New Drug Approvals

Home » PHASE 3

Category Archives: PHASE 3

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

Blog Stats

  • 4,479,548 hits

Flag and hits

Flag Counter

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

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

Archives

Categories

Recent Posts

Flag Counter

ORGANIC SPECTROSCOPY

Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL 

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

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

DR ANTHONY MELVIN CRASTO Ph.D

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

Personal Links

Verified Services

View Full Profile →

Archives

Categories

Flag Counter

Evobrutinib


Evobrutinib.png

Evobrutinib

429.5 g/mol,C25H27N5O2

  • Evobrutinib
  • 1415823-73-2
  • Evobrutinib [INN]
  • 1-(4-(((6-amino-5-(4-phenoxyphenyl)pyrimidin-4-yl)amino)methyl)piperidin-1-yl)prop-2-en-1-one
  • MSC2364447C

Evobrutinib is under investigation in clinical trial NCT03934502 (Effect of Meal Composition and Timing on Evobrutinib Bioavailability).

Evobrutinib is an inhibitor of Bruton’s tyrosine kinase (BTK) with potential antineoplastic activity. Upon administration, evobrutinib inhibits the activity of BTK and prevents the activation of the B-cell antigen receptor (BCR) signaling pathway. This prevents both B-cell activation and BTK-mediated activation of downstream survival pathways, which leads to the inhibition of the growth of malignant B-cells that overexpress BTK. BTK, a member of the Src-related BTK/Tec family of cytoplasmic tyrosine kinases, is overexpressed in B-cell malignancies; it plays an important role in B-lymphocyte development, activation, signaling, proliferation and survival.

Evobrutinib is in clinical development to investigate its potential as a treatment for multiple sclerosis (MS). It is an oral, highly selective inhibitor of Bruton’s tyrosine kinase (BTK) which is important in the development and functioning of various immune cells including B lymphocytes and macrophages.

Evobrutinib is designed to inhibit primary B cell responses such as proliferation and antibody and cytokine release, without directly affecting T cells. BTK inhibition is thought to suppress autoantibody-producing cells, which preclinical research suggests may be therapeutically useful in certain autoimmune diseases.

U.S. Patent No. 9073947 discloses a pyrimidine derivative of Evobrutinib which chemically named as l-(4-(((6-amino-5-(4-phenoxyphenyl)pyrimidin-4-yl)amino)methyl)

piperidin-l-yl)prop-2-en-l-one and pharmaceutically acceptable salts, solvates and pharmaceutical compositions thereof.

U.S. Patent No. 9073947 and ‘Journal of Medicinal Chemistry 2019, 62(17), 7643-7655’ discloses process for the preparation of Evobrutinib which involves column purifications and lyophilisation methods to provide Evobrutinib with low yield, which is not viable at large scale production.

https://www.frontiersin.org/articles/10.3389/fnume.2021.820235/full

STR4

Radiosynthesis of [11C]Evobrutinib. [11C]Evobrutinib was synthesized similarly to the Tolebrutinib example above with the following exceptions. First, the precursor 5-(4-phenoxyphenyl)-N4-(piperidin-4-ylmethyl)pyrimidine-4,6-diamine (4) (1 mg, 2.7 μmol) was used and the crude reaction mixture after the carbonylation reaction was purified by semi-preparative HPLC (column: Luna C18(2), 5 μ (250 x 9.6 mm); mobile phase: 44% MeCN in 200 mM ammonium formate; flow rate: 5 ml/min; UV: 254 nm). The [11C]1-(4-(((6-amino-5-(4-phenoxyphenyl)pyrimidin-4-yl)amino)methyl)piperidin-1-yl)prop-2-en-1-one ([11C]evobrutinib) was isolated between the 15.5 and 18 min mark of the chromatogram and this sample was collected into a dilution flask that contained 50 ml of a 2 mg/ml sodium ascorbate aqueous solution. This solution was transferred to an HLB light (30 mg) SPE cartridge. After transfer, the cartridge was eluted with 1 ml of ethanol into the sterile product vial that contained 4 ml of sterile saline. Using this method, 2.2 ± 0.6 GBq (81.4 ± 22.2 mCi) [11C]evobrutinib was isolated (n = 3), and the product was analyzed via reverse phase HPLC using the following methods. Method A described above and Method B (Isocratic and molar activity): column: Luna C18(2) 3-μm (250×4.6 mm); mobile phase Isocratic: 36% acetonitrile in aqueous 0.1% TFA; flow rate: 1.3 ml/min; UV: 254 nm. Method A was used to confirm chemical identity using a co-injection of non-radioactive standard. Radiochemical purity and molar activity were determined by Method B. [11C]Evobrutinib was confirmed by co-injection with a verified non-radioactive reference standard. Am was determined using a 4-point standard curve (analytical HPLC peak area) (Y) vs. standard concentration (X: in nmol) by comparison with an evobrutinib reference standard of known concentration (2.3 mg in 1 ml). The isolated [11C] evobrutinib was co-eluted with a non-radioactive reference standard. The sample was >99% radiochemically pure, >95% chemically pure (HPLC, UV: 254 nm), with a molar activity of 496.5 ± 74 GBq/μmol (13.4 Ci/μmol) The overall synthesis time from the end of cyclotron bombardment was 37–46 min.

Patent

U.S. Patent No. 9073947

PAPER

Journal of Medicinal Chemistry 2019, 62(17), 7643-7655

https://pubs.acs.org/doi/10.1021/acs.jmedchem.9b00794

STR3

Step 4

To a 20 mL vial was added 5-(4-phenoxyphenyl)-N-(piperidin-4-ylmethyl)pyrimidine-4,6-diamine (210.00 mg, 0.56 mmol, 1.00 equiv), sodium bicarbonate (70.48 mg, 0.84 mmol, 1.50 equiv), THF (8.00 mL, 98.74 mmol, 176.55 equiv), and water (0.80 mL, 44.41 mmol, 79.40 equiv). The mixture was cooled to 0 °C on an ice bath. Acryloyl chloride (0.15 mL, 1.83 mmol) was then added dropwise. The ice bath was removed, and the reaction was stirred at room temperature for 12 h before it was purified by silica gel chromatography (25 g KPNH silica, 0–100% methanol/ethyl acetate) to afford the title compound (A18) (21 mg, 8.7% yield) was synthesized with a similar protocol to prepared as described in the main body of the article. 1H NMR (DMSO-d6) δ 7.93 (s, 1 H), 7.40–7.08 (m, 9H), 6.76 (dd, J = 4 Hz, 1 H), 6.04 (d, J = 4 Hz, 1 H), 5.61 (d, J = 4 Hz, 1 H), 5.43 (s, 2H), 4.34 (d, J = 12 Hz, 1 H), 3.98 (d, J = 8 Hz, 1 H), 3.12 (m, 2H), 2.95 (m, 1 H), 2.56 (m, 1 H), 1.81 (m, 1 H), 1.59 (m, 2H), 0.92 (m, 2H). [ES-MS] (ESI+): m/z calcd for C25H28N5O2 [M + H]+ 430, found 430.

PATENT

STR1
STR2

Examples:

Example-1: Preparation of tert-butyl 4-(((6-amino-5-chloropyrimidin-4-yl)amino) met hy 1 jpiperid ine- 1 -carboxylate

Tert-butyl-4-(aminomethyl)piperidine-l -carboxylate (81 ml) and 1,8-diazabicyclo [5.4.0]undec-7-ene (60.34 g) were added to a mixture of 5,6-dichloropyrimidin-4-amine (50 g) in N,N-dimethylformamide (500 ml) at 25-35°C. Heated the mixture to 90-95°C and stirred for 22 hrs. Cooled the mixture to 25-30°C. Water was added to the mixture at 25-35°C and stirred for 5 hrs. Filtered the precipitated solid, washed with water and n-heptane and dried to get the title compound. Yield: 73.0 gms; Purity by HPLC: 98.7%

Example-2: Preparation of tert-butyl 4-(((6-amino-5-(4-phenoxyphenyl)pyrimidin-4-yl) amino)methyl)piperidine-l-carboxylate

(4-Phenoxyphenyl)boronic acid (75.12 g) was added to a mixture of tert-butyl 4-(((6-amino-5-chloropyrimidin-4-yl)amino)methyl)piperidine-l-carboxylate(100 g), 2-di cyclo hexylphosphino-2′,6′-dimethoxybiphenyl (12 g) and potassium carbonate (121.28 g) in 1,4-di oxane (1000 ml) at 25-30°C and stirred for 30 minutes under nitrogen atmosphere. Palladium acetate (1.96 g) was added to the mixture at 25-30°C. Heated the mixture to 100-105°C and stirred for 3 hrs. Cooled the mixture to 25-30°C. Water and ethyl acetate were added to the mixture at 25-35°C and stirred for 30 minutes. Filtered the mixture by using hyflow bed. Organic layer was separated from the filtrate. Organic layer was treated with carbon powder and distilled-off the solvent under reduced pressure, n-heptane (800 ml) was added to the obtained compound. Heated the mixture to 60-65°C and stirred for 90 minutes. Cooled the mixture to 25-30°C and stirred for 2 hrs. Filtered the precipitated solid, washed with n-heptane and dried to get the title compound. Yield: 120 gms, Purity by HPEC: 97.6% Example-3: Preparation of 5-(4-phenoxyphenyl)-N4-(piperidin-4-ylmethyl)pyrimidine-4,6-diamine

Tert-butyl-4-(((6-amino-5-(4-phenoxyphenyl)pyrimidin-4-yl)amino)methyl) piperidine- 1 -carboxylate (200 g) in methanol (600 ml) was cooled to 0-5°C. Hydrochloric acid in ethyl acetate (500 ml) was slowly added to the mixture at 0-5°C. Mixture allowed to warm to 25-30°C and stirred for 20 hours. Water was added to the mixture and treated the mixture with aqueous ammonia solution. Dichloromethane was added to the mixture at 25-30°C and stirred for 10 minutes. Layers were separated and distilled-off the organic layer under reduce pressure. Obtained compound was treated with isopropyl ether and dried to get the title compound. Yield: 150 gms, Purity by HPLC: 76.4%

Example-4: Preparation of Evobrutinib

Sodium bicarbonate (23.86 g) and water (301 ml) were added to the mixture of 5-(4-phenoxyphenyl)-N4-(piperidin-4-ylmethyl)pyrimidine-4,6-diamine (70 g) in tetrahydrofuran (2800 ml). Cooled the mixture to 0-5°C. Acryloyl chloride (23.62 g) was slowly added to the mixture. Mixture allowed to warm to 25-30°C and stirred for 20 hrs. Distilled-off the solvent from the mixture under reduced pressure. Ethyl acetate and water were added to the mixture and stirred for 10 minutes. Both the layers were separated. Organic layer was treated with aqueous hydrochloric acid solution and carbon powder. Distilled-off the organic layer under reduced pressure. Isopropyl ether was added to the mixture at 25-30°C and stirred for 14 hrs. Filtered the mixture and washed with isopropyl ether. Dried to get the title compound.

Yield: 41.8 gms, Purity by HPLC: 97.6%

str1
Flag Counter

AS ON AUG2023 4,071,221 VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@amcrasto

///////////

//////////PHASE 3, MSC2364447CM-2951MSC-2364447CZA45457L1KM2951, M 2951, Evobrutinib

wdt-2

NEW DRUG APPROVALS

ONE TIME

$10.00

TEREVALEFIM


2D chemical structure of 1070881-42-3
Structure of TEREVALEFIM

TEREVALEFIM

Molecular Formula

  • C9-H8-N2-S

Molecular Weight

  • 176.2382

RN: 1070881-42-3
UNII: GG91UXK2M5

  • 5-((E)-2-Thiophen-2-yl-vinyl)-lh-pyrazole
  • 1H-Pyrazole, 3-((1E)-2-(2-thienyl)ethenyl)-
  • ANG-3777
  • SNV-003
  • OriginatorAngion Biomedica
  • ClassAnti-ischaemics; Antifibrotics; Heart failure therapies; Pyrazoles; Small molecules; Thiophenes; Urologics; Vascular disorder therapies
  • Mechanism of ActionProto oncogene protein c met stimulants
  • Orphan Drug StatusYes – Renal failure
  • Phase IIIDelayed graft function
  • Phase IIAcute kidney injury; Acute lung injury; Renal failure
  • PreclinicalBrain injuries
  • No development reportedHeart failure
  • DiscontinuedHepatic fibrosis; Myocardial infarction; Stroke
  • 02 Aug 2022Vifor Pharma has been acquired by CSL and renamed to CSL Vifor
  • 14 Dec 2021Efficacy and adverse events data of a phase II GUARD trial in Acute kidney injury released by the company
  • 26 Oct 2021Top-line efficacy and adverse events data from the phase III trial GIFT (Graft Improvement Following Transplant) trial in Delayed graft function released by Angion Biomedica and Vifor Pharma

Terevalefim, an hepatocyte growth factor (HGF) mimetic, selectively activates the c-Met receptor.

PATENT

WO 2004/058721

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2004058721

PATENT

PCT Application No. PCT/US2003/040917, filed December 19, 2003 and published as WO2004/058721 on July 15, 2004, the entirety of which is hereby incorporated by reference, describes certain compounds that act as HGF/SF mimetics . Such compounds include terevalefim:

Terevalefim has been demonstrated to be remarkably useful for treatment of a variety of conditions including, for example, fibrotic liver disease, ischemia-reperfusion injury, cerebral infarction, ischemic heart disease, renal disease, lung fibrosis, damaged and/or ischemic organs, transplants or grafts, stroke, cerebrovascular disease, and renal fibrosis, among others (see, for example, WO 2004/058721, WO 2010/005580, US 2011/0230407, US 7879898, and WO 2009/064422, each of which is hereby incorporated by reference.) Exemplary methods of using terevalefim for, eg, treating delayed graft function after kidney transplantation and acute lung injury, are described in WO 2021/087392 and WO 2021/183774, each of which is hereby incorporated by reference. In particular, Terevalefim is or has been the subject of clinical trials for delayed graft function in recipients of a deceased donor kidney (Clinicaltrials.gov identifier: NCT02474667), acute kidney injury after cardiac surgery involving cardiopulmonary bypass (Clinicaltrials.gov identifier: NCT02771509), and COVID -19 pneumonia (Clinicaltrials.gov identifier: NCT04459676). Without wishing to be bound by any particular theory, it is believed that terevalefim’s HGF mimetic capability imparts a variety of beneficial attributes and activities.

[0035] Terevalefim has a CAS Registry No. of 1070881-42-3 and is also known by at least the following names:

● 3-[(1E)-2-(thiophen-2-yl)ethen-1-yl]-1H-pyrazole; and

● (E)-3-[2-(2-thienyl)vinyl]-1H-pyrazole.

Synthesis of Terevalefim

[0057] In some embodiments, the present disclosure provides methods for preparing compounds useful as HGF/SF mimetics, such as terevalefim. A synthesis of terevalefim is described in detail in Example 7 of WO 2004/058721 (“the ‘721 Synthesis”). The ‘721 Synthesis is depicted in Scheme 1:

The ‘721 Synthesis includes certain features which are not desirable for preparation of terevalefim at scale and/or with consistency and/or with suitable purity for use in humans. For example, the ‘721 Synthesis includes preparation of aldehyde compound 1.2, a viscous oil that is difficult to purify with standard techniques. Additionally, the ‘721 Synthesis uses a diethoxyphosphorylacetaldehyde tosylhydrazone reagent in step 1-2. As such, step 1-2 has poor atom economy and results in multiple byproducts that must be purified away from the final product of terevalefim. Step 1-2 also uses sodium hydride, a highly reactive base that can be difficult to control and often results in byproducts that must be purified away from the final product of terevalefim. Such purification steps can be costly and time-consuming. In some embodiments, the present disclosure encompasses the recognition that one or more features of the ‘721 Synthesis can be improved to increase yield and/or increase reliability and/or increase scale and/or reduce byproducts. In some embodiments, the present disclosure provides such a synthesis, as detailed herein.

[0059] In some embodiments, the present disclosure provides a synthesis of terevalefim as depicted in Scheme 2:

Scheme 2

wherein X and R 1 are defined below and in classes and subclasses as described herein.

[0060] It will be appreciated that compounds described herein, eg, compounds in Scheme 2, may be provided and/or utilized in a salt form. For example, compounds which contain a basic nitrogen atom may form a salt with a suitable acid. Alternatively and/or additionally, compounds which contain an acidic moiety, such as a carboxylic acid group, may form a salt with a suitable base. Suitable counterions are well known in the art, eg, see generally, March ‘s Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, MB Smith and J.

March, 5 th Edition, John Wiley & Sons, 2001. All forms of the compounds in Scheme 2 are contemplated by and within the scope of the present disclosure.

Step 2-1 of Scheme 2

[0061] Step 2-1 includes a condensation-elimination reaction between commercially available thiophene-2-carboxaldehyde (1.1) and acetone to provide an α,β-unsaturated ketone compound (2.1).

[0062] In some embodiments, the present disclosure provides a method comprising steps of:

(i) providing compound 1.1:

(ii) contacting compound 1.1 with acetone in the presence of a suitable base,

to compound provide 2.1:

////////

str1
Flag Counter

AS ON DEC2021 3,491,869 VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@amcrasto

/////////////////////////////////////////////////////////////////////////////

///////TEREVALEFIM, ANG-3777, SNV-003, Phase 3, Delayed graft function

C(=C\c1cccs1)/c2cc[nH]n2

wdt

NEW DRUG APPROVALS

ONE TIME

$10.00

RELACORILANT


Relacorilant.svg
Relacorilant.png

Relacorilant

  • Molecular FormulaC27H22F4N6O3S
  • Average mass586.561 Da

CAS 1496510-51-0

Phase III

UNII-2158753C7E

2158753C7E

[(4aR)-1-(4-fluorophenyl)-6-(1-methylpyrazol-4-yl)sulfonyl-4,5,7,8-tetrahydropyrazolo[3,4-g]isoquinolin-4a-yl]-[4-(trifluoromethyl)pyridin-2-yl]methanone

[(4aR)-1-(4-fluorophenyl)-6-(1-methylpyrazol-4-yl)sulfonyl-4,5,7,8-tetrahydropyrazolo[3,4-g]isoquinolin-4a-yl]-[4-(trifluoromethyl)pyridin-2-yl]methanone

Methanone, [(4aR)-1-(4-fluorophenyl)-1,4,5,6,7,8-hexahydro-6-[(1-methyl-1H-pyrazol-4-yl)sulfonyl]-4aH-pyrazolo[3,4-g]isoquinolin-4a-yl][4-(trifluoromethyl)-2-pyridinyl]-

Methanone, ((4aR)-1-(4-fluorophenyl)-1,4,5,6,7,8-hexahydro-6-((1-methyl-1H-pyrazol-4-yl)sulfonyl)-4ah-pyrazolo(3,4-g)isoquinolin-4a-yl)(4-(trifluoromethyl)-2-pyridinyl)-

релакорилант[Russian][INN]

ريلاكوريلانت[Arabic][INN]

瑞拉可兰[Chinese][INN]

  • OriginatorCorcept Therapeutics
  • ClassAntineoplastics; Fluorine compounds; Isoquinolines; Ketones; Organic sulfur compounds; Pyrazoles; Pyridines; Small molecules
  • Mechanism of ActionGlucocorticoid receptor antagonists
  • Orphan Drug StatusYes – Pancreatic cancer; Cushing syndrome
  • Phase IIICushing syndrome; Ovarian cancer; Pancreatic cancer
  • Phase IIFallopian tube cancer; Peritoneal cancer; Prostate cancer
  • Phase I/IISolid tumours
  • Phase IAdrenocortical carcinoma

Most Recent Events

  • 09 Sep 2022Subgroup analysis efficacy data from a phase-II trial in Ovarian cancer presented at the 47th European Society for Medical Oncology Congress (ESMO-2022)
  • 29 Jun 2022Phase-III clinical trials in Ovarian cancer (Combination therapy, Recurrent, Second-line therapy or greater) in USA (PO)
  • 06 Jun 2022Corcept Therapeutics announces intentions to submit a NDA for Ovarian cancer

Relacorilant (developmental code name CORT-125134) is an antiglucocorticoid which is under development by Corcept Therapeutics for the treatment of Cushing’s syndrome.[1] It is also under development for the treatment of solid tumors and alcoholism.[1][2] The drug is a nonsteroidal compound and acts as an antagonist of the glucocorticoid receptor.[1] As of December 2017, it is in phase II clinical trials for Cushing’s syndrome and phase I/II clinical studies for solid tumors, while the clinical phase for alcoholism is unknown.[1]

Relacorilant is an orally available antagonist of the glucocorticoid receptor (GR), with potential antineoplastic activity. Upon administration, relacorilant competitively binds to and blocks GRs. This inhibits the activity of GRs, and prevents both the translocation of the ligand-GR complexes to the nucleus and gene expression of GR-associated genes. This decreases the negative effects that result from excess levels of endogenous glucocorticoids, like those seen when tumors overproduce glucocorticoids. In addition, by binding to GRs and preventing their activity, inhibition with CORT125134 also inhibits the proliferation of GR-overexpressing cancer cells. GRs are overexpressed in certain tumor cell types and promote tumor cell proliferation.

CLIP

https://europepmc.org/article/pmc/pmc8175224

Relacorilant (CORT125134)118) is being developed by Corcept Therapeutics, Inc. It is an orally active, high-affinity, selective antagonist of the glucocorticoid receptor that may benefit from the modulation of cortisol activity. In structural optimization, the introduction of a trifluoromethyl group to the 4-position on the pyridyl moiety was found to increase HepG2 tyrosine amino transferase assay potency by a factor of four. Relacorilant is currently being evaluated in a phase II clinical study in patients with Cushing’s syndrome.119)

2-Bromo-4-(trifluoromethyl)pyridine (17) prepared from (E)-4-ethoxy-1,1,1-trifluorobut-3-en-2-one is employed as a key intermediate for the preparation of relacorilant as shown in Scheme 31.120)

An external file that holds a picture, illustration, etc. Object name is jps-46-2-D21-012-scheme31.jpg

Scheme31. Synthesis of relacorilant.118)

118) H. Hunt, T. Johnson, N. Ray and I. Walters (Corcept Therapeutics, Inc.): PCT Int. Appl. WO2013/177559 (2013).

119) H. J. Hunt, J. K. Belanoff, I. Walters, B. Gourdet, J. Thomas, N. Barton, J. Unitt, T. Phillips, D. Swift and E. Eaton: Identification of the Clinical Candidate (R)-(1-(4-Fluorophenyl)-6-((1-methyl-1H-pyrazol-4-yl)sulfonyl)-4,4a,5,6,7,8-hexahydro-1H-pyrazolo[3,4-g]isoquinolin-4a-yl)(4-(trifluoromethyl)pyridin-2-yl)methanone (CORT125134): A Selective Glucocorticoid Receptor (GR) Antagonist. J. Med. Chem. 60, 3405–3421 (2017). [Abstract] [Google Scholar]

120) B. Lehnemann, J. Jung and A. Meudt (Archimica GmbH): PCT Int. Appl. WO 2007/000249 (2007).

PAPER

https://pubs.acs.org/doi/abs/10.1021/acs.jmedchem.7b00162

The nonselective glucocorticoid receptor (GR) antagonist mifepristone has been approved in the U.S. for the treatment of selected patients with Cushing’s syndrome. While this drug is highly effective, lack of selectivity for GR leads to unwanted side effects in some patients. Optimization of the previously described fused azadecalin series of selective GR antagonists led to the identification of CORT125134, which is currently being evaluated in a phase 2 clinical study in patients with Cushing’s syndrome.

Abstract Image

PATENT

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2013177559

SYN

////////

str1
Flag Counter

AS ON DEC2021 3,491,869 VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@amcrasto

/////////////////////////////////////////////////////////////////////////////

Cushing’s syndrome (CS) is a metabolic disorder caused by chronic hypercortisolism. CS is associated with cardiovascular, metabolic, skeletal and psychological dysfunctions and can be fatal if left untreated. The first-line treatment for all forms of CS is a surgery. However, medical therapy has to be chosen if surgical resection is not an option or is deemed ineffective. Currently available therapeutics are either not selective and have side effects or are only available as an injection (pasireotide).

References

  1. Jump up to:a b c d “Relacorilant – Corcept Therapeutics – AdisInsight”.
  2. ^ Veneris JT, Darcy KM, Mhawech-Fauceglia P, Tian C, Lengyel E, Lastra RR, Pejovic T, Conzen SD, Fleming GF (2017). “High glucocorticoid receptor expression predicts short progression-free survival in ovarian cancer”Gynecol. Oncol146 (1): 153–160. doi:10.1016/j.ygyno.2017.04.012PMC 5955699PMID 28456378.

