New Drug Approvals

Home » 2020 » September

Monthly Archives: September 2020

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

Blog Stats

  • 4,479,833 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

Tetrahydrobiopterin,


Kuvan (Saproterin Dihydrochloride Tablets): Uses, Dosage, Side Effects, Interactions, Warning

Sapropterin

Sapropterin dihydrochloride, Dapropterin dihydrochloride, R-THBP, 6R-BH4, SUN-0588, Phenoptin, Biopten, Biobuden, Bipten

Approval:US: Dec’07, EU: Dec’08

Approval:US: Dec’07, EU: Dec’08

IUPAC Name

(6R)-2-amino-6-[(1R,2S)-1,2-dihydroxypropyl]-3,4,5,6,7,8-hexahydropteridin-4-one

SMILES

[H][C@@]1(CNC2=C(N1)C(=O)NC(N)=N2)[C@@H](O)[C@H](C)O
сапроптерин [Russian] [INN]
سابروبتيرين [INN]
沙丙蝶呤 [Chinese] [INN]
  • 17528-72-2
  • 27070-47-9
  • Sun 0588
  • 6R-BH4
  • R-THBP
  • Sapropterin
  • Sapropterina
  • sapropterinum
  • Tetrahydrobiopterin
Title: Sapropterin
CAS Registry Number: 62989-33-7
CAS Name: (6R)-2-Amino-6-[(1R,2S)-1,2-dihydroxypropyl]-5,6,7,8-tetrahydro-4(1H)-pteridinone
Additional Names: (6R)-L-erythro-tetrahydrobiopterin; dapropterin; R-THBP; 6R-BH4
Molecular Formula: C9H15N5O3
Molecular Weight: 241.25
Percent Composition: C 44.81%, H 6.27%, N 29.03%, O 19.90%
Literature References: Natural cofactor of the aromatic amino acid hydroxylases required for catecholamine and serotonin biosynthesis. Identification of cofactor activity: S. Kaufman, Proc. Natl. Acad. Sci. USA 50, 1085 (1963). Prepn of (6R,S)-BH4: B. Schircks et al., Helv. Chim. Acta 61, 2731 (1978). Chromatographic separation of diastereoisomers: S. W. Bailey, J. E. Ayling, J. Biol. Chem. 253, 1598 (1978). Absolute configuration of natural isomer: W. L. F. Armarego et al., Aust. J. Chem. 35, 785 (1982). Stereospecific synthesis: S. Matsuura et al., Heterocycles 23, 3115 (1985); H. Sakai, T. Kanai, EP 191335eidem, US 4713454 (1986, 1987 both to Shiratori; Suntory). Bioavailability: G. Kapatos, S. Kaufman, Science 212, 955 (1981). Effect on neurotransmitter monoamine biosynthesis: S. Miwa et al., Arch. Biochem. Biophys. 239, 234 (1985). LC determn in biological samples: Y. Tani, T. Ishihara, Life Sci. 46, 373 (1990). Therapeutic potential in hyperphenylalaninemia: S. Kaufman, J. Nutr. Sci. Vitaminol, Suppl., 601 (1992).
Properties: pK¢ 5.05. uv max (0.1 N HCl): 265 nm (e 14000).
pKa: pK¢ 5.05
Absorption maximum: uv max (0.1 N HCl): 265 nm (e 14000)
Derivative Type: Dihydrochloride
CAS Registry Number: 69056-38-8
Manufacturers’ Codes: SUN-0588
Trademarks: Biopten (Maruho)
Molecular Formula: C9H15N5O3.2HCl
Molecular Weight: 314.17
Percent Composition: C 34.41%, H 5.45%, N 22.29%, O 15.28%, Cl 22.57%
Properties: Crystals from HCl, mp 245-246° (dec). [a]D25 -6.81° (c = 0.665 in 0.1 M HCl). uv max (2 M HCl): 264 nm (e 16770).
Melting point: mp 245-246° (dec)
Optical Rotation: [a]D25 -6.81° (c = 0.665 in 0.1 M HCl)
Absorption maximum: uv max (2 M HCl): 264 nm (e 16770)
Therap-Cat: In treatment of hyperphenylalaninemia.
Keywords: Enzyme Cofactor
INGREDIENT UNII CAS INCHI KEY
Sapropterin dihydrochloride RG277LF5B3 69056-38-8 RKSUYBCOVNCALL-NTVURLEBSA-N

Experimental Properties

PROPERTY VALUE SOURCE
melting point (°C) 250-255 °C (hydrochloride salt) Not Available
water solubility >20 mg/mL (dichloride salt) Not Available
logP -1.7 Not Available

Tetrahydrobiopterin (BH4THB), also known as sapropterin (INN),[2][3] is a cofactor of the three aromatic amino acid hydroxylase enzymes,[4] used in the degradation of amino acid phenylalanine and in the biosynthesis of the neurotransmitters serotonin (5-hydroxytryptamine, 5-HT), melatonindopaminenorepinephrine (noradrenaline), epinephrine (adrenaline), and is a cofactor for the production of nitric oxide (NO) by the nitric oxide syntheses.[5] Chemically, its structure is that of a (dihydropteridine reductase) reduced pteridine derivative (Quinonoid dihydrobiopterin).[6]

Medical use

Tetrahydrobiopterin is available as a tablet for oral administration in the form of sapropterin dihydrochloride (BH4*2HCL).[7][8][9] It was approved for use in the United States as a tablet in December 2007[10][11] and as a powder in December 2013.[12][11] It was approved for use in the European Union in December 2008,[9] Canada in April 2010,[11] and Japan in July 2008.[11] It is sold under the brand names Kuvan and Biopten.[9][8][11] The typical cost of treating a patient with Kuvan is US$100,000 per year.[13] BioMarin holds the patent for Kuvan until at least 2024, but Par Pharmaceutical has a right to produce a generic version by 2020.[14]

Sapropterin is indicated in tetrahydrobiopterin deficiency caused by GTP cyclohydrolase I (GTPCH) deficiency, or 6-pyruvoyltetrahydropterin synthase (PTPS) deficiency.[15] Also, BH4*2HCL is FDA approved for use in phenylketonuria (PKU), along with dietary measures.[16] However, most people with PKU have little or no benefit from BH4*2HCL.[17]

Sapropterin (tetrahydrobiopterin or BH4) is a cofactor in the synthesis of nitric oxide. It is also essential in the conversion of phenylalanine to tyrosine by the enzyme phenylalanine-4-hydroxylase; the conversion of tyrosine to L-dopa by the enzyme tyrosine hydroxylase; and conversion of tryptophan to 5-hydroxytryptophan via tryptophan hydroxylase.

Sapropterin commonly known as tetrahydrobiopterin (THB or BH4) developed by BioMarin and marketed as Sapropterin dihydrochloride under the brand name of KUVAN®. It is indicated for the treatment of phenylketonuria (PKU) and tetrahydrobiopterin deficiencies. Sapropterin dihydrochloride is chemically known as (6R)-2-amino-6-[(lR, 2S)-1, 2- dihydroxypropyl]-5,6,7,8-tetrahydro-4(lH)-pteridinone dihydrochloride and structurally represented as below.

Figure imgf000002_0001

Sapropterin dihydrochloride

Due to its vital role in the conversion of L-tyrosine into L-DOPA, which is the precursor for dopamine, a deficiency in tetrahydrobiopterin can cause severe neurological disorders unrelated to toxic build-up of L-phenylalanine; dopamine is a crucial neurotransmitter, and is the precursor of norepinephrine and epinephrine. Thus, a deficiency of tetrahydrobiopterin can result in phenylketonuria (PKU) from L-phenylalanine concentrations or hyperphenylalaninemia (HP A), as well as monoamine and nitric oxide neurotransmitter deficiency or chemical imbalance. The chronic presence of PKU can result in severe brain damage, including symptoms of mental retardation, speech impediments like stuttering, slurring, seizures or convulsions and behavioural abnormalities.

In an article published in Bio Chem J 347 (1): 1-16, tetrahydrobiopterin is reported to be biosynthesized from guanosine triphosphate (GTP) by three chemical reactions mediated by the enzymes GTP cyclohydrolase I (GTPCH), 6-pyruvoyltetrahydropterin synthase (PTPS), and sepiapterin reductase (SR).

Preparation of Sapropterin is reported with a mixture of R & S isomers in Helv. Chim. Acta, 60, 1977, 211-214, by catalytic reduction of L-biopterin of formula (2). Similar process with slight modifications is also published in Hel. Chim. Acta, 61, 1978, 2731- 2738.

Figure imgf000003_0001

(2)

In another publication reported in Helv. Chim. Acta, 62, 1979, 2577-2580, separation of the diastereomers (6R) and (6S)-5,6,7,8-tetrahydro-L-biopterin is reported by fractional crystallization of corresponding tetraacetyl derivative followed by hydrolysis using aq. HC1.

In another process published in Heterocycles, 23(12), 1985, 3115-3120, Sapropterin dihydrochloride of formula (1) is prepared by catalytic hydrogenation of L- biopterin of formula (2) in the presence of Pt02 under latm hydrogen pressure in 0.1 M potassium phosphate buffer at pH 11.8 for 18hr followed by filtration and recrystallization from 8M HC1. With slight modifications in the above reaction conditions like using platinum black, aq. base solutions like tetraethylammonium hydroxide or triethylamine etc. under 100 Kg/cm2 hydrogen pressure / 0° C / pH 12.0 / 1000 rpm / 20h/3N HCl-EtOH with 85% yield is disclosed in US4713454. In another process disclosed in US4595752, L-biopterin of formula (2) is catalytically reduced in the presence of platinum oxide in aq. base / acid solutions like (10% aq. potassium carbonate, aq. sodium carbonate, aq. potassium acetate and 0.1 N aq. HCl) under bubbling of hydrogen gas for 5-30hr at room temperature followed by filtration and isolated as HCl salt of formula (1) using aq. HCl and ethanol to obtain Sapropterin dihydrochloride.

In another approach disclosed in WO2005049614, racemic isomers of Sapropterin dihydrochloride are prepared from L-neopterin.

In another process disclosed in WO2009088979, the diacetyl biopterin is hydrolysed in the presence of aq. diethyl amine-n-butanol mixture at 40°C for 16hr at pH >11.5 followed by hydrogenation in the presence of platinum black using 50 bar hydrogen pressure at 25 °C. Product of formula (1) isolated as HCl salt from ethanol or butanol.

In another process disclosed in US20130197222, Sapropterin dihydrochloride of formula (1) is prepared starting from condensation of crotonoic acid.

The process for preparation of key intermediate, L-biopterin of formula (2) is cited in the following references.

In an article published in J. Am. Chem. Soc, 1955, 77, 3167-3168, L-biopterin of formula (2) is reported to be first isolated from human urine. The melting point reported to be 250-280°C. In another article published in J. Am. Chem. Soc, 1956, 78, 5868-5871, L-biopterin of formula (2) is prepared starting from L-rhamnose. A slight modification in the reaction conditions mentioned above is disclosed in US3505329.

In the article published in Helv. Chim. Acta, 1969, 52, 1225-1228, L-biopterin of formula (2) along with 7-biopterin is synthesized by condensing 2, 4, 5-triamino-6-oxo-l, 6-dihydropyrimidine dihydrochloride with (1 -benzyl- l-phenyl-hydrazino)-5-desoxy-L- ribulose followed by oxidation of the tetrahydro derivative.

Later in the year 1974, in an article, J. Am. Chem. Soc, 1974, 96, 6781-6782, L-biopterin is reported to be prepared starting from L-rhamnose. In another approach published in Bull. Chem. Soc. Jpn., 1975, 48(12), 3767-3768, L- biopterin of formula (2) is prepared from 2, 4, 5-triamino-6-hydroxypyrimidine dihydrochloride is reacted with hydrazone derivative in aq. methanol at reflux temperature.

In another process disclosed in US5043446 (1989), L-biopterin process is claimed to be synthesized starting from D-ribose. Similar approach with slight variations in the process, later published in Liebigs Ann. Chem., 1989, 1267-1269.

In another approach published in Agric. Biol. Chem., 1989, 53, 2095-2100, L-biopterin is synthesized starting from (S)-ethyl lactate. Prior to this publication the methodology is claimed by the same authors in JP01-221380 (1989).

In another approach disclosed in US5037981 (1990), L-biopterin is synthesized from 2- methylfuran.

In the article, Synthesis, 1992, 303-308, L-biopterin is synthesized from (4S)-4(3P- Acetoxy-5-androsten-17P-ylcarbonyloxy)-2-pentynol.

In the approach published in J. Org. Chem., 1996, 61, 8698-8700, L-biopterin is synthesized from L-tartaric acid.

In the patent US7361759 (2005), L-biopterin of formula (2) is made from L-rhamnose diethyl mercaptal.

US 20120157671 application discloses the preparation of compound of formula (4a) is by reacting D-ribose of formula (3) with acetone in the presence of sulphuric acid at room temperature followed by neutralization with sodium carbonate and concentrated under vacuum.

Sapropterin | Nature Reviews Drug Discovery

Pharmaceutics 12 00323 g004 550

https://www.mdpi.com/1999-4923/12/4/323/htm

Synthesis Reference

Steven S. Gross, “Blocking utilization of tetrahydrobiopterin to block induction of nitric oxide synthesis.” U.S. Patent US5502050, issued October, 1984.

US5502050

SYN

SYN

Synthetic Reference

Hong, Hao; Gage, James; Chen, Chaoyong; Lu, Jiangping; Zhou, Yan; Liu, Shuangyong. Method for synthesizing sapropterin dihydrochloride. Assignee Asymchem Laboratories (Tianjin) Co., Ltd., Peop. Rep. China; Asymchem Life Science (Tianjin) Co., Ltd.; Tianjin Asymchem Pharmaceutical Co., Ltd.; Asymchem Laboratories (Fuxin) Co., Ltd.; Jilin Asymchem Laboratories Co., Ltd. WO 2013152609. (2013).

syn 1

EP 0191335. Aust J Chem 1984,37(2),355-66, Chem Lett 1984,5(5),735-8

Helv Chim Acta 1979,62(8),2577-80

This compound can be prepared in two related ways: 1) The catalytic hydrogenation of biopterin (I) with H2 over PtO2 aqueous K2HPO4 at pH 11.4 or aq. (Et)4NOH at pH 12 yields a solution which is acidified with HCl. After evaporation, the residue is crystallized in ethanol – HCl. 2) The acetylation of biopterin (I) with refluxing acetic anhydride gives the triacetyl derivative (II), which is hydrogenated with H2 over PtO2 in trifluoroacetic acid, yielding the (6RS)-mixture of triacetyl derivatives (III). Acetylation of (III) with refluxing acetic anhydride affords the tetracetyl (6RS)-derivative (IV), which by fractional crystallization or column chromatography of the dihydrochloride in methanol gives the desired compound as pure (6R)-isomer.

PATENT

https://patents.google.com/patent/WO2016189542A1/en

formula 1).

Figure imgf000015_0004

The present invention is shown in below scheme- 1

Figure imgf000016_0001

Experimental Section: Example-1: Preparation of (6R)-2-amino-6-[(lR, 2S)-1, 2-dihydroxypropyl]-5,6,7,8- tetrahydro-4(lH)-pteridinone dihydrochloride of formula (1):

Step (i): Preparation of 2, 3-O-isopropylidene-D-ribose of formula (4a)

Into a 5L, 4 necked round-bottomed flask equipped with a mechanical stirrer, a thermometer socket, and a condenser, were charged acetone (3.0 L), D-ribose (300.0 gm, 2.0 mole) and p-toluene sulfonic acid (11.5 gm). The solution was stirred and maintained at 20-25°C for 2.5-3.0hrs. After completion of reaction, the reaction mixture was neutralized with aq. base solution and filtered. The filtrate was evaporated to dryness to get 375.0 gm (98.8% by theory) of 2, 3-O-isopropylidene-D-ribose of formula (4a) as light brown colour oily residue. Purity: >95% by GC. Step (ii): Preparation of l-deoxy-3, 4-O-isopropylidene-D-allitol of formula (5a)

Into a 5L, 4 necked round-bottomed flask equipped with a mechanical stirrer, a thermometer socket, and a condenser, was charged, 2.0L, 3M methyl magnesium chloride and cooled to 10° C. To this stirred solution, a solution of 200gm of 2,3-0- isopropylidene-D-ribose of formula (4a) dissolved in 200 mL tetrahydrofuran was added. After completion of reaction, the reaction mixture was quenched with ammonium chloride, extracted with ethyl acetate and separated. The solvent was evaporated to dryness under vacuum to get 185gm of l-deoxy-3, 4-O-isopropylidene-D-allitol of formula (5a) as dark brown colour oily residue. The crude product was purified by crystallization from ethyl acetate/hexane mixture to get 130g (60% by theory) as white crystalline solid. Purity: >98% by GC.

JR (λ Cm-1, KBr disc): 3317.64, 2993.69-2976.90, 2926.08, 2873.26 (m) -CH3, 1074.35; 1 HNMR (400 MHz, DMSO-d6, EDl®j&¾ : (H2¾H3, J=6.8Hz, 3H),

1.148 (s, CH3, 3H), 1.290 (s,CH3), 3.415-3.357 (m, CH, 1H), 3.652-3.571 (m, CH2, 2H), 3.812-3.803 (d, 2 X CH, 2H), 4.00-3.969 (q, CH, 1H), 4.504-4.476 (t, ΟΗ, ΙΗ), 4.504- 4.476 (d, OH, 1H), 5.381-5.371 (d, OH, 1H): 13 CNMR (100 MHz, DMSO-d6, □ (ppm): 20.59, 25.35, 27.73, 63.18, 64.61 , 69.77, 76.82, 81.40, 107.31 ; Mass: 206.42 [M], 205.41 [M-l]. DSC (° C): 77.58° C Step (iii): Preparation of 5-deoxy-2, 3-O-isopropylidene-D-ribose of formula (6a)

Into a 5L 4 necked round-bottomed flask equipped with a mechanical stirrer, a thermometer socket, and a condenser, were charged, 1.6 L of water and 270 gm of sodium meta periodate. The solution was cooled to 10-20°C. To the stirred solution, a solution of 200 gm of l-deoxy-3, 4-O-isopropylidene-D-allitol of formula (5a) dissolved in 1.4 L of isopropyl ether at 25°C. After addition, the reaction mixture was maintained at 25-30° C for l-2h. After completion of reaction, the layers were separated and the organic layer was washed with water, aq. sodium bicarbonate and separated. The excess solvent was removed by distillation under vacuum to get 145 gm (85.4% by theory) of 5- deoxy-2, 3-O-isopropylidene-D-ribose of formula (6a) as yellow oil. Purity: >98% by GC.

Step (iv & v): Preparation of 5-deoxy-L-ribose phenyl hydrazone of formula (8) a) Step (iv): Preparation of 5-deoxy-L-ribose of formula (7)

Into a 2L 4 necked round-bottomed flask equipped with a mechanical stirrer, a thermometer socket, and a condenser, were charged 600ml of water and 200gm of 5- deoxy-2, 3-O-isopropylidene-D-ribose of formula (6a). To the stirred reaction mixture, 180gm of resin was charged and stirred for 8-10 h at 10-15° C. After completion of reaction, the resin was recovered and the filtrate was clarified by activated charcoal and filtered. The filtrate was distilled off under vacuum and the resulting 5-deoxy-L-ribose of formula (7) present water was directly used in the next step without further isolation and purification. The purity of 5-deoxy-L-ribose of formula (7) present in water was above 95% by TLC.

b) Step (v): Preparation of 5-deoxy-L-ribose phenyl hydrazone of formula (8)

Into a 2L 4 necked round-bottomed flask equipped with a mechanical stirrer, a thermometer socket, and a condenser, were charged the above aq. solution of 5-deoxy-L- ribose of formula (7), 5.0 mL of acetic acid. To the stirred solution, 125g of phenyl hydrazine was charged and stirred the reaction mixture for l-2h at 25-35° C. After completion of reaction, the reaction product was filtered and washed with isopropyl ether. The wet product was dried to get 190g (73.9% by theory) of 5-deoxy-L-ribose phenyl hydrazone of formula (8) as yellow colour crystalline powder. Purity: >99.0% by HPLC. Step (VI toX): Preparation of L-erythro-biopterin of formula (2)

a) Step (vi): Preparation of triacetoxy-5-deoxy-L-ribose phenylhydrazone of formula (9)

Into a 10L 4 necked round-bottomed flask equipped with a mechanical stirrer, a thermometer socket, and a guard tube, were charged 5L of ethyl acetate, 500g of 5- deoxy-L-ribose phenyl hydrazone of formula (8) and 54gm of 4-dimethylaminopyridine. The reaction mixture was cooled to 25-30° C and was added 730gm of acetic anhydride drop wise. The reaction mixture was maintained under stirring for 2-3h. After completion of reaction, the reaction mixture was washed with water, aq. sodium carbonate and water, and separated. The organic layer was used in the next stage without further isolation and purification.

b) Step (vii): Preparation of 1,2-diacetyl-biopterin of formula (10)

Into a 20L 4 necked round-bottomed flask equipped with a mechanical stirrer, a thermometer socket, addition funnel, and a condenser, were charged, the above organic layer containing triacetoxy-5-deoxy-phenyl hydrazone of formula (9) obtained in step (vi), 3.0 L methanol and 4-hydroxy-2,5,6-triaminopyrimidine base (generated from 600 gm of corresponding sulphate salt) and salt (generated from 350 gm of tetra butyl ammonium bromide and 154g of 70% perchloric acid) and 5.3L water under stirring and heated and maintained at 35-40°C for 6-8h. The reaction mixture was then cooled to 20- 25°C and added 1.0 Kg 35% aq. hydrogen peroxide drop wise. The reaction mixture was maintained for 36-40h under stirring at 25-30°C and resulting product was filtered under suction. The wet product was washed with water and utilized in the next step without further purification.

c) Step (viii): Preparation oi -erythro biopterin of formula (2)

Into a 10L 4-necked round-bottomed flask equipped with a mechanical stirrer, condenser, thermometer socket, and addition funnel, were charged 1.35 L of aq. potassium hydroxide and the above wet product obtained from step (vii). The reaction mixture was heated to 45-50° C and maintained form 2-3h and filtered. The pH of the filtrate was adjusted to neutral and the resulting product was filtered and dried to get 205 g of crude L-erythro-biopterin of formula (2) as dark brown solid. Purity: >90% by HPLC

d) Step (ix): Preparation of potassium salt oi -erythro biopterin of formula (11a) Into a 10L 4 necked round-bottomed flask equipped with a mechanical stirrer, thermometer socket, and a glass stopper, were charged 650 mL water followed by HOg of potassium hydroxide and dissolved under stirring. The potassium hydroxide solution was cooled to 25-30° C and the above crude L-erythro-biopterin of formula (2) was charged under stirring. The resulting solution was then clarified using activated carbon and filtered. The potassium salt was regenerated from the solution by the addition of 8.5L of isopropyl alcohol. The resulting salt was filtered and washed with isopropyl alcohol. The wet product of formula (11a) was utilized in the next step without further purification.

e) Step (x): Preparation of pure L-er thro biopterin of formula (2) from potassium salt of L-erythro biopterin of formula (2)

Into a 5L 4 necked round-bottomed flask equipped with mechanical stirrer, thermometer socket, and addition funnel, were charged 3.2 L of water and the above wet potassium salt of formula (11a). The reaction mixture was stirred to dissolve completely. The resulting solution was clarified using activated carbon and filtered. The pH of the filtrate was adjusted to 6.0-7.0 to get pure L-erythro-biopterin of formula (2). The product was filtered and washed with water followed by isopropyl alcohol followed by isopropyl ether to get 130g of highly pure L-erythro biopterin of formula (2) with > 98% HPLC purity Appearance: pale brown coloured solid.

1H NMR (3N DC1) 5(ppm): 1.569-1.585(d, 3H), 4.596-4.657(p, 1H), 5.325-5.337(d, 1H), 9.355(s, 1H); Mass: 238.29(M+1), 239.22(M+2).

