New Drug Approvals

Home » Posts tagged 'organic chemistry' (Page 48)

Tag Archives: organic chemistry

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

Blog Stats

  • 4,815,585 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

MAA EU -GSK submits diabetes drug Eperzan, albiglutide in EU


MAA EU =marketing authorisation application EU

MAR 08 2013

GSK submits diabetes drug albiglutide in EU

GlaxoSmithKline has announced the submission of a marketing authorisation application for albiglutide, which will have the brand name Eperzan, to the European Medicines Agency.

The filing of albiglutide, a once-weekly treatment for type 2 diabetes, comes almost two months after it was filed in the USA. The drug is a GLP-1 receptor agonist, the same class of injectable treatments dominated by Novo Nordisk’s once-a-day Victoza (liraglutide), twice-daily Byetta (exenatide) and an extended-release formulation of the latter, Bydureon. They were developed and sold by Amylin, which was then acquired by  Bristol-Myers Squibb and AstraZeneca.

The filing is based in part on a study which assessed albiglutide against Merck & Co’s DPP-4 inhibitor Januvia (sitagliptin) which showed that GSK’s drug showed clinically and statistically significant reductions in HbA1c from baseline and superiority versus the US firm’s diabetes blockbuster. However in data from a late-stage study released in November 2011, albiglutide failed to show non-inferiority to Victoza and a number of analysts believe GSK will have its work cut out to grab a decent share of the GLP-1 market.

Albiglutide is a glucagon-like peptide-1 agonist (GLP-1 agonist) drug under investigation by GlaxoSmithKline for treatment of type 2 diabetes. It is a dipeptidyl peptidase-4-resistant glucagon-like peptide-1 dimer fused to human albumin.

Albiglutide has a half-life of four to seven days, which is considerably longer than the other two GLP-1 analogs approved for market use, exenatide (Byetta) and liraglutide (Victoza).[1] [2] GLP-1 drugs are currently only available for subcutaneous administration on a daily basis, so a GLP-1 drug with a longer half-life is desirable. Such a drug would only need to be injected biweekly or weekly instead of daily, reducing the discomfort and inconvenience of GLP-1 administration considerably.

It has not yet been determined whether albiglutide is as effective an antidiabetic agent as GLP-1 drugs currently on the market, and final data remain to be published regarding the incidence of adverse effects related to the drug. To evaluate the efficacy and safety of the drug, albiglutide is undergoing eight Phase III clinical trials. Four of these trials should report useful data by end 2010.[3]

Phase 2 SMP-986(afacifenacin fumarate) Overactive bladder/ Dainippon Sumitomo Pharma and Nippon Shinyaku conclude a license agreement for SMP-986, a therapeutic agent for urology


877606-63-8 cas no of afacifenacin

any str error, mail to amcrasto@gmail.com

SMP-986 (afacifenacin fumarate)

(4S)-4-phenyl-3-(1-{[3-(trifluoromethoxy)phenyl]methyl}piperidin- 4-yl)-3,4-dihydroquinazolin-2(1H)-one muscarinic receptor antagonist

  • Developed in-house
  • SMP-986 possesses the dual pharmacological actions of muscarinic receptor antagonism (non-selective) and inhibition of the bladder afferent pathway through Na+-channel blockade. This compound is being evaluated for its ability to ease urinary urgency and reduce the frequency of both urination and incontinence. The compound has also exhibited the potential to have lower incidence of side effects related to muscarinic receptor antagonism, such as dry mouth.
  • Development stage: Phase II in the U.S. and Europe. Phase II in Japan

7 mar 2013

Dainippon Sumitomo Pharma Co., Ltd. (DSP) and Nippon Shinyaku Co., Ltd. Announce they have concluded a license agreement for exclusive rights to develop, manufacture and commercialize SMP-986 in Japan, a new therapeutic agent for overactive bladder created by DSP.

Completing Phase 2 studies in Japan, Europe and the U.S., DSP was searching for a partner with a strong presence in urology. As a result, DSP chose Nippon Shinyaku as its best partner for the Japanese market.
Under this agreement, Nippon Shinyaku obtains exclusive rights to develop, manufacture and commercialize SMP-986 in Japan for general urological diseases. In return, Nippon Shinyaku will pay DSP an upfront fee and make development milestone payments. In addition, after launch Nippon Shinyaku will also pay DSP royalties according to sales amounts and milestone payments in accordance with sales goals.
SMP-986 possesses the dual pharmacological actions of muscarinic receptor antagonism and inhibition through Na+-channel blockade. In the future, Nippon Shinyaku plans to proceed with the development of SMP-986 to obtain approval for the indication of nocturia

DCVax®-Direct Phase I/II Trial For All Inoperable Solid Tumors Is Expected To Produce Ongoing Results In 2013


BETHESDA, Md., \

March 5, 2013 Northwest Biotherapeutics  (NW Bio), a biotechnology company developing DCVax® personalized immune therapies for solid tumor cancers, announced today that its Phase I/II  DCVax-Direct clinical trial for all inoperable solid tumor cancers is planned to begin within approximately the next sixty days.  As a Phase I/II trial, this trial is not blinded and the results will be seen as the trial proceeds.  With an efficacy endpoint of tumor regression (i.e., tumor shrinkage or elimination), this innovative trial is expected to yield meaningful ongoing results by the second half of 2013.

This clinical trial is approved by the FDA for all types of solid tumor cancers (i.e., cancers in any tissues of the body), and is configured to provide rapid results.  In the Phase I portion, it will test both safety and a variety of dosing regimes, and will do so in multiple different cancers – avoiding the need to conduct separate Phase I studies in each such cancer, as is usually the case.  Then, the trial will go directly into the Phase II portion, testing for efficacy, without the need for another FDA review.

DCVax®

All immune responses start in the same way that involves initially a single cell type, the dendritic cell (DC). This cell functions like the General of an army, in that it directs all ensuing activities of the immune response. The immune system is like an army with many divisions, and multiple soldiers with various types of weapons. When an invader, which could be a virus, bacteria, or a cancer cell, encounters a DC the result is that the DC consumes or eats the invader and chops it into small pieces. In the process, the DC becomes activated and starts traveling to the lymph node. In the lymph nodes the DC elicits a cascade of events eventually involving the entire army that leads to a full-blown immune response. Importantly, the nature of the invader and the nature of the DC activation dictate the type of immune response: the DC is the general of the army who directs all the soldiers to work in synchrony, and who determines which weapons to use to best defeat the enemy. We believe that preparing the DC outside the body, as done for DCVax® products, allows the greatest degree of control and begins the immune response in the natural fashion leading to the most complete attack on the foreign invader.

Different Approaches

We believe that the optimal way to arrive at the most effective immune response is through the control of the DC. Most traditional immunization approaches, including traditional virus, specific antigen or peptide vaccines as well as some that are used for immunotherapy of cancer, try to control the already existing DC in the body, or they try to modulate only one arm of the immune system. The immunogen, i.e. the virus, specific antigen, peptide or the cancer cells used to prepare the vaccine, is in those cases injected into the body in a formulation that aims at targeting and activating local DC. Examples are viral, specific antigen or peptide vaccines formulated with adjuvant, or killed tumor cells alone or modified to produce the DC mobilizing protein GM-CSF. In these instances, it is left to chance as to whether the immunogen arrives at the DC, whether the DC are properly activated, and effectively migrate to lymph nodes to produce an effective immune response. We believe that the failure of several recent clinical trials may be the result of the inability of these other approaches to effectively mount a natural and robust immune response.

Treatments that use only a single division of the immune system may employ only large amounts of T cells, or a single (monoclonal) antibody. DCVax® products are being developed to activate all aspects of the immune response, both cellular and antibody, thus potentially providing a broader and longer lasting immune and clinical response. Northwest Biotherapeutics’ products are deigned as pure, activated DC loaded with the immunogen as would naturally occur, and that are capable of migrating to lymph nodes. The intended result is a very robust, and full immune response consisting of both a specific cellular T cell response and a specific antibody response against the cancer associated antigen. Data obtained in our Phase I and Phase II clinical trials suggest that such response may occur and may translate into a clinical benefit.

Cancer and the Immune System

Cancer cells produce many substances that shut down the immune response, as well as substances that paralyze the DC that are resident in the body. We believe therefore that the optimal time for controlling cancer growth by activating the immune system is at the time when tumor burden is low. Northwest Biotherapeutics targets patients with brain cancer following surgery, radiation and chemotherapy, and hormone independent prostate cancer patients with no detectable tumor growth. This approach aims at inducing powerful immune responses to control progression of the disease.

Manufacturing

Northwest Biotherapeutics has focused on solving many of the challenges that are typically associated with producing personalized products that consist of living cells. The Company’s new automated cell processing system allows high-throughput production of products for a fraction of the historical cost.

  • DCVax® products contain pure DC
  • The DC in DCVax® are prepared outside the body, which eliminates many uncontrollable variables, and are subjected to a potency test designed to ensure that the DC administered to the patient are capable of eliciting an immune response
  • DCVax® is used in patients with low tumor burden
  • DCVax® products can be manufactured in a cost-efficient manner

sNDA – FDA accepts AMAG Feraheme (Ferumoxytol) sNDA for review


Feraheme (ferumoxytol)

Iron(II,III) oxide

Fe3O4

CUT PASTE OF INFO….

7 MAR 2013

The US Food and Drug Administration (FDA) has accepted for review AMAG Pharmaceuticals’ supplemental new drug application (sNDA) for Feraheme (ferumoxytol) injection for Intravenous (IV) use.

The sNDA filed is to expand the indication for ferumoxytol for the treatment of iron deficiency anemia (IDA) in adult patients with chronic kidney disease (CKD), who have failed or could not take oral iron treatment.

Ferumoxytol is currently indicated for oral use for the treatment of IDA in adult patients with CKD, according to the company.

The sNDA included the data from a global phase III program, which included two phase III clinical trials such as as IDA-301 (placebo comparator) and IDA-302 (active comparator).

The trials, which enrolled 1,400 patients, evaluated the use of ferumoxytol in a broad range of adult IDA patients, all of whom had failed or could not take oral iron treatment.

Both studies achieved the primary efficacy endpoints with statistically significant improvements in hemoglobin from baseline to the 35-day.

The studies, which also included patient-reported outcomes data as pre-specified secondary and exploratory endpoints, found no new safety signals, outside of those described in the current Feraheme (ferumoxytol) label, were observed with ferumoxytol treatment in these studies, claims the company.

In response to the application, the FDA said it will complete the review of Feraheme sNDA by 21 October 2013.

 

 

Feraheme, an iron replacement product, is a non-stoichiometric magnetite (superparamagnetic iron oxide) coated with polyglucose sorbitol carboxymethylether. The overall colloidal particle size is 17-31 nm in diameter. The chemical formula of Feraheme is Fe5874O8752-C11719H18682O9933Na414 with an apparent molecular weight of 750 kDa.

Feraheme injection is an aqueous colloidal product that is formulated with mannitol. It is a black to reddish brown liquid, and is provided in single use vials containing 510 mg of elemental iron. Each mL of the sterile colloidal solution of Feraheme injection contains 30 mg of elemental iron and 44 mg of mannitol, and has low bleomycin-detectable iron. The formulation is isotonic with an osmolality of 270-330 mOsm/kg. The product contains no preservatives, and has a pH of 6 to 8.

Ferumoxytol

2D chemical structure of 1309-38-2

STRUCTURE SOURCE http://chem.sis.nlm.nih.gov/chemidplus/rn/1309-38-2

EMA DOC   http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Public_assessment_report/human/002215/WC500129751.pdf

Molecular Formulas

  • Fe.O
  • Fe3-O4

Molecular Weight

  • 231.531

Ferumoxytol [USAN]
RN: 1309-38-2

Polyglucose sorbitol carboxymethyl ether-coated non-stoichiometric magnetite. Ferumoxytol is a superparamagnetic iron oxide that is coated with a low molecular weight semi-synthetic carbohydrate, polyglucose sorbitol carboxymethyl ether. The iron oxide is a superparamagnetic form of non-stoichiometric magnetite with crystal size of 6.2 to 7.3 nm. In solution, the colloidal particle of ferumoxytol has a Stokes diameter of 18-20 nm. Molecular weight is approximately 308,000

Iron oxide (Fe3O4). It is a black ore of IRON that forms opaque crystals and exerts strong magnetism. The NANOPARTICLES; and MICROSPHERES of its mineral form, magnetite, have many biomedical applications.

Ferumoxytol is the generic ingredient in one branded drug marketed by Amag Pharms Inc and is included in one NDA. There are six patents protecting this compound and one Paragraph IV challenge. Additional information is available in the individual branded drug profile pages.

This ingredient has eleven patent family members in ten countries.

There is one drug master file entry for ferumoxytol. One supplier is listed for this compound.

Phase II

Cas 722492-56-0

Launched – 2009, Anemia, iron deficiency

7228
AMI-7228
Code-7228

A superparamagnetic iron oxide (non-stoichiometric magnetite) coated with a low molecular weight semi-synthetic carbohydrate polyglucose carboxymethyl ether; USAN (OO-74) (Advanced Magnetics, Cambridge, MA, USA)

Other Names

  • C 7228
  • Code 7228
  • Cytogen
  • Feraheme
  • Rienso

 

Superparamagnetic iron oxide coated with a low molecular weight semi-synthetic carbohydrate polyglucose sorbitol carboxymethyl ether. The iron oxide is a superparamagnetic form of non-stoichiometric magnetite with crystal size of 6.2 to 7.3 nm. In solution, the colloidal particle has a Stokes diameter of 18-20 nm

CLICK ON IMAGE

 

CLICK O IMAGE

Feraheme, an iron replacement product, is a non-stoichiometric magnetite (superparamagnetic iron oxide) coated with polyglucose sorbitol carboxymethylether. The overall colloidal particle size is 17-31 nm in diameter. The chemical formula of Feraheme is Fe5874O8752C11719H18682O9933Na414 with an apparent molecular weight of 750 kDa.

Feraheme Injection is an aqueous colloidal product that is formulated with mannitol. It is a black to reddish brown liquid, and is provided in single use vials containing 510 mg of elemental iron. Each mL of the sterile colloidal solution of Feraheme Injection contains 30 mg of elemental iron and 44 mg of mannitol, and has low bleomycin-detectable iron. The formulation is isotonic with an osmolality of 270-330 mOsm/kg. The product contains no preservatives, and has a pH of 6 to 8.

 

Ferumoxytol is AMAG Pharmaceuticals’ lead investigational compound. In 2007, the company filed a regulatory application seeking approval in the U.S. for use as an intravenous iron replacement therapeutic in patients who may be on dialysis and are suffering from anemic chronic kidney disease (CKD). In 2009, FDA approval was assigned and the product became available on the market. A regulatory application was filed in the E.U. in 2010 for this indication and a positive opinion was received in 2012. Final E.U. approval was obtained in June 2012. In 2012, AMAG Pharmaceuticals submitted a supplemental NDA to the FDA for the treatment of patients with iron-deficiency anemia (IDA) who are not candidates for oral iron, for which they received a complete response letter in January 2014. In 2013, Takeda filed for approval for this indication in the E.U. This application was withdrawn in 2015 due to safety concerns.

In terms of clinical studies, phase II trials are underway at AMAG and at Oregon Health and Science University for use in magnetic resonance angiography (MRA). The National Cancer Institute is also conducting phase II trials for the imaging of primary high-grade brain tumors and/or cerebral metastases from lung or breast cancer. Phase I clinical trials are ongoing at Dana-Farber Cancer Institute for use in magnetic resonance imaging in pediatric and adult patients with malignant sarcoma.

The drug consists of intravenously administered bioavailable iron which allows for more efficient replenishment of the body’s iron stores than oral iron supplements, without their associated common side effects. Ferumoxytol is a blood pool agent, a true intravascular contrast agent that remains in the blood stream for an extended period of time. Based on this quality, the product may be useful as a contrast agent in a wide range of applications in MRI.

In 2008, fast track designation was received in the U.S. as a diagnostic agent for vascular-enhanced magnetic resonance imaging (VE-MRI) to improve the assessment of peripheral arterial disease in patients with known or suspected chronic kidney disease. In 2010, a license, development and commercialization agreement was established between Takeda and AMAG Pharmaceuticals in Asia Pacific countries (excluding Japan, China and Taiwan), Canada, Europe, the Commonwealth of Independent States and Turkey. However, in December 2014, both companies announced the termination of this license agreement. In 2011, orphan drug designation was assigned by the FDA for use in magnetic resonance imaging in brain metastases. This designation was assigned in 2012 for use in magnetic resonance imaging to assess, and monitor treatment of solid tumor malignancies previously diagnosed in pediatric patients (age 16 years and younger).

SFDA

STR1

As announced in May 2008, we entered into a development and commercialization agreement with AMAG Pharmaceuticals, Inc. (“AMAG”) (NASDAQ:AMAG), a US biopharmaceutical company, for ferumoxytol, an intravenous iron replacement therapeutic agent being developed to treat iron deficiency anemia in CKD patients and in patients requiring hemodialysis.

Under the terms of the agreement, AMAG granted us exclusive rights to develop and commercialize ferumoxytol in the PRC, initially for CKD, and with an option to expand into additional indications. We will be responsible for the clinical development, registration, and commercialization of ferumoxytol in the PRC. We and AMAG will form a joint steering committee, with equal representation from both parties, to oversee and guide the development and commercialization of ferumoxytol in China. The agreement has an initial duration of 13 years and will be automatically renewed for a set term if minimum sales thresholds are achieved. AMAG will retain all manufacturing rights for ferumoxytol and will provide, under a separate agreement, commercial supply to us at a predetermined supply price.

Ferumoxytol was approved in June 2009 by the U.S. Food and Drug Administration to treat iron deficiency anemia in CKD patients and launched commercially in the U.S. by AMAG in July 2009. Ferumoxytol received marketing approval in Canada in December 2011 and a positive recommendation for approval from the Committee for Medicinal Products for Human Use of the European Medicines Agency in April 2012.

We have submitted the application for a registrational clinical trial for ferumoxytol to SFDA, as announced in January 2010. Once approved by the SFDA, we will commence a multi-center randomized efficacy and safety study in China with approximately 200 CKD patients, measuring the mean change in hemoglobin from baseline at Day 35 after first dose.

https://www.google.com/patents/US20100266644

Ferumoxytol is a newer parenteral iron formulation but limited information is available as to its efficacy and administration. See e.g., Landry et al. (2005) Am J Nephrol 25, 400-410, 408; and Spinowitz et al. (2005) Kidney Intl 68, 1801-1807; U.S. Pat. No. 6,599,498.

Another example of a preferred iron carbohydrate complex for use in the methods described herein is a carboxyalkylated reduced polysaccharide iron oxide complex (e.g., ferumoxytol, described in U.S. Pat. No. 6,599,498).

Another preferred iron carbohydrate complex for use in the methods described herein is a polyglucose sorbitol carboxymethyl ether-coated non-stoichiometric magnetite (e.g., “ferumoxytol”). Ferumoxytol is known in the art to be effective for treating anemia (at single unit doses lower than described herein). See e.g., Spinowitz et al. (2005) Kidney Intl 68, 1801-1807. Ferumoxytol is a superparamagnetic iron oxide that is coated with a low molecular weight semi-synthetic carbohydrate, polyglucose sorbitol carboxymethyl ether. Ferumoxytol and its synthesis are described in U.S. Pat. No. 6,599,498, incorporated herein by reference. Safety, efficacy, and pharmacokinetics of ferumoxytol are as described, for example, in Landry et al. (2005) Am J Nephrol 25, 400-410, 408; and Spinowitz et al. (2005) Kidney Intl 68, 1801-1807.

The iron oxide of ferumoxytol is a superparamagnetic form of non-stoichiometric magnetite with a crystal size of 6.2 to 7.3 nm. Average colloidal particle size can be about 30 nm, as determined by light scattering. Molecular weight is approximately 750 kD. The osmolarity of ferumoxytol is isotonic at 297 mOsm/kg and the pH is neutral. The blood half-life of ferumoxytol is approximately 10-14 hours. It has been previously reported that ferumoxytol can be given by direct intravenous push over 1-5 minutes in doses up to 1,800 mg elemental iron per minute, with maximal total dose up to 420 mg per injection. Landry et al. (2005) Am J Nephrol 25, 400-410, 408.

About Feraheme® (ferumoxytol)/Rienso

In the United States, Feraheme (ferumoxytol) Injection for Intravenous (IV) use is indicated for the treatment of iron deficiency anemia (IDA) in adult patients who have failed oral iron therapy. Feraheme received marketing approval from the FDA on June 30, 2009 for the treatment of IDA in adult chronic kidney disease (CKD) patients and was commercially launched by AMAG in the U.S. shortly thereafter.

Ferumoxytol is protected in the U.S. by five issued patents covering the composition and dosage form of the product. Each issued patent is listed in the FDA’s Orange Book. These patents are set to expire in March 2020; a request for patent term extension has been filed, which, if granted, may extend the patent term to June 2023 for one of the patents.

Ferumoxytol received marketing approval in Canada in December 2011, where it is marketed by Takeda as Feraheme, and in the European Union in June 2012 and Switzerland in August 2012, where it is marketed by Takeda as Rienso.

For additional U.S. product information, including full prescribing information, please visit www.feraheme.com.

AMAG now has five Orange Book-listed patents for ferumoxytol, with patent protection through March 2020, without patent term extension. AMAG has applied for a patent term extension for an Orange Book-listed ferumoxytol patent, which would lengthen that patent term through June 2023.

 

 

//////////Ferumoxytol, AMAG Pharmaceuticals, Phase II,  722492-56-0, Launched,  2009, Anemia, iron deficiency, 7228  , AMI-7228  , Code-7228

[Fe](O[Fe]=O)O[Fe]=O

NDA FDA-Nuvo reports FDA response to PENNSAID 2% , diclofenac sodium topical solution, 2% w/w


DICLOFENAC

 

 

PENNSAID 2%

7 MAR 2013

The US Food and Drug Administration (FDA) has issued a Complete Response Letter (CRL) to Nuvo Research’s US licensing partner, Mallinckrodt, following the review of Mallinckrodt’s New Drug Application (NDA) for diclofenac sodium topical solution, 2% w/w (PENNSAID 2%).

FDA in the letter mentioned that it requires Mallinckrodt’s complete pharmacokinetic study comparing PENNSAID 2% to original PENNSAID 1.5%.

FDA denied to review the similar pharmacokinetic studies submitted by Mallinckrodt with the NDA, as the reserve samples were not retained at the clinical site.

Pharmacokinetic studies are standard studies conducted during a drug development program to identify the total exposure or the amount of drug that reaches the blood stream after a patient receives both single and multiple doses of the product.

Mallinckrodt has suggested Nuvo that it expects to complete the study and submit the results to the FDA in the third quarter of 2013, and that it anticipates the FDA will provide a formal response to the filing within 6 months thereafter.

Nuvo’s Pain Group president Dr. Bradley Galer said with the new FDA’s letter the firm was disappointed that PENNSAID 2% will not be approved in this review cycle.

“We are pleased that the FDA has outlined a clear pathway to approval that we believe can be completed in a relatively short time frame,” Galer added.

“Upon approval, PENNSAID 2% will be the first and only topical NSAID in the U.S. featuring twice per day dosing and a metered dose pump bottle.”

Phase 3-Trius Therapeutics will soon be reporting data from its second phase III trial of Tedizolid


ChemSpider 2D Image | Torezolid | C17H15FN6O3

Tedizolid

(5R)-3-{3-fluoro-4-[6-(2-methyl-2H-tetrazol-5-yl)pyridin-3-yl]phenyl}-5-(hydroxymethyl)-1,3-oxazolidin-2-one

  • Molecular Formula: C17H15FN6O3
  • Average mass: 370.337799

856866-72-3  cas no

Torezolid (also known as TR-701 and now tedizolid[1]) is an oxazolidinone drug being developed by Trius Therapeutics (originator Dong-A Pharmaceuticals) for complicated skin and skin-structure infections (cSSSI), including those caused by Methicillin-resistantStaphylococcus aureus (MRSA).[2]

As of July 2012, tedizolid had completed one phase III trial, with another one under way. [3]Both trials compare a six-day regimen of tedizolid 200mg once-daily against a ten-day regimen of Zyvox (linezolid) 600mg twice-daily.

The prodrug of tedizolid is called “TR-701”, while the active ingredient is called “TR-700”.[4][5]

March 5 2013

Trius Therapeutics will soon be reporting data from its second phase III trial (ESTABLILSH-2) and the recently announced publication of the data from its first phase III trial (ESTABLISH-1) in the Journal of the American Medical Association (JAMA)

 

  1. “Trius grows as lead antibiotic moves forward”. 31 Oct 2011.
  2. “Trius Completes Enrollment In Phase 2 Clinical Trial Evaluating Torezolid (TR-701) In Patients With Complicated Skin And Skin Structure Infections”. Jan 2009.
  3. http://clinicaltrials.gov/ct2/results?flds=Xf&flds=a&flds=b&term=tedizolid&phase=2&fund=2&show_flds=Y
  4. PMID 19528279 In vitro activity of TR-700, the active ingredient of the antibacterial prodrug TR-701, a novel oxazolidinone antibacterial agent.
  5. PMID 19218276 TR-700 in vitro activity against and resistance mutation frequencies among Gram-positive pathogens.

Phase 1-Sangamo Presents New Clinical Data at CROI 2013 Demonstrating Persistent Immune System Improvements After Treatment With ZFN Therapeutic(R) SB-728-T


 

The gene therapy diminished the levels of virus and eradicated in patients having naturally occurring mutation of gene, found a preliminary trail of HIV treatment. The first phase of very small trail tested the SB-728-T gene treatment that is intended to interrupt theCCR5 gene used by HIV to contaminate immune system cells.

The first clinical trial using zinc-finger nucleases to provide long-term resistance to HIV-1 infection has been given the go-ahead by the US Food and Drug Administration. Sangamo BioSciences of Richmond, California, and its clinical partner, the University of Pennsylvania, have begun enrolling the first 12 people in a phase 1 clinical trial to evaluate SB-728-T, a novel zinc-finger DNA-binding nuclease that permanently disrupts the CCR5 gene on CD4+ T cells (Nat. Biotechnol. 26, 808–816, 2008

Data Demonstrate that SB-728-T Possesses Necessary Immunologic Properties to Support a ‘Functional Cure’ for HIV/AIDS

RICHMOND, Calif., March 6, 2013

Sangamo BioSciences, Inc. announced new data from its program to develop a ‘functional cure’ for  HIV/AIDS  in two presentations at the 20th Conference on Retroviruses and Opportunistic Infections (CROI), held in Atlanta from March 3 to 6, 2013.

The first presentation described data from the SB-728-T Phase 1 study (SB-728-902, Cohorts 1-3) demonstrating that SB-728-T treatment of HIV-infected subjects leads to durable reconstitution of the immune system driven by increases in total CD4+ central memory T-cells (TCM) and CCR5-protected TCM. TCM are long-lived, self-renewing cells that have the ability to remember and react against foreign antigens including HIV.  The data also showed that certain cell surface marker and gene expression profiles may predict which patients will likely respond best to SB-728-T treatment.

About Sangamo


Sangamo BioSciences, Inc. is focused on research and development of novel DNA-binding proteins for therapeutic gene regulation and genome editing. The Company has ongoing Phase 2 clinical trials to evaluate the safety and efficacy of a novel ZFP Therapeutic® for the treatment of HIV/AIDS. Sangamo’s other therapeutic programs are focused on monogenic diseases, including hemophilia, Huntington’s disease and  hemoglobinopathies such as beta-thalassemia and sickle cell anemia. Sangamo’s core competencies enable the engineering of a class of DNA-binding proteins known as zinc finger DNA-binding proteins (ZFPs).  Engineering of ZFPs that recognize a specific DNA sequence enables the creation of sequence-specific ZFP Nucleases (ZFNs) for gene modification and ZFP transcription factors (ZFP TFs) that can control gene expression and, consequently, cell function. Sangamo has entered into a strategic collaboration with Shire AG to develop therapeutics for hemophilia, Huntington’s disease and other monogenic diseases and has established strategic partnerships with companies in non-therapeutic applications of its technology including Dow AgroSciences and Sigma-Aldrich Corporation. For more information about Sangamo, visit the company’s website atwww.sangamo.com.

Phase III Study of Oral Laquinimod for Relapsing-Remitting Multiple Sclerosis


Laquinimod

5-chloro-N-ethyl-4-hydroxy-1-methyl-2-oxo-
N-phenyl-1,2-dihydroquinoline-3-carboxamide

Laquinimod is an experimental immunomodulator developed by Active Biotech and Teva. It is currently being investigated as an oral treatment for multiple sclerosis (MS).

Laquinimod is the successor of Active Biotech’s failed experimental immunomodulator linomide.[1]

The compound has been investigated in two Phase II trials using successive magnetic resonance scans (MRI). Laquinimod seems to be able to reduce the MS disease activity on MRI.[2][3] However, the response to a given dose was discrepant between both studies.[4]

Phase III studies for MS started in December 2007.[5] In 2011, Teva announced its clinical trials involving laquinimod had failed, being unable to significantly reduce relapses into MS among patients beyond a placebo.[6] However, the final results of above mentioned phase III trial proved oral laquinimod administered once daily slowed the progression of disability and reduced the rate of relapse in patients with relapsing–remitting multiple sclerosis [7]

Mar 6, 2013 –

CONCERTO Study Enrolling Patients Globally to Evaluate Impact of Laquinimod on Disability Progression

Teva Pharmaceutical Industries Ltd.  and Active Biotech  announced today enrollment of the first patient in the CONCERTO study – the third Phase III placebo-controlled study designed to evaluate the efficacy, safety and tolerability of once-daily oral laquinimod in patients with relapsing-remitting multiple sclerosis (RRMS). The primary outcome measure of CONCERTO will be confirmed disability progression as measured by the Expanded Disability Status Scale (EDSS).

“Previous Phase III studies in more than 2,400 people with RRMS suggest a unique profile of laquinimod, directly affecting the neurodegenerative processes that lead to disability progression, the main concern in the treatment of RRMS,” said CONCERTO principal investigator, Dr. Timothy Vollmer, Professor of Neurology, University of Colorado Denver, Medical Director of the Rocky Mountain Multiple Sclerosis Center, and Co-Director of the RMMSC at Anschutz. “We are currently enrolling patients in this third pivotal study to further examine the clinical benefits of laquinimod on disability progression, the primary endpoint of the CONCERTO trial, and brain atrophy, at both the previously studied 0.6 mg dose, and now a higher 1.2 mg dose.”

The multinational, randomized, double blind placebo-controlled study will aim to enroll approximately 1,800 patients at more than 300 sites globally (http://clinicaltrials.gov/show/NCT01707992). Along with the primary endpoint of time to confirmed disability progression, the study will also examine the impact of laquinimod on endpoints such as percent change in brain volume and other clinical and MRI markers of disease activity.

“For nearly 30 years, Teva has been focused on improving the lives of people with multiple sclerosis by delivering innovative treatment options that address this complex disease,” said Dr. Michael Hayden, President of Global R&D and Chief Scientific Officer at Teva Pharmaceutical Industries Ltd. “The CONCERTO study demonstrates our commitment to collaborating with MS communities worldwide to further develop laquinimod and address unmet patient needs.”

ABOUT LAQUINIMOD

Laquinimod is an oral, once-daily CNS-active immunomodulator with a novel mechanism of action being developed for the treatment of MS. In animal models laquinimod crosses the blood brain barrier to potentially have a direct effect on resident CNS inflammation and neurodegeneration. The global Phase III clinical development program evaluating oral laquinimod in MS includes two pivotal studies, ALLEGRO and BRAVO.

In addition to the MS clinical studies, laquinimod is currently in clinical development for Crohn’s disease and Lupus.

ABOUT CONCERTO

CONCERTO is a multinational, multicenter, randomized, double-blind, parallel-group, placebo-controlled study followed by an active treatment phase, to evaluate the efficacy, safety and tolerability of two doses of oral administration of laquinimod 0.6 mg/day or 1.2 mg/day in subjects with RRMS. This third Phase III laquinimod study will evaluate laquinimod in approximately 1,800 patients for up to 24 months, after which patients will continue to an active treatment period with laquinimod for an additional 24 months. The primary outcome measure will be time to confirmed disability progression as measured by the Expanded Disability Status Scale (EDSS). The study will also examine the impact of laquinimod on endpoints such as percent change in brain volume, as well as other clinical and MRI markers of disease activity.

ABOUT MULTIPLE SCLEROSIS

MS is the leading cause of neurological disability in young adults. It is estimated that more than 400,000 people in the United States are affected by the disease and that two million people may be affected worldwide. Multiple sclerosis is a degenerative disease of the central nervous system in which inflammation and axonal damage and loss result in the development of progressive disability.

ABOUT TEVA

Teva Pharmaceutical Industries Ltd. (NYSE: TEVA) is a leading global pharmaceutical company, committed to increasing access to high-quality healthcare by developing, producing and marketing affordable generic drugs as well as innovative and specialty pharmaceuticals and active pharmaceutical ingredients. Headquartered in Israel, Teva is the world’s leading generic drug maker, with a global product portfolio of more than 1,000 molecules and a direct presence in about 60 countries. Teva’s branded businesses focus on CNS, oncology, pain, respiratory and women’s health therapeutic areas as well as biologics. Teva currently employs approximately 46,000 people around the world and reached $20.3 billion in net revenues in 2012.

ABOUT ACTIVE BIOTECH

Active Biotech AB is a biotechnology company with focus on autoimmune/inflammatory diseases and cancer. Projects in or entering pivotal phase are laquinimod, an orally administered small molecule with unique immunomodulatory properties for the treatment of multiple sclerosis, TASQ for prostate cancer as well as ANYARA for use in cancer targeted therapy, primarily of renal cell cancer. In addition, laquinimod is in Phase II development for Crohn’s and Lupus. Further projects in clinical development comprise the two orally administered compounds, 57-57 for SLE & Systemic Sclerosis and RhuDex(TM) for RA. Please visit http://www.activebiotech.com for more information.

  1.  Tan IL, Lycklama à Nijeholt GJ, Polman CH et al. (April 2000). “Linomide in the treatment of multiple sclerosis: MRI results from prematurely terminated phase-III trials”. Mult Scler 6 (2): 99–104. PMID 10773855.
  2. Comi G, Pulizzi A, Rovaris M et al. (June 2008). “Effect of laquinimod on MRI-monitored disease activity in patients with relapsing-remitting multiple sclerosis: a multicentre, randomised, double-blind, placebo-controlled phase IIb study”. Lancet 371 (9630): 2085–2092. doi:10.1016/S0140-6736(08)60918-6. PMID 18572078.
  3.  Polman C, Barkhof F, Sandberg-Wollheim M et al. (March 2005). “Treatment with laquinimod reduces development of active MRI lesions in relapsing MS”. Neurology 64 (6): 987–91. doi:10.1212/01.WNL.0000154520.48391.69. PMID 15781813.
  4. Keegan BM, Weinshenker BG (June 2008). “Laquinimod, a new oral drug for multiple sclerosis”. Lancet 371 (9630): 2059–2060. doi:10.1016/S0140-6736(08)60894-6. PMID 18572062.
  5. ClinicalTrials.gov NCT00509145 Safety and Efficacy of Orally Administered Laquinimod Versus Placebo for Treatment of Relapsing Remitting Multiple Sclerosis (RRMS) (ALLEGRO)
  6. Kresege, Naomi (1 August 2011). “Teva’s Copaxone Successor Fails in Latest Clinical Trial”. Bloomberg. http://www.bloomberg.com/news/2011-08-01/teva-s-oral-multiple-sclerosis-drug-fails-to-meet-goal-of-clinical-trial.html. Retrieved 2 August 2011. “Teva Pharmaceutical Industries Ltd. (TEVA)’s experimental multiple sclerosis pill failed to reduce relapses more than placebo in a clinical trial, dealing a blow to the company’s effort to find a successor to an older drug.”
  7. (Comi et al. N Engl J Med 2012;366:1000).

EP 1073639; JP 2002513006; US 6077851; WO 9955678

5-Chloroisatoic anhydride (I) is alkylated with iodomethane and NaH to afford (II). Subsequent condensation of anhydride (II) with the malonic monoamide (III) in the presence of NaH in hot DMA furnishes the target quinoline carboxamide.

Reaction of 2-amino-6-chlorobenzoic acid (I) with phosgene and NaHCO3 in dioxane gives 5-chloroisatoic anhydride (II), which is methylated by means of iodomethane and NaH in DMF to yield 5-chloro-1-methylisatoic anhydride (III). Finally, anhydride (III) is condensed with the malonic monoamide (IV) by means of NaH in hot dimethylacetamide. Alternatively, condensation of anhydride (III) with ethoxy malonyl chloride (V) by means of NaOMe and triethylamine in dichloromethane affords 5-chloro-4-hydroxy-1-methyl-2-oxo-1,2-dihydroquinoline-3- carboxylic acid ethyl ester (VI), which is finally condensed with N-ethylaniline (VII) in refluxing toluene. Alternatively, ester (VI) is hydrolyzed by means of concentrated HCl in hot Ac2O to give the carboxylic acid (VIII), which is finally condensed with N-ethylaniline (VII) by means of SOCl2 and TEA in dichloromethane

PHASE1,Progenics Pharmaceuticals’ Novel Small Molecule Drugs Targeting PSMA Successfully Visualize Prostate Cancer, 123-I-MIP-1095


Name:  123-I-MIP-1095

Synonym:   123-I-MIP-1095;     [123I]-MIP-1095;  iodine I 123 IMP-1095;       2-(3-{l-carboxy-5-[3-(4-iodo-phenyl)-ureido]-pentyl}-ureido)-pentanedioic acid.;   [123I]-(S)-2-(3-((S)-1-carboxy-5-(3-(4-iodophenyl)ureido)pentyl)ureido)pentanedioic acid

 

IUPAC/Chemical name: 

2-(3-(1-carboxy-5-(3-(4-iodophenyl)ureido)pentyl)ureido)pentanedioic acid

Chemical Formula: C19H25123IN4O8

Exact Mass: 560.07284
Molecular Weight: 560.33

123-I-MIP-1095
An iodine 123-radiolabled small molecule that exhibits high affinity for prostate-specific membrane antigen (PSMA) with potential use in molecular imaging. 123-I-MIP-1095, a radiolabeled glutamate-urea-lysine analogue, selectively binds PSMA, which allows imaging of PSMA-expressing prostate cancer cells with gamma scintigraph. PSMA is a transmembrane glycoprotein highly expressed by malignant prostate epithelial cells and vascular endothelial cells of various solid tumors.

Synonym: iodine I 123 IMP-1095
Chemical structure: 2-(3-{l-carboxy-5-[3-(4-iodo-phenyl)-ureido]-pentyl}-ureido)-pentanedioic acid

March 5, 2013

Progenics Pharmaceuticals, Inc. (Nasdaq:PGNX) reported positive clinical data from a study of two novel radiolabeled small molecules targeting prostate-specific membrane antigen (PSMA). The imaging agents — 123I-MIP-1072 and 123I-MIP-1095 — had a high sensitivity of lesion detection in bone, tissue and the prostate gland with minimal retention in non-target tissue. The research was published as the cover article in the March issue of The Journal of Nuclear Medicine.

“Existing imaging techniques are limited in their ability to diagnose and stage prostate cancer,” said John J. Babich, Ph.D., senior author of the article “First-in-Man Evaluation of Two High-Affinity PSMA-Avid Small Molecules for Imaging Prostate Cancer.” “The approach described in this paper has the potential to assess disease status more accurately. It could help clinicians select optimal treatments and lead to better patient outcomes.”

Separate phase 1 studies were conducted under an exploratory investigational new drug (IND) application to measure the potential effectiveness of the small molecules in diagnosing and staging prostate cancer. In the first study, seven patients with documented prostate cancer were administered doses of 123I-MIP-1072 and 123I-MIP-1095, two weeks apart. In the second study, six healthy volunteers received 123I-MIP-1072 only. Whole body planar imaging and single photon emission computed tomography (SPECT)/computed tomography (CT) were performed for each group, and pharmacokinetics, tissue distribution, excretion, safety and organ radiation dose were analyzed.

Based on the data reported, Progenics is conductinga global, multi-center phase 2 trial investigating a next generation radiolabeled small molecule targeting PSMA, MIP-1404.

Mark R. Baker, chief executive officer of Progenics, said, “We recently acquired all of the rights to the compounds described in this Journal of Nuclear Medicine paper, as well as to the phase 2 stage imaging agent MIP-1404, through Progenics’ acquisition of Molecular Insight Pharmaceuticals. It is gratifying to see this expansion of our oncology pipeline demonstrating progress so soon.”

Robert J. Israel, M.D., Progenics’ senior vice president of medical affairs and clinical research, said, “We believe that MIP-1404 has excellent potential as a diagnostic radiopharmaceutical. Results to date from the study compounds and MIP-1404 show PSMA as a robust target for prostate cancer molecular imaging, and that a radiolabeled small molecule, which binds PSMA with high affinity, has the potential to detect prostate cancer throughout the body. Cancer treatment guidelines call for imaging prostate cancer with conventional bone scans or MRI. A more accurate method of imaging prostate cancer could be of great value.”

Mr. Baker further added, “Thought leaders in prostate cancer care are focused on avoiding unnecessary surgery and other invasive procedures due to the complications associated with them. Clinicians generally prefer “watchful waiting” when the cancer appears to be indolent. At the same time, some therapeutics to treat aggressive prostate cancer have recently been approved or are under development, such as Progenics’ own PSMA ADC, which currently is in phase 2 testing. Patients and their physicians would benefit from feedback on how therapeutic agents are impacting the course of cancer, and guidance on how and when to use therapeutic agents. It is clear that an improved way to visualize prostate cancer, with a high degree of specificity and sensitivity, would better inform both “watchful waiting” and the treatment of aggressive disease. We believe that data from the ongoing phase 2 trial of MIP-1404 will demonstrate its capabilities to assist prostate cancer patients and their physicians in making these critical decisions.”

About Prostate Cancer

Prostate cancer is the most common form of cancer affecting men in the United States and is the second leading cause of cancer deaths among men each year. The American Cancer Society estimates that in 2013, 238,590 new cases of prostate cancer will be diagnosed and approximately 29,720 American men will die from the disease. Accurate diagnosis and staging of prostate cancer is critical to determining appropriate patient management.

About Progenics

Progenics Pharmaceuticals, Inc. is discovering and developing innovative medicines for oncology, with a pipeline that includes product candidates in preclinical through late-stage development. Progenics’ first commercial product, Relistor® (methylnaltrexone bromide) for opioid-induced constipation, is marketed and in further development by Salix Pharmaceuticals, Ltd. for markets worldwide other than Japan, where Ono Pharmaceutical Co., Ltd. holds an exclusive license for the subcutaneous formulation. For additional information, please visit http://www.progenics.com.

Betrixaban


N-(5-chloropyridin-2-yl)-2-([4-(N,N-dimethylcarbamimidoyl)benzoyl]amino)-5-methoxybenzamide

Betrixaban:PRT-54021, PRT-021, MK-4448, PRT-054021

N- (5- chloro-2-pyridyl) -2 – [[4 – [(dimethylamino) methyl] benzoyl] amino] -5 – methoxy – benzamide

CAS 330942-05-7

MW 451.91, C23H22ClN5O3

Venous Thromboembolism (VTE)

Millennium INNOVATOR

Takeda Pharmaceutical Co Ltd

Lee’s Pharmaceutical Holdings (Hong Kong) Ltd; Portola Pharmaceuticals Inc…DEVELOPERS

Ever since post was written now, FDA approval on June 23rd, 2017

The U.S. Food and Drug Administration (FDA) has approved betrixaban for the prophylaxis of venous thromboembolism (VTE) in adults hospitalized for an acute medical illness who are at risk for thromboembolic complications (related to limited mobility or other risk factors for VTE). Betrixaban is now the fifth FDA-approved oral anticoagulant on the market.

The decision was based on data from the phase III APEX trial, a double-blind, international study that randomized 7,513 patients to receive either extended-duration betrixaban (betrixaban 160 mg orally on day 1, then 80 mg daily for 35 to 42 days, followed by a placebo injection once-daily for 6 to 14 days) or short-duration enoxaparin (enoxaparin 40 mg subcutaneously once-daily for 6 to 14 days followed by an oral placebo pill once-daily for 35 to 42 days).

Image result for betrixabanImage result for betrixabanImage result for betrixaban

Patients in the betrixaban arm experienced fewer VTE events, a composite outcome score of asymptomatic or symptomatic proximal deep vein thrombosis, non-fatal pulmonary embolism, or VTE-related death: 4.4 percent versus 6 percent (relative risk = 0.75, 95% CI 0.61-0.91).

Fifty-four percent of betrixaban-treated patients experienced at least one adverse event (AE), compared with 52 percent of those on enoxaparin. The most common AEs (observed in ≥5% of patients) associated with betrixaban were bleeding-related, and bleeding was the most common reason for treatment discontinuation.

UNII-28Z3021TMU.png

Betrixaban maleate

CAS 936539-80-9,

Molecular Weight, 567.98, Molecular Formula, C23H22ClN5O3 . C4H4O4

(2Z)-but-2-enedioic acid; N-(5-chloropyridin-2-yl)-2- [4-(N,N-dimethylcarbamimidoyl)benzamido]-5- methoxybenzamide

Image result for betrixabanImage result for betrixaban

STR2STR1

STR1

STR2STR1

https://www.accessdata.fda.gov/drugsatfda_docs/nda/2017/208383Orig1s000ChemR.pdf

FDA approval on June 23rd, 2017. FDA approved betrixaban (BEVYXXA, Portola) for the prophylaxis of venous thromboembolism (VTE) in adult patients”

Image result for betrixaban

Image result for betrixaban

Image result for betrixabanImage result for betrixaban

血栓新药Bevyxxa(betrixaban,贝曲西班)的合成_syntheticfuture_新浪博客

新浪博客690 × 529Search by image

血栓新药Bevyxxa(betrixaban,贝曲西班)的合成
str6

Conversion of the carboxylic acid compound S-1 to the acid chloride followed by reaction with the aminopyridine S-2 gives the amide compound, which is subsequently hydro-reduced to give the compound S-4 . Dimethylamine in the presence of a strong base to deprotonated proton nitrile compound to obtain amidine compounds S-6 , hydrolysis ester group to give carboxylic acid compound S-7 . S-7 and S-4 resulted in Bevyxxa ( betrixaban ) with the participation of the condensation reagent EDC .

Synthetic route reference: WO2011084519A1

STR1STR2str3str4

Betrixaban, a factor Xa (FXa) inhibitor, is chemically described as N-(5-chloropyridin-2-yl)-2[4-(N,N-dimethylcarbamimidoyl)-benzoylamino]-5-methoxybenzamide maleate. Its molecular formula (as maleate salt) is C27H26ClN5O7, which corresponds to a molecular weight of 567.98. Betrixaban (maleate salt) has the following structural formula:

BEVYXXA™ (betrixaban) Structural Formula Illustration

BEVYXXA capsules are available for oral administration in strengths of 80 mg and 40 mg of betrixaban with the following inactive ingredients: dextrose monohydrate, croscarmellose sodium, magnesium stearate, and a hard gelatin capsule.

Patents

  1. US8557852
  2. US6376515
  3. US8691847
  4. US9629831
  5. US9555023
  6. US8404724
  7. US8987463
  8. US7598276
  9. US6835739
  10. US8518977

FDA Orange Book Patents

FDA Orange Book Patents: 1 of 10 (FDA Orange Book Patent ID)
Patent 6376515
Expiration Sep 15, 2020
Applicant PORTOLA PHARMS INC
Drug Application
  1. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
  2. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
FDA Orange Book Patents: 2 of 10 (FDA Orange Book Patent ID)
Patent 6835739
Expiration Sep 15, 2020
Applicant PORTOLA PHARMS INC
Drug Application
  1. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
  2. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
FDA Orange Book Patents: 3 of 10 (FDA Orange Book Patent ID)
Patent 9555023
Expiration Nov 7, 2026
Applicant PORTOLA PHARMS INC
Drug Application
  1. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
  2. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
FDA Orange Book Patents: 4 of 10 (FDA Orange Book Patent ID)
Patent 9629831
Expiration Sep 15, 2020
Applicant PORTOLA PHARMS INC
Drug Application
  1. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
  2. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
FDA Orange Book Patents: 5 of 10 (FDA Orange Book Patent ID)
Patent 7598276
Expiration Nov 8, 2026
Applicant PORTOLA PHARMS INC
Drug Application
  1. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
  2. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
FDA Orange Book Patents: 6 of 10 (FDA Orange Book Patent ID)
Patent 8404724
Expiration Mar 29, 2031
Applicant PORTOLA PHARMS INC
Drug Application
  1. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
  2. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
FDA Orange Book Patents: 7 of 10 (FDA Orange Book Patent ID)
Patent 8518977
Expiration Sep 15, 2020
Applicant PORTOLA PHARMS INC
Drug Application
  1. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
  2. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
FDA Orange Book Patents: 8 of 10 (FDA Orange Book Patent ID)
Patent 8557852
Expiration Sep 8, 2028
Applicant PORTOLA PHARMS INC
Drug Application
  1. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
  2. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
FDA Orange Book Patents: 9 of 10 (FDA Orange Book Patent ID)
Patent 8691847
Expiration Sep 15, 2020
Applicant PORTOLA PHARMS INC
Drug Application
  1. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
  2. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
FDA Orange Book Patents: 10 of 10 (FDA Orange Book Patent ID)
Patent 8987463
Expiration Dec 28, 2030
Applicant PORTOLA PHARMS INC
Drug Application
  1. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)
  2. N208383 (Prescription Drug: BEVYXXA. Ingredients: BETRIXABAN)

////////

PHASE 3  for Venous Thromboembolism (VTE)

Patents CN1391555A, CN102336702A, CN101595092A, CN102762538A

Portola Pharmaceuticals, under license from Takeda (formerly known as Millennium Pharmaceuticals), is developing betrixaban (was reported to be in phase III in November 2015), for treating venous thrombosis

In October 2015, betrixaban was granted Fast Track designation by the FDA for extended-duration prevention of VTE or blood clots in acute medically ill patients

Betrixaban (INN, codenamed PRT-054,021) is an anticoagulant drug which acts as a direct factor Xa inhibitor.[1] It is potent, orally active and highly selective for factor Xa, being selected from a group of similar compounds for its low hERG affinity.[2] Betrixaban has undergone human clinical trials for prevention of embolism after knee surgery,[3] and prevention of stroke following atrial fibrillation,[4] with promising results.[5] Betrixaban is currently being studied in a human clinical trial for extended duration thromboprophylaxis to prevent venous thromboembolism in acute medically ill patients.[6] Joint development with Portola was discontinued in 2011 by Merck.[7] Betrixaban is now being developed by Portola Pharmaceuticals.

Long-acting, oral, direct Factor Xa Inhibitor

Description

Betrixaban is an oral small molecule anticoagulant that directly inhibits the activity of Factor Xa, an important validated target in the blood coagulation pathway.

Key Characteristics

Betrixaban has been specifically designed for chronic, once-a-day treatment. It has a half-life that supports true, once-daily dosing and a low peak-to-trough drug concentration ratio that minimizes anticoagulant variability. Betrixaban is primarily eliminated unchanged in the bile and has been studied in patients with all degrees of renal function, including those with severe renal impairment (excluding dialysis patients). Betrixaban is minimally metabolized through the Cytochrome 450 enzyme system, which may result in low potential for CYP-related drug interactions. Betrixaban is reversible with PRT4445, a universal Factor Xa inhibitor antidote that Portola is developing as a companion product.

Potential Indications

Treatment or prevention of life-threatening blood clots (venous thromboembolism; VTE) in acute medically ill patients.

Clinical Development

ClinicalTrials.gov Identifier:
NCT01583218
COMPLETION-August 2014

http://clinicaltrials.gov/ct2/show/NCT01583218

APEX Study

Portola has initiated its pivotal Phase 3 APEX Study to demonstrate the safety and efficacy of betrixaban for extended duration venous thromboembolism (VTE) prophylaxis for up to 35 days in acute medically ill patients with restricted mobility and certain risk factors. This randomized, double-blind, active-controlled, multicenter, multinational study will compare a once-daily dose of 80 mg of betrixaban for a total of 35 days (including both in the hospital and after discharge) with in-hospital administration of 40 mg of enoxaparin once daily for 6 to 14 days followed by placebo. The global study is expected to enroll approximately 6,850 patients at more than 400 study sites throughout the world. The primary objective of the trial is to demonstrate the superiority of betrixaban as compared to the current standard of care in the reduction of VTE-related events at 35 days while maintaining a favorable benefit to risk profile.

The APEX study is adequately powered to show a clinically relevant benefit with a p-value of less than 0.01 on the primary endpoint of total asymptomatic proximal DVT (as detected by ultrasound), symptomatic DVT (proximal or distal), non-fatal pulmonary embolism and VTE-related death. The first patient was enrolled in March 2012.

The safety and tolerability of betrixaban for stroke prevention was evaluated in 508 patients with atrial fibrillation in the Phase 2 EXPLORE-Xa dose-ranging study. Results were presented during a late-breaking session at the American College of Cardiology’s 59th Annual Scientific Session in March 2010. The data showed that a once-daily dose of oral betrixaban, given to patients with non-valvular atrial fibrillation or atrial flutter and at least one risk factor for stroke, reduced the incidence of major and clinically relevant non-major bleeds compared to dose-adjusted warfarin. In August 2010, additional pharmacodynamic data from a pre-specified analysis of EXPLORE-Xa showed a concentration dependent relationship and provided further evidence for the anticoagulant activity of betrixaban across all three doses studied in the clinical trial. The additional pharmacodynamic analysis provides information for dose selection for Phase 3 evaluation of betrixaban.

In 2007, positive top-line results from EXPERT were published in The Journal of Thrombosis and Haemostasis. This randomized, multi-center, Phase 2 in-hospital efficacy and safety study of the prevention of VTE compared betrixaban with enoxaparin in 215 patients undergoing knee replacement surgery.

Portola Pharmaceuticals

Betrixaban (INN, codenamed PRT-054,021) is an anticoagulant drug which acts as a direct factor Xa inhibitor.[1] It is potent, orally active and highly selective for factor Xa, being selected from a group of similar compounds for its low hERG affinity.[2] Betrixaban has undergone human clinical trials for prevention of embolism after knee surgery,[3] and prevention of stroke following atrial fibrillation,[4] with promising results.[5]

b1

b2

 

Patent Document CN1391555A first discloses a preparation method (see Scheme 1):

Figure CN104693114AD00042

 

CN101595092A  (See Scheme 2).

Figure CN104693114AD00051

 

Patent Document CN102762538A  (see Scheme 3).

[0013]

Figure CN104693114AD00061

 

 

CN104693114

Machine translated from chinese please bear with names

http://www.google.com/patents/CN104693114A?cl=en

Figure CN104693114AD00071

 

Preparation Example 1 shell song in Spanish

Figure CN104693114AD00111

  Under stirring, temperature 15 ~ 20 ° C, was added dropwise 2mol / L tetrahydrofuran solution of isopropylmagnesium chloride (available commercially available) 308ml (0 • 615mol, 5eq) to 2mol / L dimethylamine THF Solution (commercially available can) 339ml (0.677mol, 5. 5eq) to give dimethylamine reaction solution.

  Under stirring, temperature 15 ~ 20 ° C, the compound of formula II 50. 0g (0 123mol, leq.) Was mixed with 500ml of tetrahydrofuran, was added dropwise the above-described reaction solution of dimethylamine; After the addition continued at 25 The reaction was stirred for ~ 30 ° C, the progress of the reaction was monitored by HPLC. After completion of the reaction, at 15 ~ 20 ° C, the reaction solution was added to about 2mol L hydrochloric acid solution 700ml / in hydrochloric acid and then adjusting the pH to 2-3; concentrated under reduced pressure and the organic solvent was evaporated, filtered and concentrated liquid The precipitated solid, the filter cake washed with an appropriate amount of water; the filter cake was mixed by stirring with 500ml of acetone, the pH adjusted with triethylamine to 7-8; filtered; the cake at 40 ~ 45 ° C and dried under reduced pressure to give Pui Spanish song 45. 5g. . Yield: 82 0%; HPLC purity: 98.9%, of which 0.05% dechlorinated impurities VIII, IX desmethyl impurities were not detected.

Take the above Tony Qu Spanish 45.0g, at about 70 ° C under stirring dissolved in N, N- dimethylacetamide 180ml, a toluene solution of 360ml; cooling crystallization, filtration, the filter cake washed with an appropriate amount of acetone at 40 ~ 45 ° C and dried under reduced pressure; the resulting song Tony Spanish HPLC purity 99.7%.

(+) LC-MS: m / z = 452 ([M + H] +). Che NMR (400MHz, DMS0-d6) S:…. 2 96 (s, 6H), 3 83 (s, 3H), 7. 06-7 09 (dd, 1H), 7. 55-7 59 ( m, 3H), 7. 80-7. 83 (dd, 1H), 8. 21-8. 23 (d, 1H), 8. 27-8. 30 (d, 2H), 8. 37-8. 40 (d, 1H), 8. 41-8. 43 (d, 1H), 10. 54 (br., 2H).

Preparation Example 2 Tony Qu Spanish maleate

  Under stirring, temperature 0 ~ 5 ° C, dropping 2mol isopropyl magnesium chloride in tetrahydrofuran / L (available commercial available) 105ml (0 • 21mol, 8. 4eq) twenty methylamine hydrochloride 8. 91g (0 • llmol, 4. 4eq) in tetrahydrofuran 60ml of the suspension, the reaction solution obtained dimethylamine.

  Under stirring, temperature 0 ~ 5 ° C, the compound of formula II 10. 0g (0 025mol, leq.) Was mixed with 100ml of tetrahydrofuran, and then dropping the above reaction liquid dimethylamine; After the addition continued 10 The reaction was stirred for ~ 15 ° c, the progress of the reaction was monitored by HPLC. After completion of the reaction, at 10 ~ 15 ° C, the reaction solution was added to an aqueous solution of 45g and 100ml dubbed maleic acid solution; the organic solvent was evaporated under reduced pressure and concentrated, filtered concentrate precipitated solid cake was washed with the right amount of water washing. Cake at 40 ~ 45 ° C and dried under reduced pressure to give Tony Qu Spanish maleate 12.lg. . Yield: 85 4%; HPLC purity: 98.6%, which is 0.03% dechlorinated impurities VIII, IX desmethyl impurities were not detected.

Take the above shellfish Spanish song maleate 10. 0g, at about 70 ° C under stirring dissolved in a mixed solvent of ethanol 50ml and 25ml of water, dropping water 150ml; cooling crystallization, filtration, the filter cake at 40 ~ 45 ° C and dried under reduced pressure; the resulting song Tony Spanish maleate HPLC purity 99.9%.

: HNMR (400MHz, DMS〇-d6) 8: 3. 25 (s, 3H), 3. 32 (s, 3H), 3. 87 (s, 3H), 6. 02 (s, 2H) , 7. 19-7. 21 (dd, 1H), 7. 44-7. 45 (1H), 7. 75-7. 77 (d, 2H), 7. 97-9. 98 (d, 2H) , 8. 08-8. 13 (m, 3H), 8. 44-8. 45 (d, 1H), 9. 01 (br., 1H), 9. 37 (br., 1H), 11.04 (s , 1H), 11. 13 (s, 1H).

Preparation Example 3 Tony Spanish song of [0075] Example

  Under stirring, temperature 25 ~ 30 ° C, isopropylmagnesium chloride in tetrahydrofuran was added dropwise a solution of 2mol / L (available commercially available) 81ml (0 • 161mol, 7eq) to 2mol / L dimethylamine THF Solution (commercially available can) 121ml (0 • 242mol, 10. 5eq) to give dimethylamine reaction solution.

Under stirring, temperature 25 ~ 30 ° C, the hydrochloride salt of a compound of formula II 10. 0g (0 023mol, leq.) Was mixed with 100ml of tetrahydrofuran, was added dropwise the above-described reaction solution of dimethylamine; After the addition was complete The reaction continued stirring at 25 ~ 30 ° C, the progress of the reaction was monitored by HPLC. After completion of the reaction, at 15 ~ 20 ° C, the reaction solution was added to about 2mol L hydrochloric acid solution 210ml / in hydrochloric acid and then adjusting the pH to 2-3; concentrated under reduced pressure and the organic solvent was evaporated, filtered and concentrated liquid The precipitated solid, the filter cake washed with an appropriate amount of water; the filter cake with 90ml acetone was stirred and mixed, the pH adjusted with triethylamine to 7-8; filtration; cake was 45 ~ 50 ° C and dried under reduced pressure to give Pui Qu Spanish 8. 35g. Yield: 80.5%. HPLC purity: 98.7%, which is 0.03% dechlorinated impurities VIII, IX desmethyl impurities were not detected.

Preparation Example 4 shellfish Spanish song hydrochloride

  Under stirring, temperature 15 ~ 20 ° C, dropping lmol / n-amyl magnesium bromide tetrahydrofuran solution (which can be commercialized available) 75ml (0 • 075mol, 3eq) to 2mol / L of dimethyl L amine in tetrahydrofuran (commercially available can) 56ml (0 • 113mol, 4. 5eq) to give dimethylamine reaction solution.

Under stirring, temperature 15 ~ 20 ° C, the compound of formula II 10. 0g (0 025mol, leq.) Was mixed with 100ml of tetrahydrofuran, was added dropwise the above-described reaction solution of dimethylamine; After the addition continued at 25 The reaction was stirred for ~ 30 ° C, the progress of the reaction was monitored by HPLC. After completion of the reaction, at 15 ~ 20 ° C, the reaction solution was added to about 2mol L hydrochloric acid solution 100ml / in hydrochloric acid and then adjusting the pH to 2-3; concentrated under reduced pressure and the organic solvent was evaporated, filtered and concentrated liquid The precipitated solid, the filter cake washed with an appropriate amount of water. Cake at 40 ~ 45 ° C and dried under reduced pressure to give Tony Qu Spanish hydrochloride 10.lg, yield:. 82 9%; HPLC purity: 99.0%, which is 0.02% dechlorination impurity VIII, from A impurities IX was not detected.

  Take the above shellfish Spanish song hydrochloride 10. 0g, at about 70 ° C under stirring dissolved in N, N- dimethylacetamide 40ml, a toluene solution of 80ml; cooling crystallization, filtration, cake at 40 ~ 45 ° C and dried under reduced pressure; the resulting song Tony Spanish hydrochloride HPLC purity 99.8%.

Preparation 5 shellfish Spanish song of [0082] Example

Under stirring, temperature 0 ~ 5 ° C, dropping lmol / diethyl zinc toluene solution of L (available commercially oriented) 50ml (0. 050mol, 2eq) to 2mol / L dimethylamine tetrahydrofuran (commercially available can) 28ml (0. 055mol, 2. 2eq) to give dimethylamine reaction solution.

  Under stirring, temperature 0 ~ 5 ° C, the compound of formula II 10. 0g (0 025mol, leq.) Was mixed with 100ml of tetrahydrofuran, and then dropping the above reaction liquid dimethylamine; After dropping 5 continues The reaction was stirred for ~ 10 ° C, the progress of the reaction was monitored by HPLC. After completion of the reaction, in the next 5 ~ 10 ° C, the reaction mixture was added to about 2mol L dilute hydrochloric acid solution 70ml / in hydrochloric acid and then adjusting the pH to 2-3; concentrated under reduced pressure and the organic solvent was evaporated, filtered and concentrated liquid The precipitated solid, the filter cake was washed successively with a suitable amount of water; the filter cake with acetone l〇〇ml mixing, the pH adjusted with triethylamine to 7-8; filtered; the cake at 40 ~ 45 ° C under reduced pressed and dried to give Tony Qu Spanish 9. 03g. . Yield: 80 1%; HPLC purity: 99.0%, which is 0.02% dechlorinated impurities VIII, IX desmethyl impurities were not detected.

  Preparation of compounds of Formula II Preparation Example 1

Methoxy-2-nitro – (5-chloro-pyridin-2-yl) -5 – benzamide (compound V) Preparation of [0086] (1) N-

Figure CN104693114AD00131

  with stirring at room temperature, 5-methoxy-2-nitrobenzoic acid (Compound VI, can be commercially available) 250g (1. 27mol, leq) and 2-amino-5-chloropyridine (Compound VII .) 163g (l 27mol, leq) was suspended in 1700ml of acetonitrile, pyridine 301g (3 81mol, 3eq), and then phosphorus oxychloride was added dropwise 231g (l 52mol, 1 2eq);… After stirring for 1 hour the reaction 3500ml water quenching crystallization; the filter cake was washed with water 1700mlX2; dried under reduced pressure to obtain compound V349g.

  (2) 2-Amino -N- (5- chloro – pyridin-2-yl) -5-methoxy – benzamide (compound IV) is prepared

Figure CN104693114AD00132

  with stirring at room temperature, the N- (5- chloro – pyridin-2-yl) -5-methoxy-2-nitro – benzamide (Compound V) 300g (0 • 977mol, 1.Oeq) 3000ml was dissolved in acetic acid, and iron powder was added portionwise 546g (9 77mol, 10eq.); After the addition of iron stirring was continued for 3 hours, and then ethyl acetate and water 6000ml 3000ml, liquid separation; the aqueous phase was separated 3000mlX2 extracted with ethyl acetate; combined organic phases were washed with water, saturated aqueous sodium bicarbonate, saturated sodium chloride solution, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give compound IV244g.

(3) N- (5- chloro – pyridin-2-yl) -2- (4-cyano – benzoyl – amino) -5-methoxy – benzamide (compound II) is prepared

Figure CN104693114AD00141

at 10 ~ 20 ° C, a solution of a compound of formula IV 200g (0 • 72mol, 1.Oeq) and triethylamine 109g (1. 08mol, 1. 5eq) 2000ml dissolved in tetrahydrofuran, to which was added dropwise to cyano benzoyl chloride (compound III, commercially available technology) 130g (0 79mol, 1.leq.) and tetrahydrofuran solution dubbed 1000ml, HPLC monitoring progress of the reaction; after the reaction was filtered, the filter cake washed with an appropriate amount of ethanol, dried under reduced pressure to obtain compound II263g. HPLC purity: 98.7%.

  (+) LC-MS: m / z = 407 ([M + H] +). Insect NMR (400MHz, DMS0-d6) S:… 3 85 (s, 3H), 7 16-7 .19 (dd, 1H), 7. 39-7 41 (d, 1H), 7. 93- 7. 96 (d, 2H), 8. 02-8. 04 (m, 4H), 8. 13-8. 14 (d, 2H), 8. 42-8. 43 (d, 1H), 11. 06 (br. 2H).

Example 2 Preparation of the hydrochloride salt of the compound of formula II

  at 10 ~ 20 ° C, a solution of a compound of formula IV 40. 0g (0 • 14mol, 1.Oeq) was dissolved in 400ml of tetrahydrofuran, a solution of cyanobenzoyl chloride (Compound III, can be commercialized available) 24 8g (0 15mol, 1.leq) and tetrahydrofuran solution 200ml dubbed, HPLC monitoring progress of the reaction;.. After the reaction was filtered, the filter cake washed with ethanol and after an appropriate amount, and dried under reduced pressure to obtain a compound of formula II hydrochloride . HPLC purity: 99.5%.

 

 WO 2015176591

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2015176591&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=FullText

Example 1: Preparation of Spanish Preparation and Form A half-L- malic acid shellfish song

At 55 ~ 60 ℃, the shellfish song Spanish 6.0g (13.3mmol), L- malic acid 1.1g (8.0mmol) was dissolved in tetrahydrofuran 70mL / water 7mL mixed solvent acetone was added with stirring 60mL, cooled to room temperature, Crystallization. Precipitated solid was filtered, and the resulting solid at 40 ~ 45 ℃ vacuum dried to give half L- malic acid shellfish Spanish song.

1H NMR(400MHz,MeOD)δ:2.355-2.419(dd,0.5H),2.735-2.781(dd,0.5H),3.226(s,6H),3.907(s,3H),4.302-4.326(dd,0.5H),7.195-7.224(dd,1H),7.448-7.455(d,1H),7.744-7.764(d,2H),7.821-7.849(dd,1H),8.145-8.165(d,2H),8.196-8.219(d,1H),8.238-8.261(d,1H),8.323-8.329(d,1H)。

Above 1 H-NMR results, δ: 3.907 (s, 3H) attributed to shellfish Spanish song molecule methyl CH 3 , 4.302-4.326 (dd, 0.5H) attributed to L- malic acid molecule methine CH , you can determine the song title product in shellfish Spanish and L- malic acid molar ratio of 2: 1.

PATENT

http://www.google.com.na/patents/EP2101760A2?cl=en

Example 2

Preparation of the compound of Formula II

a. Gram scale preparation A slurry of the compound of Formula F (455 g, 1.0 eq.) in THF (4.67 kg,

10.3 parts) was prepared and adjusted to <10 0C. Lithium dimethyl amide was prepared as follows :hexyllithium (2.3 N/hexane, 2.45 L, 5.5 eq.) was added to dimethylamine solution (2 N/THF, 2.8 L, 5.5 eq.) maintaining <10 0C. The lithium dimethyl amide solution was charged into the slurry containing the compound of Formula F keeping the pot temperature of <10 0C. The reaction progress was monitored by in-process HPLC which confirmed that the amount of Formula F was <1.0 A%. A buffer solution of NaHCO3 (490 g, 1.1 parts, 5.7 eq.) and Na2CO3 (490 g, 1.1 parts, 4.5 eq.) in deionized water (6.6 kg, 14.51 parts) was prepared, and the above reaction mixture was transferred to this aqueous solution maintaining < 5 0C. The product precipitated out and the resulting slurry was adjusted to 20 0C over a period of 12 hr. The solid was filtered, and the resulting wet cake was washed with 3.5 kg (7.7 parts) of deionized water. The solid was filtered off using a coarse frit glass bench filter, and rinsed forwarded with cold (0-5 0C) absolute ethanol (628 g, 1.4 parts). The product was dried at 30-35 0C. Dry product was obtained in 458 g (73% yield). b. Kilogram scale preparation A slurry of the compound of Formula F (31.5 kg, 1.0 eq.) in THF (251 kg,

8.0 parts) was prepared in a 780 L Hastelloy reactor (Reactor A) and adjusted to 0 0C (-3 to 3 0C). 2 M Dimethylamine in THF (161.0 kg, 5.0 eq.) and THF (63 kg, 2 parts) were charged into a 1900 L GLMS reactor (Reactor B) and adjusted to 0 0C (-3 to 3 0C) with maximum agitation. Hexyllithium (2.3 M, 97.2 kg, 4.5 eq.) was slowly charged to Reactor B while maintaining a max temperature of 10 0C. The pump and lines were rinsed forward to Reactor B with THF (3.2 kg). The Reactor B contents were adjusted to 0 0C (-3 to 3 0C), then transferred to Reactor A while keeping Reactor A temperature < 10 0C. The Reactor B pump and lines were rinsed forward with THF (31.4 kg, 1.0 part). The Reactor A contents were adjusted to 0 0C (-3 to 3 0C), and agitated at this temperature until the reaction was complete as verified by HPLC (1-2 hrs). After about 1 hr of agitation, in-process HPLC analysis indicated that 0 A% starting material remained (in-process criteria: max 1 A%). Reactor A contents were adjusted to -5 0C (-8 to -3 0C). In-process cleaning of Reactor B with water was performed. Two previously prepared aqueous solutions (NaHCO3 (35.0 kg, 1.1 parts) in water (236 kg, 7.5 parts), and Na2CO3 (35.0 kg 1.1 parts) in water (236 kg, 7.5 parts))were charged to Reactor B and adjusted to -3 0C (0 to 6 0C). Reactor A contents were transferred to Reactor B through an insulated line, maintaining the temperature of Reactor B at -8 0C to a maximum of 5 0C. The Reactor A pump and lines were rinsed forward with cold [-5 0C (-8 to -3 0C)] THF (31.4 kg, 1.0 part). Reactor B contents were adjusted to 22 0C (19-25 0C) and agitated for ca. 3 hrs. Slurry formation was visually confirmed, and Reactor B contents were filtered onto a 30″ centrifuge fitted with a filter cloth. The Reactor B pump and lines were rinsed forward onto the 30″ centrifuge fitted with a filter cloth with drinking water (63 kg, 2 parts). The wet filter cake (66.5 kg) was transferred back to Reactor B and submitted to a slurry wash in drinking water (1005 kg, 32 parts) at 22 0C (19-25) 0C for ca. 1 hr. The product was filtered onto the 30″ centrifuge (after in-process cleaning and fitting with a filter cloth), and the Reactor B lines and pump were rinsed forward with drinking water (63 kg, 2 parts). The water rinse was sampled for test by TDS, which was found to be 0.46%. The Reactor B pump, lines and wet filter cake were further rinsed with cold [0 0C (-3 to 3 0C)] ethanol (44 kg, 1.39 parts). The wet filter cake was dried under vacuum with a maximum temperature of water bath (to heat dryer jacket) of 35 0C. In-process LOD was 0% after ca. 24 hrs of drying, and the product was discharged (24.8 kg) in 76.7% yield. HPLC showed 98 % purity, with dechlorinated impurity at 1.14 %. Example 3

Preparation of the compound of Formula F Step 1. Synthesis of 2-nitro-N-(5-chloro-pyridin-2-yl)-5-methoxy-benzamide (C)

5-Methoxy-2-nitrobenzoic acid (A) (25.0 kg, 1.0 eq.), 2-amino-5- chloropyridine (B) (16.3 kg, 1.0 eq.), and acetonitrile (87.5 kg, 3.5 parts) were charged to a 380 L GLMS reactor. The reaction mixture was adjusted to 22 0C (19-25 0C) and anhydrous pyridine (30.0 kg, 3.0 eq.) was added. The pump and lines were rinsed forward with acetonitrile (22.5 kg, 0.9 parts), and the reactor contents were adjusted to a temperature of 19-22 0C. Phosphorous oxychloride (23.3 kg, 1.20 eq.) was charged to the contents of the reactor via a metering pump, while maintaining a temperature of 25 0C (22-28 0C). The metering pump and lines were rinsed forward with acetonitrile (12.5 kg, 0.5 parts), while keeping the temperature at 25 0C (22-28 0C). The reaction mixture normally turned from a slurry to a clear solution after the addition of about 1/3 of the POCI3. At the end of the addition, it became turbid. After complete addition, the reaction mixture was agitated at 25 0C (22-28 0C) for ca. 1 hr, at which time HPLC analysis confirmed reaction completion. The solution was cooled to 15 0C (12-18 0C) and drinking water (156.3 kg, 6.25 parts) was charged slowly while keeping reaction temperature of between 12 and 30 0C. The reaction mixture was then adjusted to 22 0C (19-25 0C) and agitated for ca. 5 hrs until exotherm ceased. Formation of a slurry was visually confirmed and the contents of the reactor were filtered onto a pressure nutsche fitted with a filter cloth. The reactor, pump, and lines were washed forward onto the pressure nutsche with two portions of drinking water (62.5 kg, 2.5 parts each). The filtrate had a pH value of 7. The product (41.8 kg) was dried under vacuum with a maximum temperature of water bath (to heat dryer jacket) of 50 0C. After ca. 12 hrs, in-process LOD analysis indicated a solvent content of 0.72%. The dry product (C) was discharged (34.4 kg) with 88.2% yield and 99.1 % purity by HPLC. Step 2. Synthesis of 2-amino-N-(5-chloro-pyridin-2-yl)-5-methoxy-benzamide (D)

To a 780 L Hastelloy reactor, compound C (33 kg, 1.0 eq.), 5% platinum carbon (sulfided, 0.33 kg, 0.010 parts) and dichloromethane (578 kg, 17.5 parts) were charged. Agitation was started and reactor contents were adjusted to 22 0C (19-25 0C). The reactor was pressurized with ca. 30 psi hydrogen and the reaction mixture gently heated to 28 0C (25-31 0C). Hydrogenation of the reactor contents was performed under ca. 30 psi at 28 0C (25 to 31 0C; maximum 31 0C) until the reaction was complete by HPLC. After 16.5 hrs, the reaction was deemed complete after confirming the disappearance of starting material (0.472 A%). The contents of the reactor were circulated through a conditioned celite pad (0.2-0.5 kg celite conditioned with 20-55 kg dichloromethane) prepared in a 8″ sparkler filter to remove the platinum catalyst. The reactor and celite bed were rinsed forward with two portions of dichloromethane (83 kg, 2.5 parts each). The filtrate was transferred to and concentrated in a 570 L GLMS reactor under a atmospheric pressure to ca. 132 L (4 parts volume). Ethanol (69 kg, 2.1 parts) was charged and concentration continued under atmospheric pressure to ca. 99 L (3 parts volume). In-process NMR indicated that the dichloromethane content was 39%. Ethanol (69 kg, 2.1 parts) was charged again and concentration continued again to ca. 99 L (3 parts volume). In-process NMR indicated that the dichloromethane content was 5%. The reaction mixture was then adjusted to 3 0C (0 to 6 0C), agitated for ca. 1 hr, and the resulting slurry filtered onto a jacketed pressure nutsche fitted with a filter cloth. The reactor, pump, and lines were rinsed forward with cold [3 0C (0-6 0C)] ethanol (26 kg, 0.8 parts). The wet filter cake (36.6 kg) was dried under vacuum at 40-50 0C with a maximum temperature of water bath (to heat dryer jacket) of 50 0C. LOD analysis after 12.5 hrs indicated solvent content was at 0.1%. The dry product (D) was discharged (26.4 kg) in 89.5% yield. HPLC showed 98.4 A% purity, with dechlorinated impurity at 0.083 %. Step 3. Synthesis of N-(5-chloro-pyridin-2-yl)-2-(4-cyano-benzoyl-amino)-5-methoxy- benzamide Hydrochloride (F)

To a 780 L Hastelloy reactor, was charged 4-cyanobenzoyl chloride (E)

(17.2 kg, 1.1 eq.) and THF (92 kg, 3.5 parts). Reactor contents were agitated at 22 0C (19- 25 0C) until all of the solids had dissolved. The resulting solution was transferred to a lower receiver and the reactor was rinsed forward with THF (26 kg, 1 part). Compound D (26.4 kg, 1 eq.), THF (396 kg, 15 parts) and pyridine (2.90 kg, 0.4 eq.) were charged to a clean reactor. The pump and lines were rinsed forward with THF (34 kg, 1.3 parts). Via a metering pump, the 4-cyanobenzoyl chloride/THF solution was charged to the reactor, keeping the temperature at < 30 0C and rinsing forward with THF (ca. 10 kg). The resulting yellow-colored slurry was agitated at 22 0C (19-25 0C) for ca 2 hrs. In-process HPLC taken after 2 hrs showed a compound of Formula D content of 0%, indicating completion of the reaction. The slurry was filtered onto a pressure nutsche fitted with a filter cloth. The reactor, pump, lines and wet cake were rinsed with three portions of ethanol (ca. 15 kg each). The wet filter cake was discharged (65.4 kg) and transferred back to the reactor for slurry wash in ethanol (317 kg, 12 parts) at 22 0C (19-25 0C) for ca. 1 hr. The slurry was filtered onto the pressure nutsche and the reactor, pump, lines, and wet filter cake were rinsed with two portions of ethanol (ca. 15 kg each) and two portions of THF (ca. 15 kg each). The wet filter cake was dried under vacuum with a maximum temperature of warm glycol bath (to heat the dryer jacket) of 40 0C. After 14.5 hrs of drying, LOD was 0.75%. The dried material was milled (screen 0.125″) to give 31.8 kg of product, which was dried under vacuum for another 10.5 hrs. LOD after drying was 1.8%, and the product was discharged (31.5 kg) in 74.8% yield (expected 60-90%). HPLC showed 100 % purity.

PATENT

http://www.google.com/patents/WO2011084519A1?cl=en

U.S. Patent No. 6,376,515 B2 discloses a class of benzamide based compounds as specific factor Xa inhibitors. In particular, U.S. Patent No. 6,376,515 B2 describes a compound identified as Example 206, which is also disclosed in U.S. Patent No. 6,835,739 B2 as Example 206 and herein identified as betrixaban, which has the chemical formula of Formula I:

 

 

 

Scheme 1

Example 1: Preparation of betrixaban

[0113] Dimethylformamide (13L) and hydrochloride (18 mL) were charged into a reactor. Compound B (1 kg) was added followed by Compound A (0.88 kg).

Compound A is commercially available or, just as with Compound B may be prepared using the methods described in Examples 4 and 5. The reaction mixture was cooled between 0 °C and -10 °C. EDC (0.752 kg) was added while maintaining the temperature between -10 °C and 0 °C. The reaction mixture was stirred until the content of

Compound B is below 0.10% area by HPLC. The reaction mixture was stirred until betrixaban started to crystallize. Acetone (26 L) was then added during a period of at least 1 hr while the temperature was maintained at between -10 °C and 0 °C. The suspension was then stirred for additional 2 hrs at a temperature of between 0 °C and 10 °C. The suspension was filtered and washed with cold acetone to give a wet product betrixaban. Example 2: Preparation of a maleate salt of betrixaban

[0114] The wet betrixaban obtained above was reacted with maleic acid (0.52 x weight of maleic acid/weight of dry betrixaban) in ethanol (22.4 x volume of

liquid/weight of dry betrixaban (v/w)) and purified water (5.7 x v/w) to form a betrixaban maleate salt. The solution of the betrixaban maleate salt was filtered and concentrated under vacuum until a final volume of 5.7 x v/w. Water (2 x v/w) was then added and the mixture was back concentrated until the same volume. The procedure of adding water and distil until a final volume of 5.7 x v/w was carried out until the molar ratio between the content of ethanol and the content of betrixaban maleate salt in the mixture was lower than, or equal to, 6. Betrixaban maleate salt crystallized during the removal of ethanol. The suspension was cooled to a temperature between 19 °C and 25 °C and stirred for not less than 2 hours at this temperature range. Betrixaban maleate salt was isolated by filtration, washed with water and dried under vacuum at a maximum temperature of 40 °C until the content of water was lower than, or equal to, 0.5 % w/w by Karl-Fisher. The purity of the maleate salt was determined to be greater than 99 % by HPLC. The betrixaban maleate isolated was in a crystalline form A which was concluded based on IR, DSC and XRPD results obtained, see Figures 3-5, respectively. The major peaks of XRPD pattern of crystalline form A are also listed in Table 2. Table 2: Betrixaban Form A XRPD Peak °2-Theta (2Θ0)

Example 3: Synthesis of 2-nitro-N-(5-chloro-pyridin-2-yl)-5-methoxy-benzamide (C)

D E C

[0115] 5-Methoxy-2-nitrobenzoic acid (D) (25.0 kg, 1.0 eq.), 2-amino-5- chloropyridine (E) (16.3 kg, 1.0 eq.), and acetonitrile (87.5 kg) were charged to a 380 L glass-lined reactor. The reaction mixture was adjusted to 22 °C (19-25 °C) and anhydrous pyridine (30.0 kg, 3.0 eq.) was added. The pump and lines were rinsed forward with acetonitrile (22.5 kg), and the reactor contents were adjusted to a temperature of 19-22 °C. Phosphorous oxychloride (23.3 kg, 1.20 eq.) was charged to the contents of the reactor via a metering pump, while maintaining a temperature of 25 °C (22-28 °C). The metering pump and lines were rinsed forward with acetonitrile (12.5 kg), while keeping the temperature at 25 °C (22-28 °C). The reaction mixture normally turned from a slurry to a clear solution after the addition of about 1/3 of the POCI3. At the end of the addition, it became turbid. After complete addition, the reaction mixture was agitated at 25 °C (22- 28 °C) for ca. 1 hr, at which time HPLC analysis confirmed reaction completion. The solution was cooled to 15 °C (12-18 °C) and water (156.3 kg) was charged slowly while keeping reaction temperature of between 12 and 30 °C. The reaction mixture was then adjusted to 22 °C (19-25 °C) and agitated for ca. 5 hrs until exotherm ceased. Formation of a slurry was visually confirmed and the contents of the reactor were filtered onto a pressure nutsche fitted with a filter cloth. The reactor, pump, and lines were washed forward onto the pressure nutsche with two portions of water (62.5 kg). The filtrate had a pH value of 7. The product (41.8 kg) was dried under vacuum with a maximum temperature of water bath (to heat dryer jacket) of 50 °C. After ca. 12 hrs, in-process LOD analysis indicated a solvent content of 0.72%. The dry product (C) was discharged (34.4 kg) with 88.2% yield and 99.1 % purity by HPLC.

Exam le 4. Synthesis of 2-amino-N-(5-chloro-pyridin-2-yl)-5-methoxy-benzamide

Process A

[0116] To a 780 L Hastelloy reactor, Compound C (33 kg, 1.0 eq.), 5%> platinum carbon (sulfided, 0.33 kg) and dichloromethane (578 kg) were charged. Agitation was started and reactor contents were adjusted to 22 °C (19-25 °C). The reactor was pressurized with ca. 30 psi hydrogen and the reaction mixture gently heated to 28 °C (25-31 °C). Hydrogenation of the reactor contents was performed under ca. 30 psi at 28 °C (25 to 31 °C; maximum 31 °C) until the reaction was complete by HPLC. After 16.5 hrs, the reaction was deemed complete after confirming the disappearance of starting material (0.472 A%). The contents of the reactor were circulated through a conditioned Celite™ (diatomaceous earth; Celite Co., Santa Barbara, Ca.) pad (0.2-0.5 kg Celite™ conditioned with 20-55 kg dichloromethane) prepared in a 8″ sparkler filter to remove the platinum catalyst. The reactor and Celite™ bed were rinsed forward with two portions of dichloromethane (83 kg). The filtrate was transferred to and concentrated in a 570 L glass-lined reactor under an atmospheric pressure to ca. 132 L. Ethanol (69 kg) was charged and concentration continued under atmospheric pressure to ca. 99 L. In-process NMR indicated that the dichloromethane content was 39%. Ethanol (69 kg) was charged again and concentration continued again to ca. 99 L. In-process NMR indicated that the dichloromethane content was 5%. The reaction mixture was then adjusted to 3 °C (0 to 6 °C), agitated for ca. 1 hr, and the resulting slurry filtered onto a jacketed pressure nutsche fitted with a filter cloth. The reactor, pump, and lines were rinsed forward with cold [3 °C (0-6 °C)] ethanol (26 kg. The wet filter cake (36.6 kg) was dried under vacuum at 40-50 °C with a maximum temperature of water bath (to heat dryer jacket) of 50 °C. LOD analysis after 12.5 hrs indicated solvent content was at 0.1%. The dry product (B) was discharged (26.4 kg) in 89.5% yield. HPLC showed 98.4 A% purity, with dechlorinated impurity at 0.083 %.

Process B

[0117] To a 780 L Hastelloy reactor, Compound C (33 kg, 1.0 eq.), 5%> platinum carbon (sulfided, 0.33 kg) and dichloromethane (578 kg) were charged. Agitation was started and reactor contents were adjusted to 22 °C (19-25 °C). The reactor was pressurized with ca. 30 psi hydrogen and the reaction mixture gently heated to 26 °C (21 to 31 °C). Hydrogenation of the reactor contents was performed under ca. 30 psi at 26 °C (21 to 31 °C; maximum 31 °C) until the reaction was complete by HPLC. After 16.5 hrs, the reaction was deemed complete after confirming the disappearance of starting material (0.472 A%). The contents of the reactor were circulated through a conditioned Celite™ pad (0.2-0.5 kg Celite™ conditioned with 20-55 kg dichloromethane) prepared in a 8″ sparkler filter to remove the platinum catalyst. The reactor and Celite™ bed were rinsed forward with two portions of dichloromethane (83 kg). The filtrate was transferred to and concentrated in a 570 L glass-lined reactor under vacuum and a maximum temperature of 45 °C to ca. 132 L. Ethanol (69 kg) was charged and concentration continued under vacuum and a maximum temperature of 45 °C to ca. 132 L. In-process NMR indicated that the dichloromethane content was 39%. Ethanol (69 kg) was charged again and concentration continued again to ca. 132 L. In-process NMR indicated that the dichloromethane content was 5%. The reaction mixture was then adjusted to 3 °C (0 to 6 °C), agitated for ca. 1 hr, and the resulting slurry filtered onto a jacketed pressure nutsche fitted with a filter cloth. The reactor, pump, and lines were rinsed forward with cold [3 °C (0-6 °C)] ethanol (26 kg. The wet filter cake (36.6 kg) was dried under vacuum at 40-50 °C with a maximum temperature of water bath (to heat dryer jacket) of 50 °C. LOD analysis after 12.5 hrs indicated solvent content was at 0.1%. The dry product (B) was discharged (26.4 kg) in 89.5% yield. HPLC showed 98.4 A% purity, with dechlorinated impurity at 0.083 %.

Example 5. Synthesis of 4-(N,N-dimethylcarbamimidoyl)benzoic acid (A)

Process A

Step 1: Amidine Formation

[0118] To a tetrahydrofuran solution of 2M dimethylamine, 2.3M hexane solution of hexyllithium was slowly added over a period of at least three (3) hours while maintaining the temperature at between -8°C and -12°C. This solution was added to the tetrahydrofuran solution of ethyl-4-cyanobenzoate (F) while maintaining the temperature between -8°C and -12°C. The completion of the reaction was confirmed by HPLC, and the solution temperature was adjusted to between -8°C and 3°C. The reaction mixture was slowly added to the cold solution of aqueous sodium bicarbonate solution and the desired ethyl-4-(N,N-dimethylcarbamimidoyl)benzoate (G) was extracted with ethyl acetate. The ethyl acetate layer was dried, filtered and evaporated under vacuum to afford ethyl-4-(N,N-dimethylcarbamimidoyl)benzoate (G) as a white solid.

Step 2: Hydrolysis of ester

[0119] To a THF solution of ethyl -4(N,N-dimethylcarbamimidoyl)benzoate (G) was added an aqueous solution of lithium hydroxide (2 eq.) and the reaction mixture was stirred for 6 hr. The completion of the reaction was confirmed by HPLC. To the reaction mixture was added water, followed by extraction with ethyl acetate. The aqueous layer was acidified with 6N HCI to pH between 3-4 at which point the desired 4-(N,N- dimethylcarbamimidoyl)benzoic acid precipitated as the white solid. The white solid isolated was washed with hexane to afford 4-(N,N-dimethylcarbamimidoyl)benzoic acid as an hydrochloride salt (A).

Process B:

Step 1: Ester Formation

[0120] To a methanolic solution of 4-cyanobenzoic acid was added concentrated sulfuric acid and refluxed the reaction for at least 12 hours. The completion of the reaction was confirmed by HPLC. The solution was cooled and the solvent was evaporated. To the residue was added ethyl acetate followed by washing with 10 % sodium hydroxide solution. The ethyl acetate layer was dried, filtered and evaporated to give desired 4-methyl cyanobenzoate as a white solid.

Step 2: Dimethylamidine formation

[0121] A stream of HCI (gas) was bubbled through a 0 °C solution of 4-methyl cyanobenzoate (1 mmol) in 50 mL of ethanol until saturation. The mixture was stirred at room temperature overnight and evaporated to afford compound P. The resulting residue was treated with dimethylamine hydrochloride (0.15 eq.) in 20 mL ethanol at reflux temperature for 4 hours. The solvent was removed at reduced pressure and the residue was washed with hexane to afford desired product Q as a light yellow solid.

Step 3: Ester hydrolysis

[0122] To a THF solution of ethyl-4(N,N-dimethylcarbamimidoyl)benzoate (Q) was added an aqueous solution of lithium hydroxide (2 eq.) and the reaction mixture was stirred for 6 hours. The completion of the reaction was confirmed by HPLC. To the reaction mixture was added water, followed by extraction with ethyl acetate. The aqueous layer was acidified with 6N HC1 to pH between 3-4 at which point the desired 4- (N,N-dimethylcarbamimidoyl)benzoic acid precipitated as the white solid. The white solid isolated was washed with hexane to afford 4-(N,N-dimethylcarbamimidoyl)benzoic acid as an hydrochloride salt (A).

Example 6: Preparation of betrixaban, free base

[0123] To 100 mL round bottom flask, was added compound B (2.0 g, obtained as in Example 4), compound A (1.98 g, obtained as in example 5), 20 mL N,N- dimethylacetamide. The reaction mixture was stirred briefly so as to dissolve most of the solid, then con. HC1 (36 microliters) was added. To this thin slurry add EDC HCl (1.8 g total, Aldrich) in 3 portions, 0.6 g each, 20 min apart. The reaction mixture was stirred for 1.5 hours for complete reaction. [0124] To this reaction was added 2.3 g sodium carbonate solution in 10 mL water while the batch was cooled with water bath to keep the batch temperature 22-30 °C. Vigorous agitation was required to keep the batch well mixed. Then 10 mL water was added. The batch was stirred at 22-25 °C for 30 min. After a slurry was formed, 20 mL more water was added. The batch was stirred at 22 °C for 1 hour. The batch was filtered and the wet cake was washed with 3×5 mL water, then 5 mL acetone. The cake was dried on the funnel by suction. The weight of the dry cake is 2.95 g -2.92 g which is the crude betrixaban. To purify the crude betrixaban obtained, 1.0 g of the crude solid was mixed with 4 mL Ν,Ν-dimethylacetamide and heated to 70 °C for 30 min. Then add 8 mL toluene was added and the mixture was heated for 30 min, then cooled to 22 °C over 1 h, then cooled to 0 °C, aged at 0 °C for 2 hours, filtered, washed with 2×1 mL toluene. The cake was dried on the funnel by suction to obtain 0.88 g pure betrixaban (I).

WO2012031017A1 * Aug 31, 2011 Mar 8, 2012 Merck Sharp & Dohme Corp. CRYSTALLINE FORMS OF A FACTOR Xa INHIBITOR
WO2013033370A1 * Aug 30, 2012 Mar 7, 2013 Portola Pharmaceuticals, Inc. Prevention and treatment of thrombosis in medically ill patients
US8946269 Aug 31, 2011 Feb 3, 2015 Portola Pharmaceuticals, Inc. Crystalline forms of a factor Xa inhibitor
WO2004083174A2 * Mar 17, 2004 Sep 30, 2004 Timur Gangor Sulfonyl-amidino containing and tetrahydropyrimidino containing compounds as factor xa inhibitors
WO2008057972A1 Nov 1, 2007 May 15, 2008 Millennium Pharm Inc Methods of synthesizing pharmaceutical salts of a factor xa inhibitor
US6376515 Feb 28, 2001 Apr 23, 2002 Cor Therapeutics, Inc. Benzamides and related inhibitors of factor Xa
US6835739 Oct 15, 2003 Dec 28, 2004 Millennium Pharmaceuticals, Inc. Benzamides and related inhibitors of factor Xa
US6844367 Sep 15, 2000 Jan 18, 2005 Millennium Pharmaceuticals, Inc. Benzamides and related inhibitors of factor Xa
US61287680

References

 

 

  1. Eriksson BI, Quinlan DJ, Weitz JI (2009). “Comparative pharmacodynamics and pharmacokinetics of oral direct thrombin and factor xa inhibitors in development”. Clinical Pharmacokinetics48 (1): 1–22. PMID19071881.
  2. Zhang P, Huang W, Wang L, Bao L, Jia ZJ, Bauer SM, Goldman EA, Probst GD, Song Y, Su T, Fan J, Wu Y, Li W, Woolfrey J, Sinha U, Wong PW, Edwards ST, Arfsten AE, Clizbe LA, Kanter J, Pandey A, Park G, Hutchaleelaha A, Lambing JL, Hollenbach SJ, Scarborough RM, Zhu BY (April 2009). “Discovery of betrixaban (PRT054021), N-(5-chloropyridin-2-yl)-2-(4-(N,N-dimethylcarbamimidoyl)benzamido)-5-methoxybenzamide, a highly potent, selective, and orally efficacious factor Xa inhibitor”. Bioorganic & Medicinal Chemistry Letters19 (8): 2179–85. doi:10.1016/j.bmcl.2009.02.111. PMID19297154.
  3. Turpie AG, Bauer KA, Davidson BL, Fisher WD, Gent M, Huo MH, Sinha U, Gretler DD (January 2009). “A randomized evaluation of betrixaban, an oral factor Xa inhibitor, for prevention of thromboembolic events after total knee replacement (EXPERT)”. Thrombosis and Haemostasis101 (1): 68–76. PMID19132191.
  4. Piccini, J. P.; Lopes, R. D.; Mahaffey, K. W. (2010). “Oral factor Xa inhibitors for the prevention of stroke in atrial fibrillation”. Current Opinion in Cardiology25 (4): 312. doi:10.1097/HCO.0b013e32833a524f. PMID20520539edit
  5. Sobieraj-Teague, M.; O’donnell, M.; Eikelboom, J. (2009). “New Anticoagulants for Atrial Fibrillation”. Seminars in Thrombosis and Hemostasis35 (5): 515–24. doi:10.1055/s-0029-1234147. PMID19739042edit

Husten, Harry. “Merck Abandons Development of Factor Xa Inhibitor Betrixaban”. CardioBrief. Retrieved 11 April 2014.

Betrixaban
Betrixaban.svg
Systematic (IUPAC) name
N-(5-chloropyridin-2-yl)-2-([4-(N,N-dimethylcarbamimidoyl)benzoyl]amino)-5-methoxybenzamide
Clinical data
Legal status
  • Development terminated
Identifiers
CAS Number 330942-05-7 
ATC code None
PubChem CID: 10275777
ChemSpider 18981107 Yes
UNII 74RWP7W0J9 Yes
ChEMBL CHEMBL512351 Yes
Chemical data
Formula C23H22ClN5O3
Molecular mass 451.905 g/mol

 

/////////////CN(C)C(=N)C1=CC=C(C=C1)C(=O)NC2=C(C=C(C=C2)OC)C(=O)NC3=NC=C(C=C3)Cl

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