate formed. After 5 h the reaction appeared >97% complete by LC (@214 nm; 2.56 min.). The stirring was continued over 3 d, then the solid was collected and rinsed with a small amount of EtOAc. The resulting solid was dried under high vacuum under refluxing toluene for 2.5 h to yield 0.19 g (71%) of desired Cpd 9c as a white solid di-HCl salt.

Example 8 (S)-2-tert-Butoxycarbonylamino-3-(2,6-dimethyl-4-trifluoromethanesulfonylphenyl)-propionic acid methyl ester

Figure US20050203143A1-20050915-C00034

A. (S)-2-tert-Butoxycarbonylamino-3-(2,6-dimethyl-4-trifluoromethanesulfonylphenyl)-propionic acid methyl ester. Into a cool solution of Boc-L-(2,6-diMe)Tyr-OMe (7.0 g, 21.6 mmol; Sources: Chiramer or RSP AminoAcidAnalogues) and N-phenyltrifluoromethanesulfonimide (7.9 g, 22.0 mmol) in dichloromethane (60 mL) was added triethylamine (3.25 mL, 23.3 mmol). The resulting solution was stirred at 0° C. for 1 h and slowly warmed to rt. Upon completion, the reaction was quenched by addition of water. The separated organic phase was washed with 1 N NaOH aqueous solution, water and dried over Na2SOovernight. After filtration and evaporation, the residue was purified by flash column chromatography (eluent: EtOAc-hexane: 3:7) to give the desired product (9.74 g, 99%) as a clear oil; 1H NMR (300 MHz, CDCl3): δ 1.36 (9H, s), 2.39 (6H, s), 3.06 (2H, d, J=7.7 Hz), 3.64 (3H, s), 4.51-4.59 (1H, m), 5.12 (1H, d, J=8.5 Hz), 6.92 (2H, s); MS (ES+) (relative intensity): 355.8 (100) (M−Boc)+.

B. (S)4-(2-tert-Butoxycarbonylamino-2-methoxycarbonylethyl)-3,5-dimethylbenzoic acid. To a suspension of (S)-2-tert-butoxycarbonylamino-3-(2,6-dimethyl-4-trifluoromethanesulfonylphenyl)-propionic acid methyl ester (9.68 g, 21.3 mmol), K2CO(14.1 g, 0.102 mol), Pd(OAc)(0.48 g, 2.13 mmol) and 1,1′-bis(diphenylphosphino)ferrocene (2.56 g, 4.47 mmol) in DMF (48 mL) was bubbled in gaseous CO for 15 min. The mixture was heated to 60° C. for 8 h with a CO balloon. The cool mixture was partitioned between NaHCOand EtOAc, and filtered. The aqueous layer was separated, acidified with 10% citric acid aqueous solution, extracted with EtOAc, and finally dried over Na2SO4. Filtration and concentration of the filtrate resulted in a residue. The residue was recrystallized from EtOAc-hexanes to afford the desired product (7.05 g, 94%); 1H NMR (300 MHz, CDCl3): δ 1.36 (9H, s), 2.42 (6H, s), 3.14 (2H, J=7.4 Hz), 3.65 (3H, s), 4.57-4.59 (1H, m), 5.14 (1H, d, J=8.6 Hz), 7.75 (2H, s); MS(ES+) (relative intensity): 251.9 (100) (M−Boc)+.

C. (S)-2-tert-Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethylphenyl)propionic acid methyl ester. Into a stirring solution of (S)-4-(2-tert-butoxycarbonylamino-2-methoxycarbonylethyl)-3,5-dimethyl benzoic acid (3.00 g, 8.54 mmol), PyBOP (6.68 g, 12.8 mmol) and HOBt (1.74 g, 12.8 mmol) in DMF (36 mL) was added DIPEA (5.96 mL, 34.2 mmol) and NH4Cl (0.92 g, 17.1 mmol). The resulting mixture was stirred at rt for 40 min before being partitioned between aqueous NH4Cl solution and EtOAc. The separated organic phase was washed sequentially with 2N citric acid aqueous solution, saturated aqueous NaHCOsolution, and brine, then dried over Na2SOovernight. After filtration and concentration, the residue was purified by flash column chromatography (eluent: EtOAc) to give the product. (3.00 g, 100%); 1H NMR (300 MHz, CDCl3): δ 1.36 (9H, s), 2.39 (6H, s), 3.11 (2H, J=7.2 Hz), 3.65 (3H, s), 4.53-4.56 (1H, m), 5.12 (1H, d, J=8.7 Hz), 5.65 (1H, brs), 6.09 (1H, br s), 7.46 (2H, s); MS(ES+) (relative intensity): 250.9 (100) (M−Boc)+.

