AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

Green Pocketbook® for Research & Development Departments from ViridisChem Inc.

 TOXICITY, Uncategorized  Comments Off on Green Pocketbook® for Research & Development Departments from ViridisChem Inc.
Jul 232016
 

str1


CLICK TO PLAY THE ABOVE MEDIA CLIP……

 

Comprehensive toxicological data on raw material is practically non existent, which means designing green product development processes can be tedious and time consuming.

ViridisChem Inc., is offering a solution that will help scientists make environmentally friendly decisions throughout the product development life-cycle.

Our first product the Green Pocketbook® is a cloud based reference tool that helps scientists assess chemicals of high concern and identify safer alternatives that are less hazardous for people and environmentally friendly.

Containing more than 90 million chemical entries from the worlds best structure, reaction and literature databases, the Green Pocketbook combines the power of a search engine with user friendly decisions support tools.

str1

click  Media

Unlike other chemistry reference products, it is the only software tool in the industry that correlates physical and toxicological properties then calculates a Green Score based on these properties.  The software allows the user to visually compare multiple chemicals side by side for easy assessment and decision making.

str1

Other Features include:

Flexible Search options, search by;

  • Chemical Name, CAS#, IUPAC Name
  • Structure, or draw your own
  • Citation, Reference, Patents

Comprehensive Physical and Toxicological data

  • The software displays many of the properties contained in the MSDS plus more.
  • There over 24 physical properties and over 26 toxicological properties that can be configured and displayed to suite your preferences
  • Displays US and International regulatory concerns

str1

click  Media

 

 

Jose Castanon

Jose Castanon

Marketing Leader, Team Builder

Best,
Jose Castanon
ViridisChem Inc.
408 218 3125
josec@viridischem.com
click  Media
 
Jose Castanon is a marketing professional with over 15 years of experience in medical technology and devices. He specializes in growth strategies and bringing new technologies to market. Prior to ViridisChem Jose was the Director of Marketing for Omnicell, where he led all North American marketing activity for the Medication Automation and Analytics business line. Jose has held commercial leadership roles at Philips Healthcare, Roche and Johnson and Johnson. He holds a BS in Biology from the University of Puget Sound and an MBA from Pepperdine University.

Neelam Vaidya

Neelam Vaidya

Experienced executive with passion to bring most needed solutions to market

Neelam Vaidya is a serial entrepreneur with over 25 years’ experience in both high-tech and bio-tech industry. In 2014, Neelam and Rahul Vaidya cofounded ViridisChem Inc. with the mission to develop a portfolio of software solutions that will provide all the information and analysis capabilities scientists would need to practice “green chemistry” within their everyday research, that will result in environmentally friendly product development processes that are greener, safer, and economical. To enable this goal, the company has built in-house and proprietary
– chemical database with over 60 million chemicals,
– citation database with over 20 million citations and patents,
– reaction database with over 10 million reference reactions

It recently launched its first product Green Pocketbook that is being used both as an educational tool and as a reference guide by universities and industry scientists for their day-to-day research needs. It provides full chemical and toxicological profiles of chemicals, and offers “green scores” based on these properties through easy to understand visual charts. With inclusion of chemicals relevant to most industries, and by providing the most comprehensive information about chemicals in a very easy to use and understand manner, Green Pocketbook is proving to be a “must have” software solution for scientists from most industries for their day-to-day work.

In past she was the founder and CEO of a biotech company ChiroSolve, Inc. that offers products and services that define chiral resolution method for optically active and hard-to-separate chiral molecules. Neelam was also the founder of Software Company called Perfect Solutions that offered enterprise software products for high-tech industry. Before her entrepreneur career, Neelam has lead number high-profile enterprise infrastructure projects

ACS National Fall Symposium, 2015, Boston, MA (August 16-18, 2015)

News/Events – ViridisChem, Inc.

www.viridischem.com

ViridisChem Team

 

 

REFERENCES

http://www.viridischem.com/wp-content/uploads/2016/03/GreenPocket-Datasheet-3-10-2016_V3.pdf

https://library.stanford.edu/swain/databases/green-pocketbook

////////Green Pocketbook, Research & Development Departments, Jose Castanon,  ViridisChem Inc,

Neelam Vaidya

Share

AZD 1981

 Uncategorized  Comments Off on AZD 1981
Jul 222016
 

 

STR1

AZD1981; AZD-1981; 802904-66-1; UNII-2AD53WQ2CX; ; AZD 1981;
Molecular Formula: C19H17ClN2O3S
Molecular Weight: 388.86788 g/mol
      1H-Indole-1-acetic acid, 4-(acetylamino)-3-[(4-chlorophenyl)thio]-2-methyl-
  • 2-[4-acetamido-3-(4-chlorophenyl)sulfanyl-2-methylindol-1-yl]acetic acid
  • Originator AstraZeneca
  • Developer AstraZeneca; Johns Hopkins University
  • Class Antiasthmatics
  • Mechanism of Action Prostaglandin D2 receptor antagonists
    • Phase II Urticaria
    • Discontinued Asthma; Chronic obstructive pulmonary disease

    Most Recent Events

    • 09 Mar 2016 AZD 1981 is still in phase II trials for Urticaria in USA (PO)
    • 07 Mar 2016 Johns Hopkins University in collaboration with AstraZeneca completes a phase II trial in Urticaria in USA (PO) (NCT02031679)
    • 04 Mar 2016 Efficacy and safety data from a phase II trial in Urticaria presented at the Annual Meeting of the American Academy of Allergy, Asthma and Immunology (AAAAI-2016)

https://ncats.nih.gov/files/AZD1981.pdf

SEE

NMR

HPLC

AZD1981 is a potent, selective CRTh2 (DP2) receptor antagonist with IC50 of 4 nM, showing >1000-fold selectivity over more than 340 other enzymes and receptors, including DP1. Phase 2.

AZD1981.png

118 patients were randomised to treatment (AZD1981 n = 61; placebo n = 57); 83% of patients were male and the mean age was 63 years (range 43-83). There were no significant differences in the mean difference in change from baseline to end of treatment between AZD1981 and placebo for the co-primary endpoints of pre-bronchodilator FEV1 (AZD1981-placebo: -0.015, 95% CI: -0.10 to 0.070; p = 0.72) and CCQ total score (difference: 0.042, 95% CI: -0.21 to 0.30; p = 0.75). Similarly, no differences were observed between treatments for the other outcomes of lung function, COPD symptom score, 6-MWT, BODE index, and use of reliever medication. AZD1981 was well tolerated.

CONCLUSION:

There was no beneficial clinical effect of AZD1981, at a dose of 1000 mg twice daily for 4 weeks, in patients with moderate to severe COPD. AZD1981 was well tolerated and no safety concerns were identified.

 

STR1

 

STR1

STR1

 

 

Biological Activity

Description AZD1981 is a potent, selective CRTh2 (DP2) receptor antagonist with IC50 of 4 nM, showing >1000-fold selectivity over more than 340 other enzymes and receptors, including DP1. Phase 2.
Targets CRTh2 (DP2) receptor [1]
IC50 4 nM
In vitro AZD1981, as a potent antagonist in a disease relevant cell system, inhibits DK-PGD2-induced CD11b expression in human eosinophils with IC50 of 10 nM. [1] AZD1981 blocks DP2-mediated shape change in human eosinophils and basophils in blood, as well as DP2-mediated chemotaxis of human Th2 cells and eosinophils. Moreover, AZD1981 also blocks the binding of [3H]PGD2 to mouse, rat, guinea pig, rabbit and dog recombinant DP2. [2]
In vivo AZD1981 has high oral bioavailability in male sprague dawley rats. [1] In guinea pig hind limb model, AZD1981 (100 nM) completely inhibits DK-PGD2-induced eosinophil mobilization. [2]
Features An orally available selective DP2(CRTh2) receptor antagonist in clinical development for asthma.

Protocol(Only for Reference)

Kinase Assay: [2]

DP2 binding studies A scintillation proximity assay (SPA) following [3H]PGD2 binding to membranes of HEK cells expressing recombinant DP2 is used. The potency of AZD1981 as an antagonist is determined by quantifying its ability to displace specific radio-ligand binding. Briefly, membranes from HEK293 expressing recombinant human DP2 are pre-bound to Wheat Germ Agglutinin-coated PVT-SPA beads for 18 h at 4°C. Assays were started by the addition of 25 μL of membrane-coated beads (10 mg/mL of beads) to an assay buffer (50 mm HEPES pH 7.4 containing 5 mm MgCl2) containing 2.5 nM [3H]PGD2 in the absence or the presence of increasing concentrations of the tested compounds (50 μL final volume). Non-specific binding is determined in the same conditions but in the presence of 10 μM DK-PGD2. Plates are incubated for 2 h at room temperature, and bead-associated radioactivity is measured using a Wallac Microbeta counter. The concentration of the compounds causing 50% inhibition of binding of [3H]PGD2 to the receptor is calculated (IC50). Ki values have not been derived from IC50, as there is no evidence of a simple competitive interaction with PGD2. The same methodology is used for recombinant human, murine, rat, guinea pig, dog and rabbit DP2. Reversibility of binding to the human receptor was assessed by recovery of [3H]PGD2 binding after removal of AZD1981 by washing of the membrane-coated SPA beads. HEK-membrane-coated beads are incubated in the presence of AZD1981 for 2 h at room temperature to bind the compound to DP2. To remove the bound AZD1981, beads are centrifuged (1 min at 1300× g), and the pellet resuspended in 1 mL of assay buffer. This is repeated four times. Aliquots (30 μL) are transferred to 96-well plates, and [3H]PGD2 binding is evaluated as above. Parallel samples containing (i) 10 μM DK-PGD2 during the 2 h incubation and in the wash buffer; (ii) AZD1981 at 2 μM in the wash buffer; and (iii) vehicle are processed alongside to determine non-specific binding and the ‘no wash’ condition whilst controlling for loss of beads during the washing process. The time from first wash to end of first reading is approximately 13 min.

Animal Study: [1]

Animal Models Male sprague dawley rats.
Formulation
Dosages 1 mg/kg(i.v.), 4 mg/kg(oral)
Administration i.v. or oral administration

Conversion of different model animals based on BSA (Value based on data from FDA Draft Guidelines)

Species Mouse Rat Rabbit Guinea pig Hamster Dog
Weight (kg) 0.02 0.15 1.8 0.4 0.08 10
Body Surface Area (m2) 0.007 0.025 0.15 0.05 0.02 0.5
Km factor 3 6 12 8 5 20
Animal A (mg/kg) = Animal B (mg/kg) multiplied by  Animal B Km
Animal A Km

For example, to modify the dose of resveratrol used for a mouse (22.4 mg/kg) to a dose based on the BSA for a rat, multiply 22.4 mg/kg by the Km factor for a mouse and then divide by the Km factor for a rat. This calculation results in a rat equivalent dose for resveratrol of 11.2 mg/kg.

Rat dose (mg/kg) = mouse dose (22.4 mg/kg) × mouse Km(3)  = 11.2 mg/kg
rat Km(6)

 

References

[1] Luker T, et al. Bioorg Med Chem Lett. 2011, 21(21), 6288-6292.

[2] Schmidt JA, et al. Br J Pharmacol. 2013, 168(7), 1626-1638.

Clinical Trial Information( data from http://clinicaltrials.gov, updated on 2016-07-09)

NCT Number Recruitment Conditions Sponsor
/Collaborators
Start Date Phases
NCT02031679 Recruiting Chronic Idiopathic Urticaria Johns Hopkins University|AstraZeneca January 2014 Phase 2
NCT01311635 Completed Healthy AstraZeneca April 2011 Phase 1
NCT01254461 Completed Drug Interaction AstraZeneca February 2011 Phase 1
NCT01265641 Completed Asthma AstraZeneca January 2011 Phase 1
NCT01199341 Completed Pharmakokinetic AstraZeneca October 2010 Phase 1

Patent ID Date Patent Title
US2015210655 2015-07-30 CERTAIN (2S)-N-[(1S)-1-CYANO-2-PHENYLETHYL]-1,4-OXAZEPANE-2-CARBOXAMIDES AS DIPEPTIDYL PEPTIDASE 1 INHIBITORS
US2015072963 2015-03-12 COMPOSITIONS AND METHODS FOR REGULATING HAIR GROWTH
US2014328861 2014-11-06 Combination of CRTH2 Antagonist and a Proton Pump Inhibitor for the Treatment of Eosinophilic Esophagitis
US8772305 2014-07-08 Substituted pyridinyl-pyrimidines and their use as medicaments
US8227622 2012-07-24 Pharmaceutical Process and Intermediates 714
US2012178764 2012-07-12 Novel Compounds
US2011263614 2011-10-27 Novel compounds
US7781598 2010-08-24 Process for the preparation of substituted indoles
US7687535 2010-03-30 Substituted 3-sulfur indoles
US2009163518 2009-06-25 Novel Compounds

///////////

CC1=C(C2=C(N1CC(=O)O)C=CC=C2NC(=O)C)SC3=CC=C(C=C3)Cl

 

Share

AZD 3514 MALEATE

 Uncategorized  Comments Off on AZD 3514 MALEATE
Jul 222016
 

STR1

AZD3514; AZD 3514; AZD-3514.

CAS 1240299-33-5
Chemical Formula: C25H32F3N7O2
Exact Mass: 519.25696

1-(4-(2-(4-(1-(3-(trifluoromethyl)-7,8-dihydro-[1,2,4]triazolo[4,3-b]pyridazin-6-yl)piperidin-4-yl)phenoxy)ethyl)piperazin-1-yl)ethanone

Ethanone, 1-​[4-​[2-​[4-​[1-​[7,​8-​dihydro-​3-​(trifluoromethyl)​-​1,​2,​4-​triazolo[4,​3-​b]​pyridazin-​6-​yl]​-​4-​piperidinyl]​phenoxy]​ethyl]​-​1-​piperazinyl]

6-f4-{4-[2-f4-acetylpiperazin-l-yl)ethoxylphenyl}piperidin-l-yl)-3-( trifluoromethyr)-7,8-dihvdro [ 1 ,2,41 triazolo [4,3-bl pyridazine

6-(4-{4-[2-(4-acetylpiperazin-l- vDethoxyl phenyllpiperidin- l-vD-3-f trifluoromethyl)-7.,8-(iihv(iro [ 1 ,2,41 triazolo [4,3- blpyridazine

  • 1-[4-[2-[4-[1-[7,8-Dihydro-3-(trifluoromethyl)-1,2,4-triazolo[4,3-b]pyridazin-6-yl]-4-piperidinyl]phenoxy]ethyl]-1-piperazinyl]ethanone
  • Originator AstraZeneca
  • Class Antineoplastics
  • Mechanism of Action Androgen receptor antagonists

AZD-3514 is a potent androgen receptor downregulator with potential anticancer cancer activity. AZD3514 is being evaluated in a Phase I clinical trial in patients with castrate-resistant prostate cancer.

AZD3514 is currently in Phase I trail. This trial is looking at a new drug called AZD3514 for men who have prostate cancer that has spread to other parts of the body and is no longer responding to hormone therapy.  Doctors often use hormone therapy to treat prostate cancer. This may keep it under control for long periods of time. But researchers are looking for treatments that will help men who have prostate cancer that stops responding to hormone therapy.  Prostate cancer needs the hormone testosterone to grow. The testosterone locks into receptors on the cancer cells. AZD3514 works by breaking down these receptors so that testosterone canÂ’t tell the prostate cancer cells to grow.

img

 

 

6-(4-{4-[2-(4-Acetylpiperazin-1-yl)ethoxy]phenyl}piperidin-1-yl)-3-(trifluoromethyl)-7,8-ihydro[1,2,4]triazolo[4,3-b]pyridazine 

as a white, free flowing solid.

