Quantcast
Channel: New Drug Approvals
Viewing all 2909 articles
Browse latest View live

ブレキサノロン , Brexanolone, Allopregnanolone

$
0
0

Allopregnanolone.png

ChemSpider 2D Image | Allopregnanolone | C21H34O2

Image result for Brexanolone

Brexanolone

318.501 g/mol, C21H34O2

CAS: 516-54-1

ブレキサノロン

MFCD00003677
Pregnan-20-one, 3-hydroxy-, (3α,5α)-
Pregnan-20-one, 3-hydroxy-, (3α,5α)- [ACD/Index Name]
S39XZ5QV8Y
TU4383000
UNII:S39XZ5QV8Y
(1S,2S,7S,11S,14S,15S,5R,10R)-14-acetyl-5-hydroxy-2,15-dimethyltetracyclo[8.7.0.0<2,7>.0<11,15>]heptadecane
(+)-3a-Hydroxy-5a-pregnan-20-one
(+)-3α-Hydroxy-5α-pregnan-20-one
(3α,5α)-3-Hydroxypregnan-20-one [ACD/IUPAC Name]
10446
3211363 [Beilstein]
3a-Hydroxy-5a-pregnan-20-one

The U.S. Food and Drug Administration today approved Zulresso (brexanolone) injection for intravenous (IV) use for the treatment of postpartum depression (PPD) in adult women. This is the first drug approved by the FDA specifically for PPD. 

https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm633919.htm?utm_campaign=031919_PR_FDA%20approves%20new%20drug%20for%20post-partum%20depression&utm_medium=email&utm_source=Eloqua

March 19, 2019

Release

The U.S. Food and Drug Administration today approved Zulresso (brexanolone) injection for intravenous (IV) use for the treatment of postpartum depression (PPD) in adult women. This is the first drug approved by the FDA specifically for PPD.

“Postpartum depression is a serious condition that, when severe, can be life-threatening. Women may experience thoughts about harming themselves or harming their child. Postpartum depression can also interfere with the maternal-infant bond. This approval marks the first time a drug has been specifically approved to treat postpartum depression, providing an important new treatment option,” said Tiffany Farchione, M.D., acting director of the Division of Psychiatry Products in the FDA’s Center for Drug Evaluation and Research. “Because of concerns about serious risks, including excessive sedation or sudden loss of consciousness during administration, Zulresso has been approved with a Risk Evaluation and Mitigation Strategy (REMS) and is only available to patients through a restricted distribution program at certified health care facilities where the health care provider can carefully monitor the patient.”

PPD is a major depressive episode that occurs following childbirth, although symptoms can start during pregnancy. As with other forms of depression, it is characterized by sadness and/or loss of interest in activities that one used to enjoy and a decreased ability to feel pleasure (anhedonia) and may present with symptoms such as cognitive impairment, feelings of worthlessness or guilt, or suicidal ideation.

Zulresso will be available only through a restricted program called the Zulresso REMS Program that requires the drug be administered by a health care provider in a certified health care facility. The REMS requires that patients be enrolled in the program prior to administration of the drug. Zulresso is administered as a continuous IV infusion over a total of 60 hours (2.5 days). Because of the risk of serious harm due to the sudden loss of consciousness, patients must be monitored for excessive sedation and sudden loss of consciousness and have continuous pulse oximetry monitoring (monitors oxygen levels in the blood). While receiving the infusion, patients must be accompanied during interactions with their child(ren). The need for these steps is addressed in a Boxed Warning in the drug’s prescribing information. Patients will be counseled on the risks of Zulresso treatment and instructed that they must be monitored for these effects at a health care facility for the entire 60 hours of infusion. Patients should not drive, operate machinery, or do other dangerous activities until feelings of sleepiness from the treatment have completely gone away.

The efficacy of Zulresso was shown in two clinical studies in participants who received a 60-hour continuous intravenous infusion of Zulresso or placebo and were then followed for four weeks. One study included patients with severe PPD and the other included patients with moderate PPD. The primary measure in the study was the mean change from baseline in depressive symptoms as measured by a depression rating scale. In both placebo controlled studies, Zulresso demonstrated superiority to placebo in improvement of depressive symptoms at the end of the first infusion. The improvement in depression was also observed at the end of the 30-day follow-up period.

The most common adverse reactions reported by patients treated with Zulresso in clinical trials include sleepiness, dry mouth, loss of consciousness and flushing. Health care providers should consider changing the therapeutic regimen, including discontinuing Zulresso in patients whose PPD becomes worse or who experience emergent suicidal thoughts and behaviors.

The FDA granted this application Priority Review and Breakthrough Therapydesignation.

Approval of Zulresso was granted to Sage Therapeutics, Inc.

Allopregnanolone, also known as 5α-pregnan-3α-ol-20-one or 3α,5α-tetrahydroprogesterone (3α,5α-THP), as well as brexanolone (USAN),[1] is an endogenous inhibitory pregnane neurosteroid[2] which has been approved by the FDA as a treatment for post-partum depression. It is synthesized from progesterone, and is a potent positive allosteric modulator of the action of γ-aminobutyric acid (GABA) at GABAA receptor.[2] Allopregnanolone has effects similar to those of other positive allosteric modulators of the GABA action at GABAA receptor such as the benzodiazepines, including anxiolyticsedative, and anticonvulsant activity.[2][3][4] Endogenously produced allopregnanolone exerts a pivotal neurophysiological role by fine-tuning of GABAA receptor and modulating the action of several positive allosteric modulators and agonists at GABAA receptor.[5] The 21-hydroxylated derivative of this compound, tetrahydrodeoxycorticosterone (THDOC), is an endogenous inhibitory neurosteroid with similar properties to those of allopregnanolone, and the 3β-methyl analogue of allopregnanolone, ganaxolone, is under development to treat epilepsy and other conditions, including post-traumatic stress disorder (PTSD).[2]

Biochemistry

Biosynthesis

The biosynthesis of allopregnanolone in the brain starts with the conversion of progesterone into 5α-dihydroprogesterone by 5α-reductase type I. After that, 3α-hydroxysteroid dehydrogenase converts this intermediate into allopregnanolone.[2] Allopregnanolone in the brain is produced by cortical and hippocampus pyramidal neurons and pyramidal-like neurons of the basolateral amygdala.[6]

Biological activity

Allopregnanolone acts as a highly potent positive allosteric modulator of the GABAA receptor.[2] While allopregnanolone, like other inhibitory neurosteroids such as THDOC, positively modulates all GABAA receptor isoforms, those isoforms containing δ subunitsexhibit the greatest potentiation.[7] Allopregnanolone has also been found to act as a positive allosteric modulator of the GABAA-ρ receptor, though the implications of this action are unclear.[8][9] In addition to its actions on GABA receptors, allopregnanolone, like progesterone, is known to be a negative allosteric modulator of nACh receptors,[10] and also appears to act as a negative allosteric modulator of the 5-HT3 receptor.[11] Along with the other inhibitory neurosteroids, allopregnanolone appears to have little or no action at other ligand-gated ion channels, including the NMDAAMPAkainate, and glycine receptors.[12]

Unlike progesterone, allopregnanolone is inactive at the nuclear progesterone receptor (nPR).[12] However, allopregnanolone can be intracellularly oxidized into 5α-dihydroprogesterone, which is an agonist of the nPR, and thus/in accordance, allopregnanolone does appear to have indirect nPR-mediated progestogenic effects.[13] In addition, allopregnanolone has recently been found to be an agonist of the newly discovered membrane progesterone receptors (mPR), including mPRδmPRα, and mPRβ, with its activity at these receptors about a magnitude more potent than at the GABAA receptor.[14][15] The action of allopregnanolone at these receptors may be related, in part, to its neuroprotective and antigonadotropic properties.[14][16] Also like progesterone, recent evidence has shown that allopregnanolone is an activator of the pregnane X receptor.[12][17]

Similarly to many other GABAA receptor positive allosteric modulators, allopregnanolone has been found to act as an inhibitor of L-type voltage-gated calcium channels (L-VGCCs),[18] including α1 subtypes Cav1.2 and Cav1.3.[19] However, the threshold concentration of allopregnanolone to inhibit L-VGCCs was determined to be 3 μM (3,000 nM), which is far greater than the concentration of 5 nM that has been estimated to be naturally produced in the human brain.[19] Thus, inhibition of L-VGCCs is unlikely of any actual significance in the effects of endogenous allopregnanolone.[19] Also, allopregnanolone, along with several other neurosteroids, has been found to activate the G protein-coupled bile acid receptor (GPBAR1, or TGR5).[20] However, it is only able to do so at micromolar concentrations, which, similarly to the case of the L-VGCCs, are far greater than the low nanomolar concentrations of allopregnanolone estimated to be present in the brain.[20]

Biological function

Allopregnanolone possesses a wide variety of effects, including, in no particular order, antidepressantanxiolyticstress-reducingrewarding,[21] prosocial,[22] antiaggressive,[23]prosexual,[22] sedativepro-sleep,[24] cognitivememory-impairmentanalgesic,[25] anestheticanticonvulsantneuroprotective, and neurogenic effects.[2] Fluctuations in the levels of allopregnanolone and the other neurosteroids seem to play an important role in the pathophysiology of moodanxietypremenstrual syndromecatamenial epilepsy, and various other neuropsychiatric conditions.[26][27][28]

Increased levels of allopregnanolone can produce paradoxical effects, including negative moodanxietyirritability, and aggression.[29][30][31] This appears to be because allopregnanolone possesses biphasic, U-shaped actions at the GABAA receptor – moderate level increases (in the range of 1.5–2 nM/L total allopregnanolone, which are approximately equivalent to luteal phase levels) inhibit the activity of the receptor, while lower and higher concentration increases stimulate it.[29][30] This seems to be a common effect of many GABAA receptor positive allosteric modulators.[26][31] In accordance, acute administration of low doses of micronized progesterone (which reliably elevates allopregnanolone levels) has been found to have negative effects on mood, while higher doses have a neutral effect.[32]

During pregnancy, allopregnanolone and pregnanolone are involved in sedation and anesthesia of the fetus.[33][34]

Chemistry

Allopregnanolone is a pregnane (C21) steroid and is also known as 5α-pregnan-3α-ol-20-one, 3α-hydroxy-5α-pregnan-20-one, or 3α,5α-tetrahydroprogesterone (3α,5α-THP). It is very closely related structurally to 5-pregnenolone (pregn-5-en-3β-ol-20-dione), progesterone (pregn-4-ene-3,20-dione), the isomers of pregnanedione (5-dihydroprogesterone; 5-pregnane-3,20-dione), the isomers of 4-pregnenolone (3-dihydroprogesterone; pregn-4-en-3-ol-20-one), and the isomers of pregnanediol (5-pregnane-3,20-diol). In addition, allopregnanolone is one of four isomers of pregnanolone (3,5-tetrahydroprogesterone), with the other three isomers being pregnanolone (5β-pregnan-3α-ol-20-one), isopregnanolone(5α-pregnan-3β-ol-20-one), and epipregnanolone (5β-pregnan-3β-ol-20-one).

Derivatives

A variety of synthetic derivatives and analogues of allopregnanolone with similar activity and effects exist, including alfadolone (3α,21-dihydroxy-5α-pregnane-11,20-dione), alfaxolone (3α-hydroxy-5α-pregnane-11,20-dione), ganaxolone (3α-hydroxy-3β-methyl-5α-pregnan-20-one), hydroxydione (21-hydroxy-5β-pregnane-3,20-dione), minaxolone (11α-(dimethylamino)-2β-ethoxy-3α-hydroxy-5α-pregnan-20-one), Org 20599 (21-chloro-3α-hydroxy-2β-morpholin-4-yl-5β-pregnan-20-one), Org 21465 (2β-(2,2-dimethyl-4-morpholinyl)-3α-hydroxy-11,20-dioxo-5α-pregnan-21-yl methanesulfonate), and renanolone (3α-hydroxy-5β-pregnan-11,20-dione).

Research

Allopregnanolone and the other endogenous inhibitory neurosteroids have short terminal half-lives and poor oral bioavailability, and for these reason, have not been pursued for clinical use as oral therapies, although development as a parenteral therapy for multiple indications has been carried out. However, synthetic analogs with improved pharmacokineticprofiles have been synthesized and are being investigated as potential oral therapeutic agents.

In other studies of compounds related to allopregnanolone, exogenous progesterone, such as oral micronized progesterone (OMP), elevates allopregnanolone levels in the body with good dose-to-serum level correlations.[35] Due to this, it has been suggested that OMP could be described as a prodrug of sorts for allopregnanolone.[35] As a result, there has been some interest in using OMP to treat catamenial epilepsy,[36] as well as other menstrual cycle-related and neurosteroid-associated conditions. In addition to OMP, oral pregnenolonehas also been found to act as a prodrug of allopregnanolone,[37][38][39] though also of pregnenolone sulfate.[40]

Allopregnanolone has been under development by Sage Therapeutics as an intravenously administered drug for the treatment of super-refractory status epilepticuspostpartum depression, and essential tremor.[41] As of 19 March 2019 the FDA has approved allopregnanolone for postpartum depression.