Step (xi): Preparation of Sapropterin dihydrochloride of formula (1)

Into a 5L 4 necked round-bottomed flask equipped with mechanical stirrer, and thermometer socket, were charged 1.8L of water, 250g of L-erythro-biopterin of formula (2) followed by 800mL of 20% aq. potassium carbonate solution under stirring. The solution was then added 90g of platinum oxide catalyst. The reaction mixture was then transferred into an autoclave and pressurized with 40 bar hydrogen gas and hydrogenated at room temperature for 24-30h under stirring. After completion of reaction, the catalyst was filtered off and the pH of the filtrate was acidified with concentrated hydrochloric acid. The water was evaporated under vacuum and the resulting crude Sapropterin dihydrochloride of formula (1) was isolated as pale yellow colour solid by addition of isopropanol/l-pentanol mixture. The product was dried in a vacuum oven to get 250g of crude Sapropterin dihydrochloride of formula (1). Step (xii): Purification of Sapropterin dihydrochloride of formula (1)

Into a 2L 4 necked round-bottomed flask equipped with a mechanical stirrer, thermometer socket, and reflux condenser, were charged 1L water and 250g of Sapropterin dihydrochloride of formula (1). The contents were stirred to dissolve completely. The clear solution was treated with activated charcoal and filtered. The filtrate was distilled off completely under vacuum to afford pale yellow solid. The product was isolated from isopropanol/l-pentanol mixture to get 225.0 g (90%) pure Sapropterin dihydrochloride of formula (1) as pale yellow to off-white solid. HPLC purity is >99.9%.

Example 2: Preparation of triacetoxy-5-deoxy-L-ribose phenylhydrazone of formula

(9)

Into a 10L 4 necked round-bottomed flask equipped with a mechanical stirrer, a thermometer socket, and a guard tube, were charged 50mL of ethyl acetate, 5.0g of 5- deoxy-L-ribose phenyl hydrazone of formula (8) and 0.54g of N, N-dimethylamino pyridine. The reaction mixture was cooled to 15-20°C and was added 7.2gm of acetic anhydride drop wise. The reaction mixture was maintained under stirring for 6-8h. After completion of reaction, the reaction mixture was washed with water, aq. sodium carbonate and water, and separated. The organic layer was distilled under reduced pressure and product was isolated from n-hexane to get 6.2g of triacetoxy-5 -deoxy-L- ribose phenylhydrazone of formula (9) 79.4% yield.

Appearance: Orange coloured solid.

Melting point: 70-75 °C.

1HNMR (CDC13): 1.275-1.29 l(d, 3H), 2.039(s, 3H), 2.085-2.095(d, 6H), 5.083-5.144(m, 1H), 5.390-5.416(t, 1H), 5.589-5.619(t, 1H), 6.849-6.886(t, 1H), 6.922-6.937(t, 1H), 6.966-6.987(d, 2H), 7.221-7.242(d, 2H), 7.563(s, 1H(D20 exchangeable).

13CNMR (CDC13): 15.325, 20.816-21.053, 68.482, 71.717, 73.043, 112.759, 120.510, 129.212, 132.105, 144.049, 169.496, 169.948. Example 3: Preparation of potassium salt of L-erythro biopterin of formula (11)

Into a 1.0L 4 necked round-bottomed flask equipped with a mechanical stirrer, thermometer socket, and a glass stopper, were charged 75 mL water followed by 3.7g of potassium hydroxide and dissolved under stirring. The potassium hydroxide solution was cooled to 25-30° C and 15.0g of crude L-erythro-biopterin of formula (2) was charged under stirring. The resulting solution was then clarified using activated carbon and filtered. The potassium salt was regenerated from the solution by the addition of 500mL of ethanol. The resulting salt was filtered and washed with ethanol and dried to get 9.1g of potassium salt of L-erythro biopterin of formula (11) with 52.3% yield. HPLC <98% Appearance: Brown coloured solid.

1H NMR (D20): 1.187-1.203(d, 3H), 4.158-4.220(p, 1H), 4.731-4.745(d, 1H), 8.623(s, 1H).

13C NMR (D20): 18.198, 70.645, 76.703, 128.811, 147.875, 149.410, 156.504, 164.774, 173.731.

Mass: 276.23(M+1), 277.21(M+2), 238.29(M-K+1); DSC (° C): 313.12°

Example 4: Preparation of Sapropterin dihydrochloride of formula (1)

Into a 5L 4 necked round-bottomed flask equipped with mechanical stirrer, and thermometer socket, were charged 1.8L of water, 250g of L-erythro-biopterin of formula (2) followed by 800ml of 20% aq. potassium hydroxide solution under stirring. The solution was then added 90gm of platinum oxide catalyst. The reaction mixture was then transferred into an autoclave and pressurized with 50 bar hydrogen gas and hydrogenated at room temperature for 24-30h under stirring. After completion of reaction, the catalyst was recovered by filtration and the filtrate was acidified with concentrated hydrochloric acid. The water was evaporated under vacuum and the resulting crude Sapropterin dihydrochloride of formula (1) was isolated as pale yellow colour solid by addition of ethanol- 1 -pentanol mixture. The product was dried in a vacuum oven to get 250g of crude Sapropterin dihydrochloride of formula (1). Example 5: Purification of Sapropterin dihydrochloride of formula (1)

Into a 2L 4 necked round-bottomed flask equipped with a mechanical stirrer, thermometer socket, and reflux condenser, were charged 1L water and 250g of Sapropterin dihydrochloride of formula (1). The contents were stirred to dissolve completely and the clear solution was treated with activated charcoal and filtered. The filtrate was distilled off completely under vacuum to afford pale yellow solid. The product 225.0 g (90%) was isolated ethanol- 1 -pentanol mixture as pure Sapropterin dihydrochloride of formula (1) as pale yellow to off-white solid. HPLC purity is >99.9%.

syn

str1 str2

str1 str2 str3

PATENT

https://patents.google.com/patent/WO2016101211A1/en

was developed by Merck and was launched in the United States and the European Union in 2007 and 2008 under the trade name Kuvan. This product can be used to treat hyperphenylalaninemia (HPA) caused by tetrahydrobiopterin (BH4) deficiency. The structure is as follows:

Figure PCTCN2014094961-appb-000001

The chemical name is: (6R)-2-amino-6-[(1R,2S)-1,2-dihydroxypropyl]-5,6,7,8-tetrahydro-4(1H)-fluorenone Dihydrochloride.

The oxaprozin hydrochloride can be obtained by hydrogenation of L-erythrobiopterin. The literature Liebigs Ann. Chem. 1989, 1267-1269 reports the preparation of L-erythrobiopterin starting from L-ribose. The preparation route is as follows:

Figure PCTCN2014094961-appb-000002

Although the method is simple and easy to perform, it is a better preparation route, but the disadvantage is that the starting material L-ribose price is higher, thus causing the cost of sapropium hydrochloride to be high.

The literature for the preparation of L-erythrobiopteris is reported by the documents Helv. Chim. Acta, 1985, 1639-1643, US2011218339A, etc. The product of the acetylation reaction of the steroid compound 6 with 2,4,5-triaminopyrimidinone Cyclization in a methanol/water/pyridine system followed by aromatization with an iodine reagent to give an acetylated L- Red-type biopterin, followed by hydrolysis and deacetylation to obtain L-erythrobiopterin. The reaction equation is as follows:

Figure PCTCN2014094961-appb-000003

Among them, compound 6 is used as a key intermediate, and many methods for its preparation are reported. The method reported in J. Am. Chem. Soc. 1974, 6781-6782, J. Am. Chem. Soc. 1976, 2301-2307, etc., uses L-rhamnose as a raw material, and reacts with ethanethiol to form a corresponding shrinkage. Sulfuraldehyde, oxidizing thiol to sulfone with an oxidizing agent, removing a carbon under alkaline conditions to obtain 5-deoxy-L-arabinose, and reacting 5-deoxy-L-arabinose with phenylhydrazine to obtain a key intermediate formula 6 . The synthetic route is as follows:

Figure PCTCN2014094961-appb-000004

Although this method has been improved and improved many times, the ethanethiol used has a special malodor and requires the use of a deodorizing device, and its lower boiling point also causes inconvenience to the production.

Document J. Org. Chem. 1996, 8699-8700 reports that L-tartaric acid is used as a starting material, which is protected by hydroxyl group, carboxyl group, reduction, addition, deprotection to obtain 5-deoxy-L-ribose, 5-deoxy- The condensation of L-arabinose with phenylhydrazine gives key intermediates. The synthetic route is as follows:

Figure PCTCN2014094961-appb-000005

The reducing agent used in the route of the acid chloride to reduce the aldehyde is bis(triphenylphosphine) copper borohydride (I), which has a high price and is not favorable for the control of industrialization cost. The reaction temperature of the format reagent with carbonyl addition and lactone reduction is -78 ° C, and the energy consumption in industrial production is high. In addition, the post-treatment of the multi-step reaction uses silica gel column color The spectrum is purified and it is difficult to achieve industrialization. Therefore, this route has great disadvantages in terms of cost and operability in industrial production.

Document CN201010151443.2 reports the use of L-arabinose as a starting material to obtain L-erythrobioptery through a multi-step reaction. The preparation route is as follows:

Figure PCTCN2014094961-appb-000006

In reproducing the preparation method, we have found that the intermediate 2 is directly subjected to reduction and desulfonation reaction to prepare the intermediate 2, which has the disadvantages of low yield, low product purity, and difficulty in purification of the product. Therefore, it is necessary to find a simple, feasible and low-cost preparation route.

 scheme synthetic route includes the following steps:

Figure PCTCN2014094961-appb-000012

Example 1: Preparation of Product 1

To the reaction flask was added 10 L of anhydrous methanol, and 1.5 kg of the starting material L-arabinose was added under mechanical stirring. 250 g of concentrated sulfuric acid was added dropwise under a water bath, and the reaction was stirred for 20-24 hours. The reaction was monitored by TLC, and 350 g of sodium carbonate was added to the reaction system. Stir until pH = 7-8 and filter. The filtrate was concentrated under reduced pressure at 35 ° C to 40 ° C to dryness to yield 1.64 kg of oil, yield -100%.

Example 2: Preparation of product 2

The product 1, 4 L of pyridine and 5 L of acetonitrile were added to the reaction flask and dissolved by mechanical stirring. The mixture was cooled by stirring, and a solution of 1.8 kg of p-toluenesulfonyl 5 L acetonitrile was added dropwise at a temperature of 0 to 5 ° C. After completion of the dropwise addition, the reaction was stirred at room temperature 20-25 ° C for 4 hours. The TLC monitors the reaction.

After concentration, 12 L of ethyl acetate and 5 L of water were added to the concentrated residue, and the layers were stirred. The organic layer was washed with 1 mol/L hydrochloric acid, saturated sodium hydrogen carbonate and saturated brine and dried. Filtration and concentration of the filtrate gave 1.7 kg of pale yellow oil, yield 56.3%.

Example 3: Preparation of product 3

1.2 kg of product 2 was added to a 10 L reaction flask, dissolved with 6 L of methyl ethyl ketone, and 840 g of sodium iodide was added with stirring. After the addition, the temperature was refluxed for 12 hours, and the reaction was completed by TLC. The mixture was cooled to room temperature, filtered, and the filtrate was evaporated. It was dissolved in ethyl acetate, washed with water, and the aqueous layer was evaporated. The combined organic layers were washed with EtOAc EtOAc m.

Example 4: Preparation of product 4

To a 20 L reaction flask was added 900 g of product 3, 332 g of triethylamine dissolved in 9 L of methanol, 45 g of 10% Pd/C, vacuumed, hydrogenated twice, and hydrogenated at a constant temperature of 25-30 ° C for 16 hours. The reaction was completed by TLC, filtered, and the filtrate was concentrated under reduced pressure to give a residue. 4 L of ethyl acetate was added to the residue to precipitate a white solid. The mixture was stirred at 0 ° C for 30 min, and filtered. The filtrate was added to 2 L of a 0.4 mol/L sulfuric acid solution and the layers were separated. The aqueous layer was washed once with 50 mL of ethyl acetate to give an aqueous solution of product 4 (approximately 250 g).

Example 5: Preparation of product 5

The aqueous solution of product 4 was added to the reaction flask, and the reaction was heated at 75 ° C for 3 hours, and the reaction was completed by TLC (DCM: MeOH = 10:1). After cooling to room temperature, it was washed with 100 mL of ethyl acetate, and the aqueous layer was separated to give the product 5, i.e., about 213 g of aqueous solution of 5-deoxy-L-arabinose, which was directly reacted in the next step.

Example 6: Preparation of product 6

To the reaction flask, 2.5 L of ethyl acetate and 170 g of phenylhydrazine were added under nitrogen, and an aqueous solution of the product 5 was added dropwise with stirring at a temperature of 5 to 10 ° C (protected from light). The reaction was kept for 1 hour, and then the temperature was raised to 20-25 ° C for 30 min. The reaction was completed by TLC and the layers were separated. The aqueous layer was extracted with ethyl acetate and organic layers were combined. The organic layer was dried over anhydrous sodium sulfate and filtered.

The ethyl acetate solution of product 6 was added to the reaction flask under nitrogen, and 8 L of petroleum ether was slowly added with stirring. After the addition was completed, the mixture was cooled to -5 – 10 ° C and stirred for 1 hour, and filtered to give a beige solid. Drying under reduced pressure at 30-35 ° C gave a dry product of about 250 g, yield 71.4%.

Example 7: Preparation of product 7

To the reaction flask was added 2.5 L of ethyl acetate and 250 g of product 6. 30 g of DMAP was added with stirring. 400 ml of acetic anhydride was added dropwise at a temperature of 15 ° C, and the reaction was stirred at a temperature of 20-25 ° C for 3 hours. The reaction was monitored by TLC, and a hydrochloric acid solution was added at a temperature of 15 ° C to separate the layers. The organic layer was washed with saturated hydrochloric acid and saturated sodium hydrogen sulfate. The organic phase was separated, dried and filtered to give 371 g, m.

Example 8: Preparation of product 9

To the reaction flask was added 220 g of product 8, 2.2 L of purified water. Under stirring, 500 g of a product 7 in 5 L of methanol and 150 g of anhydrous lithium perchlorate dissolved in 1.5 L of water were added. After the addition was completed, the reaction was stirred at a temperature of 30 to 32 ° C for 20 hours. The reaction is completed and filtered. The filtrate was temperature-controlled at 15 ° C to 20 ° C, and 1 L of 30% hydrogen peroxide was added dropwise. After the addition, the reaction was kept at 20 ° C for 6 hours, and the solid was precipitated, filtered, and dried by blasting at 35-40 ° C to obtain 215 g of a brownish yellow product 9 in a yield of 47%.

Example 9: Preparation of product 10

To the reaction flask, 80 g of product 9, 400 ml of purified water, 300 ml of n-butanol, and 80 ml of diethylamine were added, and the mixture was stirred and heated to 45-50 ° C for 16 hours. After the TLC reaction is completed, the layers are separated, and the aqueous layer is separated to obtain an aqueous solution of the product 10, which is directly reacted in the next step.

Example 10: Preparation of Product I

An aqueous solution of product 10 was added to the autoclave, and 50 ml of triethylamine and 2 g of platinum dioxide were added thereto with stirring. The pressure was evacuated, the hydrogen was replaced three times, the pressure was controlled to 1.5 MPa, and the reaction was stirred at 35 ° C for 20 hours. After filtration, the filtrate was added to 30 ml of n-butanol for 5 min, and the mixture was allowed to stand to give an aqueous solution of product I. 200 ml of concentrated hydrochloric acid was added dropwise at a temperature of 10 ° C, and the aqueous solution was concentrated under reduced pressure to dryness. 500 ml of 95% ethanol was added to the crude product, and the mixture was heated to 55-60 ° C for 1 hour, then cooled to 35 ° C for 2 hours, filtered, and the filter cake was dried to give the product I35 g.

Example 11: Preparation of product 9′

To the reaction flask was added 1.25 L of ethyl acetate and 125 g of product 9. 15 g of DMAP was added with stirring. 200 ml of acetic anhydride was added dropwise at a temperature of 15 ° C, and the reaction was stirred at a temperature of 20-25 ° C for 3 hours. The reaction was monitored by TLC, and a hydrochloric acid solution was added at a temperature of 15 ° C to separate the layers. The organic layer was washed with saturated hydrochloric acid and saturated sodium hydrogen sulfate. The organic phase was separated, dried and concentrated to give 12,5 g of oil.

Example 12: Preparation of product 10

The product 9′ prepared in Example 11 was added to the reaction flask, 600 ml of purified water, 450 ml of n-butanol, and 120 ml of diethylamine were added, and the mixture was stirred and heated to 45-50 ° C for 16 hours. After the TLC reaction is completed, the layers are separated, and the aqueous layer is separated to obtain an aqueous solution of the product 10, which is directly reacted in the next step.

Example 13: Preparation of Product I

An aqueous solution of the product 10 prepared in Example 12 was added to the hydrogenation vessel, and 80 ml of triethylamine, 3 g of platinum dioxide was added thereto with stirring, and vacuum was applied thereto, and the pressure was controlled to 1.5 MPa, and the reaction was stirred at 35 ° C for 20 hours. After filtration, the filtrate was added to 45 ml of n-butanol for 5 min, and the mixture was allowed to stand to give an aqueous solution of product I. After cooling at 10 ° C, 300 ml of concentrated hydrochloric acid was added dropwise, and the aqueous solution was concentrated under reduced pressure to dryness. 750 ml of 95% ethanol was added to the crude product, and the mixture was heated to 55-60 ° C for 1 hour, then cooled to 35 ° C for 2 hours, filtered, and the filter cake was dried to give the product I 48.9 g.

///////////

https://patents.google.com/patent/US9365573B2/en

Sapropterin dihydrochloride, chemical name (6R)-2-amino-6-[(1R,2S)-1,2-dihydroxypropyl]-5,6,7,8-tetrahydro-4(1H)-pteridinone dihydrochloride, molecular formula C9H15N5O3.2HCl, and CAS registry number 69056-38-28, is a synthetic product of tetrahydrobiopterin (BH4) dihydrochloride. BHis a cofactor of Phenylalanine Hydroxylase (PAH). Tyrosine is acquired from Phenylalanine (Phe) through hydroxylation under the action of PAH which is low in activity or even inactive in PKU patients, while BHis able to activate PAH, promote normal oxidative metabolism of Phe in the bodies of the patients, and reduce the Phe levels in the bodies of some patients. On Dec. 16, 2007, the sapropterin dihydrochloride tablets produced by BioMarin Pharmaceutical Inc. in USA were approved by the Food and Drug Administration (FDA) for marketing for treatment of PKU. Because of the effective activity of sapropterin dihydrochloride, it is extremely necessary to select a route applicable to industrial production with high product purity.

At present, BHis mainly synthesized by the following methods reported in literatures:

1. Preparation using 4-hydroxy-2,5,6-triaminopyrimidine (TAP) and 5-deoxy-L-arabinose as raw materials, please see literature E. L. Patterson et al., J. Am. Chem. Soc. 78, 5868(1956).

2. Preparation using TAP and 5-deoxy-L-arabinose phenylhydrazone as raw materials, please see literature Matsuura et al., Bull. Chem. Soc. Jpn., 48,3767 (1975);

3. Preparation by reaction of raw materials hydroxyl-protected TAP and 4-acetyl-2,3-epoxypentanal through oxidation of iodine and a dehydroxylation protecting group, please see literature Matsuura et al., Chemistry of Organic Synthesis, MI/g. 46. No. 6, P570(1988).

These traditional methods for preparing BH4 have the following major disadvantages: raw materials are expensive, arabinose which can be hardly acquired is used as a carbon atom radical for asymmetric synthesis; there are multiple steps in reactions with low yield, and low product purity, 5-deoxy-L-arabinose is easily degraded in a reaction solution, and products of the synthesis routes above have low stereoselectivity. To sum up, the traditional synthesis methods are not applicable to mass industrial production. Therefore, a synthesis route, which is applicable to industrial production with high product purity, high yield and high stereoselectivity, needs to be searched urgently.

tep 10: add 0.7 kg (0.05 g/g) of palladium 5% on carbon in the presence of the methanol solution containing 1.5 kg of acetylamino-7,8-dihydropteridine

Figure US09365573-20160614-C00106


obtained in Step 9, introduce hydrogen until the pressure of the reaction kettle is 0.8±0.05 MPa, control the temperature of the system at 25±5° C. and the pressure at 0.8±0.05 MPa, react for 82 hours, after reacting thoroughly, perform quenching in 31.9 kg (9 eq) of dilute hydrochloric acid having a concentration of 15%, and perform suction filtration and drying to the system to obtain a target product, i.e. a crude product of sapropterin dihydrochloride

Figure US09365573-20160614-C00107


recrystallize and purify the crude product by 29 L (20 ml/g) of methanol at 35±5° C. to obtain 0.8 kg of a pure product, with a yield of 45%, a purity of 98.3% and an enantiomeric excess of 99.1%.

Embodiment 5: main raw material:

Figure US09365573-20160614-C00108


and X═O

Step 1: add 836 kg (0.3 eq) of a tetrahydrofuran solution contaning a samarium catalyst having a concentration of 4%, 29.2 kg (0.3 eq) of (R)-(+)-1,1′-bi-2-naphthol, 28.4 kg (0.3 eq) of triphenylphosphine oxide, and 600 kg (10 kg/kg) of a 4 A molecular sieve to a 3000 L reaction kettle, after stirring uniformly, control the system temperature at 20±5° C., add 117.4 kg (2 eq) of meta-chloroperoxybenzoic acid, add 60 kg (1 eq) of benzyl crotonate

Figure US09365573-20160614-C00109


to the system after adding meta-chloroperoxybenzoic acid, react for 32 hours while preserving the temperature, add 19.6 kg (0.3 eq) of citric acid to the system to stop the reaction, and perform centrifugation, concentration and rectification to the system to obtain 40.5 kg of (2S,3R)-2,3-epoxy-benzyl butyrate

Figure US09365573-20160614-C00110


with a yield of 62%;

Step 2: add 36.8 kg (3 eq) of acetone, and 5.4 kg (0.6 eq) of lithium chloride to a 500 L enamel vessel, control the temperature at 15±5° C., add 40.5 kg (1 eq) of (2S,3R)-2,3-epoxy-benzyl butyrate

Figure US09365573-20160614-C00111


react for 7 hours while preserving the temperature, add 422 kg (2 eq) of a potassium bicarbonate aqueous solution having a concentration of 10%, and perform liquid separation, extraction and concentration to the system to obtain 44 kg of (4S,5S)-2,2,5-trimethyl-acetonide-benzyl butyrate

Figure US09365573-20160614-C00112


with a yield of 82%;

Step 3: add 352 L (8 ml/g) of ethanol, and 44 kg (1 eq) of (4S,5S)-2,2,5-trimethyl-2,3-acetonide-benzyl butyrate

Figure US09365573-20160614-C00113


to a 1000 L reaction kettle, increase the temperature to 37±5° C., add 4.8 kg (1.5 eq) of pure water and 53.2 kg (1.5 eq) of a sodium hydroxide aqueous solution having a concentration of 20%, react for 6 hours while preserving the temperature, perform centrifugation, dissolve a filter cake in 352 L (8 ml/g) of ethanol, add 71.0 kg (3 eq) of L-α-amphetamine, preserve the temperature at 22±5° C. for 4 hours, and perform centrifugation and drying to obtain 32.4 kg of (4S,5S)-2,2,5-trimethyl-2,3-acetonide-phenylacetylamino butyrate

Figure US09365573-20160614-C00114


with a yield of 62%;

Step 4: add 48 L (6 ml/g) of 1,4-dioxane, 8 kg (1 eq) of (4S,5S)-2,2,5-trimethyl-2,3-acetonide-phenylacetylamino butyrate

Figure US09365573-20160614-C00115


to a 72 L reaction bottle, then add a dilute sulphuric acid aqueous solution having a concentration of 10% to the system to regulate the pH at 2.5±0.5, control the temperature at −5±5° C., react for 1 hour, perform liquid separation to obtain an organic phase, add 7.0 kg of (2.0 eq) N,N-diisopropylethylamine to the organic phase, and concentrate the system to obtain 4.1 kg of (4S,5S)-2,2,5-trimethyl-1,3-dioxolan-4-methanoic acid

Figure US09365573-20160614-C00116


with a yield of 93.5%;

Step 5: add 49 L (12 ml/g) of 2-methyltetrahydrofuran, 4.1 kg of 1,3-dioxolan-4-methanoic acid

Figure US09365573-20160614-C00117


and 13.1 kg (4 eq) of N,N-diisopropylethylamine to a 100 L reaction bottle, reduce the temperature to −22±5° C., add 5.5 kg (2.0 eq) of ethyl chloroformate, react for 1.8 hours while preserving the temperature, introduce a diazomethane gas for 1.8 hours, add 18.5 kg (4.5 eq) of a hydrochloride ethanol solution having a concentration of 20%, react for 1.8 hours, add potassium bicarbonate to regulate the pH value to 8.5±0.5, and perform extraction, liquid separation and concentration to obtain 4.1 kg of (4S,5S)-2,2,5-trimethyl-5-chloroacetyl-1,3-dioxolane

Figure US09365573-20160614-C00118


with a yield of 83.7%;

Step 6: add 49 L (12 ml/g) of acetone, 4.1 kg of (4S,5S)-2,2,5-trimethyl-5-chloroacetyl-1,3-dioxolane

Figure US09365573-20160614-C00119


3.4 kg (2.5 eq) of sodium azide, and 1.8 kg (0.5 eq) of potassium iodide to a 72 L bottle, react the system for 26 hours while preserving the temperature at 34±5° C., perform filtering and concentration to obtain an acetone solution containing 3.9 kg of (4S,5S)-2,2,5-trimethyl-5-(2-azidoacetyl)-1,3-dioxolane

Figure US09365573-20160614-C00120


with a yield of 91.5%;

Step 7: add 46.4 L (12 ml/g) of methyl tert-butyl ether and 1.2 kg (0.3 g/g) of Raney nickel to a 100 L reaction kettle, introduce hydrogen until the system pressure is 0.8±0.1 MPa, regulate the pH of the system to 3±0.5 with concentrated sulfuric acid, add an acetonitrile solution containing 3.9 kg (1 eq) of (4S,5S)-2,2,5-trimethyl-5-(2-azidoacetyl)-1,3-dioxolane

Figure US09365573-20160614-C00121


react at 27±5° C. for 8.5 hours, perform suction filtration and concentration to obtain 2.3 kg of (3S,4S)-1-amino-3,4-dihydroxy-2-pentanone

Figure US09365573-20160614-C00122


with a yield of 89%;

Step 8: add 23 L (10 ml/g) of propanol, 6.9 L (3 ml/g) of pure water, 0.9 kg of (0.3 eq) of potassium iodide, 4.8 kg (1.2 eq) of compound A (2-amino-6-chloro-5-nitro-3H-pyrimidin-4-one), 2.3 kg (1 eq) of (3S,4S)-1-amino-3,4-dihydroxy-2-pentanone

Figure US09365573-20160614-C00123


and 10.5 kg (6 eq) of diisopropylamine to a 50 L reaction bottle, react the system for 7 hours while preserving the temperature at 72±5° C., then add a potassium dihydrogen phosphate-dipotassium phosphate aqueous solution to regulate the pH of the system to 7.5±0.5; and filter the system to obtain 2.5 kg of 2-acetylamino-5-nitro-6-((3S,4S)-3,3-dihydroxy-2-oxo-pentylamino)-pyrimidin-4-one

Figure US09365573-20160614-C00124


with a yield of 44%;

Step 9: add 1.25 kg (1 eq) of 2-acetylamino-5-nitro-6((3S,4S)-3,3-dihydroxy-2-oxo-pentylamino)-pyrimidin-4-one

Figure US09365573-20160614-C00125


50 L (40 ml/g) of ethanol and 0.5 kg (0.4 g/g) of 10% palladium on carbon to a 100 L autoclave, introduce hydrogen until the reaction system pressure is 0.8±0.05 MPa, control the temperature of the system at 27±5° C. and the pressure at 0.8±0.05 MPa, react for 24 hours, filter the system, and regulate the pH to 11±0.5 to obtain an ethanol solution containing 1.1 kg of acetylamino-7,8-dihydropteridine

Figure US09365573-20160614-C00126


which is used directly in the next step;

Step 10: add 0.44 kg (0.4 g/g) of palladium 10% on carbon in the presence of the ethanol solution containing 1.1 kg of acetylamino-7,8-dihydropteridine

Figure US09365573-20160614-C00127


obtained in Step 9, introduce hydrogen until the pressure of the reaction kettle is 0.8±0.05 MPa, control the temperature of the system at 25±5° C. and the pressure at 0.8±0.05 MPa, react for 80 hours, after reacting thoroughly, perform quenching in 20 kg (8 eq) of dilute hydrochloric acid having a concentration of 15%, and perform suction filtration and drying to the system to obtain a target product, i.e. a crude product of sapropterin dihydrochloride

Figure US09365573-20160614-C00128


recrystallize and purify the crude product by 21.4 L (20 ml/g) of ethanol at 35±5° C. to obtain 0.4 kg of a pure product, with a yield of 46.2%, a purity of 98.5% and an enantiomeric excess of 99.2%.

Embodiment 6: main raw material:

Figure US09365573-20160614-C00129


and X═N

Step 1: add 522 kg (0.05 eq) of a tetrahydrofuran solution containing a samarium catalyst having a concentration of 2%, 9.1 kg (0.05 eq) of (R)-(+)-1,1′-bi-2-naphthol, 8.9 kg (0.05 eq) of triphenylphosphine oxide, and 567 kg (7 kg/kg) of a 4 A molecular sieve to a 3000 L reaction kettle, after stirring uniformly, control the system temperature at 8±5° C., add 57.4 kg (0.eq) of a tert-butyl hydroperoxide toluene solution having a concentration of 50%, add 81.1 kg (1 eq) of (E)-N-isopropylbut-2-enamide

Figure US09365573-20160614-C00130


to the system after adding the tert-butyl hydroperoxide toluene solution, react for 34 hours while preserving the temperature, add 6.1 kg (0.05 eq) of citric acid to the system to stop the reaction, and perform centrifugation, concentration and rectification to the system to obtain 56.1 kg of (2S,3R)-2,3-epoxy-diisopropylamido butyrate

Figure US09365573-20160614-C00131


with a yield of 61.5%;

Step 2: add 11.4 kg (0.5 eq) of acetone, and 8.8 kg (0.1 eq) of zinc bromide to a 500 L enamel vessel, control the temperature at 20±5° C., add 56.1 kg (1 eq) of (2S,3R)-2,3-epoxy-diisopropylamido butyrate

Figure US09365573-20160614-C00132


react for 8.5 hours while preserving the temperature, add 329 kg (2 eq) of a sodium bicarbonate aqueous solution having a concentration of 10%, and perform liquid separation, extraction and concentration to the system to obtain 64.7 kg of (4S,5S)-2,2,5-trimethyl-2,3-acetonide-diisopropylamido butyrate

Figure US09365573-20160614-C00133


with a yield of 82%;

Step 3: add 259 L (4 ml/g) of tetrahydrofuran, and 64.7 kg (1 eq) of (4S,5S)-2,2,5-trimethyl-2,3-acetonide-diisopropylamido butyrate

Figure US09365573-20160614-C00134


to a 1000 L reaction kettle, increase the temperature to 27±5° C., add 2.9 kg (0.5 eq) of pure water and 29.9 kg (0.5 eq) of a methanol solution of sodium methoxide having a concentration of 29.9%, react for 4 hours while preserving the temperature, perform centrifugation, dissolve a filter cake in 194 L (3 ml/g) of tetrahydrofuran, add 39 kg (1 eq) of L-α-phenylethylamine, preserve the temperature at 18±5° C. for 3.5 hours, and perform centrifugation and drying to obtain 54.3 kg of 1-phenyltehanamine (4S,5S)-2,2,5-trimethyl-1,3-dioxolane-4-carboxylate

Figure US09365573-20160614-C00135


with a yield of 60%;

Step 4: add 30 L (3 ml/g) of 2-methyltetrahydrofuran, 10 kg (1 eq) of 1-phenyltehanamine (4S,5S)-2,2,5-trimethyl-1,3-dioxolane-4-carboxylate

Figure US09365573-20160614-C00136


to a 72 L reaction bottle, then add a dilute phosphoric acid aqueous solution having a concentration of 10% to the system to regulate the pH at 1.5±0.5, control the temperature at −5±5° C., react for 1 hour, perform liquid separation to obtain an organic phase, add 3.7 kg of (0.8 eq) N,N-diisopropylethylamine to the organic phase, and concentrate the system to obtain 5.3 kg of (4S,5S)-2,2,5-trimethyl-1,3-dioxolan-4-methanoic acid

Figure US09365573-20160614-C00137


with a yield of 92.5%;

Step 5: add 42 L (8 ml/g) of 1,4-dioxane, 5.3 kg of 1,3-dioxolan-4-methanoic acid

Figure US09365573-20160614-C00138


and 8.5 kg (2 eq) of N,N-diisopropylethylamine to a 100 L reaction bottle, reduce the temperature to −10±5° C., add 4 kg (21.0 eq) of propyl chloroformate, react for 2 hours while preserving the temperature, introduce a diazomethane gas for 2 hours, add 12 kg (2 eq) of a hydrochloride ethanol solution having a concentration of 20%, react for 2 hours, add sodium hydroxide to regulate the pH value to 7.5±0.5, and perform extraction, liquid separation and concentration to obtain 5.1 kg of (4S,5S)-2,2,5-trimethyl-5-chloroacetyl-1,3-dioxolane

Figure US09365573-20160614-C00139


with a yield of 81%; Step 6: add 41 L (8 ml/g) of tetrahydrofuran, 5.1 kg of (4S,5S)-2,2,5-trimethyl-5-chloroacetyl-1,3-dioxolane

Figure US09365573-20160614-C00140


3.1 kg (1 eq) of azidotrimethylsilane, and 0.5 kg (0.1 eq) of sodium iodide to a 72 L bottle, react the system for 30 hours while preserving the temperature at 12±5° C., perform filtering and concentration to obtain an acetone solution containing 4.6 kg of (4S,5S)-2,2,5-trimethyl-5-(2-azidoacetyl)-1,3-dioxolane

Figure US09365573-20160614-C00141


with a yield of 87.5%;

Step 7: add 28 L (6 ml/g) of 1,4-dioxane and 0.23 kg (0.05 g/g) of palladium 10% on carbon to a 50 L reaction kettle, introduce hydrogen until the system pressure is 0.8±0.1 MPa, regulate the pH of the system to 3±0.5 with acetic acid, add an acetonitrile solution containing 4.6 kg (1 eq) of (4S,5S)-2,2,5-trimethyl-5-(2-azidoacetyl)-1,3-dioxolane

Figure US09365573-20160614-C00142


react at 27±5° C. for 8.5 hours, react for 8.5 hours, perform suction filtration and concentration to obtain 2.7 kg of (3S,4S)-1-amino-3,4-dihydroxy-2-pentanone

Figure US09365573-20160614-C00143


with a yield of 87.7%;

Step 8: add 16.3 L (6 ml/g) of isopropanol, 2.7 L (1 g/g) of pure water, 0.4 kg of (0.1 eq) of sodium iodide, 4.8 kg (1.0 eq) of compound A (2-amino-6-chloro-5-nitro-3H-pyrimidin-4-one), 2.7 kg (1 eq) of (3S,4S)-1-amino-3,4-dihydroxy-2-pentanone

Figure US09365573-20160614-C00144


and 8.7 kg (4 eq) of sodium carbonate to a 50 L reaction bottle, react the system for 7 hours while preserving the temperature at 45±5° C., then add an ammonium formate-ammonia aqueous solution to regulate the pH of the system to 6.5±0.5; and filter the system to obtain 2.85 kg of 2-acetylamino-5-nitro-6((3S,4S)-3,3-dihydroxy-2-oxo-pentylamino)-pyrimidin-4-one

Figure US09365573-20160614-C00145


with a yield of 42.5%;

Step 9: add 2 kg (1 eq) of 2-acetylamino-5-nitro-6-((3S,4S)-3,3-dihydroxy-2-oxo-pentylamino)-pyrimidin-4-one

Figure US09365573-20160614-C00146


60 L (30 ml/g) of ethanol and 0.2 kg (0.1 g/g) of platinum dioxide to a 100 L autoclave, introduce hydrogen until the reaction system pressure is 0.6±0.05 MPa, control the temperature of the system at 20±5° C. and the pressure at 0.6±0.05 MPa, react for 20 hours, filter the system, and regulate the pH to 11±0.5 to obtain an ethanol solution containing 1.7 kg of acetylamino-7,8-dihydropteridine

Figure US09365573-20160614-C00147


which is used directly in the next step;

Step 10: add 0.2 kg (0.1 g/g) of platinum dioxide in the presence of the ethanol solution containing 1.7 kg of acetylamino-7,8-dihydropteridine

Figure US09365573-20160614-C00148


obtained in Step 9, introduce hydrogen until the pressure of the reaction kettle is 0.6±0.05 MPa, control the temperature of the system at 15±5° C. and the pressure at 0.6±0.05 MPa, react for 75 hours, after reacting thoroughly, perform quenching in 30 kg (5 eq) of dilute hydrochloric acid having a concentration of 10%, and perform suction filtration and drying to the system to obtain a target product, i.e. a crude product of sapropterin dihydrochloride

Figure US09365573-20160614-C00149


recrystallize and purify the crude product by 17 L (10 ml/g) of butanone at 15±5° C. to obtain 0.6 kg of a pure product, with a yield of 43%, a purity of 98.4% and an enantiomeric excess of 98.9%.

Thus, it can be seen that synthesis of a sapropterin dihydrochloride compound and an intermediate thereof disclosed in a method of the present disclosure can obtain a target product with a high purity, a high enantiomeric excess, and a high yield. The synthesis method uses readily-available raw materials, significantly reduces a synthesis route of sapropterin dihydrochloride. The technological conditions are stable, and there is less pollution in the whole operation process, hence providing an effective scheme for mass industrial production of sapropterin dihydrochloride.

The above are only preferred embodiments of the present disclosure and should not be used to limit the present disclosure. For those skilled in the art, the present disclosure may have various modifications and changes. Any modifications, equivalent replacements, improvements and the like within the spirit and principle of the present disclosure shall fall within the scope of protection of the present disclosure.

///////

USA
Patent No. Patent Type Assignee Patent Expiry
(Pediatric exclusivity)
Estimated Expiry Status
US 4,713,454 Process Shiratori Pharmaceutical Co., Ltd. (Narashino, JP) Suntory Limited (Osaka, JP) NA 23-JAN-06 Expired

References

  1. Jump up to:a b “Sapropterin (Kuvan) Use During Pregnancy”Drugs.com. 17 May 2019. Retrieved 4 March 2020.
  2. ^ “Sapropterin”Drugs.com. 28 February 2020. Retrieved 4 March 2020.
  3. ^ “International Non-proprietary Names for Pharmaceutical Substances (INN)”Fimea. Retrieved 4 March 2020.
  4. Jump up to:a b Kappock TJ, Caradonna JP (November 1996). “Pterin-Dependent Amino Acid Hydroxylases”. Chemical Reviews96 (7): 2659–2756. doi:10.1021/CR9402034PMID 11848840.
  5. ^ Całka J (2006). “The role of nitric oxide in the hypothalamic control of LHRH and oxytocin release, sexual behavior and aging of the LHRH and oxytocin neurons”Folia Histochemica et Cytobiologica44 (1): 3–12. PMID 16584085.
  6. ^ Bhagavan, N. V. (2015). Essentials of Medical Biochemistry With Clinical Cases, 2nd Edition. USA: Elsevier. p. 256. ISBN 978-0-12-416687-5.
  7. ^ Schaub J, Däumling S, Curtius HC, Niederwieser A, Bartholomé K, Viscontini M, et al. (August 1978). “Tetrahydrobiopterin therapy of atypical phenylketonuria due to defective dihydrobiopterin biosynthesis”Archives of Disease in Childhood53 (8): 674–6. doi:10.1136/adc.53.8.674PMC 1545051PMID 708106.
  8. Jump up to:a b “Kuvan- sapropterin dihydrochloride tablet Kuvan- sapropterin dihydrochloride powder, for solution Kuvan- sapropterin dihydrochloride powder, for solution”DailyMed. 13 December 2019. Retrieved 4 March 2020.
  9. Jump up to:a b c “Kuvan EPAR”European Medicines Agency (EMA). 4 March 2020. Retrieved 4 March 2020.
  10. ^ “Drug Approval Package: Kuvan (Sapropterin Dihydrochloride) NDA #022181”U.S. Food and Drug Administration (FDA). 24 March 2008. Retrieved 4 March 2020Lay summary (PDF).
  11. Jump up to:a b c d e “Kuvan (sapropterin dihydrochloride) Tablets and Powder for Oral Solution for PKU”BioMarin. Retrieved 4 March 2020.
  12. ^ “Drug Approval Package: Kuvan Powder for Oral Solution (Sapropterin Dihydrochloride) NDA #205065”U.S. Food and Drug Administration (FDA). 28 February 2014. Retrieved 4 March 2020Lay summary (PDF).
  13. ^ Herper M (28 July 2016). “How Focusing On Obscure Diseases Made BioMarin A $15 Billion Company”Forbes. Retrieved 9 October 2017.
  14. ^ “BioMarin Announces Kuvan (sapropterin dihydrochloride) Patent Challenge Settlement”. BioMarin Pharmaceutical Inc. 13 April 2017. Retrieved 9 October 2017 – via PR Newswire.
  15. ^ “Tetrahydrobiopterin Deficiency”National Organization for Rare Disorders (NORD). Retrieved 9 October 2017.
  16. ^ “What are common treatments for phenylketonuria (PKU)?”NICHD. 23 August 2013. Retrieved 12 September 2016.
  17. ^ Camp KM, Parisi MA, Acosta PB, Berry GT, Bilder DA, Blau N, et al. (June 2014). “Phenylketonuria Scientific Review Conference: state of the science and future research needs”Molecular Genetics and Metabolism112 (2): 87–122. doi:10.1016/j.ymgme.2014.02.013PMID 24667081.
  18. Jump up to:a b Haberfeld, H, ed. (1 March 2017). Austria-Codex (in German). Vienna: Österreichischer Apothekerverlag. Kuvan 100 mg-Tabletten.
  19. ^ “Genetics Home Reference: GCH1”National Institutes of Health.
  20. ^ Wu G, Meininger CJ (2009). “Nitric oxide and vascular insulin resistance”. BioFactors35(1): 21–7. doi:10.1002/biof.3PMID 19319842S2CID 29828656.
  21. ^ Kaufman S (February 1958). “A new cofactor required for the enzymatic conversion of phenylalanine to tyrosine”The Journal of Biological Chemistry230 (2): 931–9. PMID 13525410.
  22. ^ Thöny B, Auerbach G, Blau N (April 2000). “Tetrahydrobiopterin biosynthesis, regeneration and functions”The Biochemical Journal. 347 Pt 1: 1–16. doi:10.1042/0264-6021:3470001PMC 1220924PMID 10727395.
  23. ^ Kuzkaya N, Weissmann N, Harrison DG, Dikalov S (June 2003). “Interactions of peroxynitrite, tetrahydrobiopterin, ascorbic acid, and thiols: implications for uncoupling endothelial nitric-oxide synthase”The Journal of Biological Chemistry278 (25): 22546–54. doi:10.1074/jbc.M302227200PMID 12692136.
  24. ^ Muller-Delp JM (November 2009). “Ascorbic acid and tetrahydrobiopterin: looking beyond nitric oxide bioavailability”Cardiovascular Research84 (2): 178–9. doi:10.1093/cvr/cvp307PMID 19744948.
  25. ^ Gori T, Burstein JM, Ahmed S, Miner SE, Al-Hesayen A, Kelly S, Parker JD (September 2001). “Folic acid prevents nitroglycerin-induced nitric oxide synthase dysfunction and nitrate tolerance: a human in vivo study”Circulation104 (10): 1119–23. doi:10.1161/hc3501.095358PMID 11535566.
  26. ^ “Search results for Kuvan”ClinicalTrials.gov. U.S. National Library of Medicine.
  27. ^ “BioMarin Initiates Phase 3b Study to Evaluate the Effects of Kuvan on Neurophychiatric Symptoms in Subjects with PKU”. BioMarin Pharmaceutical Inc. 17 August 2010.
  28. ^ Burton B, Grant M, Feigenbaum A, Singh R, Hendren R, Siriwardena K, et al. (March 2015). “A randomized, placebo-controlled, double-blind study of sapropterin to treat ADHD symptoms and executive function impairment in children and adults with sapropterin-responsive phenylketonuria”Molecular Genetics and Metabolism114 (3): 415–24. doi:10.1016/j.ymgme.2014.11.011PMID 25533024.
  29. ^ Fernell E, Watanabe Y, Adolfsson I, Tani Y, Bergström M, Hartvig P, et al. (May 1997). “Possible effects of tetrahydrobiopterin treatment in six children with autism–clinical and positron emission tomography data: a pilot study”. Developmental Medicine and Child Neurology39 (5): 313–8. doi:10.1111/j.1469-8749.1997.tb07437.xPMID 9236697S2CID 12761124.
  30. ^ Frye RE, Huffman LC, Elliott GR (July 2010). “Tetrahydrobiopterin as a novel therapeutic intervention for autism”Neurotherapeutics7 (3): 241–9. doi:10.1016/j.nurt.2010.05.004PMC 2908599PMID 20643376.
  31. ^ Channon KM (November 2004). “Tetrahydrobiopterin: regulator of endothelial nitric oxide synthase in vascular disease”. Trends in Cardiovascular Medicine14 (8): 323–7. doi:10.1016/j.tcm.2004.10.003PMID 15596110.
  32. ^ Alp NJ, Mussa S, Khoo J, Cai S, Guzik T, Jefferson A, et al. (September 2003). “Tetrahydrobiopterin-dependent preservation of nitric oxide-mediated endothelial function in diabetes by targeted transgenic GTP-cyclohydrolase I overexpression”The Journal of Clinical Investigation112 (5): 725–35. doi:10.1172/JCI17786PMC 182196PMID 12952921.
  33. ^ Khoo JP, Zhao L, Alp NJ, Bendall JK, Nicoli T, Rockett K, et al. (April 2005). “Pivotal role for endothelial tetrahydrobiopterin in pulmonary hypertension”Circulation111 (16): 2126–33. doi:10.1161/01.CIR.0000162470.26840.89PMID 15824200.
  34. ^ Cunnington C, Van Assche T, Shirodaria C, Kylintireas I, Lindsay AC, Lee JM, et al. (March 2012). “Systemic and vascular oxidation limits the efficacy of oral tetrahydrobiopterin treatment in patients with coronary artery disease”Circulation125 (11): 1356–66. doi:10.1161/CIRCULATIONAHA.111.038919PMC 5238935PMID 22315282.
  35. ^ Romanowicz J, Leonetti C, Dhari Z, Korotcova L, Ramachandra SD, Saric N, et al. (August 2019). “Treatment With Tetrahydrobiopterin Improves White Matter Maturation in a Mouse Model for Prenatal Hypoxia in Congenital Heart Disease”Journal of the American Heart Association8 (15): e012711. doi:10.1161/JAHA.119.012711PMC 6761654PMID 31331224.
  36. ^ Kraft VA, Bezjian CT, Pfeiffer S, Ringelstetter L, Müller C, Zandkarimi F, et al. (January 2020). “GTP Cyclohydrolase 1/Tetrahydrobiopterin Counteract Ferroptosis through Lipid Remodeling”ACS Central Science6 (1): 41–53. doi:10.1021/acscentsci.9b01063PMC 6978838PMID 31989025.

Further reading

External links

Tetrahydrobiopterin
INN: sapropterin
(6R)-Tetrahydrobiopterin structure.png
Clinical data
Trade names Kuvan, Biopten
Other names Sapropterin hydrochloride (JAN JP), Sapropterin dihydrochloride (USAN US)
AHFS/Drugs.com Monograph
MedlinePlus a608020
License data
Pregnancy
category
  • AU: B1[1]
  • US: C (Risk not ruled out)[1]
Routes of
administration
By mouth
ATC code
Legal status
Legal status
  • AU: S4 (Prescription only)
  • CA℞-only
  • US: ℞-only
  • In general: ℞ (Prescription only)
Pharmacokinetic data
Elimination half-life 4 hours (healthy adults)
6–7 hours (PKU patients)
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEBI
ChEMBL
PDB ligand
CompTox Dashboard (EPA)
ECHA InfoCard 100.164.121 Edit this at Wikidata
Chemical and physical data
Formula C9H15N5O3
Molar mass 241.251 g·mol−1
3D model (JSmol)

////////Sapropterin, сапроптерин سابروبتيرين , 沙丙蝶呤 , Tetrahydrobiopterin,

Sapropterin Tablets - FDA prescribing information, side effects and uses

Abametapir アバメタピル , абаметапир , أباميتابير , 阿巴甲吡 ,


Abametapir skeletal.svg

Abametapir

アバアバメタピル , абаметапир , أباميتابير 阿巴甲吡 ,

5,5′-dimethyl-2,2′-bipyridine, 6,6′-Bi-3-picoline

  • BRN 0123183
  • HA 44
  • HA-44
  • HA44
Formula
C12H12N2
CAS
1762-34-1
Mol weight
184.2371

Xeglyze, FD APPROVED 24/7/2020

Pediculicide, Metalloproteinase inhibitor
  Disease
Head lice infestation
  • Originator Hatchtech
  • DeveloperDr Reddys Laboratories; Hatchtech
  • ClassAntiparasitics; Heterocyclic compounds; Pyridines; Small molecules
  • Mechanism of ActionChelating agents; Metalloprotease inhibitors
  • Registered Pediculosis
  • 27 Jul 2020Registered for Pediculosis (In adolescents, In children, In infants, In adults) in USA (Topical)
  • 18 Jun 2020FDA assigns PDUFA action date of 12/08/2020 for Abametapir for Pediculosis (Dr Reddy’s Laboratories website, June 2020)
  • 31 Mar 2019Abametapir is still in preregistration phase for Pediculosis in USA

Abametapir is a novel pediculicidal metalloproteinase inhibitor used to treat infestations of head lice.4 The life cycle of head lice (Pediculus capitis) is approximately 30 days, seven to twelve of which are spent as eggs laid on hair shafts near the scalp.2 Topical pediculicides generally lack adequate ovicidal activity,2 including standard-of-care treatments such as permethrin, and many require a second administration 7-10 days following the first to kill newly hatched lice that resisted the initial treatment. The necessity for follow-up treatment may lead to challenges with patient adherence, and resistance to agents like permethrin and pyrethrins/piperonyl butoxide may be significant in some areas.3

Investigations into novel ovicidal treatments revealed that several metalloproteinase enzymes were critical to the egg hatching and survival of head lice, and these enzymes were therefore identified as a potential therapeutic target.1 Abemetapir is an inhibitor of these metalloproteinase enzymes, and the first topical pediculicide to take advantage of this novel target. The improved ovicidal activity (90-100% in vitro) of abemetapir allows for a single administration, in contrast to many other topical treatments, and its novel and relatively non-specific mechanism may help to curb the development of resistance to this agent.1

Abametapir was first approved for use in the United States under the brand name Xeglyze on July 27, 2020.6

Abametapir, sold under the brand name Xeglyze, is a medication used for the treatment of head lice infestation in people six months of age and older.[1][2]

The most common side effects include skin redness, rash, skin burning sensation, skin inflammation, vomiting, eye irritation, skin itching, and hair color changes.[2]

Abametapir is a metalloproteinase inhibitor.[1] Abametapir was approved for medical use in the United States in July 2020.[1][3]

Medical uses

Abametapir is indicated for the topical treatment of head lice infestation in people six months of age and older.[1][2]

History

The U.S. Food and Drug Administration (FDA) approved abametapir based on evidence from two identical clinical trials of 699 participants with head lice.[2] The trials were conducted at fourteen sites in the United States.[2]

The benefit and side effects of abametapir were evaluated in two clinical trials that enrolled participants with head lice who were at least six months old.[2]

About half of all enrolled participants was randomly assigned to abametapir and the other half to placebo.[2] Abametapir lotion or placebo lotion were applied once as a ten-minute treatment to infested hair.[2] The benefit of abametapir in comparison to placebo was assessed after 1, 7 and 14 days by comparing the counts of participants in each group who were free of live lice.[2]

SYN

Ronald Harding, Lewis David Schulz, Vernon Morrison Bowles, “Pediculicidal composition.” WIPO Patent WO2015107384A2, published July, 2015.

References

  1. Jump up to:a b c d e “Xeglyze (abametapir) lotion, for topical use” (PDF)U.S. Food and Drug Administration (FDA). Dr. Reddy’s Laboratories. Inc. Retrieved 25 July 2020.
  2. Jump up to:a b c d e f g h i “Drug Trial Snapshot: Xeglyze”U.S. Food and Drug Administration (FDA). 24 July 2020. Retrieved 6 August 2020.  This article incorporates text from this source, which is in the public domain.
  3. ^ “Abametapir: FDA-Approved Drugs”U.S. Food and Drug Administration (FDA). Retrieved 25 July 2020.

Further reading

External links

  • “Abametapir”Drug Information Portal. U.S. National Library of Medicine.
Abametapir
Abametapir skeletal.svg
Clinical data
Trade names Xeglyze
Other names Ha44
AHFS/Drugs.com Professional Drug Facts
License data
Pregnancy
category
  • US: N (Not classified yet)
Routes of
administration
Topical
Drug class PediculicideMetalloproteinase inhibitor
ATC code
  • None
Legal status
Legal status
Identifiers
CAS Number
PubChem CID
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
PDB ligand
CompTox Dashboard (EPA)
ECHA InfoCard 100.157.434 Edit this at Wikidata
Chemical and physical data
Formula C12H12N2
Molar mass 184.242 g·mol−1
3D model (JSmol)

///////Abametapir, 2020 APPROVALS, FDA 2020, Xeglyze, アバメタピル , абаметапир , أباميتابير 阿巴甲吡 , BRN 0123183, HA 44, head lice

CC1=CC=C(N=C1)C1=CC=C(C)C=N1

Bulevirtide acetate


Bulevirtide acetate

(N-Myristoyl-glycyl-L-threonyl-L-asparaginyl-L-leucyl-L-seryl-L-valyl-Lprolyl-L-asparaginyl-L-prolyl-L-leucyl-glycyl-L-phenylalanyl-L-phenylalanyl-L-prolyl-L-aspartyl-L-histidyl-Lglutaminyl-L-leucyl-L-aspartyl-L-prolyl-L-alanyl-L-phenylalanyl-glycyl-L-alanyl-L-asparaginyl-L-seryl-Lasparaginyl-L-asparaginyl-Lprolyl-L-aspartyl-L-tryptophanyl-L-aspartyl-L-phenylalanyl-L-asparaginyl-L-prolylL-asparaginyl-L-lysyl-L-aspartyl-L-histidyl-L-tryptophanyl-L-prolyl-L-glutamyl-L-alanyl-L-asparaginyl-L-lysylL-valylglycinamide, acetate salt.

molecular formula C248H355N65O72,

molecular mass is 5398.9 g/mol

ブレビルチド酢酸塩;

APROVED 2020/7/31, EU, Hepcludex

MYR GmbH

Antiviral, Entry inhibitor
  Disease
Hepatitis delta virus infection

Bulevirtide is a 47-amino acid peptide with a fatty acid, a myristoyl residue, at the N-terminus and an amidated C-terminus. The active substance is available as acetate salt. The counter ion acetate is bound in ionic form to basic groups of the peptide molecule and is present in a non-stoichiometric ratio. The chemical name of bulevirtide is (N-Myristoyl-glycyl-L-threonyl-L-asparaginyl-L-leucyl-L-seryl-L-valyl-Lprolyl-L-asparaginyl-L-prolyl-L-leucyl-glycyl-L-phenylalanyl-L-phenylalanyl-L-prolyl-L-aspartyl-L-histidyl-Lglutaminyl-L-leucyl-L-aspartyl-L-prolyl-L-alanyl-L-phenylalanyl-glycyl-L-alanyl-L-asparaginyl-L-seryl-Lasparaginyl-L-asparaginyl-Lprolyl-L-aspartyl-L-tryptophanyl-L-aspartyl-L-phenylalanyl-L-asparaginyl-L-prolylL-asparaginyl-L-lysyl-L-aspartyl-L-histidyl-L-tryptophanyl-L-prolyl-L-glutamyl-L-alanyl-L-asparaginyl-L-lysylL-valylglycinamide, acetate salt. It corresponds to the molecular formula C248H355N65O72, its relative molecular mass is 5398.9 g/mol

Bulevirtide appears as a white or off-white hygroscopic powder. It is practically insoluble in water and soluble at concentrations of 1 mg/ml in 50% acetic acid and about 7 mg/ml in carbonate buffer solution at pH 8.8, respectively. The structure of the active substance (AS) was elucidated by a combination of infrared spectroscopy (IR), mass spectrometry (MS), amino acid analysis and sequence analysis Other characteristics studied included ultraviolet (UV) spectrum, higher order structure (1D- and 2D- nuclear magnetic resonance spectroscopy (NMR)) and aggregation (Dynamic Light Scattering). Neither tertiary structure nor aggregation states of bulevirtide have been identified. With regard to enantiomeric purity, all amino acids are used in L-configuration except glycine, which is achiral by nature. Two batches of bulevirtide acetate were evaluated for enanatiomeric purity and no relevant change in configuration during synthesis was detected.

Bulevirtide is manufactured by a single manufacturer. It is a chemically synthesised linear peptide containing only naturally occurring amino acids. The manufacturing of this peptide is achieved using standard solidphase peptide synthesis (SPPS) on a 4-methylbenzhydrylamine resin (MBHA resin) derivatised with Rink amide linker in order to obtain a crude peptide mixture. This crude mixture is purified through a series of washing and preparative chromatography steps. Finally, the purified peptide is freeze-dried prior to final packaging and storage. The process involves further four main steps: synthesis of the protected peptide on the resin while side-chain functional groups are protected as applicable; cleavage of the peptide from the resin, together with the removal of the side chain protecting groups to obtain the crude peptide; purification; and lyophilisation. Two chromatographic systems are used for purification. No design space is claimed. Resin, Linker Fmoc protected amino acids and myristic acid are starting materials in line with ICH Q11. Sufficient information is provided on the source and the synthetic route of the starting materials. The active substance is obtained as a nonsterile, lyophilised powder. All critical steps and parameters were presented and clearly indicated in the description of the manufacturing process. The process description includes also sufficient information on the type of equipment for the SPPS, in-process controls (IPCs). The circumstances under which reprocessing might be performed were clearly presented. No holding times are proposed. Overall the process is sufficiently described.

The finished product is a white to off white lyophilised powder for solution for injection supplied in single-use vials. Each vial contains bulevirtide acetate equivalent to 2 mg bulevirtide. The composition of the finished product was presented. The powder is intended to be dissolved in 1 ml of water for injection per vial. After reconstitution the concentration of bulevirtide net peptide solution in the vial is 2 mg/ml. The components of the formulation were selected by literature review and knowledge of compositions of similar products available on the market at that time, containing HCl, water, mannitol, sodium carbonate, sodium hydrogen carbonate and sodium hydroxide. All excipients are normally used in the manufacture of lyophilisates. The quality of the excipients complies with their respective Ph. Eur monographs. The intrinsic properties of the active substance and the compounding formulation do not support microbiological growth as demonstrated by the stability data. No additional preservatives are therefore needed.

https://www.ema.europa.eu/en/documents/assessment-report/hepcludex-epar-public-assessment-report_en.pdf

Hepcludex is an antiviral medicine used to treat chronic (long-term) hepatitis delta virus (HDV) infection in adults with compensated liver disease (when the liver is damaged but is still able to work), when the presence of viral RNA (genetic material) has been confirmed by blood tests.

HDV is an ‘incomplete’ virus, because it cannot replicate in cells without the help of another virus, the hepatitis B virus. Because of this, patients infected with the virus always also have hepatitis B.

HDV infection is rare, and Hepcludex was designated an ‘orphan medicine’ (a medicine used in rare diseases) on 19 June 2015. For further information on the orphan designation, see EU/3/15/1500.

Hepcludex contains the active substance bulevirtide.

Bulevirtide, sold under the brand name Hepcludex, is an antiviral medication for the treatment of chronic hepatitis D (in the presence of hepatitis B).[2]

The most common side effects include raised levels of bile salts in the blood and reactions at the site of injection.[2]

Bulevirtide works by attaching to and blocking a receptor (target) through which the hepatitis delta and hepatitis B viruses enter liver cells.[2] By blocking the entry of the virus into the cells, it limits the ability of HDV to replicate and its effects in the body, reducing symptoms of the disease.[2]

Bulevirtide was approved for medical use in the European Union in July 2020.[2]

Medical uses

Bulevirtide is indicated for the treatment of chronic hepatitis delta virus (HDV) infection in plasma (or serum) HDV-RNA positive adult patients with compensated liver disease.[2][3]

Pharmacology

Mechanism of action

Bulevirtide binds and inactivates the sodium/bile acid cotransporter, blocking both viruses from entering hepatocytes.[4]

The hepatitis B virus uses its surface lipopeptide pre-S1 for docking to mature liver cells via their sodium/bile acid cotransporter (NTCP) and subsequently entering the cells. Myrcludex B is a synthetic N-acylated pre-S1[5][6] that can also dock to NTCP, blocking the virus’s entry mechanism.[7]

The drug is also effective against hepatitis D because the hepatitis D virus is only infective in the presence of a hepatitis B virus infection.[7]

References

  1. ^ Deterding, K.; Wedemeyer, H. (2019). “Beyond Pegylated Interferon-Alpha: New Treatments for Hepatitis Delta”. Aids Reviews21 (3): 126–134. doi:10.24875/AIDSRev.19000080PMID 31532397.
  2. Jump up to:a b c d e f g “Hepcludex EPAR”European Medicines Agency (EMA). 26 May 2020. Retrieved 12 August 2020. Text was copied from this source which is © European Medicines Agency. Reproduction is authorized provided the source is acknowledged.
  3. ^ “Summary of opinion: Hepcludex” (PDF)European Medicines Agency. 28 May 2020.
  4. ^ Francisco, Estela Miranda (29 May 2020). “Hepcludex”European Medicines Agency. Retrieved 6 August 2020.
  5. ^ Volz T, Allweiss L, Ben MBarek M, Warlich M, Lohse AW, Pollok JM, et al. (May 2013). “The entry inhibitor Myrcludex-B efficiently blocks intrahepatic virus spreading in humanized mice previously infected with hepatitis B virus”. Journal of Hepatology58 (5): 861–7. doi:10.1016/j.jhep.2012.12.008PMID 23246506.
  6. ^ Abbas Z, Abbas M (August 2015). “Management of hepatitis delta: Need for novel therapeutic options”World Journal of Gastroenterology21 (32): 9461–5. doi:10.3748/wjg.v21.i32.9461PMC 4548107PMID 26327754.
  7. Jump up to:a b Spreitzer H (14 September 2015). “Neue Wirkstoffe – Myrcludex B”. Österreichische Apothekerzeitung (in German) (19/2015): 12.

External links

Bulevirtide
Clinical data
Trade names Hepcludex
Other names MyrB, Myrcludex-B[1]
License data
Routes of
administration
Subcutaneous injection
ATC code
  • None
Legal status
Legal status
  • EU: Rx-only [2]
Identifiers
CAS Number
DrugBank
UNII
KEGG
ChEMBL

/////////Bulevirtide acetate, ブレビルチド酢酸塩 , orphan designation, MYR GmbH, PEPTIDE, EU 2020, 2020 APPROVALS

Nifurtimox


Nifurtimox.svg

Nifurtimox

Formula
C10H13N3O5S
CAS
23256-30-6
Mol weight
287.2923

FDA APPROVED, 2020/8/6, LAMPIT

Antiprotozoal
  Disease
Chagas disease

IUPAC Name

3-methyl-4-[(E)-[(5-nitrofuran-2-yl)methylidene]amino]-1lambda6-thiomorpholine-1,1-dione

SMILES

CC1CS(=O)(=O)CCN1\N=C\C1=CC=C(O1)[N+]([O-])=O
SYN
Danong Chen, Glenn Rice. 2013. Novel formulations of nitrofurans including nifurtimox with enhanced activity with lower toxicity.US20150140089A1
  • OriginatorBayer
  • ClassAntiprotozoals; Nitrofurans; Small molecules; Thiamorpholines; Thiazines
  • Mechanism of ActionDNA damage modulators
  • RegisteredChagas disease
  • 07 Aug 2020Registered for Chagas disease (In adolescents, In children, In infants) in USA (PO)
  • 31 Jan 2020Preregistration for Chagas disease (In infants, In children, In adolescents) in USA (PO)
  • 29 Jan 2020Bayer completes a phase I trial in Chagas disease in Argentina (PO) (NCT03334838)
Title: Nifurtimox
CAS Registry Number: 23256-30-6
CAS Name: 3-Methyl-N-[(5-nitro-2-furanyl)methylene]-4-thiomorpholinamine 1,1-dioxide
Additional Names: 4-[(5-nitrofurfurylidene)amino]-3-methylthiomorpholine-1,1-dioxide; tetrahydro-3-methyl-4-[(5-nitrofurfurylidene)amino]-2H-1,4-thiazine 1,1-dioxide; 1-[(5-nitrofurfurylidene)amino]-2-methyltetrahydro-1,4-thiazine 4,4-dioxide
Manufacturers’ Codes: Bay 2502
Trademarks: Lampit (Bayer)
Molecular Formula: C10H13N3O5S
Molecular Weight: 287.29
Percent Composition: C 41.81%, H 4.56%, N 14.63%, O 27.85%, S 11.16%
Literature References: Prepn from 5-nitrofurfural and 4-amino-3-methyltetrahydro-1,4-thiazine 1,1-dioxide: Herlinger et al., DE 1170957 corresp to US 3262930 (1964 and 1966 to Bayer). Series of articles on pharmacology and clinical findings: Arzneim.-Forsch. 22, 1563-1642 (1972). Toxicity data: K. Hoffmann, ibid. 1590.
Properties: Orange-red crystals from dil acetic acid, mp 180-182°. LD50 in mice, rats (mg/kg): 3720, 4050 by gavage (Hoffmann).
Melting point: mp 180-182°
Toxicity data: LD50 in mice, rats (mg/kg): 3720, 4050 by gavage (Hoffmann)
Therap-Cat: Antiprotozoal (Trypanosoma).
Keywords: Antiprotozoal (Trypanosoma).

Nifurtimox, sold under the brand name Lampit, is a medication used to treat Chagas disease and sleeping sickness.[1][4] For sleeping sickness it is used together with eflornithine in nifurtimox-eflornithine combination treatment.[4] In Chagas disease it is a second-line option to benznidazole.[5] It is given by mouth.[1]

Common side effects include abdominal pain, headache, nausea, and weight loss.[1] There are concerns from animal studies that it may increase the risk of cancer but these concerns have not be found in human trials.[5] Nifurtimox is not recommended in pregnancy or in those with significant kidney or liver problems.[5] It is a type of nitrofuran.[5]

Nifurtimox came into medication use in 1965.[5] It is on the World Health Organization’s List of Essential Medicines.[4] It is not available commercially in Canada.[1] It was approved for medical use in the United States in August 2020.[3] In regions of the world where the disease is common nifurtimox is provided for free by the World Health Organization (WHO).[6]

Chagas disease, caused by a parasite known as Trypanosoma cruzi (T.cruzi), is a vector-transmitted disease affecting animals and humans in the Americas. It is commonly known as American Trypanosomiasis.11

The CDC estimates that approximately 8 million people in Central America, South America, and Mexico are infected with T. cruzi, without symptoms. If Chagas disease is left untreated, life-threatening sequelae may result.11

Nifurtimox, developed by Bayer, is a nitrofuran antiprotozoal drug used in the treatment of Chagas disease. On August 6 2020, accelerated FDA approval was granted for its use in pediatric patients in response to promising results from phase III clinical trials. Continued approval will be contingent upon confirmatory data.10 A convenient feature of Bayer’s formulation is the ability to divide the scored tablets manually without the need for pill-cutting devices.10

Medical uses

Nifurtimox has been used to treat Chagas disease, when it is given for 30 to 60 days.[7][8] However, long-term use of nifurtimox does increase chances of adverse events like gastrointestinal and neurological side effects.[8][9] Due to the low tolerance and completion rate of nifurtimox, benznidazole is now being more considered for those who have Chagas disease and require long-term treatment.[5][9]

In the United States nifurtimox is indicated in children and adolescents (birth to less than 18 years of age and weighing at least 2.5 kilograms (5.5 lb) for the treatment of Chagas disease (American Trypanosomiasis), caused by Trypanosoma cruzi.[2]

Nifurtimox has also been used to treat African trypanosomiasis (sleeping sickness), and is active in the second stage of the disease (central nervous system involvement). When nifurtimox is given on its own, about half of all patients will relapse,[10] but the combination of melarsoprol with nifurtimox appears to be efficacious.[11] Trials are awaited comparing melarsoprol/nifurtimox against melarsoprol alone for African sleeping sickness.[12]

Combination therapy with eflornithine and nifurtimox is safer and easier than treatment with eflornithine alone, and appears to be equally or more effective. It has been recommended as first-line treatment for second-stage African trypanosomiasis.[13]

Pregnancy and breastfeeding

Use of nifurtimox should be avoided in pregnant women due to limited use.[5][8][14] There is limited data shown that nifurtimox doses up to 15 mg/kg daily can cause adverse effects in breastfed infants.[15] Other authors do not consider breastfeeding a contraindication during nifurtimox use.[15]

Side effects

Side effects occur following chronic administration, particularly in elderly people. Major toxicities include immediate hypersensitivity such as anaphylaxis and delayed hypersensitivity reaction involving icterus and dermatitis. Central nervous system disturbances and peripheral neuropathy may also occur.[8]

Contraindications

Nifurtimox is contraindicated in people with severe liver or kidney disease, as well as people with a background of neurological or psychiatric disorders.[5][16][20]

Mechanism of action

Nifurtimox forms a nitro-anion radical metabolite that reacts with nucleic acids of the parasite causing significant breakdown of DNA.[8] Its mechanism is similar to that proposed for the antibacterial action of metronidazole. Nifurtimox undergoes reduction and creates oxygen radicals such as superoxide. These radicals are toxic to T. cruzi. Mammalian cells are protected by presence of catalaseglutathioneperoxidases, and superoxide dismutase. Accumulation of hydrogen peroxide to cytotoxic levels results in parasite death.[8]

Manufacturing and availability

A bottle of nifurtimox

Nifurtimox is sold under the brand name Lampit by Bayer.[3] It was previously known as Bayer 2502.

Nifurtimox is only licensed for use in Argentina and Germany,[citation needed] where it is sold as 120-mg tablets. It was approved for medical use in the United States in August 2020.[3]

Research

Nifurtimox is in a phase-II clinical trial for the treatment of pediatric neuroblastoma and medulloblastoma.[21]

SYN

Nifurtimox

Synthesis of Essential Drugs

2006, Pages 559-582

Nifurtimox, 1,1-dioxide 4-[(5-nitrofuryliden)amino]-3-methylthiomorpholine (37.4.7), is made by the following scheme. Interaction of 2-mercaptoethanol with propylene oxide in the presence of potassium hydroxide gives (2-hydroxyethyl)-(2-hydroxypropylsul-fide) (37.4.3), which undergoes intramolecular dehydration using potassium bisulfate to make 2-methyl-1,4-oxithiane (37.4.4). Oxidation of this using hydrogen peroxide gives 2-methyl-1,4-oxithian-4,4-dioxide (37.4.5), which when reacted with hydrazine transforms to 4-amino-3-methyltetrahydro-1,4-thiazin-1,1-dioxide (37.4.6). Reacting this with 5-nitrofurfurol gives the corresponding hydrazone—the desired nifurtimox [58,59].

58. H. Herlinger, K.H. Heinz, S. Petersen, M.Bock, Ger. Pat. 1.170.957 (1964).

59. H. Herlinger, K.H. Heinz, S. Petersen, M. Bock, U.S. Pat. 3.262.930 (1966)

References

  1. Jump up to:a b c d e f “Nifurtimox (Systemic)”Drugs.com. 1995. Archived from the original on 20 December 2016. Retrieved 3 December 2016.
  2. Jump up to:a b “Lampit (nifurtimox) tablets, for oral use” (PDF)U.S. Food and Drug Administration(FDA). Bayer HealthCare Pharmaceuticals Inc. Retrieved 6 August 2020.
  3. Jump up to:a b c d “Lampit: FDA-Approved Drugs”U.S. Food and Drug Administration (FDA). Retrieved 6 August 2020.
  4. Jump up to:a b c World Health Organization (2019). World Health Organization model list of essential medicines: 21st list 2019. Geneva: World Health Organization. hdl:10665/325771. WHO/MVP/EMP/IAU/2019.06. License: CC BY-NC-SA 3.0 IGO.
  5. Jump up to:a b c d e f g h Bern, Caryn; Montgomery, Susan P.; Herwaldt, Barbara L.; Rassi, Anis; Marin-Neto, Jose Antonio; Dantas, Roberto O.; Maguire, James H.; Acquatella, Harry; Morillo, Carlos (2007-11-14). “Evaluation and Treatment of Chagas Disease in the United States”JAMA298 (18): 2171–81. doi:10.1001/jama.298.18.2171ISSN 0098-7484PMID 18000201.
  6. ^ “Trypanosomiasis, human African (sleeping sickness)”World Health Organization. February 2016. Archived from the original on 4 December 2016. Retrieved 7 December2016.
  7. ^ Coura JR, de Castro SL (2002). “A critical review of Chagas disease chemotherapy”Mem Inst Oswaldo Cruz97 (1): 3–24. doi:10.1590/S0074-02762002000100001PMID 11992141.
  8. Jump up to:a b c d e f g h “Nifurtimox Drug Information, Professional”http://www.drugs.comArchivedfrom the original on 2016-11-08. Retrieved 2016-11-09.
  9. Jump up to:a b Jackson, Yves; Alirol, Emilie; Getaz, Laurent; Wolff, Hans; Combescure, Christophe; Chappuis, François (2010-11-15). “Tolerance and Safety of Nifurtimox in Patients with Chronic Chagas Disease”Clinical Infectious Diseases51 (10): e69–e75. doi:10.1086/656917ISSN 1058-4838PMID 20932171.
  10. ^ Pepin J, Milord F, Mpia B, et al. (1989). “An open clinical trial of nifurtimox for arseno-resistant T. b. gambiense sleeping sickness in central Zaire”. Trans R Soc Trop Med Hyg83(4): 514–7. doi:10.1016/0035-9203(89)90270-8PMID 2694491.
  11. ^ Bisser S, N’Siesi FX, Lejon V, et al. (2007). “Equivalence Trial of Melarsoprol and Nifurtimox Monotherapy and Combination Therapy for the Treatment of Second-Stage Trypanosoma brucei gambiense Sleeping Sickness”J Infect Dis195 (3): 322–329. doi:10.1086/510534PMID 17205469.
  12. ^ Pepin J (2007). “Combination Therapy for Sleeping Sickness: A Wake-Up Call”J Infect Dis195 (3): 311–13. doi:10.1086/510540PMID 17205466.
  13. ^ Priotto G, Kasparian S, Mutombo W, et al. (July 2009). “Nifurtimox-eflornithine combination therapy for second-stage African Trypanosoma brucei gambiensetrypanosomiasis: a multicentre, randomised, phase III, non-inferiority trial”. Lancet374(9683): 56–64. doi:10.1016/S0140-6736(09)61117-Xhdl:10144/72797PMID 19559476.
  14. ^ Schaefer, Christof; Peters, Paul W. J.; Miller, Richard K. (2014-09-17). Drugs During Pregnancy and Lactation: Treatment Options and Risk Assessment. Academic Press. ISBN 9780124079014Archived from the original on 2017-09-08.
  15. Jump up to:a b “Nifurtimox use while Breastfeeding | Drugs.com”http://www.drugs.comArchived from the original on 2016-11-08. Retrieved 2016-11-07.
  16. Jump up to:a b c “Parasites – American Trypanosomiasis (also known as Chagas Disease)”U.S. Centers for Disease Control and Prevention (CDC)Archived from the original on 2016-11-06. Retrieved 2016-11-09.
  17. Jump up to:a b Forsyth, Colin J.; Hernandez, Salvador; Olmedo, Wilman; Abuhamidah, Adieb; Traina, Mahmoud I.; Sanchez, Daniel R.; Soverow, Jonathan; Meymandi, Sheba K. (2016-10-15). “Safety Profile of Nifurtimox for Treatment of Chagas Disease in the United States”Clinical Infectious Diseases63 (8): 1056–1062. doi:10.1093/cid/ciw477ISSN 1537-6591PMC 5036918PMID 27432838.
  18. ^ Castro, José A.; de Mecca, Maria Montalto; Bartel, Laura C. (2006-08-01). “Toxic side effects of drugs used to treat Chagas’ disease (American trypanosomiasis)”. Human & Experimental Toxicology25 (8): 471–479. doi:10.1191/0960327106het653oaISSN 0960-3271PMID 16937919.
  19. Jump up to:a b Estani, Sergio Sosa; Segura, Elsa Leonor (1999-09-01). “Treatment of Trypanosoma cruzi infection in the undetermined phase. Experience and current guidelines of treatment in Argentina”Memórias do Instituto Oswaldo Cruz94: 363–365. doi:10.1590/S0074-02761999000700070ISSN 0074-0276PMID 10677756.
  20. ^ “Chagas disease”World Health OrganizationArchived from the original on 2014-02-27. Retrieved 2016-11-08.
  21. ^ Clinical trial number NCT00601003 for “Study of Nifurtimox to Treat Refractory or Relapsed Neuroblastoma or Medulloblastoma” at ClinicalTrials.gov. Retrieved on July 10, 2009.

External links

  • “Nifurtimox”Drug Information Portal. U.S. National Library of Medicine.
Nifurtimox
Nifurtimox.svg
Nifurtimox 3D.png
Clinical data
Trade names Lampit[1]
Other names Bayer 2502[1]
AHFS/Drugs.com Drugs.com archive
Lampit
License data
Routes of
administration
By mouth
ATC code
Legal status
Legal status
Pharmacokinetic data
Bioavailability Low
Metabolism Liver (Cytochrome P450 oxidase (CYP) involved)
Elimination half-life 2.95 ± 1.19 hours
Excretion Kidney, very low
Identifiers
CAS Number
PubChem CID
DrugBank
ChemSpider
UNII
KEGG
ChEBI
ChEMBL
CompTox Dashboard (EPA)
ECHA InfoCard 100.041.377 Edit this at Wikidata
Chemical and physical data
Formula C10H13N3O5S
Molar mass 287.29 g·mol−1
3D model (JSmol)
Chirality Racemic mixture
Melting point 180 to 182 °C (356 to 360 °F)

///////////Nifurtimox, LAMPIT, 2020 APPROVALS, FDA 2020, ニフルチモックス, CHAGAS DISEASE, ANTI PROTOZOAL

Imlifidase


MDSFSANQEI RYSEVTPYHV TSVWTKGVTP PANFTQGEDV FHAPYVANQG WYDITKTFNG
KDDLLCGAAT AGNMLHWWFD QNKDQIKRYL EEHPEKQKIN FNGEQMFDVK EAIDTKNHQL
DSKLFEYFKE KAFPYLSTKH LGVFPDHVID MFINGYRLSL TNHGPTPVKE GSKDPRGGIF
DAVFTRGDQS KLLTSRHDFK EKNLKEISDL IKKELTEGKA LGLSHTYANV RINHVINLWG
ADFDSNGNLK AIYVTDSDSN ASIGMKKYFV GVNSAGKVAI SAKEIKEDNI GAQVLGLFTL
STGQDSWNQT N

Imlifidase

イムリフィダーゼ;

Formula
C1575H2400N422O477S6
CAS
1947415-68-0
Mol weight
35070.8397

EMA APPROVED, 2020/8/25, Idefirix

Pre-transplant treatment to make patients with donor specific IgG eligible for kidney transplantation
Immunosuppressant, Immunoglobulin modulator (enzyme)

Imlifidase is under investigation in clinical trial NCT02854059 (IdeS in Asymptomatic Asymptomatic Antibody-Mediated Thrombotic Thrombocytopenic Purpura (TTP) Patients).

Imlifidase, brand name Idefirix, is a medication for the desensitization of highly sensitized adults needing kidney transplantation, but unlikely to receive a compatible transplant.[1]

Imlifidase is a cysteine protease derived from the immunoglobulin G (IgG)‑degrading enzyme of Streptococcus pyogenes.[1] It cleaves the heavy chains of all human IgG subclasses (but no other immunoglobulins), eliminating Fc-dependent effector functions, including CDC and antibody-dependent cell-mediated cytotoxicity (ADCC).[1] Thus, imlifidase reduces the level of donor specific antibodies, enabling transplantation.[1]

The benefits with imlifidase are its ability to convert a positive crossmatch to a negative one in highly sensitized people to allow renal transplantation.[1] The most common side effects are infections and infusion related reactions.[1]

In June 2020, the Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency (EMA) recommended the approval of Imlifidase.[1][2]

Medical uses

Per the CHMP recommendation, imlifidase will be indicated for desensitization treatment of highly sensitized adult kidney transplant people with positive crossmatch against an available deceased donor.[1] The use of imlifidase should be reserved for people unlikely to be transplanted under the available kidney allocation system including prioritization programmes for highly sensitized people.[1]

History

Imlifidase was granted orphan drug designations by the European Commission in January 2017, and November 2018,[3][4] and by the U.S. Food and Drug Administration (FDA) in both February and July 2018.[5][6]

In February 2019, Hansa Medical AB changed its name to Hansa Biopharma AB.[4]

References

  1. Jump up to:a b c d e f g h i “Imlifidase: Pending EC decision”European Medicines Agency (EMA). 25 June 2020. Retrieved 26 June 2020.  This article incorporates text from this source, which is in the public domain.
  2. ^ “New treatment to enable kidney transplant in highly sensitised patients”European Medicines Agency (Press release). 26 June 2020. Retrieved 26 June 2020.  This article incorporates text from this source, which is in the public domain.
  3. ^ “EU/3/16/1826”European Medicines Agency (EMA). 12 January 2017. Retrieved 27 June 2020.  This article incorporates text from this source, which is in the public domain.
  4. Jump up to:a b “EU/3/18/2096”European Medicines Agency (EMA). 13 February 2019. Retrieved 27 June 2020.  This article incorporates text from this source, which is in the public domain.
  5. ^ “Imlifidase Orphan Drug Designation and Approval”U.S. Food and Drug Administration (FDA). 3 July 2018. Retrieved 27 June 2020.
  6. ^ “Imlifidase Orphan Drug Designation and Approval”U.S. Food and Drug Administration (FDA). 14 February 2018. Retrieved 27 June 2020.

Further reading

External links

  • “Imlifidase”Drug Information Portal. U.S. National Library of Medicine.
Imlifidase
Clinical data
Pronunciation im lif’ i dase
Trade names Idefirix
Other names HMED-IdeS
Routes of
administration
Intravenous
ATC code
Identifiers
CAS Number
DrugBank
UNII
KEGG
ChEMBL
Chemical and physical data
Formula C1575H2400N422O477S6
Molar mass 35071.36 g·mol−1

//////////Imlifidase, Idefirix, PEPTIDE, イムリフィダーゼ , 2020 APPROVALS, EMA 2020, EU 2020

Pralsetinib


Pralsetinib.png

ChemSpider 2D Image | trans-N-{(1S)-1-[6-(4-Fluoro-1H-pyrazol-1-yl)-3-pyridinyl]ethyl}-1-methoxy-4-{4-methyl-6-[(5-methyl-1H-pyrazol-3-yl)amino]-2-pyrimidinyl}cyclohexanecarboxamide | C27H32FN9O2

Pralsetinib

Formula
C27H32FN9O2
CAS
2097132-94-8
Mol weight
533.6005
Cyclohexanecarboxamide, N-[(1S)-1-[6-(4-fluoro-1H-pyrazol-1-yl)-3-pyridinyl]ethyl]-1-methoxy-4-[4-methyl-6-[(5-methyl-1H-pyrazol-3-yl)amino]-2-pyrimidinyl]-, cis
2097132-94-8 [RN]
BLU-667
BS-15942

Other Names

  • cis-N-[(1S)-1-[6-(4-Fluoro-1H-pyrazol-1-yl)-3-pyridinyl]ethyl]-1-methoxy-4-[4-methyl-6-[(5-methyl-1H-pyrazol-3-yl)amino]-2-pyrimidinyl]cyclohexanecarboxamide
  • BLU 123244
  • BLU 667
  • Pralsetinib
  • X 581238
  • cis-N-{(1S)-1-[6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl]ethyl}-1-methoxy-4-{4-methyl-6-[(5-methyl-1H-pyrazol-3-yl)amino]pyrimidin-2-yl}cyclohexane-1-carboxamide

N-[(1S)-1-[6-(4-fluoropyrazol-1-yl)pyridin-3-yl]ethyl]-1-methoxy-4-[4-methyl-6-[(5-methyl-1H-pyrazol-3-yl)amino]pyrimidin-2-yl]cyclohexane-1-carboxamide

FDA APPROVED GAVRETO, 2020/9/4

Pralsetinib, sold under the brand name Gavreto, is a medication for the treatment of metastatic RET fusion-positive non-small cell lung cancer (NSCLC).[1] Pralsetinib is a tyrosine kinase inhibitor. It is taken by mouth.[1]

The most common adverse reactions include increased aspartate aminotransferase (AST), decreased hemoglobin, decreased lymphocytes, decreased neutrophils, increased alanine aminotransferase (ALT), increased creatinine, increased alkaline phosphatase, fatigue, constipation, musculoskeletal pain, decreased calcium, hypertension, decreased sodium, decreased phosphate, and decreased platelets.[1]

Pralsetinib was approved for medical use in the United States in September 2020.[1][2][3][4]

Medical uses

Pralsetinib is indicated for the treatment of adults with metastatic RET fusion-positive non-small cell lung cancer (NSCLC) as detected by an FDA approved test.[1][4]

History

Efficacy was investigated in a multicenter, open-label, multi-cohort clinical trial (ARROW, NCT03037385) with 220 participants aged 26-87 whose tumors had RET alterations.[1][4] Identification of RET gene alterations was prospectively determined in local laboratories using either next generation sequencing, fluorescence in situ hybridization, or other tests.[1] The main efficacy outcome measures were overall response rate (ORR) and response duration determined by a blinded independent review committee using RECIST 1.1.[1] The trial was conducted at sites in the United States, Europe and Asia.[4]

Efficacy for RET fusion-positive NSCLC was evaluated in 87 participants previously treated with platinum chemotherapy.[1] The ORR was 57% (95% CI: 46%, 68%); 80% of responding participants had responses lasting 6 months or longer.[1] Efficacy was also evaluated in 27 participants who never received systemic treatment.[1] The ORR for these participants was 70% (95% CI: 50%, 86%); 58% of responding participants had responses lasting 6 months or longer.[1]

The US Food and Drug Administration (FDA) granted the application for pralsetinib priority revieworphan drug, and breakthrough therapy designations[1]and granted approval of Gavreto to Blueprint Medicines.[1]

PATENT

US 20170121312

https://patents.google.com/patent/US20170121312A1/en

    • Step 7: Synthesis of (1R,4S)—N—((S)-1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethyl)-1-methoxy-4-(4-methyl-6-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-2-yl)cyclohexane-carboxamide (Compound 129) and (1S,4R)—N—((S)-1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethyl)-1-methoxy-4-(4-methyl-6-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-2-yl)cyclohexanecarboxamide (Compound 130)

    • [0194]
      Figure US20170121312A1-20170504-C00094
    • [0195]
      The title compounds were prepared from methyl 1-methoxy-4-(4-methyl-6-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-2-yl)cyclohexanecarboxylate (192 mg, 0.53 mmol) using the same two-step procedure (hydrolysis and amide coupling) outlined in Synthetic Protocols 1 and 2, with PyBOP as the amide coupling reagent instead of HATU. The products were initially isolated as a mixture of diastereomers (190 mg), which was then dissolved in 6 mL methanol and purified by SFC (ChiralPak AD-H 21×250 mm, 40% MeOH containing 0.25% DEA in CO2, 2.5 mL injections, 70 mL/min). Peak 1 was concentrated to give (1R,4S)—N—((S)-1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethyl)-1-methoxy-4-(4-methyl-6-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-2-yl)cyclohexanecarboxamide (29 mg, 10%) as a white solid. Peak 2 was concentrated to give (1s,4R)—N—((S)-1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethyl)-1-methoxy-4-(4-methyl-6-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-2-yl)cyclohexane-carboxamide (130 mg, 46%) as a white solid.

Example 6. Synthesis of Compound 149Step 1: Synthesis of Methyl 4-(2-chloro-6-methylpyrimidin-4-yl)-1-methoxycyclohexane-1-carboxylate

    • [0196]
      Figure US20170121312A1-20170504-C00095
    • [0197]
      Methyl 4-iodo-1-methoxycyclohexanecarboxylate (3.37 g, 11.3 mmol) was dissolved in dimethylacetamide (38 mL) in a pressure vessel under a stream of N2. Rieke Zinc (17.7 mL of a 50 mg/mL suspension in THF, 13.6 mmol) was added quickly via syringe, and the vessel was capped and stirred at ambient temperature for 15 minutes. The vessel was opened under a stream of Nand 2,4-dichloro-6-methylpyrimidine (1.84 g, 11.3 mmol) was added followed by PdCl2dppf (826 mg, 1.13 mmol). The vessel was capped and heated to 80° C. for one hour, then cooled to room temperature. The reaction mixture was diluted with EtOAc, filtered through celite, and the filtrate was washed with H2O (3×), brine, dried over sodium sulfate, filtered, and concentrated. The resulting residue was purified by flash-column chromatography on silica gel (gradient elution, 0 to 50% EtOAc-hexanes) to give methyl 4-(2-chloro-6-methylpyrimidin-4-yl)-1-methoxycyclohexane-1-carboxylate (74 mg, 2.2%) as a colorless oil. MS (ES+) C14H19ClN2Orequires: 298, found: 299 [M+H]+.

Step 2: Synthesis of tert-Butyl 3-((4-(4-methoxy-4-(methoxycarbonyl)cyclohexyl)-6-methylpyrimidin-2-yl)amino)-5-methyl-1H-pyrazole-1-carboxylate

    • [0198]
      Figure US20170121312A1-20170504-C00096
    • [0199]
      Methyl 4-(2-chloro-6-methylpyrimidin-4-yl)-1-methoxycyclohexane-1-carboxylate (70.5 mg, 0.236 mmol), tert-butyl 3-amino-5-methyl-1H-pyrazole-1-carboxylate (69.8 mg, 0.354 mmol), di-tert-butyl(2′,4′,6′-triisopropyl-[1,1′-biphenyl]-2-yl)phosphine (20.0 mg, 0.2 equiv.), Pd2(dba)(21.6 mg, 0.1 equiv.), and potassium acetate (70 mg, 0.71 mmol) were combined in a vial under nitrogen and 0.98 mL dioxane was added. The reaction mixture was heated to 115° C. for 2 h, then cooled to ambient temperature. The reaction mixture was diluted with EtOAc, filtered through celite, concentrated onto silica gel, and the resulting residue was purified by flash-column chromatography on silica gel (gradient elution, 0 to 100% ethyl acetate-hexanes) to give tert-butyl 3-((4-(4-methoxy-4-(methoxycarbonyl)cyclohexyl)-6-methylpyrimidin-2-yl)amino)-5-methyl-1H-pyrazole-1-carboxylate (48 mg, 44%) as a yellow oil. MS (ES+) C23H33N5Orequires: 459, found: 460 [M+H]+.

Step 3: Synthesis of 1-Methoxy-4-(6-methyl-2-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-4-yl)cyclohexane-1-carboxylic acid

    • [0200]
      Figure US20170121312A1-20170504-C00097
    • [0201]
      Lithium hydroxide monohydrate (13 mg, 0.31 mmol) was added to a solution of tert-butyl 3-((4-(4-methoxy-4-(methoxycarbonyl)cyclohexyl)-6-methylpyrimidin-2-yl)amino)-5-methyl-1H-pyrazole-1-carboxylate (47.7 mg, 0.104 mmol) in THF/MeOH/H2O (17:1:1, 1.8 mL). The reaction mixture was heated to 60° C. and stirred for 16 h. The reaction mixture was then cooled to ambient temperature and concentrated to give crude 1-methoxy-4-(6-methyl-2-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-4-yl)cyclohexane-1-carboxylic acid (57 mg, crude) which was used in the subsequent amide coupling without any further purification. MS (ES+) C17H23N5Orequires: 345, found: 346 [M+H]+.

Step 4: Synthesis of (1s,4R)—N—((S)-1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethyl)-1-methoxy-4-(6-methyl-2-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-4-yl)cyclohexane-1-carboxamide (Compound 149)

    • [0202]
      Figure US20170121312A1-20170504-C00098
    • [0203]
      The title compound was prepared from 1-methoxy-4-(6-methyl-2-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-4-yl)cyclohexane-1-carboxylic acid (57 mg, 0.104 mmol) using the same procedured (amide coupling) outlined in Synthetic Protocols 1 and 2, with PyBOP as the amide coupling reagent instead of HATU. The products were initially isolated as a mixture of diastereomers (36 mg), which was then dissolved in 6 mL methanol-DCM (1:1) and purified by SFC (ChiralPak IC-H 21×250 mm, 40% MeOH containing 0.25% DEA in CO2, 1.0 mL injections, 70 mL/min). Peak 1 was an undesired isomer, and Peak 2 was concentrated to give (1 s,4R)—N—((S)-1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethyl)-1-methoxy-4-(6-methyl-2-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-4-yl)cyclohexane-1-carboxamide (13.4 mg, 24%) as a white solid.

Synthesis of IntermediatesExample 7. Synthesis of Ketone and Boronate IntermediatesA. Methyl 1-methoxy-4-oxocyclohexane-1-carboxylate

    • [0204]
      Figure US20170121312A1-20170504-C00099
    • [0205]
      The title compound was prepared as described in WO 2014/130810 A1 page 86.

B. Ethyl 1-ethoxy-4-oxocyclohexane-1-carboxylate

    • [0206]
      Figure US20170121312A1-20170504-C00100

Step 1: Synthesis of ethyl 8-ethoxy-1,4-dioxaspiro[4.5]decane-8-carboxylate

    • [0207]
      A solution of 1,4-dioxaspiro[4.5]decan-8-one (20.0 g, 128 mmol) in CHBr(3234 g, 1280 mmol) was cooled to 0° C. and potassium hydroxide (57.5 g, 1024 mmol) in EtOH (300 mL) was added dropwise over 2.5 hrs. After stirring the mixture for 23 h, the mixture was concentrated, and the residue was partitioned between EtOAc and H2O. The organic layer was washed with brine, dried over Na2SO4, filtered, and concentrated under reduced pressure to give crude product, which was purified by flash column chromatography on silica gel (gradient elution, PE:EA=15:1 to 10:1) to obtain the title compound (18.0 g).

Step 2: Synthesis of ethyl 1-ethoxy-4-oxocyclohexane-1-carboxylate

    • [0208]
      To a solution of ethyl 8-ethoxy-1,4-dioxaspiro[4.5]decane-8-carboxylate (10 g, 43 mmol) in 1,4-dioxane (250 mL) was added aqueous HCl (6 M, 92.5 mL), and the mixture was stirred for 23 h at ambient temperature. The mixture was then diluted with H2O and extracted with EtOAc.
    • [0209]
      The organic layers were washed with brine, dried over Na2SO4, filtered, and concentrated under reduced pressure to give a crude residue, which was purified by flash column chromatography on silica gel (PE:EA=15:1) to obtain the product (8.0 g). 1H NMR (400 MHz, DMSO) δ 4.20-4.13 (m, 2H), 3.43 (q, J=6.9 Hz, 1H), 2.48-2.39 (m, 1H), 2.24-2.12 (m, 2H), 2.10-2.01 (m, 1H), 1.22 (t, J=7.1 Hz, 2H), 1.17 (t, J=7.0 Hz, 2H).

C. Ethyl 6,6-dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)cyclohex-3-ene-1-carboxylate

    • [0210]
      Figure US20170121312A1-20170504-C00101

Step 1: Synthesis of ethyl 2,2-dimethyl-4-oxocyclohexane-1-carboxylate

    • [0211]
      A solution of methylmagnesium bromide (3M, 109.8 mL, 329.4 mmol) was added dropwise to a suspension of CuCN (14.75 g, 164.7 mmol) in diethyl ether (50 mL) at 0° C. The mixture was stirred for 30 min at 0° C. and then cooled to −78° C. The solution of ethyl 2-methyl-4-oxocyclohex-2-ene-1-carboxylate (10 g, 54.9 mmol) in diethyl ether (10 mL) was then added dropwise. The mixture was stirred between −40° C. to −20° C. for 2 h, then was warmed to ambient temperature for 16 h. The reaction mixture was carefully added to a saturated solution of ammonium chloride. The aqueous layer was extracted twice with diethyl ether, and the organic layers were combined. The combined organic layer was washed with brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by flash column chromatography on silica gel (PE:EA=10:1) to give ethyl 2,2-dimethyl-4-oxocyclohexane-1-carboxylate (1.16 g).

Step 2: Synthesis of ethyl 6,6-dimethyl-4-(((trifluoromethyl)sulfonyl)oxy)cyclohex-3-ene-1-carboxylate

    • [0212]
      Ethyl 2,2-dimethyl-4-oxocyclohexane-1-carboxylate (1.16 g, 5.85 mmol) and DIPEA (3.03 g, 23.4 mmol) were dissolved in dry toluene (2 mL) and heated at 45° C. for 10 minutes. Trifluoromethanesulfonic anhydride (6.61 g, 23.4 mmol) in DCM (20 mL) was added dropwise over 10 min and the mixture was heated at 45° C. for 2 h. The mixture was allowed to cool to room temperature, concentrated, diluted with water (60 mL) and extracted with DCM (2×40 mL). The organic layer was washed with saturated sodium bicarbonate solution (20 mL) and brine (20 mL), dried over sodium sulfate, filtered, and concentrated. The crude product was purified by flash column chromatography on silica gel (gradient elution, 0 to 100% ethyl acetate-petroleum ether) to afford ethyl 6,6-dimethyl-4-(((trifluoromethyl)sulfonyl)oxy)cyclohex-3-ene-1-carboxylate (1 g).

Step 3: Synthesis of ethyl 6,6-dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)cyclohex-3-ene-1-carboxylate

    • [0213]
      Ethyl 6,6-dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)cyclohex-3-ene-1-carboxylate (1 g, 3.03 mmol), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (1.15 g, 4.54 mmol), Pd(dppf)Cl(73.5 mg, 0.09 mmol) and potassium acetate (891 mg, 9.08 mmol) were suspended in 1,4-dioxane (20 mL). The reaction mixture was flushed with nitrogen, then heated to 100° C. for 2 h. The mixture was cooled to room temperature, filtered, and concentrated, and the resulting brown oil was purified by flash column chromatography on silica gel (gradient elution, 0 to 100% ethyl acetate-petroleum ether) to afford ethyl 6,6-dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)cyclohex-3-ene-1-carboxylate (618 mg).

D. Ethyl 6-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)cyclohex-3-ene-1-carboxylate

    • [0214]
      Figure US20170121312A1-20170504-C00102
    • [0215]
      Ethyl 6-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)cyclohex-3-ene-1-carboxylate was prepared using the same synthetic protocol as described above using ethyl 2-methyl-4-oxocyclohexane-1-carboxylate as the starting material.

E. Methyl 2-methyl-5-oxotetrahydro-2H-pyran-2-carboxylate

    • [0216]
      Figure US20170121312A1-20170504-C00103

Step 1: Synthesis of methyl 2-methyl-3,4-dihydro-2H-pyran-2-carboxylate

    • [0217]
      A mixture of acrylaldehyde (120 g, 2.14 mol), methyl methacrylate (200 g, 2.00 mol) and hydroquinone (2.2 g, 20 mmol) were heated in a sealed steel vessel at 180° C. for one h. The mixture was then cooled to ambient temperature and concentrated. The residue was purified by silica gel column chromatography (gradient elution, petroleum ether:ethyl acetate=100:1 to 80:1) to give methyl 2-methyl-3,4-dihydro-2H-pyran-2-carboxylate (70 g, 22% yield) as a pale yellow oil. 1H-NMR (400 MHz, CDCl3): δ 6.38 (d, J=6.4 Hz, 1H), 4.73-4.70 (m, 1H), 3.76 (s, 3H), 2.25-2.22 (m, 1H), 1.99-1.96 (m, 2H), 1.79-1.77 (m, 1H), 1.49 (s, 3H).

Step 2: Synthesis of methyl 5-hydroxy-2-methyltetrahydro-2H-pyran-2-carboxylate

    • [0218]
      To a solution of methyl 2-methyl-3,4-dihydro-2H-pyran-2-carboxylate (20.0 g, 128 mmol) in anhydrous tetrahydrofuran (200 mL) was added borane (67 mL, 1 M in tetrahydrofuran) dropwise at −5° C. The reaction mixture was stirred at 0° C. for 3 hours. This reaction was monitored by TLC. The mixture was quenched by a solution of sodium acetate (10.5 g, 128 mmol) in water (15 mL). Then the mixture was treated with 30% hydrogen peroxide solution (23.6 g, 208.2 mmol) slowly at 0° C. and stirred at 30° C. for 3 h. The mixture was then partitioned between saturated sodium sulfite solution and tetrahydrofuran. The aqueous layer was further extracted with tetrahydrofuran (2×). The combined organic layers were washed with saturated brine, dried over sodium sulfate and concentrated in vacuo. The residue was purified by a silica gel column chromatography (gradient elution, petroleum ether:ethyl acetate=10:1 to 1:1) to give crude methyl 5-hydroxy-2-methyltetrahydro-2H-pyran-2-carboxylate (18 g, crude) as a pale yellow oil, which used directly for next step.

Step 3: Synthesis of methyl 2-methyl-5-oxotetrahydro-2H-pyran-2-carboxylate

    • [0219]
      To a solution of methyl 5-hydroxy-2-methyltetrahydro-2H-pyran-2-carboxylate (18.0 g, 103 mmol) in anhydrous dichloromethane (200 mL) was added PCC (45.0 g, 209 mmol) in portions. The reaction mixture was stirred at ambient temperature until TLC indicated the reaction was completed. Petroleum ether (500 mL) was then added and the mixture was filtered. The filter cake was washed with petroleum ether (100 mL), and the filtrate was concentrated under vacuum to give methyl 2-methyl-5-oxotetrahydro-2H-pyran-2-carboxylate (15 g, 84% yield) as a pale yellow oil. 1H-NMR (400 MHz, CDCl3): δ 4.25 (d, J=17.6 Hz, 1H), 4.07 (d, J=17.6 Hz, 1H), 3.81 (s, 3H), 2.52-2.44 (m, 3H), 2.11-2.04 (m, 1H), 1.53 (s, 3H).

Example 8. Synthesis of Iodide IntermediatesA. Methyl 1-methoxy-4-iodocyclohexane-1-carboxylate

    • [0220]
      Figure US20170121312A1-20170504-C00104

Step 1: Synthesis of methyl 1-methoxy-4-hydroxycyclohexane-1-carboxylate

    • [0221]
      Methyl 1-methoxy-4-oxocyclohexanecarboxylate (4.00 g, 21.5 mmol) was dissolved in methanol (100 mL) and the solution was cooled to 0° C. Sodium borohydride (2.03 g, 53.7 mmol) was added in portions over 20 min. The reaction mixture was stirred for 30 min, then was quenched by addition of aqueous saturated NH4Cl solution. The quenched reaction mixture was evaporated to remove the MeOH, then the aqueous suspension was extracted with DCM (3×). The combined organic layers were dried over sodium sulfate, filtered, and concentrated to yield a residue that was purified by flash-column chromatography on silica gel (gradient elution, 5% to 100% ethyl acetate-hexanes) to afford methyl 1-methoxy-4-hydroxycyclohexane-1-carboxylate (2.00 g, 49.5%) as a colorless oil. MS (ES+) C9H16Orequires: 188, found: 211 [M+Na]+.

Step 2: Synthesis of methyl 1-methoxy-4-iodocyclohexane-1-carboxylate

    • [0222]
      Methyl 1-methoxy-4-hydroxycyclohexane-1-carboxylate (2.00 g, 10.6 mmol) was dissolved in THF (20 mL) and imidazole (723 mg, 10.6 mmol) and triphenylphosphine (3.34 g, 12.8 mmol) were added. The mixture was cooled to 0° C., and then a solution of iodine (3.24 g, 12.8 mmol) in THF (10 mL) was added dropwise over 15 min. The reaction mixture was allowed to warm to ambient temperature and was then stirred for 2 days, after which it was poured over saturated sodium thiosulfate solution and extracted with EtOAc. The organic layer was dried over sodium sulfate, filtered, concentrated, and the residue was triturated with hexane (40 mL, stir for 20 min). The mixture was filtered, and the filtrate was evaporated to provide a residue that was purified by flash-column chromatography on silica gel (gradient elution, 0 to 30% ethyl acetate-hexanes) to give the title compound (2.37 g, 75%) as a pale yellow oil. MS (ES+) C9H15IOrequires: 298, found: 299 [M+H]+.

B. Ethyl 1-ethoxy-4-iodocyclohexane-1-carboxylate

    • [0223]
      Figure US20170121312A1-20170504-C00105
    • [0224]
      The title compound was prepared as described above using ethyl 1-ethoxy-4-oxocyclohexane-1-carboxylate as a starting material. C11H19IOrequires: 326, found: 327 [M+H].

Example 9. Synthesis of Amine IntermediatesA. (S)-1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethan-1-amine

    • [0225]
      Figure US20170121312A1-20170504-C00106

Step 1: Synthesis of 1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethan-1-one

    • [0226]
      4-Fluoro-1H-pyrazole (4.73 g, 55 mmol) and potassium carbonate (17.27 g, 125 mmol) were combined and stirred in N,N-dimethylformamide (41.7 mL) for 10 minutes in an open sealed tube before addition of 2-bromo-5-acetylpyridine (10 g, 50 mmol). The reaction tube was sealed and stirred for 20 hours at 100° C. The reaction mixture was then cooled to room temperature and poured into water (˜700 mL). The mixture was sonicated and stirred for 20 minutes, after which a beige solid was isolated by filtration, washed with small amounts of water, and dried to yield 1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethan-1-one (9.81 g, 96% yield). MS: M+1=206.0.

Step 2: Synthesis of (R)—N—((S)-1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethyl)-2-methylpropane-2-sulfinamide

    • [0227]
      To a stirred room temperature solution of 1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethan-1-one (9.806 g, 47.8 mmol) in THF (96 mL) was added (R)-(+)-t-Butylsulfinamide (5.79 g, 47.8 mmol) followed by titanium (IV) ethoxide (21.8 g, 96 mmol). The solution was stirred at 75° C. on an oil bath for 15 hours. The reaction solution was cooled to room temperature and then to −78° C. (external temperature) before the next step. To the −78° C. solution was added dropwise over nearly 55 minutes L-Selectride (143 mL of 1N in THF, 143 mmol). During addition, some bubbling was observed. The reaction was then stirred after the addition was completed for 15 minutes at −78° C. before warming to room temperature. LC-MS of sample taken during removal from cold bath showed reaction was completed. The reaction was cooled to −50° C. and quenched slowly with methanol (˜10 mL), then poured into water (600 mL) and stirred. An off-white precipitate was removed by filtration, with ethyl acetate used for washes. The filtrate was diluted with ethyl acetate (800 mL), the layers were separated, and the organic layer was dried over sodium sulfate, filtered, and concentrated down. The crude was purified by silica gel chromatography to yield (R)—N—((S)-1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethyl)-2-methylpropane-2-sulfinamide (10.5 g, 99% purity, 70.3% yield) as a light yellow solid. MS: M+1=311.1.

Step 3: Synthesis of (S)-1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethan-1-amine

  • [0228]
    A solution of (R)—N—((S)-1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethyl)-2-methylpropane-2-sulfinamide (10.53 g, 33.9 mmol)) in methanol (79 mmol) and 4N HCl/dioxane (85 mL, 339 mmol) was stirred for 2.5 hours, at which point LC-MS showed reaction was complete. The reaction solution was poured into diethyl ether (300 mL) and a sticky solid was formed. The mixture was treated with ethyl acetate (200 mL) and sonicated. The solvents were decanted, and the sticky solid was treated with more ethyl acetate (˜200 mL), sonicated and stirred. The bulk of the sticky solid was converted to a suspension. A light yellow solid was isolated by filtration, washed with smaller amounts of ethyl acetate, and dried to yield (S)-1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethan-1-amine (7.419 g, 78% yield). LC-MS confirmed desired product in high purity. MS: M+1=207.1.

PATENT

CN 111440151

PATENT

CN 111362923

References

  1. Jump up to:a b c d e f g h i j k l m n “FDA approves pralsetinib for lung cancer with RET gene fusions”U.S. Food and Drug Administration (FDA). 4 September 2020. Retrieved 8 September 2020.  This article incorporates text from this source, which is in the public domain.
  2. ^ “Blueprint Medicines Announces FDA Approval of Gavreto (pralsetinib) for the Treatment of Adults with Metastatic RET Fusion-Positive Non-Small Cell Lung Cancer” (Press release). Blueprint Medicines. 4 September 2020. Retrieved 8 September 2020 – via PR Newswire.
  3. ^ “Roche announces FDA approval of Gavreto (pralsetinib) for the treatment of adults with metastatic RET fusion-positive non-small cell lung cancer”Roche (Press release). 7 September 2020. Retrieved 8 September 2020.
  4. Jump up to:a b c d “Drug Trial Snapshot: Gavreto”U.S. Food and Drug Administration. 4 September 2020. Retrieved 16 September 2020.  This article incorporates text from this source, which is in the public domain.

External links

Pralsetinib
Clinical data
Trade names Gavreto
Other names BLU-667
License data
Pregnancy
category
  • US: N (Not classified yet)
Routes of
administration
By mouth
Drug class Tyrosine kinase inhibitor
ATC code
  • None
Legal status
Legal status
Identifiers
CAS Number
PubChem CID
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
Chemical and physical data
Formula C27H32FN9O2
Molar mass 533.612 g·mol−1
3D model (JSmol)

Roche buys into Blueprint’s RET inhibitor

The deal positions pralsetinib to compete against Lilly’s Retevmo

by Lisa M. Jarvis
JULY 18, 2020 | APPEARED IN VOLUME 98, ISSUE 28
09828-buscon2-pral.jpg

Roche is investing $775 million in cash and equity for access to Blueprint Medicines’ oncology drug candidate pralsetinib, which is under review by the US Food and Drug Administration.

Pralsetinib is a small-molecule inhibitor of RET alterations—rare genetic fusions or mutations that occur at low levels across lung, thyroid, and many other cancers.

The drug will go up against Eli Lilly and Company’s Retevmo, an RET inhibitor that received FDA approval in May for certain lung and thyroid cancers. Lilly acquired Retevmo in its $8 billion purchase of Loxo Oncology in 2019, a deal to obtain Loxo’s pipeline of small molecules for genetically defined tumors.

But SVB Leerink analyst Andrew Berens points out that Retevmo has side effects: it can cause an irregular heart rhythm called QT prolongation and hemorrhagic events. That leaves room for pralsetinib, which Roche will be better able to get in front of oncologists, Berens argues. In addition to a vast commercial network, Roche brings diagnostic tools to help identify cancer patients whose tumors feature RET alterations.

The FDA has a deadline of Nov. 23 to decide on approving the drug for lung cancer.

Roche’s move lowers the likelihood of a takeover of Blueprint, which had appeared on many investors’ short lists of acquisition targets. “We were surprised by the profuse language framing this deal as ensuring Blueprint’s independence,” Piper Sandler stock analyst Christopher J. Raymond told investors in a note.

//////////Pralsetinib, GAVRETO, 2020 APPROVALS, FDA 2020

CC1=CC(=NN1)NC2=NC(=NC(=C2)C)C3CCC(CC3)(C(=O)NC(C)C4=CN=C(C=C4)N5C=C(C=N5)F)OC

Clascoterone


Cortexolone 17α-propionate.svg

Clascoterone

(1R,3aS,3bR,9aR,9bS,11aS)-1-(2-hydroxyacetyl)-9a,11a-dimethyl-7-oxo-1H,2H,3H,3aH,3bH,4H,5H,7H,8H,9H,9aH,9bH,10H,11H,11aH-cyclopenta[a]phenanthren-1-yl propanoate

Formula
C24H34O5
CAS
19608-29-8
Mol weight
402.5238

FDA APPROVED, 2020/8/26, Winlevi

クラスコステロン;

Anti-acne, Androgen receptor antagonist

Clascoterone, sold under the brand name Winlevi, is an antiandrogen medication which is used topically in the treatment of acne.[1][2][3] It is also under development for the treatment of androgen-dependent scalp hair loss.[2] The medication is used as a cream by application to the skin, for instance the face and scalp.[3]

Clascoterone is an antiandrogen, or antagonist of the androgen receptor (AR), the biological target of androgens such as testosterone and dihydrotestosterone.[4][5] It shows no systemic absorption when applied to skin.[3]

The medication, developed by Cassiopea and Intrepid Therapeutics,[2] was approved by the US Food and Drug Administration (FDA) for acne in August 2020.[6][7]

Medical uses

Clascoterone is indicated for the topical treatment of acne vulgaris in females and males age 12 years and older.[1][8] It is applied to the affected skin area in a dose of 1 mg cream (or 10 mg clascoterone) twice per day, once in the morning and once in the evening.[1] The medication should not be used ophthalmicallyorally, or vaginally.[1]

Available forms

Clascoterone is available in the form of a 1% (10 mg/g) cream for topical use.[1]

Contraindications

Clascoterone has no contraindications.[1]

Side effects

The incidences of local skin reactions with clascoterone were similar to placebo in two large phase 3 randomized controlled trials.[1][9] Suppression of the hypothalamic–pituitary–adrenal axis (HPA axis) may occur during clascoterone therapy in some individuals due to its cortexolone metabolite.[1][8] HPA axis suppression as measured by the cosyntropin stimulation test was observed to occur in 3 of 42 (7%) of adolescents and adults using clascoterone for acne.[1][8] HPA axis function returned to normal within 4 weeks following discontinuation of clascoterone.[1][8] Hyperkalemia (elevated potassium levels) occurred in 5% of clascoterone-treated individuals and 4% of placebo-treated individuals.[1]

Pharmacology

Pharmacodynamics

Clascoterone is an steroidal antiandrogen, or antagonist of the androgen receptor (AR), the biological target of androgens such as testosterone and dihydrotestosterone (DHT).[1][4][5] In a bioassay, the topical potency of the medication was greater than that of progesteroneflutamide, and finasteride and was equivalent to that of cyproterone acetate.[10] Likewise, it is significantly more efficacious as an antiandrogen than other AR antagonists such as enzalutamide and spironolactone in scalp dermal papilla cells and sebocytes in vitro.[5]\

Pharmacokinetics

Steady-state levels of clascoterone occur within 5 days of twice daily administration.[1] At a dosage of 6 g clascoterone cream applied twice daily, maximal circulating levels of clascoterone were 4.5 ± 2.9 ng/mL, area-under-the-curve levels over the dosing interval were 37.1 ± 22.3 h*ng/mL, and average circulating levels of clascoterone were 3.1 ± 1.9 ng/mL.[1] In rodents, clascoterone has been found to possess strong local antiandrogenic activity, but negligible systemic antiandrogenic activity when administered via subcutaneous injection.[10] Along these lines, the medication is not progonadotropic in animals.[10]

The plasma protein binding of clascoterone is 84 to 89% regardless of concentration.[1]

Clascoterone is rapidly hydrolyzed into cortexolone (11-deoxycortisol) and this compound is a possible primary metabolite of clascoterone based on in-vitro studies in human liver cells.[1][8] During treatment with clascoterone, cortexolone levels were detectable and generally below or near the low limit of quantification (0.5 ng/mL).[1] Clascoterone may also produce other metabolites, including conjugates.[1]

The elimination of clascoterone has not been fully characterized in humans.[1]

Chemistry

Clascoterone, also known as cortexolone 17α-propionate or 11-deoxycortisol 17α-propionate, as well as 17α,21-dihydroxyprogesterone 17α-propionate or 17α,21-dihydroxypregn-4-en-3,20-dione 17α-propionate, is a synthetic pregnane steroid and a derivative of progesterone and 11-deoxycortisol (cortexolone).[11] It is specifically the C17α propionate ester of 11-deoxycortisol.[10]

An analogue of clascoterone is 9,11-dehydrocortexolone 17α-butyrate (CB-03-04).[12]

History

C17α esters of 11-deoxycortisol were unexpectedly found to possess antiandrogenic activity.[10] Clascoterone, also known as cortexolone 17α-propionate, was selected for development based on its optimal drug profile.[10] The medication was approved by the US Food and Drug Administration (FDA) for the treatment of acne in August 2020.[6]

Two large phase 3 randomized controlled trials evaluated the effectiveness of clascoterone for the treatment of acne over a period of 12 weeks.[1][8][9] Clascoterone decreased acne symptoms by about 8 to 18% more than placebo.[1][9] The defined treatment success endpoint was achieved in about 18 to 20% of individuals with clascoterone relative to about 7 to 9% of individuals with placebo.[1][8][9] The comparative effectiveness of clascoterone between males and females was not described.[1][9]

A small pilot randomized controlled trial in 2011, found that clascoterone cream decreased acne symptoms to a similar or significantly greater extent than tretinoin 0.05% cream.[8][13] No active comparator was used in the phase III clinical trials of clascoterone for acne.[8] Hence, it’s unclear how clascoterone compares to other therapies used in the treatment of acne.[8]

The FDA approved clascoterone based on evidence from two clinical trials (Trial 1/NCT02608450 and Trial 2/NCT02608476) of 1440 participants 9 to 58 years of age with acne vulgaris.[14] The trials were conducted at 99 sites in the United States, Poland, Romania, Bulgaria, Ukraine, Georgia, and Serbia.[14]

Participants applied clascoterone or vehicle (placebo) cream twice daily for 12 weeks.[14] Neither the participants nor the health care providers knew which treatment was being given until after the trial was completed.[14] The benefit of clascoterone in comparison to placebo was assessed after 12 weeks of treatment using the Investigator’s Global Assessment (IGA) score that measures the severity of disease (on a scale from 0 to 4) and a decrease in the number of acne lesions.[14]

Society and culture

Names

Clascoterone is the generic name of the drug and its INN and USAN.[11][15]

Research

Clascoterone has been suggested as a possible treatment for hidradenitis suppurativa (acne inversa), an androgen-dependent skin condition.[16]

………………………………………………………………………….

PATENT

CN 112028956

https://patents.google.com/patent/CN112028956A/en

Example 1
Preparation of intermediate I
Wherein R is DMTr
Dissolving the compound 11-deoxycortisol (1.04g, 3.0mmol, 1eq.) in 10mL of anhydrous pyridine, dissolving dried DMTrCl (1.2-1.5eq) in 5mL of anhydrous dichloromethane, dropwise adding a dichloromethane solution of DMTrCl into the reactant solution at room temperature, and reacting for 4 hours at room temperature; the reaction was quenched with methanol and the solvent was evaporated to dryness with an oil pump to give intermediate I in 85% yield (the next reaction was carried out without work-up, the solvent environment and catalyst were similar to the reaction of this step).
1 H NMR (600MHz, CDCl 3 ) (ppm) 7.25-7.31 (m, 5H, H-DMTr), 7.15-7.18 (m, 4H, H-DMTr), 6.81-6.84 (m, 4H, H-DMTr), 5.73(1H,s,H-4),4.65(1H,dd,J=19.8,4.8Hz,H-21),4.30(1H,dd,J=19.8,4.8Hz,H-21),3.80(6H ,s),2.71(s,1H,17-OH),2.66-2.71(m,1H,H-16β),2.27-2.45(m,4H),1.19(3H,s,H-19),0.96- 1.87(m, 14H), 0.72(s, 3H, H-18).
MS + 303(DMTr protecting group fragment), 649[M + H] +
Melting point: 95-97 deg.C
Example 2:
preparation of intermediate II
Wherein R is DMTr
Under the protection of nitrogen, dissolving the intermediate product I (1eq.) in 5mL of anhydrous dichloromethane, adding DMAP (0.1eq.) into the solution, dropwise adding triethylamine (1.2eq.) and propionic anhydride or propionyl chloride (1.2eq. ), reacting at 40 ℃ for 12 hours after dropwise adding, and evaporating the solvent to obtain an intermediate product II.
Or under the protection of nitrogen, dissolving the intermediate product I (1eq.) in 5mL of anhydrous pyridine, adding DMAP (0.1eq.) into the solution, dropwise adding triethylamine (1.2eq.) and propionic anhydride or propionyl chloride (1.2eq .), reacting at 80 ℃ for 4 hours after dropwise adding, and evaporating the solvent to obtain an intermediate product II. (the reaction in the step can be directly carried out for the next step of removing DMTr protecting group to obtain the reaction after solvent evaporation without strict purification post-treatment)
1 H NMR (600MHz, CDCl 3 ) (ppm) 7.26-7.32 (m, 5H, H-DMTr), 7.14-7.18 (m, 4H, H-DMTr), 6.81-6.84 (m, 4H, H-DMTr), 5.72(1H,s,H-4),4.65(1H,dd,J=19.8,4.8Hz,H-21),4.30(1H,dd,J=19.8,4.8Hz,H-21),3.81(6H ,s),2.66-2.71(m,1H,H-16β),2.35(m,2H,-CH 2 CH 3 ),2.27-2.45(m,4H),1.19(3H,s,H-19), 1.15 (t, 3H, J=7.8Hz, -CH 2 CH 3 ), 0.96-1.87 (m, 14H), 0.72 (s, 3H, H-18);
MS + :303(DMTr protecting group fragment), 727[ M + Na [)] + ,768[M+Na+CH 3 CN] + .
Example 3:
preparation of target Compound 1 (21-hydroxy-17- (1-oxopropoxy) pregn-4-ene-3, 20-dione)
Dissolving the concentrated intermediate product II in an ethyl acetate solution, slowly dropwise adding 0.5M hydrochloric acid solution or 2% trifluoroacetic acid-ethyl acetate solution at 0 ℃, reacting for 5 minutes at 0 ℃, removing DMTr protective groups, adding 5% sodium bicarbonate aqueous solution at 0 ℃, stirring, neutralizing acid in a reaction system, washing an ethyl acetate organic layer twice by using 5% sodium bicarbonate aqueous solution, removing acid and other water-soluble impurities in the ethyl acetate organic layer, drying the ethyl acetate organic layer by anhydrous sodium sulfate, evaporating to remove part of ethyl acetate solvent, adding petroleum ether into the remaining small amount of ethyl acetate solution, and recrystallizing in a system with 10 times of solvent amount of ethyl acetate-petroleum ether (5:1) to obtain a target product with high purity of 90%. The total yield from 11-deoxycortisol is up to 70%. The final product was free of isomerized by-products by HPLC and was not found by LCMS.
1 H NMR (600 MHz, CDCl 3 ) (ppm): 5.75 (s, 1H, H-4), 4.28 (d, 1H, J=18.0 Hz, H-21), 4.23 (d, 1H, J=18.0 Hz, H-21), 3.05(s, 1H, 21-OH), 2.81-2.86(m, 1H, H-16β), 2.34-2.46(m, 3H), 2.35(m, 2H, -CH 2 CH 3 ) ,2.28-2.33(m,1H),2.03-2.07(m,1H),1.86-1.94(m,2H),1.67-1.77(m,3H),1.55-1.64(m,3H),1.35-1.46( m, 3H), 1.19(s, 3H, H-19), 1.15(t, 3H, J=7.8Hz, -CH 2 CH 3 ), 1.08-1.11(m, 1H), 1.00-1.05(m, 1H ),0.69(s,3H,H-18);
MS + : 403[M+H] + , 444[M+H+CH 3 CN] +
Melting point: 128-130 ℃.
str1
PATENT
WO 2009019138,
EXAMPLES Example 1
Alcoho lysis with CCL of cortexolone 17α, 21-dipropionate
Add butanol (0.4g, 5.45 mmoles) and CCL (17.4g, 3.86 U/mg, FLUKA) to a solution of cortexolone- 17α,21-dipropionate (0.5g, 1.09 mmoles) in toluene (50ml). Maintain the mixture under stirring, at 30 0C, following the progress of the reaction in TLC (Toluene/ethyl acetate 6/4) until the initial material is dissolved (24h). Remove the enzyme by means of filtration using a Celite layer. Recover the cortexolone 17α-propionate (0.437, 99%) after evaporation under low pressure. Through crystallisation, from diisopropyl ether you obtain a product with a purity >99% in HPLC.
1 H-NMR (500MHz, CDCl3) relevant signals δ (ppm) 5.78 (br s, 1 H, H-4), 4.32 (dd, 1 H, H-21), 4.25 (dd, IH, H-21), 1.22 (s, 3H, CH3-19), 1.17 (t, 3H, CH3), 0.72 (s, 3H5 CH3-18). P.f. 114 0C Example 2
According to the method described in example 1 prepare cortexolone- 17α- butanoate.
1H-NMR relevant signals δ (ppm) 5.78 (br s, IH, H-4), 4.32 (dd, IH, H-21), 4.26 (dd, IH, H-21), 1.23 (s, 3H, CH3-19), 0.97 (t, 3H, CH3), 0.73 (s, 3H, CH3-18). P.F. 134-136 0C
Example 3
According to the method described in the example prepare cortexolone- 17α- valerate.
1H-NMR relevant signals δ (ppm) 5.77 (br s, IH, H-4), 4.32 (dd, IH, H-21), 4.26
(dd, IH, H-21), 1.22 (s, 3H, CH3-19), 0.95 (t, 3H, CH3), 0.72 (s, 3H, CH3-18). P.f.
114 0C (diisopropyl ether).
Example 4
According to the method described in the example prepare 9,11-dehydro- cortexolone- 17α-butanoate.
1 H-NMR relevant signals δ (ppm) 5.77 (br s, IH, H-4), 5.54 (m, IH, H-9), 4.29
(dd, IH, H-21), 4.24 (dd, IH, H-21), 1.32 (s, 3H, CH3-19), 0.94(t, 3H, CH3), 0.68
(s, 3H, CH3– 18). P.f. 135-136 0C (acetone/hexane).
Example 5
Alcoho lysis with CALB of cortexolone- 17α, 21-dipropionate
Dissolve cortexolone, 17α, 2-dipropionate (0.5g, 1 .09 mmoles) in acetonitrile
(40ml), add CALB (2.3g, 2.5 U/mg Fluka) and octanol (0.875ml). Leave the mixture under stirring, at 30 0C, for 76 hrs. Remove the enzyme by means of filtration using a paper filter. Once the solvents evaporate, recover a solid
(0.4758) which upon analysis 1H-NMR shall appear made up of cortexolone- 17α- propionate at 91%.
Example 6
Crystallisation
Add the solvent (t-butylmethylether or diisopropylether) to the sample according to the ratios indicated in Table 3. Heat the mixture to the boiling temperature of the solvent, under stirring, until the sample dissolves completely. Cool to room temperature and leave it at this temperature, under stirring, for 6 hours. Filter using a buchner funnel and maintain the solid obtained, under low pressure, at a room temperature for 15 hours and then, at 400C, for 5 hours.
Example 7
Precipitation Disslove the sample in the suitable solvent (dichloromethane, acetone, ethyl acetate or ethanol) according to the ratios indicated in table 3 and then add the solvent, hexane or water, according to the ratios indicated in table 3, maintaining the mixture, under stirring, at room temperature. Recover the precipitate by filtration using a buchner funnel and desiccate as in example 6. Example 8.
Obtaining a pharmaceutical form containing the medication in a defined crystalline form.
Prepare a fluid cream containing 2 % cetylic alcohol, 16% glyceryl monostearate, 10% vaseline oil, 13 % propylene glycol, 10% poly ethylengly col with low polymerization 1.5% polysorbate 80 and 47.5 % purified water. Add 1 g of cortexolone 17α-propionate of crystalline form III to 100 g of this cream and subject the mixture to homogenisation by means of a turbine agitator until you obtain homogeneity. You obtain a cream containing a fraction of an active ingredient dissolved in the formulation vehicle and a non-dissolved fraction of an active ingredient, present as a crystal of crystalline form III. This preparation is suitable for use as a formulation vehicle for skin penetration tests on Franz cells, where a coefficient of penetration in the range of 0.04 to 0.03 cm/h is observed on the preparation. Example 9.
Obtaining the pharmaceutical form containing the medication in solvate form IV for replacing the solvent during the galenic formulation procedure Dissolve lOOg of cortexolone 17α-propionate of crystalline form III in 2500 g of propylene glycol under stirring at room temperature. Separately prepare, by using a turbo emulsifϊer raising the temperature up to about 700C, an emulsion with 250 g of Cetylic alcohol, 1500 g of glyceryl monostearate, 1000 g of liquid paraffin, 5 g of mixed tocopherols, 100 g of polysorbate 80 and 4650 g of water. After cooling the emulsion up to about 300C, add – under stirring and under negative pressure – the cortexolone 17α-propionate solution in propylene glycol. Maintain the emulsioned cream under stirring until you obtain homogeneity, making sure the temperature remains low by means the circulation of a coolant. The cream contains a dispersed crystalline fraction, made up of an active ingredient in solvate crystalline form IV, formed due to the precipitation of the active ingredient itself from the glycolic solution which contained it when the latter was added to the predominantly aqueous formulation. The DRX spectra of the crystalline form present in the cream are indicated in Fig. 30.
PAPER
Tetrahedron Letters, 49(31), 4610-4612; 2008

Abstract

Several 17α-monoesters of cortexolone and its Δ9-derivative are endowed with antiandrogenic activity. Their synthesis can be accomplished by means of a lipase-catalyzed chemoselective alcoholysis of the corresponding 17α,21-diesters.

Graphical abstract

1H NMR (500 MHz, CDCl3): selected data δ 5.78 (br s, 1H, H-4), 4.32 (dd, 1H, H-21, J18.3 and 4.9 Hz), 4.25 (dd, 1H, H-21, J18.3 and 4.9 Hz), 1.22 (s, 3H, CH3-19), 1.17 (t, 3H, CH3, J7.6 Hz), 0.72 (s, 3H, CH3-18) MP 133 °C (t-butylmethylether)

…………………………………………………………………..

PATENT

https://patents.google.com/patent/EP2503005B1/en

  • Cortexolone derivatives in which the hydroxyl group at position C-17α is esterified with short chain aliphatic or aromatic acids and the derivatives of the corresponding 9,11-dehydro derivative, are known to have an antiandrogenic effect.
  • [0002]
    EP 1421099 describes cortexolone 17α-propionate and 9,11-dehydro-cortexolone-17-α-butanoate regarding a high antiandrogenic biological activity demonstrated both “in vitro” and “in vivo” on the animal.
  • [0003]
    US3530038 discloses the preparation of a crystalline form of cortexolone-17α-propionate having a melting point of 126-129 °C and an IR spectrum with bands at (cm-1): 3500, 1732, 1713, 1655 and 1617.
  • [0004]
    A method for obtaining the above mentioned derivatives is described by Gardi et al. (Gazz. Chim. It. 63, 43 1,1963) and in the United States patent US3152154 providing for the transformation of cortexolone, or transformation of 9,11-dehydrocortexolone, in the intermediate orthoester using orthoesters available in the market as a mixture of aprotic solvents such as cyclohexane and DMF, in presence of acid catalysis (ex. PTSA.H20). The intermediate orthoester thus obtained can be used as is or upon purification by suspension in a solvent capable of solubilising impurities, preferably in alcohols. The subsequent hydrolysis in a hydroalcoholic solution, buffered to pH 4-5 preferably in acetate buffer, provides the desired monoester.
  • [0005]

    Such synthesis is indicated in the diagram 1 below

    Figure imgb0001
  • [0006]
    However, the monoesters thus obtained were, in the reaction conditions, unstable and, consequently hard to manipulate and isolate (R. Gardi et al Tetrahedron Letters, 448, 1961). The instability is above all due to the secondary reaction of migration of the esterifying acyl group from position 17 to position 21.
  • [0007]
    It is thus known that in order to obtain the above mentioned monoesters with a chemical purity in such a manner to be able to proceed to the biological tests, it is necessary to use, at the end of the synthesis, a purification process which is generally performed by means of column chromatography.
  • [0008]
    Furthermore, US3152154 describes how the hydrolysis of the diester in a basic environment is not convenient due to the formation of a mixture of 17α,21-diol, of 17- and 21 -monoesters, alongside the initial non-reacted product.
  • [0009]
    Now, it has been surprisingly discovered that an alcoholysis reaction using a lipase from Candida as a biocatalyst can be usefully applied during the preparation of 17α monoesters of cortexolone, or its 9,11-dehydroderivatives.
  • [0010]

    As a matter of fact, it has been discovered that such enzymatic alcoholysis of the 17,21-diester of the cortexolone, or of its derivative 9,11-dehydro, selectively occurs in position 21 moving to the corresponding monoester in position 17, as shown in diagram 2 below:

    Figure imgb0002
  • [0011]
    The chemoselectivity of the special enzymatic reaction in alcoholysis conditions, according to the present invention, opens new perspectives for preparation, at industrial level with higher yields, of 17α-monoesters with respect to the methods already indicated in literature.
  • [0012]
    The diesters serving as a substrate for the reaction of the invention can be prepared according to the prior art, for example following the one described in B.Turner, (Journal of American Chemical Society, 75, 3489, 1953) which provides for the esterification of corticosteroids with a linear carboxylic acid in presence of its anhydride and PTSA monohydrate.

EXAMPLES

    • Example 1

Alcoholysis with CCL of cortexolone 17α, 21-dipropionate

      • [0055]
        Add butanol (0.4g, 5.45 mmoles) and CCL (17.4g, 3.86 U/mg, FLUKA) to a solution of cortexolone-17α,21-dipropionate (0.5g, 1.09 mmoles) in toluene (50ml). Maintain the mixture under stirring, at 30 °C, following the progress of the reaction in TLC (Toluene/ethyl acetate 6/4) until the initial material is dissolved (24h). Remove the enzyme by means of filtration using a Celite layer. Recover the cortexolone 17α-propionate (0.437, 99%) after evaporation under low pressure. Through crystallisation, from diisopropyl ether you obtain a product with a purity >99% in HPLC.
      • [0056]
        1H-NMR (500MHz, CDCl3) relevant signals δ (ppm) 5.78 (br s, 1 H, H-4), 4.32 (dd, 1 H, H-21), 4.25 (dd, 1H, H-21), 1.22 (s, 3H, CH3-19), 1.17 (t, 3H, CH3), 0.72 (s, 3H, CH3-18). P.f. 114 °C

Example 2 (comparative)

      • [0057]
        According to the method described in example 1 prepare cortexolone-17α-butanoate.
      • [0058]
        1H-NMR relevant signals δ (ppm) 5.78 (br s, 1H, H-4), 4.32 (dd, 1H, H-21), 4.26 (dd, 1H, H-21), 1.23 (s, 3H, CH3-19), 0.97 (t, 3H, CH3), 0.73 (s, 3H. CH3-18). P.F. 134-136 °C

Example 3 (comparative)

According to the method described in the example prepare cortexolone-17α-valerate.

      • [0059]
        1H-NMR relevant signals δ (ppm) 5.77 (br s, 1H, H-4), 4.32 (dd, 1H, H-21), 4.26 (dd, 1H, H-21), 1.22 (s, 3H, CH3-19), 0.95 (t, 3H, CH3), 0.72 (s, 3H, CH3-18). P.f. 114 °C (diisopropyl ether).

Example 4 (comparative)

According to the method described in the example prepare 9, 11-dehydro-cortexolone-17α-butanoate.

      • [0060]
        1H-NMR relevant signals δ (ppm) 5.77 (br s, 1H, H-4), 5.54 (m, 1H, H-9), 4.29 (dd, 1H, H-21), 4.24 (dd, 1H, H-21), 1.32 (s, 3H, CH3-19), 0.94(t, 3H, CH3), 0.68 (s, 3H, CH3-18). P.f. 135-136 °C (acetone/hexane).

Example 5

Alcoholysis with CALB of cartexolone-17α, 21-dipropionate

      • [0061]
        Dissolve cortexolone, 17α, 2-dipropionate (0.5g, 1.09 mmoles) in acetonitrile (40ml), add CALB (2.3g, 2.5 U/mg Fluka) and octanol (0.875ml). Leave the mixture under stirring, at 30 °C, for 76 hrs. Remove the enzyme by means of filtration using a paper filter. Once the solvents evaporate, recover a solid (0.4758) which upon analysis 1H-NMR shall appear made up of cortexolone-17α-propionate at 91%.

Example 6

Crystallisation

      • [0062]
        Add the solvent (t-butylmethylether or diisopropylether) to the sample according to the ratios indicated in Table 3. Heat the mixture to the boiling temperature of the solvent, under stirring, until the sample dissolves completely. Cool to room temperature and leave it at this temperature, under stirring, for 6 hours. Filter using a buchner funnel and maintain the solid obtained, under low pressure, at a room temperature for 15 hours and then, at 40°C, for 5 hours.

Example 7 (comparative)

Precipitation

      • [0063]
        Disslove the sample in the suitable solvent (dichloromethane, acetone, ethyl acetate or ethanol) according to the ratios indicated in table 3 and then add the solvent, hexane or water, according to the ratios indicated in table 3, maintaining the mixture, under stirring, at room temperature. Recover the precipitate by filtration using a buchner funnel and desiccate as in example 6.

Example 8.

Obtaining a pharmaceutical form containing the medication in a defined crystalline form.

  • [0064]
    Prepare a fluid cream containing 2 % cetylic alcohol, 16% glyceryl monostearate, 10% vaseline oil, 13 % propylene glycol, 10% polyethylenglycol with low polymerization 1.5% polysorbate 80 and 47.5 % purified water. Add 1 g of cortexolone 17α-propionate of crystalline form III to 100 g of this cream and subject the mixture to homogenisation by means of a turbine agitator until you obtain homogeneity. You obtain a cream containing a fraction of an active ingredient dissolved in the formulation vehicle and a non-dissolved fraction of an active ingredient, present as a crystal of crystalline form III. This preparation is suitable for use as a formulation vehicle for skin penetration tests on Franz cells, where a coefficient of penetration in the range of 0.04 to 0.03 cm/h is observed on the preparation.

References

  1. Jump up to:a b c d e f g h i j k l m n o p q r s t u v w “Winlevi (clascoterone) cream, for topical use”(PDF). Cassiopea. Retrieved 9 September 2020.
  2. Jump up to:a b c http://adisinsight.springer.com/drugs/800026561
  3. Jump up to:a b c Kircik LH (July 2019). “What’s new in the management of acne vulgaris”Cutis104(1): 48–52. PMID 31487336.
  4. Jump up to:a b Rosette C, Rosette N, Mazzetti A, Moro L, Gerloni M (February 2019). “Cortexolone 17α-Propionate (Clascoterone) is an Androgen Receptor Antagonist in Dermal Papilla Cells In Vitro”. J Drugs Dermatol18 (2): 197–201. PMID 30811143.
  5. Jump up to:a b c Rosette C, Agan FJ, Mazzetti A, Moro L, Gerloni M (May 2019). “Cortexolone 17α-propionate (Clascoterone) Is a Novel Androgen Receptor Antagonist that Inhibits Production of Lipids and Inflammatory Cytokines from Sebocytes In Vitro”. J Drugs Dermatol18 (5): 412–418. PMID 31141847.
  6. Jump up to:a b “Cassiopea Receives FDA Approval for Winlevi (clascoterone cream 1%), First-in-Class Topical Acne Treatment Targeting the Androgen Receptor”Cassiopea (Press release). Retrieved 2020-08-30.
  7. ^ “Winlevi: FDA-Approved Drugs”U.S. Food and Drug Administration (FDA). Retrieved 9 September 2020.
  8. Jump up to:a b c d e f g h i j Barbieri, John S. (2020). “A New Class of Topical Acne Treatment Addressing the Hormonal Pathogenesis of Acne”. JAMA Dermatology156 (6): 619–620. doi:10.1001/jamadermatol.2020.0464ISSN 2168-6068PMID 32320045.
  9. Jump up to:a b c d e Hebert A, Thiboutot D, Stein Gold L, Cartwright M, Gerloni M, Fragasso E, Mazzetti A (April 2020). “Efficacy and Safety of Topical Clascoterone Cream, 1%, for Treatment in Patients With Facial Acne: Two Phase 3 Randomized Clinical Trials”JAMA Dermatol156 (6): 621–630. doi:10.1001/jamadermatol.2020.0465PMC 7177662PMID 32320027.
  10. Jump up to:a b c d e f Celasco G, Moro L, Bozzella R, Ferraboschi P, Bartorelli L, Quattrocchi C, Nicoletti F (2004). “Biological profile of cortexolone 17alpha-propionate (CB-03-01), a new topical and peripherally selective androgen antagonist”. Arzneimittelforschung54 (12): 881–6. doi:10.1055/s-0031-1297043PMID 15646372.
  11. Jump up to:a b https://chem.nlm.nih.gov/chemidplus/rn/19608-29-8
  12. ^ Celasco G, Moroa L, Bozzella R, Ferraboschi P, Bartorelli L, Di Marco R, Quattrocchi C, Nicoletti F (2005). “Pharmacological profile of 9,11-dehydrocortexolone 17alpha-butyrate (CB-03-04), a new androgen antagonist with antigonadotropic activity”. Arzneimittelforschung55 (10): 581–7. doi:10.1055/s-0031-1296908PMID 16294504.
  13. ^ Trifu V, Tiplica GS, Naumescu E, Zalupca L, Moro L, Celasco G (2011). “Cortexolone 17α-propionate 1% cream, a new potent antiandrogen for topical treatment of acne vulgaris. A pilot randomized, double-blind comparative study vs. placebo and tretinoin 0·05% cream”. Br. J. Dermatol165 (1): 177–83. doi:10.1111/j.1365-2133.2011.10332.xPMID 21428978S2CID 38404925.
  14. Jump up to:a b c d e “Drug Trial Snapshot: Winlevi”U.S. Food and Drug Administration (FDA). 26 August 2020. Retrieved 10 September 2020.  This article incorporates text from this source, which is in the public domain.
  15. ^ World Health Organization (2019). “International nonproprietary names for pharmaceutical substances (INN): recommended INN: list 82”. WHO Drug Information33 (3): 106. hdl:10665/330879.
  16. ^ Der Sarkissian SA, Sun HY, Sebaratnam DF (August 2020). “Cortexolone 17 α-proprionate for hidradenitis suppurativa”. Dermatol Ther: e14142. doi:10.1111/dth.14142PMID 32761708.

External links

  • “Clascoterone”Drug Information Portal. U.S. National Library of Medicine.
  • Clinical trial number NCT02608450 for “A Study to Evaluate the Safety and Efficacy of CB-03-01 Cream, 1% in Subjects With Facial Acne Vulgaris (25)” at ClinicalTrials.gov
  • Clinical trial number NCT02608476 for “A Study to Evaluate the Safety and Efficacy of CB-03-01 Cream, 1% in Subjects With Facial Acne Vulgaris (26)” at ClinicalTrials.gov
Clascoterone
Cortexolone 17α-propionate.svg
Clinical data
Trade names Winlevi
Other names CB-03-01; Breezula; 11-Deoxycortisol 17α-propionate; 17α-(Propionyloxy)-
deoxycorticosterone; 21-Hydroxy-3,20-dioxopregn-4-en-17-yl propionate
License data
Routes of
administration
Topical (cream)
ATC code
  • None
Legal status
Legal status
Identifiers
CAS Number
PubChem CID
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
CompTox Dashboard (EPA)
ECHA InfoCard 100.210.810 Edit this at Wikidata
Chemical and physical data
Formula C24H34O5
Molar mass 402.531 g·mol−1
3D model (JSmol)

/////////Clascoterone, クラスコステロン , FDA 2020, 2020 APPROVALS, ANTI ACNE

[H][C@@]12CC[C@](OC(=O)CC)(C(=O)CO)[C@@]1(C)CC[C@@]1([H])[C@@]2([H])CCC2=CC(=O)CC[C@]12C

Somapacitan, ソマパシタン;


FPTIPLSRLF DNAMLRAHRL HQLAFDTYQE FEEAYIPKEQ KYSFLQNPQT SLCFSESIPT
PSNREETQQK SNLELLRISL LLIQSWLEPV QFLRSVFANS CVYGASDSNV YDLLKDLEEG
IQTLMGRLED GSPRTGQIFK QTYSKFDTNS HNDDALLKNY GLLYCFRKDM DKVETFLRIV
QCRSVEGSCG F
(Disulfide bridge: 53-165, 182-189)

Somapacitan.png

2D chemical structure of 1338578-34-9

Somapacitan

FDA APPROVED, 2020/8/28, SOGROYA

Growth hormone (GH) receptor agonist

CAS: 1338578-34-9

(2S)-5-[2-[2-[2-[[(2S)-1-amino-6-[[2-[(2R)-2-amino-2-carboxyethyl]sulfanylacetyl]amino]-1-oxohexan-2-yl]amino]-2-oxoethoxy]ethoxy]ethylamino]-2-[[(4S)-4-carboxy-4-[[2-[2-[2-[4-[16-(2H-tetrazol-5-yl)hexadecanoylsulfamoyl]butanoylamino]ethoxy]ethoxy]acetyl]amino]butanoyl]amino]-5-oxopentanoic acid

Formula
C1038H1609N273O319S9
Mol weight
23305.1048
JAP ソマパシタン;

Treatment of growth hormone dificiency
albumin-binding growth hormone

UNII-8FOJ430U94

8FOJ430U94

NN8640

UNII-F1 component VTUYEWRWJTWXPQ-IWWWZYECSA-N

Q27270325

Somapacitan, also known as NNC0195-0092,3 is a growth hormone analog indicated to treat adults with growth hormone deficiency.2,6 This human growth hormone analog differs by the creation of an albumin binding site, and prolonging the effect so that it requires weekly dosing rather than daily.5

Somapacitan was granted FDA approval on 28 August 2020.7

Somapacitan

Somapacitan, sold under the brand name Sogroya, is a growth hormone medication.[2] Somapacitan is a human growth hormone analog.[1] Somapacitan-beco is produced in Escherichia coli by recombinant DNA technology.[1]

The most common side effects include: back pain, joint paint, indigestion, a sleep disorder, dizziness, tonsillitis, swelling in the arms or lower legs, vomiting, adrenal insufficiency, hypertension, increase in blood creatine phosphokinase (a type of enzyme), weight increase, and anemia.[2]

It was approved for medical use in the United States in August 2020.[2][3][4]

Somapacitan (Sogroya) is the first human growth hormone (hGH) therapy that adults only take once a week by injection under the skin; other FDA-approved hGH formulations for adults with growth hormone deficiency must be administered daily.[2]

Medical uses

Somapacitan is indicated for replacement of endogenous growth hormone in adults with growth hormone deficiency.[2]

Contraindications

Somapacitan should not be used in people with active malignancy, any stage of diabetic eye disease in which high blood sugar levels cause damage to blood vessels in the retina, acute critical illness, or those with acute respiratory failure, because of the increased risk of mortality with use of pharmacologic doses of somapacitan in critically ill individuals without growth hormone deficiency.[2]

History

Somapacitan was evaluated in a randomized, double-blind, placebo-controlled trial in 300 particpants with growth hormone deficiency who had never received growth hormone treatment or had stopped treatment with other growth hormone formulations at least three months before the study.[2] Particpants were randomly assigned to receive injections of weekly somapacitan, weekly placebo (inactive treatment), or daily somatropin, an FDA-approved growth hormone.[2] The effectiveness of somapacitan was determined by the percentage change of truncal fat, the fat that is accumulated in the trunk or central area of the body that is regulated by growth hormone and can be associated with serious medical issues.[2]

At the end of the 34-week treatment period, truncal fat decreased by 1.06%, on average, among particpants taking weekly somapacitan while it increased among particpants taking the placebo by 0.47%.[2] In the daily somatropin group, truncal fat decreased by 2.23%.[2] Particpants in the weekly somapacitan and daily somatropin groups had similar improvements in other clinical endpoints.[2]

It was approved for medical use in the United States in August 2020.[2][4] The U.S. Food and Drug Administration (FDA) granted the approval of Sogroya to Novo Nordisk, Inc.[2][4]

References

  1. Jump up to:a b c d “Sogroya (somapacitan-beco) injection, for subcutaneous use” (PDF). Retrieved 1 September 2020.
  2. Jump up to:a b c d e f g h i j k l m n o “FDA approves weekly therapy for adult growth hormone deficiency”U.S. Food and Drug Administration (FDA) (Press release). 1 September 2020. Retrieved 1 September 2020.  This article incorporates text from this source, which is in the public domain.
  3. ^ “FDA approves once-weekly Sogroya for the treatment of adult growth hormone deficiency”Novo Nordisk (Press release). 28 August 2020. Retrieved 1 September 2020.
  4. Jump up to:a b c “Sogroya: FDA-Approved Drugs”U.S. Food and Drug Administration (FDA). Retrieved 2 September 2020.

External links

Somapacitan
Clinical data
Trade names Sogroya
Other names somapacitan-beco, NNC0195-0092
License data
Routes of
administration
Subcutaneous[1]
Drug class Human growth hormone analog
ATC code
  • None
Legal status
Legal status
Identifiers
CAS Number
PubChem CID
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
Chemical and physical data
Formula C1038H1609N273O319S9
Molar mass 23305.42 g·mol−1

ClinicalTrials.gov

CTID Title Phase Status Date
NCT01706783 A Trial Investigating the Safety, Tolerability, Availability and Distribution in the Body of Once-weekly Long-acting Growth Hormone (Somapacitan) Compared to Once Daily Norditropin NordiFlex® in Adults With Growth Hormone Deficiency Phase 1 Completed 2018-05-25
NCT01973244 A Trial Investigating Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of a Single Dose of Long-acting Growth Hormone (Somapacitan) Compared to Daily Dosing of Norditropin® SimpleXx® in Children With Growth Hormone Deficiency Phase 1 Completed 2018-05-25
NCT02962440 A Trial Investigating the Absorption, Metabolism and Excretion of Somapacitan After Single Dosing in Healthy Male Subjects Phase 1 Completed 2017-06-07
CTID Title Phase Status Date
NCT02616562 Investigating Efficacy and Safety of Once-weekly NNC0195-0092 (Somapacitan) Treatment Compared to Daily Growth Hormone Treatment (Norditropin® FlexPro®) in Growth Hormone Treatment naïve Pre-pubertal Children With Growth Hormone Deficiency Phase 2 Recruiting 2020-03-25
NCT03075644 A Trial to Evaluate the Safety of Once Weekly Dosing of Somapacitan (NNC0195-0092) and Daily Norditropin® FlexPro® for 52 Weeks in Previously Human Growth Hormone Treated Japanese Adults With Growth Hormone Deficiency Phase 3 Completed 2019-10-18
NCT03905850 A Study to Compare the Uptake Into the Blood of Two Strengths of Somapacitan After Injection Under the Skin in Healthy Subjects Phase 1 Completed 2019-08-06
NCT03212131 Investigation of Pharmacokinetics, Pharmacodynamics, Safety and Tolerability of Multiple Doses of Somapacitan in Subjects With Mild and Moderate Degrees of Hepatic Impairment Compared to Subjects With Normal Hepatic Function. Phase 1 Completed 2019-05-24
NCT01514500 First Human Dose Trial of NNC0195-0092 (Somapacitan) in Healthy Subjects Phase 1 Completed 2018-05-25
CTID Title Phase Status Date
NCT03811535 A Research Study in Children With a Low Level of Hormone to Grow. Treatment is Somapacitan Once a Week Compared to Norditropin® Once a Day Phase 3 Recruiting 2020-09-03
NCT03878446 A Research Study in Children Born Small and Who Stayed Small. Treatment is Somapacitan Once a Week Compared to Norditropin® Once a Day Phase 2 Recruiting 2020-08-27
NCT02382939 A Trial to Compare the Safety of Once Weekly Dosing of Somapacitan With Daily Norditropin® FlexPro® for 26 Weeks in Previously Human Growth Hormone Treated Adults With Growth Hormone Deficiency Phase 3 Completed 2020-07-09
NCT02229851 Trial to Compare the Efficacy and Safety of NNC0195-0092 (Somapacitan) With Placebo and Norditropin® FlexPro® (Somatropin) in Adults With Growth Hormone Deficiency. Phase 3 Completed 2020-07-07
NCT03186495 Investigation of Pharmacokinetics, Pharmacodynamics, Safety and Tolerability of Multiple Doses of Somapacitan in Subjects With Various Degrees of Impaired Renal Function Compared to Subjects With Normal Renal Function Phase 1 Completed 2020-04-17

EU Clinical Trials Register

EudraCT Title Phase Status Date
2018-000232-10 A dose-finding trial evaluating the effect and safety of once-weekly treatment of somapacitan compared to daily Norditropin® in children with short stature born small for gestational age with no catch-up growth by 2 years of age or older Phase 2 Ongoing, Prematurely Ended 2019-05-15
2015-000531-32 A randomised, multinational, active-controlled,(open-labelled), dose finding, (double-blinded), parallel group trial investigating efficacy and safety of once-weekly NNC0195-0092 treatment compared to daily growth hormone treatment (Norditropin® FlexPro®) in growth hormone treatment naïve pre-pubertal children with growth hormone deficiency Phase 2 Ongoing, Completed 2015-12-10
2014-000290-39 A multicentre, multinational, randomised, open-labelled, parallel-group, active-controlled trial to compare the safety of once weekly dosing of NNC0195-0092 with daily Norditropin® FlexPro® for 26 weeks in previously human growth hormone treated adults with growth hormone deficiency Phase 3 Completed 2014-11-07
2013-002892-16 A multicentre, multinational, randomised, parallel-group, placebo-controlled (double blind) and active-controlled (open) trial to compare the efficacy and safety of once weekly dosing of NNC0195-0092 with once weekly dosing of placebo and daily Norditropin® FlexPro® in adults with growth hormone deficiency for 35 weeks, followed by a 53-week open-label extension period Phase 3 Completed 2014-10-07
2018-000231-27 A trial comparing the effect and safety of once weekly dosing of somapacitan with daily Norditropin® in children with growth hormone deficiency Phase 3 Ongoing

EU Clinical Trials Register

EudraCT Title Phase Status Date
2013-000013-20 A randomised, open-labelled, active-controlled, multinational, dose-escalation trial investigating safety, tolerability, pharmacokinetics and pharmacodynamics of a single dose of long-acting growth hormone (NNC0195-0092) compared to daily dosing of Norditropin® SimpleXx® in children with growth hormone deficiency Phase 1 Ongoing, Completed 2013-12-09

///////////Somapacitan, PEPTIDE.2020 APPROVALS, FDA 2020, ソマパシタン, NN8640

C(CCCCCCCC1=NNN=N1)CCCCCCCC(=O)NS(=O)(=O)CCCC(=O)NCCOCCOCC(=O)NC(CCC(=O)NC(CCC(=O)NCCOCCOCC(=O)NC(CCCCNC(=O)CSCC(C(=O)O)N)C(=O)N)C(=O)O)C(=O)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

Copper Cu 64 dotatate, 銅(Cu64)ドータテート;


Copper dotatate Cu-64.png

2D chemical structure of 1426155-87-4

Figure imgf000004_0001

Copper Cu 64 dotatate

銅(Cu64)ドータテート;

UNII-N3858377KC

N3858377KC

Copper 64-DOTA-tate

Copper Cu-64 dotatate

Copper dotatate Cu-64

Diagnostic (neuroendocrine tumors), Radioactive agent

Formula
C65H86CuN14O19S2. 2H
CAS:
 1426155-87-4
Mol weight
1497.1526

FDA APPROVED 2020. 2020/9/3. Detectnet

2-[4-[2-[[(2R)-1-[[(4R,7S,10S,13R,16S,19R)-10-(4-aminobutyl)-4-[[(1S,2R)-1-carboxy-2-hydroxypropyl]carbamoyl]-7-[(1R)-1-hydroxyethyl]-16-[(4-hydroxyphenyl)methyl]-13-(1H-indol-3-ylmethyl)-6,9,12,15,18-pentaoxo-1,2-dithia-5,8,11,14,17-pentazacycloicos-19-yl]amino]-1-oxo-3-phenylpropan-2-yl]amino]-2-oxoethyl]-10-(carboxylatomethyl)-7-(carboxymethyl)-1,4,7,10-tetrazacyclododec-1-yl]acetate;copper-64(2+)

Cuprate(2-)-64Cu, (N-(2-(4,10-bis((carboxy-kappaO)methyl)-7-(carboxymethyl)-1,4,7,10-tetraazacyclododec-1-yl-kappaN1,kappaN4,kappaN7,kappaN10)acetyl)-D-phenylalanyl-L-cysteinyl-L-tyrosyl-D-tryptophyl-L-lysyl-L-threonyl-L-cysteinyl-L-threoni

Copper Cu 64 dotatate, sold under the brand name Detectnet, is a radioactive diagnostic agent indicated for use with positron emission tomography (PET) for localization of somatostatin receptor positive neuroendocrine tumors (NETs) in adults.[1]

Common side effects include nausea, vomiting and flushing.[2]

It was approved for medical use in the United States in September 2020.[1][2]

History

The U.S. Food and Drug Administration (FDA) approved copper Cu 64 dotatate based on data from two trials that evaluated 175 adults.[3]

Trial 1 evaluated adults, some of whom had known or suspected NETs and some of whom were healthy volunteers.[3] The trial was conducted at one site in the United States (Houston, TX).[3] Both groups received copper Cu 64 dotatate and underwent PET scan imaging.[3] Trial 2 data came from the literature-reported trial of 112 adults, all of whom had history of NETs and underwent PET scan imaging with copper Cu 64 dotatate.[3] The trial was conducted at one site in Denmark.[3] In both trials, copper Cu 64 dotatate images were compared to either biopsy results or other images taken by different techniques to detect the sites of a tumor.[3] The images were read as either positive or negative for presence of NETs by three independent image readers who did not know participant clinical information.[3]

PATENT

https://patents.google.com/patent/WO2013029616A1/en

PATENT

https://patents.google.com/patent/US20140341807

  • Known imaging techniques with tremendous importance in medical diagnostics are positron emission tomography (PET), computed tomography (CT), magnetic resonance imaging (MRI), single photon computed tomography (SPECT) and ultrasound (US). Although today’s imaging technologies are well developed they rely mostly on non-specific, macroscopic, physical, physiological, or metabolic changes that differentiate pathological from normal tissue.
  • [0003]
    Targeting molecular imaging (MI) has the potential to reach a new dimension in medical diagnostics. The term “targeting” is related to the selective and highly specific binding of a natural or synthetic ligand (binder) to a molecule of interest (molecular target) in vitro or in vivo.
  • [0004]
    MI is a rapidly emerging biomedical research discipline that may be defined as the visual representation, characterization and quantification of biological processes at the cellular and sub-cellular levels within intact living organisms. It is a novel multidisciplinary field, in which the images produced reflect cellular and molecular pathways and in vivo mechanism of disease present within the context of physiologically authentic environments rather than identify molecular events responsible for disease.
  • [0005]
    Several different contrast-enhancing agents are known today and their unspecific or non-targeting forms are already in clinical routine. Some examples listed below are reported in literature.
  • [0006]
    For example, Gd-complexes could be used as contrast agents for MRI according to “Contrast Agents I” by W. Krause (Springer Verlag 2002, page one and following pages). Furthermore, superparamagnetic particles are another example of contrast-enhancing units, which could also be used as contrast agents for MRI (Textbook of Contrast Media, Superparamagnetic Oxides, Dawson, Cosgrove and Grainger Isis Medical Media Ltd, 1999, page 373 and following pages). As described in Contrast Agent II by W. Krause (Springer Verlag 2002, page 73 and following pages), gas-filled microbubbles could be used in a similar way as contrast agents for ultrasound. Moreover “Contrast Agents II” by W. Krause (Springer Verlag, 2002, page 151 and following pages) reports the use of iodinated liposomes or fatty acids as contrast agents for X-Ray imaging.
  • [0007]
    Contrast-enhancing agents that can be used in functional imaging are mainly developed for PET and SPECT.
  • [0008]
    The application of radiolabelled bioactive peptides for diagnostic imaging is gaining importance in nuclear medicine. Biologically active molecules which selectively interact with specific cell types are useful for the delivery of radioactivity to target tissues. For example, radiolabelled peptides have significant potential for the delivery of radionuclides to tumours, infarcts, and infected tissues for diagnostic imaging and radiotherapy.
  • [0009]
    DOTA (1,4,7,10-tetrakis(carboxymethyl)-1,4,7,10tetraazacyclododecane) and its derivatives constitute an important class of chelators for biomedical applications as they accommodate very stably a variety of di- and trivalent metal ions. An emerging area is the use of chelator conjugated bioactive peptides for labeling with radiometals in different fields of diagnostic and therapeutic nuclear oncology.
  • [0010]
    There have been several reports in recent years on targeted radiotherapy with radiolabeled somatostatin analogs.
  • [0011]
    US2007/0025910A1 discloses radiolabled somatostatin analogs primarily based on the ligand DOTA-TOC. The radionucleotide can be (64)Copper and the somatostatin analog may be octreotide, lanreotide, depreotide, vapreotide or derivatives thereof. The compounds of US2007/0025910A1 are useful in radionucleotide therapy of tumours.
  • [0012]
    US2007/0025910A1 does not disclose (64)Cu-DOTA-TATE. DOTA-TATE and DOTA-TOC differ clearly in affinity for the 5 known somatostatin receptors (SST1-SST2). Accordingly, the DOTA-TATE has a 10-fold higher affinity for the SST2 receptor, the receptor expressed to the highest degree on neuroendocrine tumors. Also the relative affinity for the other receptor subtypes are different. Furthermore, since 177Lu-DOTATATE is used for radionuclide therapy, only 64Cu-DOTATATE and not 64Cu-DOTATOC can be used to predict effect of such treatment by a prior PET scan.
  • [0013]
    There exists a need for further peptide-based compounds having utility for diagnostic imaging techniques, such as PET.

Figure US20140341807A1-20141120-C00001

    • EXAMPLE
  • [0033]
    Preparation of “Cu-Dotatate-DOTA-TATE
  • [0034]
    64Cu was produced using a GE PETtrace cyclotron equipped with a beamline. The 64Cu was produced via the 64Ni (p,n) 64Cu reaction using a solid target system consisting of a water cooled target mounted on the beamline. The target consisted of 64Ni metal (enriched to >99%) electroplated on a silver disc backing. For this specific type of production a proton beam with the energy of 16 MeV and a beam current of 20 uA was used. After irradiation the target was transferred to the laboratory for further chemical processing in which the 64Cu was isolated using ion exchange chromatography. Final evaporation from aq. HCl yielded 2-6 GBq of 64Cu as 64CuCl2 (specific activity 300-3000 TBq/mmol; RNP >99%). The labeling of 64Cu to DOTA-TATE was performed by adding a sterile solution of DOTA-TATE (0.3 mg) and Gentisic acid (25 mg) in aq Sodium acetate (1 ml; 0.4M, pH 5.0) to a dry vial containing 64CuCl2 (˜1 GBq). Gentisic acid was added as a scavenger to reduce the effect of radiolysis. The mixture was left at ambient temperature for 10 minutes and then diluted with sterile water (1 ml). Finally, the mixture was passed through a 0.22 μm sterile filter (Millex GP, Millipore). Radiochemical purity was determined by RP-HPLC and the amount of unlabeled 64Cu2+ was determined by thin-layer chromatography. All chemicals were purchased from Sigma-Aldrich unless specified otherwise. DOTA-Tyr3-Octreotate (DOTA-TATE) was purchased from Bachem (Torrance, Calif.). Nickel-64 was purchased in +99% purity from Campro Scientific Gmbh. All solutions were made using Ultra pure water (<0.07 μSimens/cm). Reversed-phase high pressure liquid chromatography was performed on a Waters Alliance 2795 Separations module equipped with at Waters 2489 UV/Visible detector and a Caroll Ramsey model 105 S-1 radioactivity detector—RP-HPLC column was Luna C18, HST, 50×2 mm, 2.5 μm, Phenomenex. The mobile phase was 5% aq. acetonitrile (0.1% TFA) and 95% aq. acetonitrile (0.1% TFA).
  • [0035]
    Thin layer chromatography was performed with a Raytest MiniGita Star TLC-scanner equipped with a Beta-detector. The eluent was 50% aq methanol and the TLC-plate was a Silica60 on Al foil (Fluka). Ion exchange chromatography was performed on a Dowex 1×8 resin (Chloride-form, 200-400 mesh).

References

  1. Jump up to:a b “FDA approval letter” (PDF). 3 September 2020. Retrieved 5 September 2020.  This article incorporates text from this source, which is in the public domain.
  2. Jump up to:a b “RadioMedix and Curium Announce FDA Approval of Detectnet (copper Cu 64 dotatate injection) in the U.S.” (Press release). Curium. 8 September 2020. Retrieved 9 September 2020 – via GlobeNewswire.
  3. Jump up to:a b c d e f g h “Drug Trials Snapshots: Detectnet”U.S. Food and Drug Administration (FDA). 3 September 2020. Retrieved 10 September 2020.  This article incorporates text from this source, which is in the public domain.

External links

Coppers Coming | Cu 64 dotatate injection is coming soonThe emerging role of copper-64 radiopharmaceuticals as cancer theranostics  - ScienceDirect

The emerging role of copper-64 radiopharmaceuticals as cancer theranostics  - ScienceDirect

The FDA has approved copper Cu 64 dotatate injection (Detectnet) for the localization of somatostatin receptor–positive neuroendocrine tumors (NETs), according to an announcement from RadioMedix Inc. and Curium Pharma.1

The positron emission tomography (PET) diagnostic agent is anticipated to launch immediately, according to Curium. Doses will be accessible through several nuclear pharmacies or through the nuclear medicine company.

“Detectnet brings an exciting advancement in the diagnosis of NETs for healthcare providers, patients, and their caregivers,” Ebrahim Delpassand MD, CEO of RadioMedix, stated in a press release. “The phase 3 results demonstrate the clinical sensitivity and specificity of Detectnet which will provide a great aid to clinicians in developing an accurate treatment approach for their [patients with] NETs.”

Copper Cu 64 dotatate adheres to somatostatin receptors with highest affinity for subtype 2 receptors (SSTR2). Specifically, the agent binds to somatostatin receptor–expressing cells, including malignant neuroendocrine cells; these cells overexpress SSTR2. The agent is a positron-producing radionuclide that possesses an emission yield that permits PET imaging.

“Perhaps most exciting is that the 12.7-hour half-life allows Detectnet to be produced centrally and shipped to sites throughout the United States,” added Delpassand. “This will help alleviate shortages or delays that have been experienced with other somatostatin analogue PET agents.”

Two single-center, open-label studies confirmed the efficacy of the diagnostic agent, according to Curium.2 In Study 1, investigators conducted a prospective analysis of 63 patients, which included 42 patients with known or suspected NETs according to histology, conventional imaging, or clinical evaluations, and 21 healthy volunteers. The majority of the participants, or 88% (n = 37) had a history of NETs at the time that they underwent imaging. Just under half of patients (44%; n = 28) were men and the majority were white (86%). Moreover, patients had a mean age of 54 years.

Images produced by the PET agent were interpreted to be either positive or negative for NET via 3 independent readers who had been blinded to the clinical data and other imaging information. Moreover, the results from the diagnostic agent were compared with a composite reference standard that was comprised of 1 oncologist’s blinded evaluation of patient diagnosis based on available histopathology results, reports of conventional imaging that had been done within 8 weeks before the PET imaging, as well as clinical and laboratory findings, which involved chromogranin A and serotonin levels.

Additionally, the percentage of patients who tested positive for disease via composite reference as well as through PET imaging was used to quantify positive percent agreement. Conversely, the percentage of participants who did not have disease per composite reference and who were determined to be negative for disease per PET imaging was used to quantify negative percent agreement.

Results showed that the percent reader agreement for positive detection in 62 scans was 91% (95% CI, 75-98) and negative detection was 97% (95% CI, 80-99). For reader 2, these percentages were 91% (95% CI, 75-98) and 80% (95% CI, 61-92), respectively, for 63 scans. Lastly, the percent reader agreement for reader 3 in 63 scans was 91% (95% CI, 75-98) positive and 90% (95% CI, 72-97) negative.

Study 2 was a retrospective analysis in which investigators examined published findings collected from 112 patients; 63 patients were male, while 43 were female. The mean age of patients included in the analysis was 62 years. All patients had a known history of NETs. Results demonstrated similar performance with the PET imaging agent.

In both safety and efficacy trials, a total of 71 patients were given a single dose of the diagnostic agent; the majority of these patients had known or suspected NETs and 21 were healthy volunteers. Adverse reactions such as nausea, vomiting, and flushing were reported at a rate of less than 2%. In all clinical experience that has been published, a total of 126 patients with a known history of NETs were given a single dose of the PET diagnostic agent. A total of 4 patients experienced nausea immediately after administration.

“Curium is excited to bring the first commercially available Cu 64 diagnostic agent to the US market,” Dan Brague, CEO of Curium, North America, added in the release. “Our unique production capabilities and distribution network allow us to deliver to any nuclear pharmacy, hospital, or imaging center its full dosing requirements first thing in the morning, to provide scheduling flexibility to the institution and its patients. We look forward to joining with healthcare providers and our nuclear pharmacy partners to bring this highly efficacious agent to the market.”

References
1. RadioMedix and Curium announce FDA approval of Detectnet (copper Cu 64 dotatate injection) in the US. News release. RadioMedix Inc and Curium. September 8, 2020. Accessed September 9, 2020. https://bit.ly/3m6iC0q.
2. Detectnet. Prescribing information. Curium Pharma; 2020. Accessed September 9, 2020. https://bit.ly/32eZxS3.

///////////////Copper Cu 64 dotatate, 銅(Cu64)ドータテート , FDA 2020, 2020 APPROVALS, Diagnostic, neuroendocrine tumors, Radioactive agent,

CC(C1C(=O)NC(CSSCC(C(=O)NC(C(=O)NC(C(=O)NC(C(=O)N1)CCCCN)CC2=CNC3=CC=CC=C32)CC4=CC=C(C=C4)O)NC(=O)C(CC5=CC=CC=C5)NC(=O)CN6CCN(CCN(CCN(CC6)CC(=O)[O-])CC(=O)[O-])CC(=O)O)C(=O)NC(C(C)O)C(=O)O)O.[Cu+2]