1H NMR (400 MHz, CDCl3): δ 1.62 (2H, m), 1.88 (2H, m), 2.02 (3H, s), 2.49 (4H, m), 2.65 – 2.78 (5H, m), 2.94 (2H, m), 3.15 (2H, t), 3.42 (2H, m), 3.57 (2H, m), 4.03 (2H, t), 4.24 (2H, m), 6.80 (2H, d), 7.06 (2H, d);

m/z = 520 [M+H]+. RT = 0.87: 99% purity.

HRMS found 520.26373,

 

Prostate cancer is the second leading cause of death from cancer among men in developed countries, and was projected to account for 25% of newly-diagnosed cases and 9% of deaths due to cancer in the USA in 2010. The androgen receptor (AR), a ligand binding transcription factor in the nuclear hormone receptor super family, is a key molecular target in the etiology and progression of prostate cancer.Binding of the endogenous AR ligand dihydrotestosterone stabilizes and protects the AR from rapid proteolytic degradation. The early stages of prostate cancer tumor growth are androgen dependent and respond well to androgen ablation,  either via surgical castration or by chemical castration with a luteinizing hormone releasing hormone agonist in combination with an AR antagonist, such as bicalutamide.

Although introduction of androgen deprivation therapy represented a major advance in prostate cancer treatment, recurrence within 1–2 years typically marks transition to the so-called castrate-resistant state, in which the tumor continues to grow in the presence of low circulating endogenous ligand and is no longer responsive to classical AR antagonists. Castrate-resistant prostate cancer (CRPC) is a largely unmet medical need with a 5-year survival rate of less than 15%. Antimitotic agents docetaxel and cabazitaxel, testosterone biosynthesis inhibitor abiraterone acetate and second generation AR antagonist enzalutamide (MDV3100) are the currently approved small-molecule drugs that have been shown to provide survival benefit.

Recent evidence from both pre-clinical and clinical studies is consistent with the importance of re-activation of AR signaling in a majority of castrate-resistant prostate tumors. It is also well established that the functional AR in castrate-resistant tumors is frequently mutated or amplified, and that over-expression can convert hormone-responsive cell lines to hormone refractory. Recent second-generation AR antagonists have been designed that retain antagonism in over-expressing cell lines, and among these agents enzalutamide has recently successfully met efficacy criteria in a large Phase III clinical trial.

By analogy with fulvestrant, an estrogen receptor (ER) downregulator approved by the FDA in 2002 for treatment of advanced breast cancer and initially characterized as a pure ER antagonist, a ligand which downregulates the AR represents one of a number of potential approaches to treatment of CRPC via a sustained reduction in tumor AR content. We recently described derivation from a novel 3-(trifluoromethyl)-[1,2,4]triazolo[4,3-b]pyridazine ligand of AR inhibitor 1 The compound also causes AR downregulation15 and high plasma levels following oral administration in pre-clinical models compensate for moderate cellular potency

Figure 1.

Structures of lead AR downregulator 1 and chemotype 2.

Structures of lead AR downregulator 1 and chemotype 2.

Scheme 3.

Synthesis of compounds 10, 11a–b, 12. Reagents and conditions: (a) ...

Synthesis of compounds 10, 11ab, 12. Reagents and conditions: (a) 2-(1-Methyl-1H-pyrazol-5-yl)ethanol,27 Ph3P, diisopropyl azodicarboxylate, THF, 20 °C; (b) 2-(4-acetylpiperazine-1-yl)ethanol,28 Ph3P, diisopropyl azodicarboxylate, THF, 20 °C; (c) H2, 10% Pd-C, MeOH, 50 °C.

PATENT

WO 2010092371

 Robert Hugh Bradbury, Gregory Richard Carr,Alfred Arthur Rabow, Korupoju Srinivasa Rao,Harikrishna Tumma,
Applicant Astrazeneca Ab, Astrazeneca Uk Limited

Preparation of 6-f4-{4-[2-f4-acetylpiperazin-l-yl)ethoxylphenyl}piperidin-l-yl)-3-

( trifluoromethyr)-7,8-dihvdro [ 1 ,2,41 triazolo [4,3-bl pyridazine

Figure imgf000079_0001

A solution of acetyl chloride (0.027 mL, 0.38 mmol) in DCM (0.5 mL) was added dropwise to 6-[4- [4- [2-(piperazin- 1 -yl)ethoxy]phenyl]piperidin- 1 -yl] -3 -(trifluoromethyl)- 7,8-dihydro-[l,2,4]triazolo[4,3-b]pyridazine (150 mg, 0.31 mmol) and triethylamine (0.088 mL, 0.63 mmol) in DCM (1 mL) cooled to 00C under nitrogen. The resulting solution was stirred at 00C for 5 minutes then allowed to warm to room temperature and stirred for 15 minutes. The reaction mixture was diluted with water (2 mL), passed through a phase separating cartridge and then the organic layer was evaporated to afford crude product. The crude product was purified by preparative HPLC (Waters XBridge Prep Cl 8 OBD column, 5μ silica, 19 mm diameter, 100 mm length), using decreasingly polar mixtures of water (containing 1% ammonia) and MeCN as eluents. Fractions containing the desired compound were evaporated to dryness to give 6-(4-{4-[2-(4-acetylpiperazin-l- yl)ethoxy]phenyl}piperidin-l-yl)-3-(trifluoromethyl)-7,8-dihydro[l,2,4]triazolo[4,3- b]pyridazine (80 mg, 49%) as a gum.

IH NMR (399.9 MHz, CDC13) δ 1.69 (2H, m), 1.95 (2H, m), 2.08 (3H, s), 2.56 (4H, m), 2.71 – 2.84 (5H, m), 3.00 (2H, m), 3.22 (2H, t), 3.48 (2H, m), 3.63 (2H, m), 4.10 (2H, t), 4.31 (2H, m), 6.86 (2H, d), 7.12 (2H, d); m/z = 520 [M+H]+.

The 6-[4-[4-[2-(piperazin- 1 -yl)ethoxy]phenyl]piperidin- 1 -yl]-3-(trifluoromethyl)-7,8- dihydro-[l,2,4]triazolo[4,3-b]pyridazine used as starting material was prepared as follows :-

Preparation of tert-butyl 4-[2-[4-(l-(benzyloxycarbonyl)-l,2,3,6-tetrahydropyridin-4- yl)phenoxy]ethyl]piperazine-l-carboxylate DIAD (12.60 mL, 64.00 mmol) was added dropwise to benzyl 4-(4-hydroxyphenyl)-5,6- dihydropyridine-l(2H)-carboxylate (obtained as described in Example 4.1, preparation of starting materials) (16.5 g, 53.34 mmol), tert-butyl 4-(2-hydroxyethyl)piperazine-l- carboxylate (CAS 77279-24-4) (14.74 g, 64.00 mmol) and triphenylphosphine (16.79 g, 64.00 mmol) in THF (150 mL) under nitrogen. The resulting solution was stirred at ambient temperature for 16 hours. The reaction mixture was evaporated to dryness then the residue was stirred in ether (200 mL) for 10 minutes at room temperature. The resulting precipitate was removed by filtration and discarded. The ether filtrate was washed with water (100 mL) followed by saturated brine (100 mL), then dried over MgSO4, filtered and evaporated to give crude product. The crude product was purified by flash silica chromatography, elution gradient 20 to 60% EtOAc in isohexane. Fractions containing the desired product were evaporated to dryness to afford tert-butyl 4-[2-[4-(l- (benzyloxycarbonyl)- 1,2,3, 6-tetrahydropyridin-4-yl)phenoxy]ethyl]piperazine-l- carboxylate (34.6 g, 82%) as a gum which was contaminated with 34% by weight triphenylphosphine oxide.

IH NMR (399.9 MHz, DMSO-d6) δ 1.40 (9H, s), 2.42 – 2.47 (6H, m), 2.71 (2H, m), 3.32 (4H, m), 3.62 (2H, m), 4.03 – 4.10 (4H, m), 5.12 (2H, s), 6.06 (IH, m), 6.92 (2H, d), 7.31 – 7.40 (7H, m); m/z = 522 [M+H]+.

Preparation of tert-butyl 4-[2-[4-(piperidin-4-yl)phenoxy]ethyl]piperazine-l- carboxylate tert-Butyl 4-[2-[4-(l-(benzyloxycarbonyl)-l,2,3,6-tetrahydropyridin-4- yl)phenoxy]ethyl]piperazine-l-carboxylate (66% pure by weight) (34.62 g, 43.80 mmol) and 5% palladium on carbon (50% wet) (4.47 g, 1.05 mmol) in MeOH (250 mL) were stirred under an atmosphere of hydrogen at 5 bar and 600C for 4 hours. The catalyst was removed by filtration and the solvents evaporated to give crude product. The crude product was purified by flash silica chromatography, eluting with 60% EtOAc in isohexane then 15% 2M ammonia/MeOH in DCM. Pure fractions were evaporated to dryness to afford tert-butyl 4-[2-[4-(piperidin-4-yl)phenoxy]ethyl]piperazine-l-carboxylate (15.42 g, 90%) as a solid. IH NMR (399.9 MHz, CDC13) δ 1.46 (9H, s), 1.62 (2H, m), 1.81 (2H, m), 2.50 – 2.59 (5H, m), 2.73 (2H, m), 2.80 (2H, t), 3.18 (2H, m), 3.44 (4H, m), 4.09 (2H, t), 6.85 (2H, d), 7.13 (2H, d); m/z = 390 [M+H]+.

Preparation of tert-butyl 4-[2-[4-[l-(3-(trifluoromethyl)-[l,2,4]triazolo[4,3- b]pyridazin-6-yl]piperidin-4-yl]phenoxy]ethyl]piperazine-l-carboxylate

DIPEA (2.348 mL, 13.48 mmol) was added to 6-chloro-3-(trifluoromethyl)- [l,2,4]triazolo[4,3-b]pyridazine (obtained as described in Monatsh. Chem. 1972, 103, 1591) (2 g, 8.99 mmol) and tert-butyl 4-[2-[4-(piperidin-4-yl)phenoxy]ethyl]piperazine-l- carboxylate (3.68 g, 9.44 mmol) in DMF (30 mL). The resulting solution was stirred at 800C for 2 hours. The reaction mixture was cooled to room temperature and the solvents evaporated to dryness. The resulting solid was triturated with water then collected by filtration, washed with ether and dried to afford tert-butyl 4-[2-[4-[l-(3-(trifluoromethyl)- [l,2,4]triazolo[4,3-b]pyridazin-6-yl]piperidin-4-yl]phenoxy]ethyl]piperazine-l -carboxylate (5.02 g, 97%) as a solid.

IH NMR (399.9 MHz, CDC13) δ 1.46 (9H, s), 1.76 (2H, m), 2.00 (2H, m), 2.54 (4H, m), 2.75 – 2.86 (3H, m), 3.11 (2H, m), 3.46 (4H, m), 4.11 (2H, m), 4.37 (2H, m), 6.87 (2H, d), 7.13 (3H, m), 7.92 (IH, d); m/z = 576 [M+H]+.

Preparation of tert-butyl 4-[2-[4-[l-[3-(trifluoromethyl)-7,8-dihydro-

[1 ,2,4] triazolo [4,3-b] pyridazin-6-yl)piperidin-4-yl] phenoxy] ethyl] piperazine- 1- carboxylate

10% Palladium on carbon (0.924 g, 0.87 mmol) was added to tert-butyl 4-[2-[4-[l-(3- (trifluoromethyl)-[l,2,4]triazolo[4,3-b]pyridazin-6-yl]piperidin-4- yl]phenoxy]ethyl]piperazine-l -carboxylate (2.5 g, 4.34 mmol) and ammonium formate (2.74 g, 43.43 mmol) in ethanol (100 mL). The resulting mixture was stirred at 78°C, with further portions of ammonium formate being added every 5 hours until the reaction was complete. The reaction mixture was cooled to room temperature and the catalyst was removed by filtration. The filtrate was evaporated to dryness, redissolved in DCM (100 mL) and the solution was washed with water (100 mL) followed by brine (50 mL), then the solvents were evaporated to afford tert-butyl 4-[2-[4-[l-[3-(trifluoromethyl)-7,8-dihydro- [l,2,4]triazolo[4,3-b]pyπdazin-6-yl)pipeπdin-4-yl]phenoxy]ethyl]piperazine-l-carboxylate (2.02O g, 81%) as a solid.

IH NMR (399.9 MHz, CDC13) δ 1.46 (9H, s), 1.69 (2H, m), 1.95 (2H, m), 2.52 (4H, m), 2.71 – 2.82 (5H, m), 3.00 (2H, m), 3.22 (2H, t), 3.45 (4H, m), 4.09 (2H, m), 4.31 (2H, m), 6.86 (2H, d), 7.12 (2H, d); m/z = 578 [M+H]+.

Preparation of 6- [4-[4- [2-(piperazin-l-yl)ethoxy] phenyl] piperidin-1-yl] -3- (trifluor omethyl)-7,8-dihydr o- [ 1 ,2,4] triazolo [4,3-b] pyridazine

TFA (10 mL) was added to tert-butyl 4-[2-[4-[l-[3-(trifluoromethyl)-7,8-dihydro- [l,2,4]triazolo[4,3-b]pyπdazin-6-yl)pipeπdin-4-yl]phenoxy]ethyl]piperazine-l-carboxylate (2.02 g, 3.50 mmol) in DCM (10 mL). The resulting solution was stirred at ambient temperature for 1 hour then added to an SCX column. The desired product was eluted from the column using 2M ammonia/MeOH and the solvents were evaporated to afford 6-[4-[4- [2-(piperazin-l-yl)ethoxy]phenyl]piperidin-l-yl]-3-(trifluoromethyl)-7,8-dihydro- [l,2,4]triazolo[4,3-b]pyridazine (1.660 g, 99%) as a solid.

IH NMR (399.9 MHz, CDC13) δ 1.68 (2H, m), 1.95 (2H, m), 2.55 (4H, m), 2.70 – 2.80 (5H, m), 2.91 (4H, m), 3.00 (2H, m), 3.22 (2H, t), 4.09 (2H, t), 4.30 (2H, m), 6.87 (2H, d), 7.11 (2H, d); m/z = 478 [M+H]+.

Example 5.2

Larger scale preparation of 6-(4-{4-[2-(4-acetylpiperazin-l- vDethoxyl phenyllpiperidin- l-vD-3-f trifluoromethyl)-7.,8-dihvdro [ 1 ,2,41 triazolo [4,3- blpyridazine

Ammonium formate (99 g, 1568.94 mmol) was added to 6-[4-[4-[2-(4-acetylpiperazin-l- yl)ethoxy]phenyl]piperidin- 1 -yl]-3-(trifluoromethyl)[ 1 ,2,4]triazolo[4,3-b]pyridazine (81.2 g, 156.89 mmol) and 10% palladium on carbon (8.35 g, 7.84 mmol) in EtOH (810 mL) under nitrogen. The resulting mixture was stirred at 700C for 6 hours, then ammonium formate (50 g) was added. The mixture was stirred at 700C for 2 hours then further portions of 10% palladium on carbon (8.35 g, 7.84 mmol) and ammonium formate (50 g) were added and stirring continued at 700C for a further 10 hours. Ammonium formate (50 g) was added and the reaction mixture was stirred at 700C for 24 hours then cooled to room temperature. The catalyst was removed by filtration and the reaction charged with further 10% palladium on carbon (8.35 g, 7.84 mmol) and stirred at 700C for 16 hours. Further ammonium formate (50 g) was added and the stirring continued for 5 hours. The reaction mixture was cooled to room temperature and a further portion of 10% palladium on carbon (8.35 g, 7.84 mmol) was added. The mixture was heated to 700C for a 30 hours, cooled to room temperature and the catalyst removed by filtration and washed with EtOH. The solvent was evaporated and the residue dissolved in DCM (500 mL) and the solution washed with water (500 mL). The aqueous layer was re-extracted with DCM (500 mL), then EtOAc (500 mL x 2). The combined extracts were dried over MgSO4, filtered and evaporated to give crude product. The crude product was purified by flash silica chromatography, elution gradient 0 to 5% MeOH in DCM. Pure fractions were evaporated to dryness to afford a gum, which was slurried with ether (300 mL) and re-evaporated. Methyl tert-butyl ether (250 mL) was added and the mixture was stirred vigorously for 3 days. The solid was collected by filtration and dried to afford 6-(4-{4-[2-(4- acetylpiperazin- 1 -yl)ethoxy]phenyl}piperidin- 1 -yl)-3-(trifluoromethyl)-7,8- dihydro[l,2,4]triazolo[4,3-b]pyridazine (60.8 g, 75%) as a solid.