External links

Clinical data
Other namesCORT-125134
Routes of
administration
By mouth
Drug classAntiglucocorticoid
Identifiers
showIUPAC name
CAS Number1496510-51-0
PubChem CID73051463
ChemSpider57617720
UNII2158753C7E
KEGGD11336
Chemical and physical data
FormulaC27H22F4N6O3S
Molar mass586.57 g·mol−1
3D model (JSmol)Interactive image
showSMILES
showInChI

//////////////Relacorilant, Phase III , Orphan Drug, Cushing syndrome, Ovarian cancer, Pancreatic cancer, релакорилант , ريلاكوريلانت , 瑞拉可兰 , 

CN1C=C(C=N1)S(=O)(=O)N2CCC3=CC4=C(CC3(C2)C(=O)C5=NC=CC(=C5)C(F)(F)F)C=NN4C6=CC=C(C=C6)F

wdt-1

NEW DRUG APPROVALS

ONE TIME

$10.00

CILENGITIDE


Cilengitide.svg
ChemSpider 2D Image | cilengitide | C27H40N8O7
Cilengitide.png
IUPAC Condensedcyclo[Arg-Gly-Asp-D-Phe-N(Me)Val]
HELMPEPTIDE1{R.G.D.[dF].[meV]}$PEPTIDE1,PEPTIDE1,5:R2-1:R1$$$
IUPACcyclo[L-arginyl-glycyl-L-alpha-aspartyl-D-phenylalanyl-N-methyl-L-valyl]

CILENGITIDE

  • Molecular FormulaC27H40N8O7
  • Average mass588.656 Da

2-[(2S,5R,8S,11S)-5-benzyl-11-[3-(diaminomethylideneamino)propyl]-7-methyl-3,6,9,12,15-pentaoxo-8-propan-2-yl-1,4,7,10,13-pentazacyclopentadec-2-yl]acetic acid188968-51-6[RN]
4EDF46E4GI
7823
циленгитид 
سيلانجيتيد 
西仑吉肽 

EMD 121974EMD-121974UNII-4EDF46E4GI

2-[(2S,5R,8S,11S)-5-benzyl-11-[3-(diaminomethylideneamino)propyl]-7-methyl-3,6,9,12,15-pentaoxo-8-propan-2-yl-1,4,7,10,13-pentazacyclopentadec-2-yl]acetic acid

Cilengitide has been in phase III clinical trials by Merck Serono and NCI for the treatment of glioblastoma multiforme. However, this research has been discontinued.

Cilengitide was originally developed by Merck KGaA in collaboration with the Technical University of Munich, then received orphan drug designation from FDA for the treatment of glioma in 2005.

Cilengitide (EMD 121974) is a molecule designed and synthesized at the Technical University Munich in collaboration with Merck KGaA in Darmstadt. It is based on the cyclic peptide cyclo(-RGDfV-), which is selective for αv integrins, which are important in angiogenesis (forming new blood vessels), and other aspects of tumor biology. Hence, it is under investigation for the treatment of glioblastoma, where it may act by inhibiting angiogenesis, and influencing tumor invasion and proliferation.[1][2]

The European Medicines Agency has granted cilengitide orphan drug status.[3]

Cilengitide seems to function by inhibiting the FAK/src/AKT pathway and inducing apoptosis in endothelial cells.[4] Preclinical studies in mice of cilengitide were able to demonstrate efficacious tumor regression.[4]

In a rat xenograft model, cilengitide was able to potentiate the cytotoxic effects of radiation when cilengitide was administered prior to radiation therapy.[5] When combined with radiation, inhibition of integrin expression by cilengitide synergistically improves the cytotoxic effects of ionizing radiation for glioblastoma.[5]

Clinical trials

Phase II studies were able to demonstrate that cilengitide as a potential monotherapy in patients with recurrent glioblastoma[6] with high intratumor drug levels when 2000 mg of cilengitide is given twice weekly.[7]

Cilengitide is well tolerated, in combination with radiation and temozolomide, at a dose of 2000 mg in patients with newly diagnosed glioblastoma, regardless of MGMT promoter status.[8] In a phase I/IIa study, the addition of cilengitide to the standard of care for newly diagnosed glioblastoma (surgical resection followed by temozolomide and radiation therapy) improves progression-free survival and overall survival in patients with MGMT promoter methylation.[9]

However, in a subsequent study, cilengitide does not seem to alter the pattern of glioblastoma progression,[10]

and in an EORTC phase III randomized, controlled, multicenter clinical trial, consisting of over 500 patients in 23 countries, the addition of cilengitide to the standard of care did not improve overall survival in patients with newly diagnosed glioblastoma and methylated MGMT promoter status [11] A phase II study, the CORE trial, is currently being conducted in patients with newly diagnosed glioblastoma and unmethylated MGMT promoter status.[12]

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@amcrasto

/////////////////////////////////////////////////////////////////////////////////////////////////////

SYN

Angewandte Chemie, International Edition, 55(4), 1540-1543; 2016

SYN

Chemistry – A European Journal, 16(18), 5385-5390, S5385/1-S5385/36; 2010

Reference:1. WO0047228A1 / US7115261B1.

2. US6001961A.Route 2

Reference:1. CN102731627A.PATENTWO/2021/224234ANTIVIRAL USE OF CILENGITIDEhttps://patentscope.wipo.int/search/en/detail.jsf?docId=WO2021224234&_cid=P20-KW0M52-85135-1

PATENThttps://patents.google.com/patent/CN102731627A/enEMD121974 (Cilengitide), the Chinese another name: ring (L-arginyl glycyl-L-aspartoyl-D-phenylalanyl-N-methyl-L-valyl) is an a kind of new classification cancer therapy drug of synthetic.Merkel company discovers that EMD121974 amalgamation radiotherapy (merging to reach assists TM to add radiotherapy) possibly prolong lifetime; Simultaneously integrate plain supressor antitumor drug as first; Got into the III clinical trial phase, its important mechanism is to grow targeting that the blood supply structure of nutrition, the growth of promotion cancer cell is provided in tumour and for tumour through line artery.The EMD121974 molecular formula is: C 274087, have following structure: 
The preparation method of cyclic peptide mainly contains liquid phase synthesis process, solid phase synthesis precursor peptide cyclization process, process for solid phase synthesis in liquid phase at present; Wherein preceding two kinds of synthesis techniques all are the cyclisation in liquid phase of synthetic precursor peptide, and this method needs reactant in extremely rare solvent, to react (10 -3~10 -4Mol/L), and intermolecular be prone to react generation line style or cyclic polymer, greatly reduced the cyclisation yield, bring trouble for follow-up purifying, and in large-scale production, produce a large amount of waste liquids, be unfavorable for suitability for industrialized production.In conjunction with the structure of EMD121974, utilize the false rare principle of benefit of solid phase, developed a kind of efficient cyclization reaction, the cyclisation time shortens to 20%~30% of liquid phase cyclisation, and the 2%-8% of solvent as liquid phase used in reaction.Embodiment 1The preparation of Fmoc-L-Asp (OtBu)-Wang ResinThe Wang Resin that takes by weighing the 10g substitution degree and be 0.5mmol/g joins in the reactor drum, adds an amount of DCM, and swelling 30min takes out DCM; 6.17g Fmoc-L-Asp-OtBu, DIC 2.40ml, HOBT2.1g are dissolved among the 30ml DMF; At 0-5 ℃ of activation 15min, activation solution is joined in the reactor drum that contains Wang Resin, behind the reaction 10min; Add DMAP 0.18g again, at 0~30 ℃ of reaction 1~5h.After reaction finishes, add sealing Wang Resin unreacted hydroxylation reagent diacetyl oxide 1ml and pyridine 0.5ml, behind the capping 1h, DMF, DCM, the CH of 80ml used in washing successively 3OH, DMF washing 2,1,1,2 times, each 1min.Through detecting, obtain the Fmoc-L-Asp that substitution degree is 0.47mmol/g (OtBu)-Wang Resin.Embodiment 2The EMD121974 precursor:The preparation of A-Wang Resin (Fmoc-D-Phe-N-Me-L-Val-L-Arg (Mtr)-Gly-L-Asp (OtBu)-Wang Resin)Fmoc-L-Asp (OtBu)-Wang Resin is joined in the reactor drum, behind DMF swelling 30min, take out solvent, the piperidines-DMF that adds 80ml 25% reacts 5min, and 80ml DMF washs 1 time (3min), and the piperidines-DMF that adds 80ml 25% reacts 15min; DMF, DCM, the CH of 80ml used in washing successively 3OH, DMF washing 2,1,1,2 times, each 1min; With 4.45g Fmoc-Gly-OH, 5.68g HBTU, 2.03g HOBt, be dissolved among the DMF of 30ml, dissolve the back and added DIEA 2.45ml; 0~5 ℃ of activation 15min; Activation solution is joined in the above-mentioned reactor drum, and behind reaction 1-3h under 0~30 ℃, reaction end detects with ninhydrin method.Adopt aforesaid method coupling Fmoc-L-Arg (Mtr)-OH, Fmoc-N-Me-L-Val, Fmoc-D-Phe-OH successively, finally obtain Fmoc-D-Phe-N-Me-L-Val-L-Arg (Mtr)-Gly-L-Asp (OtBu)-Wang Resin.Embodiment 3EMD121974 precursor peptide: the preparation of B-Wang Resin (D-Phe-N-Me-L-Val-L-Arg (Mtr)-Gly-L-Asp-Wang Resin)With volume ratio is that piperidines-DMF of 25% is the Fmoc deprotection agent of Fmoc-D-Phe-N-Me-L-Val-L-Arg (Mtr)-Gly-L-Asp (OtBu)-Wang Resin; Add piperidines-DMF 80ml of 25% first time; Reaction 5min, 80ml DMF washs 1 time (3min), adds piperidines-DMF 80ml of 25% for the second time; Behind the reaction 15min, DMF, DCM, the CH of 80ml used in washing successively 3OH, DMF washing 2,1,1,2 times, each 1min gets D-Phe-N-Me-L-Val-L-Arg (Mtr)-Gly-L-Asp (OtBu)-Wang Resin after washing finishes.80% the PhOH-DCM solution that adds volume ratio and be 100ml takes off OtBu with the TFA of catalytic amount, reacts 8h; DMF, DCM, the CH of 80ml used in washing successively 3OH, DMF washing 2,1,1,2 times, each 1min gets D-Phe-N-Me-L-Val-L-Arg (Mtr)-Gly-L-Asp-Wang Resin.Embodiment 4The preparation of EMD121974-Wang Resin (Cyclo (D-Phe-N-Me-L-Val-L-Arg-Gly-L-Asp)-Wang Rsin)In above-mentioned reactor drum, add cyclization reagent 3.9g DPPA, 2.5ml DIEA (reactant cyclization reagent amount of substance ratio is 1: 3), at 10~40 ℃ of reaction 3h, the multiple cyclization reagent reaction 3~5h (reaction end detects with ninhydrin method) that throws once above-mentioned equivalent; DMF, DCM, the CH of 80ml used in washing successively 3OH washing 2,1,3 times, each 3min gets Cyclo (D-Phe-N-Me-L-Val-L-Arg-Gly-L-Asp)-Wang Rsin.Embodiment 5The preparation of EMD121974 (Cyclo (D-Phe-N-Me-L-Val-L-Arg-Gly-L-Asp))In above-mentioned reactor drum, add the TFA/H of lytic reagent 120ml again 2Behind O/TlS (volume ratio is 95: 2.5: 2.5) the reaction 3h, suction filtration is removed resin, and filtrating slowly joins in the no water-ice ether; Static 2-5h, high speed centrifugation obtain thick peptide, prepare through high-pressure liquid phase; Lyophilize gets smart EMD121974; Its purity>99.5%, single impurity<0.2%, total recovery reaches 63%.Choosing substitution degree in the present embodiment is the Wang Resin of 0.5mmol/g, and can also choose substitution degree is the arbitrary Wang Resin and Fmoc-L-Asp-OtBu prepared in reaction Fmoc-L-Asp (the OtBu)-Wang Resin of 0.4~0.9mmol/g scope.All can realize technical scheme of the present invention, and obtain technique effect of the present invention.Above content is an EMD121974 and become one of best preferred version of route; And to further explain that the present invention did; But can not assert that practical implementation of the present invention is only limited to these explanations; Under the prerequisite that does not break away from the present invention’s design, can also make some simple deductions and replacement, all should be regarded as protection domain of the present invention. 
CLIPhttps://www.eurekaselect.net/article/2607Cilengitide, a cyclic RGD pentapeptide, is currently in clinical phase III for treatment of glioblastomas and in phase II for several other tumors. This drug is the first anti-angiogenic small molecule targeting the integrins αvβ3, αvβ5 and α5β1. It was developed by us in the early 90s by a novel procedure, the spatial screening. This strategy resulted in c(RGDfV), the first superactive αvβ3 inhibitor (100 to 1000 times increased activity over the linear reference peptides), which in addition exhibited high selectivity against the platelet receptor αIIbβ3. This cyclic peptide was later modified by N-methylation of one peptide bond to yield an even greater antagonistic activity in c(RGDf(NMe)V). This peptide was then dubbed Cilengitide and is currently developed as drug by the company Merck-Serono (Germany). This article describes the chemical development of Cilengitide, the biochemical background of its activity and a short review about the present clinical trials. The positive anti-angiogenic effects in cancer treatment can be further increased by combination with “classical” anti-cancer therapies. Several clinical trials in this direction are under investigation. 
CLIPJournal of Protein Chemistry

Schematic of the one-step chemoenzymatic synthesis of cilengitide using wild-type Mcy TE. (1) The chemically synthesised (SPPS, solid-phase peptide synthesis) mimetic substrate was condensed with benzyl mercaptane to produce pentapeptide thioester (pentapeptide-BMT). (2) Models of the substrate-O-TE acyl enzyme intermediate are marked with brackets (protein data bank, 1JMK). (3) Mechanism of TE domain catalysis: a pentapeptide -O-TE acyl-enzyme intermediate is formed by transfer of the peptidyl chain from the phosphopantethiene of the terminal peptidyl carrier protein (PCP), which was substituted by benzyl mercaptane, to the active site serine of the TE domain. For hydrolyzing TE domains, the intermediate is captured by water, generating the linear peptide; for cyclizing TE domains, an intramolecular nucleophile captures the intermediate, resulting in “cilengitide”

Schematic of the one-step chemoenzymatic synthesis of cilengitide using wild-type Mcy TE. (1) The chemically synthesised (SPPS, solid-phase peptide synthesis) mimetic substrate was condensed with benzyl mercaptane to produce pentapeptide thioester (pentapeptide-BMT). (2) Models of the substrate-O-TE acyl enzyme intermediate are marked with brackets (protein data bank, 1JMK). (3) Mechanism of TE domain catalysis: a pentapeptide -O-TE acyl-enzyme intermediate is formed by transfer of the peptidyl chain from the phosphopantethiene of the terminal peptidyl carrier protein (PCP), which was substituted by benzyl mercaptane, to the active site serine of the TE domain. For hydrolyzing TE domains, the intermediate is captured by water, generating the linear peptide; for cyclizing TE domains, an intramolecular nucleophile captures the intermediate, resulting in “cilengitide” 
PATENTWO 9745447 
WO 9745137 
DE 19534177 
WO 2000053627 
WO 2000047228 
US 20040063790 
WO 2009124754 
WO 2011079015 
 WO 2011069629 
 WO 2011144756WO 2016059622 
PATENTWO 2012062777https://patents.google.com/patent/WO2012062777A1/enSynthesis of cyclic peptidesCyclo[-Arg-Gly-Asp- 6 or 7 -Phe-Val-Ala-] (1 and 2). Resin loading. 2- chlorotrityl chloride-resin ( 1 50 m g , 1 .5m m ol/g ) was p laced i n a 20 m l polypropylene syringe fitted with a polyethylene filter disk. The resin was then washed with CH2CI2 (5 χ 0.5 min), and a solution of Fmoc-L-Gly-OH (334 mg, 1 .125 mmol, 5 equiv) and DIEA (239 μΙ_, 6.25 equiv) in CH2CI2 (2.5 ml_) was added. The mixture was then stirred for 15 min. Extra DIEA (239 μΙ_, total 12.5 mmol) was added, and the mixture was stirred for an additional 45 min. The reaction was stopped by adding 3 χ DCM/ MeOH/ DIEA (85: 10:5) and stirring for 1 0 m in. The Fmoc-L-Gly-O-resin product was subjected to the following washings/treatments with CH2CI2 (3 χ 0.5 min), DMF (3 χ 0.5 min), piperidine and DMF (5 χ 0.5 min). The loading was 0.50 mmol/g, as calculated by Fmoc determination.Peptide coupling. Fmoc-L-Arg(Pbf)-OH (243 mg, 0.375 mmol, 5 equiv), Fmoc- L-Ala-OH (1 17 mg, 0.375 mmol, 5 equiv), Fmoc-L-Val-OH ( 127 mg, 0.375 mmol, 5 equiv) and Fmoc- L-Phe-OH ( 145 mg, 0.375 mmol, 5 equiv) were added sequentially to the above obtained H-L-Gly-O-resin using HCTU (155 mg, 0.375 mmol, 5 equiv), HOBt (50 mg, 0.375 mmol, 5 equiv) and DIEA (127 μΙ_, 0.75 mmol, 10 equiv) in DMF (2.5 ml_). In all cases, after 90 min of coupling, the ninhydrin test was negative. Removal of Fmoc group and washings were performed as described in general procedures. /V-Alloc-thiazole 6 or 7 (92 mg, 0.375 mmol, 5 equiv) was coupled with HATU (143 mg, 0.375 mmol, 5 equiv), HOAt (51 mg, 0.375 mmol, 5 equiv) and DIEA (127 μΙ_, 0.75 mmol, 10 equiv) for 90 min. This coupling was repeated twice in the same conditions. The Alloc group of the peptide resin was removed with Pd (PPh3)4 (9 mg, 0.0075 mmol, 0.1 equiv) in the presence of PhSiH3 (92.5 μΙ_, 0.75 mmol, 10 equiv) in DCM for 20 min. This deprotection was repeated three times in the same conditions. After washing, the resin was treated with dry THF (2ml_) for 15 min. Meanwhile, Fmoc-L-Asp(tBu)-OH (154 mg, 0.375 mmol, 5 equiv) was added to a 68 mM solution of triphosgene in dry THF (1 .15 equiv). Sym-collidine (99.5 μΙ_, 0.75 mmol, 10 equiv) was added to the clear solution, upon which a precipitate of collidinium chloride was formed. DIEA (102 μΙ_, 0.6 mmol, 8 equiv) was added to the resin, immediately followed by addition of the suspension. This coupling was repeated four times in the same conditions. The reaction mixture was stirred at 50 °C during 48 h.Peptide cleavage. Following Fmoc deprotection, the peptidyl-resin was treated with TFA-CH2CI2 (1 :99) (5 χ 30 s). The filtrate was collected on H20 (4 ml_) and the H20 was partially removed under reduced pressure. MeCN was then added to dissolve solid that formed during the removal of H20, and the solution was lyophilized to give 12 mg and 10 mg of the linear compounds 28 and 29 respectively with a purity of > 91 % as checked by HPLC (Column A, Rt 7.43 min and Rt 7.38 min respectively, linear gradient 35%-40% ACN in 15 min.)], which was used without further purification. MALDI-TOF-MS calculated for C50H71 N11 O13S2 1098.29; found mlz 1099.29 [M + H]+, 1 121 .28 [M + Na]+, 1 137.39 [M + K]+.Synthesis in solution. Cyclization. The protected linear peptides 28 and 29 were dissolved in DMF (1 L, 10“4 M), and HOAt (9.6 mg, 0.07 mmol, 5 equiv), DIPEA (24 μΙ_, 0.14 mmol, 10 equiv), and PyAOP (36.6 mg, 0.07 mmol, 5 equiv) were added. The mixture was stirred for 24 h at room temperature, and the course of the cyclization step was then checked by HPLC (Column A, Rt 1 1 -67 min and Rt 10.70 min respectively, linear gradient 45%-55% ACN in 15 min.). The solvent was removed by evaporation under reduced pressure and the protected cycle 30 and 31 were used in the next step without further purification. MALDI-TOF-MS calculated for C50H69N11 O12S2 1080.28; found mlz 1081 .28 [M + H]+, 1 103.27 [M + Na]+, 1 1 19.38 [M + K]+.Side chain deprotection. The protected cyclopeptides 30 and 31 (14.7 mg, 19.04 pmol) were treated with TFA-H20 (95: 5) during 1 h. The solvent was removed by evaporation under reduced pressure.Peptide purification. The crude product was purified by HPLC (Symmetry C8 5 μη-Ί, 30 mm x 100 mm), gradient of MeCN (30% to 75% in 15 min) MeCN (+0.05% TFA) in water (+0.05% TFA), 20 mL/min, detection at 220 nm, to give the cyclopeptides 1 and 2 (4.5 mg, 5.8 pmol and 6.5 mg, 8.37 pmol, 7.7% and 12% yield respectively). The products were characterized by HPLC (Rt 8.99 min, and Rt 8.02 min Column A, respectively, linear gradient 0%-100% ACN in 1 5 min. ) and by MALDI-TOF-MS: calculated for C33H45N11 O9S 771 .84; found mlz 772.84 [M + H]+, 794.83 [M + Na]+, 810.94 [M + K]+.Cyc/o-[Arg-Gly-Asp-Thz1X-] (3). General procedure for cyclopeptide synthesis. Solid phase synthesis: The synthesis of the linear peptide H- Asp(tBu)-XX-Arg(Pbf)-Gly-OH was performed using Fmoc-based solid phase peptide synthesis with 2-chlorotrityl chloride resin (2.0 g, 3.2 mmol).Resin loading: Fmoc-Gly-OH (594 mg, 2.0 mmol) was attached to the resin with DIPEA in DCM at room temperature for 1 .5 h. The remaining trityl groups were capped adding 0.5 mL of MeOH for 30 min. After that, the resin was filtered and washed with DCM (2x), DMF (2x). The loading of the resin was determined by titration of the Fmoc group (Chan WC and White PD. Fmoc Solid Phase Peptide Synthesis. Oxford University Press: New York, 2000). The final loading was 2.0 mmol/g. The Fmoc group was eliminated by treatment with 20% piperidine in DMF (2X10 min). The resin was washed with DMF (3x), DCM (3x). Peptide coupling: Fmoc-Arg(Pbf)-OH (5.19 g, 8.0 mmol), DIPCDI (1.23 mL, 8.0 mmol) and HOBt (1.08 g, 8.0 mmol) were dissolved in DMF and added to the resin for 1 .5 h. The end of the coupling was monitored by ninhydrin test (free amine group) (Kaiser E et al. Anal Biochem 1970, 34:595-598). The resin was filtered and washed with DMF (3X) and DCM (3X). The Fmoc group was eliminated with 20 % piperidine in DMF (2X10 min).The coupling of the thiazole module was carried out with 8 (1 .14 g, 3.0 mmol), PyAOP (1 .56 g, 3.0 mmol) and DIPEA (1 .02 mL, 6.0 mmol) in DMF for 1 .5 h. The completion of the reaction was checked with the ninhydrin test. Finally the deprotection of the amine and coupling of the Fmoc-Asp(‘Bu)-OH were carried out under the same conditions of the second amino acid.Peptide cleavage: The resin bound peptide was treated with 2% TFA in DCM (6 x 30 sec.) The resin was washed with DCM and the combined solution was evaporated under vacuum with Et20 several times, furnishing the linear peptide 32 as a white solid. The peptide was used for the next step without purification.H PLC (gradient 20 to 80% of CH3CN in 1 5 m in): tR= 8.33 min. HPLC-MS (ES(+)): m/z 795.3.Synthesis in solution. Cyclization: The product 32 (200 mg, 0.251 mmol) was dissolved in anhydrous DMF (50 mL, 5 mM), PyAOP (262 mg, 0.503 mmol) and DIPEA (213 μί, 1 .255 mmol) were added. The reaction was monitored by HPLC. Once the reaction was finished, the DMF was evaporated under vacuum. The crude was dissolved in AcOEt and the solution was washed with NH4CISat and Na2CO3 sat. The organic layer was collected, dried over Na2SO4, filtered and concentrated under vacuum. The peptide was purified by flash chromatography (CHCIs/MeOH 8:2) furnishing the protected cyclic peptide 33 as a white solid (1 56 mg, XX%). HPLC (gradient 40 to 90% of CH3CN in 1 5 min): tR= 8.86 min. HPLC-MS (ES(+)): m/z 778.2Side chain deprotection: The protected peptide 33 (125 mg, XX mmol), was treated with 25 mL of a solution of TFA H2O (95:5). After 3 h, the solvent was evaporated under vacuum and the residue was precipitated with Et2O (4X). The Et2O solution was discarded and the white solid was lyophilized to afford 3 55 mg (XX%).