References

  1. ^ “ChemIDplus – 516-54-1 – AURFZBICLPNKBZ-SYBPFIFISA-N – Brexanolone [USAN] – Similar structures search, synonyms, formulas, resource links, and other chemical information”. NIH Toxnet. Retrieved 26 December 2017.
  2. Jump up to:a b c d e f g Reddy DS (2010). Neurosteroids: endogenous role in the human brain and therapeutic potentialsProg. Brain Res. Progress in Brain Research. 186. pp. 113–37. doi:10.1016/B978-0-444-53630-3.00008-7ISBN 9780444536303PMC 3139029PMID 21094889.
  3. ^ Reddy DS, Rogawski MA (2012). “Neurosteroids — Endogenous Regulators of Seizure Susceptibility and Role in the Treatment of Epilepsy”Jasper’s Basic Mechanisms of the Epilepsies, 4th Edition: 984–1002. doi:10.1093/med/9780199746545.003.0077ISBN 9780199746545.
  4. ^ T. G. Kokate, B. E. Svensson & M. A. Rogawski (September 1994). “Anticonvulsant activity of neurosteroids: correlation with γ-aminobutyric acid-evoked chloride current potentiation”. The Journal of Pharmacology and Experimental Therapeutics270 (3): 1223–1229. PMID 7932175.
  5. ^ Pinna, G; Uzunova, V; Matsumoto, K; Puia, G; Mienville, J. -M; Costa, E; Guidotti, A (2000-03-01). “Brain allopregnanolone regulates the potency of the GABAA receptor agonist muscimol”Neuropharmacology39 (3): 440–448. doi:10.1016/S0028-3908(99)00149-5PMID 10698010.
  6. ^ Agís-Balboa, Roberto C.; Pinna, Graziano; Zhubi, Adrian; Maloku, Ekrem; Veldic, Marin; Costa, Erminio; Guidotti, Alessandro (2006-09-26). “Characterization of brain neurons that express enzymes mediating neurosteroid biosynthesis”Proceedings of the National Academy of Sciences103 (39): 14602–14607. doi:10.1073/pnas.0606544103ISSN 0027-8424PMC 1600006PMID 16984997.
  7. ^ Mousavi Nik A, Pressly B, Singh V, Antrobus S, Hulsizer S, Rogawski MA, Wulff H, Pessah IN (2017). “Rapid Throughput Analysis of GABAA Receptor Subtype Modulators and Blockers Using DiSBAC1(3) Membrane Potential Red Dye”Mol. Pharmacol92 (1): 88–99. doi:10.1124/mol.117.108563PMC 5452057PMID 28428226.
  8. ^ Morris KD, Moorefield CN, Amin J (October 1999). “Differential modulation of the gamma-aminobutyric acid type C receptor by neuroactive steroids”. Mol. Pharmacol56 (4): 752–9. PMID 10496958.
  9. ^ Li W, Jin X, Covey DF, Steinbach JH (October 2007). “Neuroactive steroids and human recombinant rho1 GABAC receptors”J. Pharmacol. Exp. Ther323 (1): 236–47. doi:10.1124/jpet.107.127365PMC 3905684PMID 17636008.
  10. ^ Bullock AE, Clark AL, Grady SR, et al. (June 1997). “Neurosteroids modulate nicotinic receptor function in mouse striatal and thalamic synaptosomes”. J. Neurochem68 (6): 2412–23. doi:10.1046/j.1471-4159.1997.68062412.xPMID 9166735.
  11. ^ Wetzel CH, Hermann B, Behl C, et al. (September 1998). “Functional antagonism of gonadal steroids at the 5-hydroxytryptamine type 3 receptor”. Mol. Endocrinol12 (9): 1441–51. doi:10.1210/mend.12.9.0163PMID 9731711.
  12. Jump up to:a b c Mellon SH (October 2007). “Neurosteroid regulation of central nervous system development”Pharmacol. Ther116 (1): 107–24. doi:10.1016/j.pharmthera.2007.04.011PMC 2386997PMID 17651807.
  13. ^ Rupprecht R, Reul JM, Trapp T, et al. (September 1993). “Progesterone receptor-mediated effects of neuroactive steroids”. Neuron11 (3): 523–30. doi:10.1016/0896-6273(93)90156-lPMID 8398145.
  14. Jump up to:a b Thomas P, Pang Y (2012). “Membrane progesterone receptors: evidence for neuroprotective, neurosteroid signaling and neuroendocrine functions in neuronal cells”Neuroendocrinology96 (2): 162–71. doi:10.1159/000339822PMC 3489003PMID 22687885.
  15. ^ Pang Y, Dong J, Thomas P (January 2013). “Characterization, neurosteroid binding and brain distribution of human membrane progesterone receptors δ and {epsilon} (mPRδ and mPR{epsilon}) and mPRδ involvement in neurosteroid inhibition of apoptosis”Endocrinology154 (1): 283–95. doi:10.1210/en.2012-1772PMC 3529379PMID 23161870.
  16. ^ Sleiter N, Pang Y, Park C, et al. (August 2009). “Progesterone receptor A (PRA) and PRB-independent effects of progesterone on gonadotropin-releasing hormone release”Endocrinology150 (8): 3833–44. doi:10.1210/en.2008-0774PMC 2717864PMID 19423765.
  17. ^ Lamba V, Yasuda K, Lamba JK, et al. (September 2004). “PXR (NR1I2): splice variants in human tissues, including brain, and identification of neurosteroids and nicotine as PXR activators”. Toxicol. Appl. Pharmacol199 (3): 251–65. doi:10.1016/j.taap.2003.12.027PMID 15364541.
  18. ^ Hu AQ, Wang ZM, Lan DM, et al. (July 2007). “Inhibition of evoked glutamate release by neurosteroid allopregnanolone via inhibition of L-type calcium channels in rat medial prefrontal cortex”. Neuropsychopharmacology32 (7): 1477–89. doi:10.1038/sj.npp.1301261PMID 17151597.
  19. Jump up to:a b c Earl DE, Tietz EI (April 2011). “Inhibition of recombinant L-type voltage-gated calcium channels by positive allosteric modulators of GABAA receptors”J. Pharmacol. Exp. Ther337 (1): 301–11. doi:10.1124/jpet.110.178244PMC 3063747PMID 21262851.
  20. Jump up to:a b Keitel V, Görg B, Bidmon HJ, et al. (November 2010). “The bile acid receptor TGR5 (Gpbar-1) acts as a neurosteroid receptor in brain”. Glia58 (15): 1794–805. doi:10.1002/glia.21049PMID 20665558.
  21. ^ Rougé-Pont F, Mayo W, Marinelli M, Gingras M, Le Moal M, Piazza PV (July 2002). “The neurosteroid allopregnanolone increases dopamine release and dopaminergic response to morphine in the rat nucleus accumbens”. Eur. J. Neurosci16 (1): 169–73. doi:10.1046/j.1460-9568.2002.02084.xPMID 12153544.
  22. Jump up to:a b Frye CA (December 2009). “Neurosteroids’ effects and mechanisms for social, cognitive, emotional, and physical functions”Psychoneuroendocrinology. 34 Suppl 1: S143–61. doi:10.1016/j.psyneuen.2009.07.005PMC 2898141PMID 19656632.
  23. ^ Pinna G, Costa E, Guidotti A (February 2005). “Changes in brain testosterone and allopregnanolone biosynthesis elicit aggressive behavior”Proc. Natl. Acad. Sci. U.S.A102 (6): 2135–40. doi:10.1073/pnas.0409643102PMC 548579PMID 15677716.
  24. ^ Terán-Pérez G, Arana-Lechuga Y, Esqueda-León E, Santana-Miranda R, Rojas-Zamorano JÁ, Velázquez Moctezuma J (October 2012). “Steroid hormones and sleep regulation”Mini Rev Med Chem12 (11): 1040–8. doi:10.2174/138955712802762167PMID 23092405.
  25. ^ Patte-Mensah C, Meyer L, Taleb O, Mensah-Nyagan AG (February 2014). “Potential role of allopregnanolone for a safe and effective therapy of neuropathic pain”. Prog. Neurobiol113: 70–8. doi:10.1016/j.pneurobio.2013.07.004PMID 23948490.
  26. Jump up to:a b Bäckström T, Andersson A, Andreé L, et al. (December 2003). “Pathogenesis in menstrual cycle-linked CNS disorders”. Ann. N. Y. Acad. Sci1007: 42–53. doi:10.1196/annals.1286.005PMID 14993039.
  27. ^ Guille C, Spencer S, Cavus I, Epperson CN (July 2008). “The role of sex steroids in catamenial epilepsy and premenstrual dysphoric disorder: implications for diagnosis and treatment”Epilepsy Behav13 (1): 12–24. doi:10.1016/j.yebeh.2008.02.004PMC 4112568PMID 18346939.
  28. ^ Finocchi C, Ferrari M (May 2011). “Female reproductive steroids and neuronal excitability”. Neurol. Sci. 32 Suppl 1: S31–5. doi:10.1007/s10072-011-0532-5PMID 21533709.
  29. Jump up to:a b Bäckström T, Haage D, Löfgren M, et al. (September 2011). “Paradoxical effects of GABA-A modulators may explain sex steroid induced negative mood symptoms in some persons”. Neuroscience191: 46–54. doi:10.1016/j.neuroscience.2011.03.061PMID 21600269.
  30. Jump up to:a b Andréen L, Nyberg S, Turkmen S, van Wingen G, Fernández G, Bäckström T (September 2009). “Sex steroid induced negative mood may be explained by the paradoxical effect mediated by GABAA modulators”. Psychoneuroendocrinology34 (8): 1121–32. doi:10.1016/j.psyneuen.2009.02.003PMID 19272715.
  31. Jump up to:a b Bäckström T, Bixo M, Johansson M, et al. (February 2014). “Allopregnanolone and mood disorders”. Prog. Neurobiol113: 88–94. doi:10.1016/j.pneurobio.2013.07.005PMID 23978486.
  32. ^ Andréen L, Sundström-Poromaa I, Bixo M, Nyberg S, Bäckström T (August 2006). “Allopregnanolone concentration and mood–a bimodal association in postmenopausal women treated with oral progesterone”. Psychopharmacology187 (2): 209–21. doi:10.1007/s00213-006-0417-0PMID 16724185.
  33. ^ Mellor DJ, Diesch TJ, Gunn AJ, Bennet L (2005). “The importance of ‘awareness’ for understanding fetal pain”. Brain Res. Brain Res. Rev49 (3): 455–71. doi:10.1016/j.brainresrev.2005.01.006PMID 16269314.
  34. ^ Lagercrantz H, Changeux JP (2009). “The emergence of human consciousness: from fetal to neonatal life”Pediatr. Res65 (3): 255–60. doi:10.1203/PDR.0b013e3181973b0dPMID 19092726[…] the fetus is sedated by the low oxygen tension of the fetal blood and the neurosteroid anesthetics pregnanolone and the sleep-inducing prostaglandin D2 provided by the placenta (36).
  35. Jump up to:a b Andréen L, Spigset O, Andersson A, Nyberg S, Bäckström T (June 2006). “Pharmacokinetics of progesterone and its metabolites allopregnanolone and pregnanolone after oral administration of low-dose progesterone”. Maturitas54 (3): 238–44. doi:10.1016/j.maturitas.2005.11.005PMID 16406399.
  36. ^ Orrin Devinsky; Steven Schachter; Steven Pacia (1 January 2005). Complementary and Alternative Therapies for Epilepsy. Demos Medical Publishing. pp. 378–. ISBN 978-1-934559-08-6.
  37. ^ Saudan C, Desmarchelier A, Sottas PE, Mangin P, Saugy M (2005). “Urinary marker of oral pregnenolone administration”. Steroids70 (3): 179–83. doi:10.1016/j.steroids.2004.12.007PMID 15763596.
  38. ^ Piper T, Schlug C, Mareck U, Schänzer W (2011). “Investigations on changes in ¹³C/¹²C ratios of endogenous urinary steroids after pregnenolone administration”. Drug Test Anal3(5): 283–90. doi:10.1002/dta.281PMID 21538944.
  39. ^ Sripada RK, Marx CE, King AP, Rampton JC, Ho SS, Liberzon I (2013). “Allopregnanolone elevations following pregnenolone administration are associated with enhanced activation of emotion regulation neurocircuits”Biol. Psychiatry73 (11): 1045–53. doi:10.1016/j.biopsych.2012.12.008PMC 3648625PMID 23348009.
  40. ^ Ducharme N, Banks WA, Morley JE, Robinson SM, Niehoff ML, Mattern C, Farr SA (2010). “Brain distribution and behavioral effects of progesterone and pregnenolone after intranasal or intravenous administration”Eur. J. Pharmacol641 (2–3): 128–34. doi:10.1016/j.ejphar.2010.05.033PMC 3008321PMID 20570588.
  41. ^ “Brexanolone – Sage Therapeutics”. AdisInsight.

Further reading

Allopregnanolone
Skeletal formula of allopregnanolone
Ball-and-stick model of the allopregnanolone molecule
Names
IUPAC name
1-(3-Hydroxy-10,13-dimethyl-2,3,4,5,6,7,8,9,11,12,14,15,16,17-tetradecahydro-1H-cyclopenta[a]phenanthren-17-yl)ethanone
Other names
ALLO; Allo; ALLOP; AlloP; Brexanolone; 5α-Pregnan-3α-ol-20-one; 3α-Hydroxy-5α-pregnan-20-one; 3α,5α-Tetrahydroprogesterone; 3α,5α-THP; Zulresso
Identifiers
3D model (JSmol)
ChEMBL
ChemSpider
UNII
Properties
C21H34O2
Molar mass 318.501 g·mol−1
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).

//////////Brexanolone, Priority Review, Breakthrough Therapy designation, Zulresso, Sage Therapeutics Inc, FDA 2019, ブレキサノロン , Brexanolone, Allopregnanolone

CC(=O)C1CCC2C1(CCC3C2CCC4C3(CCC(C4)O)C)C


Industry-Oriented Route Evaluation and Process Optimization for the Preparation of Brexpiprazole

$
0
0
Abstract Image

Efforts toward route evaluation and process optimization for the preparation of brexpiprazole (1) are described. Starting from commercially available dihydroquinolinone 11, a three-step synthesis route composed of O-alkylation, oxidation, and N-alkylation was selected for industry-oriented process development aiming to reduce side reactions and achieve better impurity profiles. The reaction conditions of the three steps were investigated, and the control strategy for the process-related impurities was established. The optimized process was validated on the kilogram scale and now is viable for commercialization, with the results of not less than 99.90% purity of 1 (by HPLC) and not more than 0.05% of persistent impurities 15 and 16

Industry-Oriented Route Evaluation and Process Optimization for the Preparation of Brexpiprazole

Key Laboratory of Plant Resources and Chemistry in Arid Regions, Xinjiang Technical Institute of Physics and ChemistryChinese Academy of SciencesSouth Beijing Road 40−1, Urumqi, Xinjiang 830011, P. R. China
University of Chinese Academy of SciencesNo. 19A Yuquan Road, Beijing 100049, P. R. China
§CAS Key Laboratory for Receptor ResearchShanghai Institute of Materia Medica, Chinese Academy of Sciences555 Zuchongzhi Road, Shanghai 201203, P. R. China
Topharman Shanghai Co., Ltd.Building 1, No. 388 Jialilue Road, Zhangjiang Hitech Park, Shanghai 201209, P. R. China
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.8b00438
*Tel: +86-0991-3835679. Fax: +86-0991-3835679. E-mail: haji@ms.xjb.ac.cn., *Tel: +86-21-20231000-2407. Fax: +86-21-20231000-2407. E-mail: shenjingshan@simm.ac.cn.
ESI-MS: m/z = 434.22 [M + H].
1H NMR (500 MHz, DMSO-d6) δ (ppm): 11.61 (s, 1H), 7.80 (d, J = 9.4 Hz, 1H), 7.69 (d, J = 5.5 Hz, 1H), 7.61 (d, J = 8.0 Hz, 1H), 7.56 (d, J = 9.4 Hz, 1H), 7.40 (d, J = 5.5 Hz, 1H), 7.27 (d, J = 7.8 Hz, 1H), 6.87 (d, J = 7.6 Hz, 1H), 6.84–6.78 (m, 2H), 6.30 (d, J = 9.4 Hz, 1H), 4.05 (t, J = 6.4 Hz, 2H), 3.06 (brs, 4H), 2.61 (brs, 4H), 2.43 (t, J = 7.1 Hz, 2H), 1.86–1.75 (m, 2H), 1.69–1.57 (m, 2H).
13C NMR (125 MHz, DMSO-d6) δ (ppm): 162.35, 160.55, 148.36, 140.76, 140.49, 140.12, 133.47, 129.34, 125.92, 125.19, 121.99, 118.57, 116.73, 113.36, 112.11, 110.96, 98.68, 67.71, 57.47, 53.08, 51.83, 26.66, 22.81.
/////////////Brexpiprazole

Abikoviromycin

$
0
0
Abikoviromycin
ChemSpider 2D Image | abikoviromycin | C10H11NO
Abikoviromycin
  • Molecular FormulaC10H11NO
  • Average mass161.200 Da
CAS Registry Number: 31774-33-1
CAS Name: 7-Ethylidene-1a,2,3,7-tetrahydrocyclopent[b]oxireno[c]pyridine
(1aR,7E,7aS)-7-Ethylidene-1a,2,3,7-tetrahydrocyclopenta[b]oxireno[c]pyridine [ACD/IUPAC Name]
abikoviromycin
Cyclopent(b)oxireno(c)pyridine, 7-ethylidene-1a,2,3,7-tetrahydro-, (1aR,7E,7aS)-
Cyclopent[b]oxireno[c]pyridine, 7-ethylidene-1a,2,3,7-tetrahydro-, (1aR,7E,7aS)-
Additional Names: 4,4a-epoxy-5-ethylidene-2,3,4,4a-tetrahydro-5H-1-pyridine; abicoviromycin; latumcidin
Molecular Formula: C10H11NO
Molecular Weight: 161.20
Percent Composition: C 74.51%, H 6.88%, N 8.69%, O 9.93%
Literature References: Antiviral antibiotic produced by Streptomyces abikoensis and Streptomyces rubescens. Chromatographic isoln from broth cultures: Umezawa et al., Jpn. Med. J. 4, 331 (1951); C.A. 46, 7167 (1952); Umezawa, JP 54 6200 (1954 to Nippon). Identity with latumcidin: Sakagami et al., J. Antibiot. 11A, 231 (1958). Structure: Gurevich et al., Tetrahedron Lett. 1968,2209. Stereochemistry: Kono et al., J. Antibiot. 23, 572 (1970); Gurevich et al., Khim. Prir. Soedin. 7, 104 (1971), C.A. 75, 5752e (1971). Crystal and molecular structure of the selenate: Y. Kono et al., Acta Crystallogr. B27, 2341 (1971). In vitro antiviral activity: V. M. Roikhel, N. A. Zeitlenok, Antibiotiki 14, 969 (1969), C.A. 72, 19394q (1969).
Properties: Highly unstable and polymerizes promptly on isolation even at -50°; however, it can be handled in dilute solutions and in the form of its salts. uv max (neutral ethanol or 0.1N KOH): 218, 244, 289 nm (log e 3.83, 3.99, 3.94); (0.1N HCl) 236, 341 nm (log e 3.99, 4.05).
Absorption maximum: uv max (neutral ethanol or 0.1N KOH): 218, 244, 289 nm (log e 3.83, 3.99, 3.94); (0.1N HCl) 236, 341 nm (log e 3.99, 4.05)
Isolation of abikoviromycin and dihydroabikoviromycin as inhibitors of polyketide synthase involved in melanin biosynthesis by Colletotrichum lagenarium
Journal of Antibiotics (2003), 56, (9), 801-804.
purified by normal-phase HPLC (column: Senshu-Pak Aquasil SS-752N, 10×250mm, Senshu Kagaku; mobile phase: isocratic elution of nhexane: 2-propanol: H2O: triethylamine, 70:30:1:0.02; flow rate: 5ml/minutes; retention time: 9.0 minutes) to obtain 1 (6.3mg). 1:
FAB-MS (NBA matrix) m/z 162 (M+H)+; [α]20D+67.5° (c 0.025, 0.1N NaOH) [lit. [α]21D +148.9° (c 1, 0.1N NaOH)]9);
1H NMR (500MHz, CDCl3) δ 7.43 (1H, d, J=6.5Hz, 7-H), 6.53 (1H, d, J=6.5Hz, 6- H), 5.50 (1H, q, J=7.0Hz, 8-H), 3.92 (1H, s, 4-H), 3.81 (1H, dd, J=5.5, 15Hz, 2-Ha), 3.69 (1H, dt, J=5.5, 15Hz, 2-Hb), 2.19 (1H, m, 3-Ha), 1.90 (3H, d, J=7.0Hz, 9-H), 1.62 (1H, m, 3-Hb);
13C NMR (125MHz, CDCl3) δ 172.1 (C-7a), 142.3 (C-7), 136.5 (C-5), 132.8 (C-6), 119.5 (C-8), 59.5 (C-4), 54.5 (C-4a), 44.5 (C-2), 21.8 (C-3),14.0 (C-9).
////////////Abikoviromycin