IH NMR (399.9 MHz, CDC13) δ 1.62 (2H, m), 1.88 (2H, m), 2.02 (3H, s), 2.49 (4H, m), 2.65 – 2.78 (5H, m), 2.94 (2H, m), 3.15 (2H, t), 3.42 (2H, m), 3.57 (2H, m), 4.03 (2H, t), 4.24 (2H, m), 6.80 (2H, d), 7.06 (2H, d); m/z = 520 [M+H]+.

The 6-[4-[4-[2-(4-acetylpiperazin-l-yl)ethoxy]phenyl]piperidin-l-yl]-3-

(trifluoromethyl)[l,2,4]triazolo[4,3-b]pyridazine used as starting material was prepared as follows :-

Preparation of 4-(piperidin-4-yl)phenol Benzyl 4-(4-hydroxyphenyl)-5,6-dihydropyridine-l(2H)-carboxylate (obtained as described in Example 4.1, preparation of starting materials) (37.7 g, 121.86 mmol) and 5% palladium on carbon (7.6 g, 3.57 mmol) in methanol (380 mL) were stirred under an atmosphere of hydrogen at 5 bar and 25°C for 2 hours. The catalyst was removed by filtration, washed with MeOH and the solvents evaporated. The crude material was triturated with diethyl ether, then the desired product collected by filtration and dried under vacuum to afford 4-(piperidin-4-yl)phenol (20.36 g, 94%) as a solid. IH NMR (399.9 MHz, DMSO-d6) δ 1.46 (2H, m), 1.65 (2H, m), 2.45 (IH, m), 2.58 (2H, m), 3.02 (2H, m), 6.68 (2H, d), 7.00 (2H, d), 9.15 (IH, s); m/z = 178 [M+H]+.

Preparation of 4- { 1- [3-(trifluor omethyl) [1 ,2,4] triazolo [4,3-b] pyridazin-6-yl] piperidin- 4-yl}phenol

DIPEA (48.2 mL, 276.86 mmol) was added to 6-chloro-3-(trifluoromethyl)- [l,2,4]triazolo[4,3-b]pyridazine (obtained as described in Monatsh. Chem. 1972, 103, 1591) (24.65 g, 110.74 mmol) and 4-(piperidin-4-yl)phenol (20.61 g, 116.28 mmol) in DMF (200 mL). The resulting solution was stirred at 800C for 1 hour. The reaction mixture was cooled to room temperature, then evaporated to dryness and re-dissolved in DCM (1 L) and washed with water (2 x 1 L). The organic layer was washed with saturated brine (500 mL), then dried over MgSO4, filtered and evaporated to afford crude product. The crude product was triturated with ether to afford 4-{l-[3- (trifluoromethyl)[l,2,4]triazolo[4,3-b]pyridazin-6-yl]piperidin-4-yl}phenol (36.6 g, 91%) as a solid.

IH NMR (399.9 MHz, DMSO-d6) δ 1.64 (2H, m), 1.87 (2H, m), 2.75 (IH, m), 3.09 (2H, m), 4.40 (2H, m), 6.69 (2H, d), 7.05 (2H, d), 7.65 (IH, d), 8.24 (IH, d), 9.15 (IH, s); m/z = 364 [M+H]+.

Preparation of 2-(4-{l-[3-(trifluoromethyl)[l,2,4]triazolo[4,3-b]pyridazin-6- yl]piperidin-4-yl}phenoxy)ethanol

A solution of ethylene carbonate (121 g, 1376.13 mmol) in DMF (200 mL) was added dropwise to a stirred suspension of 4-{l-[3-(trifluoromethyl)[l,2,4]triazolo[4,3- b]pyridazin-6-yl]piperidin-4-yl}phenol (100 g, 275.23 mmol) and potassium carbonate (76 g, 550.45 mmol) in DMF (200 mL) at 800C over a period of 15 minutes under nitrogen.

The resulting mixture was stirred at 800C for 20 hours. The reaction mixture was cooled to room temperature, then concentrated and diluted with DCM (2 L), and washed sequentially with water (1 L) and saturated brine (500 mL). The organic layer was dried over MgSO4, filtered and evaporated to afford crude product. The crude product was purified by flash silica chromatography, elution gradient 70 to 100% EtOAc in isohexane. Fractions containing the desired product were evaporated to dryness then triturated with EtOAc (150 mL). The resulting solid was washed with further EtOAc (50 mL) and ether then dried to give 2-(4- { 1 -[3-(trifluoromethyl)[ 1 ,2,4]triazolo[4,3-b]pyridazin-6-yl]piperidin-4- yl}phenoxy)ethanol. The filtrate was evaporated and further purified by flash silica chromatography, elution gradient 70 to 100% EtOAc in isohexane. Fractions containing the desired product were evaporated to dryness then triturated with ether, dried and combined with the material previously collected to afford 2-(4- { 1 -[3-

(trifluoromethyl)[ 1 ,2,4]triazolo[4,3-b]pyridazin-6-yl]piperidin-4-yl}phenoxy)ethanol (89 g, 79%) as a solid.

IH NMR (399.9 MHz, DMSO-d6) δ 1.66 (2H, m), 1.88 (2H, m), 2.80 (IH, m), 3.10 (2H, m), 3.70 (2H, m), 3.95 (2H, t), 4.41 (2H, m), 4.85 (IH, t), 6.87 (2H, d), 7.18 (2H, d), 7.67 (IH, d), 8.25 (IH, d); m/z = 408 [M+H]+.

Preparation of 2-(4-{ 1- [3-(trifluoromethyl) [ 1 ,2,4] triazolo [4,3-b] pyridazin-6- yl] piperidin-4-yl}phenoxy)ethyl methanesulfonate

A solution of methanesulfonyl chloride (20.37 mL, 262.16 mmol) in DCM (300 mL) was added to 2-(4- { 1 -[3-(trifluoromethyl)[ 1 ,2,4]triazolo[4,3-b]pyridazin-6-yl]piperidin-4- yl}phenoxy)ethanol (89 g, 218.46 mmol) and triethylamine (60.9 mL, 436.93 mmol) in DCM (900 mL) at 00C over a period of 30 minutes under nitrogen. The resulting solution was stirred at 00C for 1 hour. The reaction mixture was diluted with DCM (1 L), and washed with water (2 L). The organic layer was dried over MgSO4, filtered and evaporated to afford 2-(4- { 1 -[3-(trifluoromethyl)[ 1 ,2,4]triazolo[4,3-b]pyridazin-6-yl]piperidin-4- yl}phenoxy)ethyl methanesulfonate (104 g, 98%) as a solid.

IH NMR (399.9 MHz, DMSO-d6) δ 1.67 (2H, m), 1.89 (2H, m), 2.83 (IH, m), 3.11 (2H, m), 3.23 (3H, s), 4.23 (2H, t), 4.41 (2H, m), 4.52 (2H, t), 6.91 (2H, d), 7.21 (2H, d), 7.66 (IH, d), 8.24 (IH, d); m/z = 486 [M+H]+. Preparation of 6-[4-[4-[2-(4-acetylpiperazin-l-yl)ethoxy]phenyl]piperidin-l-yl]-3- (trifluor omethyl) [ 1 ,2,4] triazolo [4,3-b] pyridazine DIPEA (107 mL, 613.00 mmol) was added to 2-(4-{l-[3-

(trifluoromethyl)[l,2,4]triazolo[4,3-b]pyridazin-6-yl]piperidin-4-yl}phenoxy)ethyl methanesulfonate (99 g, 204.33 mmol) and N-acetylpiperazine (28.8 g, 224.77 mmol) in DMA (500 mL). The resulting solution was stirred at 1100C for 1 hour. The reaction mixture was cooled to room temperature and the solvents were evaporated. The residue was dissolved in ethyl acetate (1 L) and the solution was washed with water (1 L). The aqueous was re-extracted with ethyl acetate (1 L) and the combined organics were washed with brine (1 L), dried over MgSO4, filtered and evaporated to give crude product. The aqueous layer was basifϊed to pH 12 with 2M NaOH, then extracted with ethyl acetate (1 L), washed with brine (IL), dried over MgSO4, filtered and evaporated to give further crude product. The crude product was purified by flash silica chromatography, elution gradient 0 to 3% MeOH in DCM then 5% MeOH in DCM. Pure fractions were evaporated to give 6-[4-[4-[2-(4-acetylpiperazin-l-yl)ethoxy]phenyl]piperidin-l-yl]-3- (trifluoromethyl)[l,2,4]triazolo[4,3-b]pyridazine (81 g, 77%) as a solid. IH NMR (399.9 MHz, DMS0-d6) δ 1.59-1.73 (2H, m), 1.87 (2H, d), 1.99 (3H, s), 2.42 (2H, t), 2.71 (2H, t), 2.76-2.86 (IH, t), 3.08 (2H, t), 3.38-3.47 (4H, m), 4.08 (2H, t), 4.41 (2H, d), 6.88 (2H, d), 7.18 (2H, d), 7.62 (IH, d), 8.26 (IH, d); m/z = 518 [M+H]+.

Example 5.5

Alternative route for the preparation of 6-(4-{4-[2-(4-acetylpiperazin-l- vDethoxyl phenyllpiperidin- l-vD-3-f trifluoromethyl)-7.,8-(iihv(iro [ 1 ,2,41 triazolo [4,3- blpyridazine Form A

Methanol (375.0 mL) was added to 6-[4-[4-[2-(4-acetylpiperazin-l- yl)ethoxy]phenyl]piperidin-l-yl]-3-(trifluoromethyl)[ 1,2,4] triazolo[4,3-b]pyridazine (25.0 g, 48 m mol) in a 2.0 L autoclave reactor and to this was added 10% Pd/C (12.5 g, 50% w/w) paste at 22-25°C under nitrogen gas atmosphere. The reaction was performed under hydrogen pressure (5.0 bar) at 500C temperature for 10.0 h. The reaction mass was cooled to room temperature and the catalyst removed by filtration. Filtered cake was washed with methanol. The solvent was evaporated and the residue was azeotropically distilled by ethylacetate (2 x 125.0 mL) at 400C under reduced pressure to 3.0 rel vol (75.0 mL). Drop wise addition of tert-butylmethylether (MTBE, 375.0 mL) to the reaction mass resulted in solid material, which was collected by filtration and washed with MTBE (50.0 mL). The material was dried under reduced pressure with nitrogen gas bleed at 500C to afford the desired product 6-(4-{4-[2-(4-acetylpiperazin-l-yl)ethoxy]phenyl}piperidin-l-yl)-3- (trifluoromethyl)-7,8-dihydro[l,2,4]triazolo [4,3-b]pyridazine (22.3 g, 88%) as a white color free flowing solid. The isolated material was confirmed by XRPD as Form A. IH NMR (400.13 MHz, CDC13): δ 1.62 (2H, m), 1.88 (2H, m), 2.02 (3H, s), 2.49 (4H, m), 2.65 – 2.78 (5H, m), 2.94 (2H, m), 3.15 (2H, t), 3.42 (2H, m), 3.57 (2H, m), 4.03 (2H, t), 4.24 (2H, m), 6.80 (2H, d), 7.06 (2H, d); m/z = 520 [M+H]+.

The 6-[4-[4-[2-(4-acetylpiperazin-l-yl)ethoxy]phenyl]piperidin-l-yl]-3- (trifluoromethyl)[ 1,2,4] triazolo[4,3-b]pyridazine used as starting material was prepared as follows :-

Preparation of 4- { 1- [3-(trifluor omethyl) [1 ,2,4] triazolo [4,3-b] pyridazin-6-yl] piperidin- 4-yl}phenol: Dimethylacetamide (250.0 mL) was added to 6-chloro-3-(trifluoromethyl)- [l,2,4]triazolo[4,3-b]pyridazine [CAS: 40971-95-7] (50.0 g, 225 m mol) at 22-25°C in a suitable round bottom flask followed by 4-(piperidin-4-yl)phenol [CAS: 62614-84-0] (60.9 g, 236 m mol) at 22-25°C. The reaction mass was stirred to obtain a clear solution. Triethylamine (79.1 mL, 561 m mol) was slowly added to the reaction mass by drop wise addition over a period of 60 min at 25-300C. Temperature was raised to 400C and the reaction mass stirred for 1.0 h. After completion of reaction, water (500.0 mL) was added to the reaction mass by drop wise addition over a period of 30 min at 40-430C. The slurry mass was stirred for 30 min at 400C and then filtered under reduced pressure. The wet material was slurry washed using water (500.0 mL) for 30 min at 400C. The solid was collected by filtration and the material washed with water (125.0 mL). The material was dried under reduced pressure with nitrogen gas bleed at 500C to afford the desired product 4-{l-[3-(trifluoromethyl)[l,2,4]triazolo[4,3-b]pyridazin-6-yl]piperidin-4-yl}phenol (75.1 g, 89.9%) as a free flowing solid. IH NMR (400.13 MHz, DMSO-d6): δ 1.64 (2H, m), 1.87 (2H, m), 2.75 (IH, m), 3.09 (2H, m), 4.40 (2H, m), 6.69 (2H, d), 7.05 (2H, d), 7.65 (IH, d), 8.24 (IH, d), 9.15 (IH, s); m/z = 364 [M+H]+.

Preparation of 6-[4-[4-[2-(4-acetylpiperazin-l-yl)ethoxy]phenyl]piperidin-l-yl]-3- (trifluor omethyl) [ 1 ,2,4] triazolo [4,3-b] pyridazine:

Dichloromethane (225.0 mL) and 4-{l-[3-(trifluoromethyl)[l,2,4]triazolo[4,3-b]pyridazin- 6-yl]piperidin-4-yl} phenol (50.0 g, 138 m mol) were charged to a suitable round bottom flask at 22-25°C. Triphenylphosphine (72.2 g, 275 m mol) and l-[4-(2-hydroxy- ethyl)piperazin-l-yl]ethanone [CAS: 83502-55-0] (47.4 g, 275 m mol) were added successively to the reaction mass and stirred for 10 min at 22-25°C. Di-isopropyl azodicarboxylate (55.65 g, 275 m mol) in dichloromethane (75.0 mL) was added to the reaction mass slowly drop wise at 25-300C over a period of 60-90 min. The resulting reaction mass was stirred for 1.0 h at 25-300C to complete the reaction. n-Heptane (600.0 mL) was introduced to the reaction mass by drop wise addition over a period of 15-30 min at 22-25°C and stirred for 30 min at the same temperature. Thus precipitated solid was filtered and washed with n-heptane (150.0 mL). The material was then suck dried for 30 min under reduced pressure. The crude material was purified by slurry washing in methanol (325.0 mL) at 22-25°C. The solid was then collected by filtration and washed with methanol (50.0 mL). The material was dired under reduced pressure with nitrogen gas bleed at 500C to afford the desired product 6-[4-[4-[2-(4-acetylpiperazin-l- yl)ethoxy]phenyl]piperidin- 1 -yl]-3-(trifluoromethyl)[ 1 ,2,4] triazolo[4,3-b]pyridazine (61.2 g, 84%) as a free flowing solid.