Peptide purification. The end product 3 was dissolved in 5 ml MilliQ water and it was filtered through a 0.2 pm filter. The cyclic peptide was purified by semipreparative RP-HPLC using acetronitrile (0.05% TFA)/water (0.1 % TFA). The HPLC sample was vacuum concentred and transformed into the hydrochloride salt lyophilized with water with 0.05% HCI.1H-NMR (500 MHz, H20:D20-d2 9: 1 , 278 K): δ = 9.29 (t, NH Gly), 9.20 (d, J = 7.24 Hz, NH Asp), 8.90 (t, J = 5.89/5.89 Hz, NH Thz), 8.46 (d, J = 8.93 Hz, NH Arg), 7.79 (s, CH Thz), 7.22 (t, J = 5.39/5.39 Hz, ΝΗε Arg), 4.75 (m, CHa Arg), 4.63 (m, CHa Asp), 4.04 (dd, J = 3.35/14.90 Hz, CHa Gly), 3.82 (dd, J = 6.69/14.96 Hz, CHa Gly), 3.17 (m, CH25 Arg), 2.89 (m, CH2p Asp), 1 .92 (m, CH p Arg), 1 .82 (m, CHP Arg), 1 .63 (m, CH2 Arg). HPLC (gradient 0 to 20% of CH3CN in 15 min): tR= 10.52 m in. HRMS (E IS) m/z calculated 468.1540

Figure imgf000047_0001

found 469.16099 (M+H)+.Cyc/o-[Arg-Gly-Asp-Thz2X-] (4). The cyclopeptide 4 was prepared according to the process followed for 3 and using bithiazole 9 (XX mg, YY mmol) instead of 8. The linear peptide 34: HPLC (gradient 0 to 100% CH3CN in 15 min.): tR = 10.34 min, HPLC-MS (ES(+)): m/z 877.81 . The protected peptide 35: HPLC (gradient 0 to 100% CH3CN in 15 min.): tR = 13.91 min, HPLC-MS (ES(+)): m/z 860.54. The final peptide 4: 1H-NMR (500 MHz, H20:D20-d2 9: 1 , 298 K): δ = 8.93 (sbroad, NH Gly), 8.82 (d, J = 7.62 Hz, NH Asp), 8.75 (t, J = 5.69/5.69 Hz, NH Thz), 8.51 (d, J = 7.62 Hz, NH Arg), 8.05 (s, CH Thz1), 7.50 (s, CH Thz2), 7.19 (t, J = 5.38/5.38 Hz, ΝΗε Arg), 4.13 (dd, J = 5.82/14.24 Hz, CH Gly), 3.87 (dd, J = 5.96/15.69 Hz, CH Gly), 3.21 (m , CH25 Arg), 2.94 (m, CH2p Asp), 1 .95 (m , CHP Arg), 1 .87 (m , CHP Arg), 1 .68 (m , CH2y Arg). HPLC (gradient 1 0 to 25% of CH3CN in 1 5 m in): tR = 8.73 min. HRMS (EIS) m/z calculated 551 .1369 (C2oH25N906S2) found 552.14392 (2M+2H)+.Cyc/o-[Arg-Gly-Asp-Thz3X-] (5). The cyclopeptide 5 was prepared according to the process for 3 and using trithiazole 10 (XX mg, YY mmol) instead of 8. The linear peptide 36: HPLC (gradient 20 to 80% of CH3CN in 15 min.): tR = 7.60 min, HPLC-MS (ES(+)): m/z 961 .23. The protected peptide 37: HPLC (gradient 20 to 80% of CH3CN in 15 m in. ): tR = 1 3.13 min, HPLC-MS (ES(+)): m/z 944.3. The final peptide 5: HPLC (gradient 10 to 30% CH3CN in 15 m in): tR = 8.26 m in. HRMS (E IS) m/z calculated 634.1 1 99 (C23H26N10O6S3) found 635.12683 (2M+2H)+1H-NMR (500 MHz, DMSO-d6 298 K): δ = 9.21 (t, J = 5.4, NH Gly), 8.72 (m, NH Asp + NH Thz), 8.37 (s, CH Thz1), 7.96 (d, J = 9.2, NHa Arg), 7.77 (s, CH Thz2), 7.68 (t, J = 6.0, ΝΗε Arg), 7.23 (s, CH Thz3), 4.83 (dd, J = 14.3, 8.5, CHa Arg), 4.72 (dd, J = 16.3, 6.6, CH Thz), 4.59 (m, CH Thz + CHa Asp), 3.89 (d, J = 1 1 .5, CH Gly), 3.59 (d, J = 9.7, CH Gly), 3.13 (dd, J = 12.6, 6.3, CH25 Arg), 2.81 (dd, J = 16.3, 4.3, CHP Asp), 2.58 (dd, J = 16.5, 8.7, CHP Asp), 1 .82 (m, CHP Arg), 1 .71 (m, CHP Arg), 1 .49 (m, CH2y Arg).Cilengitide. The cilengitide was prepared according to the method described in Dechantsreiter MA et al. (J Med Chem 1999, 42:3033-3040). 1H- NMR (500 MHz, H20:D20-d2 9: 1 , 298 K): δ = 8.55 (d, J = 8.06 Hz, NH Asp), 8.37 (d, J = 7.28 Hz, NH Arg), 8.13 ( d, J = 9.19 Hz, NH Phe), 7.97 (m, NH Gly), 7.34 (m, 2H, C6H5 Phe), 7.26 (m, 3H, C6H5 Phe), 7.22 (t, J = 5.53/5.53 Hz, ΝΗε Arg), 5.19 (dd, J = 8.58/16.02 Hz, CHa Phe), 4.56 (dd, J = 7.45/- Hz, CHa Asp), 4.34 (d, J = 10.89 Hz, CHa MeVal), 4.12 (dd, J = 7.80/14.63 Hz, CH Gly), 3.95 (dd, J = 6.84/15.33 Hz, CHa Arg), 3.54 (dd, J = 3.37/14.60 Hz, CH Gly), 3.20 (m , CH25 Arg), 3.02 (m, CH2p Phe), 2.88 (s, CH3 MeVal), 2.84 (dd, J = 7.26/16.68 Hz, CHP Asp), 2.63 (dd, J = 7.60/16.54 Hz, CHP Asp), 2.06 (m, CHP Val), 1 .91 (m, CH2p Arg), 1 .57 (m, CH2 Asp), 0.88 (d, J = 6.55 Hz, CH3 Val1), 0.56 (d, J = 6.49 Hz, CH3 Val2). 
PAPERJournal of medicinal chemistry (1999), 42(16), 3033-40.Peptide Science (2001),  Volume Date2000, 37th, 249-250. Current opinion in investigational drugs (London, England : 2000) (2003), 4(6), 741-5. Journal of medicinal chemistry (2005), 48(24), 7675-87.Peptide Science (2006), 43rd, 215-216Angewandte Chemie, International Edition (2010), 49(15), 2732-2737, S2732/1-S2732/53.Accounts of Chemical Research (2017), 50(7), 1541-1556.

References

  1. ^ Burke PA, DeNardo SJ, Miers LA, Lamborn KR, Matzku S, DeNardo GL (August 2002). “Cilengitide targeting of alpha(v)beta(3) integrin receptor synergizes with radioimmunotherapy to increase efficacy and apoptosis in breast cancer xenografts”. Cancer Research62 (15): 4263–72. PMID 12154028.
  2. ^ Goodman SL, Hölzemann G, Sulyok GA, Kessler H (February 2002). “Nanomolar small molecule inhibitors for alphav(beta)6, alphav(beta)5, and alphav(beta)3 integrins”. Journal of Medicinal Chemistry45 (5): 1045–51. doi:10.1021/jm0102598PMID 11855984.
  3. ^ Spreitzer H (October 27, 2008). “Neue Wirkstoffe – Cilengitide”. Österreichische Apothekerzeitung (in German) (22/2008): 1136–7.
  4. Jump up to:a b Yamada S, Bu XY, Khankaldyyan V, Gonzales-Gomez I, McComb JG, Laug WE (December 2006). “Effect of the angiogenesis inhibitor Cilengitide (EMD 121974) on glioblastoma growth in nude mice”. Neurosurgery59 (6): 1304–12, discussion 1312. doi:10.1227/01.NEU.0000245622.70344.BEPMID 17277694S2CID 19861713.
  5. Jump up to:a b Mikkelsen T, Brodie C, Finniss S, Berens ME, Rennert JL, Nelson K, Lemke N, Brown SL, Hahn D, Neuteboom B, Goodman SL (June 2009). “Radiation sensitization of glioblastoma by cilengitide has unanticipated schedule-dependency”International Journal of Cancer124 (11): 2719–27. doi:10.1002/ijc.24240PMID 19199360.
  6. ^ Reardon DA, Fink KL, Mikkelsen T, Cloughesy TF, O’Neill A, Plotkin S, et al. (December 2008). “Randomized phase II study of cilengitide, an integrin-targeting arginine-glycine-aspartic acid peptide, in recurrent glioblastoma multiforme”. Journal of Clinical Oncology26 (34): 5610–7. CiteSeerX 10.1.1.688.8987doi:10.1200/JCO.2008.16.7510PMID 18981465.
  7. ^ Gilbert MR, Kuhn J, Lamborn KR, Lieberman F, Wen PY, Mehta M, Cloughesy T, Lassman AB, Deangelis LM, Chang S, Prados M (January 2012). “Cilengitide in patients with recurrent glioblastoma: the results of NABTC 03-02, a phase II trial with measures of treatment delivery”Journal of Neuro-Oncology106 (1): 147–53. doi:10.1007/s11060-011-0650-1PMC 4351869PMID 21739168.
  8. ^ Nabors LB, Mikkelsen T, Hegi ME, Ye X, Batchelor T, Lesser G, Peereboom D, Rosenfeld MR, Olsen J, Brem S, Fisher JD, Grossman SA (November 2012). “A safety run-in and randomized phase 2 study of cilengitide combined with chemoradiation for newly diagnosed glioblastoma (NABTT 0306)”Cancer118 (22): 5601–7. doi:10.1002/cncr.27585PMC 3423527PMID 22517399.
  9. ^ Stupp R, Hegi ME, Neyns B, Goldbrunner R, Schlegel U, Clement PM, et al. (June 2010). “Phase I/IIa study of cilengitide and temozolomide with concomitant radiotherapy followed by cilengitide and temozolomide maintenance therapy in patients with newly diagnosed glioblastoma” (PDF). Journal of Clinical Oncology28(16): 2712–8. doi:10.1200/JCO.2009.26.6650PMID 20439646.
  10. ^ Eisele G, Wick A, Eisele AC, Clément PM, Tonn J, Tabatabai G, et al. (March 2014). “Cilengitide treatment of newly diagnosed glioblastoma patients does not alter patterns of progression”(PDF). Journal of Neuro-Oncology117 (1): 141–5. doi:10.1007/s11060-014-1365-xPMID 24442484S2CID 21636884.
  11. ^ Merck Group. “Phase III Trial of Cilengitide Did Not Meet Primary Endpoint in Patients With Newly Diagnosed Glioblastoma, Date accessed: 3/24/2014.”
  12. ^ ASCO Meeting Library. [1] “Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma and methylated O6-methylguanine-DNA methyltransferase (MGMT) gene promoter: Key results of the multicenter, randomized, open-label, controlled, phase III CENTRIC study, Date accessed: 3/24/2014
Names
IUPAC name2-[(2S,5R,8S,11S)-5-benzyl-11-{3-[(diaminomethylidene)amino]propyl}-7-methyl-3,6,9,12,15-pentaoxo-8-(propan-2-yl)-1,4,7,10,13-pentaazacyclopentadecan-2-yl]acetic acid
Identifiers
CAS Number188968-51-6 
3D model (JSmol)Interactive image
ChEMBLChEMBL429876 
ChemSpider154046 
IUPHAR/BPS6597
KEGGD03497 
MeSHCilengitide
PubChem CID176873
UNII4EDF46E4GI 
CompTox Dashboard (EPA)DTXSID9044035 
showInChI
showSMILES
Properties
Chemical formulaC27H40N8O7
Molar mass588.656 g/mol
Density1.417 g/mL
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).
☒ verify (what is  ?)
Infobox references

/////////CILENGITIDE, циленгитид , سيلانجيتيد ,西仑吉肽 , PHASE 3, EMD 121974EMD-121974UNII-4EDF46E4GI, orphan drug , MERCK, glioblastoma

CC(C)C1C(=O)NC(C(=O)NCC(=O)NC(C(=O)NC(C(=O)N1C)CC2=CC=CC=C2)CC(=O)O)CCCN=C(N)N

wdt-12

NEW DRUG APPROVALS

ONE TIME

$10.00

Verdiperstat


Verdiperstat (AZD3241) | MPO Inhibitor | MedChemExpress
Verdiperstat.png

Verdiperstat

AZD 3241; BHV-3241

CAS No. : 890655-80-8

1-(2-propan-2-yloxyethyl)-2-sulfanylidene-5H-pyrrolo[3,2-d]pyrimidin-4-one

4H-​Pyrrolo[3,​2-​d]​pyrimidin-​4-​one, 1,​2,​3,​5-​tetrahydro-​1-​[2-​(1-​methylethoxy)​ethyl]​-​2-​thioxo-

1-(2-isopropoxyethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d] pyrimidin-4-one

l-(2-Isopropoxyethyl)-2-thioxo-l,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one

  • Molecular FormulaC11H15N3O2S
  • Average mass253.321 Da

AZD-3241BHV-3421UNII-TT3345YXVRTT3345YXVRBHV-3241, WHO 10251вердиперстат [Russian] [INN]فيرديبيرستات [Arabic] [INN]维地泊司他 [Chinese] [INN]

  • OriginatorAstraZeneca
  • DeveloperAstraZeneca; Biohaven Pharmaceuticals
  • ClassAntiparkinsonians; Ethers; Organic sulfur compounds; Pyrimidinones; Small molecules
  • Mechanism of ActionPeroxidase inhibitors
  • Orphan Drug StatusYes – Multiple system atrophy
  • Phase IIIMultiple system atrophy
  • Phase II/IIIAmyotrophic lateral sclerosis
  • DiscontinuedParkinson’s disease
  • 23 Jun 20213574186: Added patent info and HE
  • 23 Jun 2021Biohaven Pharmaceuticals has patents pending for the composition of matter of verdiperstat, pharmaceutical compositions and various neurological diseases in Europe, Japan and other countries
  • 01 Nov 2020Brigham and Women’s Hospital plans a phase I trial for Multiple System Atrophy in USA , (NCT04616456)

EU/3/14/1404: Orphan designation for the treatment of multiple system atrophy

This medicine is now known as verdiperstat.

On 16 December 2014, orphan designation (EU/3/14/1404) was granted by the European Commission to Astra Zeneca AB, Sweden, for 1-(2-isopropoxyethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d] pyrimidin-4-one for the treatment of multiple system atrophy.

The sponsorship was transferred to Richardson Associates Regulatory Affairs Limited, Ireland, in March 2019.

The sponsorship was transferred to Biohaven Pharmaceutical Ireland DAC, Ireland, in September 2021.

Key facts

Active substance1-(2-isopropoxyethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d] pyrimidin-4-one (verdiperstat)
Intented useTreatment of multiple system atrophy
Orphan designation statusPositive
EU designation numberEU/3/14/1404
Date of designation16/12/2014
SponsorBiohaven Pharmaceutical Ireland DAC

VERDIPERSTAT

For Initial Indications in Multiple System Atrophy (MSA) and Amyotrophic Lateral Sclerosis (ALS)

Verdiperstat is a first-in-class, potent, selective, brain-penetrant, irreversible myeloperoxidase (MPO) enzyme inhibitor. Verdiperstat was progressed through Phase 2 clinical trials by AstraZeneca. Seven clinical studies were completed by AstraZeneca, including four Phase 1 studies in healthy subjects, two Phase 2a studies in subjects with Parkinson’s Disease, and one Phase 2b study in subjects with MSA. These Phase 2 clinical studies provide evidence that verdiperstat achieves peripheral target engagement (i.e., reduces MPO specific activity in plasma) and central target engagement in the brain and offer proof of its mechanism of action (i.e., reduce microglial activation and neuroinflamation).

A Phase 3 clinical trial to evaluate the efficacy of verdiperstat in MSA is currently ongoing. A Phase 2/3 trial to evaluate the efficacy of verdiperstat in ALS is currently ongoing as part of the HEALEY ALS Platform Trial.

Verdiperstat has received Fast Track and Orphan Drug designations by the U.S. Food and Drug Administration (FDA) and the European Medicine Agency due to the unmet medical needs in MSA.

Verdiperstat Overview

DESCRIPTIONClick to expendFirst-in-class, brain-penetrant, irreversible inhibitor of MPO

CLINICAL STATUSClick to expendOver 250 healthy volunteers and patients have been treated with verdiperstat in Phase 1 and Phase 2 studies. A Phase 3 study in MSA is currently underway and a Phase 2/3 study in ALS is currently enrolling.
Verdiperstat (AZD3241) is a selective, irreversible and orally active myeloperoxidase (MPO) inhibitor, with an IC50 of 630 nM, and can be used in the research of neurodegenerative brain disorders.

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter a

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

PATENTWO 2006062465https://patents.google.com/patent/WO2006062465A1/enExample 9 l-(2-Isopropoxyethyl)-2-thioxo-l,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one (a) 3-[(2-Isopropoxyethyl)ωnino]-lH-pyrwle-2-carboxylic acid ethyl ester Trichlorocyanuric acid (1.84 g, 7.93 mmol) was added to a solution of 2- isopropoxyethanol (0.75 g, 7.21 mmol) in CH2Cl2 (3 mL). The reaction mixture was cooled to 0 °C and TEMPO (0.022 g, 0.14 mmol) was carefully added in small portions. The mixture was stirred at r.t. for 20 minutes then filtered through Celite and washed with CH2Cl2. The filtrate was kept cold, 0 °C, during filtration. The aldehyde solution was added to a stirred mixture of 3-amino-lH-pyrrole-2-carboxylic acid ester (0.83 g, 5.41 mmol) and HOAc (0.62 mL, 10.8 mmol) at 0 °C in methanol (5 mL). The mixture was stirred for 20 minutes, then NaCNBH3 (0.34 g, 5.41 mmol) was added. After stirring at r.t for 2 h, the solution was evaporated onto silica and purified by flash column chromatography (heptane/ethyl acetate gradient; 0 to 100% ethyl acetate) to yield the title compound (0.75 g, 58%) as an oil. 1H NMR (DMSO-d6) δ ppm 10.72 (IH, br s), 6.76-6.74 (IH, m), 5.66-5.65 (IH, m), 5.34(1H, br s), 4.17 (2H, q, J=7.0 Hz), 3.59-3.49 (3H, m), 3.15 (2H, q, J=5.6 Hz), 1.26 (3H, t, J=7.0 Hz), 1.10 (3H, s), 1.08 (3H, s); MS (ESI) m/z 241 (M +1).(b) l-(2-Isopropoxyethyl)-2-thioxo-l,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one The title compound (0.17 g, 23%) was prepared in accordance with the general method B using 3-[(2-isopropoxyethyl)amino]-lH-pyrrole-2-carboxylic acid ethyl ester (0.7 g, 2.91 mmol) and ethoxycarbonyl isothiocyanate (0.40 mL, 3.50 mmol).1H NMR (DMSO-d6) δ ppm 12.74 (2H, br s), 7.35 (IH, d, J=2.8 Hz), 6.29 (IH, d, J=3.0Hz), 4.49 (2H, t, J=6.3 Hz), 3.72 (2H, t, J=6.3 Hz), 3.60-3.58 (IH, m), 1.02 (3H, s), 1.01 (3H, s);MS (ESI) m/z 254 (M +1).

/////////verdiperstat, вердиперстат , فيرديبيرستات , 维地泊司他 , WHO 10251, AZD-3241BHV-3421UNII-TT3345YXVRTT3345YXVRBHV-3241, AZD 3241, BHV 3241, BHV 3421

CC(C)OCCN1C2=C(C(=O)NC1=S)NC=C2

wdt-17

NEW DRUG APPROVALS

ONE TIME TO MAINTAIN THIS BLOG

$10.00

Rilzabrutinib


Click here for structure editor
(R)-2-(3-(4-Amino-3-(2-fluoro-4-phenoxyphenyl)-1H-pyrazolo[3,4-d]-pyrimidin-1-yl)piperidine-1-carbonyl)-4-methyl-4-(4-(oxetan-3-yl)piperazin-1-yl)pent-2-enenitrile.png
20200818lnp2-rilza.jpg

PRN 1008, Rilzabrutinib

CAS 1575591-66-0

リルザブルチニブ;

C36H40FN9O3,

MW 665.7597

2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxyphenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile

Anti-inflammatory disease, Autoimmune disease treatment

  • OriginatorPrincipia Biopharma
  • Class2 ring heterocyclic compounds; Amines; Anti-inflammatories; Fluorobenzenes; Nitriles; Phenyl ethers; Piperazines; Piperidines; Pyrazoles; Pyrimidines; Skin disorder therapies; Small molecules
  • Mechanism of ActionAgammaglobulinaemia tyrosine kinase inhibitors
  • Orphan Drug StatusYes – Idiopathic thrombocytopenic purpura; Pemphigus vulgaris
  • Phase IIIIdiopathic thrombocytopenic purpura; Pemphigus vulgaris
  • Phase IIAutoimmune disorders
  • 02 Jun 2021Efficacy data from a phase IIa trial in Ankylosing spondylitis presented at the 22nd Annual Congress of the European League Against Rheumatism (EULAR-2021)
  • 07 Apr 2021Sanofi initiates enrollment in a phase I pharmacokinetics trial in healthy volunteers in Australia (PO, Tablet, Capsule) (NCT04748926)
  • 31 Mar 2021Sanofi announces intention to seek regulatory approval for Idiopathic thrombocytopenic purpura in 2023 (Sanofi pipeline, May 2021)

CLIP

https://cen.acs.org/pharmaceuticals/drug-development/Sanofi-acquire-BTK-inhibitor-firm/98/web/2020/08

Sanofi to acquire BTK inhibitor firm Principia for $3.7 billion

Principia is testing its small-molecule compounds in multiple sclerosis and immune system diseases

Sanofi will pay $3.7 billion to acquire Principia Biopharma, a San Francisco-based biotech firm developing small molecules that inhibit Bruton tyrosine kinase (BTK). The price represents about a 75% premium over Principia’s stock market value in early July, before reports surfaced that Sanofi was interested in buying the firm.

BTK is a protein important for both normal B cell development and the proliferation of lymphomas, which are B cell cancers. AbbVie, AstraZeneca, and BeiGene all market BTK inhibitors for treating specific kinds of lymphomas. Sales of AbbVie’s inhibitor, Imbruvica, approached $4.7 billion in 2019.

Other drug firms have been eager to get in on the action as well. In January, Merck & Co. spent $2.7 billion to acquire ArQule, whose experimental noncovalent BTK inhibitor is designed to overcome resistance that some cancers develop after treatment with current covalent BTK inhibitors. Eli Lilly and Company’s $8 billion acquisition of Loxo Oncology in 2019 also included a noncovalent BTK inhibitor.

BTK is also linked to inflammation, and Principia focuses on developing BTK inhibitors for immune system diseases and multiple sclerosis. Its compound rilzabrutinib is currently in clinical trials for pemphigus and immune thrombocytopenia. In 2017, Sanofi struck a deal to develop Principia’s brain-penetrant BTK inhibitor, SAR442168, for multiple sclerosis.

Sanofi announced in April of this year that the inhibitor reduced formation of new lesions—the scarred nervous tissue that gives multiple sclerosis its name—by 85% in a Phase II clinical trial. A Phase III trial of the compound began in June.

Upon announcing its deal to acquire Principia, Sanofi said that both rilzabrutinib and SAR442168 have the potential to become a “pipeline in a product,” indicating they can be used for many immune-related and neurological diseases, respectively.

The anti-inflammatory effects of BTK inhibitors have raised interest in the drugs as treatments for people hospitalized with COVID-19. Notably, the US National Cancer Institute conducted a small study suggesting acalabrutinib may help reduce the respiratory distress and inflammation in people with COVID-19. Based on that preliminary study, AstraZeneca—which markets acalabrutinib as Calquence—is conducting a 60-person randomized trial of the drug for COVID-19.

Sanofi has not indicated interest in investigating Principia’s BTK inhibitors as COVID-19 treatments.Chemical & Engineering NewsISSN 0009-2347 
PATENTWO 2021127231https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2021127231&tab=PCTDESCRIPTION&_cid=P20-KRA0I9-18818-1

SOLID FORMS OF 2-[3-[4-AMTNO-3-(2-FT,TTORO-4-PHENOXY- PHEN¥L)PYRAZOLO[3,4 D]PYRIMIDIN l~YL]PIPERIDINE~l~CARBON¥L] 4~

METHYL-4-[4-(OXETAN-3-YL)PIPERAZIN-l-YLjPENT-2-ENENITRILE

[11 This application claims the benefit of priority to U.S. Provisional Application

No 62/951,958, filed December 20, 2019, and U.S Provisional Application No. 63/122,309, filed December 7, 2020, the contents of each of which are incorporated by reference herein in their entirety.

[2] Disclosed herein are solid forms of 2-[3-[4~amino-3~(2~fluoro-4-phenoxy-plienyl)pyrazolo[3,4-d]pyrimidin-l-yl]piperidine-l Carbonyl]~4-nietliyl-4~[4-(oxetaii~3-yl)piperazin-!~yi]pent-2~enenitriie (Compound (I)), methods of using the same, and processes for making Compound (I), including its solid forms. The solid forms of Compound (I) may be inhibitors of Bruton’s tyrosine kinase (BTK) comprising low residual solvent content.

[3| The enzyme BTK is a member of the Tec family non-receptor tyrosine kinases.