“ALL FOR DRUGS” 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

READ

ANTHONY MELVIN CRASTO
NDA
DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO …..FOR BLOG HOME CLICK HERE
Join me on Linkedin

View Anthony Melvin Crasto Ph.D's profile on LinkedIn

Join me on Facebook FACEBOOK

Join me on twitterFollow amcrasto on Twitter
Join me on google plus Googleplus

 amcrasto@gmail.com

CALL +919323115463  INDIA
//////////////

Batimastat, バチマスタット

$
0
0

Batimastat

Batimastat.svg

Batimastat
バチマスタット
Formula
C23H31N3O4S2
cas
130370-60-4
Mol weight
477.6399
Butanediamide, N4-hydroxy-N1-(2-(methylamino)-2-oxo-1-(phenylmethyl)ethyl)-2-(2-methylpropyl)-3-((2-thienylthio)methyl)-, (2R-(1(S*),2R*,3S*))-
DESCARBOXY-NOR-N(ω)-HYDROXY-L-ARGININE
DSX
EJ6675000
UNII:BK349F52C9
(2R,3S)-N4-Hydroxy-2-isobutyl-N1-[(2S)-1-(methylamino)-1-oxo-3-phenylpropan-2-yl]-3-[(2-thienylsulfanyl)methyl]succinamide
(2R,3S)-N4-hydroxy-N1-[(2S)-1-(methylamino)-1-oxo-3-phenylpropan-2-yl]-2-(2-methylpropyl)-3-[(thiophen-2-ylsulfanyl)methyl]butanediamide
(2R,3S)-N4-Hydroxy-N1-[(1S)-2-(methylamino)-2-oxo-1-(phenylmethyl)ethyl]-2-(2-methylpropyl)-3-[(2-thienylthio)methyl]butanediamide
(2S,3R)-5-Methyl-3-(((aS)-a-(methylcarbamoyl)phenethyl)carbamoyl)-2-((2-thienylthio)methyl)hexanohydroxamic Acid
[2R-[1(S*),2R*,3S*]]-N4-Hydroxy-N1-[2-(methylamino)-2-oxo-1-(phenylmethyl)ethyl]-2-(2-methylpropyl)-3-[(2-thienylthio)methyl]butane Diamide
130370-60-4 [RN]
7154
BK349F52C9
CAS Registry Number: 130370-60-4
CAS Name: (2R,3S)-N4-Hydroxy-N1-[(1S)-2-(methylamino)-2-oxo-1-(phenylmethyl)ethyl]-2-(2-methylpropyl)-3-[(2-thienylthio)methyl]butanediamide
Additional Names: (2S,3R)-5-methyl-3-[[(aS)-a-(methylcarbamoyl)phenethyl]carbamoyl]-2-[(2-thienylthio)methyl]hexanohydroxamic acid; [4-(N-hydroxyamino)-2R-isobutyl-3S-(2-thienylthiomethyl)succinyl]-L-phenylalanine-N-methylamide
Manufacturers’ Codes: BB-94
Molecular Formula: C23H31N3O4S2
Molecular Weight: 477.64
Percent Composition: C 57.84%, H 6.54%, N 8.80%, O 13.40%, S 13.43%
Literature References: Synthetic matrix metalloproteinase inhibitor. Prepn: C. Campion et al., WO 9005719eidem, US 5240958(1990, 1993 both to British Biotech.). Effect on transplanted human ovarian carcinoma: B. Davies et al., Cancer Res. 53, 2087 (1993). Inhibition of metastasis of transplanted human colorectal carcinoma: X. Wang et al., ibid. 54, 4726 (1994).
Properties: Fine white powder. mp 236-238°.
Melting point: mp 236-238°
Therap-Cat: Antineoplastic adjunct (antimetastatic agent).
Keywords: Antineoplastic Adjunct; Antimetastatic Agent; Matrix Metalloproteinase Inhibitor.

Batimastat (INN/USAN, codenamed BB-94) is an anticancer drug that belongs to the family of drugs called angiogenesis inhibitors. It acts as a matrix metalloproteinase inhibitor (MMPI) by mimicking natural MMPI peptides.

Batimastat was the first MMPI that went into clinical trials. First results of a Phase I trial appeared in 1994. The drug reached Phase III but was never marketed; mainly because it couldn’t be administered orally (as opposed to the newer and chemically similar MMPI marimastat), and injection into the peritoneum caused peritonitis.[1]

SYN

U.S. Patent 5,453,438

U.S. Patent 5,240,958

U.S. Patent 5,530,161

Image result for batimastat

SYN

US 5240958; US 5310763; WO 9005719

The treatment of D-leucine (I) with NaNO2, H2SO4 and NaBr gives 2(R)-bromo-5-methylpentanoic acid (II), which is esterified with isobutene and H2SO4 to the corresponding tert-butyl ester (III). The condensation of (III) with dibenzyl malonate (IV) by means of potassium tert-butoxide in DMF yields the malonyl derivative (V), which is treated with trifluoroacetic acid to hydrolyze the tert-butyl ester, and without isolation is condensed with L-phenylalanine methyl amide (VI) by means of hydroxybenzotriazole (HOBT) and dicyclohexylcarbodiimide (DCC), affording 4-benzyloxy-3-(benzyloxycarbonyl)-2(R)-isobutylsuccinyl-L-phenylalanine methylamide (VII). The elimination of the benzyl groups of (VII) by hydrogenolysis over Pd/C in ethanol gives the dicarboxylic acid (VIII), which by partial decarboxylation and reaction with aqueous formaldehyde and piperidine yields 4-hydroxy-2(R)-isobutyl-3-methylenesuccinyl-L-phenylalanine methylamide (IX). The addition of thiophene-2-thiol (X) to the double bond of (IX) affords 4-hydroxy-2(R)-isobutyl-3(S)-(2-thienylsulfanylmethyl)succinyl-L-phenylalanine methylamide (XI), which is finally treated with hydroxylamine and hydroxybenzotriazole in dichloromethane/DMF.

SPEC

HPLC

References

  1. ^ Rothenberg, M. L.; Nelson, A. R.; Hande, K. R. (1999). “New Drugs on the Horizon: Matrix Metalloproteinase Inhibitors”. Stem Cells17 (4): 237–240. doi:10.1002/stem.170237PMID 10437989.
Batimastat
Batimastat.svg
Clinical data
Pregnancy
category
  • N/A
Routes of
administration
Injection into pleural space or abdomen
ATC code
  • none
Legal status
Legal status
  • Never marketed
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
ECHA InfoCard 100.222.897 Edit this at Wikidata
Chemical and physical data
Formula C23H31N3O4S2
Molar mass 477.64 g/mol g·mol−1
3D model (JSmol)

//////////Batimastat, BB-94, バチマスタット  ,

[H][C@@](CC1=CC=CC=C1)(NC(=O)[C@]([H])(CC(C)C)[C@]([H])(CSC1=CC=CS1)C(=O)NO)C(=O)NC

FDA approves new oral testosterone capsule for treatment of men with certain forms of hypogonadism

$
0
0

FDA approves new oral testosterone capsule (testosterone undecanoate) for treatment of men with certain forms of hypogonadism

March 27, 2019

Release

The U.S. Food and Drug Administration today approved Jatenzo (testosterone undecanoate), an oral testosterone capsule to treat men with certain forms of hypogonadism. These men have low testosterone levels due to specific medical conditions, such as genetic disorders like Klinefelter syndrome or tumors that have damaged the pituitary gland. Jatenzo should not be used to treat men with “age-related hypogonadism,” in which testosterone levels decline due to aging, even if these men have symptoms that appear to be related to low testosterone. Jatenzo’s benefits do not outweigh its risks for that use.

“Jatenzo’s oral route of administration provides an important addition to current treatment options available for men with certain hypogonadal conditions who up until now have most commonly been treated with testosterone products that are applied to the skin or injected,” said Hylton V. Joffe, M.D, M.M.Sc., director of the Division of Bone, Reproductive and Urologic Products in the FDA’s Center for Drug Evaluation and Research. “But it’s important to emphasize that this drug should not, like other testosterone treatments, be used to treat older men with ‘age-related hypogonadism.’ The benefits of testosterone therapy, including Jatenzo, have not been established for this use, and Jatenzo’s effects on raising blood pressure can increase the risks of heart attack, stroke and cardiovascular death in this population.”

The efficacy of Jatenzo was demonstrated in a four-month clinical trial involving 166 men with hypogonadism. Study participants initially were given Jatenzo at a dose of 237 mg twice per day, and the dose was adjusted downward or upward to a maximum of 396 mg twice per day on the basis of testosterone levels. Eighty-seven percent of Jatenzo-treated men achieved an average testosterone level within the normal range, which was the primary study endpoint.

Jatenzo contains a boxed warning on its labeling stating that the drug can cause blood pressure to rise, increasing the risk of heart attack, stroke and cardiovascular death. Health care providers should consider a patient’s individual heart disease risks and ensure that blood pressure is adequately controlled before prescribing Jatenzo; they should also periodically monitor patient blood pressure during treatment. Jatenzo is currently one of two testosterone products that have this boxed warning. The FDA is requiring all testosterone product manufacturers to conduct blood pressure postmarketing trials to more clearly address whether these products increase blood pressure.

Common side effects, occurring in more than 2 percent of patients in the Jatenzo clinical trial, included headache, an increase in hematocrit (red blood cell count), a decrease in high-density lipoprotein cholesterol (“good” cholesterol), high blood pressure and nausea. An increase in prostate specific antigen (PSA) was also observed. Patients should have their hematocrit, cholesterol and PSA monitored regularly to check for changes. Those with benign prostate hyperplasia should be monitored for worsening of symptoms.

The FDA granted the approval of Jatenzo to Clarus Therapeutics.

//////////FDA 2019, Jatenzo, Clarus Therapeutics, (testosterone undecanoate,

https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm634585.htm?utm_campaign=032719_PR_FDA%20approves%20new%20oral%20testosterone%20capsule&utm_medium=email&utm_source=Eloqua

Cevimeline, セビメリン

$
0
0

Cevimeline.svg

Cevimeline

セビメリン

  • Molecular FormulaC10H17NOS
  • Average mass199.313 Da
cis-2′-Methylspiro[4-azabicyclo[2.2.2]octane-2,5′-[1,3]oxathiolane]
Evoxac [Trade name]
Spiro[1-azabicyclo[2.2.2]octane-3,5′-[1,3]oxathiolane], 2′-methyl-, (2’R,3R)-
Cevimeline
CAS Registry Number: 107233-08-9
CAS Name: (2¢R,3R)-rel-2¢-Methylspiro[1-azabicyclo[2.2.2]octane-3,5¢-[1,3]oxathiolane]
Additional Names: (±)-cis-2-methylspiro[1,3-oxathiolane-5,3¢-quinuclidine]
Molecular Formula: C10H17NOS
Molecular Weight: 199.31
Percent Composition: C 60.26%, H 8.60%, N 7.03%, O 8.03%, S 16.09%
Literature References: Muscarinic M1 and M3 receptor agonist. Prepn: A. Fisher et al., JP Kokai 61 280497eidemUS 4855290; (1986, 1989 both to State of Israel). Improved process: K. Hayashi et al., US 5571918 (1996 to Ishihara Sangyo Kaisha). Sialogogic effect in animals: H. Masunaga et al., Eur. J. Pharmacol. 339, 1 (1997). General pharmacology: H. Arisawa et al., Arzneim.-Forsch. 52, 14, 81 (2002). Clinical experience in Sjögren’s syndrome dry eye: M. Ono et al., Am. J. Ophthalmol. 138, 6 (2004); in dry mouth: K. Suzuki et al., Pharmacology 74, 100 (2005). Review of clinical pharmacokinetics and efficacy in Sjögren’s syndrome: H. Yasuda, H. Niki, Clin. Drug Invest. 22, 67-73 (2002).
Derivative Type: Hydrochloride hemihydrate
CAS Registry Number: 153504-70-2; 107220-28-0 (anhydrous)
Manufacturers’ Codes: AF-102B; SNI-2011
Trademarks: Evoxac (Daiichi)
Molecular Formula: C10H17NOS.HCl.½H2O
Molecular Weight: 244.78
Percent Composition: C 49.07%, H 7.82%, N 5.72%, O 9.80%, S 13.10%, Cl 14.48%
Properties: White to off white crystalline powder, mp 201-203°. Freely sol in alcohol, chloroform; very sol in water. Virtually insol in ether.
Melting point: mp 201-203°
Therap-Cat: Sialagogue.
Keywords: Sialagogue.

Cevimeline hydrochloride

    • Synonyms:AF-102B, SNI-2011, SNK-508, Evoxac
    • ATC:N07
  • Use:cognition disorder, treatment of Sjogren’s syndrome, muscarinic M3-receptor agonist
  • Chemical name:(2′R,3R)-rel-2′-methylspiro[1-azabicyclo[2.2.2]octane-3,5′-[1,3]oxathiolane] hydrochloride hydrate (2:2:1)
  • Formula:C10H17NOS • HCl • 1/2H2O
  • MW:489.57 g/mol
  • CAS-RN:153504-70-2
  • InChI Key:SURWTGAXEIEOGY-GHXDPTCOSA-N
  • InChI:InChI=1S/C10H17NOS.ClH/c1-8-12-10(7-13-8)6-11-4-2-9(10)3-5-11;/h8-9H,2-7H2,1H3;1H/t8-,10-;/m1./s1

Derivatives

base

  • Formula:C10H17NOS
  • MW:199.32 g/mol
  • CAS-RN:107233-08-9

anhydrous hydrochloride

  • Formula:C10H17NOS • HCl
  • MW:235.78 g/mol
  • CAS-RN:107220-28-0

Cevimeline is cis-2′-methylspiro {1-azabicyclo [2.2.2] octane-3, 5′ -[1,3] oxathiolane} hydro-chloride, hydrate (2:1). Its empirical formula is C10H17NOS•HCl•½ H2O, and its structural formula is:

Image result for Cevimeline STRUCTURECevimeline has a molecular weight of 244.79. It is a white to off white crystalline powder with a melting point range of 201 to 203°C. It is freely soluble in alcohol and chloroform, very soluble in water, and virtually insoluble in ether. The pH of a 1% solution ranges from 4.6 to 5.6. Inactive ingredients include lactose monohydrate, hydroxypropyl cellulose, and magnesium stearate.