IH NMR (400.13 MHz, DMSO-d6): δ 1.59-1.73 (2H, m), 1.87 (2H, d), 1.99 (3H, s), 2.42 (2H, t), 2.71 (2H, t), 2.76-2.86 (IH, t), 3.08 (2H, t), 3.38-3.47 (4H, m), 4.08 (2H, t), 4.41 (2H, d), 6.88 (2H, d), 7.18 (2H, d), 7.62 (IH, d), 8.26 (IH, d); m/z = 518 [M+H]+.

Example 5.8

Preparation of 6-(4-{4-[2-(4-acetylpiperazin-l-yl)ethoxy]phenyl}piperidin-l-yl)-3-(trifluor omethyl)-7,8-dihydr 0 [1 ,2,4] triazolo [4,3-b] pyridazine maleate

Figure imgf000096_0001

A clear solution of maleic acid (0.445 g, 3.84 m mol) in methanol (1.0 mL) was added to a clear solution of 6-(4-{4-[2-(4-acetylpiperazin-l-yl)ethoxy]phenyl}piperidin-l-yl)-3- (trifluoromethyl)-7,8-dihydro[l,2,4]triazolo[4,3-b]pyridazine, obtained as described in Example 5.5, (2.0 g, 3.84 m mol) in methanol (2.0 mL) at 22-25°C and the resulting clear solution heated to 500C for 30 min. The reaction mass was cooled to 22-25°C and ethylacetate (16.0 mL) added drop wise to the reaction mass at 22-25°C. The reaction mass was then stirred for 60 min at 22-25°C. The resulting white color material was collected by filtration and washed with ethylacetate (5.0 mL). The material was dried under reduced pressure with nitrogen gas bleed at 500C to afford the desired product 6-(4- {4-[2-(4-acetylpiperazin-l-yl)ethoxy]phenyl}piperidin-l-yl)-3-(trifluoromethyl)-7,8- dihydro[l,2,4]triazolo[4,3-b]pyridazine maleate (2.21 g, 90.0%) as free flowing white color material.

IH NMR (400.13 MHz, DMSO-d6): δ 1.62 (2H, m), 1.77 (2H, m), 2.02 (3H, s), 2.75 (IH, m), 2.77 (2H, m), 2.80 (2H, m), 2.95 (4H, m), 3.16 (2H, t), 3.36 (6H, m), 4.22 (4H, m), 6.08 (2H, s), 6.91 (2H, d), 7.17 (2H, d).

PAPER

Bioorg Med Chem Lett. 2013 Apr 1;23(7):1945-8

Discovery of AZD3514, a small-molecule androgen receptor downregulator for treatment of advanced prostate cancer

  • Oncology iMed, AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, UK

 

Removal of the basic piperazine nitrogen atom, introduction of a solubilising end group and partial reduction of the triazolopyridazine moiety in the previously-described lead androgen receptor downregulator 6-[4-(4-cyanobenzyl)piperazin-1-yl]-3-(trifluoromethyl)[1,2,4]triazolo[4,3-b]pyridazine (1) addressed hERG and physical property issues, and led to clinical candidate 6-(4-{4-[2-(4-acetylpiperazin-1-yl)ethoxy]phenyl}piperidin-1-yl)-3-(trifluoromethyl)-7,8-dihydro[1,2,4]triazolo[4,3-b]pyridazine (12), designated AZD3514, that is being evaluated in a Phase I clinical trial in patients with castrate-resistant prostate cancer.

Image for unlabelled figure

http://www.sciencedirect.com/science/article/pii/S0960894X13002321

 

SYNTHESIS

STR1AZD 3514

6-(4-{4-[2-(4-Acetylpiperazin-1-yl)ethoxy]phenyl}piperidin-1-yl)-3-(trifluoromethyl)-7,8-dihydro[1,2,4]triazolo[4,3-b]pyridazine AZD 3514

 

 

 

STR1

 

 

SYNTHETIC ROUTE 2ND GENERATION

STR1

 

 

STR1

SYNTHETIC ROUTE 4TH GENERATION

STR1

 

REFERENCES

1: Bradbury RH, Acton DG, Broadbent NL, Brooks AN, Carr GR, Hatter G, Hayter BR,  Hill KJ, Howe NJ, Jones RD, Jude D, Lamont SG, Loddick SA, McFarland HL, Parveen  Z, Rabow AA, Sharma-Singh G, Stratton NC, Thomason AG, Trueman D, Walker GE, Wells SL, Wilson J, Wood JM. Discovery of AZD3514, a small-molecule androgen receptor downregulator for treatment of advanced prostate cancer. Bioorg Med Chem Lett. 2013 Apr 1;23(7):1945-8. doi: 10.1016/j.bmcl.2013.02.056. Epub 2013 Feb 21. PubMed PMID: 23466225.

 

Some pics, Team at Astrazeneca , Bangalore, INDIA

Vijaykumar Sengodan Chellappan

Vijaykumar Sengodan Chellappan

 

Jagannath V, PMP®

Jagannath V, PMP®

 

Dr. Vidya Nandialath

Associate Research Scientist II at AstraZeneca India Pvt Ltd

Rifahath Mon

Rifahath Mon

Associate Research Scientist at AstraZeneca

Dr Kagita Veera Babu

Route Scouting, Process Design, Technology Transfer, Trouble shooting, QbD, Green Chemistry

Srinivasa Rao Korupoju

Srinivasa Rao Korupoju

Harikrishna Tumma Ph. D.

Harikrishna Tumma Ph. D.

 

Rashmi HV

Anandan Muthusamy

Anandan Muthusamy

Partha Pratim Bishi, PMP®

Partha Pratim Bishi,

Ranga Nc

 

 ASTAZENECA BANGALORE

 

 

///////////////AZD 3514 MALEATE, AZD 3514 , AZD-3514, Prostate cancer, Androgen receptor downregulator, AZD3514, 1240299-33-5

 

Share

Practical Implementation of the Control of Elemental Impurities: EMA’s new Guideline Draft

 regulatory  Comments Off on Practical Implementation of the Control of Elemental Impurities: EMA’s new Guideline Draft
Jul 222016
 

 

One and a half year after its publication, the ICH Q3D guideline still raises many questions. The EMA has recently published a guideline draft aiming at clarifying the practical implementation of ICH Q3D. Read more here about what is expected in a marketing authorisation application or in an application for a CEP with regard to risk assessment and the control of elemental impurities in APIs and medicinal products.

http://www.gmp-compliance.org/enews_05481_Practical-Implementation-of-the-Control-of-Elemental-Impurities-EMA-s-new-Guideline-Draft_15339,15429,15332,S-WKS_n.html

The “ICH Q3D Guideline for Elemental Impurities” was published in December 2014 as Step 4 document and released in August 2015 under No EMA/CHMP/ICH/353369/2013 as EMA’s Scientific Guideline. The guideline came into effect in June 2016 for all medicinal products currently underlying a marketing authorisation procedure (new applications).

In the meantime, it became clear that implementing in practice the requirements of this guideline has been so complex and led to some marketing authorisation procedures being delayed. The ICH has already reacted to the situation and published 7 training modules on its website. Moreover, a concept paper announces a question & answer document.

On 12 July 2016, the draft of an EMA’s guideline entitled “Implementation strategy of ICH Q3D guideline” (EMA/404489/2016) was published. The purpose of the document is to provide support for implementing ICH Q3D in the European context.

The draft comprises three chapters addressing the most important elements in relation with the implementation of the ICH Q3D requirements. The chapter “1. Different approaches to Risk Management” starts describing the two fundamental approaches to the performance of a risk assessment and the justification for a control strategy with regard to elemental impurities:

Drug Product Approach
Here, batches of the finished product are scanned by means of analytical (validated!) procedures to develop a risk-based control strategy. If – with this approach – the omission of a routine testing has to be justified, the authority expects a detailed and valid justification though, and not just analytical data from a few batches.

Component Approach
The guideline draft clearly gives its preference to this approach. The respective contribution of the different components of a medicinal product is considered with respect to the potential total impurity profile and compared to the PDE value from the risk assessment. All potential sources of impurity, for example from production equipment or from excipients of natural (mined) origin have to be considered in this assessment. This particularly applies to outsourced APIs; here, all pieces of information available from Active Substance Master Files (ASMFs) or Certificates of Suitability (CEPs) have to be used. Substances with a Ph.Eur. monograph should always comply with the elemental impurities limits of the corresponding monograph.

The chapter “2. Particulars for Intentionally Added Element(s)” deals with the common practice in many organic syntheses to add elements to increase the specificity of the chemical reaction and the yield. It is particularly critical when the last step of an API synthesis just before the end product uses a metal catalyst. In such a case, the authority expects a convincing evidence that the catalyst is purged to levels consistently below the control threshold (<30% of the PDE) by means of appropriate methods. All details about the API synthesis including the fate of the metals intentionally added have to be consistently described and documented in the marketing authorisation application or in the application for a CEP. If the routine testing of an elemental impurity is needed, the API manufacturer may determine a specification. This information will be required by the medicinal product manufacturer for his overall risk assessment.

The chapter “3. ASMF/CEP: dossier expectations and assessment strategy” explains who has to submit the risk assessment necessary for an ASMF or a CEP and how the dossier will be processed by the assessor of the regulatory authority. Basically, two scenarios are possible:

1. The API manufacturer submits a summary of a risk assessment/management for elemental impurities
Such information flows in the overall risk assessment of the medicinal product manufacturer and is assessed by the quality assessor/ CEP assessor within the marketing authorisation procedure. All data and documents used for the risk assessment should also be available for a GMP inspection.

2. The API manufacturer doesn’t perform any risk assessment/ management.
The regulatory authority basically expects a detailed description of the API synthesis including data on all metal catalysts used. This as well as the analytical routine controls on elemental impurities performed by the API manufacturer will also be assessed by the quality assessor/ CEP assessor. Nevertheless, the assessor won’t make a final conclusion in the ASMF or CEP assessment report with regard to the compliance with ICH Q3D. This will be done within the marketing authorisation procedure for the medicinal product.

The guideline draft can be commented on until 12 August 2016.

///////////ICH Q3D, Control of Elemental Impurities,  EMA, control of elemental impurities in APIs

Share

Elaboration of New USP General Chapter <1220> – Analytical Procedure Lifecycle – announced

 regulatory, USP  Comments Off on Elaboration of New USP General Chapter <1220> – Analytical Procedure Lifecycle – announced
Jul 222016
 

 

On June 24, 2016, the USP announced the elaboration of a new general chapter <1220> regarding life cycle management of analytical methods. Read more about the new general chapter  <1220> “The Analytical Procedure Lifecycle“.

SEE

http://www.gmp-compliance.org/enews_05438_Elaboration-of-New-USP-General-Chapter–1220—-Analytical-Procedure-Lifecycle—announced_15438,15608,Z-PDM_n.html

On June 24, 2016, the USP announced the elaboration of a new general chapter <1220> “The Analytical Procedure Lifecycle”. Input Deadline is July 29, 2016.

The suggested audience are drug product manufacturers, dietary supplement manufacturers, testing organizations, and drug product related regulatory agencies.

“An analytical procedure must be shown to be fit for its intended purpose. It is useful to consider the entire lifecycle of an analytical procedure when approaching development of the procedure, i.e. its design, development, qualification, and continued verification. The current concepts of validation, verification, and transfer of procedures address portions of the lifecycle but do not consider them holistically. This General Chapter intends to more fully address the entire procedure lifecycle and define concepts which may be useful.”

The approach is consistent with the concepts of Quality by Design (QbD) as described in ICH Guidelines Q8 (R2), 9, 10, and 11 and with the expected new ICH Guideline Q12 (Lifecycle Management).

Preliminary outline:
THE LIFECYCLE APPROACH

  • focal point: Analytical target profile (ATP), comparable to the Quality Target Product Profile (QTPP).

STAGE 1: PROCEDURE DESIGN, DEVELOPMENT, AND UNDERSTANDING

  • Procedure design and development,
  • Procedure understanding,
  • Preparing for qualification.

STAGE 2: PROCEDURE PERFORMANCE QUALIFICATION

STAGE 3: IMPLEMENTATION AND CONTINUED PROCEDURE PERFORMANCE VERIFICATION

  • Routine monitoring,
  • Analytical control strategy,
  • Knowledge management,
  • Change control.

Anticipated proposed design phase activities:

Two Stimuli articles are scheduled for PF 42(5) [Sep.–Oct. 2016]:

  • Analytical Target Profile: Structure and Application throughout the Analytical Lifecycle,
  • Analytical Control Strategy.

Two stimuli articles have already been published:

  • Lifecycle Management of Analytical Procedures: Method Development, Procedure Performance Qualification, and Procedure Performance Verification. PF 39(5) [Sep.–Oct. 2013],
  • Fitness for Use: Decision Rules and Target Measurement Uncertainty. PF 42(2) [Mar.–Apr. 2016].

Additionally, the USP proposed a revision of general chapter <1225> “Validation of compendial procedures” in PF 42(2) [March-April 2016].
This chapter is being revised to incorporate a section on “Lifecycle Management of Analytical Procedures”. The revision is an attempt to better align the validation concept with the recently (July 2015) issued FDA guidance “Analytical Procedures and Methods Validation for Drugs and Biologics”, which also includes a section on “Life Cycle Management of Analytical Procedures”.

Estimated proposal for the new general chapter <1220> “The Analytical Procedure Lifecycle” is PF 43(1) [Jan.–Feb. 2017].

Furthermore, an USP and ECA Joint Conference and Workshop on Lifecycle Approach of Analytical Procedures will be held November 8-9, 2016 in Prague, Czech Republic.

For more information please visit the USP website – Notices- General Chapter Prospectus – The Analytical Procedure Lifecycle.

 

 

//////////The Analytical Procedure Lifecycle, USP, chapter <1220>

Share

Sustained Local Delivery of Structurally Diverse HIV-1 Microbicides Released from Sublimation Enthalpy Controlled Matrices

 Uncategorized  Comments Off on Sustained Local Delivery of Structurally Diverse HIV-1 Microbicides Released from Sublimation Enthalpy Controlled Matrices
Jul 212016
 

Simi Gunaseelan Ph.D  Author

Assistant Professor of Pharmaceutical Sciences, The University of Texas at Tyler, TEXAS, USA

S. Gunaseelan : R. Maskiewicz (*)

Department of Pharmaceutical Sciences( School of Pharmacy Loma Linda University 11175 Campus Street, Chan Shun Pavilion 21018 Loma Linda, California 92350, USA e-mail: rmaskiewicz@llu.edu

Purpose

Use of coital-dependent products to prevent HIV-1 transmission has resulted in mixed success. We hypothesize that incorporation of antiviral drug candidates into a novel controlled delivery system will prolong their activity, making their use coital independent, thus increasing their chance of prophylactic success.

Methods

Tenofovir, emtricitabine, and C5A peptide HIV microbicides were mechanically incorporated into matrices comprising a series of subliming solids. Matrix sublimation rates and drug release rates were measured in three in vitro and one in vivo environments intended to model human vaginal interior. Antiviral activity studies evaluating matrix incorporated microbicides were performed using in vitro cell cultures and human ectocervical explants.