BTK is expressed in most hematopoietic cells, including B cells, mast cells, and macrophages BTK plays a role in the development and activation of B cells. BTK activity has been implicated in the pathogenesis of several disorders and conditions, such as B cell-related hematological cancers (e.g., non-Hodgkin lymphoma and B cell chronic lymphocytic leukemia) and autoimmune diseases (e.g., rheumatoid arthritis, Sjogren’s syndrome, pemphigus, IBD, lupus, and asthma).

[4] Compound (I), pharmaceutically acceptable salts thereof, and solid forms of any of the foregoing may inhibit BTK and be useful in the treatment of disorders and conditions mediated by BTK activity. Compound (I) is disclosed in Example 31 of WO 2014/039899 and has the following structure:

where *C is a stereochemical center. An alternative procedure for producing Compound (!) is described in Example 1 of WO 2015/127310.

[5] Compound (I) obtained by the procedures described in WO 2014/039899 and WO 2015/127310 comprises residual solvent levels well above the limits described in the International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use (“ICH”) guidelines. In general, manufacturing processes producing residual solvent levels near or above the ICH limits are not desirable for preparing active pharmaceutical ingredients (APIs).

Example 1: Spray Drying Process A

[311] A solution of Compound (I) in dichloromethane (prepared according to Example 31 on pages 86-87 of WO 2014/039899) was washed with pH 3 phosphate buffer to remove basic impurities that are more soluble than Compound (I) in the aqueous layer. The dichloromethane solution was then washed with pH 7 buffer and solvent exchanged into isopropyl acetate. The isopropyl acetate solution was then washed with pH 3 phosphate buffer, bringing Compound (I) into the aqueous layer and removing non-basic impurities. The pH of the aqueous layer was adjusted to pH 9 with 10% sodium hydroxide, and the aqueous layer was extracted with isopropyl acetate. Upon concentration under vacuum, Compound (I) was precipitated from heptane at 0 °C, filtered and dried to give a white amorphous solid as a mixture of the (E) and (Z) isomers, as wet Compound (I). Wet Compound (I) was dissolved in methanol and spray dried at dryer inlet temperature of 125 °C to 155 °C and dryer outlet temperature of 48 to 58 °C to obtain the stable amorphous Compound (I) free base with levels of isopropyl acetate and heptane below 0.5% and 0.05%, respectively.

Example 2: Spray Drying Process B


intermediate A

Compound (!)

[241] A jacketed reactor with overhead stirrer, condenser, nitrogen line, temperature probe, and recirculating fluid chiller/heater was charged with Intermediate A (20.2 kg) and Intermediate B (13.6 kg, 1.5 equiv). DCM (361.3 kg, 14.5 vol) was charged to the reactor. The mixture was agitated, and the batch cooled to 0 °C to 5 °C. The reactor was charged with pyrrolidine (18.3 kg, 6 equiv) and then charged with TMSC1 (18.6 kg, 4 eq). Stirring was continued at 0 °C to 5 °C for 0.5 to 1 hour

[242] At 0 °C to 5 °C, acetic acid (2.0 equiv) was charged to the reactor followed by water (5 equiv). Stirring was continued at 0 °C to 5 °C for 1 to 1.5 hours. Water (10 equiv) was charged to the reactor, and the solution was adjusted to 20 °C to 25 °C. The internal temperature was adjusted to 20 °C to 25 °C and the biphasic mixture was stirred for 15 to 20 mins. Stirring was stopped and phases allowed to separate for at least 0.5 h. The lower aqueous layer was removed.

[243] Water (7 vol) was charged to the reactor. The pH was adjusted to 2.8-3.3 with a 10 wt. % solution of citric acid. Stirring was continued at 0 to 5 °C for 1 to 1.5 hours. Stirring was stopped and phases allowed to separate for at least 0.5 h. The lower aqueous layer was removed.

[244] A jacketed reactor with overhead stirrer, condenser, nitrogen line, temperature probe, and recirculating fluid chiller/heater was charged with an approximately 9% solution of NaHCCri (1 vol) and the organic layer. The internal temperature was adjusted to 20 °C to 25 °C, and the biphasic mixture was stirred for 15 to 20 mins. Stirring was stopped and phases allowed to separate for at least 0.5 h. The lower aqueous layer was removed. The aqueous layer was measured to have a pH greater than 7.

[245] A jacketed reactor with overhead stirrer, condenser, nitrogen line, temperature probe and recirculating fluid chiller/heater was charged with the organic layer. The organic phase ¾s distilled under vacuum at less than 25 °C to 4 total volumes. IP AC (15 vol) was charged to the reactor. The organic phase was distilled under vacuum at less than 25 °C to 10 total volumes. Water (15 vol) followed by pH 2.3 phosphate buffer were charged to the reactor at an internal temperature of 20 °C to 25 °C. The pH adjusted to 3 Stirring was stopped and phases allowed to separate for at least 0.5 h. The organic phase was removed.

[246] The following steps were repeated twice: IP AC (5 vol) was charged to the reactor containing the aqueous layer. Stirring was continued for 0.25 to 0.5 hours. Stirring was stopped and phases allowed to separate for at least 0.5 h. The organic phase was removed. [247] IP AC (15 vol) was charged to the reactor containing the aqueous layer. A pH 10 phosphate buffer was charged to the reactor and the pH adjusted to 10 with 14% NaOH solution. Stirring was continued for 1.5 to 2 hours. Stirring was stopped and phases allowed to separate for at. least 0.5 h. The aqueous layer was discarded. The organic layer was dried over brine.

[248] The organic solution was distilled under vacuum at less than 25 °C to 5 total volumes.

[249] A jacketed reactor with overhead stirrer, condenser, nitrogen line, temperature probe and recirculating fluid chiller/heater was charged with n-heptane (20 vol). The internal temperature was adjusted to 0 to 5 °C, and the IP AC solution was added.

[250] The suspension was filtered. The filter cake was washed with n-heptane and the tray was dried at 35 °C. Compound (I) (24.6 kg) was isolated in 86% yield.

[251] Compound (1) was dissolved in methanol (6 kg) and spray dried to remove residual IP AC and n-heptane.

Example 3: Precipitation Process A

[252] A solution of Compound (I) in dichloromethane (prepared according to Example 31 on pages 86-87 of WO 2014/039899) was quenched with acetic acid and water, followed by washing with pH 3 aqueous solution to remove basic impurities that are more soluble than Compound (1) in the aqueous layer. Washing was repeated as needed to reduce impurities. Methanesulfonic acid was added to the dichloromethane solution, and the dichloromethane solution was concentrated by distillation under reduced pressure, followed by addition of 1% NaCi aqueous solution and isopropyl acetate before adjustment of pH to approximately 3 with potassium hydroxide. The isopropyl acetate layer was removed and discarded. The aqueous layer containing Compound (I) was washed with isopropyl acetate to remove hydrophobic impurities. Washing was repeated as needed to reduce related substance impurities. Residual isopropyl acetate was removed by distillation under reduced pressure. The aqueous solution containing Compound (I) was cooled to 0 to 5°C before adjusting the pH to approximately 9 with potassium hydroxide. The free base of Compound (I) was allowed to precipitate and maturate at 20 °C for 20 hours. The mixture temperature was then adjusted to 20 °C to 25 °C, and the hydrate impurity was verified to be less than 0.3% (< 0.3%). The cake of the free base of Compound (I) was filtered and washed as needed to reduce conductivity. The cake was then allowed to dry on the filter under vacuum and nitrogen swept to reduce water content by Karl-Fischer (KF < 50%) before transferring to the oven for drying. The wet cake of the free base of Compound (1) was dried under vacuum at 25 °C until water content by Karl -Fischer was less than 1.5% (KF < 1.5%), and then dehmiped by milling to yield a uniform white amorphous solid as a mixture of the (E) and (Z) isomers, with no detectible levels of isopropyl acetate or heptane.

Example 4: Precipitation Process 3B

[253] A solution of Compound (I) in dichloromethane (prepared according to Example 31 on pages 86-87 of WO 2014/039899) was quenched with acetic acid and water, followed by washing with pH 3 aqueous solution to remove basic impurities that are more soluble than Compound (I) in the aqueous layer. The washing was repeated as needed to reduce residual solvents and impurities. The dichloromethane solution was then washed with saturated sodium bicarbonate (pH > 7). Dichloromethane was removed by distillation under reduced pressure, followed by addition of water and isopropyl acetate. The pH of the aqueous layer was adjusted to pH to 2.8 – 3.3 with 2 M aqueous sulfuric acid (H2SQ4) at 0 – 5 °C, and the mixture rvas stirred and settled. After phase separation removal of the organic layer, the aqueous layer was washed with isopropyl acetate three times and the residual isopropyl acetate in aqueous layer was distilled out under vacuum at a temperature below 25 °C and the solution was basitied with 5% aqueous KOFI to pH 9 – 10 to a slurry . The resulting suspension was stirred and warmed up to 20 °C to 25 °C and aged for 20 h. The product was filtered and washed with water and dried to give white solid in 86% yield.

Example 5: Precipitation Process C

[254] A solution of Compound (I) in dichloromethane (prepared according to Example 31 on pages 86-87 of WO 2014/039899) was quenched with acetic acid and water, followed by washing to remove basic impurities that are more soluble than Compound (I) in the aqueous layer. Washing was repeated as needed to reduce impurities. Methanesulfonic acid was added to the d chloromethane solution, and the dichloromethane solution was concentrated under reduced pressure to obtain a thin oil. The concentrated oil was cooled to approximately 5°C before washing with an aqueous solution of sodium chloride. The organic phase was discarded. Washing of the aqueous layer was repeated as needed with dichloromethane to remove low level impurities. The pH of the aqueous solution was adjusted to approximately 3 with an aqueous solution of potassium hydroxide. Residual dichloromethane was removed

under reduced pressure. The level of residual acetic acid was determined by, for example, titration. The aqueous solution containing Compound (I) was cooled to a temperature between 0°C and 5°C. Acetic acid was present at 0 wt % to 8 wt. %. Acetic acid level was 0 wt % if the aqueous acid solution was washed with aqueous sodium bicarbonate or another aqueous inorganic base. Optionally, additional acetic acid was added to achieve a 0 wt.% to 8 wt. % acetic acid level. An aqueous solution of potassium hydroxide was constantly charged to the aqueous solution to obtain a pH to approximately 9.5. The free base of Compound (I) was allowed to precipitate and maturate at approximately 20 °C for least 3 hours. The cake (wet solid) of the free base of Compound (I) was filtered and washed with water. The wet cake was then dried under reduced vacuum with slight heat. Alternatively, instead of washing the wet cake with water, the wet cake was reslurried with water at approximately 15 °C for at least 1 hour before filtering. The free base of Compound (I) in the fomi of a wet cake was dried under vacuum with slight heat at 25°C.

[255] FIGs. 12-15 are example SEM images showing the variable morphologies of particles of Compound (I) during the filtration step to isolate Compound (I) based on the amount acetic acid added during the initial step in the precipitation of Compound (Ϊ) (FIG. 12: at 0 wt. % acetic acid; FIG 13: at 3 wt. % acetic acid; FIG. 14: at 5 wt. % acetic acid; FIG 15: at 8 wt. % acetic acid). Filtration speed depended on the morphology and was the fastest for 0 wt. % acetic acid. At 1 wt. % acetic acid, the filtration speed diminished considerably, improving at 2 wt. % to 3 wt. % acetic acid. Morphologies with more open holes (such as, e.g., more porous particles) resulted in improved filtration speeds, whereas more compact particles resulted in decreased filtration speed.

Example 6: Conversion of a Crystalline Form of Compound (Ϊ) to an Amorphous Form

[256] 9.8 grams of a crystalline form of Compound (I) were dissolved in approximately 20 mL of dichloromethane and approximately 120 ml. of brine solution. Then, approximately 1 equivalent of methanesulfonic acid was added. The pH w¾s approximately 2. The layers were separated. The aqueous layer was concentrated at a temperature between 0°C and 5°C to remove residual dichloromethane before slowly adding aqueous KOI I solution (approximately 5%) to adjust the pH to a value between 9 and 10. During aqueous KOH addition, an amorphous form of Compound (I) precipitated out. The slurry was slowly warmed to room temperature and then was stirred for approximately 24 hours before filtering and rinsing the wet cake with water. The wet cake was dried under vacuum with slight heat at approximately 30°C to provide 7 grams of a white to an off-white solid (87% yield and 98 4% purity). XRPD showed that the product was an amorphous solid form of Compound (I).

Example 7: Micronization of Compound (I) Particles Obtained by Precipitation Processes

[257] A fluid jet mill equipment was used during lab scale jet milling trials. The fluid jet mill equipment includes a flat cylindrical chamber with 1.5” diameter, fitted with four symmetric jet nozzles winch are tangentially positioned in the inner wall. Prior to feeding material to the fluid jet mill in each trial, the material was sieved in a 355 iim screen to remove any agglomerates and avoid blocking of the nozzles during the feed of material to the micronization chamber. The material to be processed was drawn into the grinding chamber through a vacuum created by the venturi (P vent ~ 0 5 – 1 0 bar above P grind). The feed flow rate of solids (F_feed) was controlled by a manual valve and an infinite screw volumetric feeder. Compressed nitrogen was used to inject the feed material; compressed nitrogen was also used for the jet nozzles in the walls of the milling chamber. Compressed fluid issuing from the nozzles expands from P grind and imparts very’ high rotational speeds in the chamber. Accordingly, material is accelerated by rotating and expanding gases and subjected to centrifugal forces. Particles move outward and are impacted by high velocity jets, directing the particles radially inward at very high speeds. Rapidly moving particles impact the slower moving path of particles circulating near the periphery of the chamber. Attrition takes place due to the violent impacts of particles against each other. Particles with reduced size resulting from this sequence of impacts are entrained in the circulating stream of gas and swept against the action of centrifugal force toward the outlet at the center. Larger particles in the gas stream are subjected to a centrifugal force and returned to the grinding zone. Fine particles are carried by the exhaust gas to the outlet and pass from the grinding chamber into a collector.

[258] The feeder has continuous feed rate control; however, to more precisely control the feed rate, the full scale of feed rates was arbitrary divided in 10 positions. To calibrate F feed, the feeder was disconnected from milling chamber and 10 g of Compound (I) powder was fed through the feeder operating at various feed rate positions. The mass of powder flowing through the feeder over 6 minutes was marked. The resulting feed rate was directly proportional to feeder position. After processing each of the four trials, the jet mill was stopped, micronized product removed from the container, and the milling chamber checked for any powder accumulation.

Variables/Parameters

F_feed Feed flow rate of solids [kg/h]

P grind Grinding pressure inside the

drying chamber [bar]

P vent Feed pressure in the venturi [bar]

Example 8: Residual Solvent Levels

[251] Retention of process solvents (/.<?., res dual solvents) depends on van der Waal s’ forces that are unique to and an inherent property of each molecule. Additionally, solvent retention depends how the API solid is formed, isolated, washed, and dried (i.e., during the manufacturing process). Because residual solvents may pose safety risks, pharmaceutical processes should be designed to minimize residual solvent levels (e.g , to result in residual solvent levels below the limits established in the ICH guidelines).

[252] Residual solvent analysis was performed using gas chromatography-mass spectrometry. The residual solvent levels in solid forms of Compound (I) prepared by spray drying processes described herein and precipitation processes described herein are provided in Table 2. The residual solvent levels in crude Compound (I) listed in Table 2 are comparable to the residual solvent levels in crude Compound (I) prepared according to the procedures detailed in Example 31 of WO 2014/039899 and Example 1 of WO 2015/127310.

Table 2: Residual solvent levels in solid forms of Compound (I)

PATENTWO 2015127310https://patents.google.com/patent/WO2015127310A1/enExample 1Synthesis of 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-l- yl]-piperidine-l-carbonyl]-4-m iperazin-l-yl]pent-2-enenitrile

Figure imgf000045_0001

Step 1To a solution of 3-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4- d]pyrimidin-l -yl]-l-piperidyl]-3-oxo-propanenitrile (15 g, 3.12mmol), 2-methyl-2-[4- (oxetan-3-yl)piperazin-l-yl]propanal (794.25mg, 3.74mmol) in DCM (40mL), pyrrolidine (1.54mL,18.71mmol) at 0-5 °C was added, which is followed by TMS-Cl (1.58mL,12.47mmol). The reaction mixture was stirred at 0-5 °C for 3 h and was quenched with 1 M potassium phosphate buffer (pH 3). Layers were separated and the organic layer was washed once more with 1 M potassium phosphate buffer (pH 3). The organic layer was extracted withl M potassium Phosphate buffer at pH 1.5. Layers were separated. The aqueous phase contained the desired product while the impurities stayed in the organic phase. The aqueous phase was neutralized with 1 M potassium phosphate (pH 7) and was extracted with isopropylacetate (10 volumes). Upon concentration 2-[(3R)-3-[4-amino-3-(2-fluoro-4- phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-l-yl]piperidine-l-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-l-yl]pent-2-enenitrile was obtained as a foam having >99% HPLC purity. MS (pos. ion) m/z: 666 (M+l ).The foam containing high levels of residual solvent was dissolved in 2 M HC1 and the resulting solution was placed under vacuum to remove residual organic solvents. pH of the solution was then adjusted to ~ 7 and the resulting paste was filtered and dried in vacuum without heat. This resulted in isolation of 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy- phenyl)pyrazolo[3,4-d]pyrimidin-l-yl]piperidine-l-carbonyl]-4-methyl-4-[4-(oxetan-3- yl)piperazin- l-yl]pent-2-enenitrile containing residual water up to 10%. Drying under vacuum without heat reduces the water level but lead to generation of impurities.Step 1AAlternatively, the isopropylacetate solution of 2-[(3R)-3-[4-amino-3-(2-fluoro-4- phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin- 1 -yl]piperidine- 1 -carbonyl]-4-methyl-4-[4- (oxetan-3-yl)piperazin-l -yl]pent-2-enenitrile can be concentrated to 4 vol and added to heptane (20 volume) at 0 °C. The resulting suspension was stirred at 0 °C overnight and the product was filtered, washed twice with heptane and dried at 45 °C for 2 days under vacuum to give 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-l – yl]piperidine-l-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-l-yl]pent-2-enenitrile in 85 – 90 % yield as a free flowing solid. However, the solids obtained by this method contained high residual solvents (3.9 wt% isopropylacetate and 1.7 wt% heptane). In addition, the free base form was not very stable as degradation products were observed during the drying process at less than 45 °C.Salt formationExample 2Preparation of 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)-pyrazolo[3,4-d]pyrimidin- l-yl]-piperidine-l-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)-piperazin-l-yl]pent-2-enenitrile hemisulfate and sulfate saltHemisulfate: To the solution of 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)-pyrazolo[3,4- d]pyrimidin-l-yl]-piperidine -carbonyl]-4-methyl-4-[4-(oxetan-3-yl)-piperazin-l-yl]pent-2- enenitrile (4.2 g) in EtOAc (60 mL, 15 vol) was added sulfuric acid (0.31 g, 0.17 mL, 0.5 eq) in EtOAc (20 mL, 5 vol) at ambient temperature. The suspension was stirred at ambient temperature for ~ 2 hr and then 40 °C for 4 hr and then at ambient temperature for at least 1 hr. After filtration and drying at ambient temperature under vacuum, 1.5 g of white powder was obtained. Solubility of the hemi-sulfate at ambient temperature was > 100 mg/mL in water.Sulfate saltTo the solution of 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)-pyrazolo[3,4- d]pyrimidin-l-yl]-piperidine-l-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)-piperazin-l-yl]pent-2- enenitrile (810 mg) in EtOAc (8 mL, 10 vol) was added sulfuric acid (0.06 mL, 1.0 equiv.) in EtOAc (2.5 mL, 5 vol) at ambient temperature. The resulting suspension was stirred at 40 °C for 2 hr and then cooled to ambient temperature for at least 1 hr. After filtration, solids were dried by suction under Argon for 1 h to give a white powder (0.68 g) in 69% yield.

Figure imgf000047_0001

Example 3Preparation of 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)-pyrazolo[3,4- d]pyrimidin- 1 -yl]-piperidine- 1 -carbonyl] -4-methyl-4-[4-(oxetan-3-yl)-piperazin- 1 -yl]pent-2- enenitrile hydrochlorideTo a solution of 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4- d]pyrimidin- 1 -yl]piperidine- 1 -carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin- 1 -yl]pent-2- enenitrile (100 mg, 0.15 mmol) in CH2CI2 (1ml) at ambient temperature was added 2 equivalent of HC1 (0.3 mmol, 0.15 ml of 2M HC1 in 1 : 1 dioaxane:CH2Cl2). The resulting homogeneous solution was stirred at ambient temperature for 1 h and was added dropwise to 15 volumes of ethylacetate (as compared to CH2C12) resulting in formation of a white solid. The mixtures was aged at ambient temperature for lh and placed at 2-8 C for 19 h. Upon filtration and washing of the filter cake with ethylacetate and drying a white solid was obtained. Analysis by XRPD indicated formation of an amorphous solid. Both Ή-NMR and IC analysis indicated formation of the salt. IC indicated formation mono-HCl salt.

Figure imgf000048_0001

Example 4General procedure for preparation of 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy- phenyl)pyrazolo[3,4-d]pyrimidin-l-yl]-piperidine-l-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)- piperazin-l-yl]pent-2-enenitrile mono- and di-mesylate saltsTo a solution of 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4- d]pyrimidin-l-yl]piperidine-l-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-l-yl]pent-2- enenitrile (100 mg, 0.15 mmol) in CH2C12 (1 ml) at ambient temperature was added either 1 equivalent of methanesulfonic acid (0.15 mmol, 0.2 ml of 74 mg/ml solution in CH2C12) or 2 equivalent of methanesulfonic acid (0.3 mmol, 0.4 ml of 74 mg/ml solution in CH2C12). The resulting homogeneous solution was stirred at ambient temperature for 1 h and was added dropwise to 10 volumes of antisolvents (ethylacetate, methyl tert-butylether (MTBE), or cyclohexane) (10 ml as compared to CH2C12) resulting in formation of a white solid. The mixture was aged at ambient temperature for lh and placed at 2-8 °C for 19 h. Upon filtration and washing of the filter cake with the antisolvent and drying, a white solid was obtained. Analysis by XRPD indicated formation of an amorphous solid. Both Ή-NMR and IC analysis indicated formation of the salt as well as counterion ratio.Alternatively 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]- pyrimidin- 1 -yl]piperidine- 1 -carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin- 1 -yl]pent-2- enenitrile can be dissolved in 4 volumes of isopropylacetate and added to 2 equivalent of methanesulfonic acid in 6 volumes of isopropylacetate at 0 °C to generate the dimesylate salt.

Figure imgf000049_0001

1. Theoretical mesylate content, monomesylate=12.6% and dimesylate=22.4%, NO- not determinedExample 5 General procedure for the preparation of carboxylate salt Approximately 20 mg of the compound (I) was dissolved in minimum amount of the allocated solvent system. These were then mixed with the appropriate number of equivalents of counterion dissolved or slurried in the allocated solvent.If compound (I) was insoluble in the selected solvent, slurry of the sample was used after adding 300 μί.If the acid was insoluble in the selected solvent, slurry of the acid was used after adding 300 xL.If the acid was a liquid, the acid was added to the dissolved/slurried compound (I) from a stock solution in the allocated solvent.The suspensions/ precipitates resulting from the mixtures of compound (I) were temperature cycled between ambient (ca. 22°C) and 40°C in 4 hour cycles for ca. 48 hrs (the cooling/heating rate after each 4 hour period was ca. 1 °C/min). The mixtures were visually checked and any solids present were isolated and allowed to dry at ambient conditions prior to analysis. Where no solid was present, samples were allowed to evaporate at ambient. Samples which produced amorphous material, after the treatment outlined above, were re- dissolved and precipitated using anti-solvent (ter/-butylmethylether) addition methods at ambient conditions (ca. 22°C). i.e. the selected anti-solvent was added to each solution, until no further precipitation could be observed visually or until no more anti-solvent could be added. The solvents used in this preparation were acetonitrile, acetone, isopropyl acetate, THF and MTBE. The acid used were oxalic acid, L-aspartic acid, maleic acid, malonic acid, L-tartaric acid, and fumaric acid.Example 6General procedure for preparation of 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy- phenyl)pyrazolo[3,4-d]pyrimidin-l-yl]-piperidine-l-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)- piperazin-l-yl]pent-2-enenitrile hemicitrate saltTo a solution 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]- pyrimidin- 1 -yl]piperidine- 1 -carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin- 1 -yl]pent-2- enenitrile (5 g, 7.5 mmol) in ethanol (50 ml) was added citric acid (720.5 mg, 3.76 mmol) dissolved in 2 ml of water. Mixture was stirred at ambient temperature for 15 min, additional 0.5 ml of water was added and the mixture was stirred for 1 h, concentrated in vacuo to a gum. Ethanol was added and the mixture was concentrated. This process was repeated twice more and then CH2CI2 was added to the mixture. Upon concentration a white solid was obtained which was tumble dried under reduced pressure at 40 C for 4 h, then in a vacuum oven for 19h to give 5.4 g of a solid. Analysis by XRD indicated formation of an amorphous solid 

PATENT

WO2014039899, Example 31

Rilzabrutinib (PRN1008) is an oral, reversible covalent inhibitor of Bruton’s tyrosine kinase (BTK) [1].

https://patents.google.com/patent/WO2014039899A1/enExample 31Synthesis of (R)-2-(3-(4-amino-3-(2-fluoro-4-phenoxyphenyl)- 1 H-pyrazolo[3,4-d]pyrimidin- 1 -yl)piperidine- 1 -carbonyl)-4-methyl-4-(4-(oxetan-3-yl)piperazin- 1 -yl)pent-2-enenitrile

Figure imgf000087_0002

Step 1A solution of 2-bromo-2-methyl-propanal (696.6 mg, 4.61 mmol) in DCM (10 mL) was cooled with an ice bath and l -(oxetan-3-yl)piperazine (328 mg, 2.31 mmol), diluted with 5-10 mL of DCM, was slowly added via addition funnel over a 15 min period. Next, Hunig’s base (0.4 mL, 2.31 mmol) was added and then the cooling bath was removed. The reaction mixture was stirred at room temperature overnight and the DCM layer was washed three times with 0.5N HC1. The combined aqueous layer was neutralized with NaOH to pH 10-11 and extracted with DCM. The combined organic layer was washed with brine and dried over Na?S04. Filtration and removal of solvent afforded 2-methyl-2-[4-(oxetan-3-yl)piperazin-l- yl]propanal as a light yellow liquid, which was used directly in the next step without further purification.Step 2To a cooled (0 °C) solution of 3-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)- pyrazolo[3,4-d]pyrimidin-l-yl]-l-piperidyl]-3-oxo-propanenitrile (80 mg, 0.17 mmol), was added 2-methyl-2-[4-(oxetan-3-yl)piperazin-l-yl]propanal (-108 mg, 0.51 mmol) in DCM (10 mL) followed by pyrrolidine (0.08 mL, 1.02 mmol) and TMS-C1 (0.09 raL, 0.68 mmol.) The ice bath was removed, and the reaction stirred 1 hour. Most of the solvent was removed and the residues were purified by chromatography, using 95:5 CH2Cl2:MeOH to obtain 79 mg of (R)-2-(3-(4-amino-3-(2-fluoro-4-phenoxyphenyl)-lH-pyrazolo[3,4-d]-pyrimidin-l- yl)piperidine- 1 -carbonyl)-4-methyl-4-(4-(oxetan-3-yl)piperazin- 1 -yl)pent-2-enenitrile as a white solid. MS (pos. ion) m/z: 666 (M+l).

PAPER

https://www.sciencedirect.com/science/article/abs/pii/S0223523421001781?dgcid=rss_sd_all

Therapy based on Bruton’s tyrosine kinase (BTK) inhibitors one of the major treatment options currently recommended for lymphoma patients. The first generation of BTK inhibitor, Ibrutinib, achieved remarkable progress in the treatment of B-cell malignancies, but still has problems with drug-resistance or off-target induced serious side effects. Therefore, numerous new BTK inhibitors were developed to address this unmet medical need. In parallel, the effect of BTK inhibitors against immune-related diseases has been evaluated in clinical trials. This review summarizes recent progress in the research and development of BTK inhibitors, with a focus on structural characteristics and structure-activity relationships. The structure-refinement process of representative pharmacophores as well as their effects on binding affinity, biological activity and pharmacokinetics profiles were analyzed. The advantages and disadvantages of reversible/irreversible BTK inhibitors and their potential implications were discussed to provide a reference for the rational design and development of novel potent BTK inhibitors.

Image 17

///////////////PRN-1008,  PRN 1008, Rilzabrutinib, リルザブルチニブ,
N#CC(=CC(N(C1COC1)C)(C)C)C(=O)N1CCCC1Cn1nc(c2c1ncnc2N)c1ccc(cc1F)Oc1ccccc1

Uprifosbuvir


Uprifosbuvir structure.svg
Uprifosbuvir.png
ChemSpider 2D Image | Uprifosbuvir | C22H29ClN3O9P

Uprifosbuvir

MK 3682, IDX 21437

ウプリホスブビル;

Formula C22H29ClN3O9P
CAS 1496551-77-9
Mol weight 545.9071

уприфосбувир [Russian] [INN]أوبريفوسبوفير [Arabic] [INN]乌磷布韦 [Chinese] [INN]

propan-2-yl (2R)-2-[[[(2R,3R,4R,5R)-4-chloro-5-(2,4-dioxopyrimidin-1-yl)-3-hydroxy-4-methyloxolan-2-yl]methoxy-phenoxyphosphoryl]amino]propanoate

Isopropyl (2R)-2-{[(R)-{[(2R,3R,4R,5R)-4-chloro-5-(2,4-dioxo-3,4-dihydro-1(2H)-pyrimidinyl)-3-hydroxy-4-methyltetrahydro-2-furanyl]methoxy}(phenoxy)phosphoryl]amino}propanoate

IDX-21437DB15206SB18784D10996Q27281714

Uprifosbuvir (MK-3682) is an antiviral drug developed for the treatment of Hepatitis C. It is a nucleotide analogue which acts as an NS5B RNA polymerase inhibitor. It is currently in Phase III human clinical trials.[1][2][3]

Uprifosbuvir is under investigation in clinical trial NCT02332707 (Efficacy and Safety of Grazoprevir (MK-5172) and Uprifosbuvir (MK-3682) With Elbasvir (MK-8742) or Ruzasvir (MK-8408) for Chronic Hepatitis C Genotype (GT)1 and GT2 Infection (MK-3682-011)).Hepatitis C viruss (HCV) have the newly-increased patients of 3-4 million every year, and World Health Organization (WHO) is estimated in global sense More than 200,000,000, in China more than 10,000,000 patients, HCV belongs to flaviviridae hepatovirus virus to dye person.Long-term hepatitis C virus Gently to inflammation, weight is to liver cirrhosis, hepatocarcinoma for poison infection.And during hepatitis C cirrhosis patients in decompensation, can there are various complication, such as abdomen Water abdominal cavity infection, upper gastrointestinal hemorrhage, hepatic encephalopathy, hepatorenal syndrome, liver failure etc. are showed.The side of HCV infection is treated initially Method is interferon and interferon and ribavirin combination therapy, and only 50% therapist has reaction, and interferon to the method With obvious side effect, such as flu-like symptoms, body weight lower and fatigue and weak, and interferon and ribavirin Conjoint therapy then produces sizable side effect, including haemolysis, anemia and tired etc..U.S. FDA have approved multiple HCV medicines, including the polymerization of protease inhibitor, ucleosides and non-nucleoside in recent years Enzyme inhibitor and NS5A inhibitor etc..The protease inhibitor class medicine of FDA approvals has three:VX‐950 (Telaprevir), SCH-503034 (Boceprevir) and TMC435 (Simeprevir), the shortcoming of protease inhibitor is It is also easy to produce that mutation, toxicity is big, poor bioavailability, it is effective to individual other gene type.Eggs of the Telaprevir as the first generation White enzyme inhibitor has logged out market.The second filial generation and third generation protease inhibitor of high activity and wide spectrum is mainly used as and other One of component of drug combination of hepatitis C medicine.NS5A inhibitor is the highly active anti-HCV medicament of a class.The most representative Daclatasive for having BMS, The Ombitasvir of the Ledipasvir and AbbVie of Gilead, as this kind of medicine independent medication is easy to produce drug resistance, They treat one of drug component of HCV primarily as drug combination.The AG14361 of hepatitis C is generally divided into two kinds of ucleosides and non-nucleoside.At present, clinically only Suo Feibu One ucleosides hepatitis C medicine of Wei is listed by FDA approvals, and other are still in the anti-hepatitis C virus medicine of ucleosides of clinical experimental stage Thing also has the MK-3682 (IDX21437) of Mo Shadong, the AL-335 of the ACH-3422 and Alios of Achillion drugmakers.Third Hepatitis virus have the features such as Multi-genotype and fast variation, and single medicine treatment hepatitis C has generation drug resistance fast, to part Genotype cure rate is low and the various defects such as course for the treatment of length.In order to overcome these defects, the treatment of drug combination is primarily now taken Scheme, in order to overcome these defects, primarily now takes the therapeutic scheme of drug combination, the Sovaldi conducts of FDA approval listings The key component of drug combination, for the patient of 4 type of 1 type of gene and gene be Suo Feibuwei, profit Ba Wei woodss and Polyethylene Glycol-α- The drug combination of interferon three, the course for the treatment of are 12 weeks;For 1 type of gene and the patient of 3 types, the big woods joints of Suo Feibuwei and Li Ba Medication, the course for the treatment of are respectively 12 weeks and 24 weeks.- 2016 years 2013, FDA ratified Suo Feibuwei and NS3 protein inhibitors again in succession Simeprevir shares the patient of 1 type of therapeutic gene;The NS5A inhibitor Daclatavir therapeutic genes 1 of Suo Feibuwei and BMS With the patient of 3 types.Harvoni is the patient that Suo Feibuweijia NS5A inhibitor Ledipasvir is used for 1 type of gene.Even if using Same nucleoside, the NS5A inhibitor and/or NS3 protease inhibitor for sharing varying strength can effectively extend composition of medicine Clinical application range and Shorten the Treatment Process.In June, 2016, FDA have approved Suo Feibuwei and more potent secondary NS5A inhibitor Velpatasvir shares the hepatitis C patient suitable for all gene types, it is not necessary to carry out genetic test.Just in three phases clinic Suo Feibuwei, NS5A inhibitor Velpatasvir and NS3 protease inhibitor Voxilaprevir goes for all of disease People, is try to the course for the treatment of and shortened to 8 weeks from 12 weeks.Suo Feibuwei just in clinical trial target spots different with hepatitis C virus are directed to Drug regimen (such as Suo Feibuweijia new type NS 5A inhibitor Velpatasvir and/or protease inhibitor GS5816), its knot Fruit show than single drug more wide spectrum, effectively, and can be with Shorten the Treatment Process.MSD Corp. is by MK-3682 and NS5A inhibitor Grazoprevir and/or protease inhibitor Elbasvir is used as new drug regimen, effective for all genotype of HCV, And further shorten to the course for the treatment of of 8 weeks.New deuterated nucleoside phosphoric acid ester compound disclosed in patent of the present invention, especially The double deuterated compound such as VI-1b2 in 5 ‘-position, shows than the more preferable bioavailability of former compound MK-3682 and longer partly declines Phase.In addition, this kind of novel nucleoside phosphoramidate is significantly superior to the Suo Feibuwei of clinical practice in terms of anti-hepatitis C activity, On sugared ring, chlorine atom replaces fluorine atom, and cytotoxicity is significantly reduced in surveyed cell line.By to base, sugared ring With the transformation and optimization of prodrug moiety system, the anti-hepatitis C activity of partial synthesis compound is higher than Suo Feibuwei 2-10 times, meanwhile, In the optimization of metabolism key position, synthesis compound shows that in blood plasma the higher metabolic stabilities of peso Fei Buwei and chemistry are steady It is qualitative.Therefore this kind of new deuterated nucleotide phosphate and NS5A inhibitor and/or egg as shown in formula a, a1, a2, b, b1, b2 The newtype drug combination constituted by white enzyme inhibitor is with extremely wide application prospect.Deuterium is the naturally occurring hydrogen isotope of nature, the deuterated isotopic body in common drug all containing trace.Deuterium without It is malicious, “dead”, it is safe to human body, C-D keys are more stable (6-9 times) than c h bond, hydrogen is replaced with after deuterium, can extend medicine Half-life, while pharmacologically active (shape difference of H and D is little, J Med Chem.2011,54,2529-2591) is not affected, in addition Deuterated medicine usually shows more preferable bioavailability and less toxicity, and the active ribonucleoside triphosphote of its metabolism is more stable, So deuterated nucleoside phosphoramidate will be better than corresponding nucleoside medicine in the curative effect of clinical practice.For example, 2013 It is exactly a deuterated compound that the nucleoside anti hepatitis C virus drug ACH-3422 of clinical trial is in the approval of year FDA, with non-deuterium (WO2014169278, WO are 2014169280) than having higher bioavailability and longer half-life for the former compound phase in generation. 
Based on above-mentioned present Research, we design and are prepared for the new deuterated nucleoside that compound VI-1b2 is representative Phosphoramidate.Below we will be described in the architectural feature of deuterated nucleoside phosphoramidate of our inventions, preparation method, Antiviral activity experimental result and it as anti-hepatitis c virus drug combination key component and NS5A inhibitor and/ Or the drug regimen of protease inhibitor is in the application of anti-virus aspect.

The EPA awarded the greener reaction conditions to the pharmaceutical company Merck & Co. for building a prodrug synthesis that eliminated the use of toxic reagents. Prodrugs are molecules that get metabolized by our bodies into an active pharmaceutical. Some hepatitis C and HIV medications are prodrugs and get synthesized through a method call pronucleotide (ProTide) synthesis. The method uses toxic and corrosive thionyl chloride, plus an excess of expensive pentafluorophenol that generates a lot of waste. Merck’s new method creates their target compounds in 90 to 92% yields without these reagents and eliminates the need for halogenated solvents entirely through strategic catalyst loading and the use of different starting materials from the traditional route.

20200616lnp3-structure.jpg

The design of greener chemicals award went to the development of more environmentally friendly versions of chemicals called thermoset binders, which can serve as carpet adhesives and are involved in the manufacture of mineral and fiberglass products. Generally, these chemicals are based on formaldehyde or polycarboxylic acids, and they can give off toxic formaldehyde and often use small amounts of sulfuric and hypophosphorous acid as catalysts to activate them. The insulation and commercial roofing company Johns Manville created a new binder based on the reaction between renewable dextrose, fructose, and other simple sugars, bound together by the α-carbon-containing cross-linking agent glyoxal. The reaction also uses a biodegradable acid in water as a catalyst. The binder can be made in just one step instead of the traditional multistep synthesis. Also, the synthesis can be done directly at the manufacturing site, instead of beforehand like with the traditional approach, meaning this new binder creates fewer of the health and environmental hazards that come from storage and transportation.

wdt-25

NEW DRUG APPROVALS

ONE TIME

$10.00

SYN

US 20170226146,

Paper

Organic Process Research & Development (2021), 25(3), 661-667.

https://pubs.acs.org/doi/10.1021/acs.oprd.0c00487

Abstract Image

A novel application of the synthesis of pronucleotide (ProTide) 5′-phosphoramidate monoesters promoted by aluminum-based Lewis acids is described. In the multikilogram synthesis of uprifosbuvir (MK-3682, 1), a clinical candidate for the treatment of hepatitis C, this methodology provided >100:1 diastereoselectivity at the phosphorus stereocenter and >100:1 selectivity for the 5′-mono phosphorylation over undesired bisphosphorylation side products. The high diastereoselectivity and mono/bis ratio achieved enabled elimination of the tedious workup associated with the tert-butyl magnesium chloride protocol commonly used to install this functionality in similar nucleotide prodrugs, achieving a near doubling of the isolated yield from 45% to 81%. The process development and purity control strategy of MK-3682, as well as handling of the pyrophoric reagent on scale, will also be discussed.

PAPER

Science (Washington, DC, United States) (2020), 369(6504), 725-730.

Science (Washington, DC, United States) (2017), 356(6336), 426-430.

Chemical Science (2017), 8(4), 2804-2810.

PATENT

CN 106543253

https://patents.google.com/patent/CN106543253A/zh

PATENT

WO 2014058801

https://patents.google.com/patent/WO2014058801A1/enExample 1Preparation of 2′-Chloro Nucleoside Analogs

Scheme 1

Figure imgf000136_0001
Figure imgf000136_0002

Ethyl (3R)-2-chloro-3-[(4R)-2,2-dimethyl-l,3-dioxolan-4-yl]-3-hydroxy-2- methylpropanoate (A2):

Figure imgf000137_0001

[00273] A 5 L flange flask was fitted with a thermometer, nitrogen inlet, pressure equalizing dropping funnel, bubbler, and a suba»seal. Methyl lithium solution (1.06 L, 1.6 M in diethylether, 1.7 equiv.) was added, and the solution was cooled to about -25 °C.Diisopropyl amine (238 ml, 1.7 equiv.) was added using the dropping funnel over about 40 minutes. The reaction was left stirring, allowing to warm to ambient temperature overnight. C02(s)/acetone cooling was applied to the LDA solution, cooling to about -70 °C.[00274] i?-Glyceraldehyde dimethylacetal solution (50% in DCM) was evaporated down to -100 mbar at a bath temp of 35 °C, to remove the DCM, then azeotroped with anhydrous hexane (200 ml), under the same Buchi conditions. 1H NMR was used to confirm that all but a trace of DCM remained.[00275] The fresh aldehyde (130 g, 1 mol) and ethyl 2-chloropropionionate (191 ml, 1.5 equiv.) were placed in a 1 L round bottom flask, which was filled with toluene (800 ml). This solution was cooled in a C02(s)/acetone bath, and added via cannula to the LDA solution over about 50 minutes, keeping the internal temperature of the reaction mixture cooler than -60 °C. The mixture was stirred with cooling (internal temp, slowly fell to ~ -72 °C) for 90 min, then warmed to room temperature over 30 minutes using a water bath. This solution was added to a sodium dihydrogen phosphate solution equivalent to 360 g of NaH2P04 in 1.5 L of ice/water, over about 10 minutes, with ice-bath cooling. The mixture was stirred for 20 minutes, then transferred to a sep. funnel, and partitioned. The aqueous layer was further extracted with EtOAc (2 x 1 L), and the combined organic extracts were dried over sodium sulfate. The volatiles were removed in vacuo (down to 20 mbar). The resultant oil was hydrolyzed crude.

(3R,4R,5R)-3-chIoro-4-hydroxy-5-(hydroxymethyI)-3-methyIoxoIan-2-one (A4):

Figure imgf000137_0002

H O CI[00276] The crude oil A2 was taken up in acetic acid (1.5 L, 66% in water) and heated to 90 °C over one hour, then at held at that temperature for one hour. Once the mixture had cooled to room temperature, the volatiles were removed in vacuo, and azeotroped with toluene (500 ml). The resultant oil was combined with some mixed material from an earlier synthesis and columned in two portions (each -1.25 L of silica, 38→ 75% EtOAc in DCM). The lower of the two main spots is the desired material; fractions containing this material as the major component were combined and the solvent removed in vacuo to give 82 g of orange solid whose 1 H NMR showed the material to be of about 57% purity (of the remainder 29% was the indicated epimer). This material was recrystallized fromtoluene/butanone (600 ml / -185 ml), the butanone being the ‘good’ solvent. The resultant solid was filtered washing with toluene and hexane, and dried in vacuo to give product of about 92% purity (30 g).(2R,3R,4R)-2-[(benzoyIoxy)methyI]-4-chIoro-4-methyI-5-oxooxoIan-3-yI benzoate(A5):

Figure imgf000138_0001

[00277] A 2 L 3 -neck round bottom flask was fitted with an overhead stirrer, thermometer and pressure equalizing dropping funnel (→N2). The intermediate A4 (160 mmol) in acetonitrile (1 L) was added, followed by 4-dimethylaminopyridine (3.2 mmol) and benzoyl chloride (352 mmol). Finally triethylamine (384 mmol) was added over 10 minutes using the dropping funnel. The addition of the triethylamine is accompanied by a mild exotherm, which obviated the addition of a cold water bath to keep the internal temperature below 25 °C. The reaction was stirred at ambient temperature for 2.5 hours. The reaction mixture was transferred to a sep. funnel with EtOAc (2 L) and half saturated brine (2 L), and partitioned. The aqueous layer was re-extracted with EtOAc (1 L). The combined organic layers were washed with 50%> sodium bicarbonate/25%) brine (1.5 L) and dried over sodium sulfate, to give 62 g of solid. This was recrystallized from 1.8 L of 1 : 1 toluene/trimethylpentane (95 °C), to give 52.4 g of product.[00278] 1H NMR (CDCls, 400 MHz): δ (ppm) 1.91 (s, 3H), 4.57 (dd, J= 5.12Hz and J = 12.57Hz, 1H), 4.77 (dd, J= 3.29Hz and J= 12.68Hz, 1H), 4.92-4.96 (m, 1H), 5.60 (d, J = 8.36Hz, 1H), 7.38-7.66 (m, 6H), 7.97-7.99 (m, 2H), 8.08-8.10 (m, 2H); MS (ESI) m/z= 411.1(MNa ).

3,5-Di-0-benzoyl-2-C-chloro-2-C-methyl-D-ribofuranose (A6):

Figure imgf000139_0001

[00279] To a solution of A5 (14.48 mmol) in anhydrous tetrahydrofurane (70 ml) was added under inert atmosphere at -35°C, LiAlH(OtBu)3 (1M in tetrahydrofurane, 21.7 mmol) over a 30 min period. The reaction mixture was stirred for 1 hour at -20 °C and quenched by addition of a saturated NH4C1 solution, keeping the temperature bellow 0 °C. Ethyl acetate was added and the white suspension was filtered through a pad of celite and washed with ethyl acetate. The filtrate was extracted with ethyl acetate twice. The combined organic layers were dried over anhydrous sodium sulfate, filtered and evaporated under reduced pressure. The residue was purified by chromatography on silica gel (eluent: petroleum ether/ethyl acetate 0 to 20%). The product was dried in vacuum (50 °C) overnight to afford expected intermediate as a colorless oil in 96% yield (mixture α/β: 45/55).[00280] 1H NMR (CDC13, 400 MHz): δ (ppm) 1.74 (s, 1.75HP), 1.76 (s, 1.25Ha), 4.42-4.69 (m, 3H), 5.30 (d, J= 12.8Hz, 0.55HP), 5.43-5.47 (m, 0.45Ha), 5.60 (d, J= 7.0Hz, 0.55HP), 5.78 (d, J= 7.0Hz , 0.45Ha), 7.35-7.41 (m, 2H), 7.45-7.56 (m, 3H), 7.59-7.65 (m, 1H), 7.96- 8.04 (m, 2H), 8.06-8.14 (m, 2H); MS (ESI) m/z= 413 (MNa+).3,5-Di-0-benzoyl-2-C-chloro-2-C-methyl-D-arabinofuranosyl bromide (A7):

Figure imgf000139_0002

[00281] To a solution of A6 (12.80 mmol) in anhydrous dichloromethane (80 ml) was added under inert atmosphere at -20 °C, triphenylphosphine (18.0 mmol). The reaction mixture was stirred for 15 minutes at -20 °C and CBr4 (19.20 mmol) was added. The reaction mixture was then stirred for 1 hour at -20 °C. The crude was partially concentrated under reduced pressure (bath temperature bellow 30 °C) and directly purified by chromatography on silica gel (eluent: petroleum ether/ethyl acetate 0 to 30%) to afford a mixture of β sugar A7a (1.67 g) and a sugar A7b (2.15 g) as a colorless gum in 66%> global yield.[00282] 1H NMR (CDC13, 400 MHz): β sugar δ (ppm) 1.93 (s, 3H), 4.60-4.88 (m, 3H), 6.08 (d, J= 7.9 Hz, 1H), 6.62 (s, 1H), 7.31-7.38 (m, 2H), 7.41-7.55 (m, 3H), 7.59-7.65 (m, 1H), 8.00-8.05 (m, 2H), 8.06-8.12 (m, 2H); a sugar δ (ppm) 1.88 (s, 3H), 4.66-4.89 (m, 3H), 5.37 (d, J= 4.88Hz, 1H), 6.44 (s, 1H), 7.41-7.55 (m, 4H), 7.54-7.65 (m, 2H), 8.00-8.05 (m, 2H), 8.14-8.20 (m, 2H); MS (ESI) m/z= 476/478 (MNa+).3 ,5′-Di-0-benzoyl-2′-C-chloro-2′-C-methyl-4-benzoyl-cytidine (A8):

Figure imgf000140_0001

[00283] To a suspension of N-benzoyl cytosine (9.48 mmol), and a catalytic amount of ammonium sulfate in 4-chlorobenzene (24 ml) was added HMDS (28.44 mmol). The reaction mixture was heated during 2 hours at 140 °C. The solvent was removed under inert atmosphere and the residue was taken in 4-chlorobenzene (15 ml). Then, A7b (4.74 mmol) in chlorobenzene (10 ml) was added dropwise to the reaction mixture followed by SnCl4 (14.22 mmol) dropwise. The reaction mixture was stirred at 70 °C overnight, cooled to room temperature and diluted with dichloromethane and a saturated NaHC03 solution. The white suspension was filtered through a pad of celite and washed with dichloromethane. The filtrate was extracted with dichloromethane twice. The combined organic layers were dried over anhydrous Na2S04, filtered and evaporated under reduced pressure to afford expected intermediate as a white solid in 89% yield.[00284] 1H NMR (DMSO, 400 MHz): δ (ppm) 1.58 (s, 3H), 4.68-4.81 (m, 3H), 5.68 (brs, 1H), 6.55 (brs, 1H), 7.36 (d, J= 7.84 Hz, 1H), 7.39-7.76 (m, 9H), 7.88-8.07 (m, 6H), 8.30 (d, J= 7.84 Hz, 1H); MS (ESI) m/z= 588 (MH+).3′,5′-Di-0-benzoyl-2,-C-chloro-2,-C-methyluridine (A9):

Figure imgf000140_0002

[00285] A suspension of A8 (4.19 mmol) in an acetic acid/water mixture (67 ml/17 ml, v/v), was heated at 110 °C for 3 hours. The reaction mixture was evaporated to dryness and co-evaporated with toluene (three times) to afford expected intermediate in quantitative yield as an oil which was directly used for the next step; MS (ESI) m/z= 485 (MH+). 2 -C-Chloro-2 -C-methyluridine (301):

Figure imgf000141_0001

H O CI[00286] Intermediate A9 (4.19 mmol) in 7 N methanolic ammonia (80 ml) was stirred at room temperature for 24 hours. The mixture was evaporated to dryness, diluted with water and transferred into a separatory funnel. The aqueous layer was extracted withdichloromethane and water was removed under reduced pressure. The residue was purified by flash RP18 gel chromatography (eluent: water/acetonitrile 0 to 40%) to afford pure expected compound as a white foam in 79% yield.[00287] 1H NMR (DMSO, 400 MHz): δ (ppm) 1.44 (s, 3H), 3.60-3.68 (m, 1H), 3.80-3.94 (m, 3H), 5.39 (t, J= 4.45 Hz, 1H), 5.63 (d, J= 8.26 Hz, 1H), 5.93 (d, J= 5.72 Hz, 1H), 6.21 (s, 1H), 8.16 (d, J= 8.90 Hz, 1H), 11.44 (m, 1H); MS (ESI) m/z= 277 (MH+).2′-C-Chloro-2′-C-methyl-3-benzyloxymethyluridine (Al 1):

Figure imgf000141_0002

H O CI[00288] To a solution of 301 (0.361 mmol) in anhydrous DMF (4 ml) was added at -5 °C, DBU (0.723 mmol) followed by benzyloxymethylchloride (0.542 mmol). The reaction mixture was stirred for 45 minutes between -5 °C and 5 °C. The solvent was evaporated under reduced pressure and the residue was purified by chromatography on silica gel (eluent: dichloromethane/methanol 0 to 10%) to afford pure expected intermediate as a white solid in 80% yield.[00289] 1H NMR (DMSO, 400 MHz): δ (ppm) 1.41 (s, 3H), 3.61-3.69 (m, 1H), 3.82-3.95 (m, 3H), 4.57 (s, 2H), 5.32 (s, 2H), 5.43 (t, J= 4.46Hz, 1H), 5.80 (d, J= 8.08Hz, 1H), 5.96 (d, J= 4.46 Hz, 1H), 6.23 (s, 1H), 7.22-7.36 (m, 5H), 8.25 (d, J= 8.22Hz, 1H); MS (ESI) m/z= 397 (MH+). Isopropyl (2S)-2-[[chloro(phenoxy)phosphoryl]amino]propanoate (A12a):

Figure imgf000142_0001

2,2-Dimethylpropyl (2S)-2-[[chloro(phenoxy)phosphoryl]amino]propanoate (A12b):

Figure imgf000142_0002

[00290] To a solution of aminoester, HC1 salt (0.434 mmol) in anhydrous dichloromethane (or acetonitrile) (4 ml) (3 times vacuo/nitrogen) under nitrogen was added at -30°C phenyldichlorophosphate (0.434 mmol) followed by N-methylimidazole (2.90 mmol)(or only 1.45 mmol for A12b). The reaction mixture was stirred at -30°C during 1 hour. The reaction was monitored by LC/MS (the sample was quenched by methanol or water) to check the complete formation of expected intermediate A12a [MS (ESI) m/z= 302 (MH+)(-OMe compounder A12b [MS (ESI) m/z= 314 (MH~)].Compound (A13a), (A13b) or (83ii):[00291] To the previous reaction mixture containing A12 was added All (or 302) (0.29 mmol) at -25°C under nitrogen. The reaction mixture was allowed to warm up slowly to room temperature overnight, and then diluted with dichloromethane and water (or with NaHCC”3 and EtOAc). The organic layer was extracted, dried, filtered and evaporated under reduced pressure. The crude residue was purified by chromatography on silica gel (eluent: dichloromethane/methanol 0 to 10%) (followed by preparative HPLC for A29).Compound (A13a):

Figure imgf000142_0003

[00292] Mixture of diastereoisomers; MS (ESI) m/z= 666 (MH+). Compound (A13b):

Figure imgf000143_0001

[00293] Mixture of diastereoisomers; MS (ESI) m/z= 692.3 (MH ).Compound (83ii):

Figure imgf000143_0002

[00294] Glassy solid; 1H NMR (CDCI3, 400MHz): δ (ppm) 1.19-1.24 (m, 9H), 1.35 (d, J = 7.1Hz, 3H), 3.95-4.05 (m, 1H), 4.31 (d, J= 8.1Hz, 2H), 4.41 (d, J= 9.0Hz, 1H), 4.59 (d, J = 7.1Hz, 2H), 4.98 (heptuplet, J= 6.28Hz, 1H), 6.38 (brs, 1H), 6.52 (s, 1H), 7.08-7.15 (m, 1H), 7.23-7.30 (m, 4H), 8.07 (s, 1H), 8.31 (s, 1H); 31P NMR (CDC13, 161.98 MHz): δ (ppm) 3.96 (s, IP); MS (ESI) m/z= 569.20 (MH+).Compounds (40iia) and (40iib):

Figure imgf000143_0003

[00295] To a solution of A13 (0.29 mmol) in anhydrous ethanol (6 ml) was added trifluoroacetic acid (2.9 mmol) dropwise (then 3 times vacuo/nitrogen purges), followed by Palladium hydroxide (20% on Carbon). The reaction mixture was purged 3 timesvacuo/nitrogen, and 3 times vacuo/hydrogen and then stirred under hydrogen for 5 hours. The reaction mixture was diluted with ethyl acetate and filtered through a pad of celite. The filtrate was evaporated under reduced pressure, and the crude compound was purified by preparative MS/HP LC to afford two pure compounds in 48% global yield.[00296] Compound 40ii (diastereoisomer 1): white solid; 1H NMR (CDC13, 400 MHz): δ (ppm) 1.22-1.26 (m, 6H), 1.37 (d, J= 7.08 Hz, 3H), 1.51 (s, 3H), 3.71-3.88 (m, 2H), 3.97- 4.06 (m, 1H), 4.16-4.18 (m, 1H), 4.45-4.57 (m, 2H), 4.97-5.07 (m, 1H), 5.57 (d, J= 8.20 Hz, 1H), 6.39 (s, 1H), 7.18-7.37 (m, 5H), 7.44 (d, J= 8.20 Hz, 1H), 8.40 (s, 1H); 31P NMR (CDC13, 161.98 MHz): δ (ppm) 4.20 (s, IP); MS (ESI, El+) m/z= 546 (MH+).[00297] Compound 40ii (diastereoisomer 2): white solid; 1H NMR (CDC13, 400 MHz): δ (ppm) 1.24-1.26 (m, 6H), 1.36 (d, J= 7.04 Hz, 3H), 1.59 (s, 3H), 3.69-3.77 (m, 1H), 3.91- 3.99 (m, 2H), 4.17-4.19 (m, 1H), 4.43-4.59 (m, 2H), 5.01-5.06 (m, 1H), 5.68 (d, J= 8.20 Hz, 1H), 6.42 (s, 1H), 7.21-7.39 (m, 5H), 7.60 (d, J=8.20 Hz, 1H), 8.14 (s, 1H); 31P NMR (CDC13, 161.98 MHz): δ (ppm) 3.47 (s, IP); MS (ESI) m/z= 546 (MH+).Compound 42ii:

Figure imgf000144_0001

[00298] Compound 42ii was synthesized from compound A13b (0.144 mmol) as described for compound 40ii.[00299] White solid; 1H NMR (MeOD, 400 MHz) δ (ppm) 0.94 (s, 9H), 1.40 (d, J= 7.10 Hz, 3H), 1.53 (s, 3H), 3.76 (d, J= 10.43 H, 1H), 3.86 (d, J= 10.44 H, 1H), 3.98-4.06 (m, 2H), 4.18-4.22 (m, 1H), 4.39-4.44 (m, 1H), 4.52-4.57 (m, 1H), 5.62 (d, J= 8.18 Hz, 1H), 6.40 (s, 1H), 7.20-7.29 (m, 3H), 7.36-7.41 (m, 2H), 7.74 (d, J= 8.18 Hz, 1H); 31P NMR (MeOD, 161.98 MHz) δ (ppm) 3.68 (s, IP); MS (ESI) m/z = 574.08 (MH+).

PAPER

US 20170226146

https://patents.google.com/patent/US20170226146A1/en

  • [0250]
  • [0251]
    A 3-neck 100 mL jacketed round bottom flask with nitrogen inlet and mechanical stirrer was charged with compound 4 (3.0 g, 10.8 mmol), compound 13 (0.484 g, 2.17 mmol, 0.20 equiv), 2-butanone (21 mL), and 2,6-lutidine (2.53 mL, 21.7 mmol, 2.0 equiv). The resulting slurry was cooled to −15° C., then a solution of compound 12 (7.96 g, 13.0 mmol) in 2-butanone (3 mL) was added over 14 hours. The reaction mixture was allowed to stir at −15° C. for an additional 25 hours and then warmed to 20° C. n-Heptane (16 mL) was added with stirring over a 1 hour period then the mixture was allowed to stir at 25° C. for 3 hours, then filtered through a fitted funnel. The filter cake was slurry-washed with a 3:2 mixture of 2-butanone and n-heptane (10 mL and then 15 mL), then dried by pulling nitrogen stream through the fritted funnel. The filter cake was slurried in a 10:1 mixture of water and 2-butanone (21 mL) and then filtered. This slurrying and filtration sequence was repeated two more times. The resulting filter cake was dried with nitrogen stream through the fritted funnel to provide compound 6.

Example 21Alternate Preparation of Compound A

  • [0252]
  • [0253]
    Compound 6 (0.072 mmol, 1 equiv), K2HPO(63.0 mg, 0.361 mmol) and compound 14 (5.45 mg, 0.018 mmol) were added to a 1 dram vial with 4 A mol sieves (40 mg). To the resulting mixture was added DCM (800 μl), then the resulting reaction was allowed to stir for 5 minutes. To the reaction mixture was then added compound 14 (28.7 mg, 0.094 mmol, 1.3 equiv) and the resulting reaction was allowed to stir for about 15 hours at room temperature to provide Compound A.
  • [0256]
  • [0257]
    A 100 mL reactor with nitrogen inlet and mechanical stirrer was charged with compound 4 (7.00 g, 25.3 mmol), compound 15 (0.225 g, 0.506 mmol, 0.020 equiv), 1,3-dioxolane (42 mL), and 2,6-lutidine (4.42 mL, 38.0 mmol, 1.5 equiv). The mixture was cooled to −10° C. and a 33 wt % solution of compound 12 in isopropyl acetate (29 mL, 30 mmol) was added over 1 hour. The reaction mixture was allowed to stir at −10° C. for additional 40 hours, then isopropyl acetate (28 mL) was added, and the resulting mixture was warmed to 0° C. A 10 wt % aqueous NaHSOsolution was added (14 mL), and the mixture was allowed to stir at 30° C. for 30 minutes, then the layers were separated. To the organic layer was added an aqueous solution containing 5 wt % NaHCOand 5 wt % Na2SO(21 mL). The mixture was allowed to stir at 50° C. for 6 h. The layers were separated. To the organic layer was added 10 wt % aqueous NaCl solution (21 mL). The mixture was allowed to stir at 50° C. for 30 min. The organic layer was separated, combined with isopropyl acetate (5 mL) and concentrated in vacuo to half volume at 20000 pa in a 50° C. bath. The resulting solution was solvent-switched with isopropanol (4×35 mL) to 60 g weight. The mixture was seeded with 100 mg of compound A at 60° C. The resulting slurry was allowed to stir at 55° C. for 30 minutes, then n-Heptane (35 mL) was added over 1 hour at 55° C. The resulting slurry was allowed to stir for an additional 1 hour at 55° C., then cooled to room temperature and filtered. The filter cake was washed with a 1:1 mixture of isopropanol and n-heptane (3×14 mL), followed by n-heptane (14 mL), then dried under nitrogen to provide Compound A.

PAPER

https://pubs.rsc.org/en/content/articlelanding/2021/sc/d1sc01978c#!divAbstract

Uprifosbuvir is an antiviral agent developed for treatment of chronic hepatitis C infections. Its original synthesis route requires twelve steps with an overall yield of only 1 %. Such a difficult and time-consuming synthesis approach is acceptable for the early trial phase of a new drug, but impractical for broad application as hepatitis C treatment or for repurposing against novel viral diseases.

Artis Klapars, John Y. L. Chung, and colleagues, Merck & Co., Inc., Rahway, NJ, USA, and WuXi STA, Shanghai, China, have developed a synthesis route for uprifosbuvir requiring only five steps and starting from readily available uridine. Initially, uridine is selectively oxidized after OH-acylation with pivaloyl chloride in an acyl migration/oxidation process driven by complexation with the Lewis acid BF3*OEt2 in toluene. In the second step, methylation is achieved by MeMgBr/MgCl2 in a toluene/anisole mixture where a more reactive methyl-manganese species is formed in-situ from the Grignard reagent, providing high yield and a good diastereomeric ratio (dr). Subsequently, the tertiary chloride group is introduced. Due to the high functional-group density, a cyclodehydration step is required before chlorination to avoid side reactions. The chlorination is carried out using dichlorodimethylsilane with FeCl3*6H2O and tetramethyldisiloxane as additives which avoids the hazardous use of HCl gas under pressure required in the initial synthesis. In the final step, the regioselective phosphoramidation is achieved using a chlorophosphoramidate precursor and a dimeric chiral imidazole carbamate catalyst which led to a dr of 97:3 starting from a 1:1 diastereomeric mixture of the chlorophosphoramidate reagent.

Uprifosbuvir was synthesized with an overall yield of 50 %, a vast improvement compared to the 1 % of the original synthesis route. Additionally, the newly developed synthesis steps have the potential to provide easier access to other nucleoside-based antiviral agents.


Efficient synthesis of antiviral agent uprifosbuvir enabled by new synthetic methods

Artis Klapars,  *a

This article is Open Access

Creative Commons BY license

All publication charges for this article have been paid for by the Royal Society of Chemistry

Abstract

An efficient route to the HCV antiviral agent uprifosbuvir was developed in 5 steps from readily available uridine in 50% overall yield. This concise synthesis was achieved by development of several synthetic methods: (1) complexation-driven selective acyl migration/oxidation; (2) BSA-mediated cyclization to anhydrouridine; (3) hydrochlorination using FeCl3/TMDSO; (4) dynamic stereoselective phosphoramidation using a chiral nucleophilic catalyst. The new route improves the yield of uprifosbuvir 50-fold over the previous manufacturing process and expands the tool set available for synthesis of antiviral nucleotides.

Graphical abstract: Efficient synthesis of antiviral agent uprifosbuvir enabled by new synthetic methods

Scheme 1 Synthetic approaches to uprifosbuvir 1 with the two main challenges highlighted. (a) Me2NH, AcOH, EtOH/MeOH, 80 °C, 1.5 h; (b) Ca(OH)2, water, 70 °C, 24 h, 19% over 2 steps.9

Scheme 3 Complexation-driven selective acyl migration/oxidation to access 12. (a) PivCl, pyridine, 0 °C, 16 h; (b) BF3·OEt2, PhMe, 40 °C, 10 h; (c) TEMPO, Bu4NBr, AcOOH, dioctyl sulphide, PhMe, −10 °C to 20 °C, 24 h, 83% from 5.

Scheme 6 Completion of uprifosbuvir synthesis. (a) TMS-Cl, iPrOH, 70 °C, 12 h; (b) NEt3, iPrOAc, wiped film evaporation, 80%; (c) PhOP(O)Cl2, NEt3, iPrOAc, −20 °C, 2 h, 90%; (d) C6F5OH, NEt3, iPrOAc, −5 °C to 10 °C, 18 h, 76%;26 (e) 4, 3 mol% 24, 2,6-lutidine, 1,3-dioxolane, −10 °C, 24 h, 88%; (f) 4, tBuMgCl, THF, −5 °C to 5 °C, 15 h, 50%;27 (g) 4, Me2AlCl, 2,6-lutidine, THF, 35 °C, 16 h, 81%.27

Scheme 7 Summary of uprifosbuvir synthesis. AY = assay yield; IY = isolated yield. 

https://www.rsc.org/suppdata/d1/sc/d1sc01978c/d1sc01978c1.pdf

PAPERhttps://www.sciencedirect.com/science/article/abs/pii/S0960894X17308314

References

  1. ^ Soriano V, Fernandez-Montero JV, de Mendoza C, Benitez-Gutierrez L, Peña JM, Arias A, Barreiro P (August 2017). “Treatment of hepatitis C with new fixed dose combinations”. Expert Opinion on Pharmacotherapy18 (12): 1235–1242. doi:10.1080/14656566.2017.1346609PMID 28644739S2CID 205819421.
  2. ^ Borgia G, Maraolo AE, Nappa S, Gentile I, Buonomo AR (March 2018). “NS5B polymerase inhibitors in phase II clinical trials for HCV infection”. Expert Opinion on Investigational Drugs27 (3): 243–250. doi:10.1080/13543784.2018.1420780PMID 29271672S2CID 3672885.
  3. ^ Lawitz E, Gane E, Feld JJ, Buti M, Foster GR, Rabinovitz M, et al. (September 2019). “Efficacy and safety of a two-drug direct-acting antiviral agent regimen ruzasvir 180 mg and uprifosbuvir 450 mg for 12 weeks in adults with chronic hepatitis C virus genotype 1, 2, 3, 4, 5 or 6”. Journal of Viral Hepatitis26 (9): 1127–1138. doi:10.1111/jvh.13132PMID 31108015S2CID 160014275.
 
Clinical data
Trade names Uprifosbuvir
Legal status
Legal status US: Investigational New Drug
Identifiers
showIUPAC name
CAS Number 1496551-77-9
PubChem CID 90055716
DrugBank DB15206
ChemSpider 57427403
UNII JW31KPS26S
KEGG D10996
ChEMBL ChEMBL3833371
Chemical and physical data
Formula C22H29ClN3O9P
Molar mass 545.9 g·mol−1
3D model (JSmol) Interactive image
showSMILES
showInChI

Uprifosbuvir (MK-3682) is an antiviral drug developed for the treatment of Hepatitis C. It is a nucleotide analogue which acts as an NS5B RNA polymerase inhibitor. It is currently in Phase III human clinical trials.[1][2][3]

References

  1. ^ Soriano V, Fernandez-Montero JV, de Mendoza C, Benitez-Gutierrez L, Peña JM, Arias A, Barreiro P (August 2017). “Treatment of hepatitis C with new fixed dose combinations”. Expert Opinion on Pharmacotherapy18 (12): 1235–1242. doi:10.1080/14656566.2017.1346609PMID 28644739S2CID 205819421.
  2. ^ Borgia G, Maraolo AE, Nappa S, Gentile I, Buonomo AR (March 2018). “NS5B polymerase inhibitors in phase II clinical trials for HCV infection”. Expert Opinion on Investigational Drugs27 (3): 243–250. doi:10.1080/13543784.2018.1420780PMID 29271672S2CID 3672885.
  3. ^ Lawitz E, Gane E, Feld JJ, Buti M, Foster GR, Rabinovitz M, et al. (September 2019). “Efficacy and safety of a two-drug direct-acting antiviral agent regimen ruzasvir 180 mg and uprifosbuvir 450 mg for 12 weeks in adults with chronic hepatitis C virus genotype 1, 2, 3, 4, 5 or 6”. Journal of Viral Hepatitis26 (9): 1127–1138. doi:10.1111/jvh.13132PMID 31108015S2CID 160014275.

//////////uprifosbuvir, MK 3682, ウプリホスブビル, уприфосбувирأوبريفوسبوفير , 乌磷布韦 , IDX-21437DB15206SB18784D10996Q27281714, IDX 21437, PHASE 3 
CC(C)OC(=O)C(C)NP(=O)(OCC1C(C(C(O1)N2C=CC(=O)NC2=O)(C)Cl)O)OC3=CC=CC=C3

wdt-24

NEW DRUG APPROVALS

ONE TIME

$10.00

RIDINILAZOLE


ChemSpider 2D Image | Ridinilazole | C24H16N6
Ridinilazole.svg

RIDINILAZOLE

SMT19969

  • Molecular FormulaC24H16N6
  • Average mass388.424 Da
  • ридинилазол [Russian] [INN]ريدينيلازول [Arabic] [INN]利地利唑 [Chinese] [INN]
  • リジニラゾール;

10075
2,2′-Di(4-pyridinyl)-3H,3’H-5,5′-bibenzimidazole
308362-25-6[RN]6,6′-Bi-1H-benzimidazole, 2,2′-di-4-pyridinyl-

Summit Therapeutics (formerly Summit Corp ) is developing ridinilazole the lead compound from oral narrow-spectrum, GI-restricted antibiotics, which also include SMT-21829, for the treatment of Clostridium difficile infection and prevention of recurrent disease.

Ridinilazole (previously known as SMT19969) is an investigational small molecule antibiotic being evaluated for oral administration to treat Clostridioides difficile infection (CDI). In vitro, it is bactericidal against C. difficile and suppresses bacterial toxin production; the mechanism of action is thought to involve inhibition of cell division.[1] It has properties which are desirable for the treatment of CDI, namely that it is a narrow-spectrum antibiotic which exhibits activity against C. difficile while having little impact on other normal intestinal flora and that it is only minimally absorbed systemically after oral administration.[2] At the time ridinilazole was developed, there were only three antibiotics in use for treating CDI: vancomycinfidaxomicin, and metronidazole.[1][2] The recurrence rate of CDI is high, which has spurred research into other treatment options with the aim to reduce the rate of recurrence.[3][4]

As of 2019, two phase II trials have been completed and two phase III trials comparing ridinilazole to vancomycin for CDI are expected to be completed in September 2021.[2][5][6] Ridinilazole was designated as a Qualified Infectious Disease Product (QIDP) and was granted Fast Track status by the U.S. FDA.[2] Fast Track status is reserved for drugs designed to treat diseases where there is currently a gap in the treatment, or a complete lack thereof.[7] The QIDP designation adds five more years of exclusivity for ridinazole upon approval.[8]

str1-1

PATENT

WO-2021009514

Process for preparing ridinilazole useful for treating Clostridium difficile infection. Also claimed is the crystalline form of a compound.

The present invention relates to processes for the preparation of 2,2′-di(pyridin-4-yl)-1/-/,T/-/-5,5′-bibenzo[d]imidazole (which may also be known as 5,5’-bis[2-(4-pyridinyl)-1/-/-benzimidazole], 2,2′-bis(4-pyridyl)-3/-/,3’/-/-5,5′-bibenzimidazole or 2-pyridin-4-yl-6-(2-pyridin-4-yl-3/-/-benzimidazol-5-yl)-1/-/-benzimidazole), referenced herein by the INN name ridinilazole, and pharmaceutically acceptable derivatives, salts, hydrates, solvates, complexes, bioisosteres, metabolites or prodrugs thereof. The invention also relates to various crystalline forms of ridinilazole, to processes for their preparation and to related pharmaceutical preparations and uses thereof (including their medical use and their use in the efficient large-scale synthesis of ridinilazole).

WO2010/063996 describes various benzimidazoles, including ridinilazole, and their use as antibacterials (including in the treatment of CDAD).

WO 2011/151621 describes various benzimidazoles and their use as antibacterials

(including in the treatment of CDAD).

W02007056330, W02003105846 and W02002060879 disclose various 2-amino benzimidazoles as antibacterial agents.

W02007148093 discloses various 2-amino benzothiazoles as antibacterial agents.

W02006076009, W02004041209 and Bowser et at. (Bioorg. Med. Chem. Lett., 2007, 17, 5652-5655) disclose various substituted benzimidazole compounds useful as anti-infectives that decrease resistance, virulence, or growth of microbes. The compounds are said not to exhibit intrinsic antimicrobial activity in vitro.

US 5,824,698 discloses various dibenzimidazoles as broad-spectrum antibiotics, disclosing activity against both Gram-negative and Gram-positive bacteria, including Staphylococcus spp.and Enterococcus spp. However, this document does not disclose activity against anaerobic spore-forming bacteria and in particular does not disclose activity against any Clostridioides spp. (including C. difficile).

US 2007/0112048 A1 discloses various bi- and triarylimidazolidines and bi- and

triarylamidines as broad-spectrum antibiotics, disclosing activity against both Gram negative and Gram-positive bacteria, including Staphylococcus spp., Enterococcus spp. and Clostridioides spp. However, this document does not disclose compounds of formula (I) as described herein.

Chaudhuri et al. (2007) J.Org. Chem. 72, 1912-1923 describe various bis-2-(pyridyl)-1 H-benzimidazoles (including compounds of formula I as described herein) as DNA binding agents. This document is silent as to potential antibacterial activity.

Singh et al. (2000) Synthesis 10: 1380-1390 describe a condensation reaction for producing 2,2′-di(pyridin-4-yl)-1/-/,T/-/-5,5′-bibenzo[d]imidazole using 4-pyridine

carboxaldehyde, FeCI3, 02, in DMF at 120°C.

Bhattacharya and Chaudhuri (2007) Chemistry – An Asian Journal 2: 648-655 describe a condensation reaction for producing 2,2′-di(pyridin-4-yl)-1/-/,T/-/-5,5′-bibenzo[d]imidazole using 4-pyridine carboxaldehyde and nitrobenzene at 120°C.

WO2019/068383 describes the synthesis of ridinilazole by metal-ion catalyzed coupling of 3,4,3’,4’-tetraaminobiphenyl with 4-pyridinecarboxaldehyde in the presence of oxygen, followed by the addition of a complexing agent.

PATENT

WO2010063996

claiming antibacterial compounds. Bicyclic heteroaromatic compounds, particularly bi-benzimidazole derivatives.

WO2007056330, WO2003105846 and WO2002060879 disclose various 2-amino benzimidazoles as antibacterial agents.

WO2007148093 discloses various 2-amino benzothiazoles as antibacterial agents.

WO2006076009, WO2004041209 and Bowser et al. (Bioorg. Med. Chem. Lett., 2007, 17, 5652-5655) disclose various substituted benzimidazole compounds useful as anti-infectives that decrease resistance, virulence, or growth of microbes. The compounds are said not to exhibit intrinsic antimicrobial activity in vitro.

US 5,824,698 discloses various dibenzimidazoles as broad-spectrum antibiotics, disclosing activity against both Gram-negative and Gram-positive bacteria, including Staphylococcus spp.and Enterococcus spp. However, this document does not disclose activity against anaerobic spore-forming bacteria and in particular does not disclose activity against any Clostridium spp. (including C. difficile).

US 2007/0112048 A1 discloses various bi- and triarylimidazolidines and bi- and triarylamidines as broad-spectrum antibiotics, disclosing activity against both Gram-negative and Gram-positive bacteria, including Staphylococcus spp., Enterococcus spp.

and Clostridium spp. However, this document does not disclose compounds of general formula (I) as described herein.

Chaudhuri et al. (J.Org. Chem., 2007, 72, 1912-1923) describe various bis-2-(pyridyl)-1 H-benzimidazoles (including compounds of formula I as described herein) as DNA binding agents. This document is silent as to potential antibacterial activity.

PATENT

Product PATENT, WO2010063996 ,

protection in the EP until 2029 and expire in the US in December 2029.

PAPER

https://www.frontiersin.org/articles/10.3389/fmicb.2018.01206/full

PAPER

Synthesis (2000), (10), 1380-1390.

https://www.thieme-connect.de/products/ejournals/abstract/10.1055/s-2000-7111

PAPERT

Chemistry – An Asian Journal (2007), 2(5), 648-655.

https://onlinelibrary.wiley.com/doi/abs/10.1002/asia.200700014

Studies of double‐stranded‐DNA binding have been performed with three isomeric bis(2‐(n‐pyridyl)‐1H‐benzimidazole)s (n=2, 3, 4). Like the well‐known Hoechst 33258, which is a bisbenzimidazole compound, these three isomers bind to the minor groove of duplex DNA. DNA binding by the three isomers was investigated in the presence of the divalent metal ions Mg2+, Co2+, Ni2+, Cu2+, and Zn2+. Ligand–DNA interactions were probed with fluorescence and circular dichroism spectroscopy. These studies revealed that the binding of the 2‐pyridyl derivative to DNA is dramatically reduced in the presence of Co2+, Ni2+, and Cu2+ ions and is abolished completely at a ligand/metal‐cation ratio of 1:1. Control experiments done with the isomeric 3‐ and 4‐pyridyl derivatives showed that their binding to DNA is unaffected by the aforementioned transition‐metal ions. The ability of 2‐(2‐pyridyl)benzimidazole to chelate metal ions and the conformational changes of the ligand associated with ion chelation probably led to such unusual binding results for the ortho isomer. The addition of ethylenediaminetetraacetic acid (EDTA) reversed the effects completely.

PAPER

 Journal of Organic Chemistry (2007), 72(6), 1912-1923.

https://pubs.acs.org/doi/10.1021/jo0619433

Three symmetrical positional isomers of bis-2-(n-pyridyl)-1H-benzimidazoles (n = 2, 3, 4) were synthesized and DNA binding studies were performed with these isomeric derivatives. Like bisbenzimidazole compound Hoechst 33258, these molecules also demonstrate AT-specific DNA binding. The binding affinities of 3-pyridine (m-pyben) and 4-pyridine (p-pyben) derivatized bisbenzimidazoles to double-stranded DNA were significantly higher compared to 2pyridine derivatized benzimidazole o-pyben. This has been established by combined experimental results of isothermal fluorescence titration, circular dichroism, and thermal denaturation of DNA. To rationalize the origin of their differential binding characteristics with double-stranded DNA, computational structural analyses of the uncomplexed ligands were performed using ab initio/Density Functional Theory. The molecular conformations of the symmetric head-to-head bisbenzimidazoles have been computed. The existence of intramolecular hydrogen bonding was established in o-pyben, which confers a conformational rigidity to the molecule about the bond connecting the pyridine and benzimidazole units. This might cause reduction in its binding affinity to double-stranded DNA compared to its para and meta counterparts. Additionally, the predicted stable conformations for p-, m-, and o-pyben at the B3LYP/6-31G* and RHF/6-31G* levels were further supported by experimental pKa determination. The results provide important information on the molecular recognition process of such symmetric head to head bisbenzimidazoles toward duplex DNA.

Patent

US 8975416

PATENT

WO 2019068383

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019068383

Clostridium difficile infection (CDI) is the leading cause of infectious healthcare-associated diarrhoea. CDI remains a challenge to treat clinically, because of a limited number of antibiotics available and unacceptably high recurrence rates. Because of this, there has been significant demand for creating innovative therapeutics, which has resulted in the development of several novel antibiotics.

Ridinilazole (SMT19969) is the INN name of 5,5’bis[2-(4-pyridinyl)-lH-benzimidazole], which is a promising non-absorbable small molecule antibiotic intended for oral use in the treatment of CDI. It has been shown to exhibit a prolonged post-antibiotic effect and treatment with ridinilazole has resulted in decreased toxin production. A phase 1 trial demonstrated that oral ridinilazole is well tolerated and specifically targets Clostridia whilst sparing other faecal bacteria.

Ridinilazole has the following chemical structure:

Bhattacharya & Chaudhuri (Chem. Asian J., 2007, No. 2, 648-655) report performing double-stranded DNA binding with three benzimidazole derivatives, including ridinilazole. The compounds have been prepared by dissolving the reactants in nitrobenzene, heating at 120°C for 8- 1 Oh and purifying the products by column chromatography over silica gel. The compounds were obtained in 65-70% yield. Singh et al., (Synthesis, 2000, No. 10, 1380-1390) describe a catalytic redox cycling approach based on Fe(III) and molecular oxygen as co-oxidant for providing access to benzimidazole and

imidazopyridine derivatives, such as ridinilazole. The reaction is performed at high temperatures of 120°C and the product is isolated in 91% yield by using silica flash chromatography.

Both processes are not optimal, for example in terms of yield, ease of handling and scalability. Thus, there is a need in the art for an efficient and scalable preparation of ridinilazole, which overcomes the problems of the prior art processes.

Example 1 : Preparation of crude ridinilazole free base

A solution of 3,4,3′,4′-tetraaminobiphenyl (3.28 g, 15.3 mmol) and isonicotinaldehyde (3.21 g, 30.0 mmol) in DMF (40 mL) was stirred at 23 °C for one hour. Then anhydrous ferric chloride (146 mg, 0.90 mmol), water (0.10 mL, 5.4 mmol) and additional DMF (2 mL) were added and fresh air was bubbled into the solution during vigorous stirring for 5 hours at room temperature. Next, water (80 mL) and EDTA (0.29 g) were added resulting in a brownish suspension, which was stirred overnight. The product was isolated by filtration, washed with water, and dried in a desiccator in vacuo as a brown powder (5.56 g; 95%). The addition of EDTA had held iron in solution and the crude ridinilazole contained significantly lower amounts of iron than comparative example 1.

Example 12: Formation of essentially pure ridinilazole free base

To a suspension von ridinilazole tritosylate (1 10 mg, 0.12 mmol) in water (35 mL) featuring a pH value of about 4.5 stirring at 70 °C sodium bicarbonate (580 mg, 6.9 mmol) were added and caused a change of color from orange to slightly tan. The mixture, now at a pH of about 8.5, was cooled down to room temperature and the solids were separated by filtration, washed with water (1 ML) and dried in vacuo providing 40 mg (85%) essentially pure ridinilazole as a brownish powder.

Spectroscopic analysis:

¾ NMR (DMSO-de, 300 MHz): δ 7.55 (d, J = 8.4 Hz, 2H), 7.70 (d, J = 8.4 Hz, 2H), 7.88 (s, 2H), 8.13 (d, J = 5.8 Hz, 4H), 8.72 (d, J = 5.8 Hz, 4H) ppm.

13C NMR (DMSO-d6, 75 MHz): δ 1 13.4 (2C), 1 16.4 (2C), 120.4 (4C), 121.8 (2C), 135.7 (2C), 138.7 (2C), 140.7 (2C), 141.4 (2C), 150.3 (4C), 151.1 (2C) ppm.

IR (neat): v 3033 (w), 1604 (s), 1429 (m), 1309 (m), 1217 (m), 1 1 15 (w), 998 (m), 964 (m), 824 (m), 791 (s), 690 (s), 502 (s) cm .

UV-Vis (MeOH): 257, 341 nm.

The sharp peaks in the ¾ NMR indicated that iron had been efficiently removed.

Comparative example 1 : Preparation of ridinilazole

A solution of 3,4,3′,4′-tetraaminobiphenyl (0.69 g, 3.2 mmol) and isonicotinaldehyde (0.64 g, 6.0 mmol) in DMF (20 mL) was stirred at 80°C for one hour. Then ferric chloride hexahydrate (49 mg, 0.18 mmol), water (0.10 mL, 5.4 mmol) and additional DMF (2 mL) were added and fresh air was bubbled into the solution during vigorous stirring for 10 hours at 120 °C. After cooling to room temperature water (50 mL) and the mixture was stirred for one hour. A black crude product was isolated by filtration and comprised ridinilazole and iron.

References

  1. Jump up to:a b Cho JC, Crotty MP, Pardo J (March 2019). “Clostridium difficile infection”Annals of Gastroenterology32 (2): 134–140. doi:10.20524/aog.2018.0336PMC 6394264PMID 30837785.
  2. Jump up to:a b c d Carlson TJ, Endres BT, Bassères E, Gonzales-Luna AJ, Garey KW (April 2019). “Ridinilazole for the treatment of Clostridioides difficile infection”Expert Opinion on Investigational Drugs28 (4): 303–310. doi:10.1080/13543784.2019.1582640PMID 30767587.
  3. ^ Bassères E, Endres BT, Dotson KM, Alam MJ, Garey KW (January 2017). “Novel antibiotics in development to treat Clostridium difficile infection”Current Opinion in Gastroenterology33 (1): 1–7. doi:10.1097/MOG.0000000000000332PMID 28134686These tables highlight the increased drug development directed towards CDI due to the rise in prevalence of infections and to attempt to reduce the number of recurrent infections.
  4. ^ Vickers RJ, Tillotson G, Goldstein EJ, Citron DM, Garey KW, Wilcox MH (August 2016). “Ridinilazole: a novel therapy for Clostridium difficile infection”International Journal of Antimicrobial Agents48 (2): 137–43. doi:10.1016/j.ijantimicag.2016.04.026PMID 27283730there exists a significant unmet and increasing medical need for new therapies to treat CDI, specifically those that can reduce the rate of disease recurrence.
  5. ^ Clinical trial number NCT03595553 for “Ri-CoDIFy 1: Comparison of Ridinilazole Versus Vancomycin Treatment for Clostridium Difficile Infection” at ClinicalTrials.gov
  6. ^ Clinical trial number NCT03595566 for “Ri-CoDIFy 2: To Compare Ridinilazole Versus Vancomycin Treatment for Clostridium Difficile Infection” at ClinicalTrials.gov
  7. ^ “Fast Track”. U.S. Food and Drug Administration. 2018-11-03.
  8. ^ “”HHS spurs new antibiotic development for biodefense and common infections””Public Health Emergency. U.S. Department of Health and Human Services. Retrieved 2020-12-04.
Clinical data
Other namesSMT19969
ATC codeNone
Identifiers
IUPAC name[show]
CAS Number308362-25-6
PubChem CID16659285
ChemSpider17592423
UNII06DX01190R
KEGGD11958
Chemical and physical data
FormulaC24H16N6
Molar mass388.42 g/mol
3D model (JSmol)Interactive image
SMILES[hide]c6cc(c5nc4ccc(c3ccc2nc(c1ccncc1)[nH]c2c3)cc4[nH]5)ccn6

/////////RIDINILAZOLE, SMT19969, SMT 19969, ридинилазол , ريدينيلازول , 利地利唑 , リジニラゾール , Qualified Infectious Disease Product, QIDP,  Fast Track , PHASE 3,  Clostridioides difficile infection , 

REPROXALAP


2-(3-Amino-6-chloroquinolin-2-yl)propan-2-ol.png

REPROXALAP

レプロキサラップ;

ADX-102

2-(3-amino-6-chloroquinolin-2-yl)propan-2-ol

C12H13ClN2O, 236.7 g/mol

CAS 916056-79-6

UNII-F0GIZ22IJH

2-(3-amino-6-chloroquinolin-2-yl)propan-2-ol

Phase 3 Clinical

Aldeyra Therapeutics is developing reproxalap, which binds and traps free aldehydes, formulated using Captisol technology licensed from Ligand Pharmaceuticals as an eye drop formulation, for treating acute noninfectious anterior uveitis, allergic conjunctivitis and dry eye syndrome.

PATENT

product case, WO2006127945 ,

EU states until 2026

expire US in 2029 with US154 extension.

PATENTS

WO2018170476

United States patent application serial number US 13/709,802, filed December 10, 2012 and published as US 2013/0190500 on July 25, 2013 (“the ‘500 publication,” the entirety of which is hereby incorporated herein by reference), describes certain aldehyde scavenging compounds. Such compounds include com ound A:

[0036] Compound A, (6-chloro-3-amino-2-(2-hydroxypropyl)-l-azanaphthalene), is designated as compound A in the ‘500 publication and the synthesis of compound A is described in detail at Example 5 of the ‘500 publication, and is reproduced herein for ease of reference.

Example A – General Preparation of Compound A

Compound A

[00436] The title compound was prepared according to the steps and intermediates (e.g., Scheme 1) described below and in the ‘500 publication, the entirety of which is incorporated herein by reference.

Step 1: Synthesis of Intermediate A- 1

[00437] To a 2 L round bottom flask was charged ethanol (220 mL), and pyridine (31 g, 392 mmol) and the resulting solution stirred at a moderate rate of agitation under nitrogen. To this solution was added ethyl bromopyruvate (76.6 g, 354 mmol) in a slow, steady stream. The reaction mixture was allowed to stir at 65±5° C. for 2 hours.

Step 2: Synthesis of Intermediate A-2

[00438] Upon completion of the 2-hour stir time in example 1, the reaction mixture was slowly cooled to 18-22° C. The flask was vacuum-purged three times at which time 2-amino-5-chloro-benzaldehyde (ACB) (50.0 g, 321 mmol) was added directly to the reaction flask as a solid using a long plastic funnel. Pyridine (64.0 g, 809 mmol) was added followed by an EtOH rinse (10 mL) and the reaction mixture was heated at 80±3° C. under nitrogen for about 16 hours (overnight) at which time HPLC analysis indicated that the reaction was effectively complete.

Step 3: Synthesis of Intermediate A-3

[00439] The reaction mixture from example 2 was cooled to about 70° C. and morpholine (76.0 g, 873 mmol)) was added to the 2 L reaction flask using an addition funnel. The reaction mixture was heated at 80±2° C. for about 2.5 hours at which time the reaction was considered complete by HPLC analysis (area % of A-3 stops increasing). The reaction mixture was cooled to 10-15° C. for the quench, work up, and isolation.

Step 4: Isolation of Intermediate A-3

[00440] To the 2 L reaction flask was charged water (600 g) using the addition funnel over 30-60 minutes, keeping the temperature below 15° C. by adjusting the rate of addition and using a cooling bath. The reaction mixture was stirred for an additional 45 minutes at 10-15° C. then the crude A-3 isolated by filtration using a Buchner funnel. The cake was washed with water (100 mLx4) each time allowing the water to percolate through the cake before applying a vacuum. The cake was air dried to provide crude A-3 as a nearly dry brown solid. The cake was returned to the 2 L reaction flask and heptane (350 mL) and EtOH (170 mL) were added and the mixture heated to 70±3° C. for 30-60 minutes. The slurry was cooled to 0-5° C. and isolated by filtration under vacuum. The A-3 was dried in a vacuum drying oven under vacuum and 35±3° C. overnight (16-18 hours) to provide A-3 as a dark green solid.

Step 5: Synthesis of Compound A

[00441] To a 2 L round bottom flask was charged methylmagnesium chloride (200 mL of 3.0 M solution in THF, 600 mmol). The solution was cooled to 0-5° C. using an ice bath.

[00442] A 500 mL flask (magnetic stirring) was charged with 22.8 grams A-3 from example 4 and THF (365 mL), stirred to dissolve then transferred to an addition funnel on the 2 L Reaction Flask. The A-3 solution was added drop-wise to the reaction flask over 5.75 hours, keeping the temperature of the reaction flask between 0-5° C throughout the addition. At the end of the addition the contents of the flask were stirred for an additional 15 minutes at 0-5° C. then the cooling bath was removed and the reaction was allowed to stir overnight at ambient temperature.

[00443] The flask was cooled in an ice bath and the reaction mixture was carefully quenched by adding EtOH (39.5 g, 857 mmol) drop-wise to the reaction mixture, keeping the temperature of the reaction mixture below 15° C. during the course of the addition. An aqueous solution of H4C1 (84.7 g H4C1 in 415 mL water) was then carefully added and the mixture stirred under moderate agitation for about 30 minutes then transferred to a separately funnel to allow the layers to separate. Solids were present in the aqueous phase so HO Ac (12.5 g) was added and the contents swirled gently to obtain a nearly homogeneous lower aqueous phase. The lower aqueous layer was transferred back to the 2 L reaction flask and stirred under moderate agitation with 2-methylTHF (50 mL) for about 15 minutes. The original upper organic layer was reduced in volume to approximately 40 mL using a rotary evaporator at≤40° C. and vacuum as needed. The phases in the separatory funnel were separated and the upper 2-MeTHF phase combined with the product residue, transferred to a 500 mL flask and vacuum distilled to an approximate volume of 25 mL. To this residue was added 2-MeTHF (50 mL) and distilled to an approximate volume of 50 mL. The crude compound A solution was diluted with 2-MeTHF (125 mL), cooled to 5-10° C. and 2M H2S04 (aq) (250 mL) was slowly added and the mixture stirred for 30 minutes as the temperature was allowed to return to ambient. Heptane (40 mL) was charged and the reaction mixture stirred for an additional 15 minutes then transferred to a separatory funnel and the layers were allowed to separate. The lower aqueous product layer was extracted with additional heptane (35 mL) then the lower aqueous phase was transferred to a 1 L reaction flask equipped with a mechanical stirrer and the mixture was cooled to 5-10° C. The combined organic layers were discarded. A solution of 25% NaOH(aq) was prepared (NaOH, 47 g, water, 200 mL) and slowly added to the 1 L reaction flask to bring the pH to a range of 6.5-8.5.

[00444] EtOAc (250 mL) was added and the mixture was stirred overnight. The mixture was transferred to a separatory funnel and the lower phase discarded. The upper organic layer was washed with brine (25 mL) then the upper organic product layer was reduced in volume on a rotary evaporator to obtain the crude compound A as a dark oil that solidified within a few minutes. The crude compound A was dissolved in EtOAc (20 mL) and filtered through a plug of silica gel (23 g) eluting with 3/1 heptane/EtOAc until all compound A was eluted (approximately 420 mL required) to remove most of the dark color of compound A. The solvent was removed in vacuo to provide 14.7 g of compound A as a tan solid. Compound A was taken up in EtOAc (25 mL) and eluted through a column of silica gel (72 g) using a mobile phase gradient of 7/1 heptane/EtOAc to 3/lheptane/EtOAc (1400 mL total). The solvent fractions containing compound A were stripped, compound A diluted with EtOAc (120 mL) and stirred in a flask with Darco G-60 decolorizing carbon (4.0 g) for about 1 hour. The mixture was filtered through celite using a fitted funnel, rinsing the cake with EtOAc (3 x 15 mL). The combined filtrates were stripped on a rotary evaporator and compound A dissolved in heptane (160 mL)/EtOAc(16 mL) at 76° C. The

homogeneous solution was slowly cooled to 0-5° C, held for 2 hours then compound A was isolated by filtration. After drying in a vacuum oven for 5 hours at 35° C. under best vacuum, compound A was obtained as a white solid. HPLC purity: 100% (AUC).

Example 1 – Preparation of Free Base Forms A and B of Compound A

Compound A

[00445] Compound A is prepared according to the method described in detail in Examples 1-5 of the ‘500 publication, the entirety of which is hereby incorporated herein by reference.

PATENT

example 5 [WO2018039197A1]

https://patents.google.com/patent/WO2018039197A1/en

Exam le 5: Synthesis of NS2

Figure imgf000055_0001

NS2

[00190] 2-(3-amino-6-chloroquinolin-2-yl)propan-2-ol. To a 2 L round bottom flask was charged methylmagnesium chloride (200 mL of 3.0 M solution in THF, 600 mmol). The solution was cooled to 0-5 °C using an ice bath.

[00191] A 500 mL flask (magnetic stirring) was charged with 22.8 grams A-3a from Example 4 and THF (365 mL), stirred to dissolve, and then transferred to an addition funnel on the 2 L reaction flask. The A-3a solution was added drop-wise to the reaction flask over 5.75 hours, keeping the temperature of the reaction flask between 0-5 °C throughout the addition. At the end of the addition the contents of the flask were stirred for an additional 15 minutes at 0-5 °C, then the cooling bath was removed and the reaction was allowed to stir overnight at ambient temperature.

[00192] The flask was cooled in an ice bath and the reaction mixture was carefully quenched by adding EtOH (39.5 g, 857 mmol) drop-wise to the reaction mixture, keeping the temperature of the reaction mixture below 15 °C during the course of the addition. An aqueous solution of H4CI (84.7 g H4CI in 415 mL water) was then carefully added and the mixture stirred under moderate agitation for about 30 minutes then transferred to a separatory funnel to allow the layers to separate. Solids were present in the aqueous phase so HOAc (12.5 g) was added and the contents swirled gently to obtain a nearly homogeneous lower aqueous phase. The lower aqueous layer was transferred back to the 2 L reaction flask and stirred under moderate agitation with 2-methyl-tetrahydrofuran (2-MeTHF) (50 mL) for about 15 minutes. The original upper organic layer was reduced in volume to approximately 40 mL using a rotary evaporator at < 40 °C under vacuum as needed. The phases in the separatory funnel were separated and the upper 2-MeTHF phase combined with the product residue was transferred to a 500 mL flask and vacuum distilled to an approximate volume of 25 mL. To this residue was added 2-MeTHF (50 mL) and the mixture again distilled to an approximate volume of 50 mL. The crude compound NS2 solution was diluted with 2-MeTHF (125 mL), cooled to 5-10 °C, and 2 M H2S04 (aq) (250 mL) was slowly added and the mixture stirred for 30 minutes as the temperature was allowed to return to ambient. Heptane (40 mL) was charged and the reaction mixture stirred for an additional 15 minutes then transferred to a separatory funnel, and the layers were allowed to separate. The lower aqueous product layer was extracted with additional heptane (35 mL), then the lower aqueous phase was transferred to a 1 L reaction flask equipped with a mechanical stirrer, and the mixture was cooled to 5-10 °C. The combined organic layers were discarded. A solution of 25% NaOH (aq) was prepared (NaOH, 47 g, water, 200 mL) and slowly added to the 1 L reaction flask to bring the pH to a range of 6.5 – 8.5.