D. (S)-2-tert-Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethylphenyl)propionic acid. Into an ice-cooled solution of methyl ester from Step C (2.99 g, 8.54 mmol) in THF (50 mL) was added an aqueous LiOH solution (1N, 50 mL) and stirred at 0° C. Upon consumption of the starting materials, the organic solvents were removed and the aqueous phase was neutralized with cooled 1N HCl at 0° C., and extracted with EtOAc, and dried over Na2SOovernight. Filtration and evaporation to dryness led to the title acid (S)-2-tert-butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethylphenyl)propionic acid (2.51 g, 87%); 1H NMR (300 MHz, DMSO-d6): δ 1.30 (9H, s), 2.32 (6H, s), 2.95 (1H, dd, J=8.8, 13.9 Hz), 3.10 (1H, dd, J=6.2, 14.0 Hz), 4.02-4.12 (1H, m), 7.18-7.23 (2H, m), 7.48 (2H, s), 7.80 (1H, s); MS(ES+) (relative intensity): 236.9 (6) (M−Boc)+.

…………………..

PATENTS

1.WO 2005090315

2..WO 2006099060

3.WO 2009009480

4. WO 2010062590

5.US 2011263868 *

                   12-24-2010
                          NOVEL COMPOUNDS AS OPIOID RECEPTOR MODULATORS
                    8-32-2010
                          Compounds as opioid receptor modulators
                   6-23-2010
                          Compounds as opioid receptor modulators
                   2-12-2010
                          PROCESS FOR THE PREPARATION OF OPIOD MODULATORS
                   12-9-2009
                          Process for the preparation of opioid modulators
US7629488 * Mar 6, 2006 Dec 8, 2009 Janssen Pharmaceutica N.V. Process for the preparation of opioid modulators
US7741356 * Mar 14, 2005 Jun 22, 2010 Janssen Pharmaceutica N.V. Compounds as opioid receptor modulators
US7786158 * Oct 24, 2007 Aug 31, 2010 Janssen Pharmaceutica N.V. Compounds as opioid receptor modulators
US7994206 Jul 7, 2008 Aug 9, 2011 Janssen Pharmaceutica, N.V. Crystals and process of making 5-({[2-amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid
CN1950342A Mar 14, 2005 Apr 18, 2007 詹森药业有限公司 Novel compounds as opioid receptor modulators

 

Update july 2015

Eluxadoline

Trade Name: Viberzi®

Research Code: JNJ-27018966, JNJ27018966, JNJ 27018966

Chemical Name: 5 – [[[(2S) -2-amino-3- [4- (aminocarbonyl) -2,6-dimethylphenyl ] -1- oxopropyl] [(1S) -1- (4-phenyl-1H-imidazol-2-yl) ethyl] amino] methyl] -2-methoxybenzoic acid

CAS No: 864821-90-9

MOA: mu opioid receptor agonist

Indication: Irritable bowel syndrome with diarrhea (IBS-D)

Approval Date: May 27, 2015 (US)

Originator: Furiex Pharmaceuticals Inc ( Furiex acquired Eluxadoline from Janssen in 2011 )

Developer: Forest Laboratories Inc. (acquired by Actavis PLC in 2014 )

Synthesis of Eluxadoline (Viberzi), Actavis' new drug for irritable bowel syndrome with diarrhea synthetic route diarrhea-predominant irritable bowel syndrome medication Eluxadoline (Viberzi) of