Results

Drug release rates were identical to matrix sublimation rates, and were zero order. Differences in matrix material sublimation enthalpies determined drug release and matrix erosion rates in a thermodynamically definable manner, in vitro and in vivo. Durations of release ranging from several days to several months were readily achieved. Prolonged duration of anti HIV-1 activity was shown for matrix incorporated microbicides, using ectocervical explant and cell culture models of HIV-1 infection.

Conclusion

Subliming solid matrices show promise as a delivery system providing multi month intravaginal release of a wide range of HIV-1 microbicides.

 

Sustained Local Delivery of Structurally Diverse HIV-1 Microbicides Released from Sublimation Enthalpy Controlled Matrices

Simi Gunaseelan,1 Philippe A. Gallay,2 Michael D. Bobardt,2 Charlene S. Dezzutti,3,4 Timothy Esch,3 and Richard Maskiewiczcorresponding author1

1Department of Pharmaceutical Sciences, School of Pharmacy Loma Linda University, 11175 Campus Street, Chan Shun Pavilion 21018, Loma Linda, California 92350 USA
2Department of Immunology and Microbial Science, IMM-9 The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037 USA
3Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, Pennsylvania 15213 USA
4Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 204 Craft Avenue, Pittsburgh, Pennsylvania 15213 USA
Richard Maskiewicz, Phone: +1-909-5589473, Fax: +1-909-5580446, ude.ull@zciweiksamr.
corresponding authorCorresponding author.

//////////enthalpically controlled release, HIV-1, intravaginal delivery, prolonged antiviral effect, subliming solid matrix,

Share

Lobeglitazone sulfate (Duvie)

 FDA 2014, Uncategorized  Comments Off on Lobeglitazone sulfate (Duvie)
Jul 212016
 

 

 

STR1

 

Lobeglitazone.svg

Lobeglitazone Sulfate, CKD-501, IDR-105

(Duvie®)Approved KOREA

Chong Kun Dang (Originator)

Adjunct to diet and exercise to improve glycemic control in adults with type 2 Diabetes mellitus

A dual PPARα and PPARγ agonist used to treat type 2 diabetes.

Trade Name:Duvie®MOA:Dual PPARα and PPARγ agonistIndication:Type 2 diabetes

CAS No. 607723-33-1(FREE)

CAS 763108-62-9(Lobeglitazone Sulfate)

2,4-Thiazolidinedione, 5-((4-(2-((6-(4-methoxyphenoxy)-4- pyrimidinyl)methylamino)ethoxy)phenyl)methyl)-, sulfate (1:1);

Duvie Tab.

  • Developer Chong Kun Dang; EQUIS & ZAROO
  • Class Antihyperglycaemics; Pyrimidines; Small molecules; Thiazolidinediones
  • Mechanism of Action Peroxisome proliferator-activated receptor alpha agonists; Peroxisome proliferator-activated receptor gamma agonists
  • MarketedType 2 diabetes mellitus
  • Most Recent Events

    • 01 May 2016Chong Kun Dang Pharmaceutical completes two phase I drug-interaction trials in Healthy volunteers in South Korea (PO) (NCT02824874; NCT02827890)
    • 01 Apr 2016Chong Kun Dang Pharmaceutical initiates two phase I drug-interaction trials in Healthy volunteers in South Korea (PO) (NCT02824874; NCT02827890)
    • 01 Mar 2016Chong Kun Dang completes a phase I pharmacokinetic trial in Impaired hepatic function in Healthy volunteers in South Korea, NCT02007941)
    • Lobeglitazone sulfate was approved by the Ministry of Food and Drug Safety (Korea) on July 4, 2013. It was developed and marketed as Duvie® by Chong Kun Dang Corporation.Lobeglitazone is an agonist for both PPARα and PPARγ, and it works as an insulin sensitizer by binding to the PPAR receptors in fat cells and making the cells more responsive to insulin. It is indicated as an adjunct to diet and exercise to improve glycemic control in adults with type 2 diabetes.Duvie® is available as tablet for oral use, containing 0.5 mg of free Lobeglitazone. The recommended dose is 0.5 mg once daily.

 

 

Lobeglitazone sulfate.png

Lobeglitazone (trade name Duvie, Chong Kun Dang) is an antidiabetic drug in the thiazolidinedione class of drugs. As an agonistfor both PPARα and PPARγ, it works as an insulin sensitizer by binding to the PPAR receptors in fat cells and making the cells more responsive to insulin.[3]

Chong Kun Dang

STR1

Lobeglitazone sulfate was approved by the Ministry of Food and Drug Safety (Korea) on July 4, 2013. It was developed and marketed as Duvie® by Chong Kun Dang Corporation.

Lobeglitazone is an agonist for both PPARα and PPARγ, and it works as an insulin sensitizer by binding to the PPAR receptors in fat cells and making the cells more responsive to insulin. It is indicated as an adjunct to diet and exercise to improve glycemic control in adults with type 2 diabetes.

Duvie® is available as tablet for oral use, containing 0.5 mg of free Lobeglitazone. The recommended dose is 0.5 mg once daily.

Lobeglitazone which was reported in our previous works belongs to the class of potent PPARα/γ dual agonists (PPARα EC50:  0.02 μM, PPARγ EC50:  0.018 μM, rosiglitazone; PPARα EC50:  >10 μM, PPARγ EC50:  0.02 μM, pioglitazone PPARα EC50:  >10 μM, PPARγ EC50:  0.30 μM). Lobeglitazone has excellent pharmacokinetic properties and was shown to have more efficacious in vivo effects in KKAy mice than rosiglitazone and pioglitazone.17 Due to its outstanding pharmacokinetic profile, lobeglitazone was chosen as a promising antidiabetes drug candidate.

Medical uses

Lobeglitazone is used to assist regulation of blood glucose level of diabetes mellitus type 2 patients. It can be used alone or in combination with metformin.[4]

Lobeglitazone was approved by the Ministry of Food and Drug Safety (Korea) in 2013, and the postmarketing surveillance is on progress until 2019.[4][5]

SYNTHESIS

STR1

 

Chong Kun Dang’s Modcol Flu Dry Syrup is released in four different versions: All-Day, Night, Nose and Cough. [CHONG KUN DANG]

 

STR1

 

PAPER

Org. Process Res. Dev. 2007, 11, 190-199.

Process Development and Scale-Up of PPAR α/γ Dual Agonist Lobeglitazone Sulfate (CKD-501)

Process Research and Development Laboratory, Chemical Research Group, Chong Kun Dang Pharmaceutical Cooperation, Cheonan P. O. Box 74, Cheonan 330-831, South Korea, and Department of Chemistry, Korea University, 5-1-2, Anam-Dong, Seoul 136-701, Korea
Org. Process Res. Dev., 2007, 11 (2), pp 190–199
DOI: 10.1021/op060087u

http://pubs.acs.org/doi/abs/10.1021/op060087u

Abstract Image

A scaleable synthetic route to the potent PPARα/γ dual agonistic agent, lobeglitazone (1), used for the treatment of type-2 diabetes was developed. The synthetic pathway comprises an effective five-step synthesis. This process involves a consecutive synthesis of the intermediate, pyrimidinyl aminoalcohol (6), from the commercially available 4,6-dichloropyrimidine (3) without the isolation of pyrimidinyl phenoxy ether (4). Significant improvements were also made in the regioselective 1,4-reduction of the intermediate, benzylidene-2,4-thiazolidinedione (10), using Hantzsch dihydropyridine ester (HEH) with silica gel as an acid catalyst. The sulfate salt form of lobeglitazone was selected as a candidate compound for further preclinical and clinical study. More than 2 kg of lobeglitazone sulfate (CKD-501, 2) was prepared in 98.5% purity after the GMP batch. Overall yield of 2 was improved to 52% from 17% of the original medicinal chemistry route.

Silica gel TLC Rf = 0.35 (detection:  iodine char chamber, ninhydrin solution, developing solvents:  CH2Cl2/MeOH, 20:1); mp 111.4 °C; IR (KBr) ν 3437, 3037, 2937, 2775, 1751, 1698, 1648, 1610, 1503, 1439, 1301, 1246, 1215, 1183 cm-1;

1H NMR (400 MHz, CDCl3) δ 3.09 (m, 4H), 3.29 (m, 1H), 3.76 (s, 3H), 3.97 (m, 2H), 4.14 (m, 2H), 4.86 (m, 1H), 6.06 (bs, 1H), 6.86 (m, 2H), 7.00 (m, 2H), 7.13 (m, 4H), 8.30 (s, 1H), 11.99 (s, NH);

13C NMR (100 MHz, CDCl3) δ 37.1, 38.2, 53.7, 53.8, 56.3, 62.2, 65.8, 86.0, 115.1, 116.0, 123.0, 129.8, 131.2, 145.7, 153.4, 157.9, 158.1, 161.1, 166.5, 172.4, 172.5, 176.3, 176.5;

MS (ESI)m/z (M + 1) 481.5; Anal. Calcd for C24H26N4O9S2:  C, 49.82; H, 4.53; N, 9.68; S, 11.08. Found:  C, 49.85; H, 4.57; N, 9.75; S, 11.15.

PATENT

WO03080605A1.

 

Clip
Lobeglitazone sulfate (Duvie) Lobeglitazone sulfate, an oral peroxisome proliferator-activated receptor (PPARa/c) dual agonist with IC50 = 20 and 18 nM respectively, was developed by Chong Kun Dang Pharmaceutical in Korea for the treatment of diabetes.135 This drug is differentiated from two other PPAR agonists available—pioglitazone and rosiglitazone —which lack PPARa activity.135 The most likely processscale preparation of lobeglitazone sulfate follows the route described in a process communication from Chong Kun Dang Pharmaceutical.136

Commercially available 4,6-dichloropyrimidine (152) was treated with a stoichiometric equivalent of p-methoxyphenol (153) in the presence of KF in warm DMF (Scheme 24). Upon completion of this reaction, 2-methylaminoethanol was added to the mixture to provide pyrimidine 154 in high yield.137

Next, alcohol 154 underwent a substitution reaction with p-fluorobenzaldehyde (155) under basic conditions to provide alkoxy benzaldehyde 156 which was converted to the benzylidene thiazolidindione 158 upon subjection to Knoevenagel conditions with 2,4-thiazolidinedione (157) in 90% yield.

Finally, reduction of olefin 158 was facilitated by treatment with the Hantzsch ester (159) in the presence of silica gel followed by treatment with methanolic sulfuric acid (96%) at low temperature to ultimately furnish lobeglitazone sulfate in 90% yield.

STR1
135. Jin, S. M.; Park, C. Y.; Cho, Y. M.; Ku, B. J.; Ahn, C. W.; Cha, B.-S.; Min, K. W.;Sung, Y. A.; Baik, S. H.; Lee, K. W.; Yoon, K.-H.; Lee, M.-K.; Park, S. W. Diab.Obes. Metab. 2015, 17, 599.
136. Lee, H. W.; Ahn, J. B.; Kang, S. K.; Ahn, S. K.; Ha, D.-C. Org. Process Res. Dev.2007, 11, 190.
137. Lee, H. W.; Kim, B. Y.; Ahn, J. B.; Kang, S. K.; Lee, J. H.; Shin, J. S.; Ahn, S. K.; Lee,S. J.; Yoon, S. S. Eur. J. Med. Chem. 2005, 40, 862.

 

References

  1. Lee JH, Noh CK, Yim CS, Jeong YS, Ahn SH, Lee W, Kim DD, Chung SJ. (2015). “Kinetics of the Absorption, Distribution, Metabolism, and Excretion of Lobeglitazone, a Novel Activator of Peroxisome Proliferator-Activated Receptor Gamma in Rats.”.Journal of Pharmaceutical sciences 104 (9): 3049–3059.doi:10.1002/jps.24378. PMID 25648999.
  2.  Kim JW, Kim JR, Yi S, Shin KH, Shin HS, Yoon SH, Cho JY, Kim DH, Shin SG, Jang IJ, Yu KS. (2011). “Tolerability and pharmacokinetics of lobeglitazone (CKD-501), a peroxisome proliferator-activated receptor-γ agonist: a single- and multiple-dose, double-blind, randomized control study in healthy male Korean subjects.”. Clinical therapeutics 33 (11): 1819–1830.doi:10.1016/j.clinthera.2011.09.023. PMID 22047812.
  3.  Lee JH, Woo YA, Hwang IC, Kim CY, Kim DD, Shim CK, Chung SJ. (2009). “Quantification of CKD-501, lobeglitazone, in rat plasma using a liquid-chromatography/tandem mass spectrometry method and its applications to pharmacokinetic studies.”. Journal of Pharmaceutical and Biomedical Analysis 50 (5): 872–877.doi:10.1016/j.jpba.2009.06.003. PMID 19577404.
  4.  “MFDS permission information of Duvie Tablet 0.5mg”(Release of Information). Ministry of Food and Drug Safety. Retrieved2014-10-23.
  5.  “국내개발 20번째 신약‘듀비에정’허가(20th new drug developed in Korea ‘Duvie Tablet’ was approved)”. Chong Kun Dang press release. 2013-07-04. Retrieved 2014-10-23.
Lobeglitazone
Lobeglitazone.svg
Systematic (IUPAC) name
5-[(4-[2-([6-(4-Methoxyphenoxy)pyrimidin-4-yl]-methylamino)ethoxy]phenyl)methyl]-1,3-thiazolidine-2,4-dione
Clinical data
Trade names Duvie
Routes of
administration
Oral
Legal status
Legal status
Pharmacokinetic data
Protein binding >99%[1]
Metabolism liver (CYP2C9, 2C19, and 1A2)[1]
Biological half-life 7.8–9.8 hours[2]
Identifiers
CAS Number 607723-33-1
PubChem CID 9826451
DrugBank DB09198 Yes
ChemSpider 8002194
Synonyms CKD-501
Chemical data
Formula C24H24N4O5S
Molar mass 480.53616 g/mol

Identifications:

1H NMR (Estimated) for Lobeglitazone

Experimental: 1H NMR (400 MHz, CDCl3) δ 3.12 (m, 4H), 3.45 (m, 1H), 3.83 (s, 3H), 4.00 (m, 2H), 4.16 (m, 2H), 4.50 (m, 1H), 5.84 (bs, 1H), 6.83 (m, 2H), 7.06 (m, 2H), 7.15 (m, 2H), 8.31 (s, 1H), 8.89 (bs, NH).