Image result for Cevimeline STRUCTURE

Image result for Cevimeline STRUCTURE

Cevimeline hydrochloride [USAN]
RN: 153504-70-2

 (+-)-cis-2-Methylspiro(1,3-oxathiolane-5,3′-quinuclidine) hydrochloride, hemihydrate

Cevimeline (trade name Evoxac) is a parasympathomimetic and muscarinic agonist,[1] with particular effect on M1 and M3 receptors. It is used in the treatment of dry mouth and especially associated with Sjögren’s syndrome.

Mechanism of action

By activating the M3 receptors of the parasympathetic nervous system, cevimeline stimulates secretion by the salivary glands, thereby alleviating dry mouth.

Side effects

Known side effects include nauseavomitingdiarrhea, excessive sweatingrashheadacherunny nosecoughdrowsinesshot flashesblurred vision, and difficulty sleeping.[2]

Contraindications include asthma and angle closure glaucoma.

Clip

https://www.sciencedirect.com/science/article/abs/pii/S0731708515302260

Image result for cevimeline

Image result for cevimeline

Image result for cevimeline

CLIP

https://www.sciencedirect.com/science/article/pii/S0040403913005042

Image result for cevimeline

Image result for cevimeline

Image result for cevimeline

CLIP

CLIP

  • Reaction of quinuclidin-3-one (I) with trimethylsulfoxonium iodide and NaH in DMSO gives epoxide (II), which is opened with SH2 in NaOH/water, yielding 3-hydroxy-3-(sulfanylmethyl)quinuclidine (III). The cyclization of compound (III) with acetaldehyde (IV) catalyzed by boron trifluoride ethearate or by SnCl4, POCl3, H3PO4 or p-toluenesulfonic acid affords a mixture of two diastereomeric spiroracemates, the (?-trans (V) and (?-cis (cevimeline). This mixture is separated by fractional recrystallization in acetone or by TLC chromatography, and treated with hydrochloric acid. The (?-trans-compound (V) can be isomerized to cevimeline by treatment with an acidic catalyst such as an organic sulfonic acid (trifluoromethanesulfonic acid, p-toluenesulfonic acid or methanesulfonic acid), a Lewis acid (SnCl4, FeCl3, BF3 or AlCl3) or sulfuric acid in refluxing toluene, hexane or CHCl3. Cevimeline hydrochloride hemihydrate is obtained from the above mentioned hydrochloride by a complex work-up using water, isopropanol and n-hexane.
  • Synthesis of Cevimeline Hydrochloride (EN:134916): Reaction of quinuclidin-3-one (I) with trimethylsulfoxonium iodide and NaH in DMSO gives epoxide (II), which is opened with SH2 in NaOH/water, yielding 3-hydroxy-3-(sulfanylmethyl)quinuclidine (III) (1,2). The cyclization of compound (III) with acetaldehyde (IV) catalyzed by boron trifluoride ethearate (1) or by SnCl4, POCl3, H3PO4 or p-toluenesulfonic acid (2) affords a mixture of two diastereomeric spiro-racemates, the (?-trans (V) and (?-cis (cevimeline). This mixture is separated by fractional recrystallization in acetone or by TLC chromatography, and treated with hydrochloric acid (1,2). The (?-trans-compound (V) can be isomerized to cevimeline by treatment with an acidic catalyst such as an organic sulfonic acid (trifluoromethanesulfonic acid, p-toluenesulfonic acid or methanesulfonic acid), a Lewis acid (SnCl4, FeCl3, BF3 or AlCl3) or sulfuric acid in refluxing toluene, hexane or CHCl3 (2,3). Cevimeline hydrochloride hemihydrate is obtained from the above mentioned hydrochloride by a complex work-up using water, isopropanol and n-hexane (4).(Scheme 13491601a) Description M.p. 203 C (4). Sources Discovered by Israel Institute for Biological Research, Ness-Ziona (IL) and licensed to Snow Brand Milk Products Co. Ltd. (JP). In the U.S., comarketed by Snow Brand Milk Products and Daiichi Pharmaceutical Co., Ltd. In Japan, codeveloped with Nippon Kayaku Co. Ltd. Ishihara Sangyo Co., Ltd. (JP) is the bulk supplier. References 1. Fisher, A., Heldman, E., Grunfeld, Y., Karton, I., Levy, A. (Israel Institute for Biological Research); Derivs. of quinuclidine; EP 0205247, JP 1986280497, US 4855290. 2. Hayashi, K., Tokumoto, S., Yoshizawa, H., Isogai, T. (Ishihara Sangyo Kaisha, Ltd.); Method for producing 2-methylspiro(1,3-oxathiolan-5,3′)quinuclidine; EP 0683168, US 5571918. 3. Haga, T., Koyanagi, T., Hara, K., Maeda, M., Shigehara, I. (Ishihara Sangyo Kaisha, Ltd.); Method for isomerization of trans-form 2-methylspiro(1,3-oxathiolane-5,3′)quinuclidine or acid addition salts thereof; EP 0298491, US 4861886. 4. Saito, K., Ono, T., Honda, N. (Snow Brand Milk Products Co., Ltd.); Preparation method of cis-2-methylspiro(1,3-oxathiolane-5,3′)quinuclidine hydrochloride.1/2 hydrate capable of disgregating easily; JP 1992108792.

PATENT

https://patents.google.com/patent/US8080663B2/en

The present invention refers to a novel, industrially advantageous process for the preparation of an intermediate useful for the preparation of Cevimeline hydrochloride (1, cis-2-methylspiro(1,3-oxathiolane-5,3′)quiniclidine, Scheme 1). This pharmaceutical is useful for the treatment of diseases of the central nervous system due to disturbances of central cholinergic function and autoimmune system (Sjörgen’s syndrome) and is marketed as Evoxac®.

U.S. Pat. No. 4,855,290 describes a process for preparation of 2-methylspiro(1,3-oxathiolane-5,3′)quiniclidine (1). The process comprises the preparation of the epoxide of 3-methylenequiniclidine, which is subsequently reacted with hydrogen sulfide to produce 3-hydroxy-3-mercaptomethylquiniclidine and condensed with acetaldehyde in the presence of a Lewis acid (boron trifluoride etherate) to provide 2-methylspiro(1,3-oxathiolane-5,3′)quiniclidine. This process is depicted in Scheme I.

Figure US08080663-20111220-C00001

This process suffers from major disadvantages when transiting to industrial scale. These include the use of the highly hazardous and difficult to handle hydrogen sulfide gas. Also, boron trifluoride etherate is employed during the condensation step with acetaldehyde. The boron trifluoride etherate reagent is an air and moisture sensitive Lewis acid which has to be used under anhydrous conditions, thus creating a serious disadvantage in industrial settings. Another drawback of this process is the use of sodium hydride. U.S. Pat. Nos. 5,571,918 and 4,861,886 relate to the isomerization of the trans- to cis-form of 2-methylspiro(1,3-oxathiolane-5,3′)quiniclidine but do not describe methods for its preparation. Thus, an industrially acceptable and cost-effective method for the preparation of Cevimeline hydrochloride which overcomes the deficiencies of the prior art is required.

Further and other objects of the invention will be realized by those skilled in the art from the following Summary of the Invention and Detailed Description of Preferred Embodiments of the Invention thereof.

According to one aspect of the invention, a novel process is provided for the preparation of 2-methylspiro(1,3-oxathiolane-5,3′)quiniclidine (1). The process is industrially practical, efficient, safe and economical, as well as being environmentally friendly. The general method is shown in the Scheme II.

Figure US08080663-20111220-C00002


wherein R is selected from C1 to C6 alkyl and aryl groups, most preferably a methyl, ethyl or propyl group; Ris hydrogen or a C2 to C7 alkyl or aryl carbonyl group; Ris a C1 to C6 alkyl group, preferably methyl, ethyl, propyl, or butyl group.

Figure US08080663-20111220-C00003

EXAMPLE I Preparation of the Epoxide of 3-methylenequiniclidine (3)

A mixture of the hydrochloric salt of 3-quiniclidinone (2, 120 g, 795.7 mmol) and trimethylsulfoxonium iodide (219 g, 993.3 mmol) in dimethylsulfoxide (91.0 g, 0.63 mol) was cooled to 0-5° C. in an ice/water bath under nitrogen atmosphere. A solution of potassium tert-butoxide (201 g, 1789.1 mmol) in dimethylsulfoxide (500 mL) was added dropwise over 45 minutes. The mixture was warmed gradually to room temperature and stirred for an additional 16 hours at room temperature. After cooling to 0-5° C. (ice/water bath) the mixture was poured into an ice/water mixture (500 g) and then sodium chloride (300 g) was added. The mixture was stirred for 30 minutes and extracted with toluene (3×400 mL). The toluene phase was dried over sodium sulfate, filtered and evaporated to furnish the epoxide of 3-methylenequiniclidine (60 g, 431.7 mmol, 54% yield) as a yellow oil. The product could be used in the next step neat or as toluene solution after the extraction without further purification.

1H NMR (400 MHz, CDCl3): δ=3.10 (d, 1H, J=14.6 Hz); 2.98-2.77 (m, 5H); 2.74 (d, 1H, J=4.8 Hz); 2.70 (d, 1H, J=4.8 Hz); 1.96-1.89 (m, 1H); 1.79-1.62 (m, 2H); 1.60-1.54 (m, 1H); 1.38-1.36 (m,1H).

LRMS (ES+): 140.0 (100, M+H+).

EXAMPLE II Preparation of the Thiolacetic Acid Salt of 3-hydroxy-3-acetoxymercaptomethylquiniclidine (4)

A solution of the epoxide of 3-methylenequiniclidine (3, 54 g, 388.5 mmol) in toluene (200 mL) was cooled to 0-5° C. (ice/water bath). Thiolacetic acid was added dropwise over 10-15 minutes. The mixture was stirred at 0-5° C. for 30 minutes and then allowed to come to room temperature. After stirring at room temperature for 2 hours the formed precipitate was filtered and washed with toluene (2×100 mL) to give the 3-hydroxy-3-acetoxymercaptomethylquiniclidine thiolacetic acid salt (4 wherein Ris H and R is methyl, 77 g, 264.6 mmol, 68%) as a light yellow solid. The product was used in the next step without any further purification.

1H NMR (400 MHz CD3OD): δ=3.47 (d, 1H, J=14.1 Hz); 3.37-3.18 (m, 7H); 2.40 (s, 3H); 2.38 (s, 3H); 2.36-2.27 (m, 1H), 2.14-2.05 (m, 2H); 2.03-1.93 (m, 1H); 1.81-1.78 (m, 1H).

LRMS (ES+): 216.1 (100, M−[SCOCH3]+H+).

EXAMPLE III Preparation of 2-methylspiro(1,3-oxathiolane-5,3′)quiniclidine using p-toluenesulfonic acid (1)

To a solution of 3-hydroxy-3-acetoxymercaptomethylquiniclidine thiolacetic acid salt (4 wherein Ris H and R is methyl, 3 g, 10.3 mmol) in iso-propanol (50 mL) was added p-toluenesulfonic acid monohydrate (5.9 g, 30.9 mmol) and the mixture was heated to reflux for 3.5 hours. The mixture was cooled to room temperature and acetaldehyde diethyl acetal (6.1 g, 51.5 mmol) was added. The mixture was heated to reflux and stirred for an additional 3 hours. The solvent was evaporated and the residue was dissolved in dichloromethane (50 mL). The mixture was cooled to 0-5° C. and a 25% aqueous solution of sodium hydroxide (80 mL) was added. The mixture was stirred for 10-15 minutes and the phases were separated. The aqueous phase was extracted with dichloromethane (3×50 mL). The organic phases were combined and extracted with 5% aqueous solution of sulfuric acid (3×50 mL). The acidic aqueous phases were combined and the pH was adjusted to 12 with a 25% aqueous solution of sodium hydroxide. The aqueous phase was extracted with heptane (3×50 mL) and the organic phases were combined, dried over sodium sulfate and the solvent was evaporated to give 2-methylspiro(1,3-oxathiolane-5,3′)quiniclidine (1.8 g, 9.2 mmol, 89% yield) as a 3:1 cis/trans ratio mixture of diastereomers (determined by 1H NMR).

LRMS (ES+): 200.1 (100, M+H+).

EXAMPLE IV Preparation of 2-methylspiro(1,3-oxathiolane-5,3′)quiniclidine (1) using racemic camphorsulfonic acid

In a similar experiment as Example III, racemic camphorsulfonic acid (7.2 g, 30.9 mmol) was added to a solution of 3-hydroxy-3-acetoxymercaptomethylquiniclidine thiolacetic acid salt (4 wherein Ris H and R is methyl, 3 g, 10.3 mmol) in iso-propanol (50 mL). The mixture was refluxed for 5 h, cooled to room temperature and acetaldehyde diethyl acetal (6.1 g, 51.5 mmol) was added. The mixture was refluxed for an additional an 8 hours and processed according to Example III to give 2-methylspiro(1,3-oxathiolane-5,3′)quiniclidine (1.32 g, 6.63 mmol, 64% yield) in a 3.5:1 cis/trans ratio mixture of diastereomers (determined by 1H NMR).

EXAMPLE V Preparation of 2-methylspiro(1,3-oxathiolane-5,3′)quiniclidine (1) using phenyl sulfonic acid

In a similar experiment as Example III, to a solution of 3-hydroxy-3-acetoxymercaptomethylquiniclidine thiolacetic acid salt (4 wherein Ris H and R is methyl, 3 g, 10.3 mmol) in iso-propanol (50 mL) was added phenyl sulfonic acid (4.9 g, 30.9 mmol) and the mixture was refluxed 5 h, cooled to room temperature and acetaldehyde diethyl acetal (6.1 g, 51.5 mmol) was added. The mixture was refluxed for an additional 8 hours and worked up in a manner similar to Example III to furnish 2-methylspiro(1,3-oxathiolane-5,3′)quiniclidine (1.6 g, 8.2 mmol, 80% yield) as a 2.5:1 cis/trans ratio mixture of diastereomers (determined by 1H NMR).

EXAMPLE VI Preparation of 2-methylspiro(1,3-oxathiolane-5,3′)quiniclidine (1) using p-toluenesulfonic acid in butanol

To a solution of 3-hydroxy-3-acetoxymercaptomethylquiniclidine thiolacetic acid salt (4 wherein Ris H and R is methyl, 3 g, 10.3 mmol) in butanol (100 mL) was added of p-toluenesulfonic acid monohydrate (5.9 g, 30.9 mmol) and the mixture was refluxed for 3 hours with a Dean-Stark apparatus attached to the flask. The reaction mixture was cooled to room temperature and acetaldehyde diethyl acetal (6.1 g, 51.5 mmol) was added. The mixture was heated to 80° C. for an additional 8 h and worked up according to Example III to afford 2-methylspiro(1,3-oxathiolane-5,3′)quiniclidine (1.8 g, 9.2 mmol, 89% yield) as a 3:1 cis/trans ratio mixture of diastereomers (determined by 1H NMR).

References

  1. ^ Ono M, Takamura E, Shinozaki K, et al. (July 2004). “Therapeutic effect of cevimeline on dry eye in patients with Sjögren’s syndrome: a randomized, double-blind clinical study”Am. J. Ophthalmol138 (1): 6–17. doi:10.1016/j.ajo.2004.02.010PMID 15234277.
  2. ^ [1] MedicineNet: Cevimeline. Accessed 10/12/2007
      • US 4 855 290 (Israel Institute for Biological Research; 8.8.1989; IL-prior. 10.5.1985).
      • US 4 876 260 (Israel Institute for Biological Research; 24.10.1989; USA-prior. 28.10.1987).
      • EP 683 168 (Ishihara Sangyo Kaisha; appl. 19.5.1995; J-prior. 19.5.1994).
    • Method for isomerization of trans-isomer:

      • US 4 861 886 (Ishihara Sangyo Kaisha; 29.8.1989; J-prior. 10.7.1987).
    • Method of separation:

      • IL 81 652 (Israel Institute for Biological Research; 12.5.1991; appl. 23.2.1987).
      • JP 01 290 680 (Ishihara Sangyo Kaisha; 22.11.1989; J-prior. 18.5.1988).
    • Synthesis of enantiomerically pure (S)-3-hydroxy-3-mercaptomethylquinuclidine (S)-II:

      • Bos, M.; Canesso, R.: Heterocycles (HTCYAM) 38 (8), 1889 (1994).
    • Synthesis of 3-quinuclidone:

      • Sternbach, L.H.; Kaiser, S.: J. Am. Chem. Soc. (JACSAT) 74, 2215 (1952).

External links

Cevimeline
Cevimeline.svg
Cevimeline 3D.png
Clinical data
Trade names Evoxac
AHFS/Drugs.com Monograph
MedlinePlus a608025
Pregnancy
category
  • C
Routes of
administration
By mouth (capsules)
ATC code
Legal status
Legal status
  • In general: ℞ (Prescription only)
Pharmacokinetic data
Protein binding <20%
Identifiers
CAS Number
PubChem CID
DrugBank
ChemSpider
UNII
KEGG
ChEBI
ChEMBL
Chemical and physical data
Formula C10H17NOS
Molar mass 199.31308 g/mol g·mol−1
3D model (JSmol)

/////////// Cevimeline, AF-102B, SNI-2011, SNK-508, Evoxac, セビメリン

FDA approves new oral treatment for multiple sclerosis, Mavenclad (cladribine)

$
0
0
FDA approves new oral treatment for multiple sclerosis, Mavenclad (cladribine)
The U.S. Food and Drug Administration today approved Mavenclad (cladribine) tablets to treat
relapsing forms of multiple sclerosis (MS) in adults, to include relapsing-remitting disease and active secondary progressive disease. Mavenclad is not recommended for MS patients with clinically isolated syndrome. Because of its safety profile, the use of Mavenclad is generally recommended for patients who have had an inadequate response to…

March 29, 2019

Release

The U.S. Food and Drug Administration today approved Mavenclad (cladribine) tablets to treat relapsing forms of multiple sclerosis (MS) in adults, to include relapsing-remitting disease and active secondary progressive disease. Mavenclad is not recommended for MS patients with clinically isolated syndrome. Because of its safety profile, the use of Mavenclad is generally recommended for patients who have had an inadequate response to, or are unable to tolerate, an alternate drug indicated for the treatment of MS.

“We are committed to supporting the development of safe and effective treatments for patients with multiple sclerosis,” said Billy Dunn, M.D., director of the Division of Neurology Products in the FDA’s Center for Drug Evaluation and Research. “The approval of Mavenclad represents an additional option for patients who have tried another treatment without success.”

MS is a chronic, inflammatory, autoimmune disease of the central nervous system that disrupts communications between the brain and other parts of the body. Most people experience their first symptoms of MS between the ages of 20 and 40. MS is among the most common causes of neurological disability in young adults and occurs more frequently in women than in men.

For most people, MS starts with a relapsing-remitting course, in which episodes of worsening function (relapses) are followed by recovery periods (remissions). These remissions may not be complete and may leave patients with some degree of residual disability. Many, but not all, patients with MS experience some degree of persistent disability that gradually worsens over time. In some patients, disability may progress independent of relapses, a process termed secondary progressive multiple sclerosis (SPMS). In the first few years of this process, many patients continue to experience relapses, a phase of the disease described as active SPMS. Active SPMS is one of the relapsing forms of MS, and drugs approved for the treatment of relapsing forms of MS can be used to treat active SPMS.

The efficacy of Mavenclad was shown in a clinical trial in 1,326 patients with relapsing forms of MS who had least one relapse in the previous 12 months. Mavenclad significantly decreased the number of relapses experienced by these patients compared to placebo. Mavenclad also reduced the progression of disability compared to placebo.

Mavenclad must be dispensed with a patient Medication Guide that describes important information about the drug’s uses and risks. Mavenclad has a Boxed Warning for an increased risk of malignancy and fetal harm. Mavenclad is not to be used in patients with current malignancy. In patients with prior malignancy or with increased risk of malignancy, health care professionals should evaluate the benefits and risks of the use of Mavenclad on an individual patient basis. Health care professionals should follow standard cancer screening guidelines in patients treated with Mavenclad. The drug should not be used in pregnant women and in women and men of reproductive potential who do not plan to use effective contraception during treatment and for six months after the course of therapy because of the potential for fetal harm. Mavenclad should be stopped if the patient becomes pregnant.

Other warnings include the risk of decreased lymphocyte (white blood cell) counts; lymphocyte counts should be monitored before, during and after treatment. Mavenclad may increase the risk of infections; health care professionals should screen patients for infections and treatment with Mavenclad should be delayed if necessary. Mavenclad may cause hematologic toxicity and bone marrow suppression so health care professionals should measure a patient’s complete blood counts before, during and after therapy. The drug has been associated with graft-versus-host-disease following blood transfusions with non-irradiated blood. Mavenclad may cause liver injury and treatment should be interrupted or discontinued, as appropriate, if clinically significant liver injury is suspected.

The most common adverse reactions reported by patients receiving Mavenclad in the clinical trials include upper respiratory tract infections, headache and decreased lymphocyte counts.

The FDA granted approval of Mavenclad to EMD Serono, Inc.

////////////fda 2019, Mavenclad, cladribine, multiple sclerosis, EMD Serono,

FDA approves treatment Cimzia (certolizumab pegol) for patients with a type of inflammatory arthritis

$
0
0

FDA approves treatment Cimzia (certolizumab pegol) for patients with a type of inflammatory arthritis

March 28, 2019

Release

The U.S. Food and Drug Administration today approved Cimzia (certolizumab pegol) injection for treatment of adults with a certain type of inflammatory arthritis called non-radiographic axial spondyloarthritis (nr-axSpA), with objective signs of inflammation. This is the first time that the FDA has approved a treatment for nr-axSpA.

“Today’s approval of Cimzia fulfills an unmet need for patients suffering from non-radiographic axial spondyloarthritis as there has been no FDA-approved treatments until now,” said Nikolay Nikolov, M.D., associate director for rheumatology of the Division of Pulmonary, Allergy, and Rheumatology Products in the FDA’s Center for Drug Evaluation and Research.

Nr-axSpA is a type of inflammatory arthritis that causes inflammation in the spine and other symptoms. There is no visible damage seen on x-rays, so it is referred to as non-radiographic.

The efficacy of Cimzia for the treatment of nr-axSpA was studied in a randomized clinical trial in 317 adult patients with nr-axSpA with objective signs of inflammation, indicated by elevated C-reactive protein (CRP) levels and/or sacroiliitis (inflammation of the sacroiliac joints) on MRI. The trial measured the improvement response on the Ankylosing Spondylitis Disease Activity Score, a composite scoring system that assesses disease activity including patient-reported outcomes and CRP levels. Responses were greater for patients treated with Cimzia compared to patients treated with placebo. The overall safety profile observed in the Cimzia treatment group was consistent with the known safety profile of Cimzia.

The prescribing information for Cimzia includes a Boxed Warning to advise health care professionals and patients about the increased risk of serious infections leading to hospitalization or death including tuberculosis (TB), bacterial sepsis (infection in the blood steam), invasive fungal infections (such as histoplasmosis, an infection that affects the lungs), and other infections. Cimzia should be discontinued if a patient develops a serious infection or sepsis. Health care providers are advised to perform testing for latent TB and, if positive, to start treatment for TB prior to starting Cimzia. All patients should be monitored for active TB during treatment, even if the initial latent TB test is negative. The Boxed Warning also advises that lymphoma (cancer in blood cells) and other malignancies, some fatal, have been reported in children and adolescent patients treated with tumor necrosis factor (TNF) blockers, of which Cimzia is a member. Cimzia is not indicated for use in pediatric patients. Cimzia must be dispensed with a patient Medication Guide that describes important information about the drug’s uses and risks.

Cimzia was originally approved in 2008 and is also indicated for adult patients with Crohn’s disease, moderate-to-severe rheumatoid arthritis, active ankylosing spondylitis (AS) and moderate-to-severe plaque psoriasis who are candidates for systemic therapy or phototherapy.

The FDA granted the approval of Cimzia to UCB.

 

///////////////FDA 2019, Cimzia, certolizumab pegol, inflammatory arthritis, UCB

https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm634671.htm?utm_campaign=032819_PR_FDA%20approves%20treatment%20for%20patients%20with%20a%20type%20of%20inflammatory%20arthritis&utm_medium=email&utm_source=Eloqua


Certolizumab pegol, セルトリズマブペゴル (遺伝子組換え)

$
0
0

Image result for certolizumab pegol

>Amino acid sequence of the light chain
DIQMTQSPSSLSASVGDRVTITCKASQNVGTNVAWYQQKPGKAPKALIYSASFLYSGVPY
RFSGSGSGTDFTLTISSLQPEDFATYYCQQYNIYPLTFGQGTKVEIKRTVAAPSVFIFPP
SDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLT
LSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
>Amino acid sequence of the heavy chain
EVQLVESGGGLVQPGGSLRLSCAASGYVFTDYGMNWVRQAPGKGLEWMGWINTYIGEPIY
ADSVKGRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCARGYRSYAMDYWGQGTLVTVSSAS
TKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGL
YSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCAA

Certolizumab pegol

CAS: 428863-50-7

セルトリズマブペゴル (遺伝子組換え)

CDP 870 / CDP-870 / CDP870 / PHA-738144

Formula
C2115H3252N556O673S16
Cas
428863-50-7
Mol weight
47748.8128

Reducing signs and symptoms of Crohn’s disease and treatment of moderately to severely active rheumatoid arthritis (RA).

Certolizumab pegol is a recombinant Fab’ antibody fragment against tumor necrosis factor alpha which is conjugated to an approximately 40kDa polyethylene glycol (PEG2MAL40K). Polyethylene glycol helps to delay the metabolism and elimination of the drugs. Chemically, the light chain is made up of 214 amino acid residues while the heavy chain is composed of 229 amino acid residues. The molecular mass of the Fab’ antibody fragment itself is 47.8 kDa. It is used for the treatment of rheumatoid arthritis and Crohn’s disease. FDA approved on April 22, 2008

Certolizumab pegol (CDP870, tradename Cimzia) is a biologic medication for the treatment of Crohn’s disease,[1][2] rheumatoid arthritispsoriatic arthritis and ankylosing spondylitis. It is a fragment of a monoclonal antibody specific to tumor necrosis factor alpha(TNF-α) and is manufactured by UCB.[3][4][5]

Image result for certolizumab pegol

Medical uses

Crohn’s Disease
On April 22, 2008, the U.S. FDA approved Cimzia for the treatment of Crohn’s disease in people who did not respond sufficiently or adequately to standard therapy.[4][6][7]
Rheumatoid arthritis
On June 26, 2009, the Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency (EMA) issued a positive opinion recommending that the European Commission grant a marketing authorisation for Cimzia for the treatment of rheumatoid arthritis only – the CHMP refused approval for the treatment of Crohn’s disease. The marketing authorisation was granted to UCB Pharma SA on October 1, 2009.[8]
Psoriatic arthritis
On September 27, 2013, the U.S. FDA approved Cimzia for the treatment of adult patients with active psoriatic arthritis.[9]

Method of action

Certolizumab pegol is a monoclonal antibody directed against tumor necrosis factor alpha. More precisely, it is a PEGylated Fabfragment of a humanized TNF inhibitor monoclonal antibody.[10]

Clinical trials

Crohn’s disease
Positive results have been demonstrated in two phase III trials (PRECiSE 1 and 2) of certolizumab pegol versus placebo in moderate to severe active Crohn’s disease.[1][10][11][12]
Axial spondyloarthritis
In 2013, a phase 3 double blind randomized placebo-controlled study found significantly positive results in patient self-reported questionnaires, with rapid improvement of function and pain reduction, in patients with axial spondyloarthritis.[13]
Rheumatoid arthritis
Certolizumab appears beneficial in those with rheumatoid arthritis.[14]

Side effects

Significant side effects occur in 2% of people who take the medication.[14]

References

  1. Jump up to:a b Sandborn WJ, Feagan BG, Stoinov S, et al. (July 2007). “Certolizumab pegol for the treatment of Crohn’s disease”N. Engl. J. Med357 (3): 228–38. doi:10.1056/NEJMoa067594PMC 3187683PMID 17634458.
  2. ^ Goel, Niti; Sue Stephens (2010). “Certolizumab pegol”mAbs2 (2): 137–147. doi:10.4161/mabs.2.2.11271PMC 2840232PMID 20190560.
  3. ^ Kaushik VV, Moots RJ (April 2005). “CDP-870 (certolizumab) in rheumatoid arthritis”. Expert Opinion on Biological Therapy5 (4): 601–6. doi:10.1517/14712598.5.4.601PMID 15934837.
  4. Jump up to:a b index.cfm?fuseaction=Search.Label_ApprovalHistory “Cimzia Label and Approval History” Check |url= value (help)Drugs@FDAU.S. Food and Drug Administration(FDA). Retrieved 2009-11-15.
  5. ^ “Cimzia Prescribing Information” (PDF). US Food and Drug Administration (FDA). April 2016. Retrieved 2016-08-21.
  6. ^ UCB press release – Cimzia Approved in the US for the Treatment of Moderate to Severe Crohn’s Disease. Retrieved April 22, 2008.
  7. ^ Waknine, Yael (May 1, 2008). “FDA Approvals: Patanase, Actonel, Cimzia”Medscape. Retrieved 2008-05-01.
  8. ^ “Cimzia European Public Assessment Report”European Medicines Agency. Retrieved November 15, 2009.
  9. ^ “Cimzia (certolizumab pegol) approved by the U.S. FDA for treatment of adult patients with active psoriatic arthritis”. Archived from the original on October 1, 2013. Retrieved October 1, 2013.
  10. Jump up to:a b Schreiber S. et al., Certolizumab pegol, a humanised anti-TNF pegylated FAb’ fragment, is safe and effective in the maintenance of response and remission following induction in active Crohn’s disease: a phase 3 study (precise), Gut, 2005, 54, suppl7, A82
  11. ^ Sandborn et al., Certolizumab pegol administered subcutaneously is effective and well tolerated in patients with active Crohn’s disease: results from a 26-week, placebo-controlled Phase 3 study (PRECiSE 1), Gastroenterology, 2006, 130, A107
  12. ^ “New Analysis Shows Cimzia (Certolizumab Pegol) Maintained Remission and Response in Recent Onset Crohn’s Disease” (Press release). UCB. October 23, 2006. Retrieved 2009-11-15.
  13. ^ Sieper J, Tubergen A, Coteur G, Woltering F, Landewe R (May 2013). “PMS50 – Rapid Improvements In Patient-Reported Outcomes With Certolizumab Pegol In Patients With Axial Spondyloarthritis, Including Ankylosing Spondylitis And Non-Radiographic Axial Spondyloarthritis: 24-Week Results Of A Phase 3 Double Blind Randomized Placebo-Controlled Study”. Value in Health16 (3): A227. doi:10.1016/j.jval.2013.03.1150.
  14. Jump up to:a b Ruiz Garcia, V; Jobanputra, P; Burls, A; Vela Casasempere, P; Bort-Marti, S; Bernal, JA (Sep 8, 2017). “Certolizumab pegol (CDP870) for rheumatoid arthritis in adults”(PDF)The Cochrane Database of Systematic Reviews9: CD007649. doi:10.1002/14651858.CD007649.pub4PMID 28884785.

External links

FDA approves treatment Cimzia (certolizumab pegol) for patients with a type of inflammatory arthritis

March 28, 2019

Release

The U.S. Food and Drug Administration today approved Cimzia (certolizumab pegol) injection for treatment of adults with a certain type of inflammatory arthritis called non-radiographic axial spondyloarthritis (nr-axSpA), with objective signs of inflammation. This is the first time that the FDA has approved a treatment for nr-axSpA.

“Today’s approval of Cimzia fulfills an unmet need for patients suffering from non-radiographic axial spondyloarthritis as there has been no FDA-approved treatments until now,” said Nikolay Nikolov, M.D., associate director for rheumatology of the Division of Pulmonary, Allergy, and Rheumatology Products in the FDA’s Center for Drug Evaluation and Research.

Nr-axSpA is a type of inflammatory arthritis that causes inflammation in the spine and other symptoms. There is no visible damage seen on x-rays, so it is referred to as non-radiographic.

The efficacy of Cimzia for the treatment of nr-axSpA was studied in a randomized clinical trial in 317 adult patients with nr-axSpA with objective signs of inflammation, indicated by elevated C-reactive protein (CRP) levels and/or sacroiliitis (inflammation of the sacroiliac joints) on MRI. The trial measured the improvement response on the Ankylosing Spondylitis Disease Activity Score, a composite scoring system that assesses disease activity including patient-reported outcomes and CRP levels. Responses were greater for patients treated with Cimzia compared to patients treated with placebo. The overall safety profile observed in the Cimzia treatment group was consistent with the known safety profile of Cimzia.

The prescribing information for Cimzia includes a Boxed Warning to advise health care professionals and patients about the increased risk of serious infections leading to hospitalization or death including tuberculosis (TB), bacterial sepsis (infection in the blood steam), invasive fungal infections (such as histoplasmosis, an infection that affects the lungs), and other infections. Cimzia should be discontinued if a patient develops a serious infection or sepsis. Health care providers are advised to perform testing for latent TB and, if positive, to start treatment for TB prior to starting Cimzia. All patients should be monitored for active TB during treatment, even if the initial latent TB test is negative. The Boxed Warning also advises that lymphoma (cancer in blood cells) and other malignancies, some fatal, have been reported in children and adolescent patients treated with tumor necrosis factor (TNF) blockers, of which Cimzia is a member. Cimzia is not indicated for use in pediatric patients. Cimzia must be dispensed with a patient Medication Guide that describes important information about the drug’s uses and risks.

Cimzia was originally approved in 2008 and is also indicated for adult patients with Crohn’s disease, moderate-to-severe rheumatoid arthritis, active ankylosing spondylitis (AS) and moderate-to-severe plaque psoriasis who are candidates for systemic therapy or phototherapy.

The FDA granted the approval of Cimzia to UCB.

Certolizumab pegol
Syringe with Certolizumab pegol-1800.jpg
Syringe with 200mg Certolizumab pegol
Monoclonal antibody
Type Fab’ fragment
Source Humanized (from mouse)
Target TNF alpha
Clinical data
Trade names Cimzia
AHFS/Drugs.com Consumer Drug Information
MedlinePlus a608041
License data
Pregnancy
category
  • US: B (No risk in non-human studies)
Routes of
administration
Subcutaneous
ATC code
Legal status
Legal status
Pharmacokinetic data
Elimination half-life about 11 days
Excretion Renal (PEG only)
Identifiers
CAS Number
ChemSpider
  • none
UNII
KEGG
ChEMBL
Chemical and physical data
Formula C2115H3252N556O673S16
Molar mass 47,750 g/mol g·mol−1

///////////////FDA 2019, Cimzia, certolizumab pegol, inflammatory arthritis, UCB

https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm634671.htm?utm_campaign=032819_PR_FDA%20approves%20treatment%20for%20patients%20with%20a%20type%20of%20inflammatory%20arthritis&utm_medium=email&utm_source=Eloqua

E 2212

$
0
0

str1

C25 H23 F3 N6 O, 480.48

CAS 1123197-68-1

(+) -2-{(E)-2-[5-methoxy-6-(4-methyl-1H-imidazol-1-yl)pyridin-3-yl]vinyl}-8-(2-trifluoromethylphenyl)-5,6,7,8-tetrahydro-[1,2,4]triazolo[1,5-a]pyridine

  • (+)-5,6,7,8-Tetrahydro-2-[(1E)-2-[5-methoxy-6-(4-methyl-1H-imidazol-1-yl)-3-pyridinyl]ethenyl]-8-[2-(trifluoromethyl)phenyl][1,2,4]triazolo[1,5-a]pyridine
  • (+)-2-[(E)-2-[5-Methoxy-6-(4-methyl-1H-imidazol-1-yl)pyridin-3-yl]ethenyl]-8-(2-trifluoromethylphenyl)-5,6,7,8-tetrahydro-[1,2,4]triazolo[1,5-a]pyridine

Figure

E2212

CAS 1123197-82-9

  • C25 H23 F3 N6 O . 3/2 C4 H6 O6
  • [1,2,4]Triazolo[1,5-a]pyridine, 5,6,7,8-tetrahydro-2-[(1E)-2-[6-methoxy-5-(4-methyl-1H-imidazol-1-yl)-2-pyridinyl]ethenyl]-8-[2-(trifluoromethyl)phenyl]-, (8S)-, (2S,3S)-2,3-dihydroxybutanedioate (2:3)

PATENT

https://patents.google.com/patent/US9453000B2/en

Examples 394 and 395 Synthesis of (+) and (−)-2-{(E)-2-[5-methoxy-6-(4-methyl-1H-imidazol-1-yl)pyridin-3-yl]vinyl}-8-(2-trifluoromethylphenyl)-5,6,7,8-tetrahydro-[1,2,4]triazolo[1,5-a]pyridine

Figure US09453000-20160927-C00296

230 mg of the racemic title compound was obtained from 1-amino-3-(2-trifluoromethylphenyl)piperidin-2-one (343 mg) and (E)-3-[5-methoxy-6-(4-methyl-1H-imidazol-1-yl)pyridin-3-yl]acrylic acid (500 mg) by the same method as in Examples 194 and 195. The racemic title compound (220 mg) was separated by CHIRALPAK™ IC manufactured by Daicel Chemical Industries, Ltd. (2 cm×25 cm; mobile phase: methanol) to obtain the title optically active compound with positive optical rotation and a retention time of 16 minutes (92 mg) and the title optically active compound with negative optical rotation and a retention time of 19 minutes (79 mg).

The property value of the title optically active compound with a retention time of 16 minutes is as follows.

ESI-MS; m/z 481 [M++H].

The property values of the title optically active compound with a retention time of 19 minutes are as follows.

ESI-MS; m/z 481 [M++H]. 1H-NMR (CDCl3) δ (ppm): 1.90-2.01 (m, 1H), 2.10-2.35 (m, 2H), 2.29 (s, 3H), 2.43-2.52 (m, 1H), 3.95 (s, 3H), 4.27-4.41 (m, 2H), 4.69 (dd, J=6.0, 8.4 Hz, 1H), 7.02 (d, J=8.0 Hz, 1H), 7.08 (d, J=16.4 Hz, 1H), 7.40 (dd, J=7.6, 7.6 Hz, 1H), 7.44-7.53 (m, 4H), 7.73 (d, J=8.0 Hz, 1H), 8.13 (d, J=1.6 Hz, 1H), 8.34 (s, 1H).

PATENT

WO2009028588

https://patentscope.wipo.int/search/en/detail.jsf;jsessionid=D6AD22B6CC7302560AE1ADCED305CDCE.wapp2nC?docId=WO2009028588&tab=FULLTEXT&queryString=%28PA%2Feisai%29%2520&recNum=93&maxRec=725
(+)および(-)-2-{(E)-2-[5-メトキシ-6-(4-メチル-1H-イミダゾール-1-イル)ピリジン-3-イル]ビニル}-8-(2-トリフルオロメチルフェニル)-5,6,7,8-テトラヒドロ-[1,2,4]トリアゾロ[1,5-a]ピリジンの合成
[化221]

実施例194および実施例195と同様の方法により、1-アミノ-3-(2-トリフルオロメチルフェニル)ピペリジン-2-オン(343mg)および(E)-3-[5-メトキシ-6-(4-メチル-1H-イミダゾール-1-イル)ピリジン-3-イル]アクリル酸(500mg)から、ラセミ体の表題化合物を230mg得た。ラセミ体の表題化合物(220mg)をダイセル製CHIRALPAK TM IC(2cm×25cm:移動相;メタノール)にて分取し、(+)の旋光性を有する保持時間16分の表題光学活性化合物(92mg)および(-)の旋光性を有する保持時間19分の表題光学活性化合物(79mg)を得た。
保持時間16分の表題光学活性体の物性値は以下の通りである。
ESI-MS;m/z 481[M +H].
保持時間19分の表題光学活性体の物性値は以下の通りである。
ESI-MS;m/z 481[M +H]. H-NMR(CDCl )δ(ppm):1.90-2.01(m,1H),2.10-2.35(m,2H),2.29(s,3H),2.43-2.52(m,1H),3.95(s,3H),4.27-4.41(m,2H),4.69(dd,J=6.0,8.4Hz,1H),7.02(d,J=8.0Hz,1H),7.08(d,J=16.4Hz,1H),7.40(dd,J=7.6,7.6Hz,1H),7.44-7.53(m,4H),7.73(d,J=8.0Hz,1H),8.13(d,J=1.6Hz,1H),8.34(s,1H).

Example 394 and Example 395
(+) and (−)-2-{(E) -2- [5-methoxy-6- (4-methyl-1H-imidazol-1-yl) pyridin-3-yl] Synthesis of vinyl} -8- (2-trifluoromethylphenyl) -5,6,7,8-tetrahydro- [1,2,4] triazolo [1,5-a] pyridine [Formula
221]

Example 194 and By a method similar to Example 195, 1-amino-3- (2-trifluoromethylphenyl) piperidin-2-one (343 mg) and (E) -3- [5-methoxy-6- (4-methyl-) 1 H-Imidazol-1-yl) pyridin-3-yl] acrylic acid (500 mg) gave 230 mg of the racemic title compound. Racemic title compound (220 mg) a Daicel CHIRALPAK TM IC (2 cm × 25 cm: mobile phase; methanol) was collected by min (+) title optically active compound of the retention time of 16 minutes with a optical rotation of (92 mg) The title optically active compound (79 mg) having a polarizability of (−) and a retention time of 19 minutes was obtained.
The physical property values of the title optically active substance with a retention time of 16 minutes are as follows.
ESI-MS; m / z 481 [M + + H].
The physical property values of the title optically active substance with a retention time of 19 minutes are as follows.
ESI-MS; m / z 481 [M + + H]. 1 H-NMR (CDCl 3)) Δ (ppm): 1.90 to 2.01 (m, 1 H), 2.10 to 2.35 (m, 2 H), 2.29 (s, 3 H), 2.43 to 2.52 (m) , 1 H), 3.95 (s, 3 H), 4.27-4. 41 (m, 2 H), 4.69 (dd, J = 6.0, 8.4 Hz, 1 H), 7.02 (d , J = 8.0 Hz, 1 H), 7.08 (d, J = 16.4 Hz, 1 H), 7.40 (dd, J = 7.6, 7.6 Hz, 1 H), 7.44-7. 53 (m, 4H), 7.73 (d, J = 8.0 Hz, 1 H), 8.13 (d, J = 1.6 Hz, 1 H), 8.34 (s, 1 H).

PATENT

https://patents.google.com/patent/WO2010098490A1/it

str1

As a novel compound that has an effect of reducing the production of Aβ40 and

42 and is expected as a therapeutic or prophylactic agent for Alzheimer’s disease or the like, the present inventors have found a compound represented by the following formula (1) (compound

(D): [Formula 1]

and filed a patent application for the invention (PCT/JP08/065365).

Generally, properties of salts of compounds and those crystals that are useful as pharmaceuticals are highly important for the development of pharmaceuticals, because the properties greatly affect bioavailability of drugs, purity of drug substances, formulation of preparations, and the like. Therefore, it is necessary to research which salts and crystal forms of the compound of the formula (1) are most excellent as pharmaceuticals. Specifically, since their properties depend on the character of the individual compounds, it is generally difficult to estimate salts and crystal forms for drug substances having excellent properties and it is demanded to actually make various studies for each compound.

EXAMPLES [0023] The present invention will be described in detail below with reference to reference examples and examples; however, the present invention is not limited to these reference examples and examples. [0024]

The following abbreviations are used in the following reference examples and examples.

DMF: N,N’-dimethylformamide

THF: Tetrahydrofuran

EDC: lrEmyl-S-β-dimemylammopropytycarbodiimide hydrochloride HOBT: 1-Hydroxybenzotriazole IPEA: Diisopropylethylamine [0025]

In powder X-ray diffractometry of the crystals produced in the following examples, the resulting crystals were placed on a sample stage of a powder X-ray diffractometer and analyzed under the following conditions. [0026] Measurement conditions

Sample holder: Aluminum Target: Copper

Detector: Scintillation counter Tube voltage: 50 kV Tube current: 300 mA

Slit: DS 0.5 mm (Height limiting slit 2 mm), SS Open, RS Open Scanning rate : 5 °/min

Sampling interval: 0.02° Scan range: 5 to 35° Goniometer: Horizontal goniometer [0027] Reference Example 1

Svnmesis ofr8SV2-(fE)-246-memoxy-5-(4-memyl-lH-imidazol-l-vnpyridin-2-yllvmvU-8-(2-trifluoromethylphenyl)-5,6J,8-tetrahvdro-[1.2,41triazolo[l.,5-a]pyridine

[Formula 2]

Synthesis of l-amino-3-(2-trifluoromemylphenyl)piperidin-2-one Thionyl chloride (2.72 mL) was added to a solution of 2-trifluoromethylphenylacetic acid (1.9 g) in methanol (38 mL), followed by stirring at room temperature for three hours. The reaction solution was concentrated under reduced pressure. The resulting residue was diluted with DMF. Sodium hydride (containing 40% mineral oil, 410 mg) was added under ice-cooling, followed by stirring for 10 minutes. The reaction solution was further stirred for 30 minutes and then ice-cooled again. l-Chloro-3-iodopropane (1.02 mL) was added to the reaction mixture, and the reaction solution was stirred at room temperature overnight. Water and ethyl acetate were added to the reaction mixture, and the organic layer was separated. The resulting organic layer was washed with saturated aqueous sodium chloride, dried over anhydrous magnesium sulfate and then concentrated under reduced pressure. The resulting residue was diluted with ethanol (26.6 mL). Hydrazine monohydrate (7.6 mL) was added, and the reaction solution was stirred at room temperature for two hours and then at 60°C for further three hours. The reaction mixture was concentrated under reduced pressure. Saturated aqueous sodium bicarbonate and ethyl acetate and were added to the residue, and the organic layer was separated. The resulting organic layer was washed with saturated aqueous sodium chloride, dried over anhydrous magnesium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (carrier: Chromatorex NH; elution solvent: heptane-ethyl acetate system) to obtain 1.68 g of the title compound. The property values of the compound are as follows.

ESI-MS; m/z 259 [M+H-H]. 1H-NMR (CDCl3) δ (ppm): 1.82-2.10 (m, 3H), 2.18-2.26 (m, IH), 3.58-3.76 (m, 2H), 4.07 (dd, J = 10.0, 5.6 Hz, IH), 4.60 (s, 2H), 7.24 (d, J = 7.6 Hz, IH), 7.35 (t, J = 7.6 Hz, IH), 7.51 (t, J = 7.6 Hz, IH)5 7.66 (d, J = 7.6 Hz, IH). [0028] Synthesis of (EV3-[6-methoxy-5-(4-methyl- 1 H-imidazol- 1 -yl)pyridin-2-yl]-N-f2-oxo-3 -(2-trifluoromethylphenyl)piperidin- 1 -yl]acrylamide

EDC (834 mg), HOBT (588 mg) and IPEA (2.03 mL) were added to a suspension of (E)-3-[6-methoxy-5-(4-methyl-lH-imidazol-l-yl)pyridm-2-yl]acrylic acid trifluoroacetate (1.42 g) and l-amήio-3-(2-trifluoromethylphenyl)piperidin-2-one (750 mg) in DMF (30 mL). After stirring at room temperature for 14 hours, a saturated sodium bicarbonate solution and ethyl acetate were added to the reaction solution, and the organic layer was separated. The resulting organic layer was dried over anhydrous magnesium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (carrier: Chromatorex NH; elution solvent: ethyl acetate-methanol system) to obtain 1.23 g of the title compound. The property values of the compound are as follows. ESI-MS; m/z 500 [M1H-HJ. [0029]

Synthesis of r8S>-2-(fEV2-r6-methoxy-5-r4-methyl-lH-imidazol-l-vnpyridm’2-vnvinvU-8-(2-trifluoromethvlphenvD-5.6.7.8-tetrahvdro-ri.2.41triazoloπ.5-a1pvridine Phosphorus oxychloride (24.2 mL) was added to (E)-3~[6-methoxy-5-(4-methyl-lH-imidazol-l-yl)pyridin-2-yl]-N-[2-oxo-3-(2-trifluoromethylphenyl)piperidin-l-yl]acrylamide (1.2 g). The reaction solution was stirred at 1000C for one hour and then concentrated under reduced pressure. Subsequently, the residue was diluted with acetic acid (24.2 mL) and then ammonium acetate (1.9 g) was added, followed by stirring at 1500C for two hours. The reaction solution was left to cool to room temperature and then concentrated under reduced pressure. A saturated sodium bicarbonate solution and ethyl acetate were added to the resulting residue, and the organic layer was separated. The resulting organic layer was dried over anhydrous magnesium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (carrier: Chromatorex NH; elution solvent: heptane-ethyl acetate system) to obtain a racemate of the title compound (750 mg). The resulting racemate (410 mg) was separated by CHIRALP AK™ IA manufactured by Daicel Chemical Industries, Ltd. (2 cm x 25 cm, mobile phase: hexane:ethanol = 8:2, flow rate: 10 mL/min) to obtain the title compound with a retention time of 33 minutes and negative optical rotation (170 mg) as crystals. The property values of the title compound are as follows.

1H-NMR (CDCl3) δ (ppm): 1.90-2.01 (m, IH), 2.10-2.35 (m, 2H), 2.29 (d, J = 1.2 Hz, 3H), 2.42-2.51 (m, IH), 4.03 (s, 3H), 4.28-4.41 (m, 2H), 4.70 (dd, J = 8.4, 6.0 Hz, IH), 6.92 (d, J = 8.0 Hz, IH), 6.95 (t, J = 1.2 Hz, IH), 7.01 (d, J = 7.6 Hz, IH), 7.39 (t, J = 7.6 Hz5 IH), 7.44 (d, J = 16.0 Hz, IH), 7.45 (d, J = 8.0 Hz, IH), 7.49 (t, J = 7.6 Hz, IH), 7.63 (d, J = 16.0 Hz5 IH), 7.72 (d, J = 7.6 Hz, IH), 7.76 (d, J = 1.2 Hz, IH). [0030]

(8S)-2-{(E)-2-[6-Methoxy-5-(4-methyl-lH-imidazol-l-yl)ρyridin-2-yl]vinyl}-8-(2-trifluoromethylphenyl)-5,6,7,8-tetrahydro-[l,2,4]triazolo[l,5-a]pyridine synthesized according to the above reference example was used for the following synthesis of salts. [0031] Example 1

Synthesis of r8SV2-{rEV2-[6-methoxy-5-(4-methyl-lH-imidazol-l-vπpyridin-2-vnvinvU-8-f2-trifluoromethylphenyl)-5.6.7.8-tetrahvdro-fl,2,4]triazolo[l.,5-a]pyridine 1.5 D-tartrate

(8S)-2-{(E)-2-[6-methoxy-5-(4-methyl-lH-imidazol-l-yl)pyridin-2-yl]vinyl}-8-(2-trifluoromethylphenyl)-5,6,7,8-tetrahydro-[l,2,4]triazolo[l,5-a]pyridine (33.70 mg) was dissolved in 285 μL of a D-tartaric acid-ethanol solution (110.92 mg/3 mL) with stirring at room temperature. The oil was precipitated when 1 mL of heptane was added. Accordingly, the oily substance was dissolved by adding 1 mL of ethanol. Further, 0.5 mL of heptane was added, and the mixture was transferred to a low temperature laboratory at about 50C (under shading) and continuously stirred for 24 hours. Thus, partial gelation occurred. Thereafter, the mixture was brought back to room temperature and continuously stirred, resulting in precipitation of a solid. The solid was collected by filtration through a glass filter and dried under reduced pressure at room temperature to obtain 21.25 mg of the title compound as white solid crystals. 1H-NMR (600 MHz, DMSOd6) δ (ppm): 1.96 (m, IH), 2.14 (s, 3H), 2.16 (m, 2H), 2.29 (m, IH), 3.98 (s, 3H), 4.28 (m, 2H), 4.29 (s, 3H), 4.51 (dd, J = 9, 6 Hz, IH), 7.22 (s, IH), 7.25 (brd, J = 8 Hz, IH), 7.27 (d, J = 8 Hz, IH), 7.32 (d, J = 16 Hz, IH)5 7.46 (d, J = 16 Hz, IH), 7.49 (brdd, J = 8 Hz, IH), 7.61 (brdd, J = 8 Hz5 IH), 7.77 (brd, J = 8 Hz, IH), 7.78 (d, J = 8 Hz, IH), 7.91 (s, IH). [0032] Example 2

Synthesis of (8SV2-l(Ε)-2-f6-methoxy-5-(4-methyl-lH-imidazol-l-vnpyridm-2-yllvinyl>-8-f2-trifluoromethylphenylV5,6J,8-tetrahvdro-[l ,2,4]triazolo[l ,5-a]pyridine di-D-tartrate

(8S)-2-{(E)-2-[6-methoxy-5-(4-methyl-lH-imidazol-l-yl)pyridin-2-yl]vinyl}-8-(2-trifluoromethylphenyl)-5,657,8-tetrahydro-[l ,2,4]triazolo[l ,5-a]ρyridine (810.18 mg) was dissolved in 8 mL of a D-tartaric acid-ethanol solution (751.13 mg/10 mL) with stirring at room temperature. The oil was precipitated when 2 mL of heptane was added. Accordingly, the oily substance was dissolved by ultrasonic treatment to prepare a clear solution. Several mg of crystals of the 1.5 D-tartrate prepared according to Example 1 were added, followed by stirring at room temperature. Stirring for about one hour resulted in gelation and subsequent precipitation of a solid. Further, stirring was continued while gradually adding 14 mL of heptane. A part of the suspension (2 mL) was separated and the solid was collected by filtration through a glass filter. The solid was dried under reduced pressure at room temperature to obtain 71.14 mg of the title compound as white solid crystals. 1H-NMR (400 MHz, DMSOd6) δ (ppm): 1.97 (m, IH), 2.15 (s, 3H), 2.16 (m, 2H), 2.30 (m, IH), 3.98 (s, 3H), 4.28 (m, 2H), 4.29 (s, 4H), 4.51 (dd, J = 9, 6 Hz, IH), 7.22 (brs, IH), 7.25 (brd, J = 8 Hz, IH), 7.27 (d, J = 8 Hz, IH), 7.32 (d, J = 16 Hz, IH), 7.46 (d, J = 16 Hz, IH), 7.49 (brdd, J – 8 Hz, IH), 7.61 (brdd, J = 8 Hz, IH), 7.77 (brd, J = 8 Hz, IH), 7.78 (d, J = 8 Hz, IH), 7.91 (brs, IH). [0033] Example 3

Synthesis of r8SV2-(rE)-2-r6-methoxy-5-r4-methyl-lH-imidazol-l-vnpyridin-2-yl1vinvU-8-α-trifluoromethylphenyl)-5,6J,8-tetrahydro-[1.2,4]triazolo[l,5-a]pyridine disulfate

Concentrated sulfuric acid (11.5 μL) was added to a solution of (8S)-2-{(E)-2-[6- methoxy-5-(4-methyl-lH-imidazol-l-yl)pyridin-2-yl]vinyl}-8-(2-txifluoromethylphenyl)-5,6,7,8-tetrahydro-[l52,4]triazolo[l55-a]pyridine (98.09 mg) in ethanol (1 mL), and 1 mL of ethyl acetate was added with stirring at room temperature. Since the oily portion was confirmed on the bottom of the recovery flask, the oily substance was dissolved by ultrasonic treatment. Stirring at room temperature under shading for about 30 minutes resulted in precipitation of a solid. The solid was collected by filtration through a glass filter and dried under reduced pressure at room temperature to obtain 127.94 mg of the title compound as white solid crystals. 1H-NMR (400 MHz, DMSOd6) δ (ppm): 1.97 (m, IH), 2.17 (m, 2H), 2.30 (m, IH), 2.34 (brd, J = 1 Hz, 3H), 4.01 (s, 3H), 4.29 (m, 2H), 4.52 (dd, J = 9, 6 Hz, IH)5 7.25 (brd, J = 8 Hz, IH), 7.37 (d, J = 16 Hz, IH), 7.40 (d, J = 8 Hz, IH), 7.50 (brdd, J = 8 Hz, IH), 7.55 (d, J = 16 Hz, IH), 7.61 (brdd, J = 8 Hz, IH), 7.77 (m, IH), 7.78 (m, IH), 8.00 (d, J = 8 Hz, IH), 9.36 (d, J = 2 Hz, IH). [0034] Example 4 Synthesis of (8SV2-((E)-2-[6-methoxy-5-(4-methyl-lH-imidazol-l-ylN)ρyridin-2-yllvinvU-8-(2-trifluoromethylphenyl)-5,6,7,8-tetrahvdiO-[1.2,41triazolo[l,5-a]pyridine dihydrobromide

Concentrated hydrobromic acid (24.8 μL) was added to a solution of (8S)-2-{(E)-2-[6-methoxy-5-(4-methyl-lH-imidazol-l-yl)pyridin-2-yl]vinyl}-8-(2-trifluoromethylphenyl)-5,6,7,84etrahydro-[l,254]triazolo[l55-a]pyridine (51.42 mg) m ethanol (1 mL), and 1 mL of heptane was added with stirring at room temperature. After several minutes, 1 mL of heptane was further added to the solution and stirring was continued. The solution was stirred at room temperature for one hour and then further stirred at about 50C for 20 minutes. The precipitated solid was collected by filtration through a glass filter and dried under reduced pressure at room temperature to obtain 49.24 mg of the title compound as white solid crystals. 1H-NMR (400 MHz, DMSO-d6) δ (ppm): 1.99 (m, IH), 2.17 (m, 2H), 2.30 (m, IH), 2.34 (brd, J = 1 Hz5 3H), 4.01 (s, 3H), 4.30 (m, 2H), 4.52 (dd, J = 9, 6 Hz5 IH), 7.25 (brd, J = 8 Hz5 IH), 7.37 (d, J = 16 Hz, IH), 7.40 (d, J = 7 Hz, IH)57.50 (brdd, J = 8 Hz, IH), 7.55 (d, J = 16 Hz, IH), 7.61 (brdd, J = 8 Hz5 IH), 7.77 (m, IH)5 7.78 (m, IH), 8.00 (d, J = 7 Hz, IH), 9.37 (d, J = 2 Hz, IH). [0035] Example 5

Synthesis of r8SV2-((Ε)-2-r6-methoxy-5-r4-methyl-lH-imidazol-l-yl)ρyridin-2-vnvinyl}-8-r2-trifluoromethylphenyl)-5,6J,8-tetrahvdro-[1.2,41triazolo[1.5-alpyridine hydrochloride

Concentrated hydrochloric acid (3.6 μL) was added to a solution of (8S)-2-{(E)- 2-[6-methoxy-5-(4-metiiyl-lH-imidazol-l-yl)pyridin-2-yl]vinyl}-8-(2-trifluoromethylplienyl)-5,6,7,8-te1xahydro-[l,2,4]triazolo[l,5-a]pyridme (19.80 mg) in 2-propanol (1 mL), and a total of 4 mL of heptane was added in 1 mL portions with stirring at room temperature. The solution was stirred at room temperature under shading for five days. The precipitated solid was collected by filtration through a glass filter and dried under reduced pressure at room temperature to obtain 7.45 mg of the title compound as white solid crystals. 1H-NMR (400 MHz, DMSOd6) δ (ppm): 1.97 (m, IH), 2.17 (m, 2H)5 2.30 (m, IH), 2.30 (s, 3H), 4.00 (s, 3H), 4.30 (m, 2H)5 4.52 (dd, J = 9, 6 Hz5 IH), 7.25 (brd, J – 8 Hz5 IH), 7.36 (d, J = 16 Hz5 IH), 7.37 (d5 J = 8 Hz, IH), 7.50 (brt, J = 8 Hz5 IH)5 7.53 (d, J = 16 Hz5 IH)5 7.61 (brt, J = 8 Hz5 IH)5 7.66 (brs, IH), 7.77 (brd, J = 8 Hz, IH)5 7.96 (d, J = 8 Hz5 IH), 9.06 (brs, IH). [0036] Example 6

Synthesis of (8S)-2-((ΕV2-r6-methoxy-5-(4-methyl-lH-imidazol-l-yl)pyridin-2-yl1vinvU-8-(2-trifluoromethylphenyl)-5.6,7,8-tetrahvdro-[l,2,4]triazolo[L5-a1pyridine hydrochloride Concentrated hydrochloric acid (14.3 μL) and heptane (7 mL) were added to a solution of (8S)-2-{(E)-2-[6-methoxy-5-(4-methyl-lH-imidazol-l-yl)pyridin-2-yl]vinyl}-8-(2-trifluoromethylphenyl)-5,657,8-tetrahydro-[l,2,4]triazolo[l,5-a]pyridine (79.77 mg) in 2-propanol (3 mL). A small amount of the crystals obtained in Example 5 were added as seed crystals with stirring at room temperature. The mixture was transferred to a low temperature laboratory at about 50C and stirred for one hour. Thereafter, 1 mL of heptane was further added, followed by stirring for several minutes. When the precipitated solid was collected by filtration through a glass filter, the solid was precipitated in the filtrate. The precipitated solid was collected by filtration through a glass filter and dried under reduced pressure at room temperature to obtain 38.02 mg of the title compound as white solid crystals. 1H-NMR (400 MHz, DMSO-d6) δ (ppm): 1.97 (m, IH), 2.17 (m5 2H), 2.29 (m, IH), 2.32 (brd, J = 1 Hz, 3H), 4.00 (s, 3H), 4.30 (m, 2H), 4.52 (dd, J = 9, 6 Hz, IH), 7.25 (brd, J = 8 Hz, IH), 7.37 (d, J = 16 Hz5 IH), 7.38 (d, J = 8 Hz, IH), 7.50 (brdd, J = 8 Hz, IH)5 7.54 (d, J = 16 Hz, IH), 7.61 (brdd, J = 8 Hz, IH), 7.72 (brs, IH), 7.77 (brd, J = 8 Hz, IH), 7.98 (d, J = 8 Hz5 IH)5 9.24 (brs, IH). [0037] Example 7

SvnJhesis off8SV2-f(E>2-r6-memoxy-5-(4-mefovπ trifluoromethylt>henylV5,6,7,8-tetrahvdro-[l,2,4]triazolo[l,5-a]pyridine mesylate

Mesylic acid (0.8 μL) was added to a mixed solution of (8S)-2-{(E)-2-[6- methoxy-5-(4-methyl-lH-imidazol-l-yl)pyridin-2-yl]vinyl}-8-(2-trifluoromethylphen^ tetrahydro-[l52,4]triazolo[l,5-a]pyridine (50 mg) in t-butyl methyl ether (0.8 mL)-ethaαol (0.1 mL). The mixture was solidified as a result of stirring at room temperature for two hours. The solid was collected by filtration through a glass filter. The solid was washed with t-butyl methyl ether-ethanol (8:1) and then dried under reduced pressure at room temperature to obtain 51.9 mg of the title compound as pale yellow solid crystals.

1H-NMR (DMSO-d6) δ (ppm): 1.90-2.05 (m, IH)3 2.10-2.22 (m, 2H), 2.28-2.40 (m, IH), 2.31 (s, 3H), 2.35 (s, 3H)5 4.02 (s, 3H)5 4.25-4.39 (m, 2H), 4.50-4.55 (m, IH), 7.27 (d5 J = 8.0 Hz5 IH)5 7.38 (d, J = 16.0 Hz5 IH)5 7.41 (d, J = 8.0 Hz, IH)5 7.51 (t5 J = 8.0 Hz5 IH)5 7.55 (d, J = 16.0 Hz5 IH), 7.63 (t, J = 8.0 Hz5 IH)5 7.78 (d, J = 8.0 Hz5 IH)5 7.79 (s, IH), 8.01 (d, J = 8.0 Hz5 IH), 9.37 (s, IH). [0038] Example 8 Synthesis of (8S)-2-((ΕV2-r6-methoxy-5-(4-methyl-lH-imidazol-l-vnpyridin-2-vnvinvn-8-r2-trifluoromethylphenyl)-5.6,7,8-tetrahydro-[l.,2,4|triazolo[l,5-a]pyridine diphosphate

A solution of phosphoric acid (52.8 mg) in acetonitrile (0.2 mL) was added to a solution of (8S)-2-{(E)-2-[6-methoxy-5-(4-methyl-lH-mτidazol-l-yl)ρyridin-2-yl]vinyl}-8-(2-trifluoromethylphenyl)-5565758-tetrahydro-[l5254]triazolo[l,5-a]pyridine (100 mg) in acetonitrile (0.8 mL) at room temperature. The precipitated oil was solidified as a result of stirring with spatula. The solid was collected by filtration through a glass filter. The solid was washed with ice-cold acetonitrile, air-dried at room temperature for 10 minutes and then dried under reduced pressure at room temperature to obtain 120 mg of the title compound as white solid crystals. 1H-NMR (DMSO-d6) δ (ppm): 1.90-2.05 (m, IH), 2.11-2.20 (m, 2H), 2.15 (s, 3H), 2.25-2.35 (m, IH), 3.99 (s, 3H)5 4.24-4.39 (m, 2H), 4.50-4.55 (m, IH)5 7.23 (s, IH), 7.26 (d, J = 7.0 Hz, IH), 7.28 (d, J = 8.0 Hz, IH), 7.33 (d, J = 16.0 Hz5 IH), 7.47 (d, J = 16.0 Hz5 IH), 7.51 (t, J = 7.0 Hz, IH), 7.63 (t, J = 7.0 Hz, IH), 7.78 (d, J = 7.0 Hz, IH), 7.79 (d, J = 8.0 Hz, IH), 7.90 (s, IH). [0039] Example 9 Svnmesis of(8SV2-{(E)-2-[6-memoxy-5-(4-methyl-lH-irnidazol-l-yl)pyridin-2-yl1vinvU-8-(2-trifluoromethylphenyl)-5,6,7,8-tetrahvdro-[l .2.41triazolo[l .5-a]pyridine diphosphate

A solution of phosphoric acid (13.2 mg) in ethanol (0.05 mL) was added to a mixed solution of (8S)-2-{(E)-2-[6-methoxy-5-(4-methyl-lH-imidazol-l-yl)pyridin-2-yl]vinyl}-8-(2-trifluoromethylphenyl)-5,6,7,8-tetrahydro-[l,2!,4]triazolo[l,5-a]pyridme (50 mg) in heptane (0.6 mL)-ethanol (0.15 mL) at room temperature. The reaction solution was stirred at room temperature, and the precipitated solid was collected by filtration through a glass filter. The solid was washed with heptane-ethanol (3:1) and then dried under reduced pressure at room temperature to obtain 37.6 mg of the title compound as white solid crystals. 1H-NMR (DMSOd6) δ (ppm): 1.90-2.05 (m, IH), 2.11-2.20 (m, 2H), 2.15 (s, 3H), 2.25-2.35 (m, IH), 3.99 (s, 3H), 4.24-4.39 (m, 2H), 4.50-4.55 (m, IH), 7.23 (s, IH), 7.26 (d, J = 7.0 Hz, IH), 7.28 (d, J = 8.0 Hz, IH), 7.33 (d, J = 16.0 Hz, IH), 7.47 (d, J = 16.0 Hz, IH), 7.51 (t, J = 7.0 Hz, IH), 7.63 (t, J = 7.0 Hz, IH), 7.78 (d, J = 7.0 Hz, IH), 7.79 (d, J = 8.0 Hz, IH), 7.90 (s, IH).

CLIP

Development of an Efficient Manufacturing Process for E2212 toward Rapid Clinical Introduction

 API Research Japan, Pharmaceutical Science & Technology, CFU, Medicine Development CenterEisai Co. Ltd.5-1-3-Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
 API Research Japan, Pharmaceutical Science & Technology, CFU, Medicine Development CenterEisai Co. Ltd.22-Sunayama, Kamisu-shi, Ibaraki 314-0255, Japan
§ Neurology Tsukuba Research Department, Discovery, Medicine Creation, NBGEisai Co. Ltd.5-1-3-Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
 Integrated ChemistryEisai AiM Institute4 Corporate Drive, Andover, Massachusetts 01810, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.8b00444
This article is part of the Japanese Society for Process Chemistry special issue.
Abstract Image

Process studies of E2212 (1) toward rapid clinical introduction are described. Through comprehensive route-finding studies and optimization of key condensation and cyclization steps, a racemate-based manufacturing route was established and successfully scaled-up to the hundred kilogram scale. For the rapid delivery of a drug substance containing the Z isomer for preclinical safety studies, the successful scale-up of the photoisomerization of an olefin in a flow system is also presented.

https://pubs.acs.org/doi/10.1021/acs.oprd.8b00444

E2212 (1) (18.0 kg, 92.5% yield) as a white solid. Mother liquor 3 were recycled according to the procedure described below. FTIR (cm–1, KBr) 3461, 3173, 2956, 1734, 1584, 1536, 1476, 1309, 1130, 835, 765, 752; 1H NMR (600 MHz, DMSO-d6) δ 7.91 (s, 1H), 7.78 (d, J = 8.4 Hz, 1H), 7.77 (br d, J = 8.4 Hz, 1H), 7.61 (br dd, J = 7.8, 7.8 Hz, 1H), 7.49 (br dd, J = 7.8, 7.8 Hz, 1H), 7.46 (d, J= 15.6 Hz, 1H), 7.32 (d, J = 15.6 Hz, 1H), 7.27 (d, J = 7.8 Hz, 1H), 7.25 (br d, J = 7.8 Hz, 1H), 7.22 (s, 1H), 4.51 (dd, J = 9.0, 6.0 Hz, 1H), 4.29 (s, 3H), 4.28 (m, 2H), 3.98 (s, 3H), 2.29 (m, 1H), 2.14 (s, 3H), 2.16 (m, 2H), 1.96 (m, 1H); 13C NMR (150 MHz, DMSO-d6) δ 173.3, 159.3, 155.4, 155.0, 150.1, 141.1, 137.1, 136.9, 133.6, 132.9, 131.0, 130.5, 127.6, 127.1 (q, JC–F = 30 Hz), 125.8 (q, JC–F = 5.6 Hz), 124.7 (q, JC–F = 270 Hz), 122.2, 120.7, 117.2, 116.5, 72.3, 53.7, 47.0, 37.6, 30.7, 21.3, 13.6; HRMS (ESI+) calcd for C25H23F3N6O ([M + H]+) 481.1958, found 481.1953.

 E/Z mixture of E2212 (196.0 g (containing residual n-PrOH), E:Z = 61.8:37.4 by UV (271 nm), 1.3:1.0 by 1H NMR) as an orange oil. HPLC conditions to monitor the isomerization conversion and E/Z ratio: XBridge-Shield-RP18 (5 μm, 4.6 mm × 250 mm), 1.0 mL/min, oven temperature = 40 °C, mobile phase A = 900:100:1 v/v/w H2O/MeCN/AcONH4, mobile phase B = 100:900:1 v/v/w H2O/MeCN/AcONH4, gradient (time (min)/B conc (%)) = 0/5 → 5/45 → 35/45 → 50/100 → 55/100 → 55.01/5 → 65/5 → 65.01/stop, RRT of Z form = 0.73.
From this mixture, a small portion was purified by silica gel column chromatography to give the Zisomer in free form. FTIR (cm–1, KBr) 3416, 2952, 1586, 1500, 1487, 1313, 1161, 1114, 1036, 966, 858, 769; 1H NMR (600 MHz, CDCl3) δ 7.90 (d, J = 7.9 Hz, 1H), 7.72 (d, J = 1.2 Hz, 1H), 7.70 (d, J = 7.9 Hz, 1H), 7.45 (dd, J = 7.6, 7.4 Hz, 1H), 7.37 (dd, J = 7.7, 7.6 Hz, 1H), 7.30 (d, J = 7.9 Hz, 1H), 7.02 (d, J = 7.9 Hz, 1H), 6.93 (dd, J = 1.2, 1.0 Hz, 1H), 6.73 (d, J = 13.3 Hz, 1H), 6.64 (d, J = 13.3 Hz, 1H), 4.63 (dd, J = 9.3, 5.9 Hz, 1H), 4.34 (br ddd, J = 13.0, 5.6, 4.1 Hz, 1H), 4.28 (ddd, J = 13.0, 9.9, 4.9 Hz, 1H), 3.92 (s, 3H), 2.45 (dddd, J = 13.2, 6.5, 6.5, 2.6 Hz, 1H), 2.29 (d, J= 1.0 Hz, 3H), 2.28 (m, 1H), 2.15 (m, 1H), 1.94 (dddd, J = 12.9, 11.4, 8.3, 2.6 Hz, 1H); 13C NMR (150 MHz, CDCl3) δ 159.4, 155.2, 154.4, 150.8, 140.2, 138.3, 136.6, 133.2, 131.9 131.6, 129.9, 128.5 (q, JC–F = 29.8 Hz), 127.2, 126.2 (q, JC–F = 5.6 Hz), 124.4 (q, JC–F = 274.0 Hz), 121.8, 120.1, 118.4, 116.0, 53.6, 47.3, 37.9, 31.0, 21.7, 13.6; HRMS (ESI+) calcd for C25H24F3N6O ([M + H]+) 481.1958, found 481.1960.

///////////E2212, E 2212

Cladribine, クラドリビン

$
0
0
Cladribine.svgChemSpider 2D Image | Cladribine | C10H12ClN5O3

Cladribine

クラドリビン

Leustatin

クラドリビン

RWJ 26251 / RWJ-26251

  • Molecular FormulaC10H12ClN5O3
  • Average mass285.687 Da
2-chloro-6-amino-9-(2-deoxy-β-D-erythro-pentofuranosyl)purine
2-Chlorodeoxyadenosine
4291-63-8 [RN]
6997
adenosine, 2-chloro-2′-deoxy- [ACD/Index Name]
AU7357560
CDA
(2R,3S,5R)-5-(6-Amino-2-chlor-9H-purin-9-yl)-2-(hydroxymethyl)tetrahydrofuran-3-ol
Leustatin (Trade name)
Litak (Trade name)
MLS000759397
Movectro (Trade name)
Mylinax
QA-1968
LAUNCHED, 1993, USA Ortho Biotech, Janssen Biotech

Cladribine, sold under the brand name Leustatin and Mavenclad among others, is a medication used to treat hairy cell leukemia(HCL, leukemic reticuloendotheliosis), B-cell chronic lymphocytic leukemia and relapsing-remitting multiple sclerosis.[4][5] Its chemical name is 2-chloro-2′-deoxyadenosine (2CdA).

Cladribine, a deoxyadenosine derivative developed by Ortho Biotech (currently Janssen), was first launched in the U.S. in 1993 as an intravenous treatment for hairy cell leukemia

Cladribine has been granted orphan drug designation in the U.S. in 1990 for the treatment of acute myeloid leukemia (AML) and hairy cell leukemia

As a purine analog, it is a synthetic chemotherapy agent that targets lymphocytes and selectively suppresses the immune system. Chemically, it mimics the nucleoside adenosine. However, unlike adenosine it is relatively resistant to breakdown by the enzyme adenosine deaminase, which causes it to accumulate in cells and interfere with the cell’s ability to process DNA. Cladribine is taken up cells via a transporter. Once inside a cell cladribine is activated mostly in lymphocytes, when it is triphosphorylated by the enzyme deoxyadenosine kinase (dCK). Various phosphatases dephosphorylate cladribine. Activated, triphosphorylated, cladribine is incorporated into mitochondrial and nuclear DNA, which triggers apoptosis. Non-activated cladribine is removed quickly from all other cells. This means that there is very little non-target cell loss.[4][6]

Medical uses

Cladribine is used for as a first and second-line treatment for symptomatic hairy cell leukemia and for B-cell chronic lymphocytic leukemia and is administered by intravenous or subcutaneous infusion.[5][7]

Since 2017, cladribine is approved as an oral formulation (10 mg tablet) for the treatment of RRMS in Europe, UAE, Argentina, Chile, Canada and Australia. Marketing authorization in the US was obtained in March 2019[8].

Some investigators have used the parenteral formulation orally to treat patients with HCL. It is important to note that approximately 40% of oral cladribine in bioavailable orally. It used, often in combination with other cytotoxic agents, to treat various kinds of histiocytosis, including Erdheim–Chester disease[9] and Langerhans cell histiocytosis,[10]

Cladribine can cause fetal harm when administered to a pregnant woman and is listed by the FDA as Pregnancy Category D; safety and efficacy in children has not been established.[7]

Adverse effects

Injectable cladribine suppresses the body’s ability to make new lymphocytesnatural killer cells and neutrophils (called myelosuppression); data from HCL studies showed that about 70% of people taking the drug had fewer white blood cells and about 30% developed infections and some of those progressed to septic shock; about 40% of people taking the drug had fewer red blood cells and became severely anemic; and about 10% of people had too few platelets.[7]

At the dosage used to treat HCL in two clinical trials, 16% of people had rashes and 22% had nausea, the nausea generally did not lead to vomiting.[7]

In comparison, in MS, cladribine is associated with a 6% rate of severe lymphocyte suppression (lymphopenia) (levels lower than 50% of normal). Other common side effects include headache (75%), sore throat (56%), common cold-like illness (42%) and nausea (39%)[11]

Mechanism of Action

As a purine analogue, it is taken up into rapidly proliferating cells like lymphocytes to be incorporated into DNA synthesis. Unlike adenosine, cladribine has a chlorine molecule at position 2, which renders it partially resistant to breakdown by adenosine deaminase (ADA). In cells it is phosphorylated into its toxic form, deoxyadenosine triphosphate, by the enzyme deoxycytidine kinase (DCK). This molecule is then incorporated into the DNA synthesis pathway, where it causes strand breakage. This is followed by the activation of transcription factor p53, the release of cytochrome c from mitochondria and eventual programmed cell death (apoptosis).[12] This process occurs over approximately 2 months, with a peak level of cell depletion 4–8 weeks after treatment[13]

Within the lymphocyte pool, cladribine targets B cells more than T cells. Both HCL and B-cell chronic lymphocytic leukaemia are types of B cell blood cancers. In MS, its effectiveness may be due to its ability to effectively deplete B cells, in particular memory B cells[14] In the pivotal phase 3 clinical trial of oral cladribine in MS, CLARITY, cladribine selectively depleted 80% of peripheral B cells, compared to only 40-50% of total T cells.[15] More recently, cladribine has been shown to induce long term, selective suppression of certain subtypes of B cells, especially memory B cells.[16]

Another family of enzymes, the 5´nucleotidase (5NCT) family, is also capable of dephosphorylating cladribine, making it inactive. The most important subtype of this group appears to be 5NCT1A, which is cytosolically active and specific for purine analogues. When DCK gene expression is expressed as a ratio with 5NCT1A, the cells with the highest ratios are B cells, especially germinal centre and naive B cells.[16] This again helps to explain which B cells are more vulnerable to cladribine-mediated apoptosis.

Although cladribine is selective for B cells, the long term suppression of memory B cells, which may contribute to its effect in MS, is not explained by gene or protein expression. Instead, cladribine appears to deplete the entire B cell department. However, while naive B cells rapidly move from lymphoid organs, the memory B cell pool repopulates very slowly from the bone marrow.

History

Ernest Beutler and Dennis A. Carson had studied adenosine deaminase deficiency and recognized that because the lack of adenosine deaminase led to the destruction of B cell lymphocytes, a drug designed to inhibit adenosine deaminase might be useful in lymphomas. Carson then synthesized cladribine, and through clinical research at Scripps starting in the 1980s, Beutler tested it as intravenous infusion and found it was especially useful to treat hairy