Quantcast
Channel: New Drug Approvals
Viewing all 2878 articles
Browse latest View live

Sucroferric oxyhydroxide, 含糖酸化鉄, スクロオキシ水酸化鉄

$
0
0

Image result for Sucroferric oxyhydroxide KEGG

Image result for Sucroferric oxyhydroxide

Sucroferric oxyhydroxide

Iron sucrose (USP);
Ferric oxide, saccharated;
Sucroferric oxyhydroxide;
Venofer (TN)

含糖酸化鉄;
スクロオキシ水酸化鉄

Molecular Formula: C12H29Fe5Na2O23
Molecular Weight: 866.546 g/mol
CAS: 8047-67-4

CAS REGISTRY NUMBER 12134-57-5, 8047-67-4

disodium;(2R,3R,4S,5S,6R)-2-[(2S,3S,4S,5R)-3,4-dihydroxy-2,5-bis(hydroxymethyl)oxolan-2-yl]oxy-6-(hydroxymethyl)oxane-3,4,5-triol;iron(3+);oxygen(2-);hydroxide;trihydrate

Iron sugar; Saccharated iron; Sucroferric oxyhydroxide; Saccharated iron oxide; Saccharated ferric oxide; Ferrivenin

Ferric oxyhydroxide; Ferrihydrite; Iron oxyhydroxide; P-TOL; PA-21; PA21-1; Phosphate binder – Vifor Pharma; suroferric oxyhydroxide tablets; Velphoro

NDC 49230-645-51

Iron saccharate (Sucroferric oxyhydroxide or Iron Sucrose) is used as a source of iron in patients with iron deficiency anemia with chronic kidney disease (CKD), including those who are undergoing dialysis (hemodialysis or peritoneal) and those who do not require dialysis. Due to less side effects than iron dextran, iron saccharate is more preferred in chronic kidney disease patients.

Mixture of polynuclear iron(III)-oxyhydroxide, starch and sucrose

VIFOR FRESENIUS MEDICAL CARE RENAL PHARMA FRANCE

Approved in US

Indicated for the control of serum phosphorus levels in patients with chronic kidney disease on dialysis.

THERAPEUTIC CLAIM Oral phosphate binder, treatement of elevated
phosphate levels in patients undergoing dialysis
CHEMICAL DESCRIPTIONS
1. Ferric hydroxide oxide
2. Mixture of iron(III) oxyhydroxide, sucrose, starches
3. Polynuclear iron(III) oxyhydroxide stabilized with sucrose and starches
structure
O =Fe -OH
MOLECULAR FORMULA FeHO2•xC12H22O11•y(C6H10O5)n

SPONSOR Vifor (International) Inc.
CODE DESIGNATIONS PA21
CAS REGISTRY NUMBER 12134-57-5

  • ClassFerric compounds; Hyperphosphataemia therapies
  • Mechanism of ActionPhosphate binding modulators
  • MarketedHyperphosphataemia
  • 24 Jun 2018Biomarkers information updated
  • 19 Jun 2018Kissei Pharmaceutical completes a phase III trial in Hyperphosphataemia (Treatment-experienced) in Japan (PO) (UMIN000023657)
  • 09 Jun 2017Phase-II clinical trials in Hyperphosphataemia in Austria (PO) (NCT03010072)

Image result for Sucroferric oxyhydroxide

Sucroferric oxyhydroxide is a brown, amorphous powder. The drug substance is relatively poorly defined, so that the manufacturing process is particularly important. Sucroferric oxyhydroxide is prepared by basifying a ferric chloride solution, giving a polynuclear iron(III)-oxyhydroxide suspension which is mixed with potato and maize starches and sucrose. Vifor states that the sucrose stabilises the iron core and thus maintain the high phosphate adsorption capacity while the starches function as processing aids, but they are added simultaneously and the drug substance is probably a complex mixture of species.

The solubility of the active moiety, polynuclear iron oxyhydroxide, is evidently low in the gastrointestinal (GI) tract so that iron absorption is low. Aqueous solubility at different pH has been very poorly quantified. Vifor states that the “sucrose part is soluble in water, iron(III)-oxyhydroxide/starch mixture is practically insoluble in water.” While the iron oxide particle size is important in determining the phosphate binding, it is relatively difficult to directly measure. The sucrose/starch “wrapped” drug substance particle size is established and the process is controlled, but it does not correlate well with phosphate adsorption. Sucroferric oxyhydroxide cannot be controlled in the manner of a well-defined molecular drug and some variability between batches is likely. The drug substance specification includes a phosphate adsorption test. Vifor has tested the adsorption of a range of other in vivo chemical species to sucroferric oxyhydroxide and not identified any likely to be strongly bound, or affect phosphate binding, except for oxalate. Some drugs, however, do interact, for example alendronate is strongly absorbed (and the PI warnings in that context should be generalised to all bisphosphonates, not just identify the single drug in class studied)….https://www.tga.gov.au/sites/default/files/auspar-sucroferric-oxyhydroxide-150219.pdf

EMA

Name Active substance Therapeutic area Date of authorisation / refusal Has current safety alert Status
Velphoro mixture of polynuclear iron(III)-oxyhydroxide, sucrose and starches HyperphosphatemiaRenal Dialysis 26/08/2014   Authorised

Product details

Name Velphoro
Agency product number EMEA/H/C/002705
Active substance mixture of polynuclear iron(III)-oxyhydroxide, sucrose and starches
International non-proprietary name(INN) or common name mixture of polynuclear iron(III)-oxyhydroxide, sucrose and starches
Therapeutic area HyperphosphatemiaRenal Dialysis
Anatomical therapeutic chemical (ATC) code V03AE05
Additional monitoring This medicine is under additional monitoring. This means that it is being monitored even more intensively than other medicines. For more information, see medicines under additional monitoring.

Publication details

Marketing-authorisation holder Vifor Fresenius Medical Care Renal Pharma France
Revision 5
Date of issue of marketing authorisation valid throughout the European Union 26/08/2014

Contact address:

Vifor Fresenius Medical Care Renal Pharma France
100-101 Terrasse Boieldieu
Tour Franklin- La Défense 8
92042 Paris la Défense Cedex
France

http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_Information/human/002705/WC500175254.pdf

Sucroferric oxyhydroxide (INN; trade name Velphoro, by Vifor Fresenius Medical Care Renal Pharma) is a non-calcium, iron-based phosphate binder used for the control of serum phosphorus levels in adult patients with chronic kidney disease (CKD) on haemodialysis(HD) or peritoneal dialysis (PD).[1] It is used in form of chewable tablets.

Hyperphosphatemia

In a healthy person, normal serum phosphate levels are maintained by the regulation of dietary absorptionbone formation and resorption, equilibration with intracellular stores, and renal excretion.[2] When kidney function is impaired, phosphate excretion declines. Without specific treatment, hyperphosphataemia occurs almost universally, despite dietary phosphate restriction and conventional dialysis treatment.[2][3] In patients on dialysis, hyperphosphataemia is an independent risk factor for fracturescardiovascular disease and mortality.[4][5] Abnormalities in phosphate metabolism such as hyperphosphatemia are included in the definition of the new chronic kidney disease–mineral and bone disorder (CKD-MBD).[5]

Structure and mechanism of action

Sucroferric oxyhydroxide comprises a polynuclear iron(III)-oxyhydroxide core that is stabilised with a carbohydrate shell composed of sucrose and starch.[6][7] The carbohydrate shell stabilises the iron(III)-oxyhydroxide core to preserve the phosphate adsorption capacity.

Dietary phosphate binds strongly to sucroferric oxyhydroxide in the gastrointestinal (GI) tract. The bound phosphate is eliminated in the faeces and thereby prevented from absorption into the blood. As a consequence of the decreased dietary phosphate absorption, serum phosphorus concentrations are reduced.

Medical uses

Sucroferric oxyhydroxide is approved by the United States Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for the control of serum phosphorus levels in patients with chronic kidney disease (CKD) on dialysis.[1][8]

Adverse effects

The most frequently reported adverse drug reactions reported from trials were diarrhoea and discoloured faeces.[1][8] The vast majority of gastrointestinal adverse events occurred early during treatment and abated with time under continued dosing.[1]

Interactions

Drug-interaction studies and post hoc analyses of Phase 3 studies showed no clinically relevant interaction of sucroferric oxyhydroxide with the systemic exposures to losartanfurosemideomeprazoledigoxin, and warfarin,[9] the lipid-lowering effects of statins,[10] and oral vitamin D receptor agonists.[11] According to the European label (Summary of Product Characteristics), medicinal products that are known to interact with iron (e.g. doxycycline) or have the potential to interact with Velphoro should be administered at least one hour before or two hours after Velphoro.[1] This allows sucroferric oxyhydroxide to bind phosphate as intended and be excreted without coming into contact with medications in the gut that it might interact with. According to the US prescribing information, Velphoro should not be prescribed with oral levothyroxine.[8] The combination of sucroferric oxyhydroxide and levothyroxine is contraindicated because sucroferric oxyhydroxide contains iron, which may cause levothyroxine to become insoluble in the gut, thereby preventing the intestinal absorption of levothyroxine.[12]

Chewability

The chewability of sucroferric oxyhydroxide compares well with that of Calcimagon, a calcium containing tablet used as a standard for very good chewability.[13] Tablets of sucroferric oxyhydroxide easily disintegrated in artificial saliva.

Effectiveness and phosphate binding

Clinical Phase 3 studies showed that sucroferric oxyhydroxide achieves and maintains phosphate levels in compliance with the KDOQI guidelines.[14][15] The reduction in serum phosphate levels of sucroferric oxyhydroxide-treated patients was non-inferior to that in sevelamer-treated patients. The required daily pill burden was lower with sucroferric oxyhydroxide.[14]

Sucroferric oxyhydroxide binds phosphate under empty and full stomach conditions and across the physiologically relevant pH range of the GI tract.[7]

In a retrospective, real-world study, hyperphosphatemic peritoneal dialysis patients who were prescribed to switch to sucroferric oxyhydroxide from sevelamer, lanthanum carbonate, or calcium acetate had significant reductions in serum phosphorus levels, along with a 53% decrease in the prescribed daily pill burden.[16]

Sucroferric oxyhydroxide nonproprietary drug name

https://www.ama-assn.org/resources/doc/…/sucroferricoxyhydroxide.pdf

1. February 27, 2013. N13/36. STATEMENT ON A NONPROPRIETARY NAME ADOPTED BY THE USAN COUNCIL. USAN (ZZ-19). SUCROFERRIC 

The US Food and Drug Administration has given the green light to Vifor Fresenius Medical Care Renal Pharma’s hyperphosphatemia drug Velphoro.

The approval for Velphoro (sucroferric oxyhydroxide), formerly known as PA21, is based on Phase III data demonstrated that the drug successfully controls the accumulation of phosphorus in the blood with the advantage of a much lower pill burden than the current standard of care in patients with chronic kidney disease on dialysis, namely Sanofi’s Renvela (sevelamer carbonate). read this at

http://www.pharmatimes.com/Article/13-11-28/FDA_okays_Vifor_Fresenius_phosphate_binder_Velphoro.aspx

Velphoro (PA21) receives US FDA approval for the treatment of hyperphosphatemia in Chronic Kidney Disease Patients on dialysis
Velphoro (sucroferric oxyhydroxide) has received US Food and Drug Administration (FDA) approval for the control of serum phosphorus levels in patients with Chronic Kidney Disease (CKD) on dialysis. Velphoro will be launched in the US by Fresenius Medical Care North America in 2014.

Velphoro (previously known as PA21) is an iron-based, calcium-free, chewable phosphate binder. US approval was based on a pivotal Phase III study, which met its primary and secondary endpoints. The study demonstrated that Velphoro® successfully controls hyperphosphatemia with fewer pills than sevelamer carbonate, the current standard of care in patients with CKD on dialysis. The average daily dose to control hyperphosphatemia was 3.3 pills per day after 52 weeks.

Velphoro was developed by Vifor Pharma. In 2011, all rights were transferred to Vifor Fresenius Medical Care Renal Pharma, a common company of Galenica and Fresenius Medical Care. In the US, Velphorowill be marketed by Fresenius Medical Care North America, a company with a strong marketing and sales organization, and expertise in dialysis care. The active ingredient of Velphoro is produced by Vifor Pharma in Switzerland.

Hyperphosphatemia, an abnormal elevation of phosphorus levels in the blood, is a common and serious condition in CKD patients on dialysis. Most dialysis patients are treated with phosphate binders. However, despite the availability of a number of different phosphate binders, up to 50% of patients depending on the region are still unable to achieve and maintain their target serum phosphorus levels. In some patients, noncompliance due to the high pill burden and poor tolerability appear to be key factors in the lack of control of serum phosphorus levels. On average, dialysis patients take approximately 19 pills per day with phosphate binders comprising approximately 50% of the total daily pill burden. The recommended starting dose of Velphoro is 3 tablets per day (1 tablet per meal).

Full results from the pivotal Phase III study involving more than 1,000 patients were presented at both the 50th ERA-EDTA (European Renal Association European Dialysis and Transplant Association) Congress in Istanbul, Turkey, in May 2013, and the American Society of Nephrology (ASN) Kidney Week in Atlanta, Georgia, in November 2013. Velphorowas shown to be a potent phosphate binder, with lower pill burden and a good safety profile.

Based on these data, Vifor Fresenius Medical Care Renal Pharma believes that Velphoro offers a new and effective therapeutic option for the control of serum phosphorus levels in patients with chronic kidney disease on dialysis.
The regulatory processes in Europe, Switzerland and Singapore are ongoing and decisions are expected in the first half 2014. Further submissions for approval are being prepared.

PATENT

https://patents.google.com/patent/WO2016038541A1/en

Hyperphosphatemia is associated with significant increase in morbidity and mortality, and may induce severe complications, such as hypocalcemia, decreasing of vitamin-D production and metastatic calcification. Hyperphosphatemia is also contributing to the increased incidence of cardiovascular disease among dialysis-dependent patients. The phosphate binding capacity of iron oxide hydroxides is known in the art. The possible medical application of iron hydroxides and iron oxide hydroxides as phosphate adsorbents is also described.

US 4,970,079 patent discloses a method of controlling serum phosphate level in patients by iron oxy-hydroxides which bind to ingested phosphate. US 5,514,281 patent also discloses a process for the selective elimination of inorganic phosphate from body fluids by using a polynuclear metal oxyhydroxide preferably iron (III) oxyhydroxide.

US 6,174,442 patent describes an adsorbent for phosphate and a process for the preparation thereof, which contains polynuclear β-iron hydroxide stabilized by carbohydrates and/or humic acid.

In order to obtain an iron-based compound which can be used as a pharmaceutical, it is necessary to have an iron-based compound which is stable. It is known that iron oxide- hydroxide is not a stable compound with time ageing occurs. Ageing usually not only involves crystallization but also particle enlargement. Such ageing may alter the phosphate binding of an iron oxide -hydroxide based phosphate adsorbent. Accordingly, there exists a need for a process for manufacturing of an iron containing phosphate adsorbent. The process needs to be scalable, robust and consistently producing an iron containing phosphate adsorbent of the required pharmaceutical grade.

Examples

In examples which are intended to illustrate embodiments of the invention but which are not intended to limit the scope of the invention: ) Method of Making an Iron Containing Phosphate Adsorbent

To a solution of 1.96 kg sodium carbonate dissolved in 12.5 liter water, solution of 2.5 kg iron (III) chloride hexahydrate dissolved in 17.5 liter water was added at a temperature of 5 – 10°C. The resulting mixture was stirred for 90 to 120 minutes at 5 – 10°C. (25.0×3) liter water was added to the reaction mass and raised the temperature at 15 – 20°C with stirring. Stopped the stirring, settled precipitate and the supernatant water was removed. The precipitate was filtered and washed with 1.25 liter water. A suspension of the precipitate was prepared in water. To this, 875.0 gm sucrose and 695.0 gm potato starch were added and stirred for 120 minutes at 25 – 35°C. Cooled the reaction mass at 10 – 15°C and stirred for 90 to 120 minutes. 25.0 liters cold acetone was added to the reaction mass at 10 – 15°C and stirred for 90 to 120 minutes. The final product was filtered and washed with 1.25 liter cold acetone and further dried under vacuum at 30-35°C.

Yield: 2.08 kg ) Large-scale Method of Making an Iron Containing Phosphate Adsorbent

An aqueous solution of sodium carbonate and an aqueous solution of iron (III) chloride hexahydrate were mixed at a temperature of 5 – 10°C, optionally in the presence of solvent- 1. A volume of aqueous solution of sodium carbonate necessary to maintain the pH at about 7.0 to form a colloidal suspension of ferric hydroxide. The resulting mixture was stirred for 90 to 120 minutes at 5 – 10°C. Water was added to the reaction mass with stirring. Stopped the stirring, settled precipitated product and the water was decanted or siphoned. The precipitated product was further filtered and washed with using water. Suspension of the precipitated product was prepared in the water. Subsequently, sucrose and starch were added in to the suspension and stirred for 120 minutes at 25 – 35°C. Cooled the reaction mixture at 10 – 15°C and stirred for 90 to 120 minutes. Solvent-2 was added to the reaction mixture at 10 – 15°C and stirred for 90 to 120 minutes. The product was filtered and washed with the solvent-2 and further dried under vacuum at 30-35°C. Few illustrative examples provided in Table- 1, wherein the iron containing phosphate adsorbents were prepared according to the process of example-2 using the respective combination of Solvent- 1 and Solvent-2 as given in the table:

Table-1

Figure imgf000013_0001

3) Physical Properties of an Iron Containing Phosphate Adsorbents prepared as per above example-2.

> BET active Surface Area:

· Instrument : Surface area analyzer

• Condition : Surface area (m2/gm) at N2.P/P0 = 10%

Table-2

Figure imgf000013_0002

> Phosphate Binding Capacity at pH 3.0:

· Method : Ion Chromatography Instrument : Metrohm IC equipped with pump, Injector, conductivity detector and recorder.

Column Dionex Ion Pac AS-11 (4.0 x 250mm), 13μπι

Guard column Dionex Ion Pac AG-11 (4.0 x 50mm), 13μπι

Buffer preparation Weigh accurately about 2.118g of Sodium carbonate and 180mg of Sodium hydroxide in 1700mL water.

Mobile phase preparation : Buffer and acetonitrile (1700:300).

Results: Phosphate binding of an iron containing phosphate adsorbents obtained by following the process of the present invention found in the range of 30 mg/gm to 60 mg/gm. Particle Size Distribution:

Instrument Model : Malvern Mastersizer 2000 Particle size analyzer

Sampling Unit : Hydro 2000S

Analysis Model : General Purpose

Dispersant : 0.1% Span 85 in n-Hexane

Dispersant RI : 1.380

Stirrer Speed : 2200 RPM

Absorption : 1

Particle RI : 1.5

Obscuration : 10% to 20%

Sample Measurement time : 12 seconds

Background Measurement time : 12 seconds Table-3

Particle size distribution

Example no.

d(0.9) (μηι)

3d 43.67

3e 65.37

3f 37.75

Publication numberPriority datePublication dateAssigneeTitle
US4970079A1989-06-051990-11-13Purdue Research FoundationMethod and composition of oxy-iron compounds for treatment of hyperphosphatemia
US5514281A1992-11-241996-05-07B. Braun Melsungen AgProcess for the selective elimination of inorganic phosphate from liquids by means of adsorbent materials modified with polynuclear metal oxyhydroxides
US6174442B11995-12-192001-01-16Vifor (International) AgAdsorbent for phosphate from an aqueous medium, production and use of said adsorbent
EP1932808A1 *2006-12-142008-06-18Novartis AGIron(III)-Carbohydrate based phosphate adsorbent
WO2009062993A1 *2007-11-162009-05-22Vifor (International) AgPharmaceutical compositions
WO2010015827A2 *2008-08-052010-02-11Medical Research CouncilPhosphate binding materials and their uses

Image result for Sucroferric oxyhydroxide KEGG

References

  1. Jump up to:a b c d e “Velphoro (sucroferric oxyhydroxide). Summary of Product Characteristics”(PDF). EMA. Archived from the original on October 21, 2014. Retrieved 24 October 2014.
  2. Jump up to:a b Jha V, Garcia-Garcia G, Iseki K, Li Z, Naicker S, Plattner B, Saran R, Wang AY, Yang CW (July 2013). “Chronic kidney disease: global dimension and perspectives”. Lancet382(9888): 260–72. doi:10.1016/S0140-6736(13)60687-XPMID 23727169.
  3. Jump up^ Hutchison AJ, Smith CP, Brenchley PE (September 2011). “Pharmacology, efficacy and safety of oral phosphate binders”. Nature Reviews. Nephrology7 (10): 578–89. doi:10.1038/nrneph.2011.112PMID 21894188.
  4. Jump up^ Isakova T, Gutiérrez OM, Chang Y, Shah A, Tamez H, Smith K, Thadhani R, Wolf M (February 2009). “Phosphorus binders and survival on hemodialysis”Journal of the American Society of Nephrology20 (2): 388–96. doi:10.1681/ASN.2008060609PMC 2637053Freely accessiblePMID 19092121.
  5. Jump up to:a b “KDIGO clinical practice guideline for the diagnosis, evaluation, prevention, and treatment of Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD)”. Kidney International Supplement76 (113): S1–130. August 2009. doi:10.1038/ki.2009.188PMID 19644521.
  6. Jump up^ Vifor Fresenius Medical Care Renal Pharma. Product Monograph 2015.
  7. Jump up to:a b Wilhelm M, Gaillard S, Rakov V, Funk F (April 2014). “The iron-based phosphate binder PA21 has potent phosphate binding capacity and minimal iron release across a physiological pH range in vitro”. Clinical Nephrology81 (4): 251–8. doi:10.5414/cn108119PMID 24656315.
  8. Jump up to:a b c “Highlights of Prescribing information for Velphoro”. Fresenius. September 2014.
  9. Jump up^ Chong E, Kalia V, Willsie S, Winkle P (December 2014). “Drug-drug interactions between sucroferric oxyhydroxide and losartan, furosemide, omeprazole, digoxin and warfarin in healthy subjects”Journal of Nephrology27 (6): 659–66. doi:10.1007/s40620-014-0080-1PMC 4242982Freely accessiblePMID 24699894.
  10. Jump up^ Levesque V, Chong EMF, Moneuse P (2013). “Post-hoc analysis of pharmacodynamic interaction of PA21 with statins in a Phase 3 study of PA21 in dialysis patients with hyperphosphatemia”. J Am Soc Nephrol24: 758A.
  11. Jump up^ Floege J, Botha J, Chong E et al. (31 May 2014). PA21 does not interact with oral vitamin D receptor agonists: a post hoc analysis of a Phase 3 study. ERA-EDTA congress. Amsterdam, The Netherlands. Abstract no. SP257.
  12. Jump up^ Prescribing Information. Synthroid (levothyroxine). Chicago, IL: Abbott Laboratories. March 1, 2008.
  13. Jump up^ Lanz M, Baldischweiler J, Kriwet B, Schill J, Stafford J, Imanidis G (December 2014). “Chewability testing in the development of a chewable tablet for hyperphosphatemia”. Drug Development and Industrial Pharmacy40 (12): 1623–31. doi:10.3109/03639045.2013.838583PMID 24010939.
  14. Jump up to:a b Floege J, Covic AC, Ketteler M, Rastogi A, Chong EM, Gaillard S, Lisk LJ, Sprague SM (September 2014). “A phase III study of the efficacy and safety of a novel iron-based phosphate binder in dialysis patients”Kidney International86 (3): 638–47. doi:10.1038/ki.2014.58PMC 4150998Freely accessiblePMID 24646861.
  15. Jump up^ Floege J, Covic AC, Ketteler M, Mann JF, Rastogi A, Spinowitz B, Chong EM, Gaillard S, Lisk LJ, Sprague SM (June 2015). “Long-term effects of the iron-based phosphate binder, sucroferric oxyhydroxide, in dialysis patients”Nephrology, Dialysis, Transplantation30(6): 1037–46. doi:10.1093/ndt/gfv006PMC 4438742Freely accessiblePMID 25691681.
  16. Jump up^ Kalantar-Zadeh K, Parameswaran V, Ficociello LH, Anderson L, Ofsthun NJ, Kwoh C, Mullon C, Kossmann RJ, Coyne DW (2018). “Real-World Scenario Improvements in Serum Phosphorus Levels and Pill Burden in Peritoneal Dialysis Patients Treated with Sucroferric Oxyhydroxide”American Journal of Nephrology47 (3): 153–161. doi:10.1159/000487856PMC 5906196Freely accessiblePMID 29514139.
Sucroferric oxyhydroxide
Clinical data
Trade names Velphoro
AHFS/Drugs.com Consumer Drug Information
License data
Pregnancy
category
  • US: B (No risk in non-human studies)
  • EU, Japan: No risk in non-human studies
Routes of
administration
Oral (chewable tablets)
ATC code
Legal status
Legal status
  • US: ℞-only
  • EU: Rx only
  • Japan: prescription only
Chemical and physical data
Formula Varies

FDA Orange Book Patents

FDA Orange Book Patents: 1 of 2 (FDA Orange Book Patent ID)
Patent 6174442
Expiration Dec 19, 2018
Applicant VIFOR FRESENIUS
Drug Application N205109 (Prescription Drug: VELPHORO. Ingredients: SUCROFERRIC OXYHYDROXIDE)
FDA Orange Book Patents: 2 of 2 (FDA Orange Book Patent ID)
Patent 9561251
Expiration Jan 23, 2030
Applicant VIFOR FRESENIUS
Drug Application N205109 (Prescription Drug: VELPHORO. Ingredients: SUCROFERRIC OXYHYDROXIDE)
Patent ID Title Submitted Date Granted Date
US6174442 Adsorbent for phosphate from an aqueous medium, production and use of said adsorbent
1998-06-02
2001-01-16
US9561251 PHARMACEUTICAL COMPOSITIONS
2008-11-13
2010-09-30

/////////////Sucroferric oxyhydroxide, EU 2014, Iron sugar, Saccharated iron, Sucroferric oxyhydroxide, Saccharated iron oxide, Saccharated ferric oxide, Ferrivenin, 含糖酸化鉄, スクロオキシ水酸化鉄 , NDC 49230-645-51

C(C1C(C(C(C(O1)OC2(C(C(C(O2)CO)O)O)CO)O)O)O)O.O.O.O.[OH-].[O-2].[O-2].[O-2].[O-2].[O-2].[O-2].[O-2].[O-2].[Na+].[Na+].[Fe+3].[Fe+3].[Fe+3].[Fe+3].[Fe+3]


Stiripentol, スチリペントール

$
0
0

D05928.pngStiripentol.pngChemSpider 2D Image | Stiripentol | C14H18O3Stiripentol structure.svg

Stiripentol

スチリペントール

STIRIPENTOL; Diacomit; 49763-96-4; BCX 2600; Estiripentol; Stiripentolum

CAS: 137767-55-6 49763-96-4

(E)-1-(1,3-benzodioxol-5-yl)-4,4-dimethylpent-1-en-3-ol

Molecular Formula: C14H18O3
Molecular Weight: 234.295 g/mol

UNII

R02XOT8V8I, Diacomit
fda approval 2018/8/20

Stiripentol (marketed as Diacomit by Laboratoires Biocodex) is an anticonvulsant drug used in the treatment of epilepsy. It is approved for the treatment of Dravet syndrome, an epilepsy syndrome. It is unrelated to other anticonvulsants and belongs to the group of aromatic allylic alcohols.

Medical use

It is used in some countries as an add-on therapy with sodium valproate and clobazam for treating children with Dravet syndromewhose seizures are not adequately controlled.[1][2][3] As of 2017 it was not known whether stiripentol remains useful as children become adolescents nor as they become adults.[4]

Adverse effects

Very common (more than 10% of people) adverse effects include loss of appetite, weight loss, insomnia, drowsiness, ataxiahypotonia, and dystonia.[3]

Common (between 1% and than 10% of people) adverse effects include neutropenia (sometimes severe), aggressiveness, irritability, behavior disorders, opposing behavior, hyperexcitability, sleep disorders, hyperkinesias, nausea, vomiting, and elevated gamma-glutamyltransferase.[3]

Interactions

Stiripentol inhibits several cytochrome P450 isoenzymes and so interacts with many anticonvulsants and other medicines.[3]

Pharmacology

As with most anticonvulsants, the precise mechanism of action is unknown. Regardless, stiripentol has been shown to have anticonvulsant effects of its own.

Stiripentol increases GABAergic activity. At clinically relevant concentrations, it enhances central GABA neurotransmission through a barbiturate-like effect, since it increases the duration of opening of GABA-A receptor channels in hippocampal slices.[5] It has also been shown to increase GABA levels in brain tissues by interfering with its reuptake and metabolism.[6] Specifically, it has been shown to inhibit lactate dehydrogenase, which is an important enzyme involved in the energy metabolism of neurons. Inhibition of this enzyme can make neurons less prone to fire action potentials, likely through activation of ATP-sensitive potassium channels.[7]

Stiripentol also improves the effectiveness of many other anticonvulsants, possibly due to its inhibition of certain enzymes, slowing the drugs’ metabolism and increasing blood plasma levels.[3]

Chemistry

Stiripentol is an α-ethylene alcohol; its chemical formula is 4,4-dimethyl-1-[3,4-(methylendioxy)-phenyl]-1penten-3-ol. It is chiral and is marketed as an equimolar racemic mixture. The R enantiomer appears to be around 2.5 times more active than the S enantiomer.[8]

Paper

Synthesis of the antiepileptic (R)-Stiripentol by a combination of lipase catalyzed resolution and alkene metathesis

The enantiopure (ee >99%) antiepileptic (R)-(+)-Stiripentol has been stereoselectively synthesized via cross metathesis of 5-vinylbenzo[d][1,3]dioxole 1 and (R)-(+)-4,4-dimethylpent-1-en-3-ol (R)-(+)-2. A novel one-pot two-step pathway for the synthesis of 5-vinylbenzo[d][1,3]dioxole 1 starting from 3,4-dihydroxycinnamic acid has been introduced. A lipase catalyzed kinetic resolution access to enantiopure (R)-(+)-4,4-dimethylpent-1-en-3-ol (R)-(+)-2 (ee >99%) has also been developed.

Image result for Stiripentol synthesis

Image result for Stiripentol synthesis

Stiripentol (CAS NO.: 49763-96-4), with other name of 4,4-Dimethyl-1-[(3,4-methylenedioxy)phenyl]-1-penten-3-ol, could be produced through many synthetic methods.

Following is one of the reaction routes:

Synthesis of Stiripentol

The synthesis of [14]-labeled stiripentol has been published:The reaction of 3,4-methylenedioxybromobenzene (I) with 14CO2 by means of butyllithium in ether gives 3,4-methylenedioxybenzoic acid (II), which is reduced with LiAlH4 to the corresponding benzyl alcohol (III). Oxidation of (III) with CrO3-pyridine affords the aldehyde (IV), which is condensed with methyl tert-butyl ketone (V) by means of NaOH in refluxing ethanol to give the labeled pentanone (VI). Finally, this compound is reduced to [14C]-labeled stiripentol with NaBH4 in methanol

合成路线图解说明:The condensation of 3,4-methylenedioxybenzaldehyde (I) with 3,3-dimethyl-2-butanone (II) by means of NaOH in ethanol-water gives 4,4-dimethyl-1-[(3,4-methylenedioxy)phenyl]-1-penten-3-one (III), which is reduced with NaBH4 in methanol.
合成路线图解说明:The synthesis of [14]-labeled stiripentol has been published: The reaction of 3,4-methylenedioxybromobenzene (I) with 14CO2 by means of butyllithium in ether gives 3,4-methylenedioxybenzoic acid (II), which is reduced with LiAlH4 to the corresponding benzyl alcohol (III). Oxidation of (III) with CrO3-pyridine affords the aldehyde (IV), which is condensed with methyl tert-butyl ketone (V) by means of NaOH in refluxing ethanol to give the labeled pentanone (VI). Finally, this compound is reduced to [14C]-labeled stiripentol with NaBH4 in methanol.

History

Stiripentol was discovered in 1978 by scientists at Biocodex and clinical trials started over the next few years.[8] It was originally developed for adults with focal seizures, but failed a Phase III trial.[4]

In December 2001 the European Medicines Agency (EMA) granted stiripentol orphan drug status (designation number EU/3/01/071) for the treatment of severe myoclonic epilepsy of infancy (SMEI, also known as Dravet’s syndrome) in children and in 2007, the EMA granted the drug a marketing authorisation for use of the drug as an add-on to other anti-seizure drugs.[3] It was approved in Canada for this use in 2012.[9] As of 2017 it was also approved for this use in Japan.[2]

As of 2014 it was not approved in the US, and parents of children with Dravets were paying around $1,000 for a month supply to obtain it from Europe.[10]

Stiripentol
Stiripentol structure.svg
Clinical data
Trade names Diacomit
AHFS/Drugs.com International Drug Names
License data
Routes of
administration
Oral
ATC code
Legal status
Legal status
  • AU: Unscheduled
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
ChemSpider
UNII
KEGG
ECHA InfoCard 100.051.329 Edit this at Wikidata
Chemical and physical data
Formula C14H18O3
Molar mass 234.30 g·mol−1
3D model (JSmol)

References

  1. Jump up^ Brigo, F; Igwe, SC; Bragazzi, NL (18 May 2017). “Antiepileptic drugs for the treatment of infants with severe myoclonic epilepsy”. The Cochrane Database of Systematic Reviews5: CD010483. doi:10.1002/14651858.CD010483.pub4PMID 28521067.
  2. Jump up to:a b Nickels, KC; Wirrell, EC (May 2017). “Stiripentol in the Management of Epilepsy”. CNS drugs31 (5): 405–416. doi:10.1007/s40263-017-0432-1PMID 28434133.
  3. Jump up to:a b c d e f “Diacomit (stiripentol) SPC” (PDF). EMA. 8 January 2014. Retrieved 1 October 2017. For updates see EMA index page
  4. Jump up to:a b Nabbout, R; Camfield, CS; Andrade, DM; Arzimanoglou, A; Chiron, C; Cramer, JA; French, JA; Kossoff, E; Mula, M; Camfield, PR (April 2017). “Treatment issues for children with epilepsy transitioning to adult care”. Epilepsy & Behavior69: 153–160. doi:10.1016/j.yebeh.2016.11.008PMID 28188045.
  5. Jump up^ Quilichini PP, Chiron C, Ben-Ari Y, Gozlan H (2006). “Stiripentol, a putative antiepileptic drug, enhances the duration of opening of GABA-A receptor channels”Epilepsia47 (4): 704–16. doi:10.1111/j.1528-1167.2006.00497.xPMID 16650136.
  6. Jump up^ Trojnar MK, Wojtal K, Trojnar MP, Czuczwar SJ (2005). “Stiripentol. A novel antiepileptic drug” (PDF). Pharmacological reports : PR57 (2): 154–60. PMID 15886413.
  7. Jump up^ Sada N, Lee S, Katsu T, Otsuki T, Inoue T (2015). “Targeting LDH enzymes with a stiripentol analog to treat epilepsy”Science347 (6228): 1362–67. doi:10.1126/science.aaa1299PMID 25792327.
  8. Jump up to:a b “Scientific evaluation” (PDF). EMA. 2007.
  9. Jump up^ “Stiripentol (Diacomit): For Severe Myoclonic Epilepsy in Infancy (Dravet Syndrome)” (PDF). Canadian Agency for Drugs and Technologies in Health. April 2015.
  10. Jump up^ Kossoff, E (January 2014). “Stiripentol for dravet syndrome: is it worth it?”Epilepsy Currents14 (1): 22–3. doi:10.5698/1535-7597-14.1.22PMC 3913306Freely accessiblePMID 24526870.

////////////Stiripentol, fda 2018, Diacomit, 49763-96-4, BCX 2600, Estiripentol, Stiripentolum

CC(C)(C)C(C=CC1=CC2=C(C=C1)OCO2)O

Glycopyrronium bromide, гликопиррония бромид , بروميد غليكوبيرونيوم , 格隆溴铵 , グリコピロニウム臭化物

$
0
0

Glycopyrronium bromide.svg

ChemSpider 2D Image | glycopyrronium bromide | C19H28BrNO3

Glycopyrrolate.png

Glycopyrronium bromide

гликопиррония бромид [Russian] [INN]
بروميد غليكوبيرونيوم [Arabic] [INN]
格隆溴铵 [Chinese] [INN]
グリコピロニウム臭化物

Cas 596-51-0,

  • 3-Hydroxy-1,1-dimethylpyrrolidinium bromide α-cyclopentylmandelate (6CI,7CI)
  • Pyrrolidinium, 3-[(cyclopentylhydroxyphenylacetyl)oxy]-1,1-dimethyl-, bromide (9CI)
  • Pyrrolidinium, 3-hydroxy-1,1-dimethyl-, bromide, α-cyclopentylmandelate (8CI)
  • 1,1-Dimethyl-3-hydroxypyrrolidinium bromide α-cyclopentylmandelate
  • AHR-504
  • Asecryl
  • Copyrrolate
  • Gastrodyn
  • Glycopyrrolate
  • Glycopyrrolate bromide
  • Glycopyrrone bromide
  • Glycopyrronium bromide
  • NSC 250836
  • NSC 251251
  • NSC 251252
  • NVA 237
  • Nodapton
  • Robanul
  • Robinul
  • Seebri
  • Tarodyl
  • Tarodyn
  • β-1-Methyl-3-pyrrolidyl-α-cyclopentylmandelate methobromide

CAS FREE FORM OF ABOVE 13283-82-4

3-{[Cyclopentyl(hydroxy)phenylacetyl]oxy}-1,1-dimethylpyrrolidiniumbromide
3-Hydroxy-1,1-dimethylpyrrolidinium bromide α-cyclopentylmandelate
596-51-0 [RN]

Glycopyrrolate, ATC:A03AB02

  • Use:anticholinergic, antispasmodic
  • Chemical name:3-[(cyclopentylhydroxyphenylacetyl)oxy]-1,1-dimethylpyrrolidinium bromide
  • Formula:C19H28BrNO3, MW:398.34 g/mol
  • EINECS:209-887-0
  • LD50:15 mg/kg (M, i.v.); 570 mg/kg (M, p.o.);
    709 mg/kg (R, p.o.)
Glycopyrrolate
Title: Glycopyrrolate
CAS Registry Number: 596-51-0
CAS Name: 3-[(Cyclopentylhydroxyphenylacetyl)oxy]-1,1-dimethylpyrrolidinium bromide
Additional Names: 3-hydroxy-1,1-dimethylpyrrolidinium bromide a-cyclopentylmandelate; a-cyclopentylmandelic acid ester with 3-hydroxy-1,1-dimethylpyrrolidinium bromide; 1-methyl-3-pyrrolidyl a-cyclopentylmandelate methobromide; 1-methyl-3-pyrrolidyl a-phenyl-a-cyclopentylglycolate methobromide; 3-(2-phenyl-2-cyclopentylglycoloyloxy)-1,1-dimethylpyrrolidinium bromide; glycopyrronium bromide
Manufacturers’ Codes: AHR-504
Trademarks: Nodapton; Robanul; Robinul (Robins); Tarodyl; Tarodyn
Molecular Formula: C19H28BrNO3
Molecular Weight: 398.33
Percent Composition: C 57.29%, H 7.09%, Br 20.06%, N 3.52%, O 12.05%
Literature References: Synthetic, quaternary ammonium anticholinergic. Prepn: Franko, Lunsford, J. Med. Pharm. Chem.2, 523 (1960); Lunsford, US2956062 (1960 to A. H. Robins). Pharmacodynamics: E. Kaltiala et al.,J. Pharm. Pharmacol.26, 352 (1974). Toxicology: B. V. Franko et al.,Toxicol. Appl. Pharmacol.17, 361 (1970). Clinical comparison with atropine in anaesthetic practice: F. Kongsrud, S. Sponheim, Acta Anaesthesiol. Scand.26, 620 (1982); A. I. Webb, R. M. McMurphy, Am. J. Vet. Res.48, 1733 (1987); B. V. G. Malling et al.,Br. J. Anaesth.60, 426 (1988). Brief review of pharmacology and clinical use: R. K. Mirakhur, J. W. Dundee, Anaesthesia38, 1195-1204 (1983).
Properties: White crystals from butanone, mp 193.2-194.5°. Sol in water. LD50 (72 hr.) in female mice, female rats (mg/kg): 107, 196 i.p.; in male rats (mg/kg): 1150 orally (Franko).
Melting point: mp 193.2-194.5°
Toxicity data: LD50 (72 hr.) in female mice, female rats (mg/kg): 107, 196 i.p.; in male rats (mg/kg): 1150 orally (Franko)
Therap-Cat: Antispasmodic; preanesthetic medicant.
Therap-Cat-Vet: Preanesthetic medicant.
Keywords: Antimuscarinic; Antispasmodic
ALSO
Cas 51186-83-5
  • Pyrrolidinium, 3-[(cyclopentylhydroxyphenylacetyl)oxy]-1,1-dimethyl-, bromide, (R*,S*)-(±)-
  • Pyrrolidinium, 3-[[(2R)-cyclopentylhydroxyphenylacetyl]oxy]-1,1-dimethyl-, bromide, (3S)-rel- (9CI)
  • erythro-Glycopyrronium bromide

FREE FORM OF ABOVE 740028-90-4

 

Michael Woehrmann, Lara Terstegen, Stefan Biel, Thomas Raschke, Svenja-Kathrin Cerv, Werner Zilz, Sven Untiedt, Thomas Nuebel, Uwe Schoenrock, Heiner Max, Helga Biergiesser, Yvonne Eckhard, Heike Miertsch, Heike Foelster, Cornelia Meier-Zimmerer, Bernd Traupe, Inge Kruse, “GLYCOPYRROLATE IN COSMETIC PREPARATIONS.” U.S. Patent US20090208437, issued August 20, 2009.US20090208437

 EMA
Glycopyrronium bromide, the active substance of Enurev Breezhaler, is a well known active substance, chemically designated as 3-(2-cyclopentyl-2-hydroxy-2-phenylacetoxy)-1,1-dimethylpyrrolidinium bromide or (3RS)-3-[(2SR)-(2-cyclopentyl-2-hydroxy-2-phenylacetyl)oxy]-1,1-dimethylpyrrolidinium bromide, and has the following structure:
It is a white, non-hygroscopic powder, freely soluble in water, soluble in ethanol (96%), very slightly soluble in methylene chloride. The substance is also freely soluble in simulated lung fluid (phosphate buffer pH 7.4). Glycopyrronium bromide is a quaternary ammonium salt (ionic compound) and it is completely ionized between pH 1 and 14. It is a racemic mixture of the 3R,2S and 3S,2R stereoisomers. No optical rotation is seen in solution. Only single polymorphic form (crystalline Form A) has been reported.
Glycopyrronium bromide is a medication of the muscarinic anticholinergic group. It does not cross the blood–brain barrier and consequently has no to few central effects. It is available in by mouth, intravenous, and inhalated forms.It is a synthetic quaternary amine. It was developed by Sosei and licensed to Novartis in 2005. The cation, which is the active moiety, is called glycopyrronium (INN)[1] or glycopyrrolate (USAN).In June 2018, glycopyrronium was approved by the FDA to treat excessive underarm sweating becoming the first drug developed specifically to reduce excessive sweating.[2]

Glycopyrrolate is a muscarinic antagonist used as an antispasmodic, in some disorders of the gastrointestinal tract, and to reduce salivation with some anesthetics.

Glycopyrronium (as the bromide salt glycopyrrolate) is a synthetic anticholinergic agent with a quaternary ammonium structure. A muscarinic competitive antagonist used as an antispasmodic, in some disorders of the gastrointestinal tract, and to reduce salivation with some anesthetics. In October 2015, glycopyrrolate was approved by the FDA for use as a standalone treatment for Chronic obstructive pulmonary disease (COPD), as Seebri Neohaler.

Medical uses

In anesthesia, glycopyrronium injection can be used as a before surgery in order to reduce salivarytracheobronchial, and pharyngealsecretions, as well as decreasing the acidity of gastric secretion. It is also used in conjunction with neostigmine, a neuromuscular blocking reversal agent, to prevent neostigmine’s muscarinic effects such as bradycardia.

It is also used to reduce excessive saliva (sialorrhea),[3][4][5] and Ménière’s disease.[6]

It decreases acid secretion in the stomach and so may be used for treating stomach ulcers, in combination with other medications.

It has been used topically and orally to treat hyperhidrosis, in particular, gustatory hyperhidrosis.[7][8]

In inhalable form it is used to treat chronic obstructive pulmonary disease (COPD). Doses for inhalation are much lower than oral ones, so that swallowing a dose will not have an effect.[9][10]

Side effects

Since glycopyrronium reduces the body’s sweating ability, it can even cause hyperthermia and heat stroke in hot environments. Dry mouth, difficulty urinating, headachesdiarrhea and constipation are also observed side effects of the medication. The medication also induces drowsiness or blurred vision, an effect exacerbated by the consumption of alcohol.

Pharmacology

Mechanism of action

Glycopyrronium blocks muscarinic receptors,[11] thus inhibiting cholinergic transmission.

Pharmacokinetics

Glycopyrronium bromide affects the gastrointestinal tracts, liver and kidney but has a very limited effect on the brain and the central nervous system. In horse studies, after a single intravenous infusion, the observed tendencies of glycopyrronium followed a tri-exponential equation, by rapid disappearance from the blood followed by a prolonged terminal phase. Excretion was mainly in urine and in the form of an unchanged drug. Glycopyrronium has a relatively slow diffusion rate, and in a standard comparison to atropine, is more resistant to penetration through the blood-brain barrier and placenta.[12]

Research

It has been studied in asthma.[13][14]

Image result for Glycopyrronium bromide synthesis

Synthesis

https://data.epo.org/publication-server/rest/v1.0/publication-dates/20090513/patents/ep1856041nwb1/document.html

Image result for Glycopyrronium bromide synthesis

PATENT

https://patents.google.com/patent/CN103819384A/en

Image result for Glycopyrronium bromide synthesis

Figure CN103819384AD00041

PAtent

https://patents.google.com/patent/CN103159659A/en

Image result for Glycopyrronium bromide synthesis

glycopyrrolate (I)

Methyl ethyl ketone (20mL) IOOmL three-necked flask was added 8 (4.6g, 15mmol) was, at (Γ5 ° C was added dropwise dibromomethane (2.9g, 30mmol) in butanone (5 mL) was added dropwise completed, continued The reaction was stirred for 15min, and a white solid precipitated, was allowed to stand 36h at room temperature, filtered off with suction, the filter cake was sufficiently dried to give crude ketone was recrystallized twice to give a white powdery crystals I (3.9g, 66%) mp 191~193 ° C chromatographic purity 99.8% [HPLC method, mobile phase: lmol / L triethylamine acetate – acetonitrile – water (1: 150: 49); detection wavelength: 230nm, a measurement of the area normalization method] .MS m / z: 318 ( m-BrO 1HNMR (CD3OD) δ:! 1.33~1.38 (m, 2H), 1.55~1.70 (m, 6H), 2.11~2.21 (m, 1H), 2.67~2.80 (m, 1H), 3.02 (m, 1H), 3.06 (s, 3H), 3.23 (s, 3H), 3.59~3.71 (m, 3H), 3.90 (dd, /=13.8,1H), 5.47 (m, 1H), 7.27 (t, 1H) , 7.35 (t, 2H), 7.62 (dd, 2H) .13C bandit R (DMSO) δ: 27.0, 27.4, 28.0, 31.3, 47.8, 53.8, 54.3, 66.0, 71.3, 74.6, 81.1, 126.9,128.7,129.3 , 143.2 17 5.00

Patent

https://patents.google.com/patent/WO2016204998A1/en

Image result for Glycopyrronium bromide synthesis

PATENT

https://patents.google.com/patent/EP2417106B1/en

  • Glycopyrronium bromide, also known as 3-[(cyclopentylhydroxyphenylacetyl)oxy]-1,1-dimethylpyrrolidinium bromide or glycopyrrolate, is an antimuscarinic agent that is currently administered by injection to reduce secretions during anaesthesia and or taken orally to treat gastric ulcers.
  • [0003]

    It has the following chemical structure:

    Figure imgb0001
  • [0004]
    United States patent US 2,956,062 discloses that 1-methyl-3-pyrrolidyl alpha-cyclopentyl mandelate and can be prepared from methyl alpha cyclopentylmandelate and that the methyl bromide quaternary salt can be prepared by saturating a solution of 1-methyl-3-pyrrolidyl alpha-cyclopentyl mandelate in dry ethyl acetate with methyl bromide and filtering the crystalline solid that appears on standing.
  • [0005]
    The process of US 2,956,062 for preparing 1-methyl-3-pyrrolidyl alpha-cyclopentyl mandelate involves transesterifying methyl glycolate with an amino alcohol under the influence of metallic sodium to give a glycolate intermediate. Metallic sodium is highly reactive, which poses health and safety risks that make its use undesirable on an industrial scale for commercial manufacture.
  • [0006]
    The process of US 2,956,062 requires preparing the methylester in a previous step and alkylating the amino esters in a later step to form the desired quaternary ammonium salts.
  • [0007]
    The process of US 2,956,062 provides a mixture of diastereoisomers. The relative proportions of the diastereoisomers can vary widely between batches. This variation can give rise to surprising differences when preparing dry powder formulations from glycopyrronium bromide, which can cause problems when formulating such dry powders for pharmaceutical use.
  • [0008]
    United States patent application US 2007/0123557 discloses 1-(alkoxycarbonylmethyl)-1-methylpyrrolidyl anticholinergic esters. It describes coupling (R)-cyclopentylmandelic acid with (R,S)-1-methyl-pyrrolidin-3-ol under Mitsunobu conditions to give pure (R)-stereoisomeric compounds that are reacted with a bromoacetate to give the desired esters. It should be noted however that the chemicals used in Mitsunobu reactions, typically dialkyl azodicarboxylates and triphenylphosphine, pose health, safety and ecological risks that make their use undesirable on an industrial scale for commercial manufacture. They are also generally too expensive to source and too laborious to use in commercial manufacture.
  • [0009]
    United States patent application US 2006/0167275 discloses a process for the enrichment of the R, R- or S, S-configured glycopyrronium isomers and their thienyl analogues having R, S or S, R configuration.
  • [0010]
    WO 03/087094 A2 discloses new therapeutically useful pyrrolidinium derivatives, processes for their preparation and pharmaceutical compositions containing them.

Image result for Glycopyrronium bromide synthesis

EXAMPLE Example 1 Preparation of (3S,2’R)- and (3R,2’S)-3-[(cyclopentyl-hydroxyphenylacetyl)-oxy]-1,1-dimethylpyrrolidinium bromide

  • [0071]
    30 g of cyclopentyl mandelic acid, dissolved in 135 g dimethylformamide (DMF), were treated with 27 g carbonyldiimidazole at 18°C (in portions) to form the “active amide”. After the addition of 16.9 g of 1-methyl-pyrrolidin-3-ol, the mixture was heated to 60°C within 1 hour and stirred for 18 hours at this temperature. After checking for complete conversion, the mixture was cooled and 200 g water was added. The mixture was extracted with 200 g toluene and the extract was washed with water three times. The organic phase was concentrated to obtain cyclopentyl-hydroxy-phenyl-acetic acid 1-methyl-pyrrolidin-3-yl ester as an about 50% solution in toluene, ready to use for the next step.
  • [0072]
    This solution was diluted with 120 g of n-propanol and cooled to 0°C. 16.8 g methyl bromide was introduced and the mixture was stirred for 2 hours and then gradually heated to 60°C to evaporate the excess methyl bromide into a scrubber. The mixture was then cooled to 50°C and seed crystals were added to facilitate crystallisation. The temperature was then slowly reduced over 18 hours to 15°C. The solid was then isolated by filtration to obtain 22.7 g after drying. It was composed mainly of one pair of enantiomers, a racemic mixture of (3S,2’R)- and (3R,2’S)-3-[(cyclopentyl-hydroxyphenylacetyl)-oxy]-1,1-dimethylpyrrolidinium bromide, with a purity greater than 90% (by HPLC). The other pair of diastereoisomers ((3R,2’R)- and (3S,2’S)-3-[(cyclopentyl-hydroxyphenyl-acetyl)-oxyl-1,1-dimethylpyrrolidinium bromide) remains mainly in the filtrate as those compounds are significantly more soluble in n-propanol than the other stereoisomers.
  • [0073]
    The solid obtained is further recrystallised in n-propanol (1:10 wt) to give pure (3S,2’R)- and (3R,2’S)-3-[(cyclopentyl-hydroxyphenylacetyl)-oxy]-1,1-dimethylpyrrolidinium bromide i.e. purity > 99.9% as determined by high performance liquid chromatography (HPLC).
  • [0074]

    This process is summarised in the following reaction scheme:

    Figure imgb0020

Reference Example 2 Preparation of cyclopentyl-hydroxy-phenyl-acetic acid 1-methyl-pyrrolidin-3yl-ester in toluene

  • [0075]
    1 g of cyclopentyl mandelic acid was suspended in 4.7 g of toluene and 1.5 g of carbonyldiimidazole were added as a solid. After 30 minutes 0.69 g of 1-methyl-pyrrolidin-3-ol and 20 mg of sodium tert-butylate were added. The mixture was stirred at room temperature for 18 hours then water was added. After stirring the phases were separated and the organic phase was washed with water twice and evaporated to obtain an approximately 50% solution of cyclopentyl-hydroxy-phenyl-acetic acid 1-methyl-pyrrolidin-3yl-ester in toluene.

Example 3 Preparation of 2-cyclopentyl-2-hydroxy-1-imidazol-1-yl-2-phenyl-ethanone, the active intermediate

  • [0076]
    The imidazolidyl derivative of cyclopentylmandelic acid was prepared and isolated as a solid by the following method:
  • [0077]
    10 g of cyclopentylmandelic acid were suspended in 30 ml of acetonitrile and the mixture was cooled to 0°C. 10.3 g of carbonyldiimidazole were added as a solid and the mixture was warmed to room temperature for 2 hours. Carbon dioxide evolved as a gas as a precipitate formed. The mixture was then cooled to 5°C and the solid was filtered, washed with acetonitrile and dried in vacuum at 40°C to obtain 7.3 g of pure 2-cyclopentyl-2-hydroxy-1-imidazol-1-yl-2-phenyl-ethanone.
  • [0078]

    This process is summarised in the following reaction scheme:

    Figure imgb0021
  • [0079]
    High resolution MS-spectroscopy revealed the molecular formula of the compound (as M+H) to be C16H19O2N2 with an exact mass of 271.14414 (0.14575ppm deviation from the calculated value).
    1H-NMR-spectroscopy (600MHz, DMSO-d6): 1.03-1.07 (m, 1H), 1.25-1.30 (m, 1H), 1.35-1.40 (m, 1H), 1.40-1.50 (m, 1H), 1.53-1.56 (m, 2H), 1-60-1.67 (m, 1H), 1.75-1.84 (m, 1H), 1.03 – 1.85 (8H, 8 secondary CH2-protons in the cyclopentylring, H-C11, H-C12, H-C13, H-C14); 2.7-2.9 (m, 1H, H-C10); 6.76 (1H, H-C5); 6.91 (1H, H-C4); 7.29 (1H, H-C18); 7.39 (2H, H-C17, H-C19); 7.49 (2H, H-C16, H-C20); 7.65 (1H, H-C2).
  • [0080]

    The compound was characterised by IR-spectroscopy (measured as a solid film on a BRUKER TENSOR 27 FT-IR spectrometer over a wave number range of 4000-600 cm-1 with a resolution of 4 cm-1). An assignment of the most important bands is given below:

    Wavenumber (cm-1) Assignments
    3300 ∼ 2500 O-H stretching
    3167, 3151, 3120 Imidazole CH stretching
    2956, 2868 Cyclopentyl CH stretching
    1727 C=O stretching
    1600, 1538, 1469 Aromatic rings stretching
    735 Mono-subst. benzene CH o.o.p. bending
    704 Mono-subst. benzene ring o.o.p. bending

SYN

PAPER

https://link.springer.com/article/10.1007/s41981-018-0015-4

Sequential α-lithiation and aerobic oxidation of an arylacetic acid – continuous-flow synthesis of cyclopentyl mandelic acid

Open Access

Communications

Image result for Glycopyrronium bromide synthesis

The medicinal properties of glycopyrronium bromide (glycopyrrolate, 4) were first identified in the late 1950s [1]. Glycopyrrolate is an antagonist of muscarinic cholinergic receptors and is used for the treatment of drooling or excessive salivation (sialorrhea) [2], excess sweating (hyperhidrosis) [3], and overactive bladder and for presurgery treatment. In addition, it has recently been introduced as an effective bronchodilator for the treatment of chronic obstructive pulmonary disease (COPD) for asthma patients [4]. Glycopyrrolate displays few side effects because it does not pass through the blood brain barrier. Cyclopentyl mandelic acid (CPMA, 1), or its corresponding ester derivatives, are key intermediates in the synthetic routes to 4. CPMA (1) reacts with 1-methyl-pyrrolidin-3-ol (2) to form tertiary amine 3N-Methylation of 3 by methyl bromide gives quaternary ammonium salt glycopyrrolate 4 as a racemate (Scheme 1) [5].

Scheme 1

Synthesis of glycopyrrolate 4 from CPMA (1)

CPMA (1) is a synthetically challenging intermediate to prepare (Scheme 2). Routes A to D are most likely to be the commercially applied methods because these procedures are described in patents [5]. The published descriptions for the yields of 1 range from 28 to 56% for routes A to D. Ethyl phenylglyoxylate is reacted with cyclopentyl magnesium bromide to form an ester which is then hydrolyzed (route A) [6]. Phenylglyoxylic acid can be reacted in a similar manner with cyclopentyl magnesium bromide to directly form 1 (route B) [7]. Alternatively, the inverse addition of phenyl-Grignard reagent to cyclopentyl glyoxylic acid ester is reported (route C) [8]. Cyclopentyl glyoxylic acid ester can also be reacted with cyclopentadienyl magnesium bromide which is followed by an additional hydrogenation step with Pd/C and H2 to afford 1 (route D) [910].

Scheme 2

Existing synthetic pathways to CPMA (1)

PATENT

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2018154597&recNum=&maxRec=1000&office=&prevFilter=&sortOption=&queryString=&tab=PCTDescription

EXA M PL E S

EXAM PL E 1

Scheme 1

ST E P I

To a stirred solution of N-methyl pyrrol i din- 3-ol (2, 1 equiv) and Et3N (1.2 equiv) in dichloromethane was added a solution of 2-cyclopentyl-2-oxoacetyl chloride (1, 1.1 equiv) in DCM at O °C under nitrogen atmosphere for 20 min. The resulting solution was allowed to stir at room temperature over 10h. After completion, the mixture was quenched with water and extracted with diethyl ether to afford the pure product (3A).

Similarly, the product 3A is also obtained by reaction of 2 with other reagents, phenyl oxalic acid, methyl phenyl oxalate, and phenyl hemi-oxaldehyde respectively as shown in Scheme 1.

ST E P II

3A

To a mixture of bromobenzene (2.2 equiv) and Mg metal (2.2 equiv) in TH F (15 mL) was stirred over a period of 30 min at 0 · C. To this mixture, a solution of 1 -methyl pyrrol idin-3-yl 2-cyclopentyl-2-oxoacetate (3, 1 equiv) in T HF was added in portions over a period of 30 min. Up on completion, the reaction mixture was poured into ice water and extracted with ethyl acetate. The organic layer was separated and concentrated in vacuo. The resulting residue was purified by column chromatography to afford the pure product (5).

ST E P III

To a solution of compound 5 (1 equiv) in acetonitrile and chloroform mixture (10 mL, 2:3) was added methyl bromide (4 equiv). The mixture was stirred at room temperature for 72h. The solvents were evaporated, and the resulting residue was washed with diethyl ether to afford the pure product (6) as a white solid.

EXAM PL E 2

Scheme 2

ST E P I

To a stirred solution of N-methyl pyrrol i din- 3-ol (2, 1 equiv) and Et3N (1.2 equiv) in dichloromethane was added a solution of 2- oxo-2- phenyl acetyl chloride (1.1 equiv) in dichloromethane at 0 °C under nitrogen atmosphere for 15 min. The resulting solution was allowed to stir at room temperature over 12h. After completion, the mixture was quenched with water and extracted with diethyl ether to afford the pure product (3B).

Similarly, the product 3B is also obtained by reaction of 2 with other reagents, phenyl oxalic acid, methyl phenyl oxalate, and phenyl hemi-oxaldehyde respectively as shown in Scheme 2.

ST E P II

To a mixture of cyclopentyl bromide (4, 2.2 equiv) and Mg metal (2.2 equiv) in THF (15 mL) was stirred over a period of 30 min at 0 – C. To this mixture, a solution of 1-methylpyrrolidin-3-yl-2-oxo-2-phenylacetate (3B, 1 equiv) in TH F was added in portions over a period of 30 min. Up on completion, the reaction mixture was poured into ice water and extracted with ethyl acetate. The organic layer was separated and concentrated in vacuo. The resulting residue was purified by column chromatography to afford the pure product (5).

ST E P III

To a solution of compound 5 (1 equiv) in acetonitrile and chloroform mixture (10 mL, 2:3) was added methyl bromide (4 equiv). The mixture was stirred at room temperature for 75h. The solvents were evaporated, and the resulting residue was washed with diethyl ether to afford the pure product (6) as a white solid.

The invention has been described in detail with reference to preferred embodiments thereof. However, it will be appreciated by those skilled in the art that changes may be made in these embodiments without departing from the principles and nature of the invention, the scope of which is defined in the appended claims and their equivalents.

CN102388021A *2009-04-092012-03-21诺瓦提斯公司Process for preparing pyrrolidinium salts
CN102627595A *2012-03-092012-08-08徐奎Method for preparation of glycopyrronium bromide
CN103159659A *2011-12-192013-06-19沈阳药科大学Preparation method of muscarinic receptor antagonist glycopyrronium bromide

References

  1. Jump up^ Bajaj V, Langtry JA (July 2007). “Use of oral glycopyrronium bromide in hyperhidrosis”Br. J. Dermatol157 (1): 118–21. doi:10.1111/j.1365-2133.2007.07884.xPMID 17459043.
  2. Jump up^ “FDA OKs first drug made to reduce excessive sweating”AP News. Retrieved 2018-07-02.
  3. Jump up^ Mier RJ, Bachrach SJ, Lakin RC, Barker T, Childs J, Moran M (December 2000). “Treatment of sialorrhea with glycopyrrolate: A double-blind, dose-ranging study”Arch Pediatr Adolesc Med154 (12): 1214–8. doi:10.1001/archpedi.154.12.1214PMID 11115305.
  4. Jump up^ Tscheng DZ (November 2002). “Sialorrhea – therapeutic drug options”Ann Pharmacother36 (11): 1785–90. doi:10.1345/aph.1C019PMID 12398577.[permanent dead link]
  5. Jump up^ Olsen AK, Sjøgren P (October 1999). “Oral glycopyrrolate alleviates drooling in a patient with tongue cancer”J Pain Symptom Manage18 (4): 300–2. doi:10.1016/S0885-3924(99)00080-9PMID 10534970.
  6. Jump up^ Maria, Sammartano Azia; Claudia, Cassandro; Pamela, Giordano; Andrea, Canale; Roberto, Albera (1 December 2012). “Medical therapy in Ménière’s disease”Audiological Medicine10 (4): 171–177. doi:10.3109/1651386X.2012.718413 – via Taylor and Francis+NEJM.
  7. Jump up^ Kim WO, Kil HK, Yoon DM, Cho MJ (August 2003). “Treatment of compensatory gustatory hyperhidrosis with topical glycopyrrolate”. Yonsei Med. J44 (4): 579–82. doi:10.3349/ymj.2003.44.4.579PMID 12950111.
  8. Jump up^ Kim WO, Kil HK, Yoon KB, Yoon DM (May 2008). “Topical glycopyrrolate for patients with facial hyperhidrosis”Br. J. Dermatol158 (5): 1094–7. doi:10.1111/j.1365-2133.2008.08476.xPMID 18294315.
  9. Jump up^ “EPAR – Product information for Seebri Breezhaler” (PDF). European Medicines Agency. 28 September 2012.
  10. Jump up^ Tzelepis G, Komanapolli S, Tyler D, Vega D, Fulambarker A (January 1996). “Comparison of nebulized glycopyrrolate and metaproterenol in chronic obstructive pulmonary disease”Eur. Respir. J9 (1): 100–3. doi:10.1183/09031936.96.09010100PMID 8834341.
  11. Jump up^ Haddad EB, Patel H, Keeling JE, Yacoub MH, Barnes PJ, Belvisi MG (May 1999). “Pharmacological characterization of the muscarinic receptor antagonist, glycopyrrolate, in human and guinea-pig airways”Br. J. Pharmacol127 (2): 413–20. doi:10.1038/sj.bjp.0702573PMC 1566042Freely accessiblePMID 10385241.
  12. Jump up^ Rumpler, M.J.; Colahan, P.; Sams, R.A. (2014). “The pharmacokinetics of glycopyrrolate in Standardbred horses”. J. Vet Pharmacol Ther37 (3): 260–8. doi:10.1111/jvp.12085PMID 24325462.
  13. Jump up^ Hansel TT, Neighbour H, Erin EM, et al. (October 2005). “Glycopyrrolate causes prolonged bronchoprotection and bronchodilatation in patients with asthma”Chest128 (4): 1974–9. doi:10.1378/chest.128.4.1974PMID 16236844.
  14. Jump up^ Gilman MJ, Meyer L, Carter J, Slovis C (November 1990). “Comparison of aerosolized glycopyrrolate and metaproterenol in acute asthma”Chest98 (5): 1095–8. doi:10.1378/chest.98.5.1095PMID 2225951.
Glycopyrronium bromide
Glycopyrronium bromide.svg
Clinical data
Trade names Robinul, Cuvposa, Seebri, Qbrexza, others
License data
Pregnancy
category
  • AU: B2
  • US: B (No risk in non-human studies)
ATC code
Legal status
Legal status
Identifiers
CAS Number
PubChemCID
ChemSpider
UNII
ECHA InfoCard 100.008.990 Edit this at Wikidata
Chemical and physical data
Formula C19H28BrNO3
Molar mass 398.335 g/mol
3D model (JSmol)
Glycopyrronium
Glycopyrrolate.svg
Clinical data
AHFS/Drugs.com Monograph
MedlinePlus a602014
Pregnancy
category
  • US: B (No risk in non-human studies)
Routes of
administration
By mouthintravenous, inhalation
ATC code
Legal status
Legal status
Pharmacokinetic data
Elimination half-life 0.6–1.2 hours
Excretion 85% renal, unknown amount in the bile
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
ECHA InfoCard 100.008.990 Edit this at Wikidata
Chemical and physical data
Formula C19H28NO3+
Molar mass 318.431 g/mol
3D model (JSmol)
///////////Glycopyrronium bromide, гликопиррония бромид بروميد غليكوبيرونيوم 格隆溴铵 596-51-0, Glycopyrrolate, ATC:A03AB02, Use:anticholinergic, antispasmodic, グリコピロニウム臭化物 , 
C[N+]1(CCC(C1)OC(=O)C(C2CCCC2)(C3=CC=CC=C3)O)C.[Br-]

Sodium zirconium cyclosilicate, ナトリウムジルコニウムシクロケイ酸塩

$
0
0

242800-27-7.png

ZS-9 structure.png

Image result for Sodium zirconium cyclosilicate

str1

Sodium zirconium cyclosilicate

ZS-9, ZS 9, UZSi-9

CAS 242800-27-7, H2 O3 Si . x H2 O . 2/3 Na . 1/3 Zr, Sodium zirconium cyclosilicate; Silicic acid (H2SiO3), Sodium zirconium(4+) salt (3:2:1), hydrate

USAN CAS 17141-74-1, H6 O9 Si3 . 2 Na . Zr, Silicic acid (H2SiO3), sodium zirconium(4+) salt (3:2:1), hydrate, Sodium zirconium silicate (Na2ZrSi3O9) hydrate

ナトリウムジルコニウムシクロケイ酸塩

ZrH4O6. 3H4SiO4. 2H2O. 2Na, 561.6068, AS IN kegg

Molecular Formula, H6-O9-Si3.2Na.Z, Molecular Weight, 371.5004 as in chemid plus

APPROVED FDA 2018/5/18, LOKELMA, NDA 207078

APPROVED EMA 2018/3/22, LOKELMA

ATC code: V03AE10

UNII-D652ZWF066

TREATMENT
selective cation exchanger
Treatment of hyperkalemia

Sodium zirconium cyclosilicate (ZS-9) is a selective oral sorbent that traps potassium ions throughout the gastrointestinal tract. It is being developed by ZS Pharma and AstraZeneca for the treatment of hyperkalemia (elevated serum potassium levels).[1]

The product was originated at ZS Pharma, a wholly owned subsidiary of AstraZeneca. In 2015, ZS Pharma was acquired by AstraZeneca.

Hyperkalaemia is the presence of an abnormally high concentration of potassium in the blood. Most data on the occurrence of hyperkalaemia have been obtained from studies of hospitalised patients, and the incidence ranges from 1 to 10%. There is no agreed definition of hyperkalaemia, since the raised level of potassium at which a treatment should be initiated has not been established. The European Resuscitation Council guidelines consider hyperkalaemia to be a serum potassium (S-K) level > 5.5 mmol/L, with mild elevations defined as 5.5 to 5.9 mmol/L, moderate as 6.0-6.4 mmol/L, and severe as ≥ 6.5 mmol/L. The guidelines also note that extracellular potassium levels are usually between 3.5 and 5.0 mmol/L, which is considered the normal range for adults. However, a number of recent retrospective studies have shown the risk of mortality is increased even with only modest elevations of S-K. Mortality risk has been shown to be significantly higher in chronic kidney disease (CKD) patients with S-K levels > 5.0 mmol/L. In acute myocardial infarction patients, a mean postadmission S-K ≥ 5.5 mmol/L during hospitalisation corresponded to a 12-fold increase in death compared with S-K levels between 3.5 and 4.5 mmol/L but, more importantly, S-K levels between 4.5 and 5.0 mmol/L, which is within the normal range, were associated with a 2-fold increased risk of mortality compared with S-K between 3.5 and 4.5 mmol/L.

Sodium zirconium cyclosilicate (ZS) has been developed as treatment for hyperkalaemia. The indication applied for is: Treatment of hyperkalaemia in adult patients, acute and extended use. ZS is an inorganic cation exchange crystalline compound. ZS has a high capacity to selectively entrap monovalent cations, specifically excess potassium and ammonium ions, over divalent cations such as calcium and magnesium, in the gastrointestinal tract. The high specificity of ZS for potassium is attributable to the chemical composition and diameter of the micro pores, which act in an analogous manner to the selectivity filter utilized by physiologic potassium channels. The exchange with potassium ions occurs throughout the gastrointestinal tract with onset in the upper part of the gastrointestinal tract. The trapped potassium ions are excreted from the body via the faeces, thereby reducing any excess and resolving hyperkalaemia. As claimed by the applicant, ZS demonstrates improved capacity, selectivity, and speed for entrapping excess potassium over currently available options for the treatment of hyperkalaemia. The proposed commercial formulation of ZS is a non-absorbed, insoluble, white crystalline powder for suspension with a specific particle size distribution profile. The proposed starting dose of ZS for reversal of hyperkalaemia (when serum potassium is > 5.0 mmol/l) is up to 10 g/day, divided in 3 doses (TID) to achieve normokalaemia.

EMA

The chemical name of the active substance is hydrogen sodium zirconium (IV) silicate hydrate. Due to the natural variability in the manufacturing process of the active substance, it is expected to have the formula Na~1.5H~0.5ZrSi3O9 • 2–3 H2O and relative molecular mass in the range of 390.5 – 408.5. The WHO chose not to designate an INN for the active substance, and a USAN sodium zirconium cyclosilicate is used throughout the dossier and this CHMP AR. The active substance has the following structure:

str1

Figure 1. Stick-and-ball (left) and polyhedral (right) unit cell structural representation of the main framework of the microporous sodium zirconium cyclosilicate active substance. Red = zirconium, green = silicon, blue = oxygen atoms. Cations are not pictured.

The structure of sodium zirconium cyclosilicate is a cubic cell arrangement of octahedrally coordinated Zr and tetrahedrally coordinated Si units that interconnect through oxygen bridges as Zr–O–Si and Si–O–Si. The two types of units are observed in a ratio 1:3, respectively, and repeat orderly to form a three-dimensional framework characteristic of the compound. The framework acquires its negative charge from the octahedral fractions, [ZrO6]2– , and features channels and cavities that interconnect and locate the positive ions that counter-balance the negative charge of the framework. Electrostatic interactions between the framework and the cations allow for mobility and possibility of exchange with other cations that would fit and pass the free pore openings of ~ 3.0 Å. The uniform micropore structure allows a high exchange capacity and selectivity for potassium (K+) and ammonium (NH4 +) cations, providing the compound with its distinctive ion-exchange selectivity features responsible for its mode of action. In vitro characterisation of ion selectivity of sodium zirconium cyclosilicate was provided by the applicant and considered satisfactory

The structure of sodium zirconium cyclosilicate was confirmed using synchrotron powder diffraction, standard X-ray powder diffraction, 29Si magic angle spinning solid nuclear magnetic resonance studies (29Si-MASNMR), Fourier transform infrared spectroscopy, inductive coupled plasma-optical emission spectrometry, wave dispersive X-ray microprobe analysis and thermo-gravimetric analysis. Calculations using proprietary software were also used for structure elucidation. The active substance is a white crystalline powder. Bonding interactions in the main framework are considered primarily of covalent nature, with some ionic contribution due to the difference in electronegativity between Si–O and Zr–O. The covalent bonding interactions in all directions within the crystals make sodium zirconium cyclosilicate a compound insoluble in water or in organic solvents. It is neither hygroscopic nor sensitive to light and it is resistant to heat. During the hydrothermal synthesis, the possibility that other crystalline phases are formed exists. The observed crystalline forms are controlled by the manufacturing process parameters and release specifications. Sodium zirconium cyclosilicate is considered to be a new active substance. The applicant demonstrated that neither it, nor its derivatives have ever been active substances in medicinal products authorised in the EU………http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Public_assessment_report/human/004029/WC500246776.pdf

TGA

DOC]Australian Public Assessment Report for Sodium zirconium … – TGA

Jan 29, 2018 – The sponsor has submitted an application to register a new chemical entity Lokelma,sodium zirconium cyclosilicate hydrate powder for …

The chemical formula of sodium zirconium cyclosilicate hydrate is Na~1.5H~0.5ZrSi3O9.2-3H2O.

str1

The drug substance ‘sodium zirconium cyclosilicate hydrate’ (abbreviated to ZS) is a white crystalline powder. The structure of ZS is summarised as a cubic cell arrangement of octahedrally coordinated zirconium Zr ([ZrO6]2-) and tetrahedrally coordinated silicon Si ([SiO4]0) units that interconnect through oxygen bridges as Zr-O-Si and Si-O-Si. The two types of units are observed in a ratio of 1:3, respectively, and repeat orderly to form a three dimensional framework characteristic of the compound. The framework acquires its negative charge from the octahedral fractions, [ZrO6]2- and features channels and cavities that interconnect and locate the positive ions (sodium, Na+, and hydrogen, H+) that counter balance the negative charge of the framework.

The manufacturing process is tightly controlled in terms of order of addition of starting material, reaction and crystallisation temperatures, mixing speeds and times, and minimum number of rinses, in order to meet expected yields of the drug substance of an expected quality. In process quality control tests [information redacted] are applied during the manufacturing process to ensure the formation of the correct crystalline structure and batch to batch consistency.

Sodium zirconium cyclosilicate hydrate is completely insoluble.

The drug substance forms part of a family of zirconium silicates that have specific ion exchange properties. Its mechanism of action is based on the cations within its porous crystalline structure, and their ability to freely exchange with a select group of monovalent cations, most specifically the potassium (K+) and ammonium (NH4+) cations. The pore size within the three dimensional crystalline structure has been measured at ~3Å (2.4 x 3.5 Å[1]), which is sufficiently wide enough to trap the potassium monovalent cations which have an approximate ionic diameter of 2.98Å.

The particle size of the drug substance is controlled to maintain a non-systemic mode of action. The sponsor adequately justified not routinely controlling the size of larger particles in the drug substance as differences in particle size were shown to not affect performance as measured by potassium ion exchange capacity (KEC), and there was no correlation between KEC and D90 for clinical lots manufactured.

There are two alternate zirconium silicate crystalline phases which may be formed in the reaction process; Crystalline Phase A (CPA) and Crystalline Phase B (CPB). These layered, two-dimensional structures also exhibit ion exchange properties, although their ion selectivity is less specific for the potassium K+ cations compared to the desired drug substance. PXRD techniques are used to differentiate between the desired drug substance and levels of CPA and CPB. Appropriate limits are applied in the drug substance specification to limit the content of these crystalline phases in the drug substance/drug product.

The quality of the drug substance is controlled by an acceptable specification that includes test and limits for Appearance, Identification (by FTIR and PXRD), KEC , Crystalline Phase A , Crystalline Phase B , Zirconium content , Silicon content , Hafnium content , Moisture content , Particle Size , and Elemental Impurities.

[1] 1 Å = 0.1 nm.

Image result for Sodium zirconium cyclosilicate

Background

Hyperkalemia occurs in 3 to 10% of hospitalized patients[2] but is often mild. Hyperkalemia can arise from impaired renal functionpotassium-sparing diuretics and renin–angiotensin system blockers (e.g., ACE inhibitorsangiotensin receptor blockersspironolactone) and diabetes mellitus.[2][3][4][5]

There is no universally accepted definition of what level of hyperkalemia is mild, moderate, or severe.[6] However, if hyperkalemia causes any ECG change it is considered a medical emergency[6] due to a risk of potentially fatal abnormal heart rhythms (arrhythmia) and is treated urgently.[6] serum potassium concentrations greater than 6.5 to 7.0 mmol/L in the absence of ECG changes are managed aggressively.[6]

Hyperkalemia, particularly if severe, is a marker for an increased risk of death.[2] However, there is disagreement regarding whether a modestly elevated serum potassium level directly causes significant problems. One viewpoint is that mild to moderate hyperkalemia is a secondary effect that denotes significant underlying medical problems.[2] Accordingly, these problems are both proximate and ultimate causes of death,[2] and adjustment of potassium may not be helpful. Alternatively, hyperkalemia may itself be an independent risk factorfor cardiovascular mortality.[7]

Several approaches are used in the treatment of hyperkalemia.[6] In October 2015, the U.S. Food and Drug Administration (FDA) approved patiromer which works by binding free potassium ions in the gastrointestinal tract and releasing calcium ions for exchange. Previously, the only approved product was sodium polystyrene sulfonate (Kayexalate),[8] an organic ion-exchange resin that nonspecifically binds cations (e.g., calciumpotassiummagnesium) in the gastrointestinal tract. The effectiveness of sodium polystyrene sulfonate has been questioned: a study in healthy subjects showed that laxatives alone were almost as effective in increasing potassium secretion as laxatives plus Kayexalate.[9] In addition, use of sodium polystyrene sulfonate, particularly if formulated with high sorbitol content, is uncommonly but convincingly associated with colonic necrosis.[6][8][10][11]

Mechanism of action

Cross-sections of ZS-9 pores with three different ions (K⁺ = potassium, Na⁺ = sodium, Ca²⁺ = calcium). The specificity for potassium is thought to be caused by the diameter and composition of the pores, which resembles potassium channels.

ZS-9 is a zirconium silicate. Zirconium silicates have been extensively used in medical and dental applications because of their proven safety.[12] 11 zirconium silicates were screened by an iterative optimization process. ZS-9 selectively captures potassium ions, presumably by mimicking the actions of physiologic potassium channels.[13] ZS-9 is an inorganic cation exchanger crystalline with a high capacity to entrap monovalent cations, specifically potassium and ammonium ions, in the GI tract. ZS-9 is not systemically absorbed; accordingly, the risk of systemic toxicity may be minimized.

Clinical studies

phase 2 clinical trial in 90 patients with chronic kidney disease and mild-to-moderate hyperkalemia found a significantly greater reduction in serum potassium with ZS-9 than placebo. ZS-9 was well tolerated, with a single adverse event (mild constipation).[14]

double-blindphase 3 clinical trial in 753 patients with hyperkalemia and underlying chronic kidney diseasediabetescongestive heart failure, and in patients on renin–angiotensin system blockers compared ZS-9 with placebo.[15] Patients were randomly assigned to receive either ZS-9 (1.25 g, 2.5 g, 5 g, or 10 g) or placebo 3 times daily for 48 hours (acute phase). Patients who achieved normokalemia (serum potassium of 3.5-4.9 mmol/L) were randomly assigned to receive ZS-9 or placebo once daily for 12 additional days (maintenance phase). At the end of the acute phase, serum potassium significantly decreased in the 2.5 g, 5 g, and 10 g ZS-9 groups. During the maintenance phase, once daily 5 g or 10 g ZS-9 maintained serum potassium at normal levels. Adverse events, including specifically gastrointestinal effects, were similar with either ZS-9 or placebo.[15]

double-blindphase 3 clinical trial in 258 patients with hyperkalemia and underlying chronic kidney diseasediabetescongestive heart failure, and in patients on renin–angiotensin system blockers compared ZS-9 with placebo.[16] All patients received 10 g ZS-9 three times daily for 48 hours in the initial open-label phase. Patients who achieved normokalemia (serum potassium 3.5-5.0 mEq/L) were randomly assigned to receive either ZS-9 (5 g, 10 g, or 15 g) or placebo once daily for 28 days (double-blind phase). 98% of patients (n=237) achieved normokalemia during the open-label phase. During the double-blind phase, once daily 5 g, 10 g, and 15 g ZS-9 maintained serum potassium at normal levels in a significantly higher proportion of patients (80%, 90%, and 94%, respectively) than placebo (46%). Adverse events were generally similar with either ZS-9 or placebo. Hypokalemiaoccurred in more patients in the 10 g and 15 g ZS-9 groups (10% and 11%, respectively), versus none in the 5 g ZS-9 or placebo groups.[16]

Regulatory

In the United States, regulatory approval of ZS-9 was rejected by the Food and Drug Administration in May 2016 due to issues associated with manufacturing.[17] On May 18th, 2018, the FDA approved ZS-9 (now known as Lokelma®) for treatment of adults with hyperkalemia.[18]

PATENT

WO 2012109590

PATENT

WO 2015070019

https://patents.google.com/patent/WO2015070019A1/en

The present invention relates to novel zirconium silicate (“ZS”) compositions which are preferably sodium zirconium cyclosilicates having an elevated level of ZS-9 crystalline form relative to other forms of zirconium cyclosilicates (i.e., ZS-7) and zirconium silicates (i.e., ZS-8, ZS-11). The ZS compositions are preferably sodium zirconium cyclosilicate compositions where the crystalline form has at least 95% ZS-9 relative to other crystalline forms of zirconium silicate. The ZS compositions of the present invention unexpectedly exhibit a markedly improved in vivo potassium ion absorption profile and rapid reduction in elevate levels of serum potassium.

[004] Preferably ZS compositions of the present invention are specifically formulated at particular dosages to remove select toxins, e.g., potassium ions or ammonium ions, from the gastrointestinal tract at an elevated rate without causing undesirable side effects. The preferred formulations are designed to remove and avoid potential entry of particles into the bloodstream and potential increase in pH of urine in patients. The formulation is also designed to release less sodium into the blood. These compositions are particularly useful in the therapeutic treatment of hyperkalemia and kidney disease. The present invention also relates to pharmaceutical granules, tablets, pill, and dosage forms comprising the microporous ZS as an active ingredient. In particular, the granules, tablets, pills or dosage forms are compressed to provide immediate release, delayed release, or specific release within the subject. Also disclosed are microporous ZS compositions having enhanced purity and potassium exchange capacity (“KEC”). Methods of treating acute, sub-acute, and chronic hyperkalemia have also been investigated. Disclosed herein are particularly advantageous dosing regimens for treating different forms of hyperkalemia using the microporous ZS compositions noted above. In addition, the present invention relates to methods of co-administering microporous ZS compositions in combination with other pharmacologic drugs that are known to induce, cause, or exacerbate the hyperkalemic condition.

Patent

Publication numberPriority datePublication dateAssigneeTitle
US3329480A *1963-10-181967-07-04Union Oil CoCrystalline zircono-silicate zeolites
US4581141A *1978-02-271986-04-08Purdue Research FoundationDialysis material and method for removing uremic substances
US20050220752A1 *2004-03-302005-10-06Dominique CharmotIon binding polymers and uses thereof
US20110097401A1 *2009-06-122011-04-28Meritage Pharma, Inc.Methods for treating gastrointestinal disorders
US20120213847A1 *2011-02-112012-08-23ZS Pharma, Inc.Microporous zirconium silicate for the treatment of hyperkalemia
CA2084086C *1990-05-282002-10-08Steven M. KuznickiLarge-pored molecular sieves containing at least one octahedral site and tetrahedral sites of at least one type
US5338527A *1992-08-201994-08-16UopZirconium silicate composition, method of preparation and uses thereof
US5891417A *1997-04-081999-04-06Uop LlcZirconium silicate and zirconium germanate molecular sieves and process using the same
US5888472A *1997-04-081999-03-30Uop LlcZirconium silicate molecular sieves and process using the same
EP1038580B1 *1999-03-262005-05-25Uop LlcAmmonium ion adsorption process using zirconium silicate and zirconium germanate molecular sieves
US6332985B1 *1999-03-292001-12-25Uop LlcProcess for removing toxins from bodily fluids using zirconium or titanium microporous compositions
WO2002062356A3 *2001-02-062002-09-26Ash Medical Systems IncMonovalent-selective cation exchangers as oral sorbent therapy
CN104968336A *2012-07-112015-10-07Zs制药公司Microporous zirconium silicate for the treatment of hyperkalemia in hypercalcemic patients and improved calcium-containing compositions for the treatment of hyperkalemia
KR20150074053A *2012-10-222015-07-01제트에스 파마, 인코포레이티드Microporous zirconium silicate for treating hyperkalemia
Publication numberPriority datePublication dateAssigneeTitle
WO2017066128A1 *2015-10-142017-04-20ZS Pharma, Inc.Extended use zirconium silicate compositions and methods of use thereof
Family To Family Citations
US20160038538A1 *2013-11-082016-02-11ZS Pharma, Inc.Microporous zirconium silicate for the treatment of hyperkalemia

References

  1. Jump up^ “ZS-9. A selective potassium binder”. ZS-Pharma.
  2. Jump up to:a b c d e Elliott, M. J.; Ronksley, P. E.; Clase, C. M.; Ahmed, S. B.; Hemmelgarn, B. R. (2010). “Management of patients with acute hyperkalemia”Canadian Medical Association Journal182 (15): 1631–5. doi:10.1503/cmaj.100461PMC 2952010Freely accessiblePMID 20855477.
  3. Jump up^ Stevens, M. S.; Dunlay, R. W. (2000). “Hyperkalemia in hospitalized patients”. International Urology and Nephrology32 (2): 177–80. doi:10.1023/A:1007135517950PMID 11229629.
  4. Jump up^ Navaneethan, S. D.; Yehnert, H.; Moustarah, F.; Schreiber, M. J.; Schauer, P. R.; Beddhu, S. (2009). “Weight Loss Interventions in Chronic Kidney Disease: A Systematic Review and Meta-analysis”Clinical Journal of the American Society of Nephrology4 (10): 1565–74. doi:10.2215/CJN.02250409PMC 2758256Freely accessiblePMID 19808241.
  5. Jump up^ Tamirisa, K. P.; Aaronson, K. D.; Koelling, T. M. (2004). “Spironolactone-induced renal insufficiency and hyperkalemia in patients with heart failure”. American Heart Journal148(6): 971–8. doi:10.1016/j.ahj.2004.10.005PMID 15632880.
  6. Jump up to:a b c d e f Taal, M.W.; Chertow, G.M.; Marsden, P.A.; Skorecki, K.; Yu, A.S.L.; Brenner, B.M. (2012). Brenner and Rector’s The Kidney (Chapter 17, page 672, 9th ed.). Elsevier. ISBN 978-1-4160-6193-9.
  7. Jump up^ Fang, J.; Madhavan, S.; Cohen, H.; Alderman, M. H. (2000). “Serum potassium and cardiovascular mortality”Journal of General Internal Medicine15 (12): 885–90. doi:10.1046/j.1525-1497.2000.91021.xPMC 1495719Freely accessiblePMID 11119186.
  8. Jump up to:a b Watson, M.; Abbott, K. C.; Yuan, C. M. (2010). “Damned if You Do, Damned if You Don’t: Potassium Binding Resins in Hyperkalemia”. Clinical Journal of the American Society of Nephrology5 (10): 1723–6. doi:10.2215/CJN.03700410PMID 20798253.
  9. Jump up^ Emmett, M.; Hootkins, R. E.; Fine, K. D.; Santa Ana, C. A.; Porter, J. L.; Fordtran, J. S. (1995). “Effect of three laxatives and a cation exchange resin on fecal sodium and potassium excretion”. Gastroenterology108 (3): 752–60. doi:10.1016/0016-5085(95)90448-4PMID 7875477.
  10. Jump up^ Sterns, R. H.; Rojas, M.; Bernstein, P.; Chennupati, S. (2010). “Ion-Exchange Resins for the Treatment of Hyperkalemia: Are They Safe and Effective?”. Journal of the American Society of Nephrology21 (5): 733–5. doi:10.1681/ASN.2010010079PMID 20167700.
  11. Jump up^ Kamel, K. S.; Schreiber, M. (2012). “Asking the question again: Are cation exchange resins effective for the treatment of hyperkalemia?”. Nephrology Dialysis Transplantation27(12): 4294–7. doi:10.1093/ndt/gfs293PMID 22989741.
  12. Jump up^ Denry I, Kelly JR. State of the art of zirconia for dental applications. Dental Materials. Volume 24, Issue 3, March 2008, Pages 299–307
  13. Jump up^ =Stavros, F (2014). “Characterization of Structure and Function of ZS-9, a K⁺ Selective Ion Trap”PLOS ONE9 (12): e114686. doi:10.1371/journal.pone.0114686PMC 4273971Freely accessiblePMID 25531770.
  14. Jump up^ Ash SR, et al. “Safety and efficacy of ZS-9, a novel selective cation trap, for treatment of hyperkalemia in CKD patients.” American Society of Nephrology 2013 conference, Late-Breaking Abstract.
  15. Jump up to:a b Packham DK, et al. (2014). “Sodium zirconium cyclosilicate in hyperkalemia”. New England Journal of Medicine372 (3): 222–31. doi:10.1056/NEJMoa1411487PMID 25415807.
  16. Jump up to:a b Kosiborod M, et al. (2014). “Effect of sodium zirconium cyclosilicate on potassium lowering for 28 days among outpatients with hyperkalemia”. Journal of the American Medical Association312 (21): 2223–33. doi:10.1001/jama.2014.15688PMID 25402495.
  17. Jump up^ Ben Adams (May 27, 2016). “AstraZeneca’s $2.7B hyperkalemia drug ZS-9 rejected by FDA”. FierceBiotech.
  18. Jump up^ https://www.drugs.com/history/lokelma.html
Sodium zirconium cyclosilicate
ZS-9 structure.png
Crystal structure of ZS-9. Blue spheres  =  oxygen atoms, red spheres  =  zirconium atoms, green spheres  =  silicon atoms.
Clinical data
Trade names Lokelma
Routes of
administration
Oral
ATC code
  • none
Legal status
Legal status
  • US: Rx-only
Pharmacokinetic data
Bioavailability Not absorbed
Excretion Stool
Identifiers
CAS Number
UNII
KEGG

//////////////Sodium zirconium cyclosilicate,  ナトリウムジルコニウムシクロケイ酸塩 , FDA 2018, EMA, 2018, EU 2018, ZS 9, UZSi-9

O[Si]1(O[Si](O[Si](O1)(O)O)(O)O)O.[Na+].[Na+].[Zr

Lanadelumab, ラナデルマブ

$
0
0

(Heavy chain)
EVQLLESGGG LVQPGGSLRL SCAASGFTFS HYIMMWVRQA PGKGLEWVSG IYSSGGITVY
ADSVKGRFTI SRDNSKNTLY LQMNSLRAED TAVYYCAYRR IGVPRRDEFD IWGQGTMVTV
SSASTKGPSV FPLAPSSKST SGGTAALGCL VKDYFPEPVT VSWNSGALTS GVHTFPAVLQ
SSGLYSLSSV VTVPSSSLGT QTYICNVNHK PSNTKVDKRV EPKSCDKTHT CPPCPAPELL
GGPSVFLFPP KPKDTLMISR TPEVTCVVVD VSHEDPEVKF NWYVDGVEVH NAKTKPREEQ
YNSTYRVVSV LTVLHQDWLN GKEYKCKVSN KALPAPIEKT ISKAKGQPRE PQVYTLPPSR
EEMTKNQVSL TCLVKGFYPS DIAVEWESNG QPENNYKTTP PVLDSDGSFF LYSKLTVDKS
RWQQGNVFSC SVMHEALHNH YTQKSLSLSP G
(Light chain)
DIQMTQSPST LSASVGDRVT ITCRASQSIS SWLAWYQQKP GKAPKLLIYK ASTLESGVPS
RFSGSGSGTE FTLTISSLQP DDFATYYCQQ YNTYWTFGQG TKVEIKRTVA APSVFIFPPS
DEQLKSGTAS VVCLLNNFYP REAKVQWKVD NALQSGNSQE SVTEQDSKDS TYSLSSTLTL
SKADYEKHKV YACEVTHQGL SSPVTKSFNR GEC
(dimer; dishulfide bridge: H22-H96, H149-H205, H225-L213, H231-H’231, H234-H’234, H266-H326, H372-H430, H’22-H’96, H’149-H’205, H’225-L’213, H’266-H’326, H’372-H’430, L23-L88, L133-L193, L’23-L’88, L’133-L’193)

Lanadelumab

DX 2930

Fda approved 2018/8/23, Takhzyro

Formula
C6468H10016N1728O2012S48
Cas
1426055-14-2
Mol weight
145714.225

Peptide, Monoclonal antibody
Prevention of angioedema in patients with hereditary angioedema

Immunomodulator, Plasma kallikrein inhibitor

breakthrough therapyUNII: 2372V1TKXK

Image result for Lanadelumab

Image result for Lanadelumab

Lanadelumab (INN) (alternative identifier DX-2930[1]) is a human monoclonal antibody (class IgG1 kappa)[2] that targets plasma kallikrein (pKal)[1] in order to promote prevention of angioedema in patients with hereditary angioedema.[3][4] In phase 1 clinical trialsLanadelumab was well tolerated and was reported to reduce cleavage of kininogen in the plasma of patients with hereditary angioedeman and decrease the number of patients experiencing attacks of angioedema.[1][5][6][7] As of 2017 ongoing trials for Lanadelumab include two phase 3 studies focused on investigating the utility of Lanadelumab in preventing of acute angioedema attacks in hereditary angioedema patients[8][9]

Image result for Lanadelumab

This drug was produced by Dyax Corp and currently under development by Shire.[10] Lanadelumab has been designated by the U.S. Food and Drug Administration (FDA) as a breakthrough therapy.[11]

Image result for Lanadelumab

References

  1. Jump up to:a b c Banerji, Aleena; Busse, Paula; Shennak, Mustafa; Lumry, William; Davis-Lorton, Mark; Wedner, Henry J.; Jacobs, Joshua; Baker, James; Bernstein, Jonathan A. (2017-02-23). “Inhibiting Plasma Kallikrein for Hereditary Angioedema Prophylaxis”. The New England Journal of Medicine376 (8): 717–728. doi:10.1056/NEJMoa1605767ISSN 1533-4406PMID 28225674.
  2. Jump up^ Kenniston, Jon A.; Faucette, Ryan R.; Martik, Diana; Comeau, Stephen R.; Lindberg, Allison P.; Kopacz, Kris J.; Conley, Gregory P.; Chen, Jie; Viswanathan, Malini (2014-08-22). “Inhibition of Plasma Kallikrein by a Highly Specific Active Site Blocking Antibody”The Journal of Biological Chemistry289 (34): 23596. doi:10.1074/jbc.M114.569061PMC 4156074Freely accessiblePMID 24970892.
  3. Jump up^ Statement On A Nonproprietary Name Adopted By The USAN Council – LanadelumabAmerican Medical Association.
  4. Jump up^ World Health Organization (2015). “International Nonproprietary Names for Pharmaceutical Substances (INN). Proposed INN: List 114”(PDF). WHO Drug Information29 (4).
  5. Jump up^ Chyung, Yung; Vince, Bradley; Iarrobino, Ryan; Sexton, Dan; Kenniston, Jon; Faucette, Ryan; TenHoor, Chris; Stolz, Leslie E.; Stevens, Chris (2014-10-01). “A phase 1 study investigating DX-2930 in healthy subjects”. Annals of Allergy, Asthma & Immunology113 (4): 460–466.e2. doi:10.1016/j.anai.2014.05.028ISSN 1534-4436PMID 24980392.
  6. Jump up^ “A Single Increasing Dose Study to Assess Safety and Tolerability of DX-2930 in Healthy Subjects – Full Text View – ClinicalTrials.gov”clinicaltrials.gov. Retrieved 2017-03-24.
  7. Jump up^ “Double-Blind, Multiple Ascending Dose Study to Assess Safety, Tolerability and Pharmacokinetics of DX-2930 in Hereditary Angioedema (HAE) Subjects – Full Text View – ClinicalTrials.gov”clinicaltrials.gov. Retrieved 2017-03-24.
  8. Jump up^ “Efficacy and Safety Study of DX-2930 to Prevent Acute Angioedema Attacks in Patients With Type I and Type II HAE – Full Text View – ClinicalTrials.gov”clinicaltrials.gov. Retrieved 2017-03-24.
  9. Jump up^ “Long-term Safety and Efficacy Study of DX-2930 to Prevent Acute Angioedema Attacks in Patients With Type I and Type II HAE – Full Text View – ClinicalTrials.gov”clinicaltrials.gov. Retrieved 2017-03-24.
  10. Jump up^ “Lanadelumab – AdisInsight”adisinsight.springer.com. Retrieved 2017-03-24.
  11. Jump up^ “Dyax Corp. Receives FDA Breakthrough Therapy Designation for DX-2930 for Prevention of Attacks of Hereditary Angioedema”http://www.businesswire.com. Retrieved 2017-03-24.
Lanadelumab
Monoclonal antibody
Type Whole antibody
Source Human
Target kallikrein
Clinical data
Synonyms DX-2930
ATC code
  • none
Identifiers
CAS Number
ChemSpider
  • none
UNII
Chemical and physical data
Formula C6468H10016N1728O2012S47
Molar mass 145.7 kDa

///////////Lanadelumab, Peptide, Monoclonal antibody, FDA 2018, ラナデルマブ ,Immunomodulator, Plasma kallikrein inhibitor, DX 2930,  breakthrough therapy, Takhzyro

“DRUG APPROVALS INTERNATIONAL” CATERS TO EDUCATION GLOBALLY, No commercial exploits are done or advertisements added by me. This is a compilation for educational purposes only. P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent

FDA approves new kind of treatment Lumoxiti (moxetumomab pasudotox-tdfk) for hairy cell leukemia

$
0
0
The U.S. Food and Drug Administration today approved Lumoxiti (moxetumomab pasudotox-tdfk) injection for intravenous use for the treatment of adult patients with relapsed or refractory hairy cell leukemia (HCL) who have received at least two prior systemic therapies, including treatment with a purine nucleoside analog. Lumoxiti is a CD22-directed cytotoxin and is the first of this type of treatment for patients with HCL.

September 13, 2018

Release

The U.S. Food and Drug Administration today approved Lumoxiti (moxetumomab pasudotox-tdfk) injection for intravenous use for the treatment of adult patients with relapsed or refractory hairy cell leukemia (HCL) who have received at least two prior systemic therapies, including treatment with a purine nucleoside analog. Lumoxiti is a CD22-directed cytotoxin and is the first of this type of treatment for patients with HCL.

“Lumoxiti fills an unmet need for patients with hairy cell leukemia whose disease has progressed after trying other FDA-approved therapies,” said Richard Pazdur, M.D., director of the FDA’s Oncology Center of Excellence and acting director of the Office of Hematology and Oncology Products in the FDA’s Center for Drug Evaluation and Research. “This therapy is the result of important research conducted by the National Cancer Institute that led to the development and clinical trials of this new type of treatment for patients with this rare blood cancer.”

HCL is a rare, slow-growing cancer of the blood in which the bone marrow makes too many B cells (lymphocytes), a type of white blood cell that fights infection. HCL is named after these extra B cells which look “hairy” when viewed under a microscope. As the number of leukemia cells increases, fewer healthy white blood cells, red blood cells and platelets are produced.

The efficacy of Lumoxiti was studied in a single-arm, open-label clinical trial of 80 patients who had received prior treatment for HCL with at least two systemic therapies, including a purine nucleoside analog. The trial measured durable complete response (CR), defined as maintenance of hematologic remission for more than 180 days after achievement of CR. Thirty percent of patients in the trial achieved durable CR, and the overall response rate (number of patients with partial or complete response to therapy) was 75 percent.

Common side effects of Lumoxiti include infusion-related reactions, swelling caused by excess fluid in body tissue (edema), nausea, fatigue, headache, fever (pyrexia), constipation, anemia and diarrhea.

The prescribing information for Lumoxiti includes a Boxed Warning to advise health care professionals and patients about the risk of developing capillary leak syndrome, a condition in which fluid and proteins leak out of tiny blood vessels into surrounding tissues. Symptoms of capillary leak syndrome include difficulty breathing, weight gain, hypotension, or swelling of arms, legs and/or face. The Boxed Warning also notes the risk of hemolytic uremic syndrome, a condition caused by the abnormal destruction of red blood cells. Patients should be made aware of the importance of maintaining adequate fluid intake, and blood chemistry values should be monitored frequently. Other serious warnings include: decreased renal function, infusion-related reactions and electrolyte abnormalities. Women who are breastfeeding should not be given Lumoxiti.

The FDA granted this application Fast Track and Priority Review designations. Lumoxiti also received Orphan Drug designation, which provides incentives to assist and encourage the development of drugs for rare diseases.

The FDA granted the approval of Lumoxiti to AstraZeneca Pharmaceuticals.

///////////// Lumoxiti, moxetumomab pasudotox-tdfk, fda 2018, Fast Track, Priority Review designations,  Orphan Drug designation,

Fipronil, 芬普尼 , フィプロニル

$
0
0

2D chemical structure of fipronilChemSpider 2D Image | Fipronil | C12H4Cl2F6N4OS

120068-37-3.png

Fipronil

  • Molecular Formula C12H4Cl2F6N4OS
  • Average mass 437.148 Da
(±)-5-Amino-1-(2,6-dichloro-a,a,a-trifluoro-p-tolyl)-4-trifluoromethylsulfinylpyrazole-3-carbonitrile
(±)-Fipronil
120068-37-3 [RN]
1H-Pyrazole-3-carbonitrile, 5-amino-1-[2,6-dichloro-4-(trifluoromethyl)phenyl]-4-[(trifluoromethyl)sulfinyl]-
Fluocyanobenpyrazole
T5NNJ AR BG FG DXFFF& CCN DSO&XFFF EZ &&(RS) Form [WLN]
Termidor
UNII:QGH063955F
NCGC00094574-08
QA-6027
SPECTRUM1505354
TL8000532
UNII-QGH063955F
UPCMLD-DP011:002
UQ4430250
芬普尼 [Chinese]
フィプロニル
1H-Pyrazole-3-carbonitrile, 5-amino-1-(2,6-dichloro-4-(trifluoromethyl)phenyl)-4-((trifluoromethyl)sulfinyl)-
424-610-5 [EINECS]
5-amino-1-[2,6-dichloro-4-(trifluoromethyl)phenyl]-4-(trifluoromethane)sulfinyl-1H-pyrazole-3-carbonitrile
5-amino-1-[4-(trifluoromethyl)phenyl]-4-(trifluoromethylsulfinyl)-3-pyrazolecarbonitrile
8090115 [Beilstein]
HSDB 7051; RM 1601
Fipronil
CAS Registry Number: 120068-37-3
CAS Name: 5-Amino-1-[2,6-dichloro-4-(trifluoromethyl)phenyl]-4-[(trifluoromethyl)sulfinyl]-1H-pyrazole-3-carbonitrile
Additional Names: 5-amino-3-cyano-1-(2,6-dichloro-4-trifluoromethylphenyl)-4-trifluoromethylsulfinylpyrazole; (±)-5-amino-1-(2,6-dichloro-a,a,a-trifluoro-p-tolyl)-4-trifluoromethylsulfinylpyrazole-3-carbonitrile
Manufacturers’ Codes: MB-46030
Trademarks: Frontline (Merial); Termidor (BASF)
Molecular Formula: C12H4Cl2F6N4OS
Molecular Weight: 437.15
Percent Composition: C 32.97%, H 0.92%, Cl 16.22%, F 26.08%, N 12.82%, O 3.66%, S 7.34%
Literature References: GABA-gated chloride channel blocker. Prototype of the phenylpyrazole insecticides known as fiproles. Prepn: I. G. Buntain et al., EP 295117 (1988 to May & Baker); L. R. Hatton et al., US 5232940 (1993). Mechanism of action study: L. M. Cole et al., Pestic. Biochem. Physiol. 46, 47 (1993). Comprehensive description: F. Colliot et al., Brighton Crop Prot. Conf. – Pests Dis. 1992, 29-34.
Properties: White solid, mp 200.5-201°. Vapor pressure (20°): 2.8 ´ 10-9 mm Hg. Log P (n-octanol/water): 4.0. Soly: water 2 mg/l; acetone >50%; corn oil >10,000 mg/l. LD50 in rats (mg/kg): 100 orally; >2000 dermally (Colliot); in mice (mg/kg): 32 i.p. (Cole).
Melting point: mp 200.5-201°
Log P: Log P (n-octanol/water): 4.0
Toxicity data: LD50 in rats (mg/kg): 100 orally; >2000 dermally (Colliot); in mice (mg/kg): 32 i.p. (Cole)
Use: Pesticide.
Therap-Cat-Vet: Ectoparasiticide.

APPROVED CDSCO INDIA 25.06.2018

Fipronil  50mg/134mg/268mg/402 mg spot on solution for cats and dogs , For treatment of flea and tick infestation in cats and dogs (for veterinary use only)

Fipronil is a broad-spectrum insecticide that belongs to the phenylpyrazole chemical family. Fipronil disrupts the insect central nervous system by blocking GABA-gated chloride channels and glutamate-gated chloride (GluCl) channels. This causes hyperexcitation of contaminated insects’ nerves and muscles. Fipronil’s specificity towards insects is believed to be due to its greater affinity to the GABA receptor in insects relative to mammals and its effect on GluCl channels, which do not exist in mammals.[1]

Because of its effectiveness on a large number of pests, fipronil is used as the active ingredient in flea control products for pets and home roach traps as well as field pest control for corn, golf courses, and commercial turf. Its widespread use makes its specific effects the subject of considerable attention. This includes ongoing observations on possible off-target harm to humans or ecosystems as well as the monitoring of resistance development.[2]

Use

Fipronil is or has been used in:

  • Under the trade name Regent, it is used against major lepidopteran (moth, butterfly, etc.) and orthopteran (grasshopper, locust, etc.) pests on a wide range of field and horticultural crops and against coleopteran (beetle) larvae in soils. In 1999, 400,000 hectares were treated with Regent. It became the leading imported product in the area of rice insecticides, the second-biggest crop protection market after cotton in China.[3]
  • Under the trade names Goliath and Nexa, it is employed for cockroach and ant control, including in the US. It is also used against pests of field corngolf courses, and commercial lawn care under the trade name Chipco Choice.[3]
  • It has been used under the trade name Adonis for locust control in Madagascar and Kazakhstan.[3]
  • Marketed under the names Termidor, Ultrathor, and Taurus in Africa and Australia, fipronil effectively controls termite pests, and was shown to be effective in field trials in these countries.[3]
  • Termidor has been approved for use against the Rasberry crazy ant in the Houston, Texas, area, under a special “crisis exemption” from the Texas Department of Agriculture and the Environmental Protection Agency. The chemical is only approved for use in Texascounties experiencing “confirmed infestations” of the newly discovered ant species.[4] Use of Termidor is restricted to certified pest control operators in the following states: Alaska, Connecticut, Nebraska, South Carolina, Massachusetts, Indiana, New York, and Washington.[citation needed]
  • In Australia, it is marketed under numerous trade names, including Combat Ant-Rid, Radiate and Termidor, and as generic fipronil
  • In the UK, provisional approval for five years has been granted for fipronil use as a public hygiene insecticide.[3]
  • Fipronil is the main active ingredient of Frontline TopSpot, Fiproguard, Flevox, and PetArmor (used along with S-methoprene in the ‘Plus’ versions of these products); these treatments are used in fighting tick and flea infestations in dogs and cats.
  • In New Zealand, fipronil was used in trials to control wasps (Vespula spp.), which are a threat to indigenous biodiversity.[5] It is now being used by the Department of Conservation to attempt local eradication of wasps,[6].[7][8]

Effects

Toxicity

Fipronil is classed as a WHO Class II moderately hazardous pesticide, and has a rat acute oral LD50 of 97 mg/kg.

It has moderate acute toxicity by the oral and inhalation routes in rats. Dermal absorption in rats is less than 1% after 24 h and toxicity is considered to be low. It has been found to be very toxic to rabbits.

The photodegradate MB46513 or desulfinylfipronil, appears to have a higher acute toxicity to mammals than fipronil itself by a factor of about 10.[9]

Symptoms of acute toxicity via ingestion includes sweating, nausea, vomiting, headache, abdominal pain, dizziness, agitation, weakness, and tonic-clonic seizures. Clinical signs of exposure to fipronil are generally reversible and resolve spontaneously. As of 2011, no data were available regarding the chronic effects of fipronil on humans. The U.S. EPA has classified fipronil as a group C (possible human) carcinogen based on an increase in thyroid follicular cell tumors in both sexes of the rat. However, as of 2011, no human data is available regarding the carcinogenic effects of fipronil.[10]

Two Frontline TopSpot products were determined by the New York State Department of Environmental Conservation to pose no significant exposure risks to workers applying the product. However, concerns were raised about human exposure to Frontline spray treatment in 1996, leading to a denial of registration for the spray product. Commercial pet groomers and veterinarians were considered to be at risk from chronic exposure via inhalation and dermal absorption during the application of the spray, assuming they may have to treat up to 20 large dogs per day.[3] Fipronil is not volatile, so the likelihood of humans being exposed to this compound in the air is low.[10]

In contrast to neonicotinoids such as acetamipridclothianidinimidacloprid, and thiamethoxam, which are absorbed through the skin to some extent, fipronil is not absorbed substantially through the skin.[11]

Detection in body fluids

Fipronil may be quantitated in plasma by gas chromatography-mass spectrometry or liquid chromatography-mass spectrometry to confirm a diagnosis of poisoning in hospitalised patients or to provide evidence in a medicolegal death investigation.[12]

Ecological toxicity

Fipronil is highly toxic for crustaceansinsects and zooplankton,[13] as well as beestermitesrabbits, the fringe-toed lizard, and certain groups of gallinaceous birds. It appears to reduce the longevity and fecundity of female braconid parasitoids. It is also highly toxic to many fish, though its toxicity varies with species. Conversely, the substance is relatively innocuous to passerineswildfowl, and earthworms.

Its half-life in soil is four months to one year, but much less on soil surface because it is more sensitive to light (photolysis) than water (hydrolysis).[14]

Few studies of effects on wildlife have been conducted, but studies of the nontarget impact from emergency applications of fipronil as barrier sprays for locust control in Madagascar showed adverse impacts of fipronil on termites, which appear to be very severe and long-lived. Also, adverse effects were indicated in the short term on several other invertebrate groups, one species of lizard (Trachylepis elegans), and several species of birds (including the Madagascar bee-eater).

Nontarget effects on some insects (predatory and detritivorous beetles, some parasitic wasps and bees) were also found in field trials of fipronil for desert locust control in Mauritania, and very low doses (0.6-2.0 g a.i./ha) used against grasshoppers in Niger caused impacts on nontarget insects comparable to those found with other insecticides used in grasshopper control. The implications of this for other wildlife and ecology of the habitat remain unknown, but appear unlikely to be severe.[3] Unfortunately, this lack of severity was not observed in bee species in South America. Fipronil is also used in Brazil and studies on the stingless bee Scaptotrigona postica have shown adverse reactions to the pesticide, including seizures, paralysis, and death with a lethal dose of .54 ng a.i./bee and a lethal concentration of .24 ng a.i./μl diet. These values are highly toxic in Scaptotrigona postica and bees in general.[15] Toxic baiting with fipronil has been shown to be effective in locally eliminating German wasps. All colonies within foraging range were completely eliminated within one week.[16][17][5]

In May 2003, the French Directorate-General of Food at the Ministry of Agriculture determined that a case of mass bee mortality observed in southern France was related to acute fipronil toxicity. Toxicity was linked to defective seed treatment, which generated dust. In February 2003, the ministry decided to temporarily suspend the sale of BASF crop protection products containing fipronil in France.[18] The seed treatment involved has since been banned.[citation needed] Fipronil was used in a broad spraying to control locusts in Madagascar in a program that began in 1997.[19]

Notable results from wildlife studies include:

  • Fipronil is highly toxic to fish and aquatic invertebrates. Its tendency to bind to sediments and its low water solubility may reduce the potential hazard to aquatic wildlife.[20]
  • Fipronil is toxic to bees and should not be applied to vegetation when bees are foraging.[20]
  • Based on ecological effects, fipronil is highly toxic to upland game birds on an acute oral basis and very highly toxic on a subacute dietary basis, but is practically nontoxic to waterfowl on both acute and subacute bases.[21]
  • Chronic (avian reproduction) studies show no effects at the highest levels tested in mallards (NOEC) = 1000 ppm) or quail (NOEC = 10 ppm). The metabolite MB 46136 is more toxic to the parent than avian species tested (very highly toxic to upland game birds and moderately toxic to waterfowl on an acute oral basis).[21]
  • Fipronil is very highly toxic to bluegill sunfish and highly toxic to rainbow trout on an acute basis.[21]
  • An early-lifestage toxicity study in rainbow trout found that fipronil affects larval growth with a NOEC of 0.0066 ppm and an LOEC of 0.015 ppm. The metabolite MB 46136 is more toxic than the parent to freshwater fish (6.3 times more toxic to rainbow trout and 3.3 times more toxic to bluegill sunfish). Based on an acute daphnia study using fipronil and three supplemental studies using its metabolites, fipronil is characterized as highly toxic to aquatic invertebrates.[21]
  • An invertebrate lifecycle daphnia study showed that fipronil affects length in daphnids at concentrations greater than 9.8 ppb.[21]
  • A lifecycle study in mysids shows fipronil affects reproduction, survival, and growth of mysids at concentrations less than 5 ppt.[21]
  • Acute studies of estuarine animals using oystersmysids, and sheepshead minnows show that fipronil is highly acutely toxic to oysters and sheepshead minnows, and very highly toxic to mysids. Metabolites MB 46136 and MB 45950 are more toxic than the parent to freshwater invertebrates (MB 46136 is 6.6 times more toxic and MB 45950 is 1.9 times more toxic to freshwater invertebrates).[21]

Colony collapse disorder

Fipronil is one of the main chemical causes blamed for the spread of colony collapse disorder among bees. It has been found by the Minutes-Association for Technical Coordination Fund in France that even at very low nonlethal doses for bees, the pesticide still impairs their ability to locate their hive, resulting in large numbers of forager bees lost with every pollen-finding expedition.[22] A synergistic toxic effect of fipronil with the fungal pathogen Nosema ceranae was recently reported[23]. The functional basis for this toxic effect is now understood: the synergy between fipronil and the pathogenic fungus induces changes in male physiology leading to infertility[24] A 2013 report by the European Food Safety Authorityidentified fipronil as “a high acute risk to honeybees when used as a seed treatment for maize and on July 16, 2013 the EU voted to ban the use of fipronil on corn and sunflowers within the EU. The ban took effect at the end of 2013.”[25][26]

Pharmacodynamics

Fipronil acts by binding to allosteric sites of GABAA receptors and GluCl receptors (of insects) as an antagonist (a form of noncompetitive inhibition). This prevents the opening of chloride ion channels normally encouraged by GABA, reducing the chloride ions’ ability to lower a neuron’s membrane potential. This results in an overabundance of neurons reaching action potential and likewise CNS toxicity via overstimulation.[27][28][29][30]

Acute oral LD50 (rat) 97 mg/kg
Acute dermal LD50 (rat) >2000 mg/kg

In animals and humans, fipronil poisoning is characterized by vomiting, agitation, and seizures, and can usually be managed through supportive care and early treatment of seizures, generally with benzodiazepine use.[31][32]

History

Fipronil was discovered and developed by Rhône-Poulenc between 1985 and 1987, and placed on the market in 1993 under the B2 U.S. Patent 5,232,940 B2. Between 1987 and 1996, fipronil was evaluated on more than 250 insect pests on 60 crops worldwide, and crop protection accounted for about 39% of total fipronil production in 1997. Since 2003, BASF holds the patent rights for producing and selling fipronil-based products in many countries.

2017 Fipronil eggs contamination

The 2017 Fipronil eggs contamination is an incident in Europe and South Korea involving the spread of insecticide contaminated eggs and egg products. Chicken eggs were found to contain Fipronil and distributed to 15 European Union countries, Switzerland, and Hong Kong.[33][34] Approximately 700,000 eggs are thought to have reached shelves in the UK alone.[35] Eggs at 44 farms in Taiwan were also found with excessive Fipronil levels.[36]

SYN

Figure US20130030190A1-20130131-C00009

SYN 2

SYN 3

SYN 4

PATENT

http://www.allindianpatents.com/patents/271132-a-process-for-the-synthesis-of-fipronil

5-Amino-l-(2,6-dichloro-4-trifluoromethylphenyl)-3-cyano-4-trifluoromethyl
sulfinyl pyrazole or 5-Amino-[2,6-dichloro-4-(trif]uoromethyl)phenyl]-4-[-(1 (R,S)-trifluoromethyl)sulfinyl]-1H-pyrazole-3-carbonitrile also known as Fipronil is a novel pesticide characterized by high efficiency, low toxicity and especially low residue.
There are various routes to synthesize Fipronil by oxidation of thiopyrazole with various other oxidizing agents in suitable solvents. Oxidation of sulfides is a very useful route for the preparation of sulfoxides. Literature is replete with the conversion of sulfides to sulfoxides and/or sulfones. However, most of the existing methods use expensive, toxic or rare oxidizing reagents, which are difficult to prepare, are very expensive and cannot be used on commercial scale. Many of these processes suffer from poor selectivity.
WO01/30760 describes oxidation of 5-amino-l-(2,6-dichloro-4-trifluoromethylphenyl)-3-cyano-4-trifluoromethylthio-pyrazole with trifluoro-acetic acid and hydrogen peroxide in the presence of boric acid. The quantity

of trifluoroacetic acid used is 14.5 molar equivalents. The patent also
discloses the preparation of 5-amino-1-(2,6-dichloro-4-trifluoromethyl
phenyl)-3-cyano-4-trifluoromethylthio-pyrazole from 5-amino-1-(2,6-
dichloro-4-trifluoromethyl phenyl)-3-cyano pyrazole-4-yl disulphide.
European Patent publication No.295117 describes the preparation of 5-amino-l-(2,6-dichloro-4-trifluoromethylphenyl)-3-cyano-4-trifluoromethylsulphinyl pyrazole starting from 2,6-Dichloro-4-trifluoromethylaniline to give an intermediate 5-amino-l-(2,6-dichloro-4-trifluoromethylphenyl)-3-cyano-4-trifluoromethylthiopyrazole which is oxidized with meta-chloroperbenzoic acid in chloroform to give desired product.
Oxidizing agents such as perbenzoic acids do not provide effective and regioselective oxidation of electron deficient sulfides such as trifluoromethylsulphides which are less readily oxidized than other sulfides. Trifluoroacetic acid and trichloroacetic acid are found to be very efficient and regioselective oxidation medium for oxidation of 5-amino-l-(2,6-dichloro-4-trifluoromethylphenyl)-3-cyano-4-trifluoromethylthio-pyrazole in presence of hydrogen peroxide. Trichloroacetic acid can not be used alone due to higher melting point. Trifluoroacetic acid on the other hand is very regioselective with respect to conversion and low by-products formation. However, it is expensive, water miscible, corrosive to metal as well as glass, comparatively lower boiling and it’s recovery (in anhydrous form) is complex in nature.
W000/35851/2000 talks about synthesis of 2,6-Dichloro-4-trifluoromethylaniline starting from 3,4,5-trichloro-benzotrifluoride in the presence of alkaline fluorides like lithium fluoride and ammonia in the

presence of N-methylpyrrolidone at 250°C to give 97% conversion and 87% selectivity. The main drawback of the above process is the synthesis of 3,4,5-trichlorobenzotrifluoride in high yield and purity. Chlorination of p-chlorobenzotrifluoride gives a mixture of 3,4,5-trichlorobenzotrifluoride in 72% GLC conversions, 3,4-dichloro and tetrachlorobenzotrifluoride. The process to get pure 3,4,5-isomer from this mixture by fractionation followed by crystallization is very tedious. Moreover in-spite of using very pure intermediates, substantial amount of an undesired isomer (3-amino-4,5-dichlorobenzotrifluoride) is also obtained.
Another approach to generate 3,4,5-trichlorobenzotrifluoride with high yield and purity is to perform denitrochlorination of 4-chloro-3,5-dinitrobenzotrifluoride in the presence of a catalyst as described in GB Patent 2154581A. Even though the process produces 3,4,5-trichlorobenzotrifluoide in high yield and purity, the reaction conditions are too drastic to be employed for an industrial process.
The known commercial processes for the manufacture of Fipronil uses corrosive and expensive chemical such as trifluoroaceticacid, hydrogen peroxide and m-chloroperbenzoicacid Trifluoroacetic acid is expensive and generally not used in large quantities, as well as of m-chloroperbenzoic acid is difficult to handle at commercial scale due to its un-stability and detonating effect. Also the raw material used such as 2,6-Dichloro-4-trifluoromethylaniline are not easily available or made. The overall process for the Fipronil as disclosed above is found to be unsatisfactory in one respect or the other.

Thus, there is felt a need for preparing Fipronil from easily available raw materials in a simple and economical manner at an industrial level, with high yields and purity.

PATENT

https://patents.google.com/patent/US20130030190A1/en

  • 5-Amino-1-(2,6-dichloro-4-trifluoromethylphenyl)-3-cyano-4-trifluoromethyl sulfinyl pyrazole or 5-Amino-[2,6-dichloro-4-(trifluoromethyl)phenyl]-4-[-(1(R,S)-trifluoromethyl)sulfinyl]-1H-pyrazole-3-carbonitrile also known as Fipronil is a novel pesticide characterized by high efficiency, low toxicity and especially low residue.
  • [0005]
    There are various routes to synthesize Fipronil by oxidation of thiopyrazole with various other oxidizing agents in suitable solvents. Oxidation of sulfides is a very useful route for the preparation of sulfoxides. Literature is replete with the conversion of sulfides to sulfoxides and/or sulfones. However, most of the existing methods use expensive, toxic or rare oxidizing reagents, which are difficult to prepare, are very expensive and cannot be used on commercial scale. Many of these processes suffer from poor selectivity.
  • [0006]
    WO01/30760 describes oxidation of 5-amino-1-(2,6-dichloro-4-trifluoromethylphenyl)-3-cyano-4-trifluoromethylthio-pyrazole with trifluoro-acetic acid and hydrogen peroxide in the presence of boric acid. The quantity of trifluoroacetic acid used is 14.5 molar equivalents. The patent also discloses the preparation of 5-amino-1-(2,6-dichloro-4-trifluoromethyl phenyl)-3-cyano-4-trifluoromethylthio-pyrazole from 5-amino-1-(2,6-dichloro-4-trifluoromethyl phenyl)-3-cyano pyrazole-4-yl disulphide.
  • [0007]
    European Patent publication No. 295117 describes the preparation of 5-amino-1-(2,6-dichloro-4-trifluoromethylphenyl)-3-cyano-4-trifluoromethylsulphinyl pyrazole starting from 2,6-Dichloro-4-trifluoromethylaniline to give an intermediate 5-amino-1-(2,6-dichloro-4-trifluoromethylphenyl)-3-cyano-4-trifluoromethylthiopyrazole which is oxidized with meta-chloroperbenzoic acid in chloroform to give desired product.
  • [0008]
    Oxidizing agents such as perbenzoic acids do not provide effective and regioselective oxidation of electron deficient sulfides such as trifluoromethylsulphides which are less readily oxidized than other sulfides. Trifluoroacetic acid and trichloroacetic acid are found to be very efficient and regioselective oxidation medium for oxidation of 5-amino-1-(2,6-dichloro-4-trifluoromethylphenyl)-3-cyano-4-trifluoromethylthio-pyrazole in presence of hydrogen peroxide. Trichloroacetic acid can not be used alone due to higher melting point. Trifluoroacetic acid on the other hand is very regioselective with respect to conversion and low by-products formation. However, it is expensive, water miscible, corrosive to metal as well as glass, comparatively lower boiling and it’s recovery (in anhydrous form) is complex in nature.
  • [0009]
    WO00/35851/2000 talks about synthesis of 2,6-Dichloro-4-trifluoromethylaniline starting from 3,4,5-trichloro-benzotrifluoride in the presence of alkaline fluorides like lithium fluoride and ammonia in the presence of N-methylpyrrolidone at 250° C. to give 97% conversion and 87% selectivity. The main drawback of the above process is the synthesis of 3,4,5-trichlorobenzotrifluoride in high yield and purity. Chlorination of p-chlorobenzotrifluoride gives a mixture of 3,4,5-trichlorobenzotrifluoride in 72% GLC conversions, 3,4-dichloro and tetrachlorobenzotrifluoride. The process to get pure 3,4,5-isomer from this mixture by fractionation followed by crystallization is very tedious. Moreover in-spite of using very pure intermediates, substantial amount of an undesired isomer (3-amino-4,5-dichlorobenzotrifluoride) is also obtained.
  • [0010]
    Another approach to generate 3,4,5-trichlorobenzotrifluoride with high yield and purity is to perform denitrochlorination of 4-chloro-3,5-dinitrobenzotrifluoride in the presence of a catalyst as described in GB Patent 2154581A. Even though the process produces 3,4,5-trichlorobenzotrifluoide in high yield and purity, the reaction conditions are too drastic to be employed for an industrial process.
  • [0011]
    The known commercial processes for the manufacture of Fipronil uses corrosive and expensive chemical such as trifluoroaceticacid, hydrogen peroxide and m-chloroperbenzoicacid Trifluoroacetic acid is expensive and generally not used in large quantities, as well as of m-chloroperbenzoic acid is difficult to handle at commercial scale due to its un-stability and detonating effect. Also the raw material used such as 2,6-Dichloro-4-trifluoromethylaniline are not easily available or made. The overall process for the Fipronil as disclosed above is found to be unsatisfactory in one respect or the other.
  • [0012]
    Thus, there is felt a need for preparing Fipronil from easily available raw materials in a simple and economical manner at an industrial level, with high yields and purity.

Figure US20130030190A1-20130131-C00009

    • Example 18
    • [0081]
      A mixture of 700 g of dichloroacetic acid and trichloroacetic acid was taken along with 300 g of chlorobenzene, 2 g of boric acid and 280 g of 5-amino-1-(2,6-dichloro-4-trifluoromethylphenyl)-3-cyano-4-trifluoromethyl thiopyrazole, the content were cooled to 15-20° C. Aqueous H20(44.2 g, 50%) was added and mass was stirred for 20 hrs. The mass was then processed and Fipronil was isolated by filtration. After work up as above, 269 g of Fipronil of purity 94% was obtained. The filtered Fipronil was then purified using chlorobenzene (5 ml/g) followed by mixture (1 ml/g, 80:20 v/v) of ethylacetate and chlorobenzene to get 232 g of Fipronil of greater than 97% purity.

Example 19 Purification of Fipronil

  • [0082]
    The fipronil prepared in example 18 of purity 97% was treated with a mixture (232 ml) of ethylacetate & chlorobenzene (80:20 v/v). This reaction mixture was heated to 85-90° C. & maintained for 1 hr. It was further cooled up to 30° C. in stages & filtered. Fipronil thus obtained had a purity of 98%. This cycle was repeated to obtain fipronil of above 98% purity.
  • [0083]
    The useful constituents from various streams of crystallization, leaching as above were reused and recycled, fipronil was isolated in 80-85% yield with purity of above 98%.

PATENT

CN 101250158 [2008 to Hunan Res Inst of of chemical Ind.]

WO2005/44806 A1, ; Page/Page column 7-8; 12 ;

WO2009/77853 A1, ; Page/Page column 28-29 ;

US 5,618,945 [1995, to Rhone-Poulenc]

CN 102060774

IN 178903 [1997, to Rallis India Ltd.]

WO 2009/077,853 [2009 to Vetoquinol SA ]

BG 109983 [2008 to BASF Agro B V]

US 8,507,693 [2013, to Gharda]

US 5,618,945 [1995, to Rhone-Poulenc]

WO 2007/122,440 [ 2007 to Gharda Chemicals Ltd.]

FR 2,925,493 [2009 to to Vetoquinol SA ]

CN 1176078 [ 2002 to Jiangsu Prov Inst of Pesticide]

EP 0,374,061 [ 1990 to Rhone Poulenc Agrochimie]

US 5,232,940 [1993, to May and Baker]

PAPER

Journal of the Chemical Society, Perkin Transactions 1: Organic and Bio-Organic Chemistry (1972-1999), , # 24 p. 3371 – 3376

Synthesis 2008, 11, 1682-1684

Synthesis 2007, 22, 3507-3511

Tetrahedron Letters, , 2007, 48(48), 8518-8520

Tetrahedron Letters 2008 ,49. 3463-3465

References

  1. Jump up^ Raymond-Delpech, Valérie; Matsuda, Kazuhiko; Sattelle, Benedict M.; Rauh, James J.; Sattelle, David B. (2005-09-20). “Ion channels: molecular targets of neuroactive insecticides”Invertebrate Neuroscience5 (3-4): 119–133. doi:10.1007/s10158-005-0004-9ISSN 1354-2516.
  2. Jump up^ Maddison, Jill E.; Page, Stephen W. (2008). Small Animal Clinical Pharmacology(Second ed.). Elsevier Health Sciences. p. 229. ISBN 9780702028588.
  3. Jump up to:a b c d e f g “Fipronil”. Pesticides News48: 20. 2000.
  4. Jump up^ “Rasberry Crazy Ant”. Texas A&M. 2008-04-12. Archived from the original on 2007-07-14. Retrieved 2012-08-06.
  5. Jump up to:a b “Revive Rotoiti Autumn 2011”. Department of Conservation. 2011. Retrieved 11 April2012.
  6. Jump up^ “War on wasps in Abel Tasman”. 10 February 2016.
  7. Jump up^ “Vespex: Making wide-area wasp control a reality – WWF’s Conservation Innovation Awards”wwf-nz.crowdicity.com.
  8. Jump up^ (DOC), corporatename = New Zealand Department of Conservation. “Wasp control using Vespex”http://www.doc.govt.nz.
  9. Jump up^ “Fipronil insecticide: Novel photochemical desulfinylation with retention of neurotoxicity”Proceedings of the National Academy of Sciences93: 12764–12767. doi:10.1073/pnas.93.23.12764PMC 23994Freely accessible.
  10. Jump up to:a b “Fipronil Technical Fact Sheet, National Pesticide Information Center”. Retrieved 2015-12-07.
  11. Jump up^ “Cockroach Control”. Retrieved August 10, 2016.
  12. Jump up^ R. Baselt, Disposition of Toxic Drugs and Chemicals in Man, 11th edition, Biomedical Publications, Seal Beach, CA, 2017, pp. 894-895. ISBN 978-0-692-77499-1
  13. Jump up^ “Ecotoxicity for Fipronil”. Retrieved 2010-05-03.
  14. Jump up^ Amrith S. Gunasekara & Tresca Troung (March 5, 2007). “Environmental Fate of Fipronil” (PDF). Retrieved April 16, 2016.
  15. Jump up^ Jacob, CRO; Hellen Maria Soares; Stephen Malfitano Carvalho; Roberta Cornélio Ferreira Nocelli; Osmar Malspina (2013). “Acute Toxicity of Fipronil to the Stingless Bee Scaptotrigona postica Latreille”Bulletin of Environmental Contamination and Toxicology90 (1): 69–72. doi:10.1007/s00128-012-0892-4. Retrieved 23 September 2015.
  16. Jump up^ Paula Sackmann, Mauricio Rabinovich and Juan Carlos Corley J. (2001). “Successful Removal of German Yellowjackets (Hymenoptera: Vespidae) by Toxic Baiting” (PDF). pp. 811–816.
  17. Jump up^ “Short and long-term control of Vespula pensylvanica in Hawaii by fipronil baiting”Pest Management Science68: 1026–1033. doi:10.1002/ps.3262.
  18. Jump up^ Elise Kissling; BASF SE (2003). “BASF statement regarding temporary suspension of sales of crop protection products containing fipronil in France”.
  19. Jump up^ June 2000 BBC News story “Anti-locust drive ‘created havoc'”.
  20. Jump up to:a b “Fipronil” (PDF). National Pesticides Communication Network. p. 3. Retrieved 19 June 2012.
  21. Jump up to:a b c d e f g United States Environmental Protection Agency Office of Prevention, Pesticides and Toxic Substances (1996). “Fipronil. May 1996. New Pesticide Fact Sheet. US EPA Office of Prevention, Pesticides and Toxic Substances”.
  22. Jump up^ “Colony Collapse Disorder linked to Fipronil”. Retrieved 2010-06-17.
  23. Jump up^ Aufavure J., Biron D. G., Vidau C., Fontbonne R., Roudel M., Diogon M., Viguès B., Belzunces L. P., Delbac F., Blot N. (2012) Parasite – insecticide interactions: a case study of Nosema ceranae and fipronil synergy on honeybee. Scientific Reports 2:326 – DOI: 10.1038/srep00326
  24. Jump up^ Kairo G, Biron D.G, Ben A.F, Bonnet M, Tchamitchian S, Cousin M, … & Brunet J.L (2017) Nosema ceranae, Fipronil and their combination compromise honey bee reproduction via changes in male physiology]. Scientific reports, 7(1), 8556.
  25. Jump up^ “EFSA assesses risks to bees from fipronil”. 27 May 2013. Retrieved 29 May 2013.
  26. Jump up^ Carrington, Damian (16 July 2013). “EU to ban fipronil to protect honeybees”The Guardian. London.
  27. Jump up^ Cole, L. M.; Nicholson, R. A.; Casida, J. E. (1993). “Action of Phenylpyrazole Insecticides at the GABA-Gated Chloride Channel”Pestic. Biochem. Physiol46: 47–54. doi:10.1006/pest.1993.1035.
  28. Jump up^ Ratra, G. S.; Casida, J. E. (2001). “GABA receptor subunit composition relative to insecticide potency and selectivity”. Toxicol. Lett122: 215–222. doi:10.1016/s0378-4274(01)00366-6PMID 11489356.
  29. Jump up^ WHO. Pesticide Residues in Food – 1997: Fipronil; International Programme on Chemical Safety, World Health Organization: Lyon, 1997.
  30. Jump up^ Olsen RW, DeLorey TM (1999). “Chapter 16: GABA and Glycine”. In Siegel GJ, Agranoff BW, Fisher SK, Albers RW, Uhler MD. Basic neurochemistry: molecular, cellular, and medical aspects (Sixth ed.). Philadelphia: Lippincott-Raven. ISBN 0-397-51820-X.
  31. Jump up^ Ramesh C. Gupta (2007). Veterinary Toxicology. pp. 502–503. ISBN 978-0-12-370467-2.
  32. Jump up^ Mohamed F, Senarathna L, Percy A, Abeyewardene M, Eaglesham G, Cheng R, Azher S, Hittarage A, Dissanayake W, Sheriff MH, Davies W, Buckley NA, Eddleston M., Acute human self-poisoning with the N-phenylpyrazole insecticide fipronil–a GABAA-gated chloride channel blocker, J Toxicol Clin Toxicol. 2004;42(7):955-63.
  33. Jump up^ “Eggs containing fipronil found in 15 EU countries and Hong Kong”BBC News. 2017-08-11. Retrieved 2017-08-11.
  34. Jump up^ News, ABC. “EU: 17 nations get tainted eggs, products in growing scandal”ABC News. Archived from the original on 2017-08-11. Retrieved 2017-08-11.
  35. Jump up^ Boffey, Daniel (11 August 2017). “Egg contamination scandal widens as 15 EU states, Switzerland and Hong Kong affected”. The Guardian. Retrieved 11 August 2017.
  36. Jump up^ “Eggs at 44 farms in Taiwan found with excessive insecticide levels”. Taiwan News. 26 August 2017. Retrieved 26 August 2017.

External links

Fipronil
2D chemical structure of fipronil
3D chemical structure of fipronil
Names
IUPAC name
(RS)-5-Amino-1-[2,6-dichloro-4-(trifluoromethyl)phenyl]-4-(trifluoromethylsulfinyl)pyrazole-3-carbonitrile
Other names
Fipronil
Fluocyanobenpyrazole
Termidor
Identifiers
3D model (JSmol)
ChEBI
ChEMBL
ChemSpider
ECHA InfoCard 100.102.312
KEGG
PubChem CID
UNII
Properties
C12H4Cl2F6N4OS
Molar mass 437.14 g·mol−1
Density 1.477-1.626 g/cm3
Melting point 200.5 °C (392.9 °F; 473.6 K)
Pharmacology
QP53AX15 (WHO)
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).

/////////////Fipronil, INDIA 2018, フィプロニル , HSDB 7051, RM 1601, veterinary, ind 2018

C1=C(C=C(C(=C1Cl)N2C(=C(C(=N2)C#N)S(=O)C(F)(F)F)N)Cl)C(F)(F)F

“ DRUG APPROVALS INTERNATIONAL” CATERS TO EDUCATION GLOBALLY, No commercial exploits are done or advertisements added by me. This is a compilation for educational purposes only. P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent

Bepotastine Besilate, ベポタスチンベシル酸塩

$
0
0

ChemSpider 2D Image | Bepotastine Besilate | C27H31ClN2O6SBepotastine besilate.png

Bepotastine Besilate

ベポタスチンベシル酸塩

  • Molecular FormulaC27H31ClN2O6S
  • Average mass547.063 Da
UNII:6W18MO1QR3
(+)-(S)-4-(4-((4-Chlorophenyl)(2-pyridyl)methoxy)piperidino)butyric acid monobenzenesulfonate
(S)-4-(4-((4-chlorophenyl)(pyridin-2-yl)methoxy)piperidin-1-yl)butanoic acid compound with benzenesulfonic acid (1:1)
190786-44-8 [RN]
125602-71-3 FREE FORM,
UNII: 6W18MO1QR3
1-Piperidinebutanoic acid, 4-[(S)-(4-chlorophenyl)-2-pyridinylmethoxy]-, benzenesulfonate (1:1) [ACD/Index Name]
4-{4-[(S)-(4-Chlorophenyl)(2-pyridinyl)methoxy]-1-piperidinyl}butanoic acid benzenesulfonate (1:1)
Talion [Trade name]
tau284
TAU-284DS, TAU-284
DA-5206
HL-151
SNJ-1773
    • Use:antiallergic, antihistaminic
For the symptomatic treatment of itchy eyes (caused by IgE-induced mast cell degranulation) due to allergic conjunctivitis.
10 mg Tablets  For the treatment of allergic rhinitis  27.03.2017 CDSCO

APPROVED 

USFDA

NDA 22-288 Bepotastine Besilate 1.5% Ophthalmic Solution ISTA Pharmaceuticals, Inc.

https://www.accessdata.fda.gov/drugsatfda_docs/nda/2009/02228s000_ChemR.pdf

str1

Drug Substance Bepotastine besilate is manufactured by Ube Industries and the information for the NDA is submitted through DMF #19966. Bepotastine besilate is a white crystalline powder with no odor and bitter taste. It is very soluble in but sparingly soluble in . It is stable when exposed to light, and optically active. The S-isomer is the active drug and is controlled as an impurity through synthesis. The distribution coefficient in 1-octanol is higher than in aqueous buffer in the pH 5-9 range. There are 10 potential impurities but only one impurity is above 0.1%. Two potential genotoxic impurities are controlled below . Residual is controlled below Bepotastine besilate is stable under long term storage conditions for (25ºC/60% RH) over 5 years

Bepotastine besilate was originally developed as an oral tablet dosage form and got approval in Japan in 2000 for allergic rhinitis. It is a non-sedating anti-allergic drug. The proposed NDA is an ophthalmic solution indicated for allergic conjunctivitis. Bepotastine besilate ophthalmic solution 1.5% is a sterile solution. It is an aqueous solution to be administered as drops at or near physiological pH range of tears. The formulation contains sodium chloride, monobasic sodium phosphate as dihydrate, benzalkonium chloride, sodium hydroxide and purified water; typically these components are used for , preservative action, pH adjustment,

INTRO

Bepotastine is a non-sedating, selective antagonist of the histamine 1 (H1) receptor. Bepotastine was approved in Japan for use in the treatment of allergic rhinitis and uriticaria/puritus in July 2000 and January 2002, respectively, and is marketed by Tanabe Seiyaku Co., Ltd. under the brand name Talion. It is available in oral and opthalmic dosage forms in Japan. The opthalmic solution is FDA approved since Sept 8, 2009 and is under the brand name Bepreve.

Tae Hee Ha, Chang Hee Park, Won Jeoung Kim, Soohwa Cho, Han Kyong Kim, Kwee Hyun Suh, “PROCESS FOR PREPARING BEPOTASTINE AND INTERMEDIATES USED THEREIN.” U.S. Patent US20100168433, issued July 01, 2010., US20100168433

BEPREVE® (bepotastine besilate ophthalmic solution) 1.5% is a sterile, topically administered drug for ophthalmic use. Each mL of BEPREVE contains 15 mg bepotastine besilate.

Bepotastine besilate is designated chemically as (+) -4-[[(S)-p-chloro-alpha -2pyridylbenzyl] oxy]-1-piperidine butyric acid monobenzenesulfonate. The chemical structure for bepotastine besilate is:

BEPREVE® (bepotastine besilate) Structural Formula Illustration

Bepotastine besilate is a white or pale yellowish crystalline powder. The molecular weight of bepotastine besilate is 547.06 daltons. BEPREVE ophthalmic solution is supplied as a sterile, aqueous 1.5% solution, with a pH of 6.8.

The osmolality of BEPREVE (bepotastine besilate ophthalmic solution) 1.5% is approximately 290 mOsm/kg.

ベポタスチンベシル酸塩 JP17
Bepotastine Besilate

C21H25ClN2O3▪C6H6O3S : 547.07
[190786-44-8]

Title: Bepotastine
CAS Registry Number: 190786-43-7
CAS Name: 4-[(S)-(4-Chlorophenyl)-2-pyridinylmethoxy]-1-piperidinebutanoic acid
Additional Names: betotastine
Molecular Formula: C21H25ClN2O3
Molecular Weight: 388.89
Percent Composition: C 64.86%, H 6.48%, Cl 9.12%, N 7.20%, O 12.34%
Literature References: Histamine H1-receptor antagonist. Prepn (stereochem. unspec.): A. Koda et al., EP 335586eidem, US4929618 (1989, 1990 both to Ube). Prepn of optically active salts: J. Kita et al., EP 949260 (1999 to Ube; Tanabe Seiyaku). Pharmacology: M. Kato et al., Arzneim.-Forsch. 47, 1116 (1997). Suppression of IL-5 production: O. Kaminuma et al., Biol. Pharm. Bull. 21, 411 (1998). Antiallergic activity in animal models: M. Ueno et al., Pharmacology 57, 206 (1998).
Derivative Type: Benzenesulfonate salt
CAS Registry Number: 190786-44-8
Additional Names: Bepotastine besilate
Manufacturers’ Codes: TAU-284
Trademarks: Talion (Tanabe)
Molecular Formula: C21H25ClN2O3.C6H6O3S
Molecular Weight: 547.06
Percent Composition: C 59.28%, H 5.71%, Cl 6.48%, N 5.12%, O 17.55%, S 5.86%
Properties: Pale grey prisms from acetonitrile, mp 161-163°. [a]D20 +6.0° (c = 5 in methanol).
Melting point: mp 161-163°
Optical Rotation: [a]D20 +6.0° (c = 5 in methanol)
Therap-Cat: Antihistaminic.
Keywords: Antihistaminic.

Bepotastine (TalionBepreve) is a 2nd generation antihistamine.[1] It was approved in Japan for use in the treatment of allergic rhinitisand urticaria/pruritus in July 2000 and January 2002, respectively. It is currently marketed in the United States under the brand-name Bepreve, by ISTA Pharmaceuticals.

Bepotastine besilate is a second-generation antihistamine that was launched in a tablet formulation under a collaboration between Tanabe Seiyaku and Ube in 2000 and in 2002 for the treatment of allergic rhinitis including sneeze, mucus discharge and solidified mucus, and for the treatment of urticaria, respectively. An orally disintegrating tablet was made available in Japan in 2006, while a dry syrup formulation for the treatment of allergic rhinitis was studied in clinical trials at Tanabe Seiyaku for the treatment of allergic rhinitis

Originally developed at Ube, bepotastine besilate was later licensed to Tanabe Seiyaku as part of a collaboration agreement. In 2010, rights were licensed to Dong-A and Mitsubishi Tanabe Pharma in Korea for the treatment of eye disorders.

Pharmacology

Bepotastine is available as an ophthalmic solution and oral tablet. It is a direct H1-receptor antagonist that inhibits the release of histamine from mast cells.[2] The ophthalmic formulation has shown minimal systemic absorption, between 1 and 1.5% in healthy adults.[3] Common side effects are eye irritation, headache, unpleasant taste, and nasopharyngitis.[3] The main route of elimination is urinary excretion, 75-90% excreted unchanged.[3]

Marketing history

It is marketed in Japan by Tanabe Seiyaku under the brand name Talion. Talion was co-developed by Tanabe Seiyaku and Ube Industries, the latter of which discovered bepotastine. In 2001, Tanabe Seiyaku granted Senju, now owned by Allergan, exclusive worldwide rights, with the exception of certain Asian countries, to develop, manufacture and market bepotastine for ophthalmic use. Senju, in turn, has granted the United States rights for the ophthalmic preparation to ISTA Pharmaceuticals.

Sales and patents

In 2011, ISTA pharmaceuticals experienced a 2.4% increase in net revenues from 2010, which was driven by the sales of Bepreve. Their net revenue for 2011 was $160.3 million.[4] ISTA Pharmaceuticals was acquired by Bausch & Lomb in March 2012 for $500 million.[5] Bausch & Lomb hold the patent for bepotastine besilate (https://www.accessdata.fda.gov/scripts/cder/ob/docs/temptn.cfm. On November 26, 2014, Bausch & Lomb sue Micro Labs USA for patent infringement.[6] Bausch & Lomb was recently bought out by Valeant Pharmaceuticals in May 2013 for $8.57 billion, Valeant’s largest acquisition to date, causing the company’s stock to rise 25% when the deal was announced.[7]

Clinical trials

A Phase III clinical trial was carried out in 2010 to evaluate the effectiveness of bepotastine besilate ophthalmic solutions 1.0% and 1.5%.[8] These solutions were compared to a placebo and evaluated for their ability to reduce ocular itchiness. The study was carried out with 130 individuals and evaluated after 15 minutes, 8 hours, or 16 hours. There was a reduction in itchiness at all-time points for both ophthalmic solutions. The study concluded that bepotastine besilate ophthalmic formulations reduced ocular itchiness for at least 8 hours after dosing compared to placebo. Phase I and II trials were carried out in Japan.

Studies have been performed in animals and bepotastine besilate was not found to be teratogenic in rats during fetal development, even at 3,300 times more that typical use in humans.[3] Evidence of infertility was seen in rats at 33,000 times the typical ocular does in humans.[3] The safety and efficacy has not been established in patients under 2 years of age and has been extrapolated from adults for patients under 10 years of age.[3]

SYN

EP 0335586; JP 1989242574; JP 1990025465; JP 1993294929; US 4929618

The reaction of 4-[1-(4-chlorophenyl)-1-(2-pyridyl)methoxy]piperidine (I) with ethyl 4-bromobutyrate (II) by means of K2CO3 in refluxing acetone gives the corresponding condensation product (III), which is then hydrolyzed with NaOH in ethanol/water yielding compound (IV).

SYN 2

JP 1998237070; JP 2000198784; WO 9829409

A new synthesis of betotastine has been developed: The racemic 4-[1-(4-chlorophenyl)-1-(2-pyridyl)methoxy]piperidine (I) is submitted to optical resolution with N-acyl amino acids such as N-acetyl-L-phenylalanine (preferred), N-acetyl-L-leucine, N-(benzyloxycarbonyl)-L-phenylalanine, N-(benzyloxycarbonyl)-L-valine, N-(benzyloxycarbonyl)-L-threonine, N-(benzyloxycarbonyl)-L-serine or with (2R,3R)-3-(5-chloro-2-nitrophenylsulfanyl)-2-hydroxy-3-(4-methoxyphenyl)propionic acid (preferred) or (2R,3R)-2-hydroxy-3-(4-methoxyphenyl)-3-(2-nitrophenylsulfanyl)propionic acid as chiral intermediates, yielding the (S)-isomer (II). The condensation of (II) with ethyl 4-bromobutyrate (III) by means of a base such as Na2CO3, NaHCO3, K2CO3 or KHCO3 gives the expected 4-(1-piperidinyl)butyric acid ester (IV), which is finally hydrolyzed with NaOH or KOH in aqueous ethanol or methanol.

SYN 3

A new synthesis of betotastine has been developed: The racemic 4-[1-(4-chlorophenyl)-1-(2-pyridyl)methoxy]piperidine (I) is submitted to optical resolution with N-acyl amino acids such as N-acetyl-L-phenylalanine (preferred), N-acetyl-L-leucine, N-(benzyloxycarbonyl)-L-phenylalanine, N-(benzyloxycarbonyl)-L-valine, N-(benzyloxycarbonyl)-L-threonine, N-(benzyloxycarbonyl)-L-serine or with (2R,3R)-3-(5-chloro-2-nitrophenylsulfanyl)-2-hydroxy-3-(4-methoxyphenyl)propionic acid (preferred) or (2R,3R)-2-hydroxy-3-(4-methoxyphenyl)-3-(2-nitrophenylsulfanyl)propionic acid as chiral intermediates, yielding the (S)-isomer (II). The condensation of (II) with ethyl 4-bromobutyrate (III) by means of a base such as Na2CO3, NaHCO3, K2CO3 or KHCO3 gives the expected 4-(1-piperidinyl)butyric acid ester (IV), which is finally hydrolyzed with NaOH or KOH in aqueous ethanol or methanol.

CLIP

A Novel Synthetic Method for Bepotastine, a Histamine H1 Receptor …

journal.kcsnet.or.kr

A Novel Synthetic Method for Bepotastine, a Histamine H1 Receptor Antagonist

file:///C:/Users/91200291/Downloads/B130241_549.pdf

Scheme 1. Synthesis of bepotastine l-menthyl ester N-benzyloxycarbonyl-L-aspartic acid complex (3), bepotastine besilate (4) and bepotastine calcium (5). Reagents and conditions; i) 4-bromobutanoic acid l-menthyl ester, K2CO3, acetone, reflux, 7 h, 95-99%; ii) N-benzyloxycarbonyl-L-aspartic acid (NCbzLAA), ethyl acetate, rt, 12 h, 71-73%; iii) Ethyl acetate/H2O, NaHCO3, 97-99%; iv) EtOH:H2O = 1:1, NaOH, rt, 12 h, 3.0 N-HCl Neutralization, 92- 95%; v) AcOH, reflux, 12 h, racemization 97-100%; vi) Bezensulfonic acid, acetonitrile, rt, 12 h, 64-67%; vii) NaOH, H2O, CaCl2, rt, 12 h, 86-89%.

Synthesis of (S)-Bepotastine Besilate (4). Bepotastine (50 g, 0.13 mol) was dissolved in 500 mL of acetonitrile, and benzenesulfonic acid monohydrate (20 g, 0.11 mol) was added to the reaction mixture. Bepotastine besilate (0.5 g, 1.28 mmol) was seeded in the reaction mixture and stirred at rt for 12 h. The solid precipitate was filtered and dried. The product was obtained 38 g (yield: 64%, optical purity: 99.5% ee) as a pale white crystalline powder. Melting point: 161- 163 o C. Water: 0.2% (Karl-Fischer water determination). MS: m/z 389.1 [M+H]; 1 H-NMR (300 MHz, DMSO-d6) δ 9.2 (br s, 1H), 8.5 (d, J = 4.1 Hz, 1H), 7.8 (t, J = 7.7 Hz, 1H), 7.6 (m, 3H), 7.4 (m, 4H), 7.3 (m, 4H), 5.7 (s, 1H), 3.7 (br s, 2H), 3.3 (br s, 3H), 3.1 (br s, 2H), 2.3 (t, J = 14.1 Hz, 2H), 2.2 (m, 1H), 2.0 (m, 1H), 1.8 (m, 3H), 1.7 (m, 1H); IR (KBr, cm−1 ): 3422, 2996, 2909, 2735, 2690, 2628, 1719, 1592, 1572, 1488, 1470, 1436, 1411, 1320, 1274, 1221, 1160, 1123, 1066, 1031, 1014, 996, 849, 830, 771, 759, 727, 693, 612, 564

Synthesis

Patent ID Title Submitted Date Granted Date
US9849121 AQUEOUS LIQUID PREPARATIONS AND LIGHT-STABILIZED AQUEOUS LIQUID PREPARATIONS
2014-10-10
2015-01-29
US2012225905 BEPOTASTINE COMPOSITIONS
2012-05-02
2012-09-06
US8883825 Aqueous liquid preparations and light-stabilized aqueous liquid preparations
2012-08-30
2014-11-11
Patent ID Title Submitted Date Granted Date
US6638534 Preparation capable of releasing drug at target site in intestine
2001-01-29
2003-10-28
US2010168433 PROCESS FOR PREPARING BEPOTASTINE AND INTERMEDIATES USED THEREIN
2010-07-01
US7282589 Acid addition salt of optically active piperidine compound and process for preparing the same
2004-11-04
2007-10-16
US6307052 Acid-addition salts of optically active piperidine compound and process for producing the same
2001-10-23
EP0949260 ACID-ADDITION SALTS OF OPTICALLY ACTIVE PIPERIDINE COMPOUND AND PROCESS FOR PRODUCING THE SAME
1999-10-13
2002-05-22
Patent ID Title Submitted Date Granted Date
US9446055 DISINTEGRATING PARTICLE COMPOSITION AND ORALLY RAPIDLY DISINTEGRATING TABLET
2010-08-11
2012-06-21
US2011257628 INSTRUMENT FOR ALLEVIATING ADDICTIVE DRUG CRAVING, METHOD FOR USING SAME AND METHOD FOR TREATING ADDICTIVE DRUG DEPENDENCE
2009-12-02
2011-10-20
US2013046240 BEPOTASTINE COMPOSITIONS
2011-10-06
2013-02-21
US2012328675 FILM PREPARATION CONTAINING MEDICAMENT WITH UNPLEASANT TASTE
2011-03-03
2012-12-27
US8771724 Percutaneous absorption enhancer and transdermal preparation using the same
2009-06-22
2014-07-08
Patent ID Title Submitted Date Granted Date
US6780877 Acid addition salt of optically active piperidine compound and process for preparing the same
2002-02-28
2004-08-24
US8877168 Aqueous liquid preparations and light-stabilized aqueous liquid preparations
2014-06-25
2014-11-04
US8784789 Aqueous liquid preparations and light-stabilized aqueous liquid preparations
2003-07-30
2014-07-22
US2010137367 NOVEL CRYSTALLINE BEPOTASTINE METAL SALT HYDRATE, METHOD FOR PREPARING SAME, AND PHARMACEUTICAL COMPOSITION COMPRISING SAME
2010-06-03
US8900602 Disintegrating particle composition and orally rapidly disintegrating tablet
2010-08-11
2014-12-02
Bepotastine
Bepotastine.svg
Clinical data
Trade names Bepreve
AHFS/Drugs.com International Drug Names
MedlinePlus a610012
Pregnancy
category
  • US: C (Risk not ruled out)
Routes of
administration
Oral, topical (eye drops)
ATC code
  • none
Legal status
Legal status
  • In general: ℞ (Prescription only)
Pharmacokinetic data
Bioavailability High (oral)
Minimal (topical)
Protein binding ~55%
Excretion Renal (75–90%)
Identifiers
CAS Number
PubChem CID
DrugBank
ChemSpider
UNII
KEGG
ChEBI
ChEMBL
Chemical and physical data
Formula C21H25ClN2O3
Molar mass 388.88 g/mol
3D model (JSmol)

References

    • EP 335 586 (Ube Ind.; appl. 22.3.1989; J-prior. 25.3.1988).
    • EP 485 984 (Ube Ind.; appl. 13.11.1991; J-prior. 15.11.1990).
    • WO 9 829 409 (Ube Ind.; appl. 25.12.1997; J-prior. 26.12.1996).
  • racemization :

    • JP 10 237 069 (Ube Ind.; appl. 21.2.1997).

References

  1. Jump up^ H. Takahashi; A. Ishida-Yamamoto; H. Iizuka (September 2004). “Effects of bepotastine, cetirizine, fexofenadine, and olopatadine on histamine-induced wheal-and flare-response, sedation, and psychomotor performance”Clinical and Experimental Dermatology29: 526–532. doi:10.1111/j.1365-2230.2004.01618.x.
  2. Jump up^ “Bepotastine Monograph”LexiComp.
  3. Jump up to:a b c d e f “Bepreve prescribing Information” (PDF).
  4. Jump up^ [phx.corporate-ir.net/External.File?item…t=1 “2011 Net Revenues Increase to $160.3 Million On an Adjusted Cash Net Income Basis, ISTA Posts Second Year of Profitability Company Reaffirms 2012 Financial Guidance”] Check |url= value (help).
  5. Jump up^ “Bausch & Lomb to Buy ISTA Pharmaceuticals for $500 Million”DealBook. Retrieved 2015-12-05.
  6. Jump up^ “Bausch & Lomb Inc. et al. v. Micro Labs USA, Inc. et al.”
  7. Jump up^ “Valenant pharmaceuticals eyes China with Bausch deal”.
  8. Jump up^ Macejko, Thomas T.; Bergmann, Mark T.; Williams, Jon I.; Gow, James A.; Gomes, Paul J.; McNamara, Timothy R.; Abelson, Mark B. (2010-07-01). “Multicenter Clinical Evaluation of Bepotastine Besilate Ophthalmic Solutions 1.0% and 1.5% to Treat Allergic Conjunctivitis”American Journal of Ophthalmology150 (1): 122–127.e5. doi:10.1016/j.ajo.2010.02.007.

////////////Bepotastine Besilate, ベポタスチンベシル酸塩  ,Talion , tau284, TAU-284DS, TAU-284, DA-5206
HL-151 , SNJ-1773

C1CN(CCC1OC(C2=CC=C(C=C2)Cl)C3=CC=CC=N3)CCCC(=O)O.C1=CC=C(C=C1)S(=O)(=O)O


Arterolane Maleate

$
0
0

Arterolane.svg

Image result for Arterolane Maleate

Image result for Arterolane Maleate

Arteolane Maleate

C26H40N2O8
Molecular Weight: 508.612

CAS  959520-73-1

664338-39-0 (free base)   959520-73-1 (maleate)   959520-79-7 (acetate)   664338-40-3 (tosylate)   959520-82-2 (tartrate)   959520-83-3 (citrate)

N-(2-amino-2-methylpropyl)-2-((1R,3R,4”S,5R,5’s,7R)-dispiro[adamantane-2,3′-[1,2,4]trioxolane-5′,1”-cyclohexan]-4”-yl)acetamide maleate

Dispiro[cyclohexane-1,3′-[1,2,4]trioxolane-5′,2”-tricyclo[3.3.1.13,7]decane]-4-acetamide, N-(2-amino-2-methylpropyl)-, cis-, (2Z)-2-butenedioate (1:1)

APPROVED 4.11.2017 CDSCO

Arteolane Maleate and Piperaquine phosphate Dispersible tablets (37.5 mg +187.5 mg

Indicated in children aged 6 months to 12 years for the treatment of :
– Acute uncomplicated Plasmodium falciparum malaria infection
– Acute uncomplicated Plasmodium Vivax malaria infection
 Arterolane, also known as OZ-277, is an adenosine triphosphatase inhibitor potentially for the treatment of malaria.

Arterolane.png

cas 664338-39-0 

Arterolane

664338-39-0, UNII-3N1TN351VB, OZ277, RBX-11160, NCGC00274173-01
Molecular Formula: C22H36N2O4
 Molecular Weight: 392.53224
 cis-adamantane-2-spiro-3’-8’-[[[(2’-amino-2’ methylpropyl) amino] carbonyl] methyl] 1’,2’,4’-trioxaspiro [4.5] decane
cis-adamantane-2-spiro-3′-8′-[[[(2′- amino-2′-methylpropyl)amino]carbonyl]-methyl]- 1 ‘,2′,4’-trioxaspiro[4.5]decane

Arterolane, also known as OZ277 or RBx 11160, is a substance that was tested for antimalarial activity[1] by Ranbaxy Laboratories.[2] It was discovered by US and European scientists who were coordinated by the Medicines for Malaria Venture (MMV).[3] Its molecular structure is uncommon for pharmacological compounds in that it has both a ozonide (trioxolane) group and an adamantanesubstituent.[4]

Initial results were disappointing, and in 2007 MMV withdrew support, after having invested $20M in the research;[5] Ranbaxy said at the time that it intended to continue developing the drug combination on its own.[2] Ranbaxy started a Phase II clinical trial of arterolane, in combination with piperaquine in 2009 that published in 2015.[6][7]

In 2012, Ranbaxy obtained approval to market the arterolane/piperaquine combination drug in India, under the brand name Synriam,[5]and in 2014 received approval to market it in Nigeria, Uganda, Senegal, Cameroon, Guinea, Kenya and Ivory Coast; it had already received approval in Uganda.[8]

Ranbaxy launched India’s first new drug, SynriamTM, treating Plasmodium falciparummalaria in adults. The drug provides quick relief from most malaria-related symptoms, including fever, and has a high cure rate of over 95 %.

Just one tablet per day is required, for three days, instead of two to four tablets, twice daily, for three or more days with other medicines. The drug is independent of dietary restrictions for fatty foods or milk.

Ranbaxy developed Synriam as a fixed-dose combination of arterolane maleate and piperaquine phosphate, where arterolane is the new chemical entity (NCE) that was developed as an alternative to artemisinin. It is the first recently developed antimalarial not based on artemisinin, one of the most effective treatments for malaria, which has shown problems with resistance in recent years. Arterolane was discovered by a collaborative drug discovery project funded by the Medicines for Malaria Venture. Since SynriamTM has a synthetic source, unlike artemisinin-based drugs, production can be scaled up whenever required and a consistent supply can be maintained at a low cost.

The new drug, has been approved by the Drug Controller General of India (DCGI) for marketing in India and conforms to the recommendations of the World Health Organization (WHO) for using combination therapy in malaria. Ranbaxy is also working to make it available in African, Asian and South American markets where Malaria is rampant. SynriamTM trials are ongoing for Plasmodium vivax malaria and a paediatric formulation.

Derek Lowe of the famous In the Pipeline blog had written about arterolane in 2009. At the time it was in Phase III trial, which I assumed were the trials that Ranbaxy was conducting. But it turned out that arterolane was developed by a collaboration between researchers in the US, the UK, Switzerland and Australia who were funded by the World Health Organization and Medicines for Malaria Venture (a Swiss non-profit). They published this work in Nature in 2004 and further SAR (Structure Activity Relationship) studies in J Med Chem in 2010. So Ranbaxy did not develop the drug from scratch? But the press release quotes Arun Sawhney, CEO and Managing Director of Ranbaxy which misleads people to think so: “It is indeed gratifying to see that Ranbaxy’s scientists have been able to gift our great nation its first new drug, to treat malaria, a disease endemic to our part of the world. This is a historic day for science and technology in India as well as for the pharmaceutical industry in the country. Today, India joins the elite and exclusive club of nations of the world that have demonstrated the capability of developing a new drug”. So Ranbaxy mixes a known active compound (piperaquine) with a new compound that someone else found to be active (arterolane) and claims that they developed a new drug? In an interview in LiveMint, Sawhney says, “Ranbaxy spent around $30 million on Synriam and the contribution from DST [India’s Department of Science & Technology] was Rs.5 crore. The drug went through several phases of development since the project began in 2003. We did not look at this as a commercial development. Instead, this is a CSR [Corporate Social Responsibility] venture for us.” That’s a give away because developing a new drug from scratch has to cost more than $30 million + Rs.50 million.


Ranbaxy  now taken over by sun

SynriamTM

Generic Name
Arterolane Maleate and Piperaquine Phosphate Tablets
Composition
Each film coated tablet contains: Arterolane maleate equivalent to Arterolane ……………………………150 mg Piperaquinephosphate……………750 mg
Dosage Form
Tablets
Inactive ingredients:
Microcrystalline cellulose, Crospovidone, Magnesium stearate, Hydroxypropyl methyl cellulose/Hypromellose, Titanium dioxide, Macrogol/ Polyethylene glycol, Talc, Ferric Oxide (Yellow), Ferric Oxide (Red)

Description SynriamTM is a fixed dose combination of two antimalarial active ingredients arterolane maleate and piperaquine phosphate.

Arterolane maleate is a synthetic trioxolane compound. The chemical name of arterolane maleate is cis-adamantane-2-spiro-3’-8’-[[[(2’-amino-2’ methylpropyl) amino] carbonyl] methyl] 1’,2’,4’-trioxaspiro [4.5] decane hydrogen maleate. The molecular formula is C26H40N2O8 and molecular weight is 508.61. The structural formula is as follows:

MALARIA
Malaria is one of the most prevalent and deadly parasitic diseases in the world. Up to 289 million cases of malaria may have occurred in 2010, causing between 660,000 and 1.25 million deaths, mainly in Africa and mostly of children younger than 5 years.
(WHO: http://www.who.int/malaria/publications/world_malaria_report_2012/en/index.html; Fidock, D. A. Eliminating Malaria. Science 2013, 340, 1531-1533.)

The most serious problem in malaria treatment is that the parasites causing the disease, particularly the deadly Plasmodium falciparum, have developed resistance to widely used drugs, particularly chloroquine (CQ). Currently, the most efficacious therapies are combinations of an artemisinin-type compound with a long-lasting partner drug like lumefantrine, amodiaquine or mefloquine.

Malaria, the most common parasitic disease of humans, remains a major health and economic burden in most tropical countries. Large areas of Central and South America, Hispaniola (Haiti and the Dominican Republic), Africa, the Middle East, the Indian subcontinent, Southeast Asia, and Oceania are considered as malaria-risk areas. It leads to a heavy toll of illness and death, especially amongst children and pregnant women.

According to the World Health Organization, it is estimated that the disease infects about 400 million people each year, and around two to three million people die from malaria every year. There are four kinds of malaria parasites that infect human: Plasmodium falciparum, Plasmodium vivax, Plasmodium ovale and Plasmodium malariae.

Malaria spreads from one person to another by the bite of mosquito, Anopheles gambiae, which serves as vector. When a mosquito sucks the blood of human, sporozoites are transfused into the human body together with saliva of the mosquito. The sporozoites enter into the hepatocytes, reproduce asexually and finally enter into the blood stream. The parasites continue to multiply inside the red blood cells, until they burst and release large number of merozoites. This process continues, destroying a significant number of blood cells and causing the characteristic paroxysm (“chills and fever”) associated with the disease. In the red blood cells, some of the merozoites become male or female gametocytes. These gametocytes are ingested by the mosquito when it feeds on blood. The gametocytes fuse in the vector’s gut; sporozoites are produced and are migrated to the vector’s salivary glands.

The clinical symptoms of malaria are generally associated with the bursting of red blood cells causing an intense fever associated with chills that can leave the infected individual exhausted and bedridden. More severe symptoms associated with repeat infections and/or infection by Plasmodium falciparum include anaemia, severe headaches, convulsions, delirium and, in some instances, death.

Quinine, an antimalarial compound that is extracted from the bark of cinchona tree, is one of the oldest and most effective drugs in existence. Chloroquine and mefloquine are the synthetic analogs of quinine developed in 1940’s, which due to their effectiveness, ease of manufacture, and general lack of side effects, became the drugs of choice. The downside to quinine and its derivatives is that they are short-acting and have bitter taste. Further, they fail to prevent disease relapses and are also associated with side effects commonly known as “Chinchonism syndrome” characterized by nausea, vomiting, dizziness, vertigo and deafness. However, in recent years, with the emergence of drug- resistant strains of parasite and insecticide-resistant strains of vector, the treatment and/or control of malaria is becoming difficult with these conventional drugs.

Malarial treatment further progressed with the discovery of Artemisinin

(qinghaosu), a naturally occurring endoperoxide sesquiterpene lactone isolated from the plant Artemisia annua (Meshnick et al., Microbiol. Rev. 1996, 60, p. 301-315; Vroman et al., Curr. Pharm. Design, 1999, 5, p. 101-138; Dhingra et al., 2000, 66, p. 279-300), and a number of its precursors, metabolites and semi-synthetic derivatives which have shown to possess antimalarial properties. The antimalarial action of artemisinin is due to its reaction with iron in free heme molecules of the malaria parasite, with the generation of free radicals leading to cellular destruction. This initiated a substantial effort to elucidate its molecular mechanism of action (Jefford, dv. Drug Res. 1997, 29, p. 271-325; Cumming et al., Adv. Pharmacol. 1997, 37, p. 254-297) and to identify novel antimalarial peroxides (Dong and Vennerstrom, Expert Opin. Ther. Patents 2001, 1 1, p. 1753-1760).

Although the clinically useful artemisinin derivatives are rapid acting and potent antimalarial drugs, they have several disadvantages including recrudescence,

neurotoxicity, (Wesche et al., Antimicrob. Agents. Chemother. 1994, 38, p. 1813-1819) and metabolic instability (White, Trans. R. Soc. Trop. Med. Hyg., 1994, 88, p. 41-43). A fair number of these compounds are quite active in vitro, but most suffer from low oral activity (White, Trans. R. Soc. Trop. Med. Hyg., 1994, 88, p. 41-43 and van Agtmael et al., Trends Pharmacol. Sci., 1999, 20, p. 199-205). Further all these artemisinin derivatives are conventionally obtained from plant source and are therefore expensive. As the cultivation of the plant material is dependent on many factors including the weather conditions, the supply source thus becomes finite and there are chances of varying yield and potency. This leads to quality inconsistencies and supply constraints. As malaria is more prevalent in developing countries, a switch to cheaper and effective medicine is highly desirable.

Thus there exists a need in the art to identify new peroxide antimalarial agents, especially those which are not dependent on plant source and can be easily synthesized, are devoid of neurotoxicity, and which possess improved solubility, stability and pharmacokinetic properties.

Following that, many synthetic antimalarial 1 ,2,4-trioxanes (Jefford, Adv. Drug Res. 1997, 29, p. 271-325; Cumming et al., Adv. Pharmacol. 1997, 37, p. 254-297), 1,2,4,5-tetraoxanes (Vennerstrom et al., J. Med. Chem., 2000, 43, p. 2753-2758), and other endoperoxides have been prepared. Various patents/applications disclose means and method for treating malaria using Spiro or dispiro 1,2,4-trioxolanes for example, U.S.

Patent Application No. 2004/0186168 and U.S. Patent Nos. 6,486, 199 and 6,825,230. The present invention relates to solid dosage forms of the various spiro or dispiro 1 ,2,4- trioxolanes antimalarial compounds disclosed in these patents/applications and are incorporated herein by reference.

Active compounds representing various Spiro and dispiro 1 ,2,4-trioxolane derivatives possess excellent potency, efficacy against Plasmodium parasites, and a lower degree of neurotoxicity, in addition to their structural simplicity and ease of synthesis. Furthermore, these compounds have half-lives which are believed to permit short-term treatment regimens comparing favorably to other artemisinin-like drugs. In general, the therapeutic dose of trioxolane derivative may range between about 0.1-1000 mg/kg/day, in particular between about 1-100 mg/kg/day. The foregoing dose may be administered as a single dose or may be divided into multiple doses. For malaria prevention, a typical dosing schedule could be, for example, 2.0-1000 mg/kg weekly beginning 1-2 weeks prior to malaria exposure, continued up to 1-2 weeks post-exposure.

Monotherapy with artemisinin (natural or synthetic) class of drugs might cure the patients within 3 days, however perceiving the potential threat of the malarial parasite developing resistance towards otherwise very potent artemisinin class of drugs, WHO had strictly called for an immediate halt to the provision of single-drug artemisinin malaria pills. Combination therapy in case of malaria retards the development of resistance, improve efficacy by lowering recrudescence rate, provides synergistic effect, and increase exposure of the parasite to the drugs.

Artemsinin based combinations are available in the market for a long time.

Artemether-lumafentrine (Co-artem®) was the first fixed dose antimalarial combination containing an artemisinin derivative and has been known since 1999. This combination has passed extensive safety and efficacy trials and has been approved by more than 70 regulatory agencies. Co-artem® is recommended by WHO as the first line treatment for uncomplicated malaria.

Other artemisinin based combinations include artesunate and amodiaquine (Coarsucam®), and dihydroartemisin and piperaquine (Eurartesim®). Unfortunately, all the available artemisinin based combinations have complicated dosage regimens making it difficult and inconvenient for a patient to comply completely with the total prescribed duration. For example, the dosage regimen of Co-artem® for an adult having body weight of more than 35 kg includes 6 doses over three days. The first dose comprises four tablets initially, the second dose comprises four tablets after eight hours, the third to sixth doses comprise four tablets twice for another two days; making it a total of 24 tablets. The dosage regimen of Coarsucam® for an adult having body weight of more than 36 kg or age above 14 years includes three doses over three days; each dose comprises two tablets; making it a total of six tablets. The dosage regimen of Eurartesim® for an adult having body weight between 36 kg – 75 kg includes 3 doses over three days, each dose comprises of three tablets, making it a total of nine tablets.

It is evident that the available artemisinin-based combinations have a high pill burden on patients as they need to consume too many tablets. As noted above, this may increase the possibility of missing a few doses, and, consequently, could result in reduced efficacy due to non-compliance and may even lead to development of resistance for the drug. Therefore, there is an urgent and unmet need for anti-malarial combinations with a simplified daily dosing regimen that reduces the pill burden and would increase patient compliance.

Apart from simplifying the regimen, there are certain limitations for formulators developing formulations with trioxolones, the first being their susceptibility to degradation in presence of moisture that results in reduced shelf lives. Another is their bitter taste, which can result in poor compliance of the regimen or selection of another, possibly less effective, therapeutic agent.

……………………..

http://www.google.st/patents/US6906205

Figure US06906205-20050614-C00051

……………………

http://www.google.st/patents/WO2013008218A1?cl=en

structural Formula II.

Figure imgf000013_0001

Formula II

Active compound includes one or more of the various spiro and dispiro trioxolane derivatives disclosed in U.S. Application No. 2004/0186168 and U.S. Patent Nos.

6,486,199 and 6,825,230, which are incorporated herein by reference. These trioxolanes are relatively sterically hindered on at least one side of the trioxolane heterocycle which provides better in vivo activity, especially with respect to oral administration. Particularly, spiro and dispiro 1,2,4-trioxolanes derivatives possess excellent potency and efficacy against Plasmodium parasites, and a lower degree of neurotoxicity.

The term “Active compound I” herein means cis-adamantane-2-spiro-3′-8′-[[[(2′- amino-2′-methylpropyl)amino]carbonyl]-methyl]- 1 ‘,2′,4’-trioxaspiro[4.5]decane hydrogen maleate. The Active compound I may be present in an amount of from about 5% to about 25%, w/w based on the total dosage form.

………………

http://www.google.st/patents/WO2007138435A2?cl=en

A synthetic procedure for preparing compounds of Formula I, salts of the free base c«-adamantane-2-spiro-3′-8′-[[[(2′-amino-2′-methyl propyl) amino] carbonyl] methyl]- 1 ‘, 2′, 4’-trioxaspiro [4.5] decane has been disclosed in U.S. 6,906,205.

Figure imgf000002_0001

The process for the preparation of compounds of Formula I wherein a compound of Formula II (wherein R is lower alkyl) is reacted with a compound of Formula III (wherein R is lower alkyl) to obtain compound of Formula IV;

Figure imgf000005_0001
Figure imgf000005_0002

Formula Formula IV

followed by hydrolysis of the compounds of Formula IV to give a compound of Formula V;

Figure imgf000005_0003

Formula V followed by the reaction of the compound of Formula V with an activating agent, for example, methyl chloroformate, ethyl chloroformate, propyl chloro formate, n-butyl chloro formate, isobutyl chloroformate or pivaloyl chloride leads to the formation of mixed anhydride, which is reacted in situ reaction with 1 ,2-diamino-2-methyl propane to give a compound of Formula VI; and

Figure imgf000005_0004

Formula Vl reacting the compound of Formula VI with an acid of Formula HX (wherein X can be the same as defined earlier) to give compounds of Formula I.

Example 1 : Preparation of O-methyl-2-adamantanone oxime

To a solution of 2-adamantanone (50 g, 0.3328 mol, 1 equiv.) in methanol (0.25 lit), sodium hydroxide solution (15 g, 0.3761mol, 1.13 equiv, in 50 mL water) was added followed by methoxylamine hydrochloride (37.5 g x 81.59% Purity= 30.596 g, 0.366 mol, 1.1 equiv) at room temperature under stirring. The reaction mixture was stirred at room temperature for 1 to 2 h. The reaction was monitored by HPLC. The reaction mixture was concentrated at 40- 45°C under vacuum to get a thick residue. Water (250 mL) was added at room temperature and the reaction mixture was stirred for half an hour. The white solid was filtered, washed with water (50 mL), and dried at 40 to 45°C under reduced pressure. O-methyl 2- adamantanone oxime (57 g, 95 % yield) was obtained as a white solid.

(M++l) 180, 1HNMR (400 MHz, CDCl3 ): δ 1.98 – 1.79 (m, 12H), 2.53 (s, IH), 3.46 ( s, IH), 3.81 (s, 3H).

Example 2: Preparation of 4-(methoxycarbonvmethvPcvclohexanone A high pressure autoclave was charged with a mixture of methyl (4- hydroxyphenyl)acetate (50 g, 0.30 mol), palladium ( 5g) (10 %) on carbon (50 % wet) and O- xylene (250 mL). The reaction mixture was stirred under 110 to 115 psi of hydrogen pressure for 7 to 8 h at 1400C. The reaction was monitored by HPLC. The reaction mixture was then cooled to room temperature, and the catalyst was filtered off. Filtrate was concentrated under reduced pressure to get 4-(methoxycarbonylmethyl)cyclohexanone as light yellow to colorless oily liquid (48.7 g, 97.4 %).

(M++!) 171, ‘ HNMR (400 MHz, CDCl 3): δ 1.48 – 1.51 ( m, 2H), 2.1 1-2.07 (m, 2H), 2.4- 2.23 (m, 7H), 3.7 (s, 3H).

Example 3: Preparation of methyl (Is, 4s)-dispiro [cyclohexane-l, 3′-f 1,2,4] trioxolane-5′, 2″-tricvclor3.3.1.1371decan1-4-ylacetate

A solution of O-methyl-2-adamantanone oxime (example 1) (11.06 g, 61.7 mmol, 1.5 equiv.) and 4-(methoxycarbonymethyl)cyclohexanone (example 2) (7.0 g, 41.1 mmol, 1 equiv.) in cyclohexane ( 200ml) and dichloromethane (40 mL) was treated with ozone (ozone was produced with an OREC ozone generator [0.6 L/min. O2, 60 V] passed through an empty gas washing bottle that was cooled to -780C). The solvent was removed after the reaction was complete. After removal of solvents, the crude product was purified by crystallization from 80% aqueous ethanol (200 mL) to afford the title compound as a colorless solid. Yield: 10.83 g, 78%, mp: 96-980C; 1HNMR (500 Hz3CDCl3): δ 1.20-1.33 (m, 2H), 1.61-2.09 (m, 5 21H), 2.22 (d, J = 6.8Hz, 2H), 3.67(s,3H).

Example 4: Preparation of (Is, 4s)-dispiro [cyclohexane-1, 3′-[l,2,4] trioxolane-5′, 2″- tricvclo [3.3.1.137] decanl-4-ylacetic acid

Sodium hydroxide (3.86 g, 96.57 mmol, 3 equiv.) in water (80 mL) was added to a solution of methyl (\s, 4s)-dispiro [cyclohexane-1, 3′-[l,2,4] trioxolane-5′, 2″-tricyclo

10 [3.3.1.I37] decan]-4-ylacetate (example 3) (10.83 g, 32.19 mmol, 1 equiv.) in 95% ethanol (150 mL). The mixture was stirred at 500C for about 4 h, cooled to O0C, and treated with IM hydrochloric acid (129ml, 4 equiv). The precipitate was collected by filtration, washed with 50 % aqueous ethanol (150 mL) and dried in vacuum at 40 0C to give the title compound as colorless solid. Yield: 9.952 g, 96%, mp: 146-1480C ( 95% ethanol), 1HNMR (500 Hz,

15 CDCl3): δ 1.19-1.41 (m,2H), 1.60-2.05 (m,21H), 2.27 (d, J=6.8 Hz,2H).

Example 5: Preparation of c?s-adamantane-2-spiro-3′-8′-[[[(2′-amino-2′-methyl propyl) amino] carbonyl] methyl]-! ‘, T , 4’-trioxaspiro [4.5] decane

Method A:

(Is, 4s)-dispiro[cyclohexane- 1 ,3 ‘-[ 1 ,2,4]trioxolane-5 ‘,2 ‘ ‘-tricyclo[3.3.1.137]decan]-4-

.0 ylacetic acid (example 4) (5 g ,15.5mmol, 1 equiv) was mixed with triethylamine (2.5 g , 24.8 mmol, 1.6 equiv) in 100ml of dichloromethane. The reaction mixture was cooled to – 1O0C to 00C. Ethyl chloro formate (1.68 g, 17 mmol, 1.0 equiv) in 15 mL dichloromethane was charged to the above reaction mixture at – 100C to 00C. The reaction mixture was stirred at the same temperature for 10 to 30 minutes. The resulting mixed anhydride reaction mixture

15 was added dropwise to a previously prepared solution of l,2-diamino-2-methylpropane (1.64 g, 18.6 mmol, 1.2 equiv), in 100 mL dichloromethane at -100C to O0C. The temperature of reaction mixture was raised to room temperature. The reaction mixture was stirred at the same temperature till the reaction was complete. Reaction monitoring was done by thin layer chromatography using 5 to 10% methanol in dichloromethane. The reaction was complete

>0 within 2 h. Nitrogen atmosphere was maintained throughout the reaction. Water (50 mL) was charged, organic layer was separated and washed with 10% sodium bicarbonate solution (50 mL) and water (50 mL) at room temperature. The organic layer was dried over sodium sulphate and the solvent was removed at 25 to 4O0C under reduced pressure. Hexane (50ml) was added to obtain residue under stirring at room temperature. The mixture was filtered and washed with 5 mL of chilled hexane. The solid was dried under reduced pressure at room 5 temperature.

Yield: 5.2 g (85.4 %), (M++l) 393, 1HNMR (400 MHz, DMSO-J6 ): δ 0.929 ( s, 6H), 1.105 – 1.079 (m, 2H), 1.887-1.641 (m, 21H), 2.030-2.017 (d, 2H), 2.928 (d, 2H).

Method B:

(Is, 4s)-dispiro [cyclohexane-1, 3′-[l,2,4] trioxolane-5′, 2″-tricyclo [3.3.1.I37]

10 decan]-4-ylacetic acid (example 4) (10 g, 31mmol, 1 equiv) was treated with isobutyl chloroformate (4.5 g, 33mmol, 1.1 equiv) in presence of organic base like triethyl amine (5 g, 49.6mmol, 1.6 equiv) at 00C to 7°C in 250ml of dichloromethane. The solution was stirred at O0C to 7°C for aboutlO to 30 minutes. To the above reaction mixture, previously prepared solution of l,2-diamino-2-methylpropane (3.27 g, 37 mmol, 1.2 equiv), in 50 mL of

15 dichloromethane was added at O0C to 7°C in one lot. The temperature of reaction mixture was raised to room temperature. The reaction mixture was stirred at the room temperature till reaction was over. Reaction monitoring was done by thin layer chromatography using 5 to 10% methanol in dichloromethane. Reaction was complete within 2 h. The reaction nitrogen atmosphere was maintained throughout the reaction. Water (250 mL) was charged, organic

20 layer was separated and washed with 10% sodium bicarbonate solution (200 mL) and water (100 mL) at room temperature and the solvent was removed at 25 to 4O0C under reduced pressure. Hexane (100ml) was added to the residue, under stirring, at room temperature. The mixture was filtered and washed with chilled hexane (10 mL). The resultant solid was dried under reduced pressure at room temperature. Yield: 10.63 g (87%), (M++l) 393, 1HNMR

>5 (400 MHz, DMSO-J6 ) :δ 0.928 ( s, 6H), 1.102 – 1.074 (m, 2H), 1.859-1.616 (m, 21H), 2.031- 2.013 (d, 2H), 2.94-2.925 (d, 2H). Method C:

(\s, 4s)-dispiro[cyclohexane-l,3′-[l,2,4]trioxolane-5′,2″-tricyclo[3.3.1.13>7]decan]-4- ylacetic acid (example 4) (5 g, 15.5mmol, 1 equiv) was treated with pivaloyl chloride (1.87 g, 15.5 mmol, 1 equiv) and triethylamine (2.5gm, 24.8mmol, 1.6 equiv) at -15°C to -100C in dichloromethane (125 mL). The solution was stirred at -150C to -100C for aboutlO to 30 minutes. It resulted in the formation of mixed anydride. To the above reaction mixture, previously prepared solution of 1 ,2-diamino-2-methylpropane (1.64 g, 18.6 mmol, 1.2 equiv) in 25 mL dichloromethane was added at -15°C to -100C. The temperature of reaction mixture was raised to room temperature. The reaction mixture was stirred at the room temperature till reaction was over. Reaction monitoring was done by thin layer chromatography using 5 to 10% methanol in dichloromethane. The reaction was complete within 2 h. Nitrogen atmosphere was maintained throughout the reaction. Water (125 mL) was charged, organic layer was separated and washed with 50 mL of 10% sodium bicarbonate solution and 125 mL of water, respectively at room temperature. Finally solvent was removed at 25 to 4O0C under reduced pressure. 50 mL of 5% Ethyl acetate – hexane solvent mixture was added to the residue under stirring at room temperature. The mixture was filtered and washed with 5 mL of chilled hexane. Solid was dried under reduced pressure at room temperature. Yield: 5.03 g (83 %), (M++l) 393, 1JINMR (400 MHz, OMSO-d6 ):δ 0.93 ( s, 6H), 1.113 – 1.069 (m, 2H), 1.861-1.644 (m, 21H), 2.033-2.015 (d, 2H), 2.948-2.933 (d, 2H).

Example 6: Preparation of c/s-adamantane-2-spiro-3′ -8 ‘-πT(2′-amino-2’ -methyl propyl) amino! carbonyl] methyli-l ‘, 2\ 4′-U-JoXaSpJrQ [4.51 decane maleate To a solution of c/s-adamantane-2-spiro-3′-8′-[[[(2′-amino-2’-methyl propyl) amino] carbonyl] methyl]-! ‘, 2′, 4’-trioxaspiro [4.5] decane (example 5) (60 g, 0.153 moles) in ethanol (150 mL) was added a solution of maleic acid (17.3 g, 0.15 moles, 0.98 equiv. in ethanol 90 mL) and the reaction mixture was stirred for about 1 h. To this clear solution, n- heptane (720 mL) was added at room temperature in 1 h and the reaction mixture was stirred for 3 h. It was then cooled to 0 to 100C and filtered. The cake was washed with n-heptane (60 mL) and dried under vacuum at 40-450C.

Yield: 67 g, 77.4%, mp: 1490C (decomp), (M++l) 393.5, 1HNMR (300 MHz, DMSO-^ ): δ 1.05-1.11 (2H,m), 1.18 (6H,s), 1.64-1.89 (21H,m), 2.07(2H,d), 3.21 (2H,d), 6.06 (2H,d), 7.797 (2H, bs), 8.07 (IH, t).

References

  1.  Dong, Yuxiang; Wittlin, Sergio; Sriraghavan, Kamaraj; Chollet, Jacques; Charman, Susan A.; Charman, William N.; Scheurer, Christian; Urwyler, Heinrich et al. (2010). “The Structure−Activity Relationship of the Antimalarial Ozonide Arterolane (OZ277)”. Journal of Medicinal Chemistry 53 (1): 481–91. doi:10.1021/jm901473sPMID 19924861.
  2.  Blow to Ranbaxy drug research plans at LiveMint.com, Sep 21 2007
  3.  Vennerstrom, Jonathan L.; Arbe-Barnes, Sarah; Brun, Reto; Charman, Susan A.; Chiu, Francis C. K.; Chollet, Jacques; Dong, Yuxiang; Dorn, Arnulf et al. (2004). “Identification of an antimalarial synthetic trioxolane drug development candidate”. Nature 430 (7002): 900–4.doi:10.1038/nature02779PMID 15318224.
  4.  In the Pipeline: “Ozonides As Drugs: What Will They Think Of Next?”, by Derek Lowe, November 23, 2009, at Corante.com
  5.  Indian company starts Phase III trials of synthetic artemisinin, May 4 2009, at the WorldWide Antimalarial Resistance Network
  6. http://www.nature.com/nature/journal/v430/n7002/full/nature02779.html
5-27-2011
PROCESS FOR THE PREPARATION OF DISPIRO 1,2,4-TRIOXOLANE ANTIMALARIALS (OZ277)
2-13-2009
STABLE DOSAGE FORMS OF SPIRO AND DISPIRO 1,2,4-TRIOXOLANE ANTIMALARIALS
6-15-2005
Spiro and dispiro 1,2,4-trioxolane antimalarials
11-31-2004
Spiro and dispiro 1,2,4-trixolane antimalarials

ANTIMALARIALS

http://www.rsc.org/chemistryworld/2013/03/new-antimalarial-drug-class-resistance-elq-300-quinolone

Antimalarial drugsSpeeding to a new lead

http://www.nature.com/nrd/journal/v9/n11/full/nrd3301.html

Structure of NITD609; the 1R,3Sconfiguration is fundamental for its antimalarial activity

References

  1. Jump up^ Dong, Yuxiang; Wittlin, Sergio; Sriraghavan, Kamaraj; Chollet, Jacques; Charman, Susan A.; Charman, William N.; Scheurer, Christian; Urwyler, Heinrich; et al. (2010). “The Structure−Activity Relationship of the Antimalarial Ozonide Arterolane (OZ277)”. Journal of Medicinal Chemistry53 (1): 481–91. doi:10.1021/jm901473sPMID 19924861.
  2. Jump up to:a b Blow to Ranbaxy drug research plans at LiveMint.com, Sep 21 2007
  3. Jump up^ Vennerstrom, Jonathan L.; Arbe-Barnes, Sarah; Brun, Reto; Charman, Susan A.; Chiu, Francis C. K.; Chollet, Jacques; Dong, Yuxiang; Dorn, Arnulf; et al. (2004). “Identification of an antimalarial synthetic trioxolane drug development candidate”. Nature430 (7002): 900–4. doi:10.1038/nature02779PMID 15318224.
  4. Jump up^ In the Pipeline: “Ozonides As Drugs: What Will They Think Of Next?”, by Derek Lowe; published November 23, 2009; retrieved November 17, 2015; at Sciencemag.org
  5. Jump up to:a b Akshat Rathi for Chemistry World. 3 May 2012 Ranbaxy launches new anti-malarial Synriam
  6. Jump up^ India Clinical trials registry CTRI/2009/091/000531
  7. Jump up^ Toure OA et al. Efficacy and safety of fixed dose combination of arterolane maleate and piperaquine phosphate dispersible tablets in paediatric patients with acute uncomplicated Plasmodium falciparum malaria: a phase II, multicentric, open-label study. Malar J. 2015 Nov 25;14(1):469. Clinical Trial Registry India: CTRI/2009/091/000531. PMID 26608469 PMC4660726
  8. Jump up^ Staff, Business Standard. December 16, 2014 Ranbaxy receives approval for malaria drug Synriam from 7 African countries

REFERENCES

1: Valecha N, Savargaonkar D, Srivastava B, Rao BH, Tripathi SK, Gogtay N, Kochar SK, Kumar NB, Rajadhyaksha GC, Lakhani JD, Solanki BB, Jalali RK, Arora S, Roy A, Saha N, Iyer SS, Sharma P, Anvikar AR. Comparison of the safety and efficacy of fixed-dose combination of arterolane maleate and piperaquine phosphate with chloroquine in acute, uncomplicated Plasmodium vivax malaria: a phase III, multicentric, open-label study. Malar J. 2016 Jan 27;15:42. doi: 10.1186/s12936-016-1084-1. PubMed PMID: 26818020; PubMed Central PMCID: PMC4728808.

2: Saha N, Moehrle JJ, Zutshi A, Sharma P, Kaur P, Iyer SS. Safety, tolerability and pharmacokinetic profile of single and multiple oral doses of arterolane (RBx11160) maleate in healthy subjects. J Clin Pharmacol. 2014 Apr;54(4):386-93. doi: 10.1002/jcph.232. PubMed PMID: 24242999.

3: Toure OA, Rulisa S, Anvikar AR, Rao BS, Mishra P, Jalali RK, Arora S, Roy A, Saha N, Iyer SS, Sharma P, Valecha N. Efficacy and safety of fixed dose combination of arterolane maleate and piperaquine phosphate dispersible tablets in paediatric patients with acute uncomplicated Plasmodium falciparum malaria: a phase II, multicentric, open-label study. Malar J. 2015 Nov 25;14:469. doi: 10.1186/s12936-015-0982-y. PubMed PMID: 26608469; PubMed Central PMCID: PMC4660726.

4: Toure OA, Valecha N, Tshefu AK, Thompson R, Krudsood S, Gaye O, Rao BH, Sagara I, Bose TK, Mohanty S, Rao BS, Anvikar AR, Mwapasa V, Noedl H, Arora S, Roy A, Iyer SS, Sharma P, Saha N, Jalali RK; AM–PQP Study Team.. A Phase 3, Double-Blind, Randomized Study of Arterolane Maleate-Piperaquine Phosphate vs Artemether-Lumefantrine for Falciparum Malaria in Adolescent and Adult Patients in Asia and Africa. Clin Infect Dis. 2016 Apr 15;62(8):964-71. doi: 10.1093/cid/ciw029. PubMed PMID: 26908796; PubMed Central PMCID: PMC4803108.

5: Fontaine SD, Spangler B, Gut J, Lauterwasser EM, Rosenthal PJ, Renslo AR. Drug delivery to the malaria parasite using an arterolane-like scaffold. ChemMedChem. 2015 Jan;10(1):47-51. doi: 10.1002/cmdc.201402362. PubMed PMID: 25314098; PubMed Central PMCID: PMC4420023.

6: Gupta A, Singh Y, Srinivas KS, Jain G, Sreekumar VB, Semwal VP. Development and validation of a headspace gas chromatographic method for the determination of residual solvents in arterolane (RBx11160) maleate bulk drug. J Pharm Bioallied Sci. 2010 Jan;2(1):32-7. doi: 10.4103/0975-7406.62706. PubMed PMID: 21814428; PubMed Central PMCID: PMC3146089.

7: Gautam A, Ahmed T, Sharma P, Varshney B, Kothari M, Saha N, Roy A, Moehrle JJ, Paliwal J. Pharmacokinetics and pharmacodynamics of arterolane maleate following multiple oral doses in adult patients with P. falciparum malaria. J Clin Pharmacol. 2011 Nov;51(11):1519-28. doi: 10.1177/0091270010385578. PubMed PMID: 21148048.

8: Patil C, Katare S, Baig M, Doifode S. Fixed dose combination of arterolane and piperaquine: a newer prospect in antimalarial therapy. Ann Med Health Sci Res. 2014 Jul;4(4):466-71. doi: 10.4103/2141-9248.139270. Review. PubMed PMID: 25221689; PubMed Central PMCID: PMC4160665.

9: Valecha N, Looareesuwan S, Martensson A, Abdulla SM, Krudsood S, Tangpukdee N, Mohanty S, Mishra SK, Tyagi PK, Sharma SK, Moehrle J, Gautam A, Roy A, Paliwal JK, Kothari M, Saha N, Dash AP, Björkman A. Arterolane, a new synthetic trioxolane for treatment of uncomplicated Plasmodium falciparum malaria: a phase II, multicenter, randomized, dose-finding clinical trial. Clin Infect Dis. 2010 Sep 15;51(6):684-91. doi: 10.1086/655831. PubMed PMID: 20687837.

10: Lanteri CA, Chaorattanakawee S, Lon C, Saunders DL, Rutvisuttinunt W, Yingyuen K, Bathurst I, Ding XC, Tyner SD. Ex vivo activity of endoperoxide antimalarials, including artemisone and arterolane, against multidrug-resistant Plasmodium falciparum isolates from Cambodia. Antimicrob Agents Chemother. 2014 Oct;58(10):5831-40. doi: 10.1128/AAC.02462-14. PubMed PMID: 25049252; PubMed Central PMCID: PMC4187925.

11: Valecha N, Krudsood S, Tangpukdee N, Mohanty S, Sharma SK, Tyagi PK, Anvikar A, Mohanty R, Rao BS, Jha AC, Shahi B, Singh JP, Roy A, Kaur P, Kothari M, Mehta S, Gautam A, Paliwal JK, Arora S, Saha N. Arterolane maleate plus piperaquine phosphate for treatment of uncomplicated Plasmodium falciparum malaria: a comparative, multicenter, randomized clinical trial. Clin Infect Dis. 2012 Sep;55(5):663-71. doi: 10.1093/cid/cis475. PubMed PMID: 22586253.

12: Dong Y, Wittlin S, Sriraghavan K, Chollet J, Charman SA, Charman WN, Scheurer C, Urwyler H, Santo Tomas J, Snyder C, Creek DJ, Morizzi J, Koltun M, Matile H, Wang X, Padmanilayam M, Tang Y, Dorn A, Brun R, Vennerstrom JL. The structure-activity relationship of the antimalarial ozonide arterolane (OZ277). J Med Chem. 2010 Jan 14;53(1):481-91. doi: 10.1021/jm901473s. PubMed PMID: 19924861.

13: Jourdan J, Matile H, Reift E, Biehlmaier O, Dong Y, Wang X, Mäser P, Vennerstrom JL, Wittlin S. Monoclonal Antibodies That Recognize the Alkylation Signature of Antimalarial Ozonides OZ277 (Arterolane) and OZ439 (Artefenomel). ACS Infect Dis. 2016 Jan 8;2(1):54-61. PubMed PMID: 26819968; PubMed Central PMCID: PMC4718528.

14: Fügi MA, Wittlin S, Dong Y, Vennerstrom JL. Probing the antimalarial mechanism of artemisinin and OZ277 (arterolane) with nonperoxidic isosteres and nitroxyl radicals. Antimicrob Agents Chemother. 2010 Mar;54(3):1042-6. doi: 10.1128/AAC.01305-09. PubMed PMID: 20028825; PubMed Central PMCID: PMC2825978.

15: Mossallam SF, Amer EI, El-Faham MH. Efficacy of Synriam™, a new antimalarial combination of OZ277 and piperaquine, against different developmental stages of Schistosoma mansoni. Acta Trop. 2015 Mar;143:36-46. doi: 10.1016/j.actatropica.2014.12.005. PubMed PMID: 25530543.

16: Longo M, Zanoncelli S, Brughera M, Colombo P, Wittlin S, Vennerstrom JL, Moehrle J, Craft JC. Comparative embryotoxicity of different antimalarial peroxides: in vitro study using the rat whole embryo culture model (WEC). Reprod Toxicol. 2010 Dec;30(4):583-90. doi: 10.1016/j.reprotox.2010.07.011. PubMed PMID: 20708075.

17: Abiodun OO, Brun R, Wittlin S. In vitro interaction of artemisinin derivatives or the fully synthetic peroxidic anti-malarial OZ277 with thapsigargin in Plasmodium falciparum strains. Malar J. 2013 Jan 31;12:43. doi: 10.1186/1475-2875-12-43. PubMed PMID: 23368889; PubMed Central PMCID: PMC3566918.

18: White NJ. Can new treatment developments combat resistance in malaria? Expert Opin Pharmacother. 2016 Jul;17(10):1303-7. doi: 10.1080/14656566.2016.1187134. PubMed PMID: 27191998.

19: Wang X, Dong Y, Wittlin S, Charman SA, Chiu FC, Chollet J, Katneni K, Mannila J, Morizzi J, Ryan E, Scheurer C, Steuten J, Santo Tomas J, Snyder C, Vennerstrom JL. Comparative antimalarial activities and ADME profiles of ozonides (1,2,4-trioxolanes) OZ277, OZ439, and their 1,2-dioxolane, 1,2,4-trioxane, and 1,2,4,5-tetraoxane isosteres. J Med Chem. 2013 Mar 28;56(6):2547-55. doi: 10.1021/jm400004u. PubMed PMID: 23489135.

20: Marfurt J, Chalfein F, Prayoga P, Wabiser F, Wirjanata G, Sebayang B, Piera KA, Wittlin S, Haynes RK, Möhrle JJ, Anstey NM, Kenangalem E, Price RN. Comparative ex vivo activity of novel endoperoxides in multidrug-resistant plasmodium falciparum and P. vivax. Antimicrob Agents Chemother. 2012 Oct;56(10):5258-63. doi: 10.1128/AAC.00283-12. PubMed PMID: 22850522; PubMed Central PMCID: PMC3457353.

Arterolane
Arterolane.svg
Clinical data
Routes of
administration
Oral
ATC code
Identifiers
CAS Number
PubChem CID
ChemSpider
UNII
Chemical and physical data
Formula C22H36N2O4
Molar mass 392.531 g/mol
3D model (JSmol)

////////////Arteolane Maleate, OZ-277, RBx-11160, OZ 277, RBx 11160, OZ277, RBx11160, IND 2017

O=C(NCC(C)(N)C)C[C@H](CC1)CC[C@@]21OC3(OO2)[C@H]4C[C@H]5C[C@@H]3C[C@@H](C4)C5.O=C(O)/C=C\C(O)=O

Zoledronic acid

$
0
0

 Thumb

Zoledronic acid

    • CGP-42446, ZOL-446
    • ATC:M05BA08
  • Use:antineoplastic, bone resorption inhibitor, biphosphonate
  • Chemical name:[1-hydroxy-2-(1H-imidazol-1-yl)ethylidene]bis[phosphonic acid]
  • Formula:C5H10N2O7P2
  • MW:272.09 g/mol
  • CAS-RN:118072-93-8

Derivatives

Zoledronate disodium.png

Disodium salt tetrahydrate

  • Formula:C5H8N2Na2O7P2 • 4H2O
  • MW:388.11 g/mol
  • CAS-RN:165800-07-7

Trisodium salt hydrate

  • Formula:C5H7N2Na3O7P2 • 2/5H2O
  • MW:1726.21 g/mol
  • CAS-RN:165800-08-8
Zoledronic Acid
CAS Registry Number: 118072-93-8; 165800-06-6 (monohydrate)
CAS Name: [1-Hydroxy-2-(1H-imidazol-1-yl)ethylidene]bisphosphonic acid
Additional Names: 2-(imidazol-1-yl)-1-hydroxyethane-1,1-diphosphonic acid
Manufacturers’ Codes: CGP-42446
Trademarks: Zometa (Novartis)
Molecular Formula: C5H10N2O7P2
Molecular Weight: 272.09
Percent Composition: C 22.07%, H 3.70%, N 10.30%, O 41.16%, P 22.77%
Literature References: Bisphosphonate antiresorptive agent. Prepn: JPKokai 88 150291; K. A. Jaeggi, L. Wilder, US4939130(1988, 1990 both to Ciba-Geigy). Effect on bone metabolism: J. R. Green et al.,J. Bone Miner. Res.9, 745 (1994). Determn in plasma by enzyme inhibition assay: F. Risser et al.,J. Pharm. Biomed. Anal.15, 1877 (1997). Series of articles on pharmacology and clinical experience: Br. J. Clin. Pract. Suppl.87, 15-22 (1996). Clinical trial in tumor-induced hypercalcemia: J. J. Body, Cancer80, 1699 (1997); of i.v. infusion in osteoporosis: I. R. Reid et al., N. Engl. J. Med.346, 653 (2002); in bone metastases of prostate cancer: F. Saad et al., J. Natl. Cancer Inst.94, 1458 (2002). Review of pharmacology and therapeutic use: J.-J. Body, Expert Opin. Pharmacother.4, 567-580 (2003).
Properties: Crystals from water, mp 239° (dec).
Melting point: mp 239° (dec)
Zoledronate disodium.png
disodium;hydroxy-[1-hydroxy-1-[hydroxy(oxido)phosphoryl]-2-imidazol-1-ylethyl]phosphinate;tetrahydrate
cas 165800-07-7
Derivative Type: Disodium salt tetrahydrate
CAS Registry Number: 165800-07-7
Additional Names: Zoledronate disodium
Manufacturers’ Codes: CGP-42446A
Molecular Formula: C5H8N2Na2O7P2.4H2O
Molecular Weight: 388.11
Percent Composition: C 15.47%, H 4.16%, N 7.22%, Na 11.85%, O 45.35%, P 15.96%
Derivative Type: Trisodium salt hydrate
CAS Registry Number: 165800-08-8
Additional Names: Zoledronate trisodium
Manufacturers’ Codes: CGP-42446B
Molecular Formula: (C5H7N2Na3O7P2)5.2H2O
Molecular Weight: 1726.21
Percent Composition: C 17.39%, H 2.28%, N 8.11%, Na 19.98%, O 34.29%, P 17.94%
Therap-Cat: Bone resorption inhibitor.
Keywords: Antiosteoporotic; Antipagetic; Bone Resorption Inhibitor.
INGREDIENT UNII CAS INCHI KEY
Zoledronate disodium 7D7GS1SA24 165800-07-7 IEJZOPBVBXAOBH-UHFFFAOYSA-L
Zoledronate trisodium ARL915IH66 165800-08-8 Not applicable
Zoledronic acid hemipentahydrate 1K9U67HDID Not Available AZZILOGHCMYHQY-UHFFFAOYSA-N
Zoledronic acid monohydrate 6XC1PAD3KF 165800-06-6 FUXFIVRTGHOMSO-UHFFFAOYSA-N

Zoledronate (zoledronic acid, marketed by Novartis under the trade names Zometa and Reclast) is a bisphosphonate. Zometa is used to prevent skeletal fractures in patients with cancers such as multiple myeloma and prostate cancer. It can also be used to treat hypercalcemia of malignancy and can be helpful for treating pain from bone metastases.

An annual dose of Zoledronate may also prevent recurring fractures in patients with a previous hip fracture.

Zoledronate is a single 5 mg infusion for the treatment of Paget’s disease of bone. In 2007, the FDA also approved Reclast for the treatment of postmenopausal osteoporosis.

Zoledronic acid, also known as zoledronate, is a medication used to treat a number of bone diseases.[1] This include osteoporosishigh blood calcium due to cancerbone breakdown due to cancer, and Paget’s disease of bone.[1] It is given by injection into a vein.[1]

Common side effects include feverjoint painhigh blood pressure, diarrhea, and feeling tired.[1] Serious side effects may include kidney problemslow blood calcium, and osteonecrosis of the jaw.[1] Use during pregnancy may result in harm to the baby.[1] It is in the bisphosphonate family of medications.[1] It works by blocking the activity of osteoclast cells and thus decreases the breakdown of bone.[1]

Zoledronic acid was approved for medical use in the United States in 2001.[1] It is on the World Health Organization’s List of Essential Medicines, the most effective and safe medicines needed in a health system.[3] The wholesale cost in the developing world is between 5.73 USD and 26.80 USD per vial.[4] In the United Kingdom, as of 2015, a dose costs the NHS about 220 pounds.[5]

Medical uses

Bone complications of cancer

Zoledronic acid is used to prevent skeletalfractures in patients with cancers such as multiple myeloma and prostate cancer, as well as for treating osteoporosis.[6] It can also be used to treat hypercalcemia of malignancy and can be helpful for treating pain from bone metastases.[7]

It can be given at home rather than in hospital. Such use has shown safety and quality-of-life benefits in people with breast cancer and bone metastases.[8]

Osteoporosis

Zoledronic acid may be given as a 5 mg infusion once per year for treatment of osteoporosis in men and post-menopausal women at increased risk of fracture.[9]

In 2007, the U.S. Food and Drug Administration (FDA) also approved it for the treatment of postmenopausal osteoporosis.[10][11]

Paget’s disease

A single 5 mg dose of zoledronic acid is used for the treatment of Paget’s disease.[medical citation needed][12]

Contraindications

Side effects

Side effects can include fatigueanemiamuscle achesfever, and/or swelling in the feet or legs. Flu-like symptoms are common after the first infusion, although not subsequent infusions, and are thought to occur because of its potential to activate human γδ T cells(gamma/delta T cells).

Kidneys

There is a risk of severe renal impairment. Appropriate hydration is important prior to administration, as is adequate calcium and vitamin D intake prior to Aclasta therapy in patients with preexisting hypocalcaemia, and for ten days following Aclasta in patients with Paget’s disease of the bone. Monitoring for other mineral metabolism disorders and the avoidance of invasive dental procedures for those who develop osteonecrosis of the jaw is recommended.[14]

Zoledronate is rapidly processed via the kidneys; consequently its administration is not recommended for patients with reduced renal function or kidney disease.[15] Some cases of acute renal failure either requiring dialysis or having a fatal outcome following Reclast use have been reported to the U.S. Food and Drug Administration (FDA).[16] This assessment was confirmed by the European Medicines Agency (EMA), whose Committee for Medicinal Products for Human Use (CHMP) specified new contraindications for the medication on 15 December 2011, which include hypocalcaemia and severe renal impairment with a creatinine clearance of less than 35 ml/min.[17]

Bone

A rare complication that has been recently observed in cancer patients being treated with bisphosphonates is osteonecrosis of the jaw. This has mainly been seen in patients with multiple myeloma treated with zoledronate who have had dental extractions.[18]

Atypical fractures : After approving the drug on 8 July 2009, the European Medicines Agency conducted a class review of all bisphosphonates, including Zoledronate, after several cases of atypical fractures were reported.[19] In 2008, the EMA’s Pharmacovigilance Working Party (PhVWP) noted that alendronic acid was associated with an increased risk of atypical fracture of the femur that developed with low or no trauma. In April 2010, the PhVWP noted that further data from both the published literature and post-marketing reports were now available which suggested that atypical stress fractures of the femur may be a class effect. The European Medicines Agency then reviewed all case reports of stress fractures in patients treated with bisphosphonates, relevant data from the published literature, and data provided by the companies which market bisphosphonates. The Agency recommended that doctors who prescribe bisphosphonate-containing medicines should be aware that atypical fractures may occur rarely in the femur, especially after long-term use, and that doctors who are prescribing these medicines for the prevention or treatment of osteoporosis should regularly review the need for continued treatment, especially after five or more years of use.[19]

Mechanism of action

Zoledronic acid slows down bone resorption, allowing the bone-forming cells time to rebuild normal bone and allowing bone remodeling.[20]

Research

Zoledronic acid has been found to have a direct antitumor effect and to synergistically augment the effects of other antitumor agents in osteosarcoma cells.[21]

Zoledronate has shown significant benefits versus placebo over three years, with a reduced number of vertebral fractures and improved markers of bone density.[22][11] An annual dose of zoledronic acid may also prevent recurring fractures in patients with a previous hip fracture.[9]

Zoledronate also attenuates accumulation of DNA damage in mesenchymal stem cells and protects their function.[23] Given this characteristic, its potential to affect conditions arising from stem-cell dysfunction makes it a promising medicine for a range of age-related diseases[24]

With hormone therapy for breast cancer

An increase in disease-free survival (DFS) was found in the ABCSG-12 trial, in which 1,803 premenopausal women with endocrine-responsive early breast cancer received anastrozole with zoledronic acid.[25] A retrospective analysis of the AZURE trial data revealed a DFS survival advantage, particularly where estrogen had been reduced.[26]

In a meta-analysis of trials where upfront zoledronic acid was given to prevent aromatase inhibitor-associated bone loss, active cancer recurrence appeared to be reduced.[27]

As of 2010 “The results of clinical studies of adjuvant treatment on early-stage hormone-receptor-positive breast-cancer patients under hormonal treatment – especially with the bisphosphonate zoledronic acid – caused excitement because they demonstrated an additive effect on decreasing disease relapses at bone or other sites. A number of clinical and in vitro and in vivo preclinical studies, which are either ongoing or have just ended, are investigating the mechanism of action and antitumoral activity of bisphosphonates.”[28]

A 2010 review concluded that “adding zoledronic acid 4 mg intravenously every 6 months to endocrine therapy in premenopausal women with hormone receptor-positive early breast cancer … is cost-effective from a US health care system perspective”.[29]

Synthesis

PAPER

J Med Chem 2002,45(17),3721

https://pubs.acs.org/doi/10.1021/jm020819i

Highly Potent Geminal Bisphosphonates. From Pamidronate Disodium (Aredia) to Zoledronic Acid (Zometa)

Novartis Pharma Research, Arthritis and Bone Metabolism Therapeutic Area, CH-4002 Basel, Switzerland
J. Med. Chem.200245 (17), pp 3721–3738
DOI: 10.1021/jm020819i
Abstract Image

Bisphosphonates (BPs) are pyrophosphate analogues in which the oxygen in P−O−P has been replaced by a carbon, resulting in a metabolically stable P−C−P structure. Pamidronate (1b, Novartis), a second-generation BP, was the starting point for extensive SAR studies. Small changes of the structure of pamidronate lead to marked improvements of the inhibition of osteoclastic resorption potency. Alendronate (1c, MSD), with an extra methylene group in the N-alkyl chain, and olpadronate (1h, Gador), the N,N-dimethyl analogue, are about 10 times more potent than pamidronate. Extending one of the N-methyl groups of olpadronate to a pentyl substituent leads to ibandronate (1k, Roche, Boehringer-Mannheim), which is the most potent close analogue of pamidronate. Even slightly better antiresorptive potency is achieved with derivatives having a phenyl group linked via a short aliphatic tether of three to four atoms to nitrogen, the second substituent being preferentially a methyl group (e.g., 4g4j5d, or 5r). The most potent BPs are found in the series containing a heteroaromatic moiety (with at least one nitrogen atom), which is linked via a single methylene group to the geminal bisphosphonate unit. Zoledronic acid (6i), the most potent derivative, has an ED50 of 0.07 mg/kg in the TPTX in vivo assay after sc administration. It not only shows by far the highest therapeutic ratio when comparing resorption inhibition with undesired inhibition of bone mineralization but also exhibits superior renal tolerability. Zoledronic acid (6i) has thus been selected for clinical development under the registered trade name Zometa. The results of the clinical trials indicate that low doses are both efficacious and safe for the treatment of tumor-induced hypercalcemia, Paget’s disease of bone, osteolytic metastases, and postmenopausal osteoporosis.

SYN 1

AU 8781453; EP 0275821; JP 1988150291; US 4939130

Zoledronate sodium can be prepared by reaction of 2-(1-imidazolyl)acetic acid hydrochloride (I) with PCl3, with optional presence of phosphoric acid, in refluxing chlorobenzene, followed by hydrolysis with refluxing 9N hydrochloric acid and final formation of the sodium salt by treatment with aqueous NaOH.

SYN

PAPER

https://www.sciencedirect.com/science/article/pii/S0969804311006385

Image result for zoledronic acid synthesis

Clip

https://link.springer.com/article/10.1007/s11094-015-1205-0

A One-Pot and Efficient Synthesis of Zoledronic Acid Starting from Tert-butyl Imidazol-1-yl Acetate

A one-pot synthesis of zoledronic acid in high yield is described. The procedure involves a non-aqueous ester cleavage of the tert-butyl imidazol-1-yl acetate under dry conditions in the presence of methanesulfonic acid as solubilizer and chlorobenzene as solvent to afford in situthe corresponding imidazolium methanesulfonate salt which yields zoledronic acid upon reaction with phosphoric acid and phosphorus oxychloride. A possible chemical mechanism for the synthesis of this acid is described.

Image result for zoledronic acid synthesis

Paper

https://www.beilstein-journals.org/bjoc/articles/4/42

str1 str2

Preparation of imidazol-1-yl-acetic acid tert-butyl ester (2)

To a solution of imidazole (10.0 g, 0.15 mol) in ethyl acetate (160 mL) was added powdered K2CO3 (29.0 g, 0.21 mol) followed by tert-butyl chloroacetate (25.7 mL, 0.18 mol) at room temperature and the mixture was refluxed for 10.0 h. After completion of the reaction as indicated by TLC (10% MeOH/CHCl3, I2 active), the reaction mass was quenched with cold water (80 mL) and the ethyl acetate layer was separated. The aqueous layer was extracted with ethyl acetate (2 × 80 mL) and the combined ethyl acetate layers were washed with brine, dried with anhydrous sodium sulfate and then concentrated under vacuum. The resulting solid was stirred with hexane (50 mL) at RT, filtered and washed with hexane (2 × 20 mL) to afford the title compound as an off-white solid (20.0 g, 75%). mp: 111.3–113.2 °C (Lit [10]: 111–113 °C). IR (cm−1): 3458, 3132, 3115, 2999, 2981, 2884, 1740, 1508, 1380, 1288, 1236, 1154, 1079, 908, 855, 819, 745, 662, 583; 1H NMR (300 MHz, CDCl3) δ 1.47 (s, 9H), 4.58 (s, 2H), 6.94 (s, 1H), 7.09 (s, 1H), 7.49 (s, 1H); 13C NMR (75 MHz, CDCl3) δ 27.7, 48.6, 82.9, 119.8, 129.2, 137.7, 166.3; MS (m/z) 183.0 [M+1, 100%], 127.0.

Preparation of imidazol-1-yl-acetic acid hydrochloride (6)

To a solution of imidazol-1-yl-acetic acid tert-butyl ester (2) (10.0 g, 0.05 mol) in dichloromethane (100 mL) was added titanium tetrachloride (8.0 mL, 0.07 mol) dropwise slowly at −15 to −10 °C over 1 h and the mixture was stirred at −5 to 0 °C for 2 h. Isopropyl alcohol (25 mL) was added at 0 to −10 °C over 0.5 h and the reaction mass was stirred at room temperature for 0.5 h. Additional isopropyl alcohol (125 mL) was added dropwise at room temperature over 0.5 h and the mixture was stirred for 1 h. Dichloromethane was distilled out under a low vacuum and the resulting crystalline solid precipitated was filtered to afford the title compound as an off-white crystalline solid (7.4 g, 83%). mp 200.3–202.3 °C; IR (cm−1): 3175, 3125, 3064, 2945, 2869, 2524, 2510, 1732, 1581, 1547, 1403, 1223, 1193, 1081, 780, 650; 1H NMR (300 MHz, D2O + 3-(trimethylsilyl)propionic acid sodium salt) δ 5.1 (s, 3H, -CH2– + HCl), 7.5 (br s, 2H), 8.7 (s, 1H); 13C NMR (75 MHz, D2O + 3-(trimethylsilyl)propionic acid sodium salt) 52.7, 122.4, 125.9, 138.8, 172.8; MS (m/z) 127.0 [M+1, 100%]; HCl-content: found 21.8% (along with 3.25% moisture), calcd 22.43% for C5H6N2O2·HCl.

Preparation of zoledronic acid (7)

To a suspension of imidazol-1-yl-acetic acid hydrochloride (6) (7.0 g, 0.043 mol) and phosphorous acid (9.5 g, 0.116 mol) in chlorobenzene (50 mL) was added phosphorous oxychloride (9.6 ml, 0.103 mol) at 80–85 °C over a period of 2 h then heated to 90–95 °C for 2.5 h. The reaction mass was cooled to 60–65 °C and water (100 mL) was added at the same temperature. The aqueous layer was separated, collected and refluxed for 18 h. It was then cooled to room temperature and diluted with methanol (140 mL). The mixture was cooled to 0–5 °C and stirred for 3 h. The precipitated solid was filtered, washed with cold water followed by methanol and then dried under vacuum at 60 °C for 12 h to afford the title compound (6.6 g, 57% yield) as a white solid; mp 237–239 °C (lit [1] 239 °C with decomposition).

PATENT

https://patents.google.com/patent/CN104610357A/en

Sodium Zoledronic (Zoledronate sodium, I), chemical name [1-yl light -2- (lH- imidazol-1-yl) ethylidene] bisphosphonic acid monosodium salt monohydrate, is by the Novartis (Novartis) developed imidazole heterocyclic bisphosphonates, belongs to the third generation of bisphosphonates bisphosphonate drugs, in October 2000, first marketed in Canada. Subsequently approved in the European Union, the United States more than 80 countries or regions, trade name Zometa, for the treatment of hypercalcemia of malignancy (HCM) and multiple myeloma and bone metastases of solid tumors. The drug is effective in treating cancer caused by HCM, advanced bone metastases and Paget’s disease, reduce the incidence of skeletal related events, relieve symptoms and improve quality of life, is also expected to be used to treat osteoporosis. Compared with other similar drugs, high efficacy, dosage, ease of administration, better security, etc., is currently the only FDA-approved for metastatic bone tumor effective bisphosphonate drugs.Currently bisphosphonate drugs in our country is still in the initial stages of clinical applications, but in recent years has made rapid progress, broad market prospect.

[0003] The prior art synthesis reaction conditions zoledronate sodium harsh, toxicity and use methanol, chloroform and chlorobenzene, easily exceeding the amount of residual organic solvents, low yield, low product purity, contamination environment, does not meet the medical criteria, is not conducive to industrial production. Environmental pollution has attracted increasing attention around the world today, the development of new green efficient synthesis of a pharmaceutical drug synthesis is an important issue facing the Institute. In recent years, room temperature ionic liquids as a reaction medium is environmentally friendly, has been widely used in a variety of organic synthesis reactions. Compared with traditional organic solvents, ionic liquids have very low vapor pressure, non-flammable, good thermal stability, both as a reaction medium underway catalysis, can be recycled and many other advantages.

Image result for zoledronic acid synthesis

Example 1 of zoledronic alendronate

Figure CN104610357AD00061

(1) Synthesis of imidazol-1-yl acetate were added successively imidazole (13.62g, 0.2mol) and [bmim] BF4 (IOOmL) a three-necked flask, heated with stirring warmed to 60 ° C, incubated under reflux was slowly added dropwise chlorination ethyl acetate (24. 51g, 0. 2mol), dropwise addition time is about 2h, dropwise, with stirring maintained at reflux for 16 h, the reaction monitored by TLC showed no starting material end point, completion of the reaction, cooled to room temperature, to give imidazole -1 – ethyl crude, about 24g, crude without purification, was used directly in the next reaction.

[0014] (2) Synthesis of imidazol-1-yl acetate hydrochloride A solution of 24g 1-yl imidazole prepared above was added crude ethyl necked flask, concentrated hydrochloric acid (34 mL), exotherm to 85 ° C, warmed to reflux heating was continued, the reaction was stirred at reflux for 10H, the reaction was completed, the solvent was evaporated under reduced pressure, 20ml of absolute ethanol was added to the residue, vigorously stirred for 2h, filtered off with suction, the filter cake finally at 80 ° C blast pressure and dried to give a white solid imidazol-1-yl acetate hydrochloride about 25. 65g, 79.4% overall yield.

[0015] (3) Synthesis of zoledronic acid monohydrate were added imidazol-1-yl acetate hydrochloride (17. 26g, 0. 137mol) a three-necked flask, in an ionic liquid with stirring – n-butyl-3- methylimidazolium tetrafluoroborate [bmim] BF4 (40mL) and concentration of 85% phosphoric acid solution (16mL), heating to 60 ° C was added dropwise phosphorus trichloride (30mL) , about 4h dropwise, reaction was continued under reflux for 4h at 65 ° C, the reaction was complete, cooled to 40 ° C, filtered off with suction, the filter cake was added to a molar concentration in 80mL 9mol / L hydrochloric acid, heated with stirring state the reaction was refluxed for 6h, the reaction was completed, filtered hot, the filter cake was added to a molar concentration in 80mL 9mol / L hydrochloric acid, the above-described operation is repeated to continue the combined filtrate was evaporated to dryness under reduced pressure to give a yellow oily residue was slowly added to the residue volume ratio of 1: 1 acetone – ethanol mixture 240 mL, was stirred, and the precipitated solid was 15min, filtered off with suction, the filter cake was recrystallized in 30mL of deionized water, suction filtered to give a white solid that is zoledronic acid monohydrate , about 35. 8g, yield 90.1%, determined by HPLC, purity> 98.5%.

After [0016] (4) Synthesis of zoledronic sodium phosphinate obtained above azole zoledronic acid monohydrate (46.4g, 0. 16mol) washed with water (450 mL of) was dissolved, was added sodium hydroxide (5. 6g, 0. IOmol), were refluxed for 30min, cooling and crystallization, filtration, to obtain a crude product zoledronate sodium, crude mother liquor was concentrated and then half with distilled water (410 mL), isopropanol (60 mL), heated to dissolve the combined, activated carbon bleaching, charcoal filtered off, cooling and crystallization, filtration, washed with water, dried at 40-60 ° C to about crystallization water containing one to give zoledronate sodium (42. 4g, 85%), total yield of more than 60% by HPLC assay, purity 99. 8%, mp239 ° C. IR: 711011 ^ 671 (^ 1 is the stretching vibration peak of the PC, 1643〇 ^ 1 = 0 (: stretching vibration peak, 3011〇 ^ 1 = (: – stretching vibration peak 11 ^ 1 is CN 1406〇 the stretching vibration, 1643CHT1 is C = N stretching vibration peak of 3447 (3485 ^^ (^ 1 is the stretching vibration peak of OH, 1459CHT1 symmetrical bending vibration of CH, 2830CHT1 stretching vibration of CH, 1324CHT1 is P = O the stretching vibration, 1094CHT1 stretching vibration peak of .1HNMR PO (400MHz, D20), S: 8.68 (lH, s), 7.48 (lH, s), 7.34 (lH, s), 4.67 (2H, t).

Effects [0017] Example 2 was added dropwise phosphorus trichloride fixed time on the yield other conditions remain unchanged, only the changes of phosphorus trichloride dropwise addition, dropping zoledronic Table 1 Effect of Sodium yield Experimental results show that excessive phosphorus trichloride was added dropwise, and instantly generate a large amount of gas, the reaction is very intense, the liquid splashing, a rapid rise in temperature, resulting in the low yield, if slowly added dropwise, the reaction rate is too slow, consumption too long, and therefore is the best 4h dropping time.

Figure CN104610357AD00071

Zoledronic fixed effect of sodium yield other conditions remain unchanged, imidazol-1-yl acetic acid hydrochloride with phosphorus trichloride and phosphoric acid condensation reaction temperature is zoledronic acid monohydrate embodiment the reaction temperature Example 3 Effect yield (Table 2). The results show that, with increasing temperature, increasing the yield, but at higher temperatures to reflux, shows a decreasing trend in yield, due to decomposition sake phosphorus trichloride, resulting in reduction reaction. Further, when the temperature is too high, solvent evaporation and solvent leakage losses will increase, the reflux temperature is low, the reaction rate is slow, the reaction is insufficient, therefore the yield is low, and therefore the optimum reaction temperature is about 65 ° C.

Figure CN104610357AD00072

Example 4

Figure CN104610357AD00081

Effect of the ionic liquid frequency reuse sodium zoledronic yield of the reaction medium can be recovered and reused important concern is “green chemistry” used in the present embodiment examines the sodium ionic liquid used in the synthesis of zoledronic repeated use, the experiment results shown in Table 3. Seen from Table 3, the ionic liquid after 5 subsequent to use, product yield began to decrease, the ionic liquid may be recovered and reused effectively, and repeated five times using good performance, and therefore is an ionic liquid in this reaction green solvents may be recycled.

Figure CN104610357AD00082

Example 5 different ionic liquids zoledronic same impact conditions were examined yield sodium 1-butyl-3-methylimidazolium tetrafluoroborate ionic liquids ([bmim] BF4), N- ethyl pyridinium tetrafluoroborate ([EPy] BF4), l- butyl-3-methylimidazolium hexafluorophosphate ([bmim] PF6), 1- hydroxyethyl-2,3-dimethyl imidazolium chloride (LOH), 1- propyl-3-carbonitrile methylimidazolium chloride (the LCN) and 1-carboxyethyl-3-methyl imidazolium chloride (LOOH) Effects of sodium zoledronic yield the results are shown in Table 4, the test results show little effect on the synthesis of ionic liquids yield.

Figure CN104610357AD00083

Clip

Mar 5, 2013 –

Dr. Reddy’s Laboratories  announced today that it has launched Zoledronic Acid Injection (4 mg/5 mL), a bioequivalent generic version of Zometa® (zoledronic acid) 4 mg/5 mL Injection in the US market on March 4, 2013, following the approval by the United States Food & Drug Administration (USFDA) of Dr. Reddy’s ANDA for Zoledronic Acid Injection (4 mg/5 mL).

Dr. Reddy’s Zoledronic Acid Injection 4 mg/5mL is available in a single use vial of concentrate.

Zoledronic acid (INN) or zoledronate (marketed by Novartis under the trade names ZometaZomeraAclasta and Reclast) is a bisphosphonate. Zometa is used to prevent skeletal fractures in patients with cancers such as multiple myeloma and prostate cancer, as well as for treating osteoporosis.It can also be used to treat hypercalcemia of malignancy and can be helpful for treating pain from bone metastases.

An annual dose of zoledronic acid may also prevent recurring fractures in patients with a previous hip fracture.

Reclast is a single 5 mg infusion for the treatment of Paget’s disease of bone. In 2007, the U.S. Food and Drug Administration (FDA) also approved Reclast for the treatment of postmenopausal osteoporosis.

About Dr. Reddy’s Laboratories Ltd.

Dr. Reddy’s Laboratories Ltd. (NYSE: RDY) is an integrated global pharmaceutical company, committed to providing affordable and innovative medicines for healthier lives. Through its three businesses – Pharmaceutical Services and Active Ingredients, Global Generics and Proprietary Products – Dr. Reddy’s offers a portfolio of products and services including APIs, custom pharmaceutical services, generics, biosimilars, differentiated formulations and NCEs. Therapeutic focus is on gastro-intestinal, cardiovascular, diabetology, oncology, pain management, anti-infective and pediatrics. Major markets include India, USA, Russia and CIS, Germany, UK, Venezuela, S. Africa, Romania, and New Zealand. For more information, log on to: http://www.drreddys.com

Zometa® is a registered trademark of Novartis AG

References

    • US 4 939 130 (Ciba-Geigy; 3.7.1990; CH-prior. 21.11.1986).
  • transdermal formulation:

    • EP 407 344 (Ciba-Geigy; appl. 28.6.1990; CH-prior. 7.7.1989).
  • treatment of angiogenesis:

    • WO 2 000 071 104 (Novartis AG; appl. 19.5.2000; GB-prior. 21.5.1999).

PATENT

ApplicationPriority dateFiling dateTitle
CN 2015100011672015-01-052015-01-05Preparation method for sodium zoledronic acid

ApplicationFiling dateTitle
CN 2015100011672015-01-05Preparation method for sodium zoledronic acid

References

  1. Jump up to:a b c d e f g h i j k “Zoledronic Acid”. The American Society of Health-System Pharmacists. Retrieved 8 December 2017.
  2. Jump up^ Drugs.com International trade names for zoledronic acid Page accessed Jan 14, 2015
  3. Jump up^ “WHO Model List of Essential Medicines (20th List)” (PDF). World Health Organization. March 2017. Retrieved 29 June 2017.
  4. Jump up^ “Single Drug Information”International Medical Products Price Guide. Retrieved 9 December 2017.
  5. Jump up^ British national formulary : BNF 69 (69 ed.). British Medical Association. 2015. p. 528. ISBN 9780857111562.
  6. Jump up^ National Prescribing Service (2009). “Zoledronic Acid for Osteoporosis”. Medicines Update, Available at “Archived copy”. Archived from the original on April 23, 2010. Retrieved January 20, 2010.
  7. Jump up^ http://www.health.gov.il/units/pharmacy/trufot/alonim/533.pdf Zomera prescribing information
  8. Jump up^ Wardley, A; Davidson, N; Barrett-Lee, P; et al. (May 2005). “Zoledronic acid significantly improves pain scores and quality of life in breast cancer patients with bone metastases: a randomised, crossover study of community vs hospital bisphosphonate administration”Br. J. Cancer92 (10): 1869–76. doi:10.1038/sj.bjc.6602551PMC 2361764Freely accessiblePMID 15870721.
  9. Jump up to:a b Lyles K, et al. (2007). “Zoledronic Acid and Clinical Fractures and Mortality after Hip Fracture”N. Engl. J. Med357 (18): 1799–809. doi:10.1056/NEJMoa074941PMC 2324066Freely accessiblePMID 17878149.
  10. Jump up^ “Biotech PRESS RELEASE: Novartis’s Reclast Receives FDA Approval FOR Women With Postmenopausal Osteoporosis”, FierceBiotech, A Division of Questex A FierceMarkets Publication Aug 20, 2007. Retrieved 2018-03-27
  11. Jump up to:a b Black; et al. (2007). “Once-Yearly Zoledronic Acid for Treatment of Postmenopausal Osteoporosis”NEJM356 (18): 1809–1822. doi:10.1056/nejmoa067312PMID 17476007.
  12. Jump up^ “Paget’s Disease of Bone”http://www.rheumatology.org. Retrieved 2015-07-09.
  13. Jump up^ Vondracek, S. F. (2010). “Managing osteoporosis in postmenopausal women”. American Journal of Health-System Pharmacy67 (7 Suppl 3): S9–19. doi:10.2146/ajhp100076PMID 20332498.
  14. Jump up^ http://www.nps.org.au/__data/assets/pdf_file/0006/60945/nvcaclin.pdf
  15. Jump up^ “Zometa 4mg/5ml Concentrate for Solution for Infusion”medicines.org.uk.
  16. Jump up^ “FDA Alert: Reclast (zoledronic acid): Drug Safety Communication – New Contraindication and Updated Warning on Kidney Impairment”drugs.com.
  17. Jump up^ “European Medicines Agency – Human medicines”europa.eu.
  18. Jump up^ Durie BG, Katz M, Crowley J (2005). “Osteonecrosis of the jaw and bisphosphonates”. N. Engl. J. Med353 (1): 99–102; discussion 99–102. doi:10.1056/NEJM200507073530120PMID 16000365.
  19. Jump up to:a b “European Medicines Agency – Human medicines”europa.eu.
  20. Jump up^ Aclasta label- Australia
  21. Jump up^ Koto K, Murata H, Kimura S, et al. (July 2010). “Zoledronic acid inhibits proliferation of human fibrosarcoma cells with induction of apoptosis, and shows combined effects with other anticancer agents”. Oncol. Rep24 (1): 233–9. doi:10.3892/or_00000851PMID 20514467.
  22. Jump up^ Reid IR, Brown JP, Burckhardt P, Horowitz Z, Richardson P, Trechsel U, Widmer A, Devogelaer JP, Kaufman JM, Jaeger P, Body JJ, Brandi ML, Broell J, Di Micco R, Genazzani AR, Felsenberg D, Happ J, Hooper MJ, Ittner J, Leb G, Mallmin H, Murray T, Ortolani S, Rubinacci A, Saaf M, Samsioe G, Verbruggen L, Meunier PJ (2002). “Intravenous zoledronic acid in postmenopausal women with low bone mineral density”. N. Engl. J. Med346 (9): 653–61. doi:10.1056/NEJMoa011807PMID 11870242.
  23. Jump up^ Juhi Misra, Sindhu T. Mohanty, Sanjeev Madan, James A. Fernandes, F. Hal Ebetino, R. Graham, G. Russell, Ilaria Bellantuono. (December 2015). Zoledronate attenuates accumulation of DNA damage in mesenchymal stem cells and protects their function. Stem Cells, doi:10.1002/stem.2255
  24. Jump up^ “Bone drug protects stem cells from aging.” ScienceDaily. 17 December 2015
  25. Jump up^ PMID 19213681 Gnant, Mlineritsch. Endocrine therapy plus zoledronic acid in premenopausal breast cancer. N Engl J Med 2009; 360:679-691 February 12, 2009 Full Free Text [1]
  26. Jump up^ Coleman RE, Winter MC, Cameron D, et al. (March 2010). “The effects of adding zoledronic acid to neoadjuvant chemotherapy on tumour response: exploratory evidence for direct anti-tumour activity in breast cancer”Br. J. Cancer102 (7): 1099–105. doi:10.1038/sj.bjc.6605604PMC 2853093Freely accessiblePMID 20234364.
  27. Jump up^ Brufsky A, Bundred N, Coleman R, et al. (May 2008). “Integrated analysis of zoledronic acid for prevention of aromatase inhibitor-associated bone loss in postmenopausal women with early breast cancer receiving adjuvant letrozole”. Oncologist13 (5): 503–14. doi:10.1634/theoncologist.2007-0206PMID 18515735.
  28. Jump up^ Tonyali O, Arslan C, Altundag K (November 2010). “The role of zoledronic acid in the adjuvant treatment of breast cancer: current perspectives”. Expert Opin Pharmacother11(16): 2715–25. doi:10.1517/14656566.2010.523699PMID 20977404.
  29. Jump up^ Delea TE, Taneja C, Sofrygin O, Kaura S, Gnant M (August 2010). “Cost-effectiveness of zoledronic acid plus endocrine therapy in premenopausal women with hormone-responsive early breast cancer”. Clin. Breast Cancer10 (4): 267–74. doi:10.3816/CBC.2010.n.034PMID 20705558.
Zoledronic acid
Zoledronic acid.svg
Zoledronic-acid-from-xtal-2003-3D-balls.png
Clinical data
Trade names Reclast, Zometa, others[2]
AHFS/Drugs.com Monograph
MedlinePlus a605023
License data
Pregnancy
category
Routes of
administration
Intravenous
Drug class Bisphosphonate[1]
ATC code
Legal status
Legal status
Pharmacokinetic data
Protein binding 22%
Metabolism Nil
Elimination half-life 146 hours
Excretion Kidney (partial)
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
PDB ligand
Chemical and physical data
Formula C5H10N2O7P2
Molar mass 272.09 g/mol
3D model (JSmol)

Judith Aronhime, Revital Lifshitz-Liron, “Zoledronic acid crystal forms, zoledronate sodium salt crystal forms, amorphous zoledronate sodium salt, and processes for their preparation.” U.S. Patent US20050054616, issued March 10, 2005., US20050054616

/////////////////disodium zoledronate tetrahydrate, zoledronic acid, ZOMETA, CGP-42446, CGP-42446A

OC(CN1C=CN=C1)(P(O)(O)=O)P(O)(O)=O

Diazoxide choline

$
0
0

Diazoxide choline.png

Image result for Diazoxide choline

Diazoxide choline,

RN: 1098065-76-9
UNII: 2U8NRZ7P8L

Diazoxide choline; UNII-2U8NRZ7P8L; 2U8NRZ7P8L; YLLWQNAEYILHLV-UHFFFAOYSA-N

Molecular Formula: C13H20ClN3O3S
Molecular Weight: 333.831 g/mol

Ethanaminium, 2-hydroxy-N,N,N-trimethyl-, compd. with 7-chloro-3-methyl-2H-1,2,4-benzothiadiazine dioxide (1:1)

7-Chloro-3-methyl-2H-1,2,4-benzothiadiazine dioxide compd. with 2-hydroxy-N,N,N-trimethylethanaminium (1:1)

7-chloro-3-methyl-1$l^{6},2,4-benzothiadiazin-2-ide 1,1-dioxide;2-hydroxyethyl(trimethyl)azanium

DiazoxideDiazoxide

CAS: 364-98-7 FREE FORM

2H-1,2,4-Benzothiadiazine, 7-chloro-3-methyl-, 1,1-dioxide

  • 4H-1,2,4-Benzothiadiazine, 7-chloro-3-methyl-, 1,1-dioxide (7CI)
  • 3-Methyl-7-chloro-1,2,4-benzothiadiazine 1,1-dioxide
  • 7-Chloro-3-methyl-2H-1,2,4-benzothiadiazine 1,1-dioxide
  • Diazoxide
  • Dizoxide
  • Eudemine injection
  • Hyperstat
  • Hypertonalum
  • Mutabase
  • NSC 64198
  • NSC 76130
  • Proglicem
  • Proglycem
  • SRG 95213
  • Sch 6783
Diazoxide
CAS Registry Number: 364-98-7
CAS Name: 7-Chloro-3-methyl-2H-1,2,4-benzothiadiazine 1,1-dioxide
Additional Names: 3-methyl-7-chloro-1,2,4-benzothiadiazine 1,1-dioxide
Manufacturers’ Codes: SRG-95213
Trademarks: Eudemine Injection (Schering); Proglicem (Essex); Hyperstat (Schering); Hypertonalum (Essex); Mutabase (Schering); Proglycem (Schering)
Molecular Formula: C8H7ClN2O2S
Molecular Weight: 230.67
Percent Composition: C 41.66%, H 3.06%, Cl 15.37%, N 12.14%, O 13.87%, S 13.90%
Literature References: Hypotensive agent that inhibits secretion of insulin from pancreatic beta cells. Prepn: A. A. Rubin et al.,Science 133, 2067 (1961); J. G. Topliss et al., US 2986573eidem, US 3345365 (1961, 1967 both to Schering); Raffa, Monzani, Farmaco Ed. Sci. 17, 244 (1962). Crystal and molecular structure: G. Bandoli, M. Nicolini, J. Cryst. Mol. Struct. 7, 229 (1978). Review of effect on insulin secretion: Nutr. Rev. 30, 194-198 (1972); of pharmacology and efficacy in hypertension: J. Koch-Weser, N. Engl. J. Med. 294, 1271-1274 (1976).
Properties: Crystals from dil alc, mp 330-331°. uv max (methanol): 268 nm (e 11300). Sol in alcohol and alkaline solns. Insol in water.
Melting point: mp 330-331°
Absorption maximum: uv max (methanol): 268 nm (e 11300)
Therap-Cat: Antihypoglycemic; antihypertensive.
Keywords: Antihypertensive; Thiazides and Analogs; Antihypoglycemic.

Diazoxide (INN; brand name Proglycem[1]) is a potassium channel activator, which causes local relaxation in smooth muscle by increasing membrane permeability to potassium ions. This switches off voltage-gated calcium ion channels, preventing calcium flux across the sarcolemma and activation of the contractile apparatus.

In the United States, this agent is only available in the oral form and is typically given in hospital settings.[2]

Medical uses

Diazoxide is used as a vasodilator in the treatment of acute hypertension or malignant hypertension.[3]

Diazoxide also inhibits the secretion of insulin by opening ATP-sensitive potassium channel of beta cells of the pancreas, thus it is used to counter hypoglycemia in disease states such as insulinoma (a tumor producing insulin)[4] or congenital hyperinsulinism.

Diazoxide acts as a positive allosteric modulator of the AMPA and kainate receptors, suggesting potential application as a cognitive enhancer.[5]

Side effects

The Food and Drug Administration published a Safety Announcement in July 2015 highlighting the potential for development of pulmonary hypertension in newborns and infants treated with this drug.[2]Diazoxide interferes with insulin release through its action on potassium channels.[6] Diazoxide is one of the most potent openers of the K+ ATP channels present on the insulin producing beta cells of the pancreas. Opening these channels leads to hyperpolarization of cell membrane, a decrease in calcium influx, and a subsequently reduced release of insulin.[7] This mechanism of action is the mirror opposite of that of sulfonylureas, a class of medications used to increase insulin release in Type 2 Diabetics. Therefore, this medicine is not given to non-insulin dependent diabetic patients.

SYN

Medicinal Chemistry Research, 12(9), 457-470; 2004

PATENT

WO 2009006483

https://patents.google.com/patent/WO2009006483A1/enIt

PATENT

US 20120238554

PATENT

WO 2013130411

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2013130411&recNum=95&docAn=US2013027676&queryString=Telmisartan%20OR%20Hydrochlorothiazide&maxRec=4800

able 16. Characterization of Forms A and B of Diazoxide Choline Salt In

Screening Study

Experiment Form A Form B

*Maj or peaks (2-Θ):

Form A (9.8, 10.5, 14.9, 17.8, 17.9, 18.5, 19.5, 22.1, 22.6, 26.2, 29.6, 31.2);

Form B (8.9, 10.3, 12.0, 18.3, 20.6, 24.1, 24.5, 26.3, 27.1, 28.9).

** Unique FTIR (ATR) absorbances (cm 1):

Form A (2926, 2654, 1592, 1449, 1248);

Form B (3256, 2174, 2890, 1605, 1463, 1235).

6.1.5.1. Solubility Screen in organic solvents.

[00725] Diazoxide choline, prepared in MEK using choline hydroxide as 50 wt % solution in water (see above) displayed some solubility in the following solvents:

acetonitrile, acetone, ethanol, IPA, MEK, DMF, and methanol. These solvents were chosen due to differences in functionality, polarity, and boiling points and their ability to dissolve diazoxide. Other solvents which showed poor ability to dissolve salts were used as antisolvents and in slurry experiments where some solubility was observed: dioxane, MTBE, EtOAc, IP Ac, THF, water, cyclohexane, heptane, CH2C12, and toluene.

[00726] Solvents for crystallizations during screening were chosen based on the solubility screen summarized in Table 17. Crystallizations of diazoxide choline from all conditions afforded a total of two forms, A and B. Forms A and B were found to be anhydrous polymorphs of diazoxide choline. Form B was observed to be generated from most solvents used. It was difficult to isolate pure Form A on large scales (>50 mg) as conditions observed to produce Form A on a smaller scale (approximately 50 mg or less) were found to result in Form B or mixtures of both forms on larger scales. Based on room-temperature slurry experiments, anhydrous Form B was found to be the most thermodynamically stable form in this study. Form A readily converted to Form B in all slurry solvents utilized.

Table 17. Solubility Screen for Diazoxide Choline Salt

Solvent Cmpd Solvent Cone. Temp. Soluble

(mg) (mL) (mg/niL) (°C)

CH2CI2 1.3 5.00 0.26 55 Partially

Toluene 1.4 5.00 0.28 55 No

.1.5.2. Single-Solvent Crystallizations

[00727] Fast cooling procedure: Diazoxide (approximately 20 mg) was weighed out into vials and enough solvent (starting with 0.25 mL) was added until the material completely dissolved at elevated temperature. After hot filtration the vials were placed in a refrigerator (4 °C) for 16 hours. After the cooling-process the samples were observed for precipitates which were isolated by filtration. Vials not demonstrating precipitates were evaporated down to dryness using a gentle stream of nitrogen. All solids were dried in vacuo at ambient temperature and 30 in. Hg.

[00728] Slow cooling procedure: Diazoxide (approximately 30 mg of choline salt) was weighed out into vials and enough solvent was added until the material went into solution at elevated temperature. After hot filtration the vials were then slowly cooled to room temperature at the rate of 20 °C/h and stirred at room temperature for 1-2 hours. All solids were dried in vacuo at ambient temperature and 30 in. Hg.

[00729] Based on the initial solubility study, seven solvents were selected for the fast-cooling crystallization: acetonitrile, acetone, ethanol, IPA, MEK, DMF, and methanol. Table 18 shows a list of the solvents that were used and the amount of solvent needed to dissolve the material. After the cooling-process precipitates were noticed in samples # 2, 3, 5, and 6, the solids were isolated by filtration. The other samples (# 1, 4, and 7) were evaporated down to dryness using a gentle stream of nitrogen. The diazoxide choline salts were found to be consistent with Form A by XRPD analysis for all solids with the exception of sample #2 (consistent with the freeform) and sample #5 (consistent with Form B with preferred orientation observed).

Table 18. Single- Solvent Crystallization of Diazoxide Choline Salt Using Fast- Cooling Procedure

[00730] In accordance with the data obtained from fast-cooling experiments, four solvents which showed precipitation of solids were chosen for the slow-cooling experiments: MeOH, EtOH, MeCN, and IPA (Table 19). All obtained analyzable solids of the choline salt were found to be consistent with Form B by XRPD with the exception of Entry #1 which was consistent with diazoxide freeform and Entry #2 which was not analyzable. Mother liquor of Entry #2 was concentrated to dryness and the residual solids were analyzed by XRPD and found to be Form B material. As a result of obtaining freeform material from the single- solvent crystallizations in methanol, three more alcohols were tested for the single- solvent crystallizations using fast- and slow-cooling procedures. Tables 20 and 21 provide a list of the solvents that were used and the amount of solvent needed to dissolve the material. XRPD patterns of the fast-cooling procedure showed freeform of diazoxide from isobutanol, Form B from isoamyl alcohol, and Form A from tert-amyl alcohol compared to the slow-cooling procedure, which afforded Form B material from all three solvents.

Table 19. Single-Solvent Crystallization of Diazoxide Choline Salt Using Slow- Cooling Procedure

Table 20. Single- Solvent Crystallization of Diazoxide Choline Salt Using Fast- Cooling Procedure

Table 21. Single-Solvent Crystallization of Diazoxide Choline Salt Using Slow- Cooling Procedure

[00731] The results of the choline salt single- solvent fast- and slow-cooling crystallizations (see Tables 19 to 21) indicated that Form A was more likely to be isolated with fast-cooling profiles and Form B with slow-cooling profiles.

6.1.5.3. Binary Solvent Crystallizations

[00732] Binary- solvent crystallizations of the choline salt were performed using four primary solvents (MeOH, EtOH, IPA, and MeCN) and nine cosolvents (MTBE, EtOAc, IPAc, THF, c-hexane, heptane, toluene, CH2CI2, and dioxane) with a fast-cooling profile (supra). XRPD patterns showed that Form B was obtained from mixtures of MeOH with MTBE, EtOAc, IPAc, toluene, and dioxane. As shown in Table 22, Form A was obtained from mixtures of MeOH with THF and with CH2CI2 after evaporating the solvent to dryness. The mixtures of MeOH with cyclohexane and heptane provided the freeform of diazoxide. All solids obtained from fast-cooling procedures with EtOH, IPA, and MeCN as primary solvents provided Form B material.

Table 22. Binary-Solvent Crystallizations of Choline Salt of Diazoxide Using Fast- Cooling Procedure and MeOH as a Primary Solvent

* Solids were dissolved at 62 °C.

** Freeform of diazoxide.

[00733] Binary- solvent recrystallizations of the choline salt with the slow-cooling procedure were performed using two primary solvents (IPA and MeCN) and nine cosolvents (MTBE, EtOAc, IPAc, THF, c-hexane, heptane, toluene, CH2C12, and dioxane). All solids obtained from a slow-cooling procedure with IPA and MeCN as primary solvents provided Form B material based on XRPD analysis. The results of

binary- solvent crystallizations indicated that Form B was the most thermodynamic ally stable form of diazoxide choline.

6.1.5.4. Binary Solvent Crystallizations Using Water as a Cosolvent

[00734] In an attempt to investigate the formation of hydrates of the choline salt, experiments was performed using fast- and slow-cooling procedures and water as a cosolvent.

[00735] The fast cooling procedure (supra) was used with the exception of using different primary solvents which were miscible with water: acetone, acetonitrile, DMF, IPA, i-BuOH, i-AmOH, and t-AmOH. Water was utilized in these crystallizations as a cosolvent. All solids obtained from the fast-cooling procedure with water as the cosolvent provided diazoxide freeform material by XRPD analysis.

[00736] To compare the results obtained from the fast-cooling procedure a set of experiments was performed using a slow-cooling procedure and water as a cosolvent. All obtained solids were analyzed by XRPD and afforded patterns consistent with diazoxide freeform. Without wishing to be bound by theory, these results suggest that the conditions used for crystallization caused dissociation of the choline salt. A small amount of a second crop was obtained in each sample, but only two samples were analyzable by XRPD and indicated that the samples were freeform material. All mother liquors were evaporated to dryness and the residual solids were also analyzed by XRPD to afford patterns consistent with Form B of the choline salt.

6.1.5.5. Metastable Zone Width Estimation

[00737] Form B: To produce a robust process, an understanding of the solubility profiles of the various solid forms under consideration is required. From a practical standpoint, this involves the measurement of the metastable zone width (MSZW) of pure forms, whereby the saturation and supersaturation curves of the different forms are generated over a well defined concentration and temperature range. This knowledge can then be used to design a crystallization protocol that should ideally favor a selective crystal growth of the desired form.

[00738] Form B of diazoxide choline salt showed moderate solubility in a solvent mixture made of MeCN/MeOH/MtBE (10: 1: 12, volume ratios). The wide width of the metastable zone as shown in Table 23 gives many seeding options. During the MSZW measurement, aliquots from the crystallizing material were withdrawn and analyzed by XRPD to ensure that no form conversion occurred during the experiment. Indeed, the material remained unchanged during the test.

Table 23. Meta-Stable Zone Width For Form B Diazoxide Choline Salt in

MeCN/MeOH/MtBE (10:1:12) (v/v).

[00739] Form A: The metastable zone width for Form could not be estimated because this polymorphic form converted during the experiment to Form B.

6.1.5.6. Crystallization of Form A of Diazoxide Choline Salt

[00740] The choline salt of diazoxide (160.3 mg) was dissolved in 1 mL of IPA at 55 °C which was then passed through a Millipore 0.45 μΜ filter into a clean vial. This vial was placed in freezer a -20 °C overnight. Solids were not noticed and the flask was scratched with a micro- spatula. The vial was placed back in the freezer and nucleation was noticed after ten minutes. The solids were collected by vacuum filtration and washed with 1 mL of MtBE. The solids were dried in vacuo at 40 °C and 30 in. Hg to afford 70 mg (43.6% recovery) of Form A as determined by XRPD.

6.1.5.7. 500-mg Scale Crystallization of Form B of Diazoxide Choline Salt

[00741] The choline salt of diazoxide (524.3 mg) was dissolved in 3 mL of IPA at 78 °C and this solution was then cooled to 55 °C for the addition of MtBE. The MtBE (4 mL) was added until nucleation was observed. After nucleation the batch was allowed to cool to room temperature at a rate of 20 °C /h. The solids were collected by vacuum filtration and washed with 1 mL of MtBE. The solids were dried in vacuo at 40 °C and 30 in. of Hg to afford 426.7 mg (81.3% recovery) of Form B as determined by XRPD.

6.1.5.8. 2-g Scale Crystallization of Form B of Diazoxide Choline Salt

[00742] The choline salt of diazoxide (2.0015 g) was dissolved in 5.5 mL of IPA at 78 °C to afford a clear solution. This solution was passed through a Millipore Millex FH 0.45 μΜ filter. This solution was then cooled to 55 °C. MtBE was added in 1 mL portions, with a two minute interval between portions. Nucleation was noted after the second addition of MtBE. This suspension was allowed to cool to room temperature at a rate of 20 °C /h and stirred at this temperature for 16 hours. The solids were collected by vacuum filtration and washed with 1 mL of MtBE. The solids were dried in vacuo at 40 °C and 30 in. of Hg to afford 1.6091 g (80.4% recovery) of Form B as determined by XRPD.

6.1.5.9. Detection of Form Impurities

[00743] Mixtures of diazoxide choline Forms A and B were prepared by adding a minor amount of Form A to Form B. Samples were lightly ground by hands with a mortar and pestle for approximately one minute. Samples were then analyzed by XRPD analysis. XRPD analysis was found to be suitable for detecting 5% of Form A in Form B.

References

  1. Jump up^ Diazoxide, drugs.com
  2. Jump up to:a b “FDA Drug Safety Communication: FDA warns about a serious lung condition in infants and newborns treated with Proglycem (diazoxide)” (Press release). Food and Drug Administration. July 16, 2015. Retrieved 2015-07-19.
  3. Jump up^ van Hamersvelt HW, Kloke HJ, de Jong DJ, Koene RA, Huysmans FT (August 1996). “Oedema formation with the vasodilators nifedipine and diazoxide: direct local effect or sodium retention?”. Journal of Hypertension14 (8): 1041–5. doi:10.1097/00004872-199608000-00016PMID 8884561.closed access publication – behind paywall
  4. Jump up^ Huang Q, Bu S, Yu Y, et al. (January 2007). “Diazoxide prevents diabetes through inhibiting pancreatic beta-cells from apoptosis via Bcl-2/Bax rate and p38-beta mitogen-activated protein kinase”Endocrinology148 (1): 81–91. doi:10.1210/en.2006-0738PMID 17053028.open access publication – free to read
  5. Jump up^ Randle, John C.R.; Biton, Catherine; Lepagnol, Jean M. (15 November 1993). “Allosteric potentiation by diazoxide of AMPA receptor currents and synaptic potentials”. European Journal of Pharmacology247 (3): 257–65. doi:10.1016/0922-4106(93)90193-DPMID 8307099.closed access publication – behind paywall
  6. Jump up^ Panten, Uwe; Burgfeld, Johanna; Goerke, Frank; Rennicke, Michael; Schwanstecher, Mathias; Wallasch, Andreas; Zünkler, Bernd J.; Lenzen, Sigurd (1989-04-15). “Control of insulin secretion by sulfonylureas, meglitinide and diazoxide in relation to their binding to the sulfonylurea receptor in pancreatic islets”Biochemical Pharmacology38 (8): 1217–1229. doi:10.1016/0006-2952(89)90327-4.
  7. Jump up^ Doyle, Máire E.; Egan, Josephine M. (2003-03-01). “Pharmacological Agents That Directly Modulate Insulin Secretion”Pharmacological Reviews55 (1): 105–131. doi:10.1124/pr.55.1.7ISSN 1521-0081PMID 12615955.
Patent ID Title Submitted Date Granted Date
US2013309301 SALTS OF POTASSIUM ATP CHANNEL OPENERS AND USES THEREOF
2012-11-07
2013-11-21
US2013040942 SALTS OF POTASSIUM ATP CHANNEL OPENERS AND USES THEREOF
2012-07-06
2013-02-14
US2010028429 SALTS OF POTASSIUM ATP CHANNEL OPENERS AND USES THEREOF
2010-02-04
US9381202 SALTS OF POTASSIUM ATP CHANNEL OPENERS AND USES THEREOF
2013-03-18
2013-08-29
US2010256360 SALTS OF POTASSIUM ATP CHANNEL OPENERS AND USES THEREOF
2010-10-07
Patent ID Title Submitted Date Granted Date
US2012238554 SALTS OF POTASSIUM ATP CHANNEL OPENERS AND USES THEREOF
2012-02-27
2012-09-20
US7799777 SALTS OF POTASSIUM ATP CHANNEL OPENERS AND USES THEREOF
2007-08-16
2010-09-21
US2009062264 SALTS OF POTASSIUM ATP CHANNEL OPENERS AND USES THEREOF
2009-03-05
US9765043 SALTS OF POTASSIUM ATP CHANNEL OPENERS AND USES THEREOF
2014-08-22
2014-12-11
US7572789 SALTS OF POTASSIUM ATP CHANNEL OPENERS AND USES THEREOF
2009-06-11
2009-08-11

//////////////Diazoxide choline

CC1=NC2=C(C=C(C=C2)Cl)S(=O)(=O)[N-]1.C[N+](C)(C)CCO

CC1=NC2=C(C=C(C=C2)Cl)S(=O)(=O)[N-]1.C[N+](C)(C)CCO

Glycopyrrolate Tosylate

$
0
0

Glycopyrrolate Tosylate.pngFigure US20130211101A1-20130815-C00001

Glycopyrrolate Tosylate

Molecular Formula: C26H35NO6S
Molecular Weight: 489.627 g/mol

(1,1-dimethylpyrrolidin-1-ium-3-yl) 2-cyclopentyl-2-hydroxy-2-phenylacetate;4-methylbenzenesulfonate

CAS 873295-46-6 , C19 H28 N O3 . C7 H7 O3 S, Pyrrolidinium, 3-[(2-cyclopentyl-2-hydroxy-2-phenylacetyl)oxy]-1,1-dimethyl-, 4-methylbenzenesulfonate (1:1)

Glycopyrronium tosylate monohydrate

Molecular Formula, C19-H28-N-O3.C7-H8-O3-S.H2-O, Molecular Weight, 508.6522

https://chem.nlm.nih.gov/chemidplus/structure/1624259-25-1?maxscale=30&width=300&height=300

CAS 1624259-25-1, C19 H28 N O3 . C7 H7 O3 S . H2 O, Pyrrolidinium, 3-[(2-cyclopentyl-2-hydroxy-2-phenylacetyl)oxy]-1,1-dimethyl-, 4-methylbenzenesulfonate, hydrate (1:1:1)

Dermira (Originator)

DRM-04
DRM-04B

  • DRM-04 tosylate monohydrate
  • DRM04
  • DRM04 tosylate
  • Glycopyrronium tosylate
  • Glycopyrronium tosylate monohydrate
  • Glycopyrronium tosylate [USAN]
  • UNII-1PVF6JLU7B
  • UNII-X2N5209428

In 2018, the product was approved in the U.S. for the treatment of primary axillary hyperhidrosis in adult and pediatric patients 9 years of age and older.

In 2016, Maruho signed an exclusive license agreement with Dermina for product development and marketing in Japan for the treatment of axillary hyperhidrosis.

PATENT

https://patents.google.com/patent/US8558008B2/en

PATENT

https://patents.google.com/patent/US20130211101A1/en

  • Glycopyrrolate is a quaternary ammonium cation of the muscarinic anticholinergic group. Glycopyrrolate, typically as a bromide salt, has been used in the treatment of a variety of conditions including diarrhea (U.S. Pat. Nos. 6,214,792 and 5,919,760), urinary incontinence (U.S. Pat. Nos. 6,204,285 and 6,063,808), and anxiety (U.S. Pat. No. 5,525,347). Additionally, U.S. Pat. No. 5,976,499 discloses a method for diagnosing cystic fibrosis in a patient by, in part, stimulating sweat production through the injection of a glycopyrrolate solution into a patient. Glycopyrrolate has also been used for the treatment of hyperhidrosis in US 20100276329.
  • [0002]
    Glycopyrrolate has previously been made available as a bromide salt or an acetate salt. The bromide salt of glycopyrrolate is sold as Rubinol®. The term “glycopyrrolate” as used in the label for Rubinol® refers to the bromide salt which is more formally referred to as glycopyrronium bromide.
    • Example 6 Glycopyrrolate Tosylate
    • [0124]
      In a dark room, silver tosylate (3.5 g) was dissolved in water (˜100 mL) by sonication. The solution was heated to approximately 40° C. and additional water was added (˜15 mL). An equimolar amount of glycopyrrolate bromide (5 g) (mixture of R,S and S,R diastereomers) was added and immediately resulted in a yellow precipitate. The slurry was stirred at approximately 40° C. overnight, and then slowly cooled while stirring to ambient temperature. At ambient temperature, the solids were vacuum filtered and the wet cake was washed three times with approximately 10 mL of water. The mother liquor was collected and filtered two times through a 0.2 μm nylon filter with glass microfiber (GMF). A clear solution was observed after filtration and was lyophilized at approximately −50° C. After 6 days, a mixture of white, needle-like and slightly sticky, glassy solids was observed. Toluene (˜20 mL) was added, and the slurry was briefly sonicated and then stirred at ambient temperature. Additional toluene (˜80 mL) was added for easier stirring, and the mixture was allowed to stand at ambient conditions for 1 day. Solids of glycopyrrolate tosylate were collected by vacuum filtration and vacuum drying at ambient temperature for 1 day.

Example 7 Preparation of Glycopyrrolate Tosylate

    • [0125]
      A slurry of equimolar amounts of glycopyrrolate acetate and p-toluenesulfonic acid was prepared in isopropanol (1 mL). The mixture was stirred at ambient temperature. Additional isopropanol (0.5 mL) was added to improve stirring, and the mixture was stirred overnight. Solids of glycopyrrolate tosylate were isolated by vacuum filtration and analyzed.

Example 8 Preparation of Glycopyrrolate Tosylate Form D

    • [0126]
      Glycopyrrolate tosylate (1.0569 g) made from Example 6 was dissolved in 4 mL ACN/H2O (50/50 vol/vol) by sonication. The solution was filtered through 0.2 μm nylon filter into a clean vial. The solvent was allowed to partially evaporate from an open vial under ambient conditions. Further evaporation was subsequently performed under nitrogen gas flow. A gel resulted which was vacuum dried at 40° C. for 1 day. Toluene (5 mL) was added and the mixture was sonicated for approximately 10 minutes causing white solids to precipitate. The mixture was stirred at ambient temperature for 1 day. The solids were isolated by vacuum filtration and the wet cake was washed with approximately 10 mL of toluene. The solids were vacuum dried at ambient temperature for 1 day. After vacuum drying the solids were placed in a vial which remained uncapped and placed inside a relative humidity chamber (˜97%). The chamber was placed inside an oven at 41° C. After 6 days, the solids were analyzed by XRPD showing Form D.

Example 9 Single Crystal Preparation of Form D

    • [0127]
      Glycopyrrolate tosylate (54.9 mg) made from Example 6 was dissolved in EtOAc/DMF (87/13 vol/vol) at approximately 55° C. at 24 mg/ml. The solution was hot filtered through a 0.2 μm nylon filter into a pre-warmed vial. The vial containing the solution was first placed in a dry ice/acetone bath and then in a freezer (approximately −25 to −10° C.). After 3 days, the solution was re-heated to approximately 50° C. and additional EtOAc was added for 96/4 EtOAc/DMF (vol/vol) at 7 mg/ml. The solution was quickly removed from elevated temperature and placed in the freezer. Solids were isolated by decanting the solvent and drying the solids under ambient conditions.
    • [0128]
      Single Crystal Data Collection
    • [0129]
      A colorless chunk of C26H37NO7S [C7H7O3S, C19H28NO3, H2O] having approximate dimensions of 0.23×0.20×0.18 mm, was mounted on a fiber in random orientation. Preliminary examination and data collection were performed with Cu Kα radiation (λ=1.54184 Å) on a Rigaku Rapid II diffractometer equipped with confocal optics. Refinements were performed using SHELX97.

Example 10 Preparation of Dehydrated Form D

    • [0130]
      A mixture of glycopyrrolate tosylate solids, including Form C and Form D, and a trace amount of silver tosylate was kept over P2Oat ambient temperature for 18 days. The resulting solids were composed of a mixture of dehydrated Form D with a trace of silver tosylate as shown by XRPD analysis.

Example 11 Preparation of Form C Glycopyrrolate Tosylate

    • [0131]
      Glycopyrrolate tosylate Form D, containing trace amounts of Form C and silver tosylate, was heated on an Anton Paar TTK 450 stage and XRPD patterns were collected in situ in the range 3.5-26° (2θ). All heating steps were at approximately 10° C./min. The stage was heated in incremental steps of 20° C. from 25 to 125° C. At each step, an XRPD pattern was collected over approximately 4 minutes. The stage was then heated to 135° C. and an XRPD pattern was collected over approximately 16 minutes and after heating further to 145° C., a pattern was collected in approximately 31 minutes. The sample was subsequently cooled to 25° C. at approximately 24° C./min, upon which a final XRPD pattern was collected over approximately 16 min. The XRPD pattern of this final pattern was indexed as Form C.

Example 12 Preparation of Form C Glycopyrrolate Tosylate

    • [0132]
      Glycopyrrolate tosylate Form D from Example 6 was heated to an approximate temperature in the range 143-149° C. under a continuous nitrogen purge for approximately 3.3 hours. The vial containing the solids was capped, placed on a lab bench and allowed to cool down to room temperature. At room temperature, the vial was placed in a jar containing P2O5. The sample was prepared for XRPD analysis under nitrogen which confirmed production of Form C.

Example 13 Preparation of Form C Glycopyrrolate Tosylate

    • [0133]
      Glycopyrrolate tosylate (59.5 mg) from Example 6 was dissolved in acetone at approximately 50° C. at 27 mg/ml. The solution was hot filtered through a 0.2 μm nylon filter into a pre-warmed vial. The vial was capped and left on the hot plate which was subsequently turned off to allow the sample to cool slowly to ambient temperature. At ambient temperature the solution was stirred causing white solids to precipitate. The solids were isolated by vacuum filtration and the wet cake was washed with approximately 2 ml of acetone. XRPD analysis resulted in Form C.

Example 14 Amorphous Glycopyrrolate Tosylate

  • [0134]
    Glycopyrrolate tosylate from Example 6 was melted and cooled repeatedly until the majority of the solids had the appearance of a glass by microscopy. XRPD analysis indicated that the “glassy” sample was observed to be amorphous. A 2.2% weight loss was observed by TGA from 25 to 250° C. of the amorphous glycopyrrolate tosylate. The onset of the glass transition temperature was measured at 11.6° C.

In a dark room, silver tosylate (3.5 g) was dissolved in water (~ 100 mL) by sonication. The solution was heated to approximately 40°C. and additional water was added (-15 mL). An equimolar amount of glycopyrrolate bromide (5 g) (mixture of R,S and S,R diastereomers) was added and imme diately resulted in a yellow precipitate. The slurry was stirred at approximately 40°C. overnight, and then slowly cooled while stirring to ambient temperature. At ambient tempera ture, the solids were vacuum filtered and the wet cake was washed three times with approximately 10 mL of water. The mother liquor was collected and filtered two times through a 0.2 pm nylon filter with glass microfiber (GMF). A clear solution was observed after filtration and was lyophilized at approximately -50°C. After 6 days, a mixture of white, needle-like and slightly sticky, glassy solids was observed. Toluene (-20 mL) was added, and the slurry was briefly sonicated and then stirred at ambient temperature. Additional toluene (-80 mL) was added for easier stirring, and the mix ture was allowed to stand at ambient conditions for 1 day. Solids of glycopyrrolate tosylate were collected by vacuum filtration and vacuum drying at ambient temperature for 1 day. Glycopyrrolate Tosylate.

PAtent

https://patents.google.com/patent/CN103159659A/en

Image result for Glycopyrronium bromide synthesis

glycopyrrolate (I)

Methyl ethyl ketone (20mL) IOOmL three-necked flask was added 8 (4.6g, 15mmol) was, at (Γ5 ° C was added dropwise dibromomethane (2.9g, 30mmol) in butanone (5 mL) was added dropwise completed, continued The reaction was stirred for 15min, and a white solid precipitated, was allowed to stand 36h at room temperature, filtered off with suction, the filter cake was sufficiently dried to give crude ketone was recrystallized twice to give a white powdery crystals I (3.9g, 66%) mp 191~193 ° C chromatographic purity 99.8% [HPLC method, mobile phase: lmol / L triethylamine acetate – acetonitrile – water (1: 150: 49); detection wavelength: 230nm, a measurement of the area normalization method] .MS m / z: 318 ( m-BrO 1HNMR (CD3OD) δ:! 1.33~1.38 (m, 2H), 1.55~1.70 (m, 6H), 2.11~2.21 (m, 1H), 2.67~2.80 (m, 1H), 3.02 (m, 1H), 3.06 (s, 3H), 3.23 (s, 3H), 3.59~3.71 (m, 3H), 3.90 (dd, /=13.8,1H), 5.47 (m, 1H), 7.27 (t, 1H) , 7.35 (t, 2H), 7.62 (dd, 2H) .13C bandit R (DMSO) δ: 27.0, 27.4, 28.0, 31.3, 47.8, 53.8, 54.3, 66.0, 71.3, 74.6, 81.1, 126.9,128.7,129.3 , 143.2 17 5.00

Patent

https://patents.google.com/patent/WO2016204998A1/en

Image result for Glycopyrronium bromide synthesis

PAPER

https://link.springer.com/article/10.1007/s41981-018-0015-4

Sequential α-lithiation and aerobic oxidation of an arylacetic acid – continuous-flow synthesis of cyclopentyl mandelic acid

Open Access

Communications

Image result for Glycopyrronium bromide synthesis

The medicinal properties of glycopyrronium bromide (glycopyrrolate, 4) were first identified in the late 1950s [1]. Glycopyrrolate is an antagonist of muscarinic cholinergic receptors and is used for the treatment of drooling or excessive salivation (sialorrhea) [2], excess sweating (hyperhidrosis) [3], and overactive bladder and for presurgery treatment. In addition, it has recently been introduced as an effective bronchodilator for the treatment of chronic obstructive pulmonary disease (COPD) for asthma patients [4]. Glycopyrrolate displays few side effects because it does not pass through the blood brain barrier. Cyclopentyl mandelic acid (CPMA, 1), or its corresponding ester derivatives, are key intermediates in the synthetic routes to 4. CPMA (1) reacts with 1-methyl-pyrrolidin-3-ol (2) to form tertiary amine 3N-Methylation of 3 by methyl bromide gives quaternary ammonium salt glycopyrrolate 4 as a racemate (Scheme 1) [5].

Scheme 1

Synthesis of glycopyrrolate 4 from CPMA (1)

CPMA (1) is a synthetically challenging intermediate to prepare (Scheme 2). Routes A to D are most likely to be the commercially applied methods because these procedures are described in patents [5]. The published descriptions for the yields of 1 range from 28 to 56% for routes A to D. Ethyl phenylglyoxylate is reacted with cyclopentyl magnesium bromide to form an ester which is then hydrolyzed (route A) [6]. Phenylglyoxylic acid can be reacted in a similar manner with cyclopentyl magnesium bromide to directly form 1 (route B) [7]. Alternatively, the inverse addition of phenyl-Grignard reagent to cyclopentyl glyoxylic acid ester is reported (route C) [8]. Cyclopentyl glyoxylic acid ester can also be reacted with cyclopentadienyl magnesium bromide which is followed by an additional hydrogenation step with Pd/C and H2 to afford 1 (route D) [910].

Scheme 2

Existing synthetic pathways to CPMA (1)

Publication numberPriority datePublication dateAssigneeTitle
WO2014134510A1 *2013-02-282014-09-04Dermira, Inc.Glycopyrrolate salts
US8859610B22013-02-282014-10-14Dermira, Inc.Crystalline glycopyrrolate tosylate
US9006462B22013-02-282015-04-14Dermira, Inc.Glycopyrrolate salts
US20160052879A1 *2014-08-202016-02-25Dermira, Inc.Process for production of glycopyrronium tosylate
Family To Family Citations
WO2018026869A12016-08-022018-02-08Dermira, Inc.Processes for making, and methods of using, glycopyrronium compounds
Patent ID Title Submitted Date Granted Date
US9440056 DEVICE AND METHOD FOR DISPENSING A DRUG
2015-09-29
2016-03-31
US2016058735 METHODS OF TREATING HYPERHIDROSIS
2015-08-27
2016-03-03
Patent ID Title Submitted Date Granted Date
US2017157088 GLYCOPYRROLATE SALTS
2017-02-21
US9610278 Glycopyrrolate Salts
2016-01-07
2016-04-28
US2016052879 PROCESS FOR PRODUCTION OF GLYCOPYRRONIUM TOSYLATE
2015-08-19
2016-02-25
US9006461 CRYSTALLINE GLYCOPYRROLATE TOSYLATE
2013-09-11
2014-08-28
US2016243345 DEVICE AND METHOD FOR DISPENSING A DRUG
2016-05-04
2016-08-25
Patent ID Title Submitted Date Granted Date
US2016354315 DOSAGE FORMS AND USE THEREOF
2016-06-03
US9259414 Glycopyrrolate Salts
2015-03-10
2015-07-16
US9006462 Glycopyrrolate Salts
2014-08-29
2014-12-18
US8558008 Crystalline glycopyrrolate tosylate
2013-02-28
2013-10-15
US8859610 Crystalline glycopyrrolate tosylate
2013-09-11
2014-10-14

///////////Glycopyrrolate Tosylate, DRM-04 , DRM-04B , FDA 2018, Qbrexza

CC1=CC=C(C=C1)S(=O)(=O)[O-].C[N+]1(CCC(C1)OC(=O)C(C2CCCC2)(C3=CC=CC=C3)O)C

Tasimelteon, タシメルテオン

$
0
0

ChemSpider 2D Image | Tasimelteon | C15H19NO2

Tasimelteon.png

Tasimelteon

N-([(1R,2R)-2-(2,3-Dihydro-1-benzofuran-4-yl)cyclopropyl]methyl)propanamide,

609799-22-6 [RN]
8985
Hetlioz [Trade name]
N-{[(1R,2R)-2-(2,3-Dihydro-1-benzofuran-4-yl)cyclopropyl]methyl}propanamide [ACD/IUPAC Name]
Propanamide, N-[[(1R,2R)-2-(2,3-dihydro-4-benzofuranyl)cyclopropyl]methyl]- [ACD/Index Name]
SHS4PU80D9

609799-22-6 cas, BMS-214778; VEC-162, ATC:N05CH03

  • Use:Treatment of sleep disorder; Melatonin receptor agonist
  • (1R,2R)-N-[2-(2,3-dihydrobenzofuran-4-yl)cyclopropylmethyl]propanamide
  • Formula:C15H19NO2, MW:245.3 g/mol
  • Hetlioz Vanda Pharmaceuticals, 2014

Approved fda 2014

EMA

Tasimelteon is a white to off-white crystalline powder, it is non hygroscopic, soluble in water across relevant pH values and freely soluble in alcohols, cyclohexane, and acetonitrile. Conducted in vivo studies demonstrate that tasimelteon is highly permeable substance. Photostability testing and testing on stress conditions demonstrated that the active substance degrades in light.

Tasimelteon exhibits stereoisomerism due to the presence of two chiral centres. Active substance is manufactured as a single, trans-1R,2R isomer. Enantiomeric purity is controlled routinely during manufacture of active substance intermediates by chiral HPLC/specific optical rotation and additionally controlled in the active substance. Stability data indicates tasimelteon is isomerically stable.

Polymorphism has been observed in polymorphic screening studies for tasimelteon and two forms have been identified. The thermodynamically more stable form has been chosen for development and the manufacturing process consistently yields active substance of single, desired polymorphic form. It was demonstrated that milling of the active substance does not affect polymorphic form. Polymorphism is additionally controlled in active substance release and shelf-life specifications using X-ray powder diffraction analysis.

Tasimelteon is synthesized in nine main steps using linear synthesis and using commercially available well-defined starting materials with acceptable specifications. Three intermediates are isolated for control of active substance quality including stereochemical control. The active substance is isolated by slow recrystallisation or precipitation of tasimelteon from an ethanol/water mixture which ensures the formation of desired polymorphic form. Up to two additional, optional recrystallisations may be performed for unmilled tasimelteon to ensure that milled tasimelteon active substance is of high purity. Seed crystals complying with active substance specifications can be used optionally. Active substance is jet milled (micronised) to reduce and control particle size, which is critical in finished product performance with regards to content uniformity and dissolution…….http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Public_assessment_report/human/003870/WC500190309.pdf

launched in 2014 in the U.S. by Vanda Pharmaceuticals for the treatment of non-24-hour sleep-wake disorder in totally blind subjects. In 2015, the European Committee for Medicinal Products of the European Medicines Agency granted approval for the same indication.  In 2010 and 2011, orphan drug designations were assigned for the treatment of non-24 hour sleep/wake disorder in blind individuals without light perception in the U.S. and the E.U., respectively.

Tasimelteon (trade name Hetlioz) is a drug approved by the U.S. Food and Drug Administration (FDA)[2] in January 2014 for the treatment of non-24-hour sleep–wake disorder (also called Non-24, N24 and N24HSWD).[3] In June 2014, the European Medicines Agency accepted an EU filing application for tasimelteon[4] and in July 2015, the drug was approved in Europe for the treatment of non-24-hour sleep-wake rhythm disorder in totally blind adults,[5] but not in the rarer case of non-24 in sighted people.

Tasimelteon is a selective agonist for the melatonin receptors MT1 and MT2, similar to other members of the melatonin receptor agonistclass of which ramelteon (2005) and agomelatine (2009) were the first approved.[6] As a treatment for N24HSWD, as with melatonin or other melatonin derivatives, the patient may experience improved sleep timing while taking the drug. Reversion to baseline sleep performance occurs within a month of discontinuation.[7]

Image result for TASIMELTEON DRUG FUTURE

Development

Tasimelteon (previously known as BMS-214,778) was developed for the treatment of insomnia and other sleep disorders. A phase II trial on circadian rhythm sleep disorders was concluded in March 2005.[8] A phase III insomnia trial was conducted in 2006.[9] A second phase III trial on insomnia, this time concerning primary insomnia, was completed in June 2008.[10] In 2010, the FDA granted orphan drug status to tasimelteon, then regarded as an investigational medication, for use in totally blind adults with N24HSWD.[11] (Through mechanisms such as easing the approval process and extending exclusivity periods, orphan drug status encourages development of drugs for rare conditions that otherwise might lack sufficient commercial incentive.)

On completion of Phase III trials, interpretations of the clinical trials by the research team concluded that the drug may have therapeutic potential for transient insomnia in circadian rhythm sleep disorders.[12] A year-long (2011–2012) study at Harvard tested the use of tasimelteon in blind subjects with non-24-hour sleep-wake disorder. The drug has not been tested in children nor in any non-blind people.

FDA approval

In May 2013 Vanda Pharmaceuticals submitted a New Drug Application to the Food and Drug Administration for tasimelteon for the treatment of non-24-hour sleep–wake disorder in totally blind people. It was approved by the FDA on January 31, 2014 under the brand name Hetlioz.[3] In the opinion of Public Citizen, an advocacy group, the FDA erroneously allowed it to be labelled without stating that it is only approved for use by totally blind people.[13] However, FDA updated its press release on Oct. 2, 2014 to clarify the approved use of Hetlioz, which includes both sighted and blind individuals. The update did not change the drug labeling (prescribing information).[14]

Toxicity

Experiments with rodents revealed fertility impairments, an increase in certain cancers, and serious adverse events during pregnancy at dosages in excess of what is considered the “human dose”.[15][16]

As expected, advisors to the US Food and Drug Administration have recommended approval of Vanda Pharmaceuticals’ tasimelteon, to be sold as Hetlioz, for the treatment of non-24-hour disorder in the totally blind.http://www.pharmatimes.com/Article/13-11-14/FDA_panel_backs_Vanda_body_clock_drug_for_blind.aspx

The master body clock controls the timing of many aspects of physiology, behavior and metabolism that show daily rhythms, including the sleep-wake cycles, body temperature, alertness and performance, metabolic rhythms and certain hormones which exhibit circadian variation. Outputs from the

suprachiasmatic nucleus (SCN) control many endocrine rhythms including those of melatonin secretion by the pineal gland as well as the control of Cortisol secretion via effects on the hypothalamus, the pituitary and the adrenal glands. This master body clock, located in the SCN, spontaneously generates rhythms of approximately 24.5 hours. These non-24-hour rhythms are synchronized each day to the 24-hour day-night cycle by light, the primary environmental time cue which is detected by specialized cells in the retina and transmitted to the SCN via the retino-hypothalamic tract. Inability to detect this light signal, as occurs in most totally blind individuals, leads to the inability of the master body clock to be reset daily and maintain entrainment to a 24-hour day.

Non-24-Hour Disorder, Non-24, also referred to as Non-24-Hour Sleep-Wake Disorder, (N24HSWD) or Non-24-Hour Disorder, is an orphan indication affecting approximately 65,000 to 95,000 people in the U.S. and 140,000 in Europe. Non- 24 occurs when individuals, primarily blind with no light perception, are unable to synchronize their endogenous circadian pacemaker to the 24-hour light/dark cycle. Without light as a synchronizer, and because the period of the internal clock is typically a little longer than 24 hours, individuals with Non-24 experience their circadian drive to initiate sleep drifting later and later each day. Individuals with Non-24 have abnormal night sleep patterns, accompanied by difficulty staying awake during the day. Non-24 leads to significant impairment, with chronic effects impacting the social and occupational functioning of these individuals.

In addition to problems sleeping at the desired time, individuals with Non-24 experience excessive daytime sleepiness that often results in daytime napping.

The severity of nighttime sleep complaints and/or daytime sleepiness complaints varies depending on where in the cycle the individual’s body clock is with respect to their social, work, or sleep schedule. The “free running” of the clock results in approximately a 1-4 month repeating cycle, the circadian cycle, where the circadian drive to initiate sleep continually shifts a little each day (about 15 minutes on average) until the cycle repeats itself. Initially, when the circadian cycle becomes desynchronous with the 24h day-night cycle, individuals with Non-24 have difficulty initiating sleep. As time progresses, the internal circadian rhythms of these individuals becomes 180 degrees out of synchrony with the 24h day-night cycle, which gradually makes sleeping at night virtually impossible, and leads to extreme sleepiness during daytime hours.

Eventually, the individual’s sleep-wake cycle becomes aligned with the night, and “free-running” individuals are able to sleep well during a conventional or socially acceptable time. However, the alignment between the internal circadian rhythm and the 24-hour day-night cycle is only temporary.

In addition to cyclical nighttime sleep and daytime sleepiness problems, this condition can cause deleterious daily shifts in body temperature and hormone secretion, may cause metabolic disruption and is sometimes associated with depressive symptoms and mood disorders.

It is estimated that 50-75% of totally blind people in the United States (approximately 65,000 to 95,000) have Non-24. This condition can also affect sighted people. However, cases are rarely reported in this population, and the true rate of Non-24 in the general population is not known.

The ultimate treatment goal for individuals with Non-24 is to entrain or synchronize their circadian rhythms into an appropriate phase relationship with the 24-hour day so that they will have increased sleepiness during the night and increased wakefulness during the daytime. Tasimelteon

Tasimelteon is a circadian regulator which binds specifically to two high affinity melatonin receptors, Mella (MT1R) and Mellb (MT2R). These receptors are found in high density in the suprachiasmatic nucleus of the brain (SCN), which is responsible for synchronizing our sleep/wake cycle. Tasimelteon has been shown to improve sleep parameters in prior clinical studies, which simulated a desynchronization of the circadian clock. Tasimelteon has so far been studied in hundreds of individuals and has shown a good tolerability profile.

Tasimelteon has the chemical name: tr ns-N-[[2-(2,3-dihydrobenzofuran- 4-yl)cycloprop-lyl] methyl] propanamide, has the structure of Formula I:

Figure imgf000008_0001

Formula I

and is disclosed in US 5856529 and in US 20090105333, both of which are incorporated herein by reference as though fully set forth.

Tasimelteon is a white to off-white powder with a melting point of about 78°C (DSC) and is very soluble or freely soluble in 95% ethanol, methanol, acetonitrile, ethyl acetate, isopropanol, polyethylene glycols (PEG-300 and PEG- 400), and only slightly soluble in water. The native pH of a saturated solution of tasimelteon in water is 8.5 and its aqueous solubility is practically unaffected by pH. Tasimelteon has 2-4 times greater affinity for MT2R relative to MTIR. It’s affinity (¾) for MTIR is 0.3 to 0.4 and for MT2R, 0.1 to 0.2. Tasimelteon is useful in the practice of this invention because it is a melatonin agonist that has been demonstrated, among other activities, to entrain patients suffering from Non-24.

Metabolites of tasimelteon include, for example, those described in “Preclinical Pharmacokinetics and Metabolism of BMS-214778, a Novel

Melatonin Receptor Agonist” by Vachharajani et al., J. Pharmaceutical Sci., 92(4):760-772, which is hereby incorporated herein by reference. The active metabolites of tasimelteon can also be used in the method of this invention, as can pharmaceutically acceptable salts of tasimelteon or of its active metabolites. For example, in addition to metabolites of Formula II and III, above, metabolites of tasimelteon also include the monohydroxylated analogs M13 of Formula IV, M12 of Formula V, and M14 of Formula VI.

Formula IV

Figure imgf000010_0001

Formula V

MO

Figure imgf000010_0002

Formula VI

Thus, it is apparent that this invention contemplates entrainment of patients suffering free running circadian rhythm to a 24 hour circadian rhythm by administration of a circadian rhythm regulator (i.e., circadian rhythm modifier) capable of phase advancing and/or entraining circadian rhythms, such as a melatonin agonist like tasimelteon or an active metabolite oftasimelteon or a pharmaceutically acceptable salt thereof. Other MT1R and MT2R agonists, i.e., melatonin agonists, can have similar effects on the master body clock. So, for example, this invention further contemplates the use of melatonin agonists such as but not limited to melatonin, N-[l-(2,3-dihydrobenzofuran-4- yl)pyrrolidin-3-yl]-N-ethylurea and structurally related compounds as disclosed in US 6,211,225, LY-156735 ((R)-N-(2-(6-chloro-5-methoxy-lH-indol- 3yl) propyl) acetamide) (disclosed in U.S. Patent No. 4,997,845), agomelatine (N- [2-(7-methoxy-l-naphthyl)ethyl]acetamide) (disclosed in U.S. Patent No.

5,225,442), ramelteon ((S)-N-[2-(l,6,7,8-tetrahydro-2H-indeno- [5,4-b] furan-8- yl)ethyl]propionamide), 2-phenylmelatonin, 8-M-PDOT, 2-iodomelatonin, and 6- chloromelatonin.

Additional melatonin agonists include, without limitation, those listed in U.S. Patent Application Publication No. 20050164987, which is incorporated herein by reference, specifically: TAK-375 (see Kato, K. et al. Int. J.

Neuropsychopharmacol. 2000, 3 (Suppl. 1): Abst P.03.130; see also abstracts P.03.125 and P.03.127), CGP 52608 (l-(3-allyl-4-oxothiazolidine-2-ylidene)-4- met- hylthiosemicarbazone) (See Missbach et al., J. Biol. Chem. 1996, 271, 13515-22), GR196429 (N-[2-[2,3,7,8-tetrahydro-lH-fur-o(2,3-g)indol-l- yl] ethyl] acetamide) (see Beresford et al., J. Pharmacol. Exp. Ther. 1998, 285, 1239-1245), S20242 (N-[2-(7-methoxy napth-l-yl) ethyl] propionamide) (see Depres-Brummer et al., Eur. J. Pharmacol. 1998, 347, 57-66), S-23478 (see Neuropharmacology July 2000), S24268 (see Naunyn Schmiedebergs Arch. June 2003), S25150 (see Naunyn Schmiedebergs Arch. June 2003), GW-290569, luzindole (2-benzyl-N-acetyltryptamine) (see U.S. Patent No. 5,093,352), GR135531 (5-methoxycarbonylamino-N-acetyltrypt- amine) (see U.S. Patent Application Publication No. 20010047016), Melatonin Research Compound A, Melatonin Agonist A (see IMSWorld R&D Focus August 2002), Melatonin

Analogue B (see Pharmaprojects August 1998), Melatonin Agonist C (see Chem. Pharm. Bull. (Tokyo) January 2002), Melatonin Agonist D (see J. Pineal Research November 2000), Melatonin Agonist E (see Chem. Pharm. Bull. (Tokyo) Febrary 2002), Melatonin Agonist F (see Reprod. Nutr. Dev. May 1999), Melatonin Agonist G (see J. Med. Chem. October 1993), Melatonin Agonist H (see Famaco March 2000), Melatonin Agonist I (see J. Med. Chem. March 2000), Melatonin Analog J (see Bioorg. Med. Chem. Lett. March 2003), Melatonin Analog K (see MedAd News September 2001), Melatonin Analog L, AH-001 (2-acetamido-8- methoxytetralin) (see U.S. Patent No. 5,151,446), GG-012 (4-methoxy-2- (methylene propylamide)indan) (see Drijfhout et al., Eur. J. Pharmacol. 1999, 382, 157-66), Enol-3-IPA, ML-23 (N-2,4-dinitrophenyl-5-methoxy-tryptamine ) (see U.S. Patent No. 4,880,826), SL-18.1616, IP-100-9 (US 5580878), Sleep Inducing Peptide A, AH-017 (see U.S. Patent No. 5,151,446), AH-002 (8-methoxy- 2-propionamido-tetralin) (see U.S. Patent No. 5,151,446), and IP-101.

Metabolites, prodrugs, stereoisomers, polymorphs, hydrates, solvates, and salts of the above compounds that are directly or indirectly active can, of course, also be used in the practice of this invention.

Melatonin agonists with a MT1R and MT2R binding profile similar to that of tasimelteon, which has 2 to 4 time greater specificity for MT2R, are preferred.

Tasimelteon can be synthesized by procedures known in the art. The preparation of a 4-vinyl-2,3-dihydrobenzofuran cyclopropyl intermediate can be carried out as described in US7754902, which is incorporated herein by reference as though fully set forth.

Pro-drugs, e.g., esters, and pharmaceutically acceptable salts can be prepared by exercise of routine skill in the art.

In patients suffering a Non-24, the melatonin and Cortisol circadian rhythms and the natural day/night cycle become desynchronized. For example, in patients suffering from a free-running circadian rhythm, melatonin and Cortisol acrophases occur more than 24 hours, e.g., >24.1 hours, prior to each previous day’s melatonin and Cortisol acrophase, respectively, resulting in desynchronization for days, weeks, or even months, depending upon the length of a patient’s circadian rhythm, before the melatonin, Cortisol, and day /night cycles are again temporarily synchronized.

Chronic misalignment of Cortisol has been associated with metabolic, cardiac, cognitive, neurologic, neoplastic, and hormonal disorders. Such disorders include, e.g., obesity, depression, neurological impairments.

Structure-activity relationship
SAR
Figure : Melatonin receptor agonists. The applied colors indicate the mutual properties with the general melatonin receptor agonists pharmacophore.

INTRODUCTION

Tasimelteon has the chemical name: trans-N-[[2-(2,3-dihydrobenzofuran-4-yl)cycloprop-1yl]methyl]propanamide, has the structure of Formula I:

Figure US20130197076A1-20130801-C00001

and is disclosed in U.S. Pat. No. 5,856,529 and in US 20090105333, both of which are incorporated herein by reference as though fully set forth.

Tasimelteon is a white to off-white powder with a melting point of about 78° C. (DSC) and is very soluble or freely soluble in 95% ethanol, methanol, acetonitrile, ethyl acetate, isopropanol, polyethylene glycols (PEG-300 and PEG-400), and only slightly soluble in water. The native pH of a saturated solution of tasimelteon in water is 8.5 and its aqueous solubility is practically unaffected by pH. Tasimelteon has 2-4 times greater affinity for MT2R relative to MT1R. It’s affinity (Ki) for MT1R is 0.3 to 0.4 and for MT2R, 0.1 to 0.2. Tasimelteon is useful in the practice of this invention because it is a melatonin agonist that has been demonstrated, among other activities, to entrain patients suffering from Non-24.

SYNTHESIS

(1R-trans)-N-[[2 – (2,3-dihydro-4 benzofuranyl) cyclopropyl] methyl] propanamide PATENT: BRISTOL-MYERS SQUIBB PRIORITY DATE: 1996 HYPNOTIC

Synthesis Tasimelteon

PREPARATION OF XV

XXIV D-camphorsulfonic acid IS REACTED WITH THIONYL CHLORIDE TO GIVE

…………XXV (1S, 4R) -7,7-dimethyl-2-oxo-bicyclo [2.2.1] heptane-1-methanesulfonyl chloride

TREATED WITH

XXVI ammonium hydroxide

TO GIVE

XXVII (1S, 4R) -7,7-dimethyl-2-oxo-bicyclo [2.2.1] heptane-1-methanesulfonamide

TREATED WITH AMBERLYST15

….XXVIII (3aS, 6R) -4,5,6,7-tetrahydro-8 ,8-dimethyl-3H-3a ,6-methano-2 ,1-benzisothiazole-2 ,2-dioxide

TREATED WITH LAH, ie double bond is reduced to get

…..XV (3aS, 6R, 7aR)-hexahydro-8 ,8-dimethyl-3H-3a ,6-methano-2 ,1-benzisothiazole-2 ,2-dioxide

Intermediate

I 3-hydroxybenzoic acid methyl ester

II 3-bromo-1-propene

III 3 – (2-propenyloxy) benzoic acid methyl ester

IV 3-hydroxy-2-(2-propenyl) benzoic acid methyl ester

V 2,3-dihydro-4-hydroxy-2-benzofurancarboxylic acid methyl ester

VI benzofuran-4-carboxylic acid methyl ester

VII benzofuran-4-carboxylic acid

VIII 2,3-dihydro-4-benzofurancarboxylic acid

IX 2,3-dihydro-4-benzofuranmethanol

X 2,3-dihydro-4-benzofurancarboxaldehyde

XI Propanedioic acid

XII (E) -3 – (2,3-dihydro-4-benzofuranyl) propenoic acid

XIII thionyl chloride

XIV (E) -3 – (2,3-dihydro-4-benzofuranyl) propenoyl chloride

XV (3aS, 6R, 7aR)-hexahydro-8 ,8-dimethyl-3H-3a ,6-methano-2 ,1-benzisothiazole-2 ,2-dioxide

XVI (3aS,6R,7aR)-1-[(E)-3-(2,3-dihydro-4-benzofuranyl)-1-oxo-2-propenyl]hexahydro-8,8-dimethyl-3H-3a,6-methano-2,1-benzisothiazole-2,2-dioxide

XVII (3aS,6R,7aR)-1-[[(1R,2R)-2-(2,3-dihydro-4-benzofuranyl)cyclopropyl]carbonyl]hexahydro-8,8-dimethyl-3H-3a,6-methano-2,1-benzisothiazole-2,2-dioxide

XVIII [R-(R *, R *)] -2 – (2,3-dihydro-4-benzofuranyl) cyclopropanemethanol

XIX [R-(R *, R *)] -2 – (2,3-dihydro-4-benzofuranyl) cyclopropanecarboxaldehyde

XX hydroxylamine hydrochloride

XXI [R-(R *, R *)] -2 – (2,3-dihydro-4-benzofuranyl) cyclopropanecarbaldehyde oxime

XXII [R-(R *, R *)] -2 – (2,3-dihydro-4-benzofuranyl) cyclopropanemethanamine

XXIII propanoyl chloride

XXIV D-camphorsulfonic acid

XXV (1S, 4R) -7,7-dimethyl-2-oxo-bicyclo [2.2.1] heptane-1-methanesulfonyl chloride

XXVI ammonium hydroxide

XXVII (1S, 4R) -7,7-dimethyl-2-oxo-bicyclo [2.2.1] heptane-1-methanesulfonamide

XXVIII (3aS, 6R) -4,5,6,7-tetrahydro-8 ,8-dimethyl-3H-3a ,6-methano-2 ,1-benzisothiazole-2 ,2-dioxide

Bibliography

– Patents: Benzofuran and dihydrobenzofuran melatonergic agents: US5856529 (1999)

Priority: US19960032689P, 10 Dec. 1996 (Bristol-Myers Squibb Company, U.S.)

– Preparation III (quinazolines): US2004044015 (2004) Priority: EP20000402845, 13 Oct. 2000

– Preparation of VII (aminoalkylindols): Structure-Activity Relationships of Novel Cannabinoid Mimetics Eissenstat et al, J.. Med. Chem. 1995, 38, 3094-3105

– Preparation XXVIII: Towson et al. Organic Syntheses, Coll. Vol. 8, p.104 (1993) Vol. 69, p.158 (1990)

– Preparation XV: Weismiller et al. Organic Syntheses, Coll. Vol. 8, p.110 (1993) Vol. 69, p.154 (1990).

– G. Birznieks et al. Melatonin agonist VEC-162 Improves sleep onset and maintenance in a model of transient insomnia. Sleep 2007, 30, 0773 Abstract.

-. Rajaratnam SM et al, The melatonin agonist VEC-162 Phase time immediately advances the human circadian system, Sleep 2006, 29, 0159 Abstract.

-. AK Singh et al, Evolution of a manufacturing route for a highly potent drug candidate, 229th ACS Natl Meet, March 13-17, 2005, San Diego, Abstract MEDI 576.

– Vachharajani NN et al, Preclinical pharmacokinetics and metabolism of BMS-214778, a novel melatonin receptor agonist, J Pharm Sci. 2003 Apr; 92 (4) :760-72.

. – JW Scott et al, Catalytic Asymmetric Synthesis of a melotonin antagonist; synthesis and process optimization. 223rd ACS Natl Meet, April 7-11, Orlando, 2002, Abstract ORGN 186.

SYNTHESIS CONSTRUCTION AS IN PATENT

WO1998025606A1

GENERAL SCHEMES

Reaction Scheme 1

Figure imgf000020_0001

The syntheses of the 4-aryl-propenoic acid derivatives, 2 and 3, are shown in Reaction Scheme 1. The starting aldehydes, 1 , can be prepared by methods well known to those skilled in the art. Condensation of malonic acid with the aldehydes, 1, in solvents such as pyridine with catalysts such as piperidine or pyrrolidine, gives the 4-aryl- propenoic acid, 2. Subsequent conversion of the acid to the acid chloride using reagents such as thionyl chloride, phosphoryl chloride, or the like, followed by reaction with N,0-dimethyl hydroxylamine gives the amide intermediate 3 in good yields. Alternatively, aldehyde 1 can be converted directly to amide 3 using reagents such as diethyl (N-methoxy- N-methyl-carbamoylmethyl)phosphonate with a strong base such as sodium hydride.

Reaction Scheme 2

Figure imgf000020_0002

The conversion of the amide intermediate 3 to the racemic, trans- cyclopropane carboxaldehyde intermediate, 4, is shown in Reaction Scheme 2. Intermediate 3 was allowed to react with cyclopropanating reagents such as trimethylsulfoxonium iodide and sodium hydride in solvents such as DMF, THF, or the like. Subsequent reduction using reagents such as LAH in solvents such as THF, ethyl ether, or the like, gives the racemic, trans-cyclopropane carboxaldehyde intermediates, 4.

Reaction Scheme 3

Figure imgf000021_0001

Racemic cyclopropane intermediate 5 (R = halogen) can be prepared from intermediate 2 as shown in Reaction Scheme 3. Intermediate 2 was converted to the corresponding allylic alcohol by treatment with reducing agents such as sodium borohydride plus iodine in solvents such as THF. Subsequent acylation using reagents such as acetic anhydride in pyridine or acetyl chloride gave the allylic acetate which was allowed to react with cyclopropanating reagents such as sodium chloro-difluoroacetate in diglyme to provide the racemic, trans- cyclopropane acetate intermediates, 5. Reaction Scheme 4

Figure imgf000022_0001

The conversion of the acid 2 to the chiral cyclopropane carboxaldehyde intermediate, (-)-(trans)-4, is shown in Reaction Scheme 4. Intermediate 2 is condensed with (-)-2,10-camphorsultam under standard conditions, and then cyclopropanated in the presence of catalysts such as palladium acetate using diazomethane generated from reagents such as 1-methyl-3-nitro-1-nitrosoguanidine. Subsequent reduction using reagents such as LAH in solvents such as THF, followed by oxidation of the alcohol intermediates using reagents such as DMSO/oxalyl chloride, or PCC, gives the cyclopropane carboxaldehyde intermediate, (-)-(trans)-4, in good yields. The enantiomer, (+)-(trans)-4, can also be obtained employing a similar procedure using (+)-2,10- camphorsultam in place of (-)-2,10-camphorsultam.

When it is desired to prepare compounds of Formula I wherein m = 2, the alcohol intermediate may be activated in the conventional manner such as with mesyl chloride and treated with sodium cyanide followed by reduction of the nitrile group with a reducing agent such as LAH to produce the amine intermediate 6.

Reaction Scheme 5

Figure imgf000023_0001
Figure imgf000023_0002

Reaction Scheme 5 shows the conversion of intermediates 4 and 5 to the amine intermediate, 7, and the subsequent conversion of 6. or 7 to compounds of Formula I. The carboxaldehyde intermediate, 4, is condensed with hydroxylamine and then reduced with reagents such as LAH to give the amine intermediate, 7. The acetate intermediate 5 is hydrolyzed with potassium hydroxide to the alcohol, converted to the mesylate with methane sulfonyl chloride and triethyl amine in CH2CI2and then converted to the azide by treatment with sodium azide in solvents such as DMF. Subsequent reduction of the azide group with a reducing agent such as LAH produced the amine intermediate 7. Further reaction of 6 or 7 with acylating reagents gives compounds of Formula I. Suitable acylating agents include carboxylic acid halides, anhydrides, acyl imidazoles, alkyl isocyanates, alkyl isothiocyanates, and carboxylic acids in the presence of condensing agents, such as carbonyl imidazole, carbodiimides, and the like. Reaction Scheme 6

Figure imgf000024_0001

Reaction Scheme 6 shows the alkylation of secondary amides of Formula I (R2 = H) to give tertiary amides of Formula I (R2 = alkyl). The secondary amide is reacted with a base such as sodium hydride, potassium tert-butoxide, or the like, and then reacted with an alkylating reagent such as alkyl halides, alkyl sulfonate esters, or the like to produce tertiary amides of Formula I.

Reaction Scheme 7

Figure imgf000024_0002

Reaction Scheme 7 shows the halogenation of compounds of Formula I. The carboxamides, i (Q1 = Q2 = H), are reacted with excess amounts of halogenating agents such as iodine, N-bromosuccinimide, or the like to give the dihalo-compounds of Formula I (Q1 = Q2 = halogen). Alternatively, a stoichiometric amount of these halogenating agents can be used to give the monohalo-compounds of Formula I (Q1 = H, Q2 = halogen; or Q1 = halogen, Q2 = H). In both cases, additives such as lead IV tetraacetate can be used to facilitate the reaction. Biological Activity of the Compounds

The compounds of the invention are melatonergic agents. They have been found to bind human melatonergic receptors expressed in a stable cell line with good affinity. Further, the compounds are agonists as determined by their ability, like melatonin, to block the forskolin- stimulated accumulation of cAMP in certain cells. Due to these properties, the compounds and compositions of the invention should be useful as sedatives, chronobiotic agents, anxiolytics, antipsychotics, analgesics, and the like. Specifically, these agents should find use in the treatment of stress, sleep disorders, seasonal depression, appetite regulation, shifts in circadian cycles, melancholia, benign prostatic hyperplasia and related conditions

EXPERIMENTAL PROCEDURES

SEE ORIGINAL PATENT FOR CORECTIONS

Preparation 1

Benzofuran-4-carboxaldehyde

Step 1 : N-Methoxy-N-methyl-benzofuran-4-carboxamide

A mixture of benzofuran-4-carboxylic acid [Eissenstat, et al.. J. Medicinal Chemistry, 38 (16) 3094-3105 (1995)] (2.8 g, 17.4 mmol) and thionyl chloride (25 mL) was heated to reflux for 2 h and then concentrated in vacuo. The solid residue was dissolved in ethyl acetate (50 mL) and a solution of N,O-dimethylhydroxylamine hydrochloride (2.8 g) in saturated NaHC03(60 mL) was added with stirring. After stirring for 1.5 h, the ethyl acetate layer was separated. The aqueous layer was extracted with ethyl acetate. The ethyl acetate extracts were combined, washed with saturated NaHCO3 and concentrated in vacuo to give an oil (3.2 g, 95.4%).

Step 2: Benzofuran-4-carboxaldehyde

A solution of N-methoxy-N-methyl-benzofuran-4-carboxamide (3.2 g, 16.6 mmol) in THF (100 mL) was cooled to -45°C and then LAH (0.7 g, 18.7 mmol) was added. The mixture was stirred for 15 min, allowed to warm to -5°C, and then recooled to -45°C. Saturated KHS04 (25 mL) was added with vigorous stirring, and the mixture was allowed to warm to room temperature. The precipitate was filtered and washed with acetone. The filtrate was concentrated in vacuo to give an oil (2.3 g, 94%). Preparation 2

2,3-Dihydrobenzofuran-4-carboxaldehyde

Step 1 : 2,3-Dihydrobenzofuran-4-carboxylic acid

Benzofuran-4-carboxylic acid (10.0 g, 61 .7 mmol) was hydrogenated (60 psi) in acetic acid (100 mL) over 10% Pd/C (2 g) for 12 hr. The mixture was filtered and the filtrate was diluted with water (500 mL) to give 2,3- dihydrobenzofuran-4-carboxylic acid as a white powder (8.4 g, 83%). A sample was recrystallized from isopropanol to give fine white needles (mp: 185.5-187.5°C).

Step 2: (2,3-Dihydrobenzofuran-4-yl)methanol

A solution of 2,3-dihydrobenzofuran-4-carboxylic acid (10 g, 61 mmol) in THF (100 mL) was stirred as LAH (4.64 g, 122 mmol) was slowly added. The mixture was heated to reflux for 30 min. The mixture was cooled and quenched cautiously with ethyl acetate and then with 1 N HCI (150 mL). The mixture was then made acidic with 12 N HCI until all the inorganic precipitate dissolved. The organic layer was separated, and the inorganic layer was extracted twice with ethyl acetate. The organic layers were combined, washed twice with brine, and then concentrated in vacuo. This oil was Kϋgelrohr distilled to a clear oil that crystallized upon cooling (8.53 g, 87.6%).

Step 3: 2.3-Dihydrobenzofuran-4-carboxaldehyde

DMSO (8.10 mL, 1 14 mmol) was added at -78°C to a stirred solution of oxalyl chloride in CH2CI2 (40 mL of a 2M solution). A solution of (2,3- dihydrobenzofuran-4-yl)methanol (8.53 g, 56.9 mmol) in CH2CI2 (35 mL) was added dropwise, and the solution stirred at -78°C for 30 min. Triethyl amine (33 mL, 228 mmol) was added cautiously to quench the reaction. The resulting suspension was stirred at room temperature for 30 min and diluted with CH2CI2 (100 mL). The organic layer was washed three times with water, and twice with brine, and then concentrated in vacuo to an oil (8.42 g, 100%) that was used without purification.

Preparation 16

(±)-(trans)-2-(2,3-Dihyd robenzofuran-4-yl)cyclopropane- carboxaldehyde

Step 1 : (±Htrans)-N-Methoxy-N-methyl-2-(2.3-dihydrobenzofuran-4- yhcyclopropanecarboxamide

Trimethylsulfoxonium iodide (9.9 g, 45 mmol) was added in small portions to a suspension of sodium hydride (1 .8 g, 45 mmol) in DMF (120 mL). After the foaming had subsided (10 min), a solution of (trans)- N-methoxy-N-methyl-3-(2,3-dihydrobenzofuran-4-yl)propenamide (3.5 g, 15 mmol) in DMF (60 mL) was added dropwise, with the temperature maintained between 35-40°C. The mixture was stirred for 3 h at room temperature. Saturated NH4CI (50 mL) was added dropwise and the mixture was extracted three times with ethyl acetate. The organic extracts were combined, washed with H2O and brine, dried over K2CO3, and concentrated in vacuo to give a white wax (3.7 g, 100%).

Step 2: (±)-(trans)- 2-(2.3-Dihydrobenzofuran-4-yl)cyclopropane- carboxaldehyde

A solution of (±)-(trans)-N-methoxy-N-methyl-2-(2,3-dihydrobenzofuran- 4-yl)cyclopropanecarboxamide (3.7 g, 15 mmol) in THF (10 mL) was added dropwise to a rapidly stirred suspension of LAH (683 mg, 18 mmol) in THF (50 mL) at -45°C, maintaining the temperature below -40°C throughout. The cooling bath was removed, the reaction was allowed to warm to 5°C, and then the reaction was immediately recooled to -45°C. Potassium hydrogen sulfate (3.4 g, 25.5 mmol) in H20 (50 mL) was cautiously added dropwise, the temperature maintained below – 30°C throughout. The cooling bath was removed and the suspension was stirred at room temperature for 30 min. The mixture was filtered through Celite and the filter cake was washed with ether. The combined filtrates were then washed with cold 1 N HCI, 1 N NaOH, and brine. The filtrates were dried over MgSO4, and concentrated in vacuo to give a clear oil (2.6 g, 99%).

Preparation 18

(-)-(trans)-2-(2.3-Dihydrobenzofuran-4-yl)cyclopropane-carboxaldehyde

Step 1 : (-Htrans)-N-[3-(2.3-Dihvdrobenzofuran-4-yl)-propenoyll-2.10- camphorsultam

To a solution of (-)-2,10-camphorsultam (8.15 g, 37.9 mmol) in 50 mL toluene at 0°C was added sodium hydride (1.67 g, 41.7 mmol). After stirring for 0.33 h at 0°C and 0.5 h at 20°C and recooling to 0°C, a solution of 3-(2,3-dihydrobenzofuran-4-yl)-2-propenoyl chloride
(37.9 mmol), prepared in situ from the corresponding acid and thionyl chloride (75 mL), in toluene (50 mL), was added dropwise. After stirring for 18 h at 20°C, the mixture was diluted with ethyl acetate and washed with water, 1 N HCI, and 1 N NaOH. The organic solution was dried and concentrated in vacuo to give 15.8 g of crude product. Recrystallization form ethanol-methanol (600 mL, 1 :1) gave the product (13.5 g, 92%, mp 199.5-200°C).

Step 2: (-)-N-[[(trans)-2-(2,3-Dihydrobenzofuran-4-yl)-cyclopropylj- carbonylj-2, 10-camphorsultam

1 -Methyl-3-nitro-1 -nitrosoguanidine (23.88g 163 mmol) was added in portions to a mixture of 10 N sodium hydroxide (60 mL) and ether (200 mL) at 0°C. The mixture was shaken vigorously for 0.25 h and the ether layer carefully decanted into a solution of (-)-N-[3-(2,3-dihydrobenzofuran-4-yl)-2-propenoyl]-2,10-camphorsultam (9.67 g, 25 mmol) and palladium acetate (35 mg) in methylene chloride (200 mL). After stirring for 18 h, acetic acid (5 mL) was added to the reaction and the mixture stirred for 0.5 h. The mixture was washed with 1 N HCI, 1 N NaOH and brine. The solution was dried, concentrated in vacuo and the residue crystallized twice from ethanol to give the product (6.67 g, 66.5%, mp 157-159°C).

Step 3: (-)-(trans)-2-(2,3-Dihydrobenzofuran-4-yl)cyclopropane- methanol

A solution of (-)-N-[(trans)-2-(2,3-dihydrobenzofuran-4-yl)cyclo-propanecarbonylj-2,10-camphorsultam (4.3 g, 10.7 mmol) in THF (50 mL) was added dropwise to a mixture of LAH (0.81 g, 21.4 mmol) in THF (50 mL) at -45°C. The mixture was stirred for 2 hr while it warmed to 10°C. The mixture was recooled to -40°C and hydrolyzed by the addition of saturated KHS0 (20 mL). The mixture was stirred at room temperature for 30 minutes and filtered. The precipitate was washed twice with acetone. The combined filtrate and acetone washes were concentrated in vacuo. The gummy residue was dissolved in ether, washed with 1 N NaOH and 1 N HCI, and then dried in vacuo to give the product (2.0 g, 98.4%).

Step 4: (-)-(trans)-2-(2.3-Dihydrobenzofuran-4-yl)cyclopropane- carboxaldehyde DMSO (1.6 g, 21 mmol) was added to oxalyl chloride in CH2CI2(7.4 mL of 2 M solution, 14.8 mmole) at -78°C. The (-)-(trans)-2-(2,3-dihydrobenzofuran-4-yl)-cyclopropylmethanol (2.0 g, 10.5 mmol) in CH2CI2(15 mL) was added. The mixture was stirred for 20 min and then triethylamine (4.24 g, 42 mmol) was added. The mixture was warmed to room temperature and stirred for 30 min. The mixture was diluted with CH2CI2 and washed with water, 1 N HCI, and then 1 N NaOH. The organic layer was dried and concentrated iι> vacuo to give the aldehyde product (1.98 g, 100%).

Preparation 24

(-)-(trans)-2-(2.3-Dihydrobenzofuran-4-yl)cyclopropane-methanamine A mixture of (-)-(trans)-2-(2,3-dihydrobenzofuran-4-yl)cyclopropane-carboxaldehyde (1.98 g, 10.5 mmol), hydroxylamine hydrochloride (2.29 g, 33 mmol), and 30% NaOH (3.5 mL, 35 mmol), in 5:1
ethanol/water (50 mL) was heated on a steam bath for 2 h. The solution was concentrated in vacuo. and the residue mixed with water. The mixture was extracted with CH2CI2. The organic extracts were dried and concentrated in vacuo to give a solid which NMR analysis showed to be a mixture of the cis and trans oximes. This material was dissolved in THF (20 mL) and added to solution of alane in THF [prepared from LAH (1.14 g, 30 mmol) and H2S04 (1.47 g, 15 mmol) at 0°Cj. The reaction was stirred for 18 h, and quenched successively with water (1.15 mL), 15% NaOH (1.15 mL), and then water (3.45 mL). The mixture was filtered and the filtrate was concentrated in vacuo. The residue was mixed with ether and washed with water and then 1 N HCI. The acid washes were made basic and extracted with CH2CI . The extracts were dried and concentrated in vacuo to give the amine product (1.4 g, 70.5%). The amine was converted to the fumarate salt in ethanol (mp: 197-198°C).
Anal. Calc’d for C12H15NO • C4H404: C, 62.94; H, 6.27; N, 4.59.
Found: C, 62.87; H, 6.31 ; N, 4.52.

FINAL PRODUCT TASIMELTEON

Example 2

(-)-(trans)-N-[[2-(2,3-Dihydrobenzofuran-4-yl)cycloprop-1-yl]methyl]propanamide

This compound was prepared similar to the above procedure using propionyl chloride and (-)-(trans)-2-(2,3-dihydrobenzofuran-4-yl)- cyclopropanemethanamine to give an oil that solidified upon standing to an off-white solid (61 %, mp: 71-72°C). IR (NaCI Film): 3298, 1645, 1548, 1459, 1235 cm“1.

Mo5 : -17.3°

Anal. Calc’d for C15H19N02: C, 73.44; H, 7.87; N, 5.71 . Found: C, 73.28; H, 7.68; N, 5.58

SYNTHESIS

Synthesis Path

SYN

Tasimelteon (Hetlioz)Tasimelteon, which is marketed by Vanda Pharmaceuticals as Hetlioz and developed in partnership with Bristol-Myers Squibb,is a drug that was approved by the US FDA in January 2014 for the treatment of non-24-hour sleep–wake disorder (also called Non-24, N24 and N24HSWD).234 Tasimelteon is a melatonin MT1
and MT2 receptor agonist; because it exhibits a greater affinity to the MT2 receptor than MT1, is also known as Dual Melatonin
Receptor Agonist.234 Two randomized controlled trials (phases II
and III) demonstrated that tasimelteon improved sleep latency
and maintenance of sleep with a shift in circadian rhythms, and
therefore has the potential to treat patients with transient insomnia
associated with circadian rhythm sleep disorders.235 Preclinical
studies showed that the drug has similar phase-shifting properties
to melatonin, but with less vasoconstrictive effects.236 The most
likely scale preparation of the drug, much of which has been published
in the chemical literature, is described below in Scheme 44.
Activation of commercial bis-ethanol 250 with 2.5 equivalents
of the Vilsmeier salt 251 followed by treatment with base resulted
an intramolecular cyclization reaction with the proximal phenol
and concomitant elimination of the remaining imidate to deliver
the vinylated dihydrobenzofuran 252 in 76% yield.237 Interestingly,
this reaction could be performed on multi-kilogram scale, required
no chromatographic purification, and generated environmentallyfriendly
DMF and HCl as byproducts.237 Sharpless asymmetric
dihydroxylation of olefin 252 delivered diol 253 in 86% yield and
impressive enantioselectivity (>99% ee). This diol was then activated
with trimethylsilyl chloride and then treated with base to generate epoxide 254.238 Next, a modified Horner–Wadsworth–
Emmons reaction involving triethylphosphonoacetate (TEPA, 255)
was employed to convert epoxide 254 to cyclopropane 256.239
The reaction presumably proceeds through removal of the acidic
TEPA proton followed by nucleophilic attack at the terminal epoxide
carbon. The resulting alkoxide undergoes an intramolecular
phosphoryl transfer reaction resulting in an enolate, which then attacked the newly formed phosphonate ester in an SN2 fashion
resulting in the trans-cyclopropane ester, which was ultimately
saponified and re-acidified to furnish cyclopropane acid 256.239
Conversion of this acid to the corresponding primary amide preceded
carbonyl reduction with sodium borohydride. The resulting
amine was acylated with propionyl chloride to furnish tasimelteon
(XXXI) as the final product in 86% yield across the four-step
sequence.

PATENTS

US2010261786 10-15-2010 PREDICTION OF SLEEP PARAMETER AND RESPONSE TO SLEEP-INDUCING COMPOUND BASED ON PER3 VNTR GENOTYPE
US2009209638 8-21-2009 TREATMENT FOR DEPRESSIVE DISORDERS
US6060506 5-10-2000 Benzopyran derivatives as melatonergic agents
US5981571 11-10-1999 Benzodioxa alkylene ethers as melatonergic agents
WO9825606 6-19-1998 BENZODIOXOLE, BENZOFURAN, DIHYDROBENZOFURAN, AND BENZODIOXANE MELATONERGIC AGENTS
WO2007137244A1 * May 22, 2007 Nov 29, 2007 Gunther Birznieks Melatonin agonist treatment
US4880826 Jun 25, 1987 Nov 14, 1989 Nava Zisapel Melatonin antagonist
US4997845 May 10, 1990 Mar 5, 1991 Eli Lilly And Company β-alkylmelatonins as ovulation inhibitors
US5093352 May 16, 1990 Mar 3, 1992 Whitby Research, Inc. Antidepressant agents
US5151446 Mar 28, 1991 Sep 29, 1992 Northwestern University Substituted 2-amidotetralins as melatonin agonists and antagonists
US5225442 Jan 3, 1992 Jul 6, 1993 Adir Et Compagnie Compounds having a naphthalene structure
US5580878 Jun 7, 1995 Dec 3, 1996 Interneuron Pharmaceuticals, Inc. Substituted tryptamines phenalkylamines and related compounds
US5856529 Dec 9, 1997 Jan 5, 1999 Bristol-Myers Squibb Company Benzofuran and dihydrobenzofuran melatonergic agents
US6211225 Jun 6, 2000 Apr 3, 2001 Bristol-Meyers Squibb Heterocyclic aminopyrrolidine derivatives as melatonergic agents
US7754902 May 18, 2006 Jul 13, 2010 Vanda Pharmaceuticals, Inc. Ruthenium(II) catalysts for use in stereoselective cyclopropanations
US20010047016 Apr 12, 2001 Nov 29, 2001 Gregory Oxenkrug Method for treating depression
US20050164987 Dec 22, 2004 Jul 28, 2005 Barberich Timothy J. Melatonin combination therapy for improving sleep quality
US20090105333 May 22, 2007 Apr 23, 2009 Gunther Birznieks Melatonin agonist treatment

extra info

Org. Synth.199069, 154
(−)-D-2,10-CAMPHORSULTAM
[3H-3a,6-Methano-2,1-benzisothiazole, 4,5,6,7-tetrahydro-8,8-dimethyl-2,2-dioxide, (3aS)-]
Submitted by Michael C. Weismiller, James C. Towson, and Franklin A. Davis1.
Checked by David I. Magee and Robert K. Boeckman, Jr..
1. Procedure
(−)-2,10-Camphorsultam. A dry, 2-L, three-necked, round-bottomed flask is equipped with a 1.5-in egg-shaped Teflon stirring bar, a 250-mL addition funnel, and a 300-mL Soxhlet extraction apparatus equipped with a mineral oil bubbler connected to an inert-gas source. The flask is charged with 600 mL of dry tetrahydrofuran (THF) (Note 1) and6.2 g (0.16 mol) of lithium aluminum hydride (Note 2). Into the 50-mL Soxhlet extraction thimble is placed 35.0 g (0.16 mol) of (−)-(camphorsulfonyl)imine (Note 3) and the reaction mixture is stirred and heated at reflux. After all of the(camphorsulfonyl)imine has been siphoned into the reaction flask (3–4 hr), the mixture is allowed to cool to room temperature. The unreacted lithium aluminum hydride is cautiously hydrolyzed by dropwise addition of 200 mL of 1 Nhydrochloric acid via the addition funnel (Note 4). After the hydrolysis is complete the contents of the flask are transferred to a 1-L separatory funnel, the lower, silver-colored aqueous layer is separated, and the upper layer placed in a 1-L Erlenmeyer flask. The aqueous phase is returned to the separatory funnel and washed with methylene chloride (3 × 100 mL). After the reaction flask is rinsed with methylene chloride (50 mL), the organic washings are combined with the THF phase and dried over anhydrous magnesium sulfate for 10–15 min. Filtration through a 300-mL sintered-glass funnel of coarse porosity into a 1-L round-bottomed flask followed by removal of the solvent on arotary evaporator gives 33.5 g (95%) of the crude (−)-2,10-camphorsultam. The crude sultam is placed in a 250-mL Erlenmeyer flask and crystallized from approximately 60 mL of absolute ethanol. The product is collected on a 150-mL sintered-glass funnel of coarse porosity and dried in a vacuum desiccator to give 31.1 g (88%) of the pure sultam. A second crop of crystals can be gained by evaporating approximately half the filtrate; the residue is crystallized as above to give 1.4 g (4%). The combined yield of white crystalline solid, mp 183–184°C, [α]D −30.7° (CHCl3, c 2.3) is92% (Note 5) and (Note 6).
2. Notes
1. Tetrahydrofuran (Aldrich Chemical Company, Inc.) was distilled from sodium benzophenone.
2. Lithium aluminum hydride was purchased from Aldrich Chemical Company, Inc.
3. (−)-(Camphorsulfonyl)imine, [(7S)-(−)-10,10-dimethyl-5-thia-4-azatricyclo[5.2.1.03,7]dec-3-ene 5,5-dioxide] was prepared by the procedure of Towson, Weismiller, Lal, Sheppard, and Davis, Org. Synth., Coll. Vol. VIII1993, 104.
4. The addition must be very slow at first (1 drop/5 sec) until the vigorous reaction has subsided.
5. The NMR spectrum of (−)-2,10-camphorsultam is as follows: 1H NMR (CDCl3) δ: 0.94 (s, 3 H, CH3), 1.14 (s, 3 H, CH3), 1.33 (m, 1 H), 1.47 (m,, 1 H), 1.80–2.05 (5 H), 3.09 (d, 1 H, J = 14), 3.14 (d, 1 H, J = 14), 3.43 (m, 1 H), 4.05 (br s, 1 H, NH); 13C NMR (CDCl3) δ: 20.17 (q, CH3), 26.51 (t), 31.55 (t), 35.72 (t), 44.44 (d), 47.15 (s), 50.08 (t), 54.46 (s), 62.48 (d).
6. Checkers obtained material having the same mp (183–184°C) and [α]D − 31.8° (CHCl3c 2.3).
3. Discussion
(−)-2,10-Camphorsultam was first prepared by the catalytic hydrogenation of (−)-(camphorsulfonyl)imine overRaney nickel.2 Lithium aluminum hydride reduction was used by Oppolzer and co-workers in their synthesis of the sultam.3,4 However, because of the low solubility of the sultam in tetrahydrofuran, a large amount of solvent was required.4 In the procedure described here the amount of solvent is significantly reduced by using a Soxhlet extractor to convey the imine slowly into the reducing medium.5
Oppolzer’s chiral auxiliary,6 (−)-2,10-camphorsultam, is useful in the asymmetric Diels–Alder reaction,3,4 and for the preparation of enantiomerically pure β-substituted carboxylic acids7 and diols,8 in the stereoselective synthesis of Δ2-isoxazolines,9 and in the preparation of N-fluoro-(−)-2,10-camphorsultam, an enantioselective fluorinating reagent.10

References and Notes
  1. Department of Chemistry, Drexel University, Philadelphia, PA 19104.
  2. Shriner, R. L.; Shotton, J. A.; Sutherland, H. J. Am. Chem. Soc.193860, 2794.
  3. Oppolzer, W.; Chapuis, C.; Bernardinelli, G. Helv. Chim. Acta198467, 1397.
  4. Vandewalle, M.; Van der Eycken, J.; Oppolzer, W.; Vullioud, C. Tetrahedron198642, 4035.
  5. Davis, F. A.; Towson, J. C.; Weismiller, M. C.; Lal, G.; Carroll,, P. J. J. Am. Chem. Soc.1988110, 8477.
  6. Oppolzer, W. Tetrahedron198743, 1969.
  7. Oppolzer, W.; Mills, R. J.; Pachinger, W.; Stevenson, T. Helv. Chim. Acta198669, 1542; Oppolzer, W.; Schneider, P. Helv. Chim. Acta198669, 1817; Oppolzer, W.; Mills, R. J.; Réglier, M. Tetrahedron Lett.198627, 183; Oppolzer, W.; Poli. G.Tetrahedron Lett.198627, 4717; Oppolzer, W.; Poli, G.; Starkemann, C.; Bernardinelli, G. Tetrahedron Lett.198829, 3559.
  8. Oppolzer, W.; Barras, J-P. Helv. Chim. Acta198770, 1666.
  9. Curran, D. P.; Kim, B. H.; Daugherty, J.; Heffner, T. A. Tetrahedron Lett.198829, 3555.
  10. Differding, E.; Lang, R. W. Tetrahedron Lett.198829, 6087.

Org. Synth.199069, 158
(+)-(2R,8aS)-10-(CAMPHORYLSULFONYL)OXAZIRIDINE
[4H-4A,7-Methanooxazirino[3,2-i][2,1]benzisothiazole, tetrahydro-9,9-dimethyl-, 3,3-dioxide, [4aS-(4aα,7α,8aR*)]]
Submitted by James C. Towson, Michael C. Weismiller, G. Sankar Lal, Aurelia C. Sheppard, Anil Kumar, and Franklin A. Davis1.
Checked by David I. Magee and Robert K. Boeckman, Jr..
1. Procedure
A. (+)-(1S)-10-Camphorsulfonamide. Into a 2-L, two-necked, round-bottomed flask, equipped with a 250-mL dropping funnel, a magnetic stirring bar, and a reflux condenser fitted with an outlet connected to a disposable pipettedipped in 2 mL of chloroform in a test tube for monitoring gas evolution, were placed 116 g (0.5 mol) ofcamphorsulfonic acid (Note 1) and 750 mL of reagent-grade chloroform. The suspension of camphorsulfonic acid was heated to reflux and 71.4 g (43.77 mL, 0.6 mol, 1.2 equiv) of freshly distilled thionyl chloride was added dropwise over a 1-hr period. Heating was continued until gas evolution (sulfur dioxide and hydrogen chloride) had ceased (approximately 9–10 hr). The resultant solution of camphorsulfonyl chloride in chloroform was converted tocamphorsulfonamide without further purification.
In a 5-L, two-necked, round-bottomed flask fitted with a 250-mL dropping funnel and a mechanical stirrer was placed a solution of 1.6 L of reagent-grade ammonium hydroxide solution and the flask was cooled to 0°C in an ice bath. The solution of the crude camphorsulfonyl chloride, prepared in the preceding section, was added dropwise to the ammonium hydroxide solution at 0–10°C over a period of 1 hr. The reaction mixture was warmed to room temperature, stirred for 4 hr, the organic layer separated, and the aqueous layer was extracted with methylene chloride (3 × 250 mL). The combined organic layers were washed with brine (250 mL) and dried over anhydrousmagnesium sulfate. Removal of the solvent on the rotary evaporator gave 104.0 g (90%) of the crudecamphorsulfonamide (Note 2) and (Note 3).
B. (−)-(Camphorsulfonyl)imine. A 1-L, round-bottomed flask is equipped with a 2-in. egg-shaped magnetic stirring bar, a Dean–Stark water separator, and a double-walled condenser containing a mineral oil bubbler connected to an inert gas source. Into the flask are placed 5 g of Amberlyst 15 ion-exchange resin (Note 4) and 41.5 g of the crude(+)-(1S)-camphorsulfonamide in 500 mL of toluene. The reaction mixture is heated at reflux for 4 hr. After the reaction flask is cooled, but while it is still warm (40–50°C), 200 mL of methylene chloride is slowly added to dissolve any(camphorsulfonyl)imine that crystallizes. The solution is filtered through a 150-mL sintered glass funnel of coarse porosity an the reaction flask and filter funnel are washed with an additional 75 mL of methylene chloride.
Isolation of the (−)-(camphorsulfonyl)imine is accomplished by removal of the toluene on the rotary evaporator. The resulting solid is recrystallized from absolute ethanol (750 mL) to give white crystals, 34.5–36.4 g (90–95%), mp225–228°C; [α]D −32.7° (CHCl3, c 1.9) (Note 5).
C. (+)-(2R, 8aS)-10-Camphorylsulfonyloxaziridine. A 5-L, three-necked, round-bottomed Morton flask is equipped with an efficient mechanical stirrer, a 125-mm Teflon stirring blade, a Safe Lab stirring bearing (Note 6), and a 500-mL addition funnel. Into the flask are placed the toluene solution of (−)-(camphorsulfonyl)imine (39.9 g, 0.187 mol)prepared in Step B and a room-temperature solution of 543 g (3.93 mol, 7 equiv based on oxone) of anhydrouspotassium carbonate dissolved in 750 mL of water. The reaction mixture is stirred vigorously and a solution of 345 g (0.56 mol, 6 equiv of KHSO5) of oxone dissolved in 1250 mL of water is added dropwise in three portions over 45 min(Note 7) and (Note 8). Completion of the oxidation is determined by TLC (Note 9) and the reaction mixture is filtered through a 150-mL sintered-glass funnel of coarse porosity to remove solids. The filtrate is transferred to a 3-L separatory funnel, the toluene phase is separated and the aqueous phase is washed with methylene chloride (3 × 100 mL). The filtered solids and any solids remaining in the Morton flask are washed with an additional 200 mL of methylene chloride. The organic extracts are combined and washed with 100 mL of saturated sodium sulfite, dried over anhydrousmagnesium sulfate for 15–20 min, filtered, and concentrated on the rotary evaporator. The resulting white solid is crystallized from approximately 500 mL of hot 2-propanol to afford, after drying under vacuum in a desiccator, 35.9 g(84%) of white needles, mp 165–167°C, [α]D +44.6° (CHCl3, c 2.2) (Note 10) and (Note 11).
(−)-(2S,8aR)-10-(camphorylsulfonyl)oxaziridine is prepared in a similar manner starting from (−)-10-camphorsulfonic acid; mp 166–167°C, [α]D +43.6° (CHCl3, c 2.2).
2. Notes
1. (1S)-(+)-10-Camphorsulfonic acid was purchased from Aldrich Chemical Company, Inc.
2. The crude sulfonamide is contaminated with 5–10% of the (camphorsulfonyl)imine, the yield of which increases on standing.
3. The 1H NMR spectrum of (+)-(1S)-10-camphorsulfonamide is as follows: (CDCl3) δ: 0.93 (s, 3 H, CH3), 1.07 (s, 3 H, CH3), 1.40–2.50 (m, 7 H), 3.14 and 3.53 (AB quartet, 2 H, CH2-SO2J = 15.1), 5.54 (br s, 2 H, NH2).
4. Amberlyst 15 ion-exchange resin is a strongly acidic, macroreticular resin purchased from Aldrich Chemical Company, Inc.
5. The spectral properties of (−)-(camphorsulfonyl)imine are as follows: 1H NMR (CDCl3) δ: 1.03 (s, 3 H, CH3), 1.18 (s, 3 H, CH3), 1.45–2.18 (m, 6 H), 2.65 (m, 1 H), 3.10 and 3.28 (AB quartet, 2 H, CH2-SO2J = 14.0); 13C NMR (CDCl3) δ: 19.01 (q, CH3), 19.45 (q, CH3), 26.64 (t), 28.44 (t), 35.92 (t), 44.64 (d), 48.00 (s), 49.46 (t), 64.52 (s), 195.52 (s); IR (CHCl3) cm−1: 3030, 2967, 1366. Checkers obtained material having identical melting point and [α]D−32.3° (CHCl3, c 1.8).
6. The SafeLab Teflon bearing can be purchased from Aldrich Chemical Company, Inc. A glass stirring bearing lubricated with silicone grease is unsatisfactory because the dissolved salts solidify in the shaft, causing freezing.
7. Efficient stirring is important and indicated by a milky white appearance of the solution.
8. Occasionally batches of oxone purchased from Aldrich Chemical Company, Inc., have exhibited reduced reactivity in this oxidation. Oxone exposed to moisture prior to use also gives reduced reactivity in this oxidation. If this occurs, oxone is added until oxidation is complete as determined by TLC (Note 9). Potassium carbonate is added as needed to maintain the pH at approximately 9.0. Oxone stored in the refrigerator under an inert atmosphere has shown no loss in reactivity for up to 6 months.
9. Oxidation is generally complete after addition of the oxone solution. The oxidation is monitored by TLC as follows. Remove approximately 0.5 mL of the toluene solution from the nonstirring solution, spot a 250-μm TLC silica gel plate, elute with methylene chloride, and develop with 10% molybdophosphoric acid in ethanol and heating(camphorsulfonyl)imine Rf = 0.28 and (camphorylsulfonyl)oxaziridine Rf = 0.62. If (camphorsulfonyl)imine is detected, stirring is continued at room temperature until the reaction is complete (see (Note 8)). If the reaction mixture takes on a brownish color after addition of oxone and has not gone to completion after 30 min, the reaction mixture is filtered through a 150-mL sintered-glass funnel of coarse porosity, and the solids are washed with 50 mL of methylene chloride. The aqueous/organic extracts are returned to the 5-L Morton flask and stirred vigorously and 52 g (0.08 mol, 1 equiv KHSO5) of oxone is added over 5 min and stirring continued until oxidation is complete (approximately 10–15 min).
10. The submitters employed a toluene solution of crude imine prepared in Part B and obtained somewhat higher yields (90–95%). However, the checkers obtained yields in this range on one half the scale using isolatedsulfonylimine.
11. The spectral properties of (+)-(camphorsulfonyl)oxaziridine are as follows: 1H NMR (CDCl3) δ: 1.03 (s, 3 H, CH3), 1.18 (s, 3 H, CH3), 1.45–2.18 (m, 6 H), 2.65 (d, 1 H), 3.10 and 3.28 (AB quartet, 2 H, CH2-SO2J = 14.0); 13C NMR (CDCl3) δ: 19.45 (q, CH3), 20.42 (q, CH3), 26.55 (t), 28.39 (t), 33.64 (t), 45.78 (d), 48.16 (s), 48.32 (t), 54.07 (s), 98.76 (s). The checkers obtained material (mp 165–167°C) having [α]D +44.7° (CHCl3, c 2.2).
3. Discussion
Camphorsulfonamide, required for the preparation of the (camphorsulfonyl)imine, was previously prepared in two steps. The first step involved conversion of camphorsulfonic acid to the sulfonyl chloride with PCl5 or SOCl2. The isolated sulfonyl chloride was converted in a second step to the sulfonamide by reaction with ammonium hydroxide. This modified procedure is more efficient because it transforms camphorsulfonic acid directly to camphorsulfonamide, avoiding isolation of the camphorsulfonyl chloride.
(Camphorsulfonyl)imine has been reported as a by-product of reactions involving the camphorsulfonamide.2,3,4,5Reychler in 1898 isolated two isomeric camphorsulfonamides,2 one of which was shown to be the(camphorsulfonyl)imine by Armstrong and Lowry in 1902.3 Vandewalle, Van der Eycken, Oppolzer, and Vullioud described the preparation of (camphorsulfonyl)imine in 74% overall yield from 0.42 mol of the camphorsulfonyl chloride.6 The advantage of the procedure described here is that, by using ammonium hydroxide, the camphorsulfonyl chloride is converted to the sulfonamide in >95% yield.7 The sulfonamide is of sufficient purity that it can be used directly in the cyclization step, which, under acidic conditions, is quantitative in less than 4 hr. These modifications result in production of the (camphorsulfonyl)imine in 86% overall yield from the sulfonyl chloride.
In addition to the synthesis of enantiomerically pure (camphorylsulfonyl)oxaziridine7 and its derivatives,8 the(camphorsulfonyl)imine has been used in the preparation of (−)-2,10-camphorsultam (Oppolzers’ auxiliary),6,9 (+)-(3-oxocamphorysulfonyl) oxaziridine,10 and the N-fluoro-2,10-camphorsultam, an enantioselective fluorinating reagent.11
The N-sulfonyloxaziridines are an important class of selective, aprotic oxidizing reagents.121314 Enantiomerically pure N-sulfonyloxaziridines have been used in the asymmetric oxidation of sulfides to sulfoxides (30–91% ee),15selenides to selenoxides (8–9% ee).16 disulfides to thiosulfinates (2–13% ee),5 and in the asymmetric epoxidation of alkenes (19–65% ee).17,18 Oxidation of optically active sulfonimines (R*SO2N=CHAr) affords mixtures of N-sulfonyloxaziridine diastereoisomers requiring separation by crystallization and/or chromatography.3
(+)-(Camphorylsulfonyl)oxaziridine described here is prepared in four steps from inexpensive (1S)-(+)- or (1R)-(+)-10-camphorsulfonic acid in 77% overall yield.7 Separation of the oxaziridine diastereoisomers is not required because oxidation is sterically blocked from the exo face of the C-N double bond in the (camphorsulfonyl)imine. In general, (camphorsulfonyl)oxaziridine exhibits reduced reactivity compared to other N-sulfonyloxaziridines. For example, while sulfides are asymmetrically oxidized to sulfoxides (3–77% ee), this oxaziridine does not react with amines or alkenes.7 However, this oxaziridine is the reagent of choice for the hydroxylation of lithium and Grignard reagents to give alcohols and phenols because yields are good to excellent and side reactions are minimized.19 This reagent has also been used for the stereoselective oxidation of vinyllithiums to enolates.20
The most important synthetic application of the (camphorylsulfonyl)oxaziridines is the asymmetric oxidation of enolates to optically active α-hydroxy carbonyl compounds.14,21,22,23,24 Chiral, nonracemic α-hydroxy carbonylcompounds have been used extensively in asymmetric synthesis, for example, as chiral synthons, chiral auxiliaries, and chiral ligands. This structural array is also featured in many biologically active natural products. This oxidizing reagent gives uniformly high chemical yields regardless of the counterion, and stereoselectivities are good to excellent (50–95% ee).9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24 Since the configuration of the oxaziridine three-membered ring controls the stereochemistry, both α-hydroxy carbonyl optical isomers are readily available. Representative examples of the asymmetric oxidation of prochiral enolates by (+)-(2R,8aS)-camphorylsulfonyl)oxaziridine are given in Tables I and II.

This preparation is referenced from:

  • Org. Syn. Coll. Vol. 8, 110
  • Org. Syn. Coll. Vol. 9, 212
  • References and Notes
    1. Department of Chemistry, Drexel University, Philadelphia, PA 19104.
    2. Reychler, M. A. Bull. Soc. Chim. III188919, 120.
    3. Armstrong, H. E.; Lowry, T. M. J. Chem. Soc., Trans.190281, 1441.
    4. Dauphin, G.; Kergomard, A.; Scarset, A. Bull. Soc. Chim. Fr.1976, 862.
    5. Davis, F. A.; Jenkins, Jr., R. H.; Awad, S. B.; Stringer, O. D.; Watson, W. H.; Galloy, J. J. Am. Chem. Soc.1982104, 5412.
    6. Vandewalle, M.; Van der Eycken, J.; Oppolzer, W.; Vullioud, C. Tetrahedron198642, 4035.
    7. Davis, F. A.; Towson, J. C.; Weismiller, M. C.; Lal, S.; Carroll, P. J. J. Am. Chem. Soc.1988110, 8477.
    8. Davis, F. A.; Weismiller, M. C.; Lal, G. S.; Chen, B. C.; Przeslawski, R. M. Tetrahedron Lett.198930, 1613.
    9. Oppolzer, W. Tetrahedron198743, 1969.
    10. Glahsl, G.; Herrmann, R. J. Chem. Soc., Perkin Trans. I1988, 1753.
    11. Differding, E.; Lang, R. W. Tetrahedron Lett.198829, 6087.
    12. For recent reviews on the chemistry of N-sulfonyloxaziridines, see: (a) Davis, F. A.; Jenkins, Jr., R. H. in “Asymmetric Synthesis,” Morrison, J. D., Ed.; Academic Press: Orlando, FL, 1984, Vol. 4, Chapter 4;
    13. Davis, F. A.; Haque, S. M. in “Advances in Oxygenated Processes,” Baumstark, A. L., Ed.; JAI Press: London, Vol. 2;
    14. Davis, F. A.; Sheppard, A. C. Tetrahedron198945, 5703.
    15. Davis, F. A.; McCauley, Jr., J. P.; Chattopadhyay, S.; Harakal, M. E.; Towson, J. C.; Watson, W. H.; Tavanaiepour, I. J. Am. Chem. Soc.1987109, 3370.
    16. Davis, F. A.; Stringer, O. D.; McCauley, Jr., J. M. Tetrahedron198541, 4747.
    17. Davis, F. A.; Chattopadhyay, S. Tetrahedron Lett.198627, 5079.
    18. Davis, F. A.; Harakal, M. E.; Awad, S. B. J. Am. Chem. Soc.1983105, 3123.
    19. Davis, F. A.; Wei, J.; Sheppard, A. C.; Gubernick S. Tetrahedron Lett.198728, 5115.
    20. Davis, F. A.; Lal, G. S.; Wei, J. Tetrahedron Lett.198829, 4269.
    21. Davis, F. A.; Haque, M. S.; Ulatowski, T. G.; Towson, J. C. J. Org. Chem.198651, 2402.
    22. Davis, F. A.; Haque, M. S. J. Org. Chem.198651, 4083; Davis, F. A.; Haque, M. S.; Przeslawski, R. M. J. Org. Chem.198954, 2021.
    23. Davis, F. A.; Ulatowski, T. G.; Haque, M. S. J. Org. Chem.198752, 5288.
    24. Davis, F. A.; Sheppard, A. C., Lal, G. S. Tetrahedron Lett.198930, 779.
    25. Davis, F. A.; Sheppard, A. C.; Chen, B. C.; Haque, M. S. J. Am. Chem. Soc.1990112, 6679.

a US 5 856 529 (Bristol-Myers Squibb; 5.1.1999; appl. 9.12.1997; USA-prior. 10.12.1996).

    • b US 7 754 902 (Vanda Pharms.; 13.7.2010; appl. 18.5.2006).
  • treatment of circadian rhythm disorders:

    • US 8 785 492 (Vanda Pharms.; 22.7.2014; appl. 25.1.2013; USA-prior. 26.1.2012).
  • synthesis cis-isomer:

    • US 6 214 869 (Bristol-Myers Squibb; 10.4.2001; appl. 25.5.1999; USA-prior. 5.6.1998).

Patents

  1. USUS5856529 A
  2. USUS8785492 B2
  3. US5856529
  4. US8785492
  5. US9060995
  6. US9549913
  7. US9539234
  8. US9730910
  9. USRE46604
  10. US9855241

References

  1. Jump up^ “Tasimelteon Advisory Committee Meeting Briefing Materials”(PDF). Vanda Pharmaceuticals Inc. November 2013.
  2. Jump up^ “FDA transcript approval minutes” (PDF). FDA. November 14, 2013.
  3. Jump up to:a b Food and Drug Administration (January 31, 2014). “FDA approves Hetlioz: first treatment for non-24 hour sleep-wake disorder”. FDA.
  4. Jump up^ “tasimelteon (Hetlioz) UKMi New Drugs Online Database”. Retrieved August 6, 2014.
  5. Jump up^ “HETLIOZ® Receives European Commission Approval for the Treatment of Non-24-Hour Sleep-Wake Disorder in the Totally Blind”MarketWatch. PR Newswire. 7 July 2015. Retrieved 8 July 2015.
  6. Jump up^ Vachharajani, Nimish N.; Yeleswaram, Krishnaswamy; Boulton, David W. (April 2003). “Preclinical pharmacokinetics and metabolism of BMS-214778, a novel melatonin receptor agonist”. Journal of Pharmaceutical Sciences92 (4): 760–72. doi:10.1002/jps.10348PMID 12661062.
  7. Jump up^ Sack, R. L.; Brandes, R. W.; Kendall, A. R.; Lewy, A. J. (2000). “Entrainment of Free-Running Circadian Rhythms by Melatonin in Blind People”. New England Journal of Medicine343 (15): 1070–7. doi:10.1056/NEJM200010123431503PMID 11027741.
  8. Jump up^ “Safety and Efficacy of VEC-162 on Circadian Rhythm in Healthy Adult Volunteers”. ClinicalTrials.gov. |accessdate=May 15, 2014
  9. Jump up^ “VEC-162 Study in Healthy Adult Volunteers in a Model of Insomnia”. ClinicalTrials.gov. Retrieved May 15, 2014.
  10. Jump up^ “VEC-162 Study in Adult Patients With Primary Insomnia”. ClinicalTrials.gov. Retrieved May 15, 2014.
  11. Jump up^ Lynne Lamberg. “Improving Sleep and Alertness in the Blind (Part 5)”Matilda Ziegler Magazine for the Blind. Retrieved May 15, 2014.
  12. Jump up^ Shantha MW Rajaratnam; Mihael H Polymeropoulos; Dennis M Fisher; Thomas Roth; Christin Scott; Gunther Birznieks; Elizabeth B Klerman (2009-02-07). “Melatonin agonist tasimelteon (VEC-162) for transient insomnia after sleep-time shift: two randomised controlled multicentre trials”The Lancet373 (9662): 482–491. doi:10.1016/S0140-6736(08)61812-7PMID 19054552. Retrieved 2010-02-23.
  13. Jump up^ Carome, Michael (1 July 2015). “Outrage of the Month: FDA Makes Major Blunder After Approving Drug for Rare Sleep Disorder”Huffington Post. Retrieved 8 July 2015.
  14. Jump up^ Food and Drug Administration (January 31, 2014). “FDA NEWS RELEASE: FDA approves Hetlioz: first treatment for non-24 hour sleep–wake disorder in blind individuals”. FDA.
  15. Jump up^ “Side Effects Drug Center: Hetlioz Clinical Pharmacology”. RxList. February 10, 2014.
  16. Jump up^ “Side Effects Drug Center: Hetlioz Warnings and Precautions”. RxList. February 10, 2014. In animal studies, administration of tasimelteon during pregnancy resulted in developmental toxicity (embryofetal mortality, neurobehavioral impairment, and decreased growth and development in offspring) at doses greater than those used clinically.
Tasimelteon
Tasimelteon 2.svg
Tasimelteon ball-and-stick model.png
Clinical data
Trade names Hetlioz
License data
Pregnancy
category
  • US:C (Risk not ruled out)
Routes of
administration
Oral
ATC code
Legal status
Legal status
Pharmacokinetic data
Bioavailability not determined in humans[1]
Protein binding 89–90%
Metabolism extensive hepatic, primarily CYP1A2 and CYP3A4-mediated
Elimination half-life 0.9–1.7 h / 0.8–5.9 h (terminal)
Excretion 80% in urine, 4% in feces
Identifiers
CAS Number
PubChemCID
IUPHAR/BPS
ChemSpider
UNII
ChEBI
ECHA InfoCard 100.114.889Edit this at Wikidata
Chemical and physical data
Formula C15H19NO2
Molar mass 245.32 g/mol
3D model (JSmol)

ANTHONY MELVIN CRASTO

DR ANTHONY MELVIN CRASTO Ph.D

amcrasto@gmail.com

MOBILE-+91 9323115463

GLENMARK SCIENTIST , NAVIMUMBAI, INDIA

//////////////BMS-214778, VEC-162, Tasimelteon, Hetlioz, FDA 2014, 609799-22-6 , BMS-214778, VEC-162, ATC N05CH03, タシメルテオン , EU 2015, VANDA, BMS, orphan drug designations
CCC(=O)NCC1CC1C2=C3CCOC3=CC=C2

Chemical and physical properties 

Tasimelteon has two stereogenic centers. Besides the medically used trans-1 R , 2 R isomer (in the picture above left), there are thus three further stereoisomers that do not arise in the synthesis.

Tasimelteon stereoisomerism.svg

Tasimelteon is a white to off-white crystalline non-hygroscopic substance, soluble in water at physiologically relevant pH levels and readily soluble in alcohols, cyclohexane and acetonitrile. The compound occurs in two crystal forms. It is an anhydrate melting at 74 ° C and a hemihydrate . [4] The hemihydrate is from about 35 ° C the water of hydration and converts thereby in the anhydrate form to. [4] The anhydrate crystallizes in a monoclinic lattice with the space group P 2 1 , and the hemihydrate crystallizes in a tetragonal lattice with the space group P 4 3 21 2. [4]

4  Kaihang Liu, Zhou Xinbo, Zhejing Xu, Bai Hongzhen, Jianrong Zhu Jianming Gu, Guping Tang, Liu Xingang, Hu Xiurong: anhydrate and hemihydrate of Tasimelteon: Synthesis, structure, and pharmacokinetic study in J. Pharm. Biomed. Anal. 151 (2018) 235-243, doi : 10.1016 / j.jpba.2017.12.035 .

Technetium (99mTc) tetrofosmin, テトロホスミンテクネチウム (99mTc)

$
0
0

99mTc-tetrofosmin structure.svg

Thumb

Technetium Tc-99m tetrofosmin.png

Technetium (99mTc) tetrofosmin, 99mTc-Tetrofosmin

テトロホスミンテクネチウム (99mTc)

Formula C36H80O10P4Tc
Molar mass 895.813 g/mol
CAS Number

UNII42FOP1YX93

2-[bis(2-ethoxyethyl)phosphanyl]ethyl-bis(2-ethoxyethyl)phosphane;technetium-98;dihydrate

Technetium Tc 99m tetrofosmin; Technetium Tc-99m tetrofosmin; TECHNETIUM TC-99M TETROFOSMIN KIT; Tc-99m tetrofosmin; Technetium-99 tetrofosmin; Technetium (99mTc) tetrofosmin

Title: Tetrofosmin
CAS Registry Number: 127502-06-1
CAS Name: 6,9-Bis(2-ethoxyethyl)-3,12-dioxa-6,9-diphosphatetradecane
Additional Names: ethylenebis[bis(2-ethoxyethyl)phosphine]
Manufacturers’ Codes: P53
Molecular Formula: C18H40O4P2
Molecular Weight: 382.46
Percent Composition: C 56.53%, H 10.54%, O 16.73%, P 16.20%
Literature References: Prepn: J. D. Kelly et al., EP 337654eidem, US 5045302 (1989, 1991 both to Amersham). Pharmacology and determn of radiochemical purity: idem et al., J. Nucl. Med. 34, 222 (1993). Clinical biodistribution: B. Higley et al., ibid. 30. Clinical trial as a myocardial perfusion imaging agent: B. L. Zaret et al., Circulation 91, 313 (1995).
Derivative Type: 99mTc-Complex
CAS Registry Number: 127455-27-0
Additional Names: 99mTc tetrofosmin; [99mTc(tetrofosmin)2O2]+
Manufacturers’ Codes: PPN1011
Trademarks: Myoview (GE Healthcare)
Molecular Formula: C36H80O10P499mTc

Technetium Tc-99m Tetrofosmin is a radiopharmaceutical consisting of tetrofosmin, composed of two bidentate diphosphine ligands chelating the metastable radioisotope technetium Tc-99 (99mTc), with potential imaging activity upon SPECT (single photon emission computed tomography). Upon administration, technetium Tc 99m tetrofosmin is preferentially taken up by, and accumulates in, myocardial cells. Upon imaging, myocardial cells can be visualized and changes in ischemia and/or perfusion can be detected.

Technetium Tc-99m tetrofosmin is a drug used in nuclear myocardial perfusion imaging. The radioisotope, technetium-99m, is chelated by two 1,2-bis[di-(2-ethoxyethyl)phosphino]ethane ligands which belong to the group of diphosphines and which are referred to as tetrofosmin. It is a lipophilic technetium phosphine dioxo cation that was formulated into a freeze-dried kit which yields an injection.[A31592] Technetium Tc-99m tetrofosmin was developed by GE Healthcare and FDA approved on February 9, 1996.

Technetium Tc-99m tetrofosmin is a drug used in nuclear myocardial perfusion imaging. The radioisotope, technetium-99m, is chelated by two 1,2-bis[di-(2-ethoxyethyl)phosphino]ethane ligands which belong to the group of diphosphines and which are referred to as tetrofosmin. It is a lipophilic technetium phosphine dioxo cation that was formulated into a freeze-dried kit which yields an injection.[1] Technetium Tc-99m tetrofosmin was developed by GE Healthcare and FDA approved on February 9, 1996.

Technetium (99mTc) tetrofosmin is a drug used in nuclear medicine cardiac imaging. It is sold under the brand name Myoview (GE Healthcare). The radioisotopetechnetium-99m, is chelated by two 1,2-bis[di-(2-ethoxyethyl)phosphino]ethane ligands which belong to the group of diphosphines and which are referred to as tetrofosmin.[1][2]

Image result for Technetium (99mTc) tetrofosmin synthesis

Tc-99m tetrofosmin is rapidly taken up by myocardial tissue and reaches its maximum level in approximately 5 minutes. About 66% of the total injected dose is excreted within 48 hours after injection (40% urine, 26% feces). Tc-99m tetrofosmin is indicated for use in scintigraphic imaging of the myocardium under stress and rest conditions. It is used to determine areas of reversible ischemia and infarcted tissue in the heart. It is also indicated to detect changes in perfusion induced by pharmacologic stress (adenosinelexiscandobutamine or persantine) in patients with coronary artery disease. Its third indication is to assess left ventricular function (ejection fraction) in patients thought to have heart disease. No contraindications are known for use of Tc-99m tetrofosmin, but care should be taken to constantly monitor the cardiac function in patients with known or suspected coronary artery disease. Patients should be encouraged to void their bladders as soon as the images are gathered, and as often as possible after the tests to decrease their radiation doses, since the majority of elimination is renal. The recommended dose of Tc-99m tetrofosmin is between 5 and 33 millicuries (185-1221 megabecquerels). For a two-dose stress/rest dosing, the typical dose is normally a 10 mCi dose, followed one to four hours later by a dose of 30 mCi. Imaging normally begins 15 minutes following injection.[3]

Image result for Technetium (99mTc) tetrofosmin synthesis

Amersham (formerly Nycomed Amersham , now GE Healthcare ) has developed and launched 99mTc-tetrofosmin (Myoview) as an injectable nuclear imaging agent for ischemic heart disease in several major territories and for use in detecting breast tumors

Technetium (99mTc) tetrofosmin is a drug used in nuclear medicine cardiac imaging. It is sold under the brand name Myoview (GE Healthcare). The radioisotope, technetium-99m, is chelated by two 1, 2-bis-[bis-(2-ethoxyethyl)phosphino] ethane ligands, which belong to the group of diphosphines and which are referred to as tetrofosmin and has the structural Formula 1 :

Formula 1

99mTc -based radiopharmaceuticals are commonly used in diagnostic nuclear medicine, especially for in vivo imaging (e.g. via immunoscintigraphy or radiolabeling). Usually cold kits are manufactured in advance in accordance with strict requirements of Good Manufacturing Practice (GMP) Guidelines, containing the chemical ingredients (e.g. 99mTc -coordinating ligands, preservatives) in lyophilized form. The radioactive isotope 99mTc (ti/2 = 6h) is added to those kits shortly before application to the patient via intravenous or subcutaneous injection.

Tc-99m tetrofosmin is rapidly taken up by myocardial tissue and reaches its maximum level in approximately 5 minutes. About 66% of the total injected dose is excreted within 48 hours after injection (40% urine, 26% feces). Tc-99m tetrofosmin is indicated for use in scintigraphic imaging of the myocardium under stress and rest conditions. It is used to determine areas of reversible ischemia and infarcted tissue in the heart. It is also indicated to detect changes in perfusion induced by pharmacologic stress (adenosine, lexiscan, dobutamine or persantine) in patients with coronary artery disease. Its third indication is to assess left ventricular function (ejection fraction) in patients thought to have heart disease. No contraindications are known for use of Tc-99m tetrofosmin, but care should be taken to constantly monitor the cardiac function in patients with known or suspected coronary artery disease. Patients should be encouraged to void their bladders as soon as the images are gathered, and as often as possible after the tests to decrease their radiation doses, since the majority of elimination is renal. The recommended dose of Tc-99m tetrofosmin is between 5 and 33 millicuries (185-1221 megabecquerels). For a two-dose stress/rest dosing, the typical dose is normally a 10 mCi dose, followed one to four hours later by a dose of 30 mCi. Imaging normally begins 15 minutes following injection.

99mTc -Tetrofosmin is also described to be useful for tumor diagnostics, in particular of breast cancer and parathyroid gland cancer, and for multidrug resistance (MDR) research.

US5045302 discloses 99mTc-coordinating diphosphine ligands (L), wherein one preferred example thereof is the ether functionalized diphosphine ligand l,2-bis[bis(2-ethoxy- ethyl)phosphino]ethane according to Formula 1, called tetrofosmin (“P53”), that forms a dimeric cationic technetium (V) dioxo phosphine complex, [TCO2L2] with 99mTc, useful as myocardial imaging agent. Example 1 of said patent described the process for preparing tetrofosmin by reacting ethyl vinyl ether, bis(diphosphino)ethane in the presence of a-azo-isobutyronitrile (AIBN) in a fischer pressure-bottle equipped with a teflon stirring bar followed by removal of volatile materials and non-distillable material obtained, as per below mentioned Scheme 1.

Scheme 1

Formula 2 Formula 3 Formula 1

CN 1184225 C discloses tetrofosmin salts containing chloride or bromide or aryl sulfonates as negatively charged counter ions, which can be used for the preparation of a 99mTc- Tetrofosmin radiopharmaceutical composition. According to this patent tetrofosmin hydrochloride is a viscous liquid. Own experiments of the inventors of the present invention revealed that the halide salts of tetrofosmin are hygroscopic oils, which are complicated to handle, e.g. when weighed. The oily and hygrospcopic

properties of tetrofosmin hydrochloride hampers its use in pharmaceutical preparations. Attempts to synthesize the subsalicylate salt of tetrofosmin failed because the starting material sulfosalicylic acid was not soluble in ether in the concentration specified in the patent (3.4 g in 15 ml).

WO2006/064175A1 discloses tetrofosmin was converted to tetrofosmin subsalicylate by reaction with 2.3 to 2.5 molar equivalents of 5-sulfosalicyclic acid at room temperature in ethanol, followed by recrystallisation from ethanol/ether.

WO2015/114002A1 relates to tetrafluoroborate salt of tetrafosmin and its process for the preparation thereof. Further this application also discloses one-vial and two vial kit formulation with tetrafluoroborate salt of tetrafosmin.

The article Proceedings of the International Symposium, 7th, Dresden, Germany, June 18-22, 2000 by Amersham Pharmacia Biotech UK Limited titled “The synthesis of [14C]tetrofosmin, a compound vital to the development of Myoview, Synthesis and Applications of Isotopically Labelled Compounds” disclosed a process for the preparation of tetrofosmin as per below mentioned Scheme 2:

Scheme 2

Formula 1A Formula 7

The starting material was bis(2- ethoxyethyl)benzylphosphine of Formula 4 . This was prepared from benzyl phosphonate, PhCH2P(0)(OEt)2 by reduction with lithium aluminium hydride to give the intermediate benzylphosphine, PhCH2PH2, followed by a photolysis reaction in the presence of ethyl vinyl ether to give compound of Formula 4. The compound of Formula 4 in acetonitrile was treated with dibromo[U-14C]ethane to give compound of Formula 6, further it was treated with excess of 30% aqueous sodium hydroxide in ethanol. The mixture was stirred at room temperature for 24 hours. The solvent was removed and the residue was treated with excess concentrated hydrochloric acid at 0°C. Aqueous work up gave compound of Formula 7. Then compound of Formula 7 in dry benzene was treated with hexachlorodisilane and hydrolysed with excess 30% aqueous sodium hydroxide at 0°C. Aqueous work up followed by flash column chromatography on silica gave [bisphosphinoethane- 1,2-14C]tetrofosmin of formula 1A.

The article Polyhedron (1995), 14(8), 1057-65, titled “Synthesis and characterization of Group 10 metal complexes with a new trifunctional ether phosphine. The X-ray crystal structures of bis[bis(2-ethoxyethyl)benzylphosphine]dichloronickel(II) and bis[bis(2-ethoxyethyl)benzylphosphine]chlorophenylnickel(II)” disclosed the process for the preparation of bis(2-ethoxyethyl)benzylphosphine as per below mentioned Scheme 3:

Scheme 3

Formula 8 Formula 9 Formula 4

The compound bis(2-ethoxyethyl)benzylphosphine of Formula 4 was prepared by first reduction of diethylbenzylphosphonate of Formula 8 using lithium aluminium hydride to obtain benzyl phosphine of Formula 9 followed by radical catalysed coupling reaction with ethyl vinyl ether carried out by using UV photolysis.

Tetrofosmin is extremely sensitive to atmospheric oxygen, which makes synthesis of the substance, as well as manufacturing and handling of the kit complicated as the substance has constantly to be handled in an oxygen free atmosphere.

High purity and stability under dry and controlled conditions are pivotal requirements for chemical compounds used as active ingredients in pharmaceuticals.

The processes disclosed in prior art for the preparation of compound of Formula 4 involves that coupling reaction of benzyl phosphine of Formula 9 with ethyl vinyl ether carried out by using photolytic conditions. Such technology is expensive as it requires separate instruments including isolated facility (to avoid the UV radiation exposure etc.), also it is not suitable for commercial scale production.

PATENT

WO-2018162964

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2018162964&tab=PCTDESCRIPTION&maxRec=1000

Example 1

Preparation of benzyl phosphine:

A mixture of lithium aluminium hydride (25 g) in methyl tertiary butyl ether (MTBE) (800 ml) was cooled to 0 to 5°C and added a solution of diethylbenzylphosphonate in methyl tertiary butyl ether (100 g in 200ml). The temperature of reaction mixture was raised to 25 to 30 °C and stirred for 14 to 16 hour. After completion of the reaction, the reaction mixture was cooled to 0 to 5°C and 6N hydrochloric acid was added slowly. Further raised the temperature of reaction mixture to 25 to 30 °C and stirred for 30-45 minutes. The layers were separated, the aqueous layer was extracted with MTBE (250ml) and the combined organic layer was washed with deoxygenated water. The organic layer was dried over sodium sulfate and concentrated to obtain the title compound as non-distillable liquid.

Example 2

Preparation of benzylbis(2-ethoxyethyl)phosphane:

To a mixture of benzyl phosphine (obtained from example 1) and vinyl ethyl ether (250 ml) in pressure RB flask was added a-azo-isobutyronitrile (AIBN) (1.5g). The resulting reaction mixture was maintained at 80 to 90°C for 14 to 16 hours. The mixture was cooled to 20 to 30°C and AIBN (0.5g) added, then continued to heat the reaction mixture at 80 to 90°C for 6 to 7 hours. After completion of the reaction, the reaction mixture was allowed to cool to room temperature and distilled under vacuum to obtain title compound as an oil (107 g).

Example 3

Preparation of Ethane- 1,2-diylbis (benzylbis(2-ethoxyethyl) phosphonium) bromide:

To a mixture of benzylbis(2-ethoxyethyl)phosphane 107.g) in acetonitrile (100ml) in pressure bottle was added 1, 2-dibromoethane (30.5 g). The reaction mixture was maintained at 80 to 90°C for 20 to 25 hours. After completion of the reaction, the reaction mass was cooled to room temperature and stirred for 45 to 60 minutes to obtain the solid. To the solid obtained was added methyl tertiary butyl ether (MTBE) (500ml) and stirred at room temperature for 2 to 3 hour. The reaction mass was filtered, washed with MTBE and suck dried. Further the filtered solid was heated in acetone (400ml) at 50 to 55°C for 2 to 3 hour. Then cooled the reaction mixture to room temperature, stirred, filtered and washed with acetone to obtain the title compound as white solid. (85g)

Example 4

Preparation of Ethane- 1, 2-diylbis (bis (2-ethoxy ethyl) phosphine oxide):

To a mixture of Ethane- 1,2-diylbis (benzylbis(2-ethoxyethyl) phosphonium) bromide (80g) in ethanol (480 ml) was added an aq. solution of sodium hydroxide ( 48g in 160 ml water) at room temperature. The reaction mass was maintained at 25 to 35°C for 10 to 12 hour. After completion of the reaction, the reaction mass was cone, under vacuum to obtained the residue. The residue was dissolved in deoxygenated water (400 ml) and washed with MTBE (400 ml x 2). The layers were separated, the aqueous layer was cooled to 10 to 20°C and 6N hydrochloric acid (200 ml) was added slowly. Then extracted the aqueous layer with dichloromethane (2000 ml), washed the organic layer with deoxygenated water (160 ml), dried the organic layer using sodium sulfate, filtered, and distilled under vacuum to obtain the residue. Further MTBE (160 ml x 2) was added to the residue and continued distillation under vacuum, degassed to obtain the solid. To the obtained solid, MTBE (400 ml) was added and heated at 45 to 50°C for 1-2 hour, further slowly cooled the reaction mass to 25 to 30°C, filtered the solid product. Again MTBE (400 ml) was added to the solid product and heated at 45 to 50°C for 1-2 hour, further slowly cooled the reaction mass to 25 to 30°C, filtered, washed with MTBE and dried under vacuum to obtain the title compound as white solid (32g).

Example 5

Preparation of tetrofosmin free base:

To a mixture of ethane- 1, 2-diylbis (bis (2-ethoxyethyl) phosphine oxide (18g) in toluene (180ml) in pressure RB flask argon/nitrogen gas was purged for 5 minute and hexachlorodisilane (30g) was added. The reaction mixture was heated to 80 to 90°C, stirred for 10 to 12 hour, further slowly cooled to -5 to 0°C and slowly added 30% aqueous sodium hydroxide solution (45g sodium hydroxide in 150 ml deoxygenated water) the temperature of reaction mixture was raised to 25 to 30°C and stirred for 1 to 2 hour. The layers were separated and the aq. layer was extracted with Toluene (180 ml). The combined organic layer was washed with deoxygenated water (180 ml). Further dried the organic layer using sodium sulfate, distilled under vacuum to obtain the residue of tetrofosmin free base (15.5g).

Example 6

Preparation of tetrofosmin disulfosalicylate salt:

To the residue of tetrofosmin free base (15.5g) was added an aq. solution of 5-sulfosalicylic acid dihydrate (21.6g in 75ml deoxygenated water) and stirred at 25 to 30°C for 25 to 30 minutes. Further heated the reaction mass to 55 to 60°C, stirred for 15 to 30 minute, slowly cooled the reaction mass to 10 to 15°C and stirred for 1-2 hour. Filtered, washed with chilled deoxygenated water, and dried under vacuum to obtain the title compound as white solid. (30g).

Example 7

Preparation of Form J of tetrofosmin disulfosalicylate salt:

An aq. solution of 5-sulfosalicylic acid dihydrate (21.6g in 75ml deoxygenated water) was added slowly into tetrofosmin free base (15.5g) and stirred at room temperature for 30 to 40 minutes. The temperature of reaction mixture was further raised to 50 to 60°C, stirred for 20 to 30 minute, cooled the reaction mass to 10 to 15°C and stirred for 1-2 hour. Filtered, washed with chilled deoxygenated water, and dried under vacuum to obtain the title compound.

PATENT

EP337654 ,

PATENT

US9549999

FDA Orange Book Patents

FDA Orange Book Patents: 1 of 1 (FDA Orange Book Patent ID)
Patent 9549999
Expiration Mar 10, 2030
Applicant GE HEALTHCARE
Drug Application
  1. N020372 (Prescription Drug: MYOVIEW 30ML. Ingredients: TECHNETIUM TC-99M TETROFOSMIN KIT)
  2. N020372 (Prescription Drug: MYOVIEW. Ingredients: TECHNETIUM TC-99M TETROFOSMIN KIT)

References

  1. Jump up^ Kelly JD, Alan M. Forster AM, Higley B, et al. (February 1993). “Technetium-99m-Tetrofosmin as a new radiopharmaceutical for myocardial perfusion imaging”Journal of Nuclear Medicine34 (2): 222–227. PMID 8429340.
  2. Jump up^ Elhendy A, Schinkel AF, et al. (December 2005). “Risk stratification of patients with angina pectoris by stress 99mTc-tetrofosmin myocardial perfusion imaging”Journal of Nuclear Medicine46 (12): 2003–2008. PMID 16330563.
  3. Jump up^ Myoview package insert. Arlington Heights, IL: GE Healthcare, 2006, Aug.
Technetium (99mTc) tetrofosmin
99mTc-tetrofosmin structure.svg
Clinical data
Routes of
administration
Intravenous
ATC code
Pharmacokinetic data
Bioavailability N/A
Identifiers
CAS Number
Chemical and physical data
Formula C36H80O10P4Tc
Molar mass 895.813 g/mol
Patent ID Title Submitted Date Granted Date
US9549999 RADIOPHARMACEUTICAL COMPOSITION
2010-09-23

External links

Myoview Prescribing Information Page

//////////99mTc-Tetrofosmin, Technetium (99mTc) tetrofosmin, テトロホスミンテクネチウム (99mTc)

CCOCCP(CCOCC)CCP(CCOCC)CCOCC.CCOCCP(CCOCC)CCP(CCOCC)CCOCC.O.O.[Tc]

A call to (green) arms: a rallying cry for green chemistry and engineering for CO2 capture, utilisation and storage

$
0
0

Green Chemistry International

Graphical abstract: A call to (green) arms: a rallying cry for green chemistry and engineering for CO2 capture, utilisation and storage

A call to (green) arms: a rallying cry for green chemistry and engineering for CO2 capture, utilisation and storage

 Author affiliations

Abstract

Chemists, engineers, scientists, lend us your ears… Carbon capture, utilisation, and storage (CCUS) is among the largest challenges on the horizon and we need your help. In this perspective, we focus on identifying the critical research needs to make CCUS a reality, with an emphasis on how the principles of green chemistry (GC) and green engineering can be used to help address this challenge. We identify areas where GC principles can readily improve the energy or atom efficiency of processes or reduce the environmental impact. Conversely, we also identify dilemmas where the…

View original post 115 more words


Efonidipine, エホニジピン

$
0
0

Efonidipine structure.svg

ChemSpider 2D Image | Efonidipine | C34H38N3O7P

Efonidipine.png

Efonidipine

  • Molecular FormulaC34H38N3O7P
  • Average mass631.655 Da
  • エホニジピン
  • CAS 111011-63-3; FREE FORM
(±)-Efonidipine
Image result for Efonidipine
Molecular Formula: C36H45ClN3O8P
Molecular Weight: 714.193 g/mol

LD50:> 5 g/kg (R, p.o.)

  • Synonyms:NZ-105
  • ATC:C08CA
Efonidipine hydrochloride monoethanolate  111011-76-8 [RN],エホニジピン塩酸塩エタノール付加物
CAS 111011-63-3; FREE FORM
2-(N-Benzylanilino)ethyl (±)-1,4-dihydro-2,6-dimethyl-4-(m-nitrophenyl)-5-phosphononicotinate Cyclic 2,2-Dimethyltrimethylene Ester
2-[Benzyl(phenyl)amino]ethyl 5-(5,5-dimethyl-2-oxido-1,3,2-dioxaphosphinan-2-yl)-2,6-dimethyl-4-(3-nitrophenyl)-1,4-dihydro-3-pyridinecarboxylate
2-[Benzyl(phenyl)amino]ethyl 5-(5,5-dimethyl-2-oxido-1,3,2-dioxaphosphinan-2-yl)-2,6-dimethyl-4-(3-nitrophenyl)-1,4-dihydropyridine-3-carboxylate
3-Pyridinecarboxylic acid, 5-(5,5-dimethyl-2-oxido-1,3,2-dioxaphosphorinan-2-yl)-1,4-dihydro-2,6-dimethyl-4-(3-nitrophenyl)-, 2-[phenyl(phenylmethyl)amino]ethyl ester
40ZTP2T37Q
5-(5,5-Dimethyl-1,3,2-dioxaphosphorinan-2-yl)-1,4-dihydro-2,6-dimethyl-4-(3-nitrophenyl)-3-pyridinecarboxylic Acid 2-[Phenyl(phenylmethyl)amino]ethyl Ester P-Oxide
Landel [Trade name]
UNII:40ZTP2T37Q
2-(N-benzylanilino)ethyl 5-(5,5-dimethyl-2-oxo-1,3,2$l^{5}-dioxaphosphinan-2-yl)-2,6-dimethyl-4-(3-nitrophenyl)-1,4-dihydropyridine-3-carboxylate

Efonidipine Hydrochloride Ethanolate Bulk & Tablets 10 mg/20mg/40mg, 

Indicated for the management of
• Hypertension
• Renal parenchymal hypertension
• Angina
CDSCO approved INDIA 28.08.2017 
Launched – 1994, Shionogi Zeria
Efonidipine (INN) is a dihydropyridine calcium channel blocker marketed by Shionogi & Co. of Japan. It was launched in 1995, under the brand name Landel. The drug blocks both T-type and L-type calcium channels [A7844, A32001]. It has also been studied in atherosclerosis and acute renal failure [A32001]. This drug is also known as NZ-105, and several studies have been done on its pharmacokinetics in animals [L1456].

Efonidipine (INN) is a dihydropyridine calcium channel blocker marketed by Shionogi & Co. of Japan. It was launched in 1995, under the brand name Landel (ランデル). The drug blocks both T-type and L-type calcium channels.[1] Drug Controller General of India (DCGI) granted approval to M/s. Zuventus pharma Ltd for marketing efonidipine under brand name Efnocar in India .[2]

Structure Activity Relationship

Efonidipine is a dual Calcium Channel Blocker (L & T-type). It has a unique chemical structure. The phosphonate moiety (Figure 1) at the C5 position of the dihydropyridine ring is considered to be important for the characteristic pharmacological profile of the drug. (figure-1)

Figure-1:Efonidipine: Chemical Structure

Mechanism of action

Efonidipine, a new generation dihydropyridine (DHP) calcium channel blocker, inhibits both L-type and T-type calcium channels.[1]

Pharmacodynamics

  • Efonidipine exhibits antihypertensive effect through vasodilatation by blocking L-type and T-type calcium channels.[1]
  • Efonidipine has a negative chronotropic effect. Working on sino atrial node cells by inhibiting T-type calcium channel activation, Efonidipine prolongs the late phase-4 depolarization of the sino atrial node action potential and suppresses an elevated HR. The negative chronotropic effect of Efonidipine decreases heart rate, myocardial oxygen demand and increases coronary blood flow.[3]
  • Efonidipine increases coronary blood flow by blocking L & T-type calcium channels and attenuates myocardial ischaemia.[4]
  • By reducing synthesis and secretion of aldosterone, Efonidipine prevents hypertrophy and remodeling of cardiac myocytes.[5]
  • Efonidipine increases glomerular filtration rate without increasing intra-glomerular pressure and filtration fraction. This prevents hypertension induced renal damage.[6]
  • Efonidipine prevents Rho-kinase and NFB induced renal parenchymal fibrosis and provides long term renal protection.[7][8]
  • Efonidipine suppresses renin secretion from the juxta glomerular apparatus in the kidneys.[9]
  • Efonidipine enhances sodium excretion from the kidneys by suppressing aldosterone synthesis and secretion from the adrenal glands. Aldosterone induced renal parenchymal fibrosis is suppressed by Efonidipine.[5]
  • Efonidipine prevents NFB induced hypertrophy and inflammation in the renal vasculature and protects the kidneys.[7]
  • Efonidipine protects against endothelial dysfunction due to its anti-oxidant activity and by restoring NO bioavailability.[10][11]
  • Efonidipine has anti-atherogenic activity and protects the blood vessels from atherosclerosis.[12]
  • Efonidipine lowers blood pressure in cerebral resistance vessels and prevents hypertension induced brain damage.[4]

Pharmacokinetics

Absorption

Peak plasma concentration is achieved in about 1.5 to 3.67 hours after administration. Half life is approximately 4 hours. The pharmacokinetic parameters of Efonidipine are depicted in Table-1.

Table 1: PK Parameters in Adult Healthy Male Subjects

Variable Efonidipine
Mean Range
Cmax(ng/ml) 36.25 9.66-66.91
Tmax (hour) 2.59 1.50-3.67
T1/2 (hour) 4.18 2.15-6.85

*Data on file

Long Duration of Action

Efonidipine has a slow onset and a long duration of action. This unique characteristic of Efonidipine is because of the following reasons:[13]

  1. High lipophilicity of Efonidipine allows it to enter the phospholipid rich cell membrane and access the dihydropyridine binding site of the Ca2+ channels.
  2. Tight binding to the dihydropyridine receptors.
  3. The dissociation constant of Efonidipine from dihydropyridine receptors is very low (0.0042/min/nM), signifying very slow dissociation from the receptors. This explains the long duration of action of Efonidipine.

Metabolism

Efonidipine is primarily metabolized in the liver. The important metabolites are N-dephenylated Efonidipine (DPH), deaminated Efonidipine (AL) and N-debenzylated Efonidipine (DBZ). DBZ and DPH exhibit activity as calcium antagonists. The vasodilating properties of DBZ and DPH were about two-thirds and one-third respectively than that of the parent compound. Results suggest that the majority of the pharmacological effect after oral dosing of Efonidipine hydrochloride in man is due to unchanged compound and its metabolites make a small contribution to the pharmacological effect.[14]

Elimination

Biliary route is the main pathway of excretion. No significant amount of unchanged drug was excreted in urine. In the urine collected for 24 h after an oral dosing, 1.1 % of the dose was excreted as deaminated Efonidipine, and 0.5% as a pyridine analogue of deaminated Efonidipine.

Indications

  • Essential hypertension and renal parenchymal hypertension
  • Angina

Dosage and Administration

  • Essential hypertension and renal parenchymal hypertension: 20-40 mg orally once daily. A dose of up to 80mg/day is seen to be safe and effective in clinical trials.[15][16]
  • Angina: 40 mg/day.

Contraindications

  • Contraindicated in patients hypersensitive to Efonidipine or any of the excipients
  •  It is also contraindicated in pregnancy and lactation.

Precautions

  •  Should be administered with caution in patients with hepatic impairment
  • Dose adjustment may be required in elderly as hypotension can occur
  •  Efonidipine may worsen clinical condition in patients with sinus bradycardia, sinus arrest or sinus node dysfunction
  • As dizziness can occur due to hypotensive action, one should be careful while operating machines, with aerial work platforms and driving of a motor vehicle
  • Drug should not be stopped abruptly. Discontinuation should be gradual and under supervision of a qualified physician

Drug Interactions

  • Other anti-hypertensive agents: Efonidipine enhances the antihypertensive action additively and may produce hypotension and shock. Blood pressure should be monitored regularly to adjust dose of concomitant drugs.
  •  Cimetidine: Cimetidine inhibits CYP450 enzymes involved in metabolism of CCBs. Blood concentration of calcium channel antagonists increase leading to higher incidence of side effects (hot flushes).
  • Grape fruit juice: Grapefruit juice suppresses enzymes metabolizing calcium channel antagonists (cytochrome P450) and reduces the clearance. Thus, there is a possibility that blood concentration of the drug may increase and the anti-hypertensive effect is enhanced.
  • Tacrolimus: Efonidipine inhibits metabolic enzymes involved in Tacrolimus metabolism and reduces its clearance. So, increase in blood concentration of Tacrolimus can occur.

Adverse Drug Reactions

The common side effects are hot flushes, facial flushing and headache. In addition, elevation in serum total cholesterol, ALT (SGPT), AST (SGOT) and BUN may occur. Frequent urination, pedal edema, increased triglycerides occurs in less than 0.1%.[17]

Lesser incidence of pedal edema (< 0.1%)

One common adverse effect of the L-type Ca2+ channel blockers like Amlodipine is vasodilatory Pedal edema. Combined L-/T-type Ca2+ channel blockers, such as Efonidipine, display antihypertensive efficacy similar to their predecessors (Amlodipine) with much less propensity of pedal edema formation. Efonidipine equalizes the hydrostatic pressure across the capillary bed through equal arteriolar and venular dilatation, thus reducing vasodilatory edema. These incremental microcirculatory benefits of efonidipine over the conventional L-type Ca2+ channel blockers (Amlodipine) are likely attributed to their additional T-type Ca2+ channel blocking properties and the increased presence of T-type Ca2+channels in the microvasculature (e.g. arterioles, capillaries, venules etc).[18]

Among the CCBs, Efonidipine (<0.1%)[17] has lowest incidence of pedal edema compared to amlodipine ( 5-16%)[19], cilnidipine (5%)[20], benidipine (5%)[21] and azelnidipine (15.5%).[22]

Use in Special Population

Administration to Elderly

The drug should be started at low dose (20 mg/day) in elderly. Patient should be carefully observed for development of hypo-tension. Dose may be halved if there is intolerance to the 20 mg/day dosage regimen.

Pregnancy and Lactation

The drug should not be administered to pregnant women and women suspected of being pregnant. Administration to lactating women should be avoided unless benefit significantly surpasses the risk to the child. Mothers on Efonidipine treatment should avoid breast feeding.

Pediatric Use

Safety of Efonidipine in low birth weight infants, newborns, infants and children has not been established.

Efonidipine-The Best in Class

Efonidipine is unique among clinically available CCBs. Its antihypertensive efficacy is superior or at par with other CCBs. But, in terms of pleiotropic effects leading to enhanced cerebral, cardiac and renal protection, Efonidipine scores over the other CCBs.

Advantages over Amlodipine

1.      Better renoprotection by:

  • Dual channel blockade [1]
  • Prevention of Rho-kinase and NFkB induced tubulointerstitial fibrosis[23][24]
  • Reduction of synthesis and secretion of aldosterone from the adrenal cortex[25]

2.       Preferred in  angina  with hypertension due to negative chronotropic action[26]

3.       Better control of reflex tachycardia[3]

4.       Reduces cardiac remodelling, arterial stiffness and prevents atherogenesis[27]

5.       More useful in patients with diabetes & nephropathy[28]

6.       Better protection  against cardiac hypertrophy by significant reduction in LVMI[29]

7.       Less adverse effects compared to Amlodipine[30]

8.       Reduces endothelial dysfunction and oxidative stress(anti-oxidant property)[10]

Advantages over Cilnidipine

1.       Strong negative chronotropic effect (less tachycardia) compared to Cilnidipine[3]

2.       Significant improvement in exercise tolerance.[31]Better choice in hypertensive patients with angina.

3.       Better BP control by marked urinary Na+ excretion[32]

4.       Better renoprotection by:

  • a.      Suppression of plasma renin release[33]
  • b.     Prevention of Rho-kinase and NFkB induced tubulointerstitial fibrosis[34][35]
  • c.      Reduction of synthesis and secretion of aldosterone from the adrenal cortex[5]

5.       Better choice in diabetic hypertensives[36]

6.       Prevents cardiac remodelling by suppression of aldosterone secretion[5]

7.       Superior anti-oxidant activity[10]

8.       Less adverse effects compared to Cilnidipine[30]

Advantages over Benidipine

L & T-type CCBs have invoked a lot of interest in the management of hypertension because of their unique pharmacological profile. Several novel agents have been developed including Azelnidipine, Barnidipine, Benidipine, Efonidipine, Manidipine and Nilvadipine. Among all the agents, Efonidipine has emerged as the best among its peers. The advantages of Efonidipine over Benidipine are summarized below.

1. More selective blockade of T-type calcium channels [37][38]

2. More balanced renal arteriolar dilatation than benidipine[37][38]

3. Superior anti-proteinuric effect [15]

4. Greater reduction of serum aldosterone [39]

5. Renoprotection by reducing plasma renin unlike Benidipine [39]

6. Greater negative chronotropic effect

7. Efonidipine has anti-platelet activity[12]

8. Efonidipine reduces Insulin Resistance [40]

9. Significantly lower incidence of pedal edema & constipation compared to Benidipine

A new synthesis of efonidipine has been described: The cyclization of 2,2-dimethylbutane-1,4-diol (I) with triethyl phosphite (II) by heating at 100 C gives 2-methoxy-5,5-dimethyl-1,3,2-dioxaphosphorinan (III), which, by treatment with iodoacetone (IV) in refluxing ether, yields 2-acetonyl-5,5-dimethyl-1,3,2-dioxaphosphorinan-2-one (V). The condensation of (V) with 3-nitrobenzaldehyde (VI) by means of piperidine in acetic acid affords 3-(5,5-dimethyl-2-oxo-1,3,2-dioxaphosphorinan-2-yl)-4-(3-nitrophenyl)-3-buten-2-one (VII), which is finally cyclized with 3-amino-2-propenoic acid 2-(N-benzyl-N-phenylamino)ethyl ester (VIII) in refluxing toluene.ReferencesChem Pharm Bull 1992,40(9),2362

A new synthesis for (4S)-efonidipine has been described: The reaction of 5,5-dimethyl-2-(2-oxopropyl)-1,3,2-dioxaphosphorinan-2-one (I) with dimorpholino(3-nitrophenyl)methane (II) by means of trifluoroacetic acid in hot toluene gives 5,5-dimethyl-2-[1-acetyl-2-(3-nitrophenyl)vinyl]-1,3,2-dioxaphosphorina n-2-one (III), which is cyclized with 3-aminocrotonic acid 2(S)-methoxy-2-phenylethyl ester (IV) in refluxing toluene; the recrystallization of the resulting product affords 5-(5,5-dimethyl-2-oxo-1,3,2-dioxaphosphorinan-2-yl)-2,6-dimethyl-4(S)-(3-nitrophenyl)-1,4-dihydropyridine-3-carboxylic acid 2(S)-methoxy-2-phenylethyl ester (V). The protection of the NH group of (V) with chloromethyl methyl ether and NaH in THF yields the N-methoxymethyl derivative (VI), which is transesterified with 2-(N-benzyl-N-methylamino)ethanol (VII) and NaH in DMSO, giving the protected final product (VIII). Finally, this compound is deprotected with HCl in ethanol.

An enantioselective synthesis of efonidipine has been described: The enantioselective hydrolysis of 5-(5,5-dimethyl-2-oxo-1,3,2-dioxaphosphorinan-2-yl)-2,6-dimethyl-4-(3-n itrophenyl)-1,4-dihydropyridine-3-carboxylic acid propionyloxymethyl ester (I) with lipase AH in 2,5-dimethyltetrahydrofuran saturated with water gives the corresponding free acid of the (S)-isomer (III), while the propionyloxymethyl ester of the (R)-isomer (II) remains undisturbed. After chromatographic separation, the (R)-ester (II) is hydrolyzed with NaOH in methanol to the (R)-acid (IV). Finally, both enantiomerically pure acids (III) and (IV) are separately esterified with 2-(N-benzyl-N-phenylamino)ethanol in the usual way

CLIP

PAPER

Synthesis of 1,4-dihydropyridine-5-phosphonates and their calcium antagonistic and antihypertensive activities: Novel calcium-antagonist 2-[benzyl(phenyl)amino]ethyl 5-(5,5-dimethyl-2-oxo-1,3,2-dioxaphosphorinan-2-yl)-1,4-dihydro-2, 6-dimethyl-4-(3-nitrophenyl)-3-pyridinecarboxylate hydrochloride ethanol (NZ-105) and its crystal structure
Chem Pharm Bull 1992, 40(9): 2362

PATENT

IN 201501586

http://ipindiaservices.gov.in/PatentSearch/PatentSearch/ViewPDF

  1. Jump up to:a b c d Tanaka H, Shigenobu K (2002). “Efonidipine hydrochloride: a dual blocker of L- and T-type Ca2+ channels”. Cardiovasc. Drug Rev20 (1): 81–92. PMID 12070536.
  2. Jump up^http://www.cdsco.nic.in/writereaddata/Minutes%20of%2034th%20SEC%20Cardiovascular%20&%20Renal%2008_11_2016.pdf
  3. Jump up to:a b c Masumiya H, Shijuku T, Tanaka H, Shigenobu K. Inhibition of myo¬cardial L- and T-type Ca2+ currents by efonidipine: possible mecha¬nism for its chronotropic effect. Eur J Pharmacol. 1998; 349: 351-7.
  4. Jump up to:a b Masuda Y, Tanaka S. Efonidipine Hydrochloride: A New Calcium Antagonist. Cardiovascular Drug Reviews. 1994; 12 ( 2): 123-135.
  5. Jump up to:a b c d Ikeda K, Isaka T, Fujioka K, Manome Y, Tojo K. Suppression of Aldosterone Synthesis and Secretion by Ca2+ Channel Antagonists. International Journal of Endocrinology. 2012.
  6. Jump up^ Hayashi K, et al. T-Type Ca Channel Blockade as a Determinant of Kidney Protection. Keio J Med. 2010; 59(3): 84-95
  7. Jump up to:a b Hayashi M, Yamaji Y, Nakazato Y, Saruta T. The effects of calcium channel blockers on nuclear factor kappa B activation in the mesangial cells. Hypertens Res. 2000;23:521–525.
  8. Jump up^ Sugano N, Sugano N, Wakino S, Tatematsu S, Homma K, Yoshioka K, Hasegawa K, Utsunomiya Y, Tokudome G, Hosoya T, Saruta T, Hayashi K. Role of T-type Ca2 channels (TCCs) as a determinant of Rho-kinase activation and epithelial-mesenchymal transition (EMT) in renal injury. J Hypertens. 2006;24(suppl 6):128
  9. Jump up^ Baylis C, Qiu C, Engels K. Comparison of L-type and mixed L- and T-type calcium channel blockers on kidney injury caused by deoxycorticosterone-salt hypertension in rats. Am J Kidney Dis. 2001;38: 1292–1297.
  10. Jump up to:a b c Sasaki H, Saiki A, Endo K, Ban N, Yamaguchi T, Kawana H, Nagayama D, Ohhira M, Oyama T, Miyashita Y, Shirai K. Protective effects of efonidipine, a T- and L-type calcium channel blocker, on renal function and arterial stiffness in type 2 diabetic patients with hypertension and nephropathy. J Atheroscler Thromb. 2009 Oct; 16(5): 568-75.
  11. Jump up^ Oshima T, Ozono R, Yano Y, Higashi Y, Teragawa H, Miho N, Ishida T, Ishida M, Yoshizumi M, Kambe M. Beneficial effect of T-type calcium channel blockers on endothelial function in patients with essential hypertension. Hypertens Res. 2005 Nov;28(11):889-94.
  12. Jump up to:a b Nomura S, Kanazawa S, Fukuhara S. Effects of efonidipine on platelet and monocyte activation markers in hypertensive patients with and without type 2 diabetes mellitus. J Hum Hypertens. 2002 Aug;16(8):539-47.
  13. Jump up^ Yamashita T, Masuda Y, et al. NZ-105, a New 1,4-Dihydropyridine Derivative: Correlation between Dihydropyridine Receptor Binding and Inhibition of Calcium Uptake in Rabbit Aorta. Japan J Pharmacol. 1991; 57: 337-348.
  14. Jump up^ Nakabeppu H, et al.Metabolism of Efonidipine in Man.Xenobiotica.1995 Aug;229-239.
  15. Jump up to:a b Hayashi K, Kumagai H, Saruta T. Effects of efonidipine and ACE inhibitors on proteinuria in human hypertension with renal impairment. Am J Hypertens. 2003;16:116 –122
  16. Jump up^  Oh IY, Seo MK, Lee HY, Kim SG, Kim KS, Kim WH, Hyon MS, Han KR, Lim SJ, Kim CH. Beneficial Effect of Efonidipine, an L- and T-Type Dual Calcium Channel Blocker, on Heart Rate and Blood Pressure in Patients With Mild-to-Moderate Essential Hypertension. Korean Circ J. 2010 Oct;40(10):514-9.
  17. Jump up to:a b http://www.kegg.jp/medicus-bin/japic_med?japic_code=00044638. Missing or empty |title= (help)
  18. Jump up^ Ge W, Ren J. Combined L-/T-type calcium channel blockers: ready for prime time. Hypertension. 2009 Apr;53(4):592-4. doi: 10.1161/HYPERTENSIONAHA.108.127548. Epub 2009 Feb 23. PubMed PMID 19237678.
  19. Jump up^ Osterloh IH. An update on the safety of amlodipine. J Cardiovasc Pharmacol.1991;17 Suppl 1:S65-8.
  20. Jump up^ http://www.kegg.jp/medicus-bin/japic_med?japic_code=00062065. Missing or empty |title= (help)
  21. Jump up^ http://www.kegg.jp/medicus-bin/japic_med?japic_code=00005939. Missing or empty |title= (help)
  22. Jump up^ Takihata M, Nakamura A, Kondo Y, Kawasaki S, Kimura M, Terauchi Y. Comparison of Azelnidipine and Trichlormethiazide in Japanese Type 2 Diabetic Patients with Hypertension: The COAT Randomized Controlled Trial. PLoS One. 2015 May 4;10(5):e0125519.
  23. Jump up^ Song I, KimD, Choi S, Sun M, Kim Y, Shin HS. Role of the α1g T-type calcium channel in spontaneous absence seizures in mutant mice. J Neurosci. 2004; 24: 5249–5257.
  24. Jump up^ Lory P, Bidaud I, Chemin J. T-Type calcium channels in differentiation and proliferation. Cell Calcium. 2006; 40: 135–146.
  25. Jump up^ Ikeda K, Isaka T, Fujioka K, Manome Y, Tojo K. Suppression of Aldosterone Synthesis and Secretion by Ca2+ Channel Antagonists. International Journal of Endocrinology. 2012.
  26. Jump up^ Oh IY, Seo MK, Lee HY, Kim SG, Kim KS, Kim WH, Hyon MS, Han KR, Lim SJ, Kim CH. Beneficial Effect of Efonidipine, an L- and T-Type Dual Calcium Channel Blocker, on Heart Rate and Blood Pressure in Patients With Mild-to-Moderate Essential Hypertension. Korean Circ J. 2010 Oct;40(10):514-9.
  27. Jump up^ Catena C, Colussi G, Marzano L, Sechi LA. Aldosterone and the heart: from basic research to clinical evidence. Horm Metab Res. 2012;44:181– 187. 
  28. Jump up^  Sasaki H, Saiki A, Endo K, Ban N, Yamaguchi T, Kawana H, Nagayama D, Ohhira M, Oyama T, Miyashita Y, Shirai K. Protective effects of efonidipine, a T- and L-type calcium channel blocker, on renal function and arterial stiffness in type 2 diabetic patients with hypertension and nephropathy. J Atheroscler Thromb. 2009 Oct; 16(5): 568-75.
  29. Jump up^  Saito T, Fujii K, Takizawa T, Toyosaki T, Kuwabara Y, Kobayashi S, Ichikawa H, Karaki A, Yamazaki Y, Iwata J, Yamada K, Tomiya H, Takeda K, Inagaki Y. Effects of the new calcium antagonist efonidipine hydrochloride on resting and exercise hemodynamics in patients with stable effort angina. Arzneimittelforschung. 1996 Sep;46(9):861-7.
  30. Jump up to:a b Saruta T. Current status of calcium antagonists in Japan. Am J Cardiol. 1998;82:32R-34R.
  31. Jump up^ Okayama S, Imagawa K, Naya N, Iwama H, Somekawa S, Kawata H, Horii M, Nakajima T, Uemura S, Saito Y. Blocking T-type Ca2+ channels with efonidipine decreased plasma aldosterone concentration in healthy volunteers. Hypertens Res. 2006 Jul;29(7):493-7.
  32. Jump up^ Honda M, Hayashi K, Matsuda H, Kubota E, Tokuyama H, Okubo K, Ozawa Y, Saruta T.  Divergent natriuretic action of calcium channel antagonists in mongrel dogs: renal haemodynamics as a determinant of natriuresis. Clinical Science. 2001; 101: 421–427
  33. Jump up^ Wagner C, Kramer KB, Hinder M, Kieninger M, Kurtz A. T-type and L-type calcium channel blockers exert opposite effects on renin secretion and renin gene expression in conscious rats. Br J Pharmacol. 1998;124: 579 –585. 
  34. Jump up^ Song I, KimD, Choi S, Sun M, Kim Y, Shin HS. Role of the α1g T-type calcium channel in spontaneous absence seizures in mutant mice. J Neurosci. 2004; 24: 5249–5257.
  35. Jump up^ Lory P, Bidaud I, Chemin J. T-Type calcium channels in differentiation and proliferation. Cell Calcium. 2006; 40: 135–146.
  36. Jump up^  Ando K, Ueshima K, Tanaka S, Kosugi S, Sato T, Matsuoka H, Nakao K, Fujita T. Comparison of the antialbuminuric effects of L-/N-type and L-type calcium channel blockers in hypertensive patients with diabetes and microalbuminuria: the study of assessment for kidney function by urinary microalbumin in randomized (SAKURA) trial. Int J Med Sci. 2013 Jul 30;10(9):1209-16.
  37. Jump up to:a b Hayashi K, Wakino S, Sugano N, Ozawa Y, Homma K, Saruta T. Ca2+ Channel Subtypes and Pharmacology in the Kidney. Circ Res. 2007;100:342-353.
  38. Jump up to:a b  Hayashi K, Ozawa Y, Fujiwara K, Wakino S, Kumagai H, Saruta T. Role of actions of calcium antagonists on efferent arterioles with special references to glomerular hypertension. Am J Nephrol. 2003 Jul-Aug;23(4):229-44.
  39. Jump up to:a b Tani S, Takahashi A, Nagao K, Hirayama A. Effects of the T/L-type calcium channel blocker benidipine on albuminuria and plasma aldosterone concentration. A pilot study involving switching from L-type calcium channel blockers to benidipine. Int Heart J. 2014;55(6):519-25
  40. Jump up^  Li M.  Role of T-Type Ca2+ Channels in Basal Insulin Release. T-type Calcium Channels in Basic and Clinical Science. Springer Vienna. 2015; 137-150. 
Efonidipine
Efonidipine structure.svg
Clinical data
Trade names Landel (ランデル)
AHFS/Drugs.com International Drug Names
Routes of
administration
Oral
ATC code
  • none
Legal status
Legal status
  • In general: ℞ (Prescription only)
Identifiers
CAS Number
PubChem CID
ChemSpider
UNII
ChEMBL
Chemical and physical data
Formula C34H38N3O7P
Molar mass 631.65 g/mol
3D model (JSmol)
Title: Efonidipine
CAS Registry Number: 111011-63-3
CAS Name: 5-(5,5-Dimethyl-1,3,2-dioxaphosphorinan-2-yl)-1,4-dihydro-2,6-dimethyl-4-(3-nitrophenyl)-3-pyridinecarboxylic acid 2-[phenyl(phenylmethyl)amino]ethyl ester, P-oxide
Additional Names: 2-(N-benzylanilino)ethyl(±)-1,4-dihydro-2,6-dimethyl-4-(m-nitrophenyl)-5-phosphononicotinate, cyclic 2,2-dimethyltrimethylene ester
Molecular Formula: C34H38N3O7P
Molecular Weight: 631.66
Percent Composition: C 64.65%, H 6.06%, N 6.65%, O 17.73%, P 4.90%
Literature References: Dihydropyridine calcium channel blocker. Prepn: K. Seto et al., WO 8704439idem et al., US 4885284(1987, 1989 both to Nissan); and crystal structure: R. Sakoda et al., Chem. Pharm. Bull. 40, 2362 (1992). Stereoselective synthesis of enantiomers and crystal structure of (S)-form: idem et al., ibid. 2377. Pharmacology: C. Shudo et al., J. Pharm. Pharmacol. 45,525 (1993). Mechanism of action study: T. Yamashita et al., Jpn. J. Pharmacol. 57, 337 (1991). Clinical study: T. Saito et al., Curr. Ther. Res. 52, 113 (1992).
Properties: Crystals from ethyl acetate, mp 169-170° (Sakoda); also reported as mp 155-156° (Seto).
Melting point: mp 169-170° (Sakoda); mp 155-156° (Seto)
Derivative Type: Hydrochloride
CAS Registry Number: 111011-53-1
Molecular Formula: C34H38N3O7P.HCl
Molecular Weight: 668.12
Percent Composition: C 61.12%, H 5.88%, N 6.29%, O 16.76%, P 4.64%, Cl 5.31%
Properties: LD50 in mice (mg/kg): >600 orally (Seto).
Toxicity data: LD50 in mice (mg/kg): >600 orally (Seto)
Derivative Type: Hydrochloride ethanol
CAS Registry Number: 111011-76-8
Manufacturers’ Codes: NZ-105
Trademarks: Landel (Zeria)
Molecular Formula: C34H38N3O7P.C2H5OH.HCl
Molecular Weight: 714.18
Percent Composition: C 60.54%, H 6.35%, N 5.88%, O 17.92%, P 4.34%, Cl 4.96%
Properties: Yellow crystals from aq ethanol, mp 151° (dec).
Melting point: mp 151° (dec)
Derivative Type: (S)- or (R)-Form
Properties: Pale yellow crystals from ethanol, mp 190-192°. [a]D25 + or -7.0° resp (c = 0.50 in chloroform).
Melting point: mp 190-192°
Optical Rotation: [a]D25 + or -7.0° resp (c = 0.50 in chloroform)

(R)-base

  • Formula:C34H38N3O7P
  • MW:631.67 g/mol
  • CAS-RN:128194-13-8

(S)-base

  • Formula:C34H38N3O7P
  • MW:631.67 g/mol
  • CAS-RN:128194-12-7
Therap-Cat: Antihypertensive.
Keywords: Antihypertensive; Dihydropyridine Derivatives; Calcium Channel Blocker; Dihydropyridine Derivatives.

///////////Efonidipine, エホニジピン, IND 2017, Landel , NZ 105, Efonidipine Hydrochloride Ethanolate

CC1=C(C(C(=C(N1)C)P2(=O)OCC(CO2)(C)C)C3=CC(=CC=C3)[N+](=O)[O-])C(=O)OCCN(CC4=CC=CC=C4)C5=CC=CC=C5

FDA approves a new antibacterial drug to treat a serious lung disease using a novel pathway to spur innovation

$
0
0

FDA approves a new antibacterial drug to treat a serious lung disease using a novel pathway to spur innovation

First drug granted approval under FDA’s Limited Population Pathway for Antibacterial and Antifungal Drugs, instituted to spur development of antibiotics for unmet medical needs

The U.S. Food and Drug Administration today approved a new drug, Arikayce (amikacin liposome inhalation suspension), for the treatment of lung disease caused by a group of bacteria, Mycobacterium avium complex (MAC) in a limited population of patients with the disease who do not respond to conventional treatment (refractory disease).

MAC is a type of nontuberculous mycobacteria (NTM) commonly found in water and soil. Symptoms of disease in patients with MAC include persistent cough, fatigue, weight loss, night sweats, and occasionally shortness of breath and coughing up of blood.

September 28, 2018

Release

The U.S. Food and Drug Administration today approved a new drug, Arikayce (amikacin liposome inhalation suspension), for the treatment of lung disease caused by a group of bacteria, Mycobacterium avium complex (MAC) in a limited population of patients with the disease who do not respond to conventional treatment (refractory disease).

MAC is a type of nontuberculous mycobacteria (NTM) commonly found in water and soil. Symptoms of disease in patients with MAC include persistent cough, fatigue, weight loss, night sweats, and occasionally shortness of breath and coughing up of blood.

“As bacteria continue to grow impervious to currently available antibiotics, we need to encourage the development of drugs that can treat resistant infections. That means utilizing novel tools intended to streamline development and encourage investment into these important endeavors,” said FDA Commissioner Scott Gottlieb, M.D. “This approval is the first time a drug is being approved under the Limited Population Pathway for Antibacterial and Antifungal Drugs, and it marks an important policy milestone. This pathway, advanced by Congress, aims to spur development of drugs targeting infections that lack effective therapies. We’re seeing a lot of early interest among sponsors in using this new pathway, and it’s our hope that it’ll spur more development and approval of antibacterial drugs for treating serious or life-threatening infections in limited populations of patients with unmet medical needs.”

Arikayce is the first drug to be approved under the Limited Population Pathway for Antibacterial and Antifungal Drugs, or LPAD pathway, established by Congress under the 21st Century Cures Act to advance development and approval of antibacterial and antifungal drugs to treat serious or life-threatening infections in a limited population of patients with unmet need. Approval under the LPAD pathway may be supported by a streamlined clinical development program. These programs may involve smaller, shorter or fewer clinical trials. As required for drugs approved under the LPAD pathway, labeling for Arikayce includes certain statements to convey that the drug has been shown to be safe and effective only for use in a limited population.

Arikayce also was approved under the Accelerated Approval pathway. Under this approach, the FDA may approve drugs for serious or life-threatening diseases or conditions where the drug is shown to have an effect on a surrogate endpoint that is reasonably likely to predict a clinical benefit to patients. The approval of Arikayce was based on achieving three consecutive negative monthly sputum cultures by month six of treatment. The sponsor of Arikayce will be required by the FDA to conduct an additional, post-market study to describe the clinical benefits of Arikayce.

The safety and efficacy of Arikayce, an inhaled treatment taken through a nebulizer, was demonstrated in a randomized, controlled clinical trial where patients were assigned to one of two treatment groups. One group of patients received Arikayce plus a background multi-drug antibacterial regimen, while the other treatment group received a background multi-drug antibacterial regimen alone. By the sixth month of treatment, 29 percent of patients treated with Arikayce had no growth of mycobacteria in their sputum cultures for three consecutive months compared to 9 percent of patients who were not treated with Arikayce.

The Arikayce prescribing information includes a Boxed Warning regarding the increased risk of respiratory conditions including hypersensitivity pneumonitis (inflamed lungs), bronchospasm (tightening of the airway), exacerbation of underlying lung disease and hemoptysis (spitting up blood) that have led to hospitalizations in some cases. Other common side effects in patients taking Arikayce were dysphonia (difficulty speaking), cough, ototoxicity (damaged hearing), upper airway irritation, musculoskeletal pain, fatigue, diarrhea and nausea.

The FDA granted this application Fast Track, Breakthrough Therapy, Priority Review, and Qualified Infectious Disease Product (QIDP) designations. QIDP designation is given to antibacterial products that treat serious or life-threatening infections under the Generating Antibiotic Incentives Now (GAIN) title of the FDA Safety and Innovation Act. Arikayce also received Orphan Drug designation, which provides additional incentives to assist and encourage the development of drugs for rare diseases.

The FDA granted approval of Arikayce to Insmed, Inc. of Bridgewater, NJ.

/////////////////// Arikayce, amikacin liposome inhalation suspension, fda 2018, Fast Track, Breakthrough Therapy, Priority Review, and Qualified Infectious Disease Product, QIDP, Generating Antibiotic Incentives Now, GAIN,

AKN 028

$
0
0

img

AKN-028
CAS 1175017-90-9
Chemical Formula: C17H14N6
Molecular Weight: 302.33

N-3-(1H-indol-5-yl)-5-pyridin-4-yl-pyrazine-2,3-diamine

N2-(1H-indol-5-yl)-6-(pyridin-4-yl)pyrazine-2,3-diamine

  • Originator Swedish Orphan Biovitrum
  • Developer Akinion Pharmaceuticals
  • Class Antineoplastics; Small molecules
  • Mechanism of Action Fms-like tyrosine kinase 3 inhibitors; Proto oncogene protein c-kit inhibitors
  • Phase I/II Acute myeloid leukaemia
  • 01 Mar 2016 Akinion Pharmaceuticals terminates phase I/II trial in Acute myeloid leukaemia in Czech Republic, Poland, Sweden and United Kingdom (NCT01573247)
  • 17 Sep 2015 AKN 028 is still in phase I/II trials for Acute myeloid leukaemia in Czech Republic, Poland and Sweden
  • 09 Apr 2014 AKN 028 is still in phase I/II trials for Acute myeloid leukaemia in Czech Republic, Poland and Sweden

AKN-028, a novel tyrosine kinase inhibitor (TKI), is a potent FMS-like receptor tyrosine kinase 3 (FLT3) inhibitor (IC(50)=6 nM), causing dose-dependent inhibition of FLT3 autophosphorylation. Inhibition of KIT autophosphorylation was shown in a human megakaryoblastic leukemia cell line overexpressing KIT. In a panel of 17 cell lines, AKN-028 showed cytotoxic activity in all five AML cell lines included. AKN-028 triggered apoptosis in MV4-11 by activation of caspase 3. In primary AML samples (n=15), AKN-028 induced a clear dose-dependent cytotoxic response (mean IC(50) 1 μM). However, no correlation between antileukemic activity and FLT3 mutation status, or to the quantitative expression of FLT3, was observed. Combination studies showed synergistic activity when cytarabine or daunorubicin was added simultaneously or 24 h before AKN-028. In mice, AKN-028 demonstrated high oral bioavailability and antileukemic effect in primary AML and MV4-11 cells, with no major toxicity observed in the experiment. (source: Blood Cancer J. 2012 Aug 3;2:e81. doi: 10.1038/bcj.2012.28.)

SYN

WO 2013/089636

Clip

Development of a Synthesis of Kinase Inhibitor AKN028

 R&D DepartmentMagle Chemoswed, P.O. Box 839, SE 201 80 Malmö, Sweden
 Recipharm OT ChemistryVirdings Allé 32 B, SE 754 50 Uppsala, Sweden
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.8b00092
*Telephone: +46 704473035. E-mail: johan.docera@gmail.com
Abstract Image

The novel tyrosine kinase inhibitor AKN028 has demonstrated promising results in preclinical trials. An expedient protocol for the synthesis of the compound at kilogram scale is described, including an SNAr reaction with high regioselectivity and a Suzuki coupling. Furthermore, an efficient method for purification and removal of residual palladium is described.

yellow or faint-orange powder. Mp 300 °C (dec.);

IR 3133 broad, 1689, 1597, 1554, 1480 cm–11H NMR (DMSO-d6) δ 11.01 (s, 1H), 8.62–8.50 (m, 2H), 8.22 (s, 1H), 8.15 (s, 1H), 8.06 (s, 1H), 7.89–7.82 (m, 2H), 7.39 (d, J = 2.0 Hz, 2H), 7.32 (t, J = 2.7 Hz, 1H), 6.77 (s, 2H), 6.42 (dd, J1 = 8.7 Hz, J2 = 2.0 Hz, 1H);

13C NMR (DMSO-d6) δ 149.9, 145.2, 145.0, 139.6, 132.8, 132.4, 132.2, 128.4, 127.6, 125.6, 118.7, 116.1, 111.2, 111.0, 101.0.

PATENT

 WO 2009095399

https://patentscope.wipo.int/search/ko/detail.jsf;jsessionid=074E97C06EF8C2088428DECCA2CD2EBA.wapp1nB?docId=WO2009095399&recNum=208&office=&queryString=&prevFilter=%26fq%3DOF%3AWO%26fq%3DICF_M%3A%22C07D%22%26fq%3DDP%3A2009&sortOption=Pub+Date+Desc&maxRec=3425

PATENT

WO 2013089636

https://patents.google.com/patent/WO2013089636A1/ko

Protein kinases are involved in the regulation of cellular metabolism, proliferation, differentiation and survival. The FLT-3 (fms-like tyrosine kinase) receptor is a member of the class III subfamily of receptor tyrosine kinases and has been shown to be involved in various disorders such as haematological disorders, proliferative disorders, autoimmune disorders and skin disorders.

In order to function effectively as an inhibitor, a kinase inhibitor needs to have a certain profile regarding its target specificity and mode of action. Depending on factors such as the disorder to be treated, mode of administration etc. the kinase inhibitor will have to be designed to exhibit suitable properties. For instance, compounds exhibiting a good plasma stability are desirable since this will provide a pharmacological effect of the compounds extending over time. Another example is oral administration of the inhibitor which may require that the inhibitor is transformed into a prodrug in order to improve the bioavailability.

WO 2009/095399 discloses pyrazine compounds acting as inhibitors of protein kinases, especially FTL3, useful in the treatment of haematological disorders, proliferative disorders, autoimmune disorders and skin disorders. This document discloses methods for manufacturing such compounds. However these methods are not suitable for large scale processes and the chemical yields are moderate. Furthermore, the compounds obtained by these methods are in amorphous form.

n one aspect of the invention, there is provided a process for preparing a compound of formula (I)

said process comprises the steps of:

a) reacting a compound of formula (1) with a compound of formula (2) in an inert solvent and in the presence of an (C1-6alkyl)3amine, providing a compound of formula (3):


, b) Suzuki coupling of a compound of formula (3) and a compound of formula (4) in an inert solvent and in the presence of a palladium catalyst and a base, providing a crude product comprising a compound of formula (I) and palladium

and

c) removing the palladium from the crude product in step b).

The compound of formula (I) may be obtained in amorphous or crystalline form using the processes outlined below.

Step 1:

Reaction of 2-amino-3,5-dibromopyrazine (1) and 5-aminoindole (2) in a

nucleophilic substitution reaction in the presence of a C1-6alkylamine and an inert polar solvent yields 3-bromo-N-3-(1H-indol-5-yl)-pyrazine-2,3-diamine (3). Examples of inert polar solvents are DMSO, water and NEP. Examples of (C1-6alkyl)3amine are triethylamine, trimethylamine and tributylamine. The reaction may be performed at reflux temperature or at about 100-130°C.

Step 2:

A Suzuki coupling of 3-bromo-N-3-(1H-indol-5-yl)-pyrazine-2,3-diamine) (3) and 4- pyridyl-boronic acid (4) in an inert polar solvent in the presence of a palladium catalyst and a base yields N-3-(1H-indol-5-yl)-5-pyridin-4-yl-pyrazine-2,3-diamine (I) in amorphous form. Examples of inert solvents are DMF, water and DMA. Examples of palladium catalysts are Pd(dppf) and Pd(OAc)2-DTB-PPS. Example of a base is

K2CO3 The reaction may be performed under inert and oxygen-free atmosphere such as nitrogen or argon.

Heating may take place during step 1 and/or step 2. Steps 1 and 2 may be performed at reflux or in a temperature range of from 100 to 140°C, such as from 105 to 135°C, such as from 110 to 130°C, such as from 130-135°C, such as from 110-115ºC.

Step 3:

A compound of formula (I), also denominated N-3-(1H-indol-5-yl)-5-pyridin-4-yl-pyrazine-2,3-diamine, in amorphous form may be dissolved in acetic acid (HOAc) after which potassium hydroxide (KOH) is added. The compound of formula (I) in amorphous form may be obtained from the process outlined in steps 1 and 2.

Alternatively, the compound of formula (I) may be obtained according to the process described in WO 2009/095399. The obtained crystalline form is removed from the slurry by, for instance, filtration. Step 3 may be repeated. Step 3 may be performed at a temperature of about 40°C followed by cooling to room temperature.

The process for preparing a compound according to formula (I) may comprise an additional step (step i) between step 2 and step 3 in order to remove palladium from the crude product of the compound of formula (I). The step comprises; forming a slurry comprising an acid and the compound according to formula (I) in a solvent, adding a siloxane compound to said slurry, removing the solvent from the slurry and adding an organic solvent, such as DMF and/or toluene, to the solid formed whereby a mixture is formed and then potassium hydroxide is added to the formed mixture, Alternatively, palladium may be removed from the crude product comprising (I) using a palladium scavenger such as TMT and/or 3-mercaptopropyl ethyl sulfide silica.

The crystalline form of the compound according to formula (I) may also be prepared from an amorphous form of the compound according to formula (I) by dissolving said amorphous form of the compound in a solvent mixture of

dichloromethane/methanol followed by evaporation of the solvent in a rotary evaporator. The amorphous form of the compound of formula (I) may obtained using the process disclosed in WO 2009/095399.

Example 1. Preparation of 5-Bromo-N-3-(1H-indol-5-yl)-pyrazine-2,3-diamine (compound 3)

DMSO (10 L, 11 kg), 2-amino-3,5-dibromopyrazine (1) (4.5 kg, 17.8 mol, 1 eq.), 5- amino indole (2) (3.06 kg, 23.15 mol, 1.3 eq.) and triethylamine (7.4 L, 5.4 kg, 53.36 mol, 3 eq.) were charged to a reactor. The reaction mixture was heated to 95°C while agitated. After 12 hours, the heating was discontinued and the conversion was 88% of 2-amino-3,5-dibromopyrazine. The reaction was heated again to 95°C and

agitated for an additional 2.5 hours. There was no improvement in conversion. The reaction mixture was agitated at ambient temperature overnight. Triethylamine (3.5 kg) was removed under vacuum and the remaining reaction mixture was transferred to a stainless steel container from which it was charged into another reactor.

Subsequently, 18.4 kg of 50% acetic acid (aq.) was introduced over a period of 20 minutes under agitation, followed by purified water (61 L) charged over a period time of 60 minutes. The slurry was then filtered and the isolated material was washed with 2 x 20 L of 1% acetic acid (aq.).

The isolated 3-bromo-N-3-(1H-indol-5-yl)-pyrazine-2,3-diamine) (3) was transferred to a drying cabinet and dried to invariable weight at 40 ±3°C, (19 hours), to afford 4.36 kg, 14.34 mol, 81 % yield, with a purity of 96% by HPLC.

The reaction temperature in the batch record was set to be 130-135°C. However, at 95°C the reaction mixture was at reflux.

Example 2. Preparation of N-3-(1H-indol-5-yl)-5-pyridin-4-yl-pyrazine-2,3- diamine (compound I)

To a reactor was charged N,N-dimethylformamide (46.7 L, 45 kg), 4-pyridylboronic acid (4) (2.64 kg, 21.5 mol, 1.5 eq.) and 5-bromo-N-3-(1H-indol-5-yl)-pyrazine-2,3- diamine (3) (4.36 kg, 14.3 mol). The reactor was then flushed with nitrogen prior to the charging of Pd(dppf)Cl2-catalyst (0.47 kg, 0.55 mol, 0.04 eq.). To reactor was then charged, over a period of 20 minutes, 24.9 kg of a 2 M solution of potassium carbonate (aq.). The reactor was flushed with nitrogen and heated under agitation to 110-115°C for 1.5 hours, after which 98.3% conversion of (3) was showed. The reaction mixture was quenched by addition of purified water (180 L) under vigorous agitation. The precipitated material was isolated on a hastalloy filter and washed with purified water (50 L), The isolated material was transferred to a drying cabinet and dried to invariable weight at 40 ±3°C (18 hours), to afford a compound of formula (5), i.e. a compound of formula (!) also denominated N-3-(1H-lndol-5-yl)-5-pyhdin-4-yl-pyrazine-2,3-diamine, (3.64 kg, 12.1 mol, 85 % yield).

During the process precipitated material was observed in the solutions, after the reactions, in both steps not previously seen in lab-scale. These impurities were not removed.

Example 3. Purification and crystallisation

In order to remove residual solvents from the material, two consecutive re-precipitations of the material from acetic acid were performed. This also gave crystallinity of the isolated substance. The purification is performed in order to remove palladium.

Purification

To a 1 L round bottomed flask was added 37.8 g of a compound according to formula (I) followed by 600 mL 2 M HOAc (aq.). The material was stirred at RT until a clear, dark red solution was obtained. To the solution was added 30 g Hyflo Super Celite and the slurry was filtered. The filter cake was washed with 25 mL 2 M HOAc

(aq) and 2×35 mL purified water. The obtained filtrate was transferred to a 2 L round bottomed flask containing 950 mL of Me-THF. The mixture was then stirred and heated to 40°C for 30 minutes. To the solution was then added 290 mL 8 M KOH (aq.) at 40°C and pH in the solution was 14.

The aqueous phase was removed and the organic phase washed with 2×100 mL of purified water. The remaining organic phase was then transferred to a 2 L round bottomed flask, followed by 95 mL of DMF, 20 g scavenger 3-Mercaptopropyl ethyl sulphide silica, Phosphonics LTD and 20 g scavenger 2-Mercaptoethyl ethyl sulfide silica purchased from Phosphonics LTD. The solution was vigorously stirred and heated at 60°C. A sample was withdrawn from the slurry after 12 hours, and showed 6 ppm of palladium remaining in the solution. The mixture was allowed to cool and was then filtered to remove the scavenger. The round bottomed flask and filter were rinsed with a mixture of 90 mL Me-THF and 10 mL DMF. Me-THF was then removed on a rotary evaporator and the remaining slurry was azeotropically dried with two portions of 100 mL toluene. To the remaining slurry was then added 85 mL of DMF to a total of 185 mL DMF (5ml DMF/g substance). To the clear solution was then added, slowly, while agitated, 1500 mL of toluene which produced a heavy precipitate. The slurry was filtered off and washed with 2×50 mL of toluene where after the material was dried overnight at 35°C under vacuum to afford 30.9 g of a compound according to formula (I) in a yield of 82%.

Crystallisation:

Example i

1. First re-precipitation

The N-3-(1H-indol-5-yl)-5-pyridin-4-yl-pyrazine-2,3-diamine material (30.9 g) was added to a 1 L round bottomed flask and 450 mL 2 M HOAc (aq.) was added. The slurry was agitated and heated to 40°C for 1 hour, until the material had dissolved. To the solution was then added 158 mL 8 M KOH (aq.) at 40°C. The pH in the solution was 11.4. The slurry was then allowed to cool to 25°C and filtered. The filter cake was washed with 3x 80 mL of purified water and the material was dried overnight at 95°C under vacuum to afford 28.7g N-3-(1H-indol-5-yl)-5-pyridin-4-yl- pyrazine-2,3-diamine in a yield of 93%.

2. Second re-precipitation

N-3-(1H-indol-5-yl)-5-pyridin-4-yl-pyrazine-2,3-diamine material (28.7 g) was added to a 1L round bottomed flask and 430 mL 2 M HOAc (aq) was added. The slurry was agitated and heated to 40°C for 1 hour, until the material had dissolved. To the solution was then added 15 mL 8M KOH (aq) at 40°C. The pH in the solution was 12.3. The slurry was then allowed to cool to 25°C and filtered. The filter cake was washed with 5×50 mL of purified water, and the solid was then dried overnight at 95°C under vacuum to afford 28.3 g N-3-(1H-indol-5-yl)-5-pyridin-4-yl-pyrazine-2,3- diamine in a yield of 99%.

Example ii

The N-3-(1H-indol-5-yl)-5-pyridin-4-yl-pyrazine-2,3-diamine material (2.1 kg, 7 mol) was added to a reactor, followed by 2M HOAc (aq.) (59.6 L, 60.2 kg) . The solution in the reactor was then heated to 40°C and stirred for 20 minutes. To the clear solution was then charged, slowly, 30% KOH (aq.) (25 kg) under vigorous agitation. The slurry was agitated for 15 minutes. pH in the solution was 6.2, and a total of 1.5 kg 30% KOH (aq.) was then added to the solution to give pH 12.1. The precipitated material was isolated on a Hastelloy filter and washed with purified water (5×30 L). The solid was then transferred to a drying cabinet and dried to invariable weight at 85 ±3°C under vacuum (16 hours; a sample was withdrawn after 16 hours, showing 1400 ppm HOAc and 75 ppm DMF), to afford N-3-(1H-indol-5-yl)-5-pyridin-4-yl-pyrazine-2,3-diamine (2.0 kg, 7 mol, 95 % yield).

Hence, N-3-(1H-indol-5-yl)-5-pyridin-4-yl-pyrazine-2,3-diamine is obtained in an uniform crystalline form, which was achieved by precipitating the product from aqueous acetic acid by introduction of aqueous potassium hydroxide.

Example 5. Synthesis of 5-Bromo-N-3-(1H-indol-5-yl)-pyrazine-2,3-diamine (compound 3)

2-Amino-3,5-dibromopyrazine (45 g, 1.0 eq.), 5-aminoindole (30,6 g, 1.3 eq.), 67.5 mL NEP, i.e. 1-ethyl-2-pyrrolidone, and 74.5 mL triethylamine were added to a 250 mL reactor. The jacket temperature was set to 130°C and the reaction mixture was stirred for 22 h. HPLC after 22 h showed 87% conversion of the 2-amino-3,5-dibromopyrazine. After 24 h HPLC showed 92% conversion and the reaction slurry was cooled to 80°C and quenched by addition of addition of 50% HOAc(aq) and water. The obtained slurry was then allowed to cool to room temperature over night while agitated. The material was isolated on a glass filter funnel and was washed with water. The material was dried at 80 °C under vacuum until dry to afford 71% of the compound 5-bromo-N-3-(1H-indol-5-yl)-pyrazine-2,3-diamine as a dark brown powder. The purity was 99.8% as measured by HPLC.

Example 6. Synthesis of N-3-(1H-indol-5-yl)-5-pyridin-4-yl-pyrazine-2,3-diamine (Compound I)

5-Bromo-N-3-(1H-indol-5-yl)-pyrazine-2,3-diamine (15.0 g, 49 mmol, 1.0 eq.), 4-pyridyl boronic acid (6.6 g, 59 mmol, 1.2 eq.), Pd(OAc)2 (166 mg, 0.74 mmol, 0.015 eq.), DTB-PPS, i.e. 3-(di-tert-butylphosphino)propane-1-sulfonic acid, (199 mg, 0.74 mmol, 0.015 eq.), and DMA, i.e. N,N-dimethylacetamide, (75 mL) were added to a three-necked round-bottomed flask equipped with a mechanical stirrer,

thermometer, and a nitrogen atmosphere. Through a septa was added 2M K2CO3 (aq) (27 ml, 54 mmol, 1.1 eq.) with a syringe. The temperature was increased to 100 °C. Samples for HPLC-analysis of the conversion were drawn and when the conversion had reached 100% the temperature was cooled to 25 °C. At that temperature a water solution of 0.5 M L-cysteine (150 ml) was added by a syringe pump over 1 hour with a rate of 2.5 mL/minute. After 3 hours maturing time at room temperature the material was isolated on a glass filter funnel and was washed with water. The material was dried at 40 °C under vacuum over the weekend, and 15 grams of N-3-(1H-indol-5-yl)-5-pyridin-4-yl-pyrazine-2,3-diamine (101%) were obtained as a brown powder.

Example 7. Purification of N-3-(1H-indol-5-yl)-5-pyridin-4-yl-pyrazine-2,3-diamine (Compound I)

The crude (7.0 g, 23 mmol) and 2M HOAc (98 mL) was added to a 250 mL round-bottomed flask. To this was added TMT, i.e. trithiocyanuric acid, (1.4 g) and SPM32, i.e. 3-mercaptopropyl ethyl sulfide silica, (1.4 g). The mixture was stirred in room temperature for 24 hours. After 24 hour a polish filtration through hyflo super cel was performed. To the clear filtrate was added 50 mL 5 M KOH(aq) under 15 minutes to precipitate the product. After 18 hours maturing time at room temperature the material was isolated on a glass filter funnel and was washed with 2×20 mL water. The first was being a slurry wash and the second a displacement wash. The material was dried at 40 °C under vacuum over the weekend, and 3.9 grams (56%) was obtained as a light yellow powder. The Pd content was 3.7 ppm.

PATENT

US 8436171

PATENT

WO 2016008433

PATENT

WO 2016015604

PATENT

WO 2016015597

PATENT

WO 2016015605

PATENT

WO 2016015598

PATENT

WO 2017146794

PATENT

WO 2017146795

https://patents.google.com/patent/WO2017146795A1/en

PATENT

US 20180071302

REFERENCES

1: Eriksson A, Hermanson M, Wickström M, Lindhagen E, Ekholm C, Jenmalm Jensen A, Löthgren A, Lehmann F, Larsson R, Parrow V, Höglund M. The novel tyrosine kinase  inhibitor AKN-028 has significant antileukemic activity in cell lines and primary cultures of acute myeloid leukemia. Blood Cancer J. 2012 Aug 3;2:e81. doi: 10.1038/bcj.2012.28. PubMed PMID: 22864397; PubMed Central PMCID: PMC3432483.

////////////AKN028 , AKN-028 , AKN 028, phase 2, Swedish Orphan Biovitrum,  Akinion Pharmaceuticals,  Acute myeloid leukaemia

NC1=NC=C(C2=CC=NC=C2)N=C1NC3=CC4=C(NC=C4)C=C3

Sarecycline , サレサイクリン

$
0
0

Sarecycline.svg

ChemSpider 2D Image | Sarecycline | C24H29N3O8

Sarecycline

サレサイクリン

MW 487.5024, MF C24H29N3O8 FREE FORM

Paratek  INNOVATOR

FDA 2018/10/1 APPROVED SEYSARA, ALMIRALL, for the oral treatment of inflammatory lesions of non-nodular moderate to severe acne vulgaris in patients 9 years of age and older

(4S,4aS,5aR,12aS)-4-(dimethylamino)-3,10,12,12a-tetrahydroxy-7-[(methoxymethylamino)methyl]-1,11-dioxo-1,4,4a,5,5a,6,11,12a-octahydrotetracene-2-carboxamide
(4S,4aS,5aR,12aS)-4-(Dimethylamino)-3,10,12,12a-tetrahydroxy-7-{[methoxy(methyl)amino]methyl}-1,11-dioxo-1,4,4a,5,5a,6,11,12a-octahydro-2-tetracenecarboxamide
1035654-66-0 [RN] FREE FORM
2-Naphthacenecarboxamide, 4-(dimethylamino)-1,4,4a,5,5a,6,11,12a-octahydro-3,10,12,12a-tetrahydroxy-7-[(methoxymethylamino)methyl]-1,11-dioxo-, (4S,4aS,5aR,12aS)-
94O110CX2E
9743

P005672, 

  • P 005672

Sarecycline hydrochloride.png

CAS 1035979-44-2 HCl

Molecular Formula C24 H29 N3 O8 . Cl H
 Molecular Weight 523.963

P-005672
PTK-AR-01
SC-1401
WC-3035

Sarecycline (trade name Seysara; development code WC-3035) is a tetracycline-derived antibiotic. In the United States, it was approved by the FDA in October 2018 for the treatment of moderate to severe acne vulgaris.[1]

Paratek Pharmaceuticals, Inc. licensed the US rights to sarecycline for the treatment of acne in the United States to Actavis, a subsidiary of Allergan, while retaining rights in the rest of the world.[2]

Allergan initiated a Phase 3 study in December 2014 evaluating the efficacy and safety of sarecycline tablets 1.5 mg/kg per day taken orally for 12 weeks versus placebo in the treatment of acne vulgaris.[3] Two phase 3 randomized, multi-center, double-blind, placebo-controlled studies evaluating the efficacy and safety of sarecycline in moderate to severe acne reported positive results on 27 March 2017.[4]

SYN

US 2016/0200671

PATENT

WO 2008079363

PATENT

WO 2008079339

PATENT

WO 2012155146

EXAMPLES

[00104] The following examples illustrate the synthesis of the compounds described herein.

Synthesis of (4S,4aS,5aR,12aS)-4-dimethylamino-3,10,12,12a-tetrahydroxy-7-[(methoxy(methyl)amino)-methyl]-l,ll-dioxo-l,4,4a,5,5a,6,ll,12a-octahydro-naphthacene-2-carboxylic acid amide (“the free base”).

[00105] A solution of 7-formylsancycline TFA salt (2.23 g) and N,0-dimethylhydroxylamine hydrochloride (780 mg) in N,N-dimethylacetamide (15 mL) was stirred for 10 minutes at room temperature under argon atmosphere. To this solution was added sodium cyanoborohydride (302 mg). The solution was stirred for 5 minutes and monitored by LC-MS. The reaction mixture was poured into diethyl ether, and the resulting precipitates were collected by filtration under vacuum. The crude product was purified by prep-HPLC using a C18 column (linear gradient 10-40% acetonitrile in 20 mM aqueous triethanolamine, pH 7.4). The prep-HPLC fractions were collected, and the organic solvent (acetonitrile) was evaporated under reduced pressure. The resulting aqueous solution was loaded onto a clean PDVB SPE column, washed with distilled water, then with a 0.1 M sodium acetate solution followed by distilled water. The product was eluted with

acetonitrile. The eluent was concentrated under reduced pressure, 385 mg was obtained as free base.

Synthesis of crystalline mono hydrochloride salt of (4S,4aS,5aR,12aS)-4-dimethylamino-3,10,12,12a-tetrahydroxy-7-[(methoxy(methyl)amino)-methyl]-l,ll-dioxo-l,4,4a,5,5a,6,ll,12a-octahydro-naphthacene-2-carboxylic acid amide (the “Crystalline Mono Hydrochloride Salt”).

[00106] Crude (4S,4aS,5aR,12aS)-4-dimethylamino-3, 10,12,12a-tetrahydroxy-7-[(methoxy(methyl)amino)-methyl]-l ,ll-dioxo-l,4,4a,5,5a,6,l l ,12a-octahydro-naphthacene-2-carboxylic acid amide (lOOg, app. 35% assay) was purified on preparative column chromatography. The desired fractions (8-10 liters) were combined and the pH was adjusted to 7.0-7.5 using ammonium hydroxide. This aqueous solution was extracted 3 times with dichloromethane (4 liters each time). The dichloromethane layers were combined and concentrated under reduced pressure. The residue was suspended in ethanol (800 ml) and 20 ml water was added. The pH was gradually adjusted to pH 1.6-1.3 using 1.25M hydrochloric acid in methanol and the mixture was stirred for 20-60 minutes at which point the free base was completely dissolved. The solution was concentrated under reduced pressure to 200-250 ml and was seeded with (4S,4aS,5aR,12aS)-4-dimethylamino-3,10, 12, 12a-tetrahydroxy-7-[(methoxy(methyl)amino)-methyl]- 1, 11-dioxo-l,4,4a,5,5a,6,l l,12a-octahydro-naphthacene-2-carboxylic acid amide mono HQ crystals (100-200 mg). The stirring was continued for 2-18 hours while the slurry was kept at <5°C. The resulting crystals were filtered, washed with ethanol (50 mL) and dried under reduced pressure to a constant weight. 20g crystalline (4S,4aS,5aR,12aS)-4-dimethylamino-3,10, 12, 12a-tetrahydroxy-7-[(methoxy(methyl)amino)-methyl]- 1, 11-dioxo-l,4,4a,5,5a,6,l l,12a-octahydro-naphthacene-2-carboxylic acid amide mono hydrochloride was isolated in > 90% purity and > 90% assay.

Synthesis of crystalline mono mesylate salt of (4S,4aS,5aR,12aS)-4-dimethylamino-3,10,12,12a-tetrahydroxy-7-[(methoxy(methyl)amino)-methyl]-l,ll-dioxo-l,4,4a,5,5a,6,ll,12a-octahydro-naphthacene-2-carboxylic acid (the “Crystalline Mesylate Salt”).

[00107] (4S,4aS,5aR,12aS)-4-dimethylamino-3, 10,12, 12a-tetrahydroxy-7-[(methoxy(methyl)amino)-methyl]-l,ll-dioxo-l,4,4a,5,5a,6,l l,12a-octahydro-naphthacene-2-carboxylic acid amide free base (74mg) was suspended in ethanol (740μ1) and heated with stirring to 60°C (bath temperature). Methane sulfonic acid (1.1 eq, 167μ1 as 1M solution in THF) was added and most of the solid dissolved. After five minutes, the suspension was cooled to ambient temperature over approximately 1.75 hours (uncontrolled in oil bath). By 53 °C, solid had precipitated which was filtered at ambient temperature under reduced pressure. A further portion of ethanol (200μ1) was added to aid filtration, as the suspension was viscous. The cake was washed with n-hexane (400μ1) and air dried on filter for approximately 30 minutes to yield 59 mg (67% yield) of yellow solid.

Synthesis of crystalline mono sulfate salt of (4S,4aS,5aR,12aS)-4-dimethylamino-3,10,12,12a-tetrahydroxy-7-[(methoxy(methyl)amino)-methyl]-l,ll-dioxo-l,4,4a,5,5a,6,ll,12a-octahydro-naphthacene-2-carboxylic acid (the “Crystalline Sulfate Salt”).

[00108] (4S,4aS,5aR,12aS)-4-dimethylamino-3, 10,12, 12a-tetrahydroxy-7-[(methoxy(methyl)amino)-methyl]-l,l l-dioxo-l,4,4a,5,5a,6,l l,12a-octahydro-naphthacene-2-carboxylic acid amide free base (86mg) was suspended in ethanol (500μ1) and heated with stirring to 63 °C (bath temperature) at which temperature most of the free base had dissolved. Sulfuric acid (1.1 eq, 194μ1 as 1M solution in water) was added and all of the solid dissolved. The solution was cooled to ambient temperature over approximately 1.75 hours (uncontrolled in oil bath) at which temperature no solid had precipitated. Methyl t-butyl ether (MtBE) was added as an antisolvent (4 x 50μ1). Each addition caused a cloud point, but the solid re-dissolved on stirring. The solution was stirred with a stopper for approximately 3 hours after which time solid precipitated. The solid was filtered under reduced pressure and washed with MtBE (3 x 200μ1) and air dried on filter for

approximately 45 minutes to yield 93 mg (90% yield) of yellow solid.

COMPARATIVE EXAMPLE 1

Synthesis of amorphous bis hydrochloride salt of (4S,4aS,5aR,12aS)-4-dimethylamino-3,10,12,12a-tetrahydroxy-7-[(methoxy(methyl)amino)-methyl]-l,ll-dioxo-l,4,4a,5,5a,6,ll,12a-octahydro-naphthacene-2-carboxylic acid amide.

[00109] (4S,4aS,5aR,12aS)-4-dimethylamino-3, 10,12, 12a-tetrahydroxy-7-[(methoxy(methyl)amino)-methyl]-l,l l-dioxo-l,4,4a,5,5a,6,l l,12a-octahydro-naphthacene-2-carboxylic acid amide free base (1 g) was suspended in methanol (50 mL). The freebase was converted to the hydrochloride salt by adding an excess of methanolic HCl followed by under reduced pressure evaporation to give 1.1 g yellow solid: MS (Mz+1 = 488). 1H NMR (300 MHz, CD30D) δ 7.46 (d, 1H, J = 8.6 Hz), 6.81 (d, 1H, J = 8.6 Hz), 4.09 (d, 1H, J = 1.0 Hz), 3.79 (d, 1H, J = 13.1 Hz), 3.73 (d, 1H, J = 13.1 Hz), 3.36 (m, 1H), 3.27 (s, 3H), 3.08-2.95 (8H), 2.61 (s, 3H), 2.38 (t, 1H, J = 14.8), 2.22 (m, 1H), 1.64 (m, 1H). An XRPD pattern is shown in Figure 10 and a TGA and DSC curve overlaid are shown in Figure 11.

COMPARATIVE EXAMPLE 2

Synthesis of amorphous mono hydrochloride salt of (4S,4aS,5aR,12aS)-4- dimethylamino-3,10,12,12a-tetrahydroxy-7-[(methoxy(methyl)amino)-methyl]-l,ll- dioxo-l,4,4a,5,5a,6,ll,12a-octahydro-naphthacene-2-carboxylic acid amide.

[00110] A sample of Crystalline Mono Hydrochloride Salt (2.09 g) was dissolved in water (250 ml, 120 vols), filtered and frozen in a -78°C bath. Water was removed from the solidified sample using a lyophilizer for 110 hours to yield the amorphous mono hydrochloride salt as a fluffy yellow solid, that was confirmed to be amorphous by XRPD analysis .

PATENT

US 20130302442

PATENT

WO 2015153864

PATENT

WO 2018051102

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2003075857

References

External links

Sarecycline
Sarecycline.svg
Clinical data
Trade names Seysara
Identifiers
CAS Number
PubChem CID
ChemSpider
UNII
Chemical and physical data
Formula C24H29N3O8
Molar mass 487.51 g·mol−1
3D model (JSmol)

////////////Sarecycline, Seysara, WC-3035 FDA 2018, サレサイクリン , P-005672 , PTK-AR-01 , SC-1401, WC-3035,

Revefenacin, ревефенацин , ريفيفيناسين , 瑞维那新 ,

$
0
0

Revefenacin.png

Revefenacin; 864750-70-9; TD-4208; UNII-G2AE2VE07O; G2AE2VE07O; TD-4208; GSK-1160724;

160724; GSK 1160724; TD-4028; YUPELRI

Molecular Formula: C35H43N5O4
Molecular Weight: 597.76 g/mol

[1-[2-[[4-[(4-carbamoylpiperidin-1-yl)methyl]benzoyl]-methylamino]ethyl]piperidin-4-yl] N-(2-phenylphenyl)carbamate

TD-4208
UNII:G2AE2VE07O
ревефенацин [Russian] [INN]
ريفيفيناسين [Arabic] [INN]
瑞维那新 [Chinese] [INN]

Revefenacin is under investigation for the treatment of Chronic Obstructive Pulmonary Disease (COPD).

  • Originator Theravance
  • Developer Theravance Biopharma
  • Class Antiasthmatics; Biphenyl compounds; Carbamates; Piperidines
  • Mechanism of Action Muscarinic receptor antagonists
  • Preregistration Chronic obstructive pulmonary disease
  • 17 Sep 2018 Efficacy data from two replicate 12-week phase III trials and a 12-month safety trial in Chronic obstructive pulmonary disease (COPD) presented at the European Respiratory Society International Congress (ERS-2018)
  • 31 May 2018 Theravance Biopharma in collaboration with Theravance Biopharma initiates enrolment in a phase III trial for Chronic obstructive pulmonary disease in USA (NCT03573817)
  • 18 May 2018Efficacy and adverse events data from a phase I trial in Chronic obstructive pulmonary disease presented at the 114th International Conference of the American Thoracic Society

The compound was licensed to GlaxoSmithKline by Theravance for the inhalation treatment of chronic obstructive pulmonary disease in 2004. The rights were returned in 2009. In 2014, Theravance Biopharma spun-off from Theravance. In 2015, Theravance Biopharma and Mylan enter in a co development agreement for the global development and commercialization of the once-daily nebulizer for the treatment of chronic obstructive pulmonary disease and other respiratory diseases.

SYN

WO 2012009166

SYN OF INT

STR1

FINAL

STR1

PAPER
Discovery of (R)-1-(3-((2-Chloro-4-(((2-hydroxy-2-(8-hydroxy-2-oxo-1,2-dihydroquinolin-5-yl)ethyl)amino)methyl)-5-methoxyphenyl)amino)-3-oxopropyl)piperidin-4-yl (1,1′-biphenyl)-2-ylcarbamate (TD-5959, GSK961081, batefenterol): First-in-class dual pharmacology multivalent muscarinic antagonist and 2 agonist (MABA) for the treatment of chronic obstructive pulmonary disease (COPD)
J Med Chem 2015, 58(6): 2609

Discovery of (R)-1-(3-((2-Chloro-4-(((2-hydroxy-2-(8-hydroxy-2-oxo-1,2-dihydroquinolin-5-yl)ethyl)amino)methyl)-5-methoxyphenyl)amino)-3-oxopropyl)piperidin-4-yl [1,1′-Biphenyl]-2-ylcarbamate (TD-5959, GSK961081, Batefenterol): First-in-Class Dual Pharmacology Multivalent Muscarinic Antagonist and β2 Agonist (MABA) for the Treatment of Chronic Obstructive Pulmonary Disease (COPD)

Departments of Medicinal Chemistry, Pharmacology, §Drug Metabolism and Pharmacokinetics, and Molecular and Cellular Biology, Theravance Biopharma, Inc., 901 Gateway Boulevard, South San Francisco, California 94080, United States
J. Med. Chem.201558 (6), pp 2609–2622
DOI: 10.1021/jm501915g
*Phone: 650-808-3737. E-mail: ahughes@theravance.com
Abstract Image

Through application of our multivalent approach to drug discovery we previously reported the first discovery of dual pharmacology MABA bronchodilators, exemplified by 1. Herein we describe the subsequent lead optimization of both muscarinic antagonist and β2 agonist activities, through modification of the linker motif, to achieve 24 h duration of action in a guinea pig bronchoprotection model. Concomitantly we targeted high lung selectivities, low systemic exposures and identified crystalline forms suitable for inhalation devices. This article culminates with the discovery of our first clinical candidate 12f (TD-5959, GSK961081, batefenterol). In a phase 2b trial, batefenterol produced statistical and clinically significant differences compared to placebo and numerically greater improvements in the primary end point of trough FEV1 compared to salmeterol after 4 weeks of dosing in patients with moderate to severe chronic obstructive pulmonary disease (COPD).

PATENT

WO 2006099165

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2006099165

FIG. 18 shows a PXRD pattern of Form I of the crystalline freebase of the compound of formula I. This crystalline freebase is further characterized by the DSC trace in FIG. 19, the TGA trace in FIG. 20, the DMS trace in FIG. 21, and the micrographic image in FIG. 22.
FIG. 23 shows a PXRD pattern of Form II of the crystalline freebase of the compound of formula I. This crystalline freebase is further characterized by the DSC trace in FIG. 24, the TGA trace in FIG. 25, and the DMS trace in FIG. 26.

PREPARATION 1
Biphenyl-2-ylcarbamic Acid Piperidin-4-yl Ester
Biphenyl-2-isocyanate (97.5 g, 521 mmol) and 4-hydroxy-N-benzylpiperidine (105 g, 549 mmol) were heated together at 70 0C for 12 hours. The reaction mixture was then cooled to 50 0C and ethanol (1 L) was added and then 6M HCl (191 mL) was added slowly. The resulting mixture was then cooled to ambient temperature and ammonium formate (98.5 g, 1.56 mol) was added and then nitrogen gas was bubbled through the solution vigorously for 20 minutes. Palladium on activated carbon (20 g, 10 wt% dry basis) was then added and the reaction mixture was heated at 40 0C for 12 hours, and then filtered through a pad of Celite. The solvent was then removed under reduced pressure and IM HCl (40 mL) was added to the crude residue. The pH of the mixture was then adjusted with IO N NaOH to pH 12. The aqueous layer was extracted with ethyl acetate (2 x 150 mL) and the organic layer was dried (magnesium sulfate), filtered and the solvent removed under reduced pressure to give 155 g of the title intermediate (100% yield). HPLC (10-70) Rt = 2.52; m/z: [M + H+] calc’d for C18H20N2O2 297.15; found 297.31
PREPARATION 2
iV-Benzyl-iV-methylaminoacetaldehvde
To a 3-necked 2-L flask was added N-benzyl-N-methylethanolamine (30.5 g, 0.182 mol), DCM (0.5 L), DIPEA (95 mL, 0.546 mol) and DMSO (41 mL, 0.728 mol).

Using an ice bath, the mixture was cooled to about -10 °C and sulfur trioxide pyridine-complex (87 g, 0.546 mol) was added in 4 portions over 5 minute intervals. The reaction was stirred at -10 0C for 2 hours. Before removing the ice-bath, the reaction was quenched by adding water (0.5 L). The aqueous layer was separated and the organic layer was washed with water (0.5 L) and brine (0.5 L) and then dried over magnesium sulfate and filtered to provide the title compound which was used without further purification.
PREPARATION 3
Biphenyl-2-ylcarbamic Acid l-[2-(Εenzylmethylammo)ethyllpiperidin-4-yl Ester
To a 2-L flask, containing the product of Preparation 2 in DCM (0.5 L) was added the product of Preparation 1 (30 g, 0.101 mol) followed by sodium triacetoxyborohydride (45 g, 0.202 mol). The reaction mixture was stirred overnight and then quenched by the addition of 1 N hydrochloric acid (0.5 L) with vigorous stirring. Three layers were observed and the aqueous layer was removed. After washing with IN NaOH (0.5 L)3 a homogenous organic layer was obtained which was then washed with a saturated solution of aqueous NaCl (0.5 L), dried over magnesium sulfate, filtered and the solvent removed under reduced pressure. The residue was purified by dissolving it in a minimal amount of isopropanol and cooling this solution to 0 °C to form a solid which was collected and washed with cool isopropanol to provide 42.6 g of the title compound (95% yield). MS m/z: [M + H+] calc’d f for C28H33N3O2444.3; found 444.6. Rf=3.5l min (10-70 ACN:H2O, reverse phase HPLC).
PREPARATION 3 A
Biphenyl-2-ylcarbamic Acid l-f2-(Benzylmethylammo)ethyllpiperidin-4-yl Ester
The title compound was prepared by mesylation of iV-benzyl-N-methyl
ethanolamine, which was then reacted with biphenyl-2-ylcarbamic acid piperidin-4-yl ester in an alkylation reaction.
A 500 mL flask (reactor flask) was charged with N-benzyl-iV-methylethanolamine (24.5 mL), DCM (120 mL), NaOH (80 mL; 30wt%) and tetrabutylammonium chloride. Mixing at low speed throughout the reaction, the mixture was cooled to -10 °C (cooling bath), and the addition funnel charged with DCM (30 mL) and mesyl chloride (15.85 mL), which was added drop wise at a constant rate over 30 minutes. The addition was exothermic, and stirring was continued for 15 minutes while the temperature equilibrated back to -10 0C. The reaction was held for at least 10 minutes to ensure full hydrolysis of the excess mesyl chloride.
A 250 mL flask was charged with biphenyl-2-ylcarbamic acid piperidin-4-yl ester (26 g; prepared as described in Preparation 1) and DCM (125 mL), stirred for 15 minutes at room temperature, and the mixture chilled briefly to 10 0C to form a slurry. The slurry was then charged into the reactor flask via the addition funnel. The cooling bath was removed and the reaction mixture was warmed to 5 °C. The mixture was transferred to a separatory funnel, the layers allowed to settle, and the aqueous layer removed. The organic layer was transferred back to the reactor flask, stirring resumed, the mixture held to room
temperature, and the reaction monitored by HPLC for a total of 3.5 hours.
The reactor flask was charged with NaOH (IM solution; 100 mL), stirred, and the layers allowed to settle. The organic layer was separated, washed (NaCl satd. solution), its volume partially reduced under vacuum, and subjected to repeated IPA washings. The solids were collected and allowed to air-dry (25.85 g, 98% purity). Additional solids were obtained from further processing of the mother liquor (volume reduction, EPA, cooling).
PREPARATION 4
Biphenyl-2-ylcarbamic Acid l-(2-Methylaminoethyl)piperidin-4-yl Ester
To a Parr hydrogenation flask was added the product of Preparation 3 (40 g, 0.09 mol) and ethanol (0.5 L). The flask was flushed with nitrogen gas and palladium on activated carbon (15g, 10 wt% (dry basis), 37% wt/wt) was added along with acetic acid (20 mL). The mixture was kept on the Parr hydrogenator under a hydrogen atmosphere (-50 psi) for 3 hours. The mixture was then filtered and washed with ethanol. The filtrate was condensed and the residue was dissolved in a minimal amount of DCM. Isopropyl acetate (10 volumes) was added slowly to form a solid which was collected to provide 22.0 g of the title compound (70% yield). MS m/z: [M + H+] calc’d for C21H27N3O2 354.2; found 354.3. R/=2.96 min (10-70 ACNrH2O, reverse phase HPLC).
PREPARATION 5
Biphenyl-2-ylcarbamic Acid l-{2-[(4-Formylbenzoyr)
methylaminol ethyll piperidin-4- yl Ester
To a three-necked 1-L flask was added 4-carboxybenzaldehyde (4.77 g,
31.8 mmol), EDC (6.64 g, 34.7 mmol), HOBT (1.91 g, 31.8 mmol), and DCM (200 mL). When the mixture was homogenous, a solution of the product of Preparation 4 (10 g, 31.8 mmol) in DCM (100 mL) was added slowly. The reaction mixture was stirred at room temperature for approximately 16 hours and then washed with water (1 x 100 mL), IN HCl (5 x 60 mL), IN NaOH (1 x 100 mL) brine (1 x 5OmL)3 dried over sodium sulfate, filtered and concentrated to afford 12.6 g of the title compound (92% yield; 85% purity based on HPLC). MS m/z: [M + H+] calc’d for C29H31N3O4 486.2; found 486.4. i?y=3.12 min (10-70 ACNiH2O, reverse phase HPLC).
EXAMPLE 1
Biphenyl-2-ylcarbamic Acid 1 -(2- { |4-(4-Carbamoylpiperidin- 1 -ylmethvD
benzoylimethylamino) ethyl’)piperidin-4-vl Ester

To a three-necked 2-L flask was added isonipecotamide (5.99 g, 40.0 mmol), acetic acid (2.57 mL), sodium sulfate (6.44 g) and isopropanol (400 mL). The reaction mixture was cooled to 0-10 0C with an ice bath and a solution of biphenyl-2-ylcarbamic acid l-{2-[(4-formylbenzoyl)methylamino]ethyl}piperidin-4-yl ester (11 g, 22.7 mmol; prepared as described in Preparation 5) in isopropanol (300 mL) was slowly added. The reaction mixture was stirred at room temperature for 2 hours and then cooled to 0-10 0C. Sodium triacetoxyborohydride (15.16 g, 68.5 mmol) was added portion wise and this mixture was stirred at room temperature for 16 hours. The reaction mixture was then concentrated under reduced pressure to a volume of about 50 mL and this mixture was acidified with IN HCl (200 mL) to pH 3. The resulting mixture was stirred at room temperature for 1 hour and then extracted with DCM (3 x 250 mL). The aqueous phase was then cooled to 0-5 °C with an ice bath and 50% aqueous NaOH solution was added to adjust the pH of the mixture to 10. This mixture was then extracted with isopropyl acetate (3 x 300 mL) and the combined organic layers were washed with water (100 mL), brine (2 x 50 mL), dried over sodium sulfate, filtered and concentrated to afford 10.8 g of the title compound (80% yield. MS m/z: [M + H+] calc’d for C35H43N5O4 598.3; found 598.6. Rj=232 min (10-70 ACNiH2O, reverse phase HPLC).

EXAMPLE 2
Crystalline Diphosphate Salt of Biphenyl-2-ylcarbamic Acid l-(2-{[4-(4- Carbamoylpiperidin-l-ylmethyl)benzoyl1methylamino>ethyDpiperidin-4-yl Ester
500 mg of biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoylpiρeridin-l-ylmethyl) benzoyl]methylamino}ethyl)piperidin-4-yl ester (0.826 mmol of 96% pure material;
prepared as described in Example 1) was taken up in 5 ml of water and 1.5 ml of IM phosphoric acid. The pH was adjusted to approximately pH 5.3 with an additional 0.25ml of IM phosphoric acid (equaling 2.1 molar equivalents). The clear solution was filtered through a 0.2 micron filter, frozen and lyophilized to dryness to yield an amorphous diphosphate salt.
20 mg of the amorphous diphosphate salt was dissolved in 2 ml of IPA: ACN (1:1). 0.1 ml of water was added and the mixture heated to 60 °C under stirring. Almost all of the solids dissolved. The suspension was allowed to cool to ambient temperature, under stirring, overnight. The resulting crystals were collected by filtration and air-dried for 20 minutes to give the title compound (18.5 mg, 93% yield) as a white crystalline solid.
When examined under a microscope using polarized light, the crystals exhibited some birefringence.
EXAMPLE 3
Crystalline Diphosphate Salt of Biphenyl-2-ylcarbamic Acid l-(2-{|4-(4- Carbamoylpiperidin-l-vhτiethyl)benzoyl]methylamino}ethyl)piperidin-4-yl Ester
5.0 g of biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoylpiperidin-l-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester (freebase; prepared as described in Example 1) was combined with 80 ml of IPA:ACN (1:1). 4.0 ml of water was added and the mixture heated to 50 °C under stirring, forming a clear solution. To this was added dropwise at 50 °C, 16 ml IM phosphoric acid. The resulting cloudy solution was stirred at 50 °C for 5 hours, then allowed to cool to ambient temperature, under slow stirring, overnight. The resulting crystals were collected by filtration and air-dried for 1 hour, then under vacuum for 18 hours, to give the title compound (5.8 g, 75% yield) as a white crystalline solid (98.3% purity by HPLC).

EXAMPLE 4
Crystalline Monosulfate Salt of Biphenyl-2-ylcarbamic Acid l-(2-{[4-(4- Carbamoylpiperidm-l-ylmethvπbenzoyllmethylamino>ethyl)piperidm-4-yl Ester
442 mg of biphenyl-2-ylcarbamic acid l-(2-{[4-(4-Carbamoylpiperidin-l-ylmethyl) benzoyl]methylamino} ethyl)piperidin-4-yl ester (0.739 mmol of 96% pure material;
prepared as described in Example 1) was taken up in 5 ml of H2OrACN (1 : 1) and 1.45 ml of IN sulfuric acid was added slowly, while monitoring the pH. The pH was adjusted to approx. pH 3.3. The clear solution was filtered through a 0.2 micron filter, frozen and lyophilized to dryness to yield a monosulfate salt.
30.3 mg of the monosulfate salt was dissolved in 1.65 ml of IPA:ACN (10:1). The suspension was heated by placing the vial in a pre-heated 60 °C water bath for 30 minutes. A viscous material was formed and the heat increased to 70 °C for 30 minutes. Since the material remained viscous, the heat was lowered to 60 0C and the mixture heated for an additional hour. The heat was turned off and the mixture was allowed to cool to room temperature. After 4 days, the material appeared to be solid, and the sample was allowed to sit for an additional nine days. The solid was then filtered and dried using a vacuum pump for 1 hour to give the title compound (23 mg, 76% yield).
EXAMPLE 5
Crystalline Monosulfate Salt of Biphenyl-2-ylcarbamic Acid l-(2-{[~4-(4- Carbamoylpiperidin-l-ylmethyl)benzoyl]methylamino>ethyl)piperidin-4-yl Ester
161 g of the monosulfate salt (prepared as described in Example 4) was dissolved in 8.77 ml of IPA:ACN (10:1). The suspension was heated by placing the vial in a pre-heated 70 °C water bath for 1.5 hours. Oil droplets formed within 5 minutes. The heat was lowered to 60 °C and the mixture heated for an additional 1.5 hours, followed by heating at 50 °C for 40 minutes, at 40 °C for 40 minutes, then at 30 0C for 45 minutes. The heat was turned off and the mixture was allowed to slowly cool to room temperature. The next day, the material was viewed under a microscope and indicated needles and plates. The material was then heated at 40 °C for 2 hours, at 35 0C for 30 minutes, and then at 30 °C for 30 minutes. The heat was turned off and the mixture was allowed to slowly cool to room temperature. The solid was then filtered and dried using a vacuum pump for 1 hour to give the title compound (117 mg, 73% yield).

EXAMPLE 6
Crystalline Dioxalate Salt of Biphenyl-2-ylcarbamic Acid l-(2-{|4-(4-Carbamoylpiperidin- 1 -ylmethyl)benzoyl]methylamino> ethyl)piperidin-4-yl Ester
510 mg of biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoylpiperidin-l-ylmethyl)benzoyl]methylamino} ethyl)piperidin-4-yl ester (0.853 mmol of 96% pure material; prepared as described in Example 1) was taken up in 5 ml of H2O:ACN (1:1) and 1.7 ml of IM aqueous oxalic acid was added slowly, while monitoring the pH. The pH was adjusted to approx. pH 3.0. The clear solution was filtered through a 0.2 micron filter, frozen and lyophilized to dryness to yield a dioxalate salt.
31.5 mg of the dioxalate salt was dissolved in 2.76 ml of 94%IPA/6%H20. The mixture was stirred in a pre-heated 60 °C water bath for 2.5 hours. After 25 minutes, all of the sample was in solution. The heat was turned off and the mixture was allowed to cool to room temperature. The next day, a small amount of viscous material was present. The vial was refrigerated at 4 °C. After 4 days, the viscous material was still present. The vial was then placed at room temperature and observed one month later. The material appeared to be solid, and was observed to be crystalline under a microscope. The solid was then filtered and dried using a vacuum pump for 1 hour to give the title compound (20 mg, 63.5% yield).
EXAMPLE 7
Crystalline Dioxalate Salt of Biphenyl-2-ylcarbamic Acid l-(2-{T4-(4-Carbamoylpiperidin- 1 -ylmethyl)benzoyl]methylammo) ethvDpiperidin-4-yl Ester
150 mg of the dioxalate salt (prepared as described in Example 6) was dissolved in 13.1 ml of 94%IPA/6%H20. The mixture was stirred in a pre-heated 60 °C water bath for 2.5 hours. The heat was turned off and the mixture was allowed to cool to room
temperature. The vial was refrigerated at 4 °C. After 6 days, an oily material was observed with what appeared to be a crystal on the side of the vial. The vial was then allowed to reach room temperature, at which point seeds (crystalline material from Example 6) were added and allowed to sit for 16 days. During this time, more crystals were observed to come out of solution. The solid was then filtered and dried using a vacuum pump for 14 hours to give the title compound (105 mg, 70% yield).

EXAMPLE 8
Crystalline Freebase Biphenyl-2-ylcarbamic Acid l-(2-(f4-(4-Carbamoylpiperidin-l- ylmethvDbenzoyl]methylaniino}ethyl)piperidin-4-yl Ester (Form T)
109 mg of biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoylpiperidin-l-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester (prepared as described in
Example 1) was dissolved in 0.56 ml of H2O: ACN (1:1). The suspension was left in a vial (cap loosely placed on top) to allow for a slower evaporation time. The vial was placed under a nitrogen flow environment, although the nitrogen was not used for evaporation, only for the environment. A precipitate was visible within 1 day, which was observed to be crystalline under a microscope. The solid was then placed on a high vacuum line to remove all solvent to give the title compound. Quantitative recovery, 97.8% pure by HPLC.

In an alternate procedure, after dissolving in H2O: ACN (1:1) (approximately 350 mg/mL), the vial was stored at 5 0C, and the precipitate was visible at day 2. The solid was filtered, rinsed with water, and dried on high vacuum overnight. Recovery was 55%, with the solid having 98.2% purity and the liquid having 92.8% purity.
EXAMPLE 9
Crystalline Freebase Biphenyl-2-ylcarbamic Acidl-(2-{J4-(4-Carbamoylpiperidin- l-yhiaethyl)benzoyllmethylammo|ethvDpiperidin-4-yl Ester (Form T)
50.4 mg of biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoylpiperidin-l-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester (prepared as described in
Example 1) was dissolved in 0.144 ml of H2O:ACN (1:1). The suspension was left in vial (cap loosely placed on top) to allow for a slower evaporation time. The vial was refrigerated at 4 0C for 6 days. A precipitate was visible after 2 days. The solid was filtered and placed on a high vacuum line to remove all solvent and give the title compound as a white solid (27.8 mg, 55.2 % yield).
EXAMPLE 10
Crystalline Freebase Biphenyl-2-ylcarbamic Acid l-(2-{[4-(4-Carbamoylpiperidin- l-vhnethvDbenzoyl]methylamino>ethvDpiperidin-4-yl Ester (Form T)
230 mg of biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoylpiperidin-l-yhnethyl)benzoyl]methylamino}ethyl)piρeridin-4-yl ester (prepared as described in
Example 1) was dissolved in 0.2 ml of H2O:ACN (1:1), using slight heat. The mixture was then heated in a 70 °C water bath for 2 hours. The heat was turned off and the mixture was allowed to cool to room temperature, then refrigerated at 4 °C for 1 hour. 50 μl of water was then added (oiled out), followed by the addition of 40 μl of ACN to get the sample back into solution. Seeds (crystalline material from Example 8) were added under slow stirring at room temperature. Crystals started to form ,and the mixture was allowed to sit overnight, with slow stirring. The next day, a heat cool cycle was applied (30 °C for 10 minutes, 40 0C for 10 minutes, then 50 °C for 20 minutes). The heat was turned off and the mixture allowed to cool overnight, with slow stirring. The next day, a second heat/cool cycle was applied (60 0C for 1 hour, with dissolving observed at 70 °C). The heat was turned off and the mixture allowed to cool overnight, with slow stirring. The next day, crystals were present and a third heat cool cycle was applied (60 0C for 3 hours). The heat was turned off and the mixture allowed to cool overnight, with slow stirring. The next day, a heat cool cycle was applied (60 °C for 3 hours, slow cool, then 60 °C for 3 hours). The heat was turned off and the mixture allowed to cool overnight, with slow stirring. After 3 days, the solid was filtered and placed on a high vacuum line to remove all solvent and give the title compound.
EXAMPLE 11
Crystalline Freebase Biphenyl-2-ylcarbamic Acid l-(2-{[4-(4-Carbamoylpiperidin- l-ylmethyl)benzoyl]methylamino|ethyl)piperidin-4-yl Ester (Form JD
70 mg of biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoylpiperidin-l-yhnethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester (prepared as described in
Example 1) was dissolved in 0.1 mL ACN. After addition of 0.3 ml MTBE, the solution appeared cloudy. An additional 50 μl of ACN was added to clarify the solution (155 mg/ml ACN:MTBE = 1 :2). The mixture was left in the vial and capped. Crystals appeared by the next day. The solid was then filtered and placed on a high vacuum line to remove all solvent and give the title compound.

PATENT

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2011008809

U.S. Patent Publication No. 2005/0203133 to Mammen et al. discloses novel biphenyl compounds that are expected to be useful for treating pulmonary disorders such as chronic obstructive pulmonary disease (COPD) and asthma. In particular, the compound biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoylpiperidin-l-ylmethyl) benzoyl]methylamino}ethyl)piperidin-4-yl ester is specifically described in this application as possessing muscarinic receptor antagonist or anticholinergic activity.

The chemical structure of biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoyl piperidin- 1 -ylmethyl)benzoyl]methylamino } ethyl)piperidin-4-yl ester is represented by formula I:

I

The compound of formula I has been named using the commercially-available AutoNom software (MDL, San Leandro, California).

Therapeutic agents useful for treating pulmonary or respiratory disorders are advantageously administered directly into the respiratory tract by inhalation. In this regard, several types of pharmaceutical inhalation devices have been developed for administering therapeutic agents by inhalation including dry powder inhalers (DPI),

metered-dose inhalers (MDI) and nebulizer inhalers. When preparing pharmaceutical compositions and formulations for use in such devices, it is highly desirable to have a crystalline form of the therapeutic agent that is neither hygroscopic nor deliquescent and which has a relatively high melting point thereby allowing the material to be micronized without significant decomposition. Although crystalline freebase forms of the compound of formula I have been reported in U.S. Patent Publication No. 2007/0112027 to Axt et al. as Form I and Form II, the crystalline freebase forms of the present invention have different and particularly useful properties, including higher melting points

One aspect of the invention relates to crystalline freebase forms of biphenyl-2-ylcarbamic acid 1 -(2- { [4-(4-carbamoylpiperidin- 1 -ylmethyl)benzoyl]methy lamino } ethyl) piperidin-4-yl ester characterized by a powder x-ray diffraction pattern comprising diffraction peaks at 2Θ values of 6.6±0.1, 13.1±0.1, 18.6±0.1, 19.7±0.1, and 20.2±0.1.

Another aspect of the invention relates to a crystalline freebase of biphenyl-2-ylcarbamic acid 1 -(2- { [4-(4-carbamoylpiperidin- 1 -ylmethyl)benzoyl]methy lamino } ethyl) piperidin-4-yl ester, designated as form III, which is characterized by a powder x-ray diffraction pattern comprising diffraction peaks at 2Θ values of 6.6±0.1, 13. l±O.l,

18.6±0.1, 19.7±0.1, and 20.2±0.1; and further characterized by having five or more additional diffraction peaks at 2Θ values selected from 8.8=1=0.1, 10. l±O.l, 11.4±0.1, l l.β±O.l, 14.8±0.1, 15.2±0.1, lβ.l±O.l, 16.4±0.1, 16.9±0.1, 17.5±0.1, 18.2±0.1, 19.3±0.1, 19.9±0.1, 20.8±0.1, 21. l±O.l, 21.7±0.1, and 22.3±0.1.

Still another aspect of the invention relates to a crystalline freebase of biphenyl-2-ylcarbamic acid 1 -(2- { [4-(4-carbamoylpiperidin- 1 -ylmethyl)benzoyl]methy lamino } ethyl) piperidin-4-yl ester, designated as form IV, which is characterized by a powder x-ray diffraction pattern comprising diffraction peaks at 2Θ values of 6.6±0.1 , 13. l±O.1 ,

18.6=1=0.1, 19.7=1=0.1, and 20.2±0.1; and further characterized by having five or more additional diffraction peaks at 2Θ values selected from 10.6±0.1, 15.0=1=0.1, lβ.O±O.l, 17.3±0.1, 17.7±0.1, 20.9±0.1, 21.4±0.1, 22.6±0.1, 24.6±0.1, and 27.8±0.1.

Preparation 1

Biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoylpiperidin-l- ylmethvDbenzovHmethylaminol ethyDpiperidin-4-yl Ester The diphosphate salt of biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoylpiperidin-l-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester (16 g) was dissolved in a biphasic mixture of water (100 mL) and EtOAc (200 mL). NaOH (2 N, 75 mL) was added over a period of 5 minutes. The mixture was then stirred for 30 minutes. The phases were separated and the aqueous phase was extracted with EtOAc (200 mL). The combined organic phases were concentrated. DCM (100 mL) was added, and the mixture evaporated to dryness. The solids were dried in an oven for about 48 hours to yield the title compound (9.6 g).

EXAMPLE 1

Crystalline Freebase of Biphenyl-2-ylcarbamic Acid l-(2-{r4-(4-Carbamoylpiperidin-l- ylmethyl)benzoyllmethylamino|ethyl)piperidin-4-yl Ester (Form III) Biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoylpiperidin-l-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester (102.4 mg) was dissolved in MeCN (500 μL). The solution was stirred at room temperature for 80 minutes and a white solid precipitate formed. The mixture was placed in the shaker block to thermocycle (0-40 0C in one hour blocks) for 48 hours. A white, dense, immobile solid was observed. MeCN (500 μL) was added to mobilize the slurry. The mixture was then placed back in the shaker block for 2 hours. The solids were isolated by vacuum filtration using a sinter funnel, then placed in the piston dryer at 40 0C under full vacuum for 15.5 hours, to yield 76.85 mg of the title crystalline compound.

EXAMPLE 2

Crystalline Freebase of Biphenyl-2-ylcarbamic Acid l-(2-{r4-(4-Carbamoylpiperidin-l- ylmethyl)benzoyllmethylamino|ethyl)piperidin-4-yl Ester (Form III) Diphosphate salt of biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoyl-piperidin-l-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester (C3sH43NsO4»2H3PO4; MW 793.75; 632.9 g) was slurried in isopropyl acetate (11.08 L) and water (6.33 L) at room temperature under nitrogen. The suspension was warmed to 53±3 0C and 1OM NaOH (317 mL) was added to the stirred mixture, while maintaining the temperature of the mixture above 50 0C. The mixture was stirred for approximately 5 minutes at 53±3 0C before allowing the layers to settle. The layers were then separated and the aqueous layer was removed. Water (3.16 L) was added to the organic layer while maintaining the temperature of the mixture above 50 0C. The mixture was stirred for 5 minutes at 53±3 0C before allowing the layers to settle. The layers were separated and the water layer was removed. Isopropyl acetate (6.33 L) was added and then about 10 volumes of distillate were collected by atmospheric distillation. This step was repeated with additional isopropyl acetate (3.2 L). After the second distillation, the temperature of the clear solution was reduced to 53±3 0C, then seeded with a suspension of the biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoylpiperidin-l-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester crystalline freebase (Form III; 3.2 g) in isopropyl acetate (51 mL). The resulting suspension was stirred at 53±3 0C for 2 hours, then cooled to 10±3 0C over 4 hours. The suspension was stirred at 10±3 0C for at least 2 hours and then the solids were collected by filtration. The resulting filter cake was washed with isopropyl acetate (2 x 1.9 L) and the product was dried in vacuo at 50 0C to yield the title crystalline compound (C3SH43NsO4; MW 597.76; 382.5 g, 80.3% yield).

EXAMPLE 3

Recrystallization of Crystalline Freebase of Biphenyl-2-ylcarbamic Acid l-(2-{[4-(4- Carbamoylpiperidin- 1 -ylmethyDbenzoyllmethylaminol ethyl)piperidin-4-yl Ester (Form

III)

Crystalline freebase of biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoylpiperidin-l-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester (Form III; C35H43N5O4; MW 597.76; 372.5 g) was slurried in toluene (5.6 L) at 20±3 0C under nitrogen. The suspension was warmed to 82±3 0C, and held at this temperature until complete dissolution was observed. The solution was then clarified into the crystallizer vessel, followed by rinsing with toluene (373 μL). Solids were observed in the crystallizer vessel, and the vessel was re-heated to 82±3 0C to effect dissolution, then cooled to 58±3 0C and seeded with a pre-sonicated (approximately 1 minute) of crystalline freebase (Form III; 1.9 g) in toluene (8 μL). The resulting suspension was allowed to stand at 58±3 0C for at least 4 hours, then cooled to 20±3 0C over 2 hours (approximate cooling rate of 0.33 °C/min). The suspension was stirred at 20±3 0C for at least 1 hour, then the solids were collected by filtration. The resulting filter cake was washed with toluene (2 x 1.2 L) and the product was dried in vacuo at 52±3 0C to yield the title crystalline compound (345.3 g, 92.7% yield).

EXAMPLE 4

Crystalline Freebase of Biphenyl-2-ylcarbamic Acid l-(2-{r4-(4-Carbamoylpiperidin-l- ylmethyl)benzoyllmethylamino|ethyl)piperidin-4-yl Ester (Form IV) Biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoylpiperidin-l-ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester (prepared as described in Preparation 1; 2.5 g) was dissolved in MeCN (10 mL) to yield a viscous oily pale yellow material. Additional MeCN (5 mL) was added to dilute the material. The solution was seeded with crystalline freebase (20 mg; Form III prepared as described in Example 1) and stirred at room temperature for 90 minutes. A large amount of white precipitate (small crystals) was observed. The slurry was analyzed under a polarized light microscope and found to be birefringent.

Additional MeCN (3 mL) was added and the slurry was placed in a Metz SynlO block to thermocycle (0-40 0C in one hour blocks) at 800 rpm overnight. The Metz SynlO is a 10 position parallel reaction station that is static. Agitation of the solution/slurry was by a cross magnetic stirrer bar. The shaker block was a separate piece of equipment that was heated and cooled by an external Julabo bath. The material was removed at 0 0C. It was observed that the slurry had settled out, leaving a pale yellow solution above the white precipitate. The slurry was stirred and placed back in the shaker block to thermocycle.

The material was removed at 40 0C, and stirred at a high agitation rate at room temperature for 80 minutes. The slurry was again analyzed and found to be birefringent. The filter cake was isolated by vacuum filtration using a sinter funnel. MeCN (3 mL) was used to wet the filter paper and the filter cake was washed with MeCN prior to filtration. The cake was deliquored under vacuum for 40 minutes to yield 2.3 g of a flowing white powder. The material was placed in a piston dryer at 400C for 65 hours, to yield 2.2 g of the title crystalline compound as a white powder (99.6% purity).

PATENT

https://patentscope.wipo.int/search/en/detail.jsf;jsessionid=0049F6A3F9FB8C7273B825D49F2465F6.wapp1nA?docId=WO2005087738&tab=PCTDESCRIPTION&maxRec=1000

Example 1
Biphenyl-2-ylcarbamic Acid l-(2-{[4-(4-Carbamoylpiperidin-l- ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl Ester

To a three-necked 2-L flask was added isonipecotamide (5.99 g, 40.0 mmol), acetic acid (2.57 mL), sodium sulfate (6.44 g) and LPA (400 mL). The reaction mixture was cooled to 0-10°C with an ice bath and a solution ofthe product of Preparation 5 (11 g, 22.7 mmol) in LPA (300 mL) was slowly added. The reaction mixture was stined at room temperature for 2 hours and then cooled to 0-10°C. Sodium triacetoxyborohydride (15.16 g, 68.5 mmol) was added portion wise and this mixture was stined at room temperature for 16 h. The reaction mixture was then concentrated under reduced pressure to a volume of about 50 mL and this mixture was acidified with IN HCl (200 mL) to pH 3. The resulting mixture was stined at room temperature for 1 hour and then extracted with DCM (3 x 250 mL). The aqueous phase was then cooled to 0-5°C with an ice bath and 50% aqueous NaOH solution was added to adjust the pH ofthe mixture to 10. This mixture was then extracted with isopropyl acetate (3 x 300 mL) and the combined organic layers were washed with water (100 mL), brine (2 x 50 mL), dried over sodium sulfate, filtered and concentrated to afford 10.8 g ofthe title compound (80% yield. MS m/z: [M + H“1”] calcd for C35H43N5O4, 598.3; found, 598.6. Rf = 2.32 min (10-70 ACN: H2O, reverse phase HPLC).

Example 1A
Biphenyl-2-ylcarbamic acid l-(2- {[4-(4-carbamoylpiperidin-l-ylmethyl)benzoyl] methylamino} ethyl)piperidin-4-yl ester was also prepared as a diphosphate salt using the following procedure :
5.0 g ofthe product of Example 1 was combined with 80 ml of IPA:ACN (1:1). 4.0 ml of water was added and the mixture heated to 50°C under stining, forming a clear solution. To this was added dropwise at 50°C, 16 ml 1M phosphoric acid. The resulting cloudy solution was stined at 50°C for 5 hours, then allowed to cool to ambient temperature, under slow stirring, overnight. The resulting crystals were collected by filtration and air-dried for 1 hour, then under vacuum for 18 hours, to give the diphosphate salt ofthe title compound (5.8 g, 75% yield) as a white crystalline solid (98.3% purity by HPLC).

Example IB
Biphenyl-2-ylcarbamic acid 1 -(2- { [4-(4-carbamoylpiperidin- 1 -ylmethyl)benzoyl] methylamino }ethyl)piperidin-4-yl ester was also prepared as a monosulfate salt using the following procedure.
442 mg ofthe product of Example 1 (0.739 mmol of 96% pure material) was taken up in 5 ml of H2O:ACN (1:1) and 1.45 ml of IN sulfuric acid was added slowly, while monitoring the pH. The pH was adjusted to approx. pH 3.3. The clear solution was filtered through a 0.2 micron filter, frozen and lyophilized to dryness. 161 g of the lyophilized material was dissolved in 8.77 ml of IPA:ACN (10:1). The suspension was heated by placing the vial in a pre-heated 70°C water bath for 1.5 hours. Oil droplets formed within 5 minutes. The heat was lowered to 60°C and the mixture heated for an additional 1.5 hours, followed by heating at 50°C for 40 minutes, at 40°C for 40 minutes, then at 30°C for 45 minutes. The heat was turned off and the mixture was allowed to slowly cool to room temperature. The next day, the material was viewed under a microscope and indicated needles and plates. The material was then heated at 40°C for 2 hours, at 35°C for 30 minutes, and then at 30°C for 30 minutes. The heat was turned off and the mixture was allowed to slowly cool to room temperature. The solid was then filtered and dried using a vacuum pump for 1 hour to give the monosulfate salt ofthe title compound (117 mg, 73% yield).

Example IC
Biphenyl-2-ylcarbamic acid l-(2- {[4-(4-carbamoylpiperidin-l-ylmethyl)benzoyl] methylamino} ethyl)piperidin-4-yl ester was also prepared as a dioxalate salt using the following procedure.
510 mg ofthe product of Example 1 (0.853 mmol of 96% pure material) was taken up in 5 ml of H2O:ACN (1:1) and 1.7 ml of 1M aqueous oxalic acid was added slowly, while monitoring the pH. The pH was adjusted to approx. pH 3.0. The clear solution was filtered through a 0.2 micron filter, frozen and lyophilized to dryness. 150 mg ofthe lyophilized material was dissolved in 13.1 ml of 94%IPA/6%H20. The mixture was stined in a pre-heated 60°C water bath for 2.5 hours. The heat was turned off and the mixture was allowed to cool to room temperature. The vial was refrigerated at 4°C. After 6 days, an oily material was observed with what appeared to be a crystal on the side ofthe vial. The vial was then allowed to reach room temperature, at which point seeds (synthesis described below) were added and allowed to sit for 16 days. During this time, more crystals were observed to come out of solution. The solid was then filtered and dried using a vacuum pump for 14 hours to give the dioxalate salt ofthe title compound (105 mg, 70% yield).
Seed Synthesis
510 mg ofthe product of Example 1 (0.853 mmol of 96% pure material) was taken up in 5 ml of H2O:ACN (1:1) and 1.7 ml of 1M aqueous oxalic acid was added slowly, while monitoring the pH. The pH was adjusted to approx. pH 3.0. The clear solution was filtered through a 0.2 micron filter, frozen and lyophilized to dryness to yield a dioxalate salt. 31.5 mg of this dioxalate salt was dissolved in 2.76 ml of 94%IPA/6%H20. The mixture was stined in a pre-heated 60°C water bath for 2.5 hours. After 25 minutes, all of the sample was in solution. The heat was turned off and the mixture was allowed to cool to room temperature. The next day, a small amount of viscous material was present. The vial was refrigerated at 4°C. After 4 days, the viscous material was still present. The vial was then placed at room temperature and observed one month later. The material appeared to be solid, and was observed to be crystalline under a microscope. The solid was then » filtered and dried using a vacuum pump for 1 hour to give the dioxalate salt (20 mg, 63.5% yield).

Example ID
Biphenyl-2-ylcarbamic acid 1 -(2- { [4-(4-carbamoylpiperidin- 1 -ylmethyl)benzoyl] methylamino} ethyl)piperidin-4-yl ester was also prepared as a freebase crystal using the following procedure.
230 mg ofthe product of Example 1 was dissolved in 0.2 ml of H O:ACN (1:1), using slight heat. The mixture was then heated in a 70°C water bath for 2 hours. The heat was turned off and the mixture was allowed to cool to room temperature, then refrigerated at 4°C for 1 hour. 50 μl of water was then added (oiled out), followed by the addition of 40 μl of ACN to get the sample back into solution. Seeds (synthesis described below) were added under slow stirring at room temperature. Crystals started to form ,and the mixture was allowed to sit overnight, with slow stirring. The next day, a heat cool cycle was applied (30°C for 10 minutes, 40°C for 10 minutes, then 50°C for 20 minutes). The heat was turned off and the mixture allowed to cool overnight, with slow stirring. The next day, a second heat/cool cycle was applied (60°C for 1 hour, with dissolving observed at 70°C). The heat was turned off and the mixture allowed to cool overnight, with slow stirring. The next day, crystals were present and a third heat cool cycle was applied (60°C for 3 hours). The heat was turned off and the mixture allowed to cool overnight, with slow stirring. The next day, a heat cool cycle was applied (60°C for 3 hours, slow cool, then 60°C for 3 hours). The heat was turned off and the mixture allowed to cool overnight, with slow stirring. After 3 days, the solid was filtered and placed on a high vacuum line to remove all solvent and give a freebase crystal ofthe title compound.

Seed Synthesis
109 mg ofthe product of Example 1 was dissolved in 0.56 ml of H2O:ACN (1:1). The suspension was left in a vial (cap loosely placed on top) to allow for a slower evaporation time. The vial was placed under a nitrogen flow environment, although the nitrogen was not used for evaporation, only for the environment. A precipitate was visible within 1 day, which was observed to be crystalline under a microscope. The solid was then placed on a high vacuum line to remove all solvent to give the freebase crystal.
Quantitative recovery, 97.8% pure by HPLC.

Example IE
Biphenyl-2-ylcarbamic acid 1 -(2- { [4-(4-carbamoylpiperidin- 1 -ylmethyl)benzoyl] methylamino} ethyl)piperidin-4-yl ester was also prepared as a freebase crystal using the following alternate procedure.
70 mg ofthe product of Example 1 was dissolved in 0.1 mL ACN. After addition of 0.3 ml MTBE, the solution appeared cloudy. An additional 50 μl of ACN was added to clarify the solution (155 mg/ml ACNMTBE = 1 :2). The mixture was left in the vial and capped. A solid appeared by the next day. The solid was then filtered and placed on a high vacuum line to remove all solvent and give a freebase crystal ofthe title compound.

PATENT

https://patents.google.com/patent/WO2012009166A1/en

U.S. Patent No. 7,228,657 to Mammen et al. discloses novel biphenyl compounds that are expected to be useful for treating pulmonary disorders such as chronic obstructive pulmonary disease and asthma. In particular, the compound biphenyl-2-ylcarbamic acid 1- (2- {[4-(4-carbamoylpiperidin-l-ylmethyl)benzoyl]methylamino}-ethyl)piperidin-4-yl ester is specifically described in this application as possessing muscarinic receptor antagonist or anticholiner ic activity, and is represented by formula I:

Figure imgf000002_0001

The compound of formula I is synthesized from the compound 8, which is described as being prepared from the oxidation of 2-(benzylmethylamino)ethanol to the aldehyde intermediate followed by reductive amination with biphenyl-2-yl-carbamic acid piperidin- 4-yl ester and debenzylation:

Figure imgf000003_0001
Figure imgf000003_0002

However, while this procedure performs well on small scale, the aldehyde intermediate is difficult to scale up due to its instability, and low yields were typically observed.

Thus, a need exists for an efficient process of preparing compound 8 as a pure material with high chemical purity and good overall yield, without having to isolate intermediates. This invention addresses those needs.

Therapeutic agents useful for treating pulmonary or respiratory disorders are advantageously administered directly into the respiratory tract by inhalation. In this regard, several types of pharmaceutical inhalation devices have been developed for administering therapeutic agents by inhalation including dry powder inhalers, metered- dose inhalers, and nebulizer inhalers. When preparing pharmaceutical compositions and formulations for use in such devices, it is highly desirable to have a crystalline form of the therapeutic agent that is neither hygroscopic nor deliquescent and which has a relatively high melting point thereby allowing the material to be micronized without significant decomposition.

A crystalline diphosphate of the compound of formula I has been reported in U.S. Patent No. 7,700,777 to Axt et al, and a crystalline freebase (identified as Form III) is described in U.S. Patent Application Publication No. 201 1/0015163 to Woollham. All of the aforementioned disclosures are incorporated herein by reference.

The compound of formula I is described as being prepared by reacting compound 8 with 4-carboxybenzaldehyde to form the aldehyde core 10:

Figure imgf000004_0001

which is then isolated prior to being combined with isonipicotamide in the presence of a reducing agent to form the compound of formula I. The crystalline diphosphate is prepared by contacting the separated and purified compound of formula I with phosphoric acid. The crystalline freebase (Form III) can then be prepared from the crystalline diphosphate.

A need also exists for an efficient process of preparing the crystalline freebase (Form III). It is desirable to develop a process that does not first require preparation of the crystalline diphosphate. This invention addresses those needs.

Figure imgf000011_0001
Figure imgf000013_0001
Figure imgf000014_0001

Preparation 1

Biphenyl-2-yl-carbamic acid piperidin-4-yl Ester

Figure imgf000018_0001

Biphenyl-2-isocyanate (97.5 g, 521 mmol) and 1 -benzylpiperidin-4-ol (105 g, 549 mmol) were heated together at 70°C for 12 hours. The mixture was then cooled to 50°C and EtOH (1 L) was added, followed by the slow addition of 6M HC1 (191 mL). The resulting mixture was then cooled to ambient temperature. Ammonium formate (98.5 g, 1.6 mol) was added and then nitrogen gas was bubbled through the solution vigorously for 20 minutes. Palladium on activated carbon (20 g, 10 wt% dry basis) was added and the mixture was heated at 40°C for 12 hours, and then filtered. The solvent was removed under reduced pressure and 1M HC1 (40 mL) was added to the crude residue. The pH of the mixture was adjusted with 10 N NaOH to pH 12. The aqueous layer was extracted with EtOAc (2×150 mL), and the organic layer was dried over MgS04, filtered and the solvent removed under reduced pressure to yield the title compound (155 g). HPLC (10-70) ¾ = 2.52; m/z: [M + H+] calcd for Ci8H2202 297.15; found 297.3.

EXAMPLE 1

Step A: (2,2-Dimethoxyethyl)methylcarbamic Acid Benzyl Ester

Figure imgf000018_0002

K2CO3 (13.8 g, 100 mmol, 1.76 eq.) and H20 (46 mL) were mixed to form a homogeneous solution. The solution was cooled to 20°C. N-methylaminoacetaldehyde dimethylacetal (12.8 mL, 100 mmol, 1.8 eq) and MeTHF (50 mL) were added. The resulting mixture was cooled to 2°C. Benzyl chloroformate (8.1 mL, 56.7 mmol, 1.0 eq.) was added by syringe over 10 minutes (addition was exothermic). The mixture was maintained at room temperature until completion of the reaction. The layers were separated and the organic layer was washed with IN HC1 (50 mL) and used directly in the next step.

Step B: Methyl-(2-oxoethyl)carbamic Acid Benzyl Ester

Figure imgf000019_0001

The mixture from the previous step was combined with a 3N HC1 solution (70 mL), and the resulting mixture was stirred for 18 hours at 22°C to yield a clear homogeneous pale yellow solution. Solid aHC03 was added to the solution to bring the pH to neutral. The layers were separated and the aqueous layer was back-extracted with MeTHF (20 mL). The organic layers were combined and washed with a saturated aHC03 solution (50 mL). The layers were separated and the organic layer was dried over Na2S04, filtered and concentrated to dryness to afford the title compound (1 1.9 g) as a pale yellow oil.

Step C: Biphenyl-2-yl-carbamic acid l-[2-(benzyloxycarbonyl

methylamino)ethyl]piperidin-4-yl Ester

Figure imgf000019_0002

Biphenyl-2-yl-carbamic acid piperidin-4-yl ester (31.1 g, 105 mmol, 1.0 eq.) and MeTHF (150 mL) were mixed. A solution of methyl-(2-oxoethyl)carbamic acid benzyl ester (23 g, 113.4 mmol, 1.05 eq.) in MeTHF (150 mL) was prepared and added to the ester mixture. The resulting mixture was heated to 30°C for a few minutes, then cooled to room temperature over 1 hour. The mixture was then cooled to 3°C and the temperature maintained for 1 hour. NaHB(OAc)3 (35.1 g, 170 mmol, 2.0 eq.) was added portion-wise while maintaining the internal temperature at 7±1°C. After addition, the mixture was allowed to warm to room temperature until the reaction was complete. A saturated solution of aHC03 (3000 mL) was added, stirred for 20 minutes, and the layers separated. This was repeated, after which the organic layer was dried over a2S04. The material was filtered, concentrated and dried under high vacuum to afford the title compound (43 g) as a thick colorless to pale yellow oil, which was used directly in the next step without purification.

Step D: Biphenyl-2-yl-carbamic acid l-(2-methylaminoethyl)piperidin-4-yl Ester

Figure imgf000020_0001

Biphenyl-2-yl-carbamic acid l-[2-(benzyloxycarbonyl methylamino)ethyl] piperidin-4-yl ester (53 g, 105 mmol, 1 eq.), MeOH (250 mL), and MeTHF (50 mL) were combined under nitrogen. 10% palladium on carbon (0.8 g) was added and hydrogen was bubbled into the mixture for 1 minute. The reaction vessel was sealed and stirred under hydrogen at atmospheric pressure for three hours. The mixture was then filtered, and the solids were washed MeTHF (10 mL).

The filtrate and washes were combined and concentrated under reduced pressure (250 mL removed). MTBE (100 mL) was added, and the solution again concentrated under reduced pressure (100 mL removed). MTBE (200 mL) was added and the solution was seeded with a few milligrams of biphenyl-2-yl-carbamic acid l-(2-methylaminoethyl) piperidin-4-yl ester, and the mixture was maintained for 3 hours. The solids were collected and the vessel and filter cake were washed with MTBE (2×15 mL). The material was dried to yield 13.2 g of the title compound (99.5% pure). This process was repeated to yield the title compound (12.5 g, 98.6% pure). The filtrate and washes were combined and concentrated under reduced pressure. MTBE (150 mL) was added and the solution was seeded with a few milligrams of biphenyl-2-yl-carbamic acid l-(2-methylaminoethyl) piperidin-4-yl ester, and the mixture was maintained for 20 hours. The solids were collected and the vessel and filter cake were washed with MTBE (2×15 mL). The material was dried to yield the title compound (5 g, 90% pure).

A portion of the three crops (13 g , 12 g, 4.5 g, respectively) were combined taken up in IPA (90 mL). The resulting slurry was heated to 45°C, then cooled to room temperature over 1 hour. The slurry was stirred for 5 hours at 25°C. The solids were collected and washed with IPA (2×15 mL). The solids were then dried for 1 hour to yield the title compound (25 g, >99% pure).

EXAMPLE 2

All volumes and molar equivalents are given relative to biphenyl-2-yl-carbamic acid piperidin-4-yl ester.

Step A: (2,2-Dimethoxyethyl)methylcarbamic Acid Benzyl Ester K2C03 (8.4 kg, 60 mol, 1.8 eq.) and H20 (49.3 kg, 2.6 volumes) were placed in the reaction vessel and stirred. N-methylaminoacetaldehyde dimethylacetal (6.5 kg, 54 mol, 1.6 eq) and MeTHF (20.2 kg, 2.9 volumes) were added. The resulting mixture was cooled to 5°C. Benzyl chloroformate (6.8 kg, 37.6 mol, 1.1 eq.) was added over a period of about 30 minutes, while maintaining the temperature below 10°C. The feed line was rinsed with MeTHF (4.3 kg). The mixture was then maintained at 5°C and stirred for 1 hour. The layers were separated and the organic layer was washed with IN HC1 (14.3 kg, 1 1.7 mol, 1.4 volumes) and used directly in the next step.

Step B: Methyl-(2-oxoethyl)carbamic Acid Benzyl Ester

The mixture from the previous step was combined with water (23.4 kg,

2.9 volumes) and 30% hydrochloric acid (13.1 kg, 107.7 mol, 1.1 volumes). Water (5.1 kg) was used to rinse the feed line. The temperature was adjusted to 25-30°C, and the reaction was run for 16-24 hours. A 25% NaOH solution (1 1.8 kg, 71.1 mol, 2.2 eq.) was added to the solution to adjust the pH and obtain phase separation.

The layers were separated and the aqueous layer was back-extracted with MeTHF

(10.0 kg, 1.1 volumes). The aqueous layer was discarded and the organic layers were combined. MeTHF (4.4 kg) was used to rinse the feed line. The organics were washed with a saturated aHC03 solution (14.6 kg, 15.6 mol, 1.1 volumes). The layers were separated and the organic layer was dried over a2S04 (2.5 kg, 17.6 mol) for 60-90 minutes. The drying agent was filtered off and the remaining solids were washed with

MeTHF (8.8 kg, 1 volume). The reaction vessel was washed with water and MeOH before continuing with the next step.

Step C: Biphenyl-2-yl-carbamic acid l-[2-(benzyloxycarbonyl

methylamino) ethyl Jpiperidin-4-yl Ester

The product from the previous step (in MeTHF) and biphenyl-2-yl-carbamic acid piperidin-4-yl ester (10.0 kg, 32.6 mol, 1.0 eq.) in MeTHF (28.5 kg) were placed in the reaction vessel and heated to 30°C for one hour. The mixture was then cooled to 5°C. NaHB(OAc)3 (10.0 kg, 45.8 mol, 1.4 eq.) was added portion wise over a period of 40 minutes while maintaining the temperature below 20°C. The mixture was then stirred for 30 minutes. Additional NaHB(OAc)3 (0.5 kg) was added the reaction allowed to progress to completion. A saturated solution of NaHCC^ (14.3 kg, 15.3 mol, 1.1 volumes) was added and stirred for 10 minutes. The aqueous phase was separated and discarded. A 33% NaOH solution (15.8 kg, 129.9 mol, 4.0 eq.) was added to the reaction mixture to adjust the H to be in the range of 8-12. Water (40 kg) was added in two portions, after which phase separation occurred. A saturated NaHCC (7.1 kg, 7.6 mol, 0.7 volumes) was added to the reaction mixture and stirred for 10 minutes. The aqueous phase was separated and discarded. Additional water (4.9 kg) was added to dissolve any remaining salts and a vacuum distillation was conducted at a maximum temperature of 45°C to remove part of the solvent (7.2 volumes). MeOH (56.1 kg, 7.2 volumes) was added to the reaction mixture before continuing with the next step.

Step D: Biphenyl-2-yl-carbamic acid l-(2-methylaminoethyl)piperidin-4-yl Ester

10% palladium on carbon (0.4 kg, 0.03 wt%, Degussa type 101 NE/W) was added to the reaction mixture. A hydrogenation reaction was performed to remove the benzyloxycarbonyl protective group, with reaction conditions at 30±5°C and 4 bar pressure. The reaction was run until completion. The mixture was then filtered and the filter cake was washed with MeOH (8.0 kg, 1.0 volume). The reaction was continued in a clean vessel, which was charged with the product solution (in MeTHF/MeOH) from the hydrogenation reaction. 3-Mercaptopropyl silica (0.6 kg, 0.07 wt%, Silicycle) was added. MeOH (4.8 kg) was used to rinse the feed line. The reaction mixture was stirred for 14-72 hours at 25±5°C. Activated carbon (0.7 kg, 0.07 wt%) was added and the mixture stirred for 30 minutes. The mixture was filtered and the filter cake was washed with MeOH (1.0 volume). The reaction was continued in a clean vessel, which was charged with the product solution (in MeTHF/MeOH), and MeOH (4.2 kg) was used to rinse the feed line. The mixture was heated to 40-45°C and a vacuum distillation was performed to bring the final volume to 5.6 volumes (removal of methanol).

2-propanol (40.2 kg, 5.0 volumes) was added and distillation continued until the volume was reduced to 2.5 volumes. The solids were then isolated by filtration and washed with MTBE (1.5 volumes) to yield the product as a wet cake (8.6 kg, 96.8% purity). The cake was charged to the reaction vessel and additional 2-propanol

(1.9 volumes) was added. The mixture was warmed to 40±5°C, and maintained at that temperature for 2 hours. The mixture was then slowly cooled over a minimum of 4 hours to 20°C, then actively cooled to 5-10°C, followed by stirring for 2 hours. The product was filtered and the resulting cake washed with MTBE (1.0 volume). The solids were then dried under atmospheric conditions to yield the title compound (6.6 kg, 98.5% purity).

EXAMPLE 3

Crystalline Freebase of Biphenyl-2-yl-carbamic Acid l- {2-r(4-carbamoylbenzoyl) methylaminolethyllpiperidin-4-yl Ester (Form III)

Biphenyl-2-yl-carbamic acid l-{2-[(4-formylbenzoyl)

methylamino ] ethyl }piperidin-4-yl Ester

Figure imgf000023_0001

4-Carboxybenzaldehyde (9 g, 60 mmol, 1.0 eq.) and biphenyl-2-yl-carbamic acid 1-

(2-methylaminoethyl)piperidin-4-yl ester (21.2 g, 60 mmol, 1.0 eq.) were combined in MeTHF (115 mL). The mixture was stirred for 0.5 hours, forming a thick slurry.

Additional MeTHF (50 mL) was added to form a free-flowing slurry. 4-(4,6-dimethoxy- l,3,5-triazin-2-yl)-4-methylmorpholinium chloride (18 g, 63 mmol, 1.1 eq., 97% pure) was added in two portions and the funnel rinsed with additional MeTHF (50 mL). The mixture was stirred at room temperature overnight. MeCN (50 mL) was added and the mixture was filtered. The solids were washed with MeTHF (30 mL). The filtrate and washes were combined and a saturated aHC03 solution (100 mL) was added and stirred for 10 minutes. The layers were separated and a saturated NaCl solution (100 mL) was added and stirred for 10 minutes. The layers were separated and the aqueous layer discarded. The resulting solution was concentrated under reduced pressure and held at room temperature for three days, then used directly in the next step.

Step B: Biphenyl-2-yl-carbamic acid l-{2-[(4-carbamoylbenzoyl)

meth lamino] ethyl}piperidin-4-yl ester (non-isolated form)

Figure imgf000023_0002

Isonipecotamide (15.4, 120 mmol, 2.0 eq.) and IPA (200 mL) were added to the solution of biphenyl-2-yl-carbamic acid l-{2-[(4-formylbenzoyl)methylamino]ethyl} piperidin-4-yl ester from the previous step. Liquid (200 mL) was distilled off and additional IPA (400 mL) was added under reduced pressure at 60°C. Liquid (400 mL) was distilled off over a period of 1.5 hours and additional IPA (600 mL) was added. Liquid (100 mL) was distilled off and the remaining solution was cooled to 30°C to yield a hazy white mixture, which was then added to Na2S04 (18 g). The flask was rinsed with IPA (100 mL) and added to the solution. The resulting mixture was cooled to room

temperature and AcOH (20 mL, 360 mmol, 6.0 eq.) was added. The mixture was cooled to 18°C with an ice bath and NaHB(OAc)3 (38.2 g, 180 mmol, 3.0 eq.) was added over 5 minutes. The mixture was allowed to warm up to 25°C and was maintained at that temperature for 2 hours. Solvent was removed under reduced pressure, and the remaining material was used directly in the next step.

Step C: Biphenyl-2-yl-carbamic acid l-{2-[(4-carbamoylbenzoyl)

methylamino]ethyl}piperidin-4-yl ester (isolated solid)

iPrOAc (300 mL) was added to the material, followed by the addition of water (200 mL). The pH of the solution was adjusted to pH 1 with 3N HC1 (-150 mL). The layers were separated and the organic layer was discarded. The aqueous layer was collected, and iPrOAc (300 mL) was added. The pH of the solution was adjusted to basic pH with 50 wt% NaOH (-100 mL). The resulting mixture was stirred for 15 minutes and the layers were separated. The organic layer was filtered and seeded with micronized crystalline freebase of biphenyl-2-yl-carbamic acid l- {2-[(4-carbamoylbenzoyl) methylamino]ethyl}piperidin-4-yl ester (Form III; prepared as described in U.S. Patent Application Publication No. 201 1/0015163 to Woollham) and stirred overnight at room temperature to yield a white slurry. Stirring was continued for 8 hours at room temperature and for 16 hours at 5°C (cold room). The mixture was slowly filtered under pressure. The cake was washed with cold iPrOAc (2×20 mL) and dried under nitrogen to yield a white solid (27.5 g). The material was further dried in a vacuum oven at 30°C for 24 hours to yield 25.9 g.

Step D: Crystalline Freebase of Biphenyl-2-yl-carbamic Acid l-{2-[ ( 4- carbamoylbenzoyl)methylamino]ethyl}piperidin-4-yl Ester (Form III) The white solid (5 g, 60 mmol, 1.0 eq.) was dissolved in toluene (75 mL) and the resulting mixture was heated to 82°C to yield a clear solution. The solution was filtered. The solids were washed with toluene (2 x 5 mL), and the filtrate and washes were combined. The mixture was cooled to 60°C and seeded with micronized crystalline freebase of biphenyl-2-yl-carbamic acid l-{2-[(4-carbamoylbenzoyl)methylamino]ethyl} piperidin-4-yl ester (Form III; prepared as described in Example 3 in U.S. Patent

Application Publication No. 201 1/0015163 to Woollham). The mixture was maintained at 55°C for 2 hours, then cooled to room temperature on an oil bath overnight (~16 hours). The resulting slurry was then filtered and the cake was dried for 3 hours to yield a solid while material (4.6 g). The material was further dried in a vacuum oven at 30°C for 24 hours (exhibited no further weight loss) to yield the title compound (4.6 g).

The product was analyzed by powder x-ray diffraction, differential scanning calorimetry and thermal gravimetric analysis, and was determined to be the crystalline freebase (Form III) of biphenyl-2-ylcarbamic acid l-(2-{[4-(4-carbamoylpiperidin-l- ylmethyl)benzoyl]methylamino}ethyl)piperidin-4-yl ester described in U.S. Patent Application Publication No. 201 1/0015163 to Woollham.

US20050113417A1 *2003-11-212005-05-26Mathai MammenCompounds having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
WO2006099165A1 *2005-03-102006-09-21Theravance, Inc.Crystalline forms of a biphenyl compound
US7228657B22003-07-102007-06-12Controlled Environments LimitedClimate control for a greenhouse
US20110015163A12009-07-152011-01-20Grahame WoollamCrystalline freebase forms of a biphenyl compound
Family To Family Citations
JP4555283B2 *2003-02-142010-09-29セラヴァンス, インコーポレーテッドβ2 adrenergic receptor agonist activity and biphenyl derivatives having muscarinic receptor antagonist activity
CN1930125B *2004-03-112010-07-21施万制药Biphenyl compounds useful as muscarinic receptor antagonists
US7659403B2 *2005-03-102010-02-09Theravance, Inc.Biphenyl compounds useful as muscarinic receptor antagonists
Patent ID Title Submitted Date Granted Date
US9226896 CRYSTALLINE FREEBASE FORMS OF A BIPHENYL COMPOUND
2014-11-19
2015-06-18
US9656993 CRYSTALLINE FORMS OF A BIPHENYL COMPOUND
2015-12-18
2016-06-16
US7700777 Crystalline forms of a biphenyl compound
2007-12-27
2010-04-20
Patent ID Title Submitted Date Granted Date
US9415041 Crystalline freebase forms of a biphenyl compound
2015-12-01
2016-08-16
US9249099 CRYSTALLINE FORMS OF A BIPHENYL COMPOUND
2014-11-25
2015-06-04
US8921396 Crystalline freebase forms of a biphenyl compound
2013-08-22
2014-12-30
US7521041 Biphenyl compounds useful as muscarinic receptor antagonists
2008-04-24
2009-04-21
US2007112027 Crystalline forms of a biphenyl compound
2007-05-17
Patent ID Title Submitted Date Granted Date
US8017783 Biphenyl compounds useful as muscarinic receptor antagonists
2008-03-20
2011-09-13
US7550595 Biphenyl compounds useful as muscarinic receptor antagonists
2007-12-20
2009-06-23
US9283183 BIPHENYL COMPOUNDS USEFUL AS MUSCARINIC RECEPTOR ANTAGONISTS
2014-11-12
2015-06-18
US2010048622 CRYSTALLINE FORMS OF A BIPHENYL COMPOUND
2010-02-25
US9452161 Biphenyl compounds useful as muscarinic receptor antagonists
2016-02-05
2016-09-27
Patent ID Title Submitted Date Granted Date
US8754225 PROCESS FOR PREPARING A BIPHENYL-2-YLCARBAMIC ACID
2012-01-19
US8921395 Crystalline forms of a biphenyl compound
2014-03-19
2014-12-30
US8716313 Crystalline forms of a biphenyl compound
2013-01-14
2014-05-06
US8557997 Biphenyl compounds useful as muscarinic receptor antagonists
2012-08-23
2013-10-15
US8541451 CRYSTALLINE FREEBASE FORMS OF A BIPHENYL COMPOUND
2011-01-20
Patent ID Title Submitted Date Granted Date
US8377965 CRYSTALLINE FORMS OF A BIPHENYL COMPOUND
2010-10-07
US8242137 CRYSTALLINE FORMS OF A BIPHENYL COMPOUND
2010-01-28
2012-08-14
US2017204061 BIPHENYL COMPOUNDS USEFUL AS MUSCARINIC RECEPTOR ANTAGONISTS
2016-08-30
US9765028 CRYSTALLINE FREEBASE FORMS OF A BIPHENYL COMPOUND
2016-07-11
US9035061 PROCESS FOR PREPARING A BIPHENYL-2-YLCARBAMIC ACID
2013-11-26
2014-05-01
Patent ID Title Submitted Date Granted Date
US7803812 BIPHENYL COMPOUNDS USEFUL AS MUSCARINIC RECEPTOR ANTAGONISTS
2009-09-10
2010-09-28
US7910608 Biphenyl compounds useful as muscarinic receptor antagonists
2009-01-15
2011-03-22
US7491736 Biphenyl compounds useful as muscarinic receptor antagonists
2007-12-20
2009-02-17
US7585879 Biphenyl compounds useful as muscarinic receptor antagonists
2007-11-15
2009-09-08
US7288657 Biphenyl compounds useful as muscarinic receptor antagonists
2005-09-15
2007-10-30
Patent ID Title Submitted Date Granted Date
US8912334 Biphenyl compounds useful as muscarinic receptor antagonists
2013-09-11
2014-12-16
US8273894 Biphenyl compounds useful as muscarinic receptor antagonists
2012-04-03
2012-09-25
US8173815 BIPHENYL COMPOUNDS USEFUL AS MUSCARINIC RECEPTOR ANTAGONISTS
2011-12-29
2012-05-08
US8053448 BIPHENYL COMPOUNDS USEFUL AS MUSCARINIC RECEPTOR ANTAGONISTS
2011-06-02
2011-11-08
US8034946 BIPHENYL COMPOUNDS USEFUL AS MUSCARINIC RECEPTOR ANTAGONISTS
2010-09-30
2011-10-11

/////////TD-4208, UNII:G2AE2VE07O, ревефенацин ريفيفيناسين 瑞维那新 , GSK 1160724, revefenacin, PHASE 3

CN(CCN1CCC(CC1)OC(=O)NC2=CC=CC=C2C3=CC=CC=C3)C(=O)C4=CC=C(C=C4)CN5CCC(CC5)C(=O)N

Viewing all 2878 articles
Browse latest View live


<script src="https://jsc.adskeeper.com/r/s/rssing.com.1596347.js" async> </script>