///Lobeglitazone Sulfate, CKD-501, Duvie®,  Approved KOREA, Chong Kun Dang, A dual PPARα and PPARγ agonist , type 2 diabetes, CKD 501, 763108-62-9, 607723-33-1, IDR-105

CN(CCOC1=CC=C(C=C1)CC2C(=O)NC(=O)S2)C3=CC(=NC=N3)OC4=CC=C(C=C4)OC.OS(=O)(=O)O

Share

Novel Intravaginal Delivery of Antiretroviral-based Microbicides for HIV prevention

 Uncategorized  Comments Off on Novel Intravaginal Delivery of Antiretroviral-based Microbicides for HIV prevention
Jul 212016
 

view above or
See ppt at

Biography

Gunaseelan’s research expertise is in drug delivery. For the past 10 years she has been working towards developing ‘Novel Drug Delivery Systems’ for HIV Prevention & HIV and Cancer Therapeutics. Her research works resulted in 5 patents, more than 20 publications in high-impact journals and presentations in 20 national and international conferences. Her dedication towards research work at Rutgers University School of Pharmacy New Jersey, led her to be a recipient of Merit Award for 3 consecutive years. She is currently a journal reviewer for Advanced Drug Delivery Reviews, Pharmaceutical Research, and Controlled Release Society Meeting Abstracts

STR1

Abstract

Objectives: Microbicides, products applied vaginally or rectally, are effective at preventing HIV transmission. However, many products (e.g., peptides, antiretroviral drugs) are reactive or incompatible in the existing diffusion/hydrolysis/dissolution based delivery systems. To overcome the issues of extended delivery and product compatibility, the use of a novel subliming solid matrix-based delivery system is described here. Methods: The microbicides C5A, tenofovir fumarate, emtricitabine, dapivarine, UC-781 and IQP0528 were employed as representatives of a range of molecular structures and physicochemical properties. Hydrophobic, chemically inert subliming solid matrices, utilized for microbicide formulations and achieving a defined range of sustained release rates, included norbornane, hexamethylcyclotrisiloxane, perfluoroundecane, perfluorododecane and cyclododecane. Rates of matrix sublimation and concomitant microbicide release were determined in vitro. Formulations were tested for cellular toxicity, and durations of anti-HIV-1 activity by constant release of microbicides from the sublimable matrices. Results: Subliming solid matrices release microbicides by surface erosion achieved through sublimation. Zero order sustained microbicide release was achieved in vitro, at rates independent of microbicide structures and properties, and controlled exclusively by sublimation enthalpies of each hydrophobic matrix material. The matrices provided prolongation of anti-HIV-1 activity relative to bolus microbicide administration, when evaluated in cultured human ectocervical tissue, macrophages, and TZM reporter cells. No evidence of matrix toxicity was observed after continuous exposure to macrophages, T-lymphocytes, PBMC cells and ectocervical explants. Implications: Subliming matrices offer unique attributes that will allow steady-state delivery of any microbicide, over durations ranging from weeks to months, by employing, simple, stable, and readily available matrix materials, suggesting novel delivery capabilities.

Speaker Presentations

 

Share

Synmr Chemicals Pvt Ltd, the first manufacturers of NMR Solvents in India

 companies, spectroscopy, SYNTHESIS  Comments Off on Synmr Chemicals Pvt Ltd, the first manufacturers of NMR Solvents in India
Jul 202016
 
 
“Synmr Chemicals Pvt Ltd” (Formerly known as Synovation Chemicals Pvt Ltd) are the first manufacturers of NMR Solvents in India. This would benefit the Indian Science community as they no longer would have to depend on Imports, high pricing and uncertain supply.
Please do assist “Synmr” and promote our “Make in India” endeavor


Synmr Chemicals Pvt Ltd (Previously known as
Synovation Chemicals and Sourcing Pvt Ltd) is engaged in the manufacture of NMR
(Deuterated solvents).
With permission of Heavy Water Board, we can now
offer NMR Solvents manufactured in India.
 
They are the first manufactures of NMR solvents in
India and the following products have been developed and up scaled.
 
1. Chloroform D 99.8%
2.   DMSO D6 99.8%
3.   Methyl Iodide D3 99.5%
4.   Acetone D6 99.8%
5.   Acetonitrile D3 99.8%
In the Pipeline
·
Methanol
D4
·
Ethanol
D6
 





















We kindly request you to send your enquiries to

 
Suresh R Iyer
suresh@synmr.in           Contact Number +9193212 58158
dinesh@synmr.in           Contact Number +9198454 04105
Dr Sankar Iyer       sankar@synovationchemicals.in       +91 94490 63877  
Website is
www.synmr.in

nmr@synovationchemicals.in

Promote our NMR solvents and thus encourage MAKE IN INDIA.

 

 

Thanks 
Regards….?
Suresh R Iyer

————————————————————————————————————————

Team

 

INFO FROM LITERATURE OR NET

//////

Share

Eliglustat tartrate (Cerdelga) エリグルスタット酒石酸塩 依利格鲁司特 エリグルスタット,サーデルガ

 FDA 2014, Uncategorized  Comments Off on Eliglustat tartrate (Cerdelga) エリグルスタット酒石酸塩 依利格鲁司特 エリグルスタット,サーデルガ
Jul 192016
 

Eliglustat tartrate (Cerdelga) エリグルスタット酒石酸塩

依利格鲁司特

エリグルスタット,サーデルガ

FOR TREATMENT OF GAUCHERS DISEASE

ELIGLUSTAT; Cerdelga; Genz 99067; Genz-99067; UNII-DR40J4WA67; GENZ-112638;

CAS 491833-29-5 FREE FORM

Molecular Formula: C23H36N2O4
Molecular Weight: 404.54294 g/mol

N-[(1R,2R)-1-(2,3-dihydro-1,4-benzodioxin-6-yl)-1-hydroxy-3-pyrrolidin-1-ylpropan-2-yl]octanamide

N-[(1R,2R)-1-(2,3-dihydro-1,4-benzodioxin-6-yl)-1-hydroxy-3-pyrrolidin-1-ylpropan-2-yl]octanamide;(2R,3R)-2,3-dihydroxybutanedioic acid
Mechanism of Action: glucosylceramide synthase inhibitor
Indication: Type I Gaucher Disease
Date of Approval: August 19, 2014 (US)

US patent number:US6916802 , US7196205 , US7615573
Patent Expiration Date: Apr 29, 2022 (US6916802, US7196205, US7615573)
Exclusivity Expiration Date:Aug 19, 2019(NCE), Aug 19, 2021 (ODE)
Originator:University of Michigan
Developer: Genzyme, a unit of Sanofi

Eliglustat, marketed by Genzyme as CERDELGA, is a glucosylceramide synthase inhibitor indicated for the long-term treatment of type 1 Gaucher disease. Patients selected for treatment with Eliglustat undergo an FDA approved genotype test to establish if they are CYP2D6 EM (extensive metabolizers), IM (intermediate metabolizers), or PM (poor metabolizers), as the results of this test dictate the dosage of Eliglustat recommended. Eliglustat was approved for use by the FDA in August 2014.

Eliglustat (INN, USAN;[1] trade name Cerdelga) is a treatment for Gaucher’s disease developed by Genzyme Corp that was approved by the FDA August 2014.[2] Commonly used as the tartrate salt, the compound is believed to work by inhibition ofglucosylceramide synthase.[3][4] According to an article in Journal of the American Medical Association the oral substrate reduction therapy resulted in “significant improvements in spleen volume, hemoglobin level, liver volume, and platelet count” in untreated adults with Gaucher disease Type 1.[5]

Cerdelga, capsule, 84 mg/1, oralGenzyme Corporation, 2014-09-03, Us

ELIGLUSTAT.pngELIGLUSTAT

ChemSpider 2D Image | Eliglustat tartrate | C50H78N4O14

Eliglustat tartrate

  • Molecular FormulaC50H78N4O14
  • Average mass959.173 Da
  • UNII-N0493335P3
  • Butanedioic acid, 2,3-dihydroxy-, (2R,3R)-, compd. with N-[(1R,2R)-2-(2,3-dihydro-1,4-benzodioxin-6-yl)-2-hydroxy-1-(1-pyrrolidinylmethyl)ethyl]octanamide (1:2)
  •  eliglustat hemitartrate
  •  eliglustat L-tartrate

CAS 928659-70-5

 

CERDELGA (eliglustat) capsules contain eliglustat tartrate, which is a small molecule inhibitor of glucosylceramide synthase that resembles the ceramide substrate for the enzyme, with the chemical name N-((1R,2R)-1-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-1- hydroxy-3-(pyrrolidin-1-yl)propan-2-yl)octanamide (2R,3R)-2,3-dihydroxysuccinate. Its molecular weight is 479.59, and the empirical formula is C23H36N2O4+½(C4H6O6) with the following chemical structure:

 

CERDELGA (eliglustat) Structural Formula Illustration

Each capsule of CERDELGA for oral use contains 84 mg of eliglustat, equivalent to 100 mg of eliglustat tartrate (hemitartrate salt). The inactive ingredients are microcrystalline cellulose, lactose monohydrate, hypromellose and glyceryl behenate, gelatin, candurin silver fine, yellow iron oxide, and FD&C blue 2.

Cost

In 2014, the annual cost of Cerdelga hard gelatin capsules taken orally twice a day was $310,250. Genzyme’s flagship Imiglucerase(brand name Cerezyme) cost about $300,000 for the infusions if taken twice a month.[6] Manufacturing costs for Cerdelga are slightly lower than for Cerezyme. Genzyme’s maintains higher prices for orphan drugs—most often paid for by insurers— in order to remain financially sustainable.[6]

Chemically Eliglustat is named N-[(1 R,2R)-2-(2,3-dihydro-1 ,4-benzodioxin-6-yl)-2-hydroxy-1 -(1 -pyrrolidinylmethyl)ethyl]-Octanamide(2R!3R)-2,3-dihydroxybutanedioate and the hemitartarate salt of eliglustat has the structural formula as shown in Formula I.

Formula I

Eliglustat hemitartrate (Genz-1 12638), currently under development by Genzyme, is a glucocerebroside (glucosylceramide) synthase inhibitor for the treatment of Gaucher disease and other lysosomal storage disorders. Eliglustat hemitartrate is orally active with potent effects on the primary identified molecular target for type 1 Gaucher disease and other glycosphingolipidoses, appears likely to fulfill high expectations for clinical efficacy. Gaucher disease belongs to the class of lysosomal diseases known as glycosphingolipidoses, which result directly or indirectly from the accumulation of glycosphingolipids, many hundreds of which are derived from glucocerebroside. The first step in glycosphingolipid biosynthesis is the formation of glucocerebroside, the primary storage molecule in Gaucher disease, via glucocerebroside synthase (uridine diphosphate [UDP] – glucosylceramide glucosyl transferase). Eliglustat hemitartrate is based on improved inhibitors of glucocerebroside synthase, and is currently under development by Genzyme.

U.S. patent No. 7,196,205 discloses a process for the preparation of Eliglustat or a pharmaceutically acceptable salt thereof.

U.S. patent No. 6855830, 7265228, 7615573, 7763738, 8138353, U.S. patent application publication No. 2012/296088 discloses process for preparation of Eliglustat and intermediates thereof.

U.S. patent application publication No. 2013/137743 discloses (i) a hemitartrate salt of Eliglustat, (ii) a hemitartrate salt of Eliglustat, wherein at least 70% by weight of the salt is crystalline, (iii) a hemitartrate salt of Eliglustat, wherein at least 99% by weight of the salt is in a single crystalline form.

It has been disclosed earlier that the amorphous forms in a number of drugs exhibit different dissolution characteristics and in some cases different bioavailablity patterns compared to crystalline forms [Konne T., Chem pharm Bull., 38, 2003(1990)]. For some therapeutic indications one bioavailabihty pattern may be favoured over another. An amorphous form of Cefuroxime axetil is a good example for exhibiting higher bioavailability than the crystalline form.

 

CLIP

Eliglustat tartrate, developed and marketed by Genzyme Corporation (a subsidiary of Sanofi), was approved by the US FDA in August 2014 for the treatment of nonneuropathic (type 1) Gaucher disease (GD1) in both treatment-naïve and treatment-experienced adult patients.98

It is the first oral treatment to be approved for first-line use in patients with Gaucher disease type 1, which is a rare lysosomal storage disease characterized by accumulation of lipid glucosylceramide (GL-1) due to insufficient production of the enzyme glucosylceramidase.99,100

Clinical complications include hepatosplenomegaly, anemia, thrombocytopenia, and bone involvement.101 Eliglustat is a specific inhibitor of glucosylceramide synthase with an IC50 of 10 ng/mL and acts as substrate reduction therapy for GD1;102 it has demonstrated non-inferiority to enzyme replacement therapy, which is the current standard of care, in Phase III trials.99

While the process-scale route has not yet been disclosed,103 the largest scale route to eliglustat tartrate reported to date is described in Scheme 15.104

Condensation of commercially available S-(+)-2-phenyl glycinol (87) with phenyl bromoacetate (88) in acetonitrile in the presence of N,N-diisopropylethylamine (DIPEA) provided morpholin-2-one 89 upon treatment with HCl.Neutralization with NaHCO3 followed by coupling with aldehyde 90 in refluxing EtOAc/toluene yielded oxazine adduct 91, which was isolated as a precipitate from methyl-tert-butyl ether (MTBE).

The stereochemistry of the three new stereocenters in 91 can be rationalized through the cycloaddition of an ylide intermediate in the sterically-preferred S-configuration (generated by the reaction of the morpholinone 89 with aldehyde 90) with a second equivalent of the aldehyde. With the morpholinone in a chair conformation in which the phenyl group is equatorial, endo axial approach of the dipolarophile to the less-hindered face of the ylide and subsequent ring flip of the morpholinone ring to a boat conformation positions all exocyclic aryl substituents in a pseudoequatorial configuration. 105

Opening of oxazine 91 with pyrrolidine in refluxing THF followed by addition of HCl in refluxing MeOH gave amide 92, which was reduced to amine 93 using LiAlH4 in refluxing THF.

Subsequent hydrogenation with Pd(OH)2 in EtOH cleaved the phenylethanol group to give the free amine, which was converted to dioxalate salt 94 by treatment with oxalic acid in methyl isobutylketone (MIBK). Subjection of aminoethanol 94 to aqueous sodium hydroxide followed by coupling with palmitic acid Nhydroxysuccinimide (NHS)-ester (95) gave eliglustat as the corresponding freebase (96) in 9.5% overall yield from 87.

Salt formation with L-tartaric acid (0.5 equiv) then provided eliglustat tartrate (XII).106

STR1

STR1

98. http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm410585.htm.
99. Poole, R. M. Drugs 2014, 74, 1829.
100. Kaplan, P. Res. Rep. Endocr. Disord. 2014, 4, 1.
101. Pastores, G. M.; Hughes, D. Clin. Invest. 2014, 4, 45.
102. Shayman, J. A. Drugs Future 2010, 35, 613.
103. Javed, I.; Dahanukar, V. H.; Oruganti, S.; Kandagatla, B. WO Patent2,015,059,679, 2015.
104. Hirth, B.; Siegel, C. WO Patent 2,003,008,399, 2003.
105. Anslow, A. S.; Harwood, L. M.; Phillips, H.; Watkin, D.; Wong, L. F. Tetrahedron:Asymmetry 1991, 2, 1343.
106. Liu, H.; Willis, C.; Bhardwaj, R.; Copeland, D.; Harianawala, A.; Skell, J.;Marshall, J.; Kochling, J.; Palace, G.; Peterschmitt, J.; Siegel, C.; Cheng, S. WO Patent 2,011,066,352, 2011.

CLIP

TAKEN FROM

http://www.xinbiaopin.com/a/zuixindongtai/huaxuepinshuju/2015/0310/2383.html

str1

Nmr predict

N-[(1R,2R)-1-(2,3-dihydro-1,4-benzodioxin-6-yl)-1-hydroxy-3-pyrrolidin-1-ylpropan-2-yl]octanamide NMR spectra analysis, Chemical CAS NO. 491833-29-5 NMR spectral analysis, N-[(1R,2R)-1-(2,3-dihydro-1,4-benzodioxin-6-yl)-1-hydroxy-3-pyrrolidin-1-ylpropan-2-yl]octanamide H-NMR spectrum

13 C NMR

N-[(1R,2R)-1-(2,3-dihydro-1,4-benzodioxin-6-yl)-1-hydroxy-3-pyrrolidin-1-ylpropan-2-yl]octanamide NMR spectra analysis, Chemical CAS NO. 491833-29-5 NMR spectral analysis, N-[(1R,2R)-1-(2,3-dihydro-1,4-benzodioxin-6-yl)-1-hydroxy-3-pyrrolidin-1-ylpropan-2-yl]octanamide C-NMR spectrum

CAS NO. 491833-29-5, N-[(1R,2R)-1-(2,3-dihydro-1,4-benzodioxin-6-yl)-1-hydroxy-3-pyrrolidin-1-ylpropan-2-yl]octanamide

C-NMR spectral analysis

 

str1

 

str1

PATENT

http://www.google.com/patents/WO2013059119A1?cl=en

Figure imgf000024_0001

http://www.google.com/patents/US7196205

Compound 7

(1R,2R)-Nonanoic acid[2-(2′,3′-dihydro-benzo[1,4]dioxin-6′-yl)-2-hydroxy-1-pyrrolidin-1-ylmethyl-ethyl]-amide

Figure US07196205-20070327-C00026

This compound was prepared by the method described for Compound 6 using Nonanoic acid N-hydroxysuccinimide ester. Analytical HPLC showed this material to be 98.4% pure. mp 74–75° C.