[00193] EtOAc (250 mL) was added and the mixture was stirred overnight. The mixture was transferred to a separatory funnel and the lower phase discarded. The upper organic layer was washed with brine (25 mL), then the upper organic product layer was reduced in volume on a rotary evaporator to obtain a obtain the crude compound NS2 as a dark oil that solidified within a few minutes. The crude compound NS2 was dissolved in EtOAc (20 mL) and filtered through a plug of silica gel (23 g) eluting with 3/1 heptane/EtOAc until all compound NS2 was eluted (approximately 420 mL required) to remove most of the dark color of compound NS2. The solvent was removed in vacuo to provide 14.7 g of compound NS2 as a tan solid. Compound NS2 was taken up in EtOAc (25 mL) and eluted through a column of silica gel (72g) using a mobile phase gradient of 7/1 heptane/EtOAc to 3/1 heptane/EtOAc (1400 mL total). The solvent fractions containing compound NS2 were evaporated. Compound NS2 was diluted with EtOAc (120 mL) and stirred in a flask with Darco G-60 decolorizing carbon (4.0 g) for about 1 hour. The mixture was filtered through celite using a firtted funnel, rinsing the cake with EtOAc (3 x 15 mL). The combined filtrates were evaporated on a rotary evaporator and compound NS2 dissolved in heptane (160 mL)/EtOAc (16 mL) at 76 °C. The homogeneous solution was slowly cooled to 0-5 °C, held for 2 hours, then compound NS2 was isolated by filtration. After drying in a vacuum oven for 5 hours at 35 °C under best vacuum, compound NS2 was obtained as a white solid. HPLC purity: 100% (AUC); HPLC (using standard conditions): A-2: 7.2 minutes; A-3 : 11.6 minutes.

Preparation of ACB

Figure imgf000057_0001

[00194] After a N2 atmosphere had been established and a slight stream of N2 was flowing through the vessel, platinum, sulfided, 5 wt. % on carbon, reduced, dry (9.04 g, 3.0 wt. % vs the nitro substrate) was added to a 5 L heavy walled pressure vessel equipped with a large magnetic stir-bar and a thermocouple. MeOH (1.50 L), 5-chloro-2-nitrobenzaldehyde (302.1 g, 1.63 mol), further MeOH (1.50 L) and Na2C03 (2.42 g, 22.8 mmol, 0.014 equiv) were added. The flask was sealed and stirring was initiated at 450 rpm. The solution was evacuated and repressurized with N2 (35 psi), 2x. The flask was evacuated and repressurized with H2 to 35 psi. The temperature of the solution reached 30 °C w/in 20 min. The solution was then cooled with a water bath. Ice was added to the water bath to maintain a temperature below 35 °C. Every 2h, the reaction was monitored by evacuating and repressurizing with N2 (5 psi), 2x prior to opening. The progress of the reaction could be followed by TLC: 5-Chloro-2-nitrobenzaldehyde (Rf = 0.60, CH2CI2, UV) and the intermediates (Rf = 0.51, CH2CI2, UV and Rf = 0.14, CH2CI2, UV) were consumed to give ACB (Rf = 0.43, CH2CI2, UV). At 5 h, the reaction had gone to 98% completion (GC), and was considered complete. To a 3 L medium fritted funnel was added celite (ca. 80 g). This was settled with MeOH (ca. 200 mL) and pulled dry with vacuum. The reduced solution was transferred via cannula into the funnel while gentle vacuum was used to pull the solution through the celite plug. This was chased with MeOH (4 x 150 mL). The solution was transferred to a 5 L three-necked round-bottom flask. At 30 °C on a rotavap, solvent (ca. 2 L) was removed under reduced pressure. An N2 blanket was applied. The solution was transferred to a 5L four-necked round-bottomed flask equipped with mechanical stirring and an addition funnel. Water (2.5 L) was added dropwise into the vigorously stirring solution over 4 h. The slurry was filtered with a minimal amount of vacuum. The collected solid was washed with water (2 x 1.5 L), 2-propanol (160 mL) then hexanes (2 x 450 mL). The collected solid (a canary yellow, granular solid) was transferred to a 150 x 75 recrystallizing dish. The solid was then dried under reduced pressure (26-28 in Hg) at 40°C overnight in a vacuum-oven. ACB (> 99% by HPLC) was stored under a N2 atmosphere at 5°C.

PATENT

WO-2020223717

Process for preparing reproxalap as acetaldehyde dehydrogenase inhibitor useful for treating ocular diseases and cancer.

PATENT

WO-2020223685

Novel crystalline forms of reproxalap (compound 1; designated as Forms A and B) as acetaldehyde dehydrogenase inhibitor useful for treating ocular diseases and cancer.

PATENT

WO 2020123730

//////////REPROXALAP, レプロキサラップ  , ADX-102, Phase 3 Clinical

CC(C)(C1=C(C=C2C=C(C=CC2=N1)Cl)N)O

Odevixibat


img

Odevixibat.png

Odevixibat

A-4250, AR-H 064974

CAS 501692-44-0

BUTANOIC ACID, 2-(((2R)-2-((2-((3,3-DIBUTYL-2,3,4,5-TETRAHYDRO-7-(METHYLTHIO)-1,1-DIOXIDO-5-PHENYL-1,2,5-BENZOTHIADIAZEPIN-8-YL)OXY)ACETYL)AMINO)-2-(4-HYDROXYPHENYL)ACETYL)AMINO)-, (2S)-

(2S)-2-[[(2R)-2-[[2-[(3,3-dibutyl-7-methylsulfanyl-1,1-dioxo-5-phenyl-2,4-dihydro-1λ6,2,5-benzothiadiazepin-8-yl)oxy]acetyl]amino]-2-(4-hydroxyphenyl)acetyl]amino]butanoic acid

Molecular Formula C37H48N4O8S2
Molecular Weight 740.929
        • UPDATE 7/20/2021FDA APPROVED, To treat pruritus,

      Bylvay

    • New Drug Application (NDA): 215498
      Company: ALBIREO PHARMA INC
  • Orphan Drug Status Yes – Primary biliary cirrhosis; Biliary atresia; Intrahepatic cholestasis; Alagille syndrome
  • New Molecular Entity Yes
  • Phase III Biliary atresia; Intrahepatic cholestasis
  • Phase II Alagille syndrome; Cholestasis; Primary biliary cirrhosis
  • No development reported Non-alcoholic steatohepatitis
  • 22 Jul 2020 Albireo initiates an expanded-access programme for Intrahepatic cholestasis in USA, Canada, Australia and Europe
  • 14 Jul 2020 Phase-III clinical trials in Biliary atresia (In infants, In neonates) in Belgium (PO) after July 2020 (EudraCT2019-003807-37)
  • 14 Jul 2020 Phase-III clinical trials in Biliary atresia (In infants, In neonates) in Germany, France, United Kingdom, Hungary (PO) (EudraCT2019-003807-37)

UPDATE Bylvay, FDA APPROVED2021/7/20 AND EMA 2021/7/16

Odevixibat, sold under the trade name Bylvay, is a medication for the treatment of progressive familial intrahepatic cholestasis (PFIC).[1]

The most common side effects include diarrhea, abdominal pain, hemorrhagic diarrhea, soft feces, and hepatomegaly (enlarged liver).[1]

Odevixibat is a reversible, potent, selective inhibitor of the ileal bile acid transporter (IBAT).[1][2]

In May 2021, the Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency (EMA) recommended granting a marketing authorization in the European Union for odevixibat for the treatment of PFIC in people aged six months or older.[1][3]

A-4250 (odevixibat) is a selective inhibitor of the ileal bile acid transporter (IBAT) that acts locally in the gut. Ileum absorbs glyco-and taurine-conjugated forms of the bile salts. IBAT is the first step in absorption at the brush-border membrane. A-4250 works by decreasing the re-absorption of bile acids from the small intestine to the liver, whichreduces the toxic levels of bile acids during the progression of the disease. It exhibits therapeutic intervention by checking the transport of bile acids. Studies show that A-4250 has the potential to decrease the damage in the liver cells and the development of fibrosis/cirrhosis of the liver known to occur in progressive familial intrahepatic cholestasis. A-4250 is a designated orphan drug in the USA for October 2012. A-4250 is a designated orphan drug in the EU for October 2016. A-4250 was awarded PRIME status for PFIC by EMA in October 2016. A-4250 is in phase II clinical trials by Albireo for the treatment of primary biliary cirrhosis (PBC) and cholestatic pruritus. In an open label Phase 2 study in children with cholestatic liver disease and pruritus, odevixibat showed reductions in serum bile acids and pruritus in most patients and exhibited a favorable overall tolerability profile.

str1

albireo_logo_nav.svg

Odevixibat is a highly potent, non-systemic ileal bile acid transport inhibitor (IBATi) that has has minimal systemic exposure and acts locally in the small intestine. Albireo is developing odevixibat to treat rare pediatric cholestatic liver diseases, including progressive familial intrahepatic cholestasisbiliary atresia and Alagille syndrome.

With normal function, approximately 95 percent of bile acids released from the liver into the bile ducts to aid in liver function are recirculated to the liver via the IBAT in a process called enterohepatic circulation. In people with cholestatic liver diseases, the bile flow is interrupted, resulting in elevated levels of toxic bile acids accumulating in the liver and serum. Accordingly, a product capable of inhibiting the IBAT could lead to a reduction in bile acids returning to the liver and may represent a promising approach for treating cholestatic liver diseases.

The randomized, double-blind, placebo-controlled, global multicenter PEDFIC 1 Phase 3 clinical trial of odevixibat in 62 patients, ages 6 months to 15.9 years, with PFIC type 1 or type 2 met its two primary endpoints demonstrating that odevixibat reduced serum bile acids (sBAs) (p=0.003) and improved pruritus (p=0.004), and was well tolerated with a low single digit diarrhea rate. These topline data substantiate the potential for odevixibat to be first drug for PFIC patients. The Company intends to complete regulatory filings in the EU and U.S. no later than early 2021, in anticipation of regulatory approval, issuance of a rare pediatric disease priority review voucher and launch in the second half of 2021.

Odevixibat is being evaluated in the ongoing PEDFIC 2 open-label trial (NCT03659916) designed to assess long-term safety and durability of response in a cohort of patients rolled over from PEDFIC 1 and a second cohort of PFIC patients who are not eligible for PEDFIC 1.

Odevixibat is also currently being evaluated in a second Phase 3 clinical trial, BOLD (NCT04336722), in patients with biliary atresia. BOLD, the largest prospective intervention trial ever conducted in biliary atresia, is a double-blind, randomized, placebo-controlled trial which will enroll approximately 200 patients at up to 75 sites globally to evaluate the efficacy and safety of odevixibat in children with biliary atresia who have undergone a Kasai procedure before age three months. The company also anticipates initiating a pivotal trial of odevixibat for Alagille syndrome by the end of 2020.

For more information about the PEDFIC 2 or BOLD studies, please visit ClinicalTrials.gov or contact medinfo@albireopharma.com.

The odevixibat PFIC program, or elements of it, have received fast track, rare pediatric disease and orphan drug designations in the United States. In addition, the FDA has granted orphan drug designation to odevixibat for the treatment of Alagille syndrome, biliary atresia and primary biliary cholangitis. The EMA has granted odevixibat orphan designation, as well as access to the PRIority MEdicines (PRIME) scheme for the treatment of PFIC. Its Paediatric Committee has agreed to Albireo’s odevixibat Pediatric Investigation Plan for PFIC. EMA has also granted orphan designation to odevixibat for the treatment of biliary atresia, Alagille syndrome and primary biliary cholangitis.

PATENT

https://patents.google.com/patent/US9694018B1/en

Example 5

1,1-Dioxo-3,3-dibutyl-5-phenyl-7-methylthio-8-(N—{(R)-α-[N—((S)-1-carboxypropyl) carbamoyl]-4-hydroxybenzyl}carbamoylmethoxy)-2,3,4,5-tetrahydro-1,2,5-benzothiadiazepine, Mw. 740.94.

This compound is prepared as described in Example 29 of WO3022286.

PATENT

https://patents.google.com/patent/WO2003022286A1/sv

Example 29

1,1-Dioxo-3,3-dibutyl-5-phenyl-7-methylthio-8-(N-((R)-α-[N-((S)- 1-carboxypropyl) carbamoyl]-4-hydroxybenzyl}carbamoylmethoxy)-2,3,4,5-tetrahydro-1,2,5-benzothiadiazepine

A solution of 1,1-dioxo-3,3-dibutyl-5-phenyl-7-methylthio-8-[N-((R)-α-carboxy-4-hydroxybenzyl)carbamoylmethoxy]-2,3,4,5-tetrahydro-1,2,5-benzothiadiazepine (Example 18; 0.075 g, 0.114 mmol), butanoic acid, 2-amino-, 1,1-dimethylethyl ester, hydrochloride, (2S)-(0.031 g, 0.160 mmol) and Ν-methylmorpholine (0.050 ml, 0.457 mmol) in DMF (4 ml) was stirred at RT for 10 min, after which TBTU (0.048 g, 0.149 mmol) was added. After 1h, the conversion to the ester was complete. M/z: 797.4. The solution was diluted with toluene and then concentrated. The residue was dissolved in a mixture of DCM (5 ml) and TFA (2 ml) and the mixture was stirred for 7h. The solvent was removed under reduced pressure. The residue was purified by preparative HPLC using a gradient of 20-60% MeCΝ in 0.1M ammonium acetate buffer as eluent. The title compound was obtained in 0.056 g (66 %) as a white solid. ΝMR (400 MHz, DMSO-d6): 0.70 (3H, t), 0.70-0.80 (6H, m), 0.85-1.75 (14H, m), 2.10 (3H, s), 3.80 (2H, brs), 4.00-4.15 (1H, m), 4.65 (1H, d(AB)), 4.70 (1H, d(AB)), 5.50 (1H, d), 6.60 (1H, s), 6.65-7.40 (11H, m), 8.35 (1H, d), 8.50 (1H, d) 9.40 (1H, brs).

PATENT

https://patents.google.com/patent/US20140323412A1/en

PATENT

https://patents.google.com/patent/WO2013063526A1/e

PATENT

https://patents.google.com/patent/WO2019245448A1/en

The compound l,l-dioxo-3,3-dibutyl-5-phenyl-7-methylthio-8-(A/-{(R)-a-[A/-((S)-l-carboxypropyl) carbamoyl]-4-hydroxybenzyl}carbamoylmethoxy)-2,3,4,5-tetrahydro-l,2,5-benzothiadiazepine (odevixibat; also known as A4250) is disclosed in WO 03/022286. The structure of odevixibat is shown below.

Figure imgf000002_0001

As an inhibitor of the ileal bile acid transporter (IBAT) mechanism, odevixibat inhibits the natural reabsorption of bile acids from the ileum into the hepatic portal circulation. Bile acids that are not reabsorbed from the ileum are instead excreted into the faeces. The overall removal of bile acids from the enterohepatic circulation leads to a decrease in the level of bile acids in serum and the liver. Odevixibat, or a pharmaceutically acceptable salt thereof, is therefore useful in the treatment or prevention of diseases such as dyslipidemia, constipation, diabetes and liver diseases, and especially liver diseases that are associated with elevated bile acid levels.

According to the experimental section of WO 03/022286, the last step in the preparation of odevixibat involves the hydrolysis of a tert-butyl ester under acidic conditions. The crude compound was obtained by evaporation of the solvent under reduced pressure followed by purification of the residue by preparative HPLC (Example 29). No crystalline material was identified.

Amorphous materials may contain high levels of residual solvents, which is highly undesirable for materials that should be used as pharmaceuticals. Also, because of their lower chemical and physical stability, as compared with crystalline material, amorphous materials may display faster

decomposition and may spontaneously form crystals with a variable degree of crystallinity. This may result in unreproducible solubility rates and difficulties in storing and handling the material. In pharmaceutical preparations, the active pharmaceutical ingredient (API) is for that reason preferably used in a highly crystalline state. Thus, there is a need for crystal modifications of odevixibat having improved properties with respect to stability, bulk handling and solubility. In particular, it is an object of the present invention to provide a stable crystal modification of odevixibat that does not contain high levels of residual solvents, that has improved chemical stability and can be obtained in high levels of crystallinity.

Example 1

Preparation of crystal modification 1

Absolute alcohol (100.42 kg) and crude odevixibat (18.16 kg) were charged to a 250-L GLR with stirring under nitrogen atmosphere. Purified water (12.71 kg) was added and the reaction mass was stirred under nitrogen atmosphere at 25 ± 5 °C for 15 minutes. Stirring was continued at 25 ± 5 °C for 3 to 60 minutes, until a clear solution had formed. The solution was filtered through a 5.0 m SS cartridge filter, followed by a 0.2 m PP cartridge filter and then transferred to a clean reactor.

Purified water (63.56 kg) was added slowly over a period of 2 to 3 hours at 25 ± 5 °C, and the solution was seeded with crystal modification 1 of odevixibat. The solution was stirred at 25 ± 5 °C for 12 hours. During this time, the solution turned turbid. The precipitated solids were filtered through centrifuge and the material was spin dried for 30 minutes. The material was thereafter vacuum dried in a Nutsche filter for 12 hours. The material was then dried in a vacuum tray drier at 25 ± 5 °C under vacuum (550 mm Hg) for 10 hours and then at 30 ± 5 °C under vacuum (550 mm Hg) for 16 hours. The material was isolated as an off-white crystalline solid. The isolated crystalline material was milled and stored in LDPE bags.

An overhydrated sample was analyzed with XRPD and the diffractogram is shown in Figure 2.

Another sample was dried at 50 °C in vacuum and thereafter analysed with XRPD. The diffractogram of the dried sample is shown in Figure 1.

The diffractograms for the drying of the sample are shown in Figures 3 and 4 for 2Q ranges 5 – 13 ° and 18 – 25 °, respectively (overhydrated sample at the bottom and dry sample at the top).

References

  1. Jump up to:a b c d “First treatment for rare liver disease”European Medicines Agency (EMA) (Press release). 21 May 2021. Retrieved 21 May 2021. Text was copied from this source which is © European Medicines Agency. Reproduction is authorized provided the source is acknowledged.
  2. ^ “Odevixibat”Albireo Pharma. Retrieved 21 May 2021.
  3. ^ “Bylvay: Pending EC decision”European Medicines Agency (EMA). 19 May 2021. Retrieved 21 May 2021.

External links

  • “Odevixibat”Drug Information Portal. U.S. National Library of Medicine.

ClinicalTrials.gov

CTID Title Phase Status Date
NCT04336722 Efficacy and Safety of Odevixibat in Children With Biliary Atresia Who Have Undergone a Kasai HPE (BOLD) Phase 3 Recruiting 2020-09-02
NCT04483531 Odevixibat for the Treatment of Progressive Familial Intrahepatic Cholestasis Available 2020-08-25
NCT03566238 This Study Will Investigate the Efficacy and Safety of A4250 in Children With PFIC 1 or 2 Phase 3 Active, not recruiting 2020-03-05
NCT03659916 Long Term Safety & Efficacy Study Evaluating The Effect of A4250 in Children With PFIC Phase 3 Recruiting 2020-01-21
NCT03608319 Study of A4250 in Healthy Volunteers Under Fasting, Fed and Sprinkled Conditions Phase 1 Completed 2018-09-19
CTID Title Phase Status Date
NCT02630875 A4250, an IBAT Inhibitor in Pediatric Cholestasis Phase 2 Completed 2018-03-29
NCT02360852 IBAT Inhibitor A4250 for Cholestatic Pruritus Phase 2 Terminated 2017-02-23
NCT02963077 A Safety and Pharmakokinetic Study of A4250 Alone or in Combination With A3384 Phase 1 Completed 2016-11-16

EU Clinical Trials Register

EudraCT Title Phase Status Date
2019-003807-37 A Double-Blind, Randomized, Placebo-Controlled Study to Evaluate the Efficacy and Safety of Odevixibat (A4250) in Children with Biliary Atresia Who Have Undergone a Kasai Hepatoportoenterostomy (BOLD) Phase 3 Ongoing 2020-07-29
2015-001157-32 An Exploratory Phase II Study to demonstrate the Safety and Efficacy of A4250 Phase 2 Completed 2015-05-13
2014-004070-42 An Exploratory, Phase IIa Cross-Over Study to Demonstrate the Efficacy Phase 2 Ongoing 2014-12-09
2017-002325-38 An Open-label Extension Study to Evaluate Long-term Efficacy and Safety of A4250 in Children with Progressive Familial Intrahepatic Cholestasis Types 1 and 2 (PEDFIC 2) Phase 3 Ongoing
2017-002338-21 A Double-Blind, Randomized, Placebo-Controlled, Phase 3 Study to Demonstrate Efficacy and Safety of A4250 in Children with Progressive Familial Intrahepatic Cholestasis Types 1 and 2 (PEDFIC 1) Phase 3 Ongoing, Completed

.

Odevixibat
Odevixibat structure.png
Clinical data
Trade names Bylvay
Routes of
administration
By mouth
ATC code
  • None
Identifiers
CAS Number
  • 501692-44-0
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
Chemical and physical data
Formula C37H48N4O8S2
Molar mass 740.93 g·mol−1
3D model (JSmol)

////////////odevixibat, Orphan Drug Status, phase 3, Albireo, A-4250, A 4250, AR-H 064974

CCCCC1(CN(C2=CC(=C(C=C2S(=O)(=O)N1)OCC(=O)NC(C3=CC=C(C=C3)O)C(=O)NC(CC)C(=O)O)SC)C4=CC=CC=C4)CCCC

publicationnumber
US-2020046635-A1
US-2020046636-A1 US-2020046757-A1 US-2020046758-A1 US-2020002299-A1 WO-2019245448-A1 WO-2019245449-A1 US-2019046451-A1 US-2019070217-A1
US-10441605-B2
US-2017224720-A1
US-2017224721-A1
US-2018264029-A1
US-2018360869-A1
US-2018360870-A1
US-2018360871-A1
WO-2017133517-A1
US-2017143738-A1
US-2017143783-A1
EP-2968230-A2
EP-2968262-A1
US-2014271734-A1
US-2014275090-A1
WO-2014144485-A1
WO-2014144485-A9
WO-2014144650-A2
EP-2770990-A1
EP-2771003-A1
EP-2771003-B1
EP-3266457-A1
EP-3278796-A1
US-10512657-B2
US-2013108573-A1
US-2013109671-A1
US-2013338093-A1
US-2014243281-A1
US-2014323412-A1
US-2016310518-A1
US-2017368085-A1
US-2019169217-A1
US-2020069715-A1
WO-2013063512-A1
WO-2013063526-A1
EP-2739286-A2
WO-2013020108-A2
EP-2637646-B1
EP-2637668-B1
EP-3023102-A1
EP-3023102-B1
EP-3400944-A1
US-10000528-B2
US-10011633-B2
US-10093697-B2
US-10221212-B2
US-2012114588-A1
US-2013225511-A1
US-2013236541-A1
US-2015031636-A1
US-2015031637-A1
US-2016193277-A1
US-2016194353-A1
US-2017182059-A1
US-2017182115-A1
US-2018022776-A1
US-2018030088-A1
US-2018030089-A1
US-2018162904-A1
US-2018362577-A1
US-9688720-B2
US-9694018-B1
US-10555950-B2
US-2016220577-A1
US-9339480-B2
WO-2008039829-A2
US-2009069285-A1
US-7842684-B2
WO-2007051995-A2
EP-1896408-A1
EP-1896409-A1
EP-1896457-A1
US-2010048529-A1
US-2010048530-A1
US-2010137273-A1
US-2010152156-A1
US-2010168039-A1
US-2010168075-A1
US-7893048-B2
US-7906502-B2
WO-2006137792-A1
WO-2006137794-A1
WO-2006137795-A1
US-2010216759-A1
US-7863265-B2
US-2008194494-A1
US-2009186834-A1
WO-2006102674-A2
US-2009005321-A1
EP-1831151-A1
US-2008114064-A1
WO-2006065214-A1
EP-1699759-A1
US-2007142304-A1
US-2008064676-A1
US-2010099657-A2
US-7871998-B2
WO-2005061452-A1
EP-1638922-A1
EP-1638926-A1
EP-1638930-A1
EP-1675820-A2
EP-1676833-A1
US-2005148656-A1
US-2006142389-A1
US-2006178432-A1
US-2006194879-A1
US-2006258866-A1
US-2007099928-A1
US-2007099997-A1
US-2007244198-A1
US-7309720-B2
WO-2004110984-A1
WO-2004113270-A2
WO-2004113276-A1
WO-2004113283-A1
EP-1610770-A1
EP-1610770-B1
EP-1894564-A2
US-2006199797-A1
US-7514421-B2
WO-2004089350-A1
EP-1572626-A1
US-2005131068-A1
WO-2004056748-A1
EP-1539120-A1
US-2006083790-A1
WO-2004006899-A1
EP-1521742-A1
US-2005239766-A1
US-7470678-B2
WO-2004005247-A1
EP-1517679-A1
EP-1517679-B1
EP-1517883-A1
EP-1517883-B1
EP-1517883-B8
US-2005222261-A1
US-2005256198-A1
US-2005267149-A1
US-7351858-B2
US-7355069-B2
US-7521461-B2
WO-2004000294-A1
WO-2004000790-A1
EP-1478368-A1
US-2005124557-A1
WO-03061663-A1
EP-1458672-A1
EP-1458672-B1
EP-1458673-A1
EP-1458673-B1
EP-1458677-A1
EP-1458677-B1
US-2005113362-A1
US-2005171204-A1
US-2005215630-A1
US-2005282822-A1
US-7256307-B2
US-7276539-B2
US-7488844-B2
US-7514471-B2
WO-03051821-A1
WO-03051822-A1
WO-03051826-A1
EP-1427423-B1
EP-1427423-B9
US-2005038009-A1
US-7132416-B2