1H NMR (CDCl3) δ 6.86–6.76 (m, 3H), 5.83 (d, J=7.3 Hz, 1H), 4.90 (d, J=3.3 Hz, 1H), 4.24 (s, 4H), 4.24–4.18 (m, 1H), 2.85–2.75 (m, 2H), 2.69–2.62 (m, 4H), 2.10 (t, J=7.3 Hz, 2H), 1.55–1.45 (m, 2H), 1.70–1.85 (m, 4H), 1.30–1.15 (m, 10H), 0.87 (t, J=6.9 Hz, 3H) ppm.

Intermediate 4(1R,2R)-2-Amino-1-(2′,3′-dihydro-benzo[1,4]dioxin-6′-yl)-3-pyrrolidin-1-yl-propan-1-ol

Figure US07196205-20070327-C00023

Intermediate 3 (5.3 g, 13.3 mmol) was dissolved in methanol (60 mL). Water (6 mL) and trifluoroacetic acid (2.05 m/L, 26.6 mmol, 2 equivalents) were added. After being placed under nitrogen, 20% Palladium hydroxide on carbon (Pearlman’s catalysis, Lancaster or Aldrich, 5.3 g) was added. The mixture was placed in a Parr Pressure Reactor Apparatus with glass insert. The apparatus was placed under nitrogen and then under hydrogen pressure 110–120 psi. The mixture was stirred for 2–3 days at room temperature under hydrogen pressure 100–120 psi. The reaction was placed under nitrogen and filtered through a pad of celite. The celite pad was washed with methanol (100 mL) and water (100 mL). The methanol was removed by rotoevaporation. The aqueous layer was washed with ethyl acetate three times (100, 50, 50 mL). A 10 M NaOH solution (10 mL) was added to the aqueous layer (pH=12–14). The product was extracted from the aqueous layer three times with methylene chloride (100, 100, 50 mL). The combined organic layers were dried with Na2SO4, filtered and rotoevaporated to a colorless oil. The foamy oil was vacuum dried for 2 h. Intermediate 4 was obtained in 90% yield (3.34 g).

Intermediate 3(1R,2R,1″S)-1-(2′,3′-Dihydro-benzo[1,4]dioxin-6′-yl)-2-(2″-hydroxy -1′-phenyl-ethylamino)-3-pyrrolidin-1-yl-propan-1-ol

Figure US07196205-20070327-C00022

To a 3-neck flask equipped with a dropping funnel and condenser was added LiAlH4 (Aldrich, 1.2 g, 31.7 mmol, 2.5 equivalents) and anhydrous THF (20 mL) under nitrogen. A solution of Intermediate 2 (5.23 g, 12.68 mmol) in anhydrous THF (75 mL) was added dropwise to the reaction over 15–30 minutes. The reaction was refluxed under nitrogen for 9 hours. The reaction was cooled in an ice bath and a 1M NaOH solution was carefully added dropwise. After stirring at room temperature for 15 minutes, water (50 mL) and ethyl acetate (75 mL) was added. The layers were separated and the aqueous layer was extracted twice with ethyl acetate (75 mL). The combined organic layers were washed with saturated sodium chloride solution (25 mL). After drying with Na2SO4 the solution was filtered and rotoevaporated to yield a colorless to yellow foamy oil. Intermediate 3 was obtained in 99% yield (5.3 g).

PATENT

WO 2016001885

EXAMPLES

Example 1 : Preparation of amorphous form of eliglustat hemitartarate.

500mg of eliglustat hemitartarate was dissolved in 14 mL of dichloromethane at 26°C and stirred for 15 min. The solution is filtered to remove the undissolved particles and the filtrate is distilled under reduced pressure at 45°C. After distillation the solid was dried under vacuum at 45°C.

PATENT

str1

 

PAPER

Journal of Medicinal Chemistry (2012), 55(9), 4322-4335

 

 

OLD CLIPS

Genzyme Announces Positive New Data from Two Phase 3 Studies for Oral Eliglustat Tartrate for Gaucher Disease


Eliglustat tartrate (USAN)

CAS:928659-70-5
February 15, 2013
Genzyme , a Sanofi company (EURONEXT: SAN and NYSE: SNY), today announced positive new data from the Phase 3 ENGAGE and ENCORE studies of eliglustat tartrate, its investigational oral therapy for Gaucher disease type 1. The results from the ENGAGE study were presented today at the 9th Annual Lysosomal Disease Network WORLD Symposium in Orlando, Fla. In conjunction with this meeting, Genzyme also released topline data from its second Phase 3 study, ENCORE. Both studies met their primary efficacy endpoints and together will form the basis of Genzyme’s registration package for eliglustat tartrateThe data presented at this year’s WORLD symposium reinforce our confidence that eliglustat tartrate may become an important oral option for patients with Gaucher disease”The company is developing eliglustat tartrate, a capsule taken orally, to provide a convenient treatment alternative for patients with Gaucher disease type 1 and to provide a broader range of treatment options for patients and physicians. Genzyme’s clinical development program for eliglustat tartrate represents the largest clinical program ever focused on Gaucher disease type 1 with approximately 400 patients treated in 30 countries.“The data presented at this year’s WORLD symposium reinforce our confidence that eliglustat tartrate may become an important oral option for patients with Gaucher disease,” said Genzyme’s Head of Rare Diseases, Rogerio Vivaldi MD. “We are excited about this therapy’s potential and are making excellent progress in our robust development plan for bringing eliglustat tartrate to the market.”ENGAGE Study Results:In ENGAGE, a Phase 3 trial to evaluate the safety and efficacy of eliglustat tartrate in 40 treatment-naïve patients with Gaucher disease type 1, improvements were observed across all primary and secondary efficacy endpoints over the 9-month study period. Results were reported today at the WORLD Symposium by Pramod Mistry, MD, PhD, FRCP, Professor of Pediatrics & Internal Medicine at Yale University School of Medicine, and an investigator in the trial.The randomized, double-blind, placebo-controlled study had a primary efficacy endpoint of improvement in spleen size in patients treated with eliglustat tartrate. Patients were stratified at baseline by spleen volume. In the study, a statistically significant improvement in spleen size was observed at nine months in patients treated with eliglustat tartrate compared with placebo. Spleen volume in patients treated with eliglustat tartrate decreased from baseline by a mean of 28 percent compared with a mean increase of two percent in placebo patients, for an absolute difference of 30 percent (p<0.0001).

Genzyme

Eliglustat tartate (Genz-112638)

What is Eliglustat?

  • Eliglustat is a new investigational phase 3 compound from Genzyme Corporation that is being studied for type 1 Gaucher Disease.
  • Eliglustat works as a substrate reduction therapy by reducing glucocerebroside. formation.
  • This product is an oral agent (i.e. a pill) that is taken once or twice a day in contrast to an IV infusion for enzyme replacement therapy. Enzyme replacement therapy focuses on replenishing the enzyme that is deficient in Gaucher Disease and breaks down glucocerebroside that accumulates.
  • The clinical trials for eliglustat tartate are sponsored by Genzyme Corporation.

Eliglustat tartrate (Genz-1 12638) is a glucocerebroside (glucosylceramide) synthase inhibitor for the treatment of gaucher disease and other lysosomal storage disorders, which is currently under development.

Eliglustat is chemically known as 1 R, 2R-Octanoic acid [2-(2′, 3′-dihydro-benzo [1 , 4] dioxin-6′-yl)-2-hydroxy-1 -pyrrolidin-1 -ylmethyl]-ethyl]-amide, having a structural formula I depicted here under.

Formula I

Eliglustat hemitartrate (Genz-1 12638) development by Genzyme, is a glucocerebroside (glucosylceramide) synthase inhibitor for the treatment of Gaucher disease and other lysosomal storage disorders. Eliglustat hemitartrate is orally active with potent effects on the primary identified molecular target for type 1 Gaucher disease and other glycosphingolipidoses, appears likely to fulfill high expectations for clinical efficacy.

Gaucher disease belongs to the class of lysosomal diseases known as glycosphingolipidoses, which result directly or indirectly from the accumulation of glycosphingolipids, many hundreds of which are derived from glucocerebroside. The first step in glycosphingolipid biosynthesis is the formation of glucocerebroside, the primary storage molecule in Gaucher disease, via glucocerebroside synthase (uridine diphosphate [UDP] – glucosylceramide glucosyl transferase). Eliglustat hemitartrate is based on improved inhibitors of glucocerebroside synthase.

U.S. patent No. 7,196,205 (herein described as US’205) discloses a process for the preparation of eliglustat or a pharmaceutically acceptable salt thereof. In this patent, eliglustat was synthesized via a seven-step process involving steps in that sequence:

(i) coupling S-(+)-2-phenyl glycinol with phenyl bromoacetate followed by column chromatography for purification of the resulting intermediate,

(ii) reacting the resulting (5S)-5-phenylmorpholin-2-one with 1 , 4-benzodioxan-6-carboxaldehyde to obtain a lactone,

(iii) opening the lactone of the oxazolo-oxazinone cyclo adduct via reaction with pyrrolidine,

(iv) hydrolyzing the oxazolidine ring, (v) reducing the amide to amine to obtain sphingosine like compound, (vi) reacting the resulting amine with octanoic acid and N-hydroxysuccinimide to obtain crude eliglustat, (vii) purifying the crude eliglustat by repeated isolation for four times from a mixture of ethyl acetate and n-heptane.

U.S. patent No. 6855830, 7265228, 7615573, 7763738, 8138353, U.S. patent application publication No. 2012/296088 disclose processes for preparation of eliglustat and intermediates thereof.

U.S. patent application publication No. 2013/137743 discloses (i) a hemitartrate salt of eliglustat, (ii) a hemitartrate salt of eliglustat, wherein at least 70% by weight of the salt is crystalline, (iii) a hemitartrate salt of Eliglustat, wherein at least 99% by weight of the salt is in a single crystalline form.

https://patentscope.wipo.int/search/en/detail.jsf;jsessionid=234E6BE008E68831F6875FB703760826.wapp2nA?docId=WO2015059679&recNum=1&office=&queryString=FP%3A%28dr.+reddy%27s%29&prevFilter=%26fq%3DCTR%3AWO&sortOption=Pub+Date+Desc&maxRec=364

WO 2015059679

Process for the preparation of eliglustat free base – comprising the reaction of S-(+)-phenyl glycinol with phenyl-alpha-bromoacetate to obtain 5-phenylmorpholin-2-one, which is further converted to eliglustat.
Dr Reddy’s Laboratories Ltd
New crystalline eliglustat free base Form R1 and a process for its preparation are claimed. Also claimed is a process for the preparation of eliglustat free base which comprises the reaction of S-(+)-phenyl glycinol with phenyl-alpha-bromoacetate to obtain 5-phenylmorpholin-2-one, which is further converted to eliglustat.Further eliglustat oxalate, its crystalline form, and a process for the preparation of crystalline eliglustat oxalate, are claimed.

Eliglustat tartrate (Genz-1 12638) is a glucocerebroside (glucosylceramide) synthase inhibitor for the treatment of gaucher disease and other lysosomal storage disorders, which is currently under development.

Eliglustat is chemically known as 1 R, 2R-Octanoic acid [2-(2′, 3′-dihydro-benzo [1 , 4] dioxin-6′-yl)-2-hydroxy-1 -pyrrolidin-1 -ylmethyl]-ethyl]-amide, having a structural formula I depicted here under.

Formula I

Eliglustat hemitartrate (Genz-1 12638) development by Genzyme, is a glucocerebroside (glucosylceramide) synthase inhibitor for the treatment of Gaucher disease and other lysosomal storage disorders. Eliglustat hemitartrate is orally active with potent effects on the primary identified molecular target for type 1 Gaucher disease and other glycosphingolipidoses, appears likely to fulfill high expectations for clinical efficacy.

Gaucher disease belongs to the class of lysosomal diseases known as glycosphingolipidoses, which result directly or indirectly from the accumulation of glycosphingolipids, many hundreds of which are derived from glucocerebroside. The first step in glycosphingolipid biosynthesis is the formation of glucocerebroside, the primary storage molecule in Gaucher disease, via glucocerebroside synthase (uridine diphosphate [UDP] – glucosylceramide glucosyl transferase). Eliglustat hemitartrate is based on improved inhibitors of glucocerebroside synthase.

U.S. patent No. 7,196,205 (herein described as US’205) discloses a process for the preparation of eliglustat or a pharmaceutically acceptable salt thereof. In this patent, eliglustat was synthesized via a seven-step process involving steps in that sequence:

(i) coupling S-(+)-2-phenyl glycinol with phenyl bromoacetate followed by column chromatography for purification of the resulting intermediate,

(ii) reacting the resulting (5S)-5-phenylmorpholin-2-one with 1 , 4-benzodioxan-6-carboxaldehyde to obtain a lactone,

(iii) opening the lactone of the oxazolo-oxazinone cyclo adduct via reaction with pyrrolidine,

(iv) hydrolyzing the oxazolidine ring, (v) reducing the amide to amine to obtain sphingosine like compound, (vi) reacting the resulting amine with octanoic acid and N-hydroxysuccinimide to obtain crude eliglustat, (vii) purifying the crude eliglustat by repeated isolation for four times from a mixture of ethyl acetate and n-heptane.

U.S. patent No. 6855830, 7265228, 7615573, 7763738, 8138353, U.S. patent application publication No. 2012/296088 disclose processes for preparation of eliglustat and intermediates thereof.

U.S. patent application publication No. 2013/137743 discloses (i) a hemitartrate salt of eliglustat, (ii) a hemitartrate salt of eliglustat, wherein at least 70% by weight of the salt is crystalline, (iii) a hemitartrate salt of Eliglustat, wherein at least 99% by weight of the salt is in a single crystalline form.

Example 1 : Preparation of 5-phenyl morpholine-2-one hydrochloride

To a (S) + phenyl glycinol (100g) add N, N-diisopropylethylamine (314ml) and acetonitrile (2000ml) under nitrogen atmosphere at room temperature. It was cooled to 10- 15° C. Phenyl bromoacetate (172.4g) dissolved in acetonitrile (500ml) was added to the above solution at 15° C over a period of 30 min. The reaction mixture is allowed to room temperature and stirred for 16-20h. Progress of the reaction was monitored by thin layer chromatography. After completion of the reaction, the reaction mixture was concentrated under reduced pressure at a water bath

temperature less than 25° C to get a residue. The residue was dissolved in ethyl acetate (1000ml) and stirred for 1 h at 15-20°C to obtain a white solid. The solid material obtained was filtered and washed with ethyl acetate (200ml). The filtrate was dried over anhydrous sodium sulphate (20g) and concentrated under reduced pressure at a water bath temperature less than 25° C to give crude compound (1000g) as brown syrup. The Crude brown syrup is converted to HCI salt by using HCI in ethyl acetate to afford 5-phenyl morpholine-2-one hydrochloride (44g) as a white solid. Yield: 50%, Mass: m/z = 177.6; HPLC (% Area Method): 90.5%

Example 2: Preparation of (1 R,3S,5S,8aS)-1 ,3-Bis-(2′,3′-dihydro-benzo[1 ,4] dioxin-6′-yl)-5-phenyl-tetrahydro-oxazolo[4,3-c][1 ,4]oxazin-8-one.

5-phenyl morpholine-2-one hydrochloride (100g) obtained from above stage 1 is dissolved in toluene (2500ml) under nitrogen atmosphere at 25-30°C. 1 ,4-benzodioxane-6-carboxaldehyde (185.3g) and sodium sulphate (400g) was added to the above solution and the reaction mixture was heated at 100-105°C for 72h. Progress of the reaction was monitored by thin layer chromatography. After completion of reaction, the reaction mixture was concentrated under reduced pressure at a water bath temperature less than 25° C to get a residue. The residue was cooled to 10°C, ethyl acetate (2700ml) and 50% sodium bisulphate solution (1351 ml) was added to the residue and stirred for 1 h at 10°C to obtain a white solid. The obtained white solid was filtered and washed with ethyl acetate. The separated ethyl acetate layer was washed with water (1000ml), brine (1000ml) and dried over anhydrous sodium sulphate. The organic layer was concentrated under reduced pressure at a water bath temperature of 45-50°C to get a crude material. The obtained crude material is triturated with diethyl ether (1500ml) to get a solid material which is filtered and dried under vacuum at room temperature for 2-3h to afford (1 R,3S,5S,8aS)-1 ,3-Bis-(2′,3′-dihydro-benzo[1 ,4]dioxin-6′-yl)-5-phenyl-tetrahydro-oxazolo[4,3-c][1 ,4]oxazin-8-one (148g) as a yellow solid. Yield: 54%, Mass: m/z = 487.7; HPLC (% Area Method): 95.4 %

Example 3: Preparation of (2S,3R,1 “S)-3-(2′,3′-(Dihydro-benzo[1 ,4]dioxin-6′-yl)-3-hydroxy-2-(2″-hydroxy-1 ”^henyl-ethy^

(1 R,3S,5S,8aS)-1 !3-Bis-(2′!3′-dihydro-benzo[1 ,4]dioxin-6′-yl)-5-phenyl-tetrahydro-oxazolo[4,3-c][1 ,4]oxazin-8-one (70g) obtained from above stage 2 was dissolved in chloroform (1400ml) at room temperature. It was cooled to 0-5°C and pyrrolidone (59.5ml) was added at 0-5°C over a period of 30 minutes. The reaction mixture was allowed to room temperature and stirred for 16-18h. Progress of the reaction was monitored by thin layer chromatography. After completion of reaction, the reaction mixture was concentrated under reduced pressure at a water bath temperature of 40-45°C to obtain a crude. The obtained crude was dissolved in methanol (1190ml) and 1 N HCI (1 190ml) at 10-15° C, stirred for 10 minutes and heated at 80-85°C for 7h. Progress of the reaction was monitored by thin layer chromatography. After completion of reaction, methanol was concentrated under reduced pressure at a water bath temperature of 50-55°C.The aqueous layer was extracted with ethyl acetate and the organic layer was washed with 1 N HCI (50ml). The aqueous layer was basified with saturated sodium bicarbonate solution up to pH 8-9 and extracted with ethyl acetate (3x70ml). The combined organic layers was washed with brine (100ml), dried over anhydrous sodium sulphate and concentrated under reduced pressure at a water bath temperature of 50-55°C to afford (2S,3R,1″S)-3-(2′,3′-(Dihydro-benzo[1 ,4]dioxin-6′-yl)-3-hydroxy-2-(2″-hydroxy-1 “-phenyl-ethylamino)-1 -pyrrolidin-1 -yl-propan-1 -one (53g) as a yellow foamy solid. Yield: 90%, Mass: m/z = 412.7, HPLC (% Area Method): 85.1 %

Example 4: Preparation of (1 R,2R,1 “S)-1-(2′,3′-(Dihydro-benzo[1 ,4]dioxin-6′-yl)2-hydroxy-2-(2”-hydroxy-1 ‘-phenyl-ethylamino)-3-pyrrolidin-1-yl-propan-1-ol.

(2S,3R,1 “S)-3-(2′,3′-(Dihydro-benzo[1 ,4]dioxin-6’-yl)-3-hydroxy-2-(2”-hydroxy-1 “-phenyl-ethylamino)-1 -pyrrolidin-1 -yl-propan-1 -one (2.5g) obtained from above stage 3 dissolved in Tetrahydrofuran (106ml) was added to a solution of Lithium aluminium hydride (12.2g) in tetrahydrofuran (795ml) at 0°C and the reaction mixture was heated at 60-65°C for 10h. Progress of the reaction was monitored by thin layer chromatography. After completion of reaction, the reaction mixture was cooled to 5- 10°C and quenched in saturated sodium sulphate solution (100ml) at 5-10°C. Ethyl acetate was added to the reaction mass and stirred for 30-45 min. The obtained solid is filtered through celite bed and washed with ethyl acetate. Filtrate was dried over anhydrous sodium sulphate and concentrated under reduced pressure at a water bath temperature of 50°C to afford (1 R,2R, 1″S)-1 -(2′,3′-(Dihydro-benzo[1 ,4]dioxin-6′-yl)2-hydroxy-2-(2″-hydroxy-1 ‘-phenyl-ethylamino)-3-pyrrolidin-1 -yl-propan-1 -ol (43.51 g) as a yellow gummy liquid. The crude is used for the next step without further purification. Yield: 85%, Mass: m/z = 398.7, HPLC (% Area Method): 77 %

Example 5: Preparation of (1 R, 2R)-2-Amino-1-(2′, 3′-dihydro-benzo [1 , 4] dioxin-6′-yl)-3-pyrrolidin-1 -yl-propan-1 -ol.

(1 R,2R,1 “S)-1 -(2′,3′-(Dihydro-benzo[1 ,4]dioxin-6’-yl)2-hydroxy-2-(2”-hydroxy-1 ‘-phenyl-ethylamino)-3-pyrrolidin-1 -yl-propan-1 -ol (40g) obtained from above stage 4 was dissolved in methanol (400ml) at room temperature in a 2L hydrogenation flask. Trifluoroacetic acid (15.5ml) and 20% Pd (OH) 2(40g) was added to the above solution under nitrogen atmosphere. The reaction mixture was hydrogenated under H2, 10Opsi for 16-18h at room temperature. Progress of the reaction was monitored by thin layer chromatography. After completion of reaction, the reaction mixture was filtered through celite bed and washed with methanol (44ml) and water (44ml). Methanol was concentrated under reduced pressure at a water bath temperature of 50-55°C and the aqueous layer was washed with ethyl acetate. The aqueous layer was basified with 10M NaOH till the PH reaches 12-14 and then extracted with dichloromethane (2x125ml). The organic layer was dried over anhydrous sodium sulphate (3gm) and concentrated under reduced pressure at a water bath temperature of 45°C to obtain a gummy liquid. The gummy liquid was triturated with methyl tertiary butyl ether for 1 h to get a white solid, which is filtered and dried under vacuum at room temperature to afford (1 R, 2R)-2-Amino-1 -(2′, 3′-dihydro-benzo [1 , 4] dioxin-6′-yl)-3-pyrrolidin-1 -yl-propan-1 -ol (23g) as a white solid. Yield: 82.3%, Mass (m/zj: 278.8, HPLC (% Area Method): 99.5%, Chiral HPLC (% Area Method): 97.9%

Example 6: Preparation of Eliglustat {(1 R, 2R)-Octanoic acid[2-(2′,3′-dihydro-benzo [1 , 4] dioxin-6′-yl)-2-hydroxy-1 -pyrrolidin-1-ylmethyl-ethyl]-amide}.

(1 R, 2R)-2-Amino-1 -(2′, 3′-dihydro-benzo [1 , 4] dioxin-6′-yl)-3-pyrrolidin-1 -yl-propan-1 -ol (15g) obtained from above stage 5 was dissolved in dry dichloromethane (150ml) at room temperature under nitrogen atmosphere and cooled to 10-15° C. Octanoic acid N-hydroxy succinimide ester (13.0 g)was added to the above reaction mass at 10-15° C and stirred for 15 min. The reaction mixture was stirred at room temperature for 16h-18h. Progress of the reaction was monitored by thin layer chromatography. After completion of reaction, the reaction mixture was cooled to 15°C and diluted with 2M NaOH solution (100 ml_) and stirred for 20 min at 20 °C. The organic layer was separated and washed with 2M sodium hydroxide (3x90ml).The organic layer was dried over anhydrous sodium sulphate (30g) and concentrated under reduced pressure at a water bath temperature of 45°C to give the crude compound (20g).The crude is again dissolved in methyl tertiary butyl ether (25 ml_) and precipitated with Hexane (60ml). It is stirred for 10 min, filtered and dried under vacuum to afford Eliglustat as a white solid (16g). Yield: 74%, Mass (m/zj: 404.7 HPLC (% Area Method): 97.5 %, ELSD (% Area Method): 99.78%, Chiral HPLC (% Area Method): 99.78 %.

Example 7: Preparation of Eliglustat oxalate.

Eliglustat (5g) obtained from above stage 6 is dissolved in Ethyl acetate (5ml) at room temperature under nitrogen atmosphere. Oxalic acid (2.22g) dissolved in ethyl acetate (5ml) was added to the above solution at room temperature and stirred for 14h. White solid observed in the reaction mixture was filtered and dried under vacuum at room temperature for 1 h to afford Eliglustat oxalate as a white solid (4g). Yield: 65.46%, Mass (m/zj: 404.8 [M+H] +> HPLC (% Area Method): 95.52 %, Chiral HPLC (% Area Method): 99.86 %

References

  1.  Eligustat (PDF), AMA By subscription only
  2. FDA approves new drug to treat a form of Gaucher disease, U.S. Food and Drug Administration, 19 August 2015, retrieved 18 July 2015
  3. Lee, L.; Abe, A.; Shayman, J. A. (21 May 1999). “Improved Inhibitors of Glucosylceramide Synthase”. Journal of Biological Chemistry 274(21): 14662–14669. doi:10.1074/jbc.274.21.14662.
  4.  Shayman, JA (1 August 2010). “Eliglustat Tartrate: Glucosylceramide Synthase Inhibitor Treatment of Type 1 Gaucher Disease.”. Drugs of the future 35 (8): 613–620. PMID 22563139.
  5.  Pramod K. Mistry, Elena Lukina, Hadhami Ben Turkia, Dominick Amato, Hagit Baris, Majed Dasouki, Marwan Ghosn, Atul Mehta, Seymour Packman, Gregory Pastores, Milan Petakov, Sarit Assouline, Manisha Balwani, Sumita Danda, Evgueniy Hadjiev, Andres Ortega, Suma Shankar, Maria Helena Solano, Leorah Ross, Jennifer Angell, M. Judith Peterschmitt (17 February 2015), “Effect of Oral Eliglustat on Splenomegaly in Patients With Gaucher Disease Type 1: The ENGAGE Randomized Clinical Trial”, Journal of the American Medical Association 313 (7): 695–706, doi:10.1001/jama.2015.459
  6.  Robert Weisman (2 September 2014), New Genzyme pill will cost patients $310,250 a year, The Boston Globe, retrieved 18 July 2015

FDA Orange Book Patents

FDA Orange Book Patents: 1 of 3
Patent 6916802
Expiration Apr 29, 2022
Applicant GENZYME CORP
Drug Application N205494 (Prescription Drug: CERDELGA. Ingredients: ELIGLUSTAT TARTRATE)
FDA Orange Book Patents: 2 of 3
Patent 7196205
Expiration Apr 29, 2022
Applicant GENZYME CORP
Drug Application N205494 (Prescription Drug: CERDELGA. Ingredients: ELIGLUSTAT TARTRATE)
FDA Orange Book Patents: 3 of 3
Patent 7615573
Expiration Apr 29, 2022
Applicant GENZYME CORP
Drug Application N205494 (Prescription Drug: CERDELGA. Ingredients: ELIGLUSTAT TARTRATE)
Patent ID Date Patent Title
US8003617 2011-08-23 Methods of Treating Diabetes Mellitus
US2010298317 2010-11-25 METHOD OF TREATING POLYCYSTIC KIDNEY DISEASES WITH CERAMIDE DERIVATIVES
US7763738 2010-07-27 SYNTHESIS OF UDP-GLUCOSE: N-ACYLSPHINGOSINE GLUCOSYLTRANSFERASE INHIBITORS
US7615573 2009-11-10 Synthesis of UDP-glucose: N-acylsphingosine glucosyltransferase inhibitors
US2009105125 2009-04-23 Methods of Treating Fatty Liver Disease
US7265228 2007-09-04 Synthesis of UDP-glucose: N-acylsphingosine glucosyltransferase inhibitors
US7196205 2007-03-27 Synthesis of UDP-glucose: N-acylsphingosine glucosyltransferase inhibitors
US6855830 2005-02-15 Synthesis of UDP-glucose: N-acylsphingosine glucosyltransferase inhibitors
Patent ID Date Patent Title
US2016068519 2016-03-10 INHIBITORS OF THE ENZYME UDP-GLUCOSE: N-ACYL-SPHINGOSINE GLUCOSYLTRANSFERASE
US2015148534 2015-05-28 SYNTHESIS OF UDP-GLUCOSE: N-ACYLSPHINGOSINE GLUCOSYL TRANSFERASE INHIBITORS
US2015051261 2015-02-19 Methods of Treating Fatty Liver Disease
US8779163 2014-07-15 Synthesis of UDP-Glucose: N-acylsphingosine glucosyl transferase inhibitors
US2013137743 2013-05-30 AMORPHOUS AND A CRYSTALLINE FORM OF GENZ 112638 HEMITARTRATE AS INHIBITOR OF GLUCOSYLCERAMIDE SYNTHASE
US2013095089 2013-04-18 GLUCOSYLCERAMIDE SYNTHASE INHIBITORS AND THERAPEUTIC METHODS USING THE SAME
US2012322786 2012-12-20 2-ACYLAMINOPROPOANOL-TYPE GLUCOSYLCERAMIDE SYNTHASE INHIBITORS
US8138353 2012-03-20 SYNTHESIS OF UDP-GLUCOSE: N-ACYLSPHINGOSINE GLUCOSYLTRANSFERASE INHIBITORS
US2012022126 2012-01-26 Method Of Treating Diabetes Mellitus
US8003617 2011-08-23 Methods of Treating Diabetes Mellitus
Eliglustat
Eliglustat.svg
Systematic (IUPAC) name
N-[(1R,2R)-1-(2,3-Dihydro-1,4-benzodioxin-6-yl)-1-hydroxy-3-(1-pyrrolidinyl)-2-propanyl]octanamide
Clinical data
Trade names Cerdelga
Legal status
Legal status
Identifiers
CAS Number 491833-29-5
ATC code A16AX10 (WHO)
PubChem CID 23652731
ChemSpider 28475348
ChEBI CHEBI:82752 Yes
Chemical data
Formula C23H36N2O4
Molar mass 404.543 g/mol

 

Patent Number Pediatric Extension Approved Expires (estimated)
US6916802 No 2002-04-29 2022-04-29 Us
US7196205 No 2002-04-29 2022-04-29 Us
US7615573 No 2002-04-29 2022-04-29 Us

///////////491833-29-5, 928659-70-5, eliglustat hemitartrate, eliglustat L-tartrate, ELIGLUSTAT,  Cerdelga,  Genz 99067,  Genz-99067,  UNII-DR40J4WA67,  GENZ-112638, エリグルスタット酒石酸塩 , FDA 2014,  GAUCHERS DISEASE, 依利格鲁司特, エリグルスタット,サーデルガ

CCCCCCCC(=O)N[C@H](CN1CCCC1)[C@@H](C2=CC3=C(C=C2)OCCO3)O

Share
Follow

Get every new post on this blog delivered to your Inbox.

Join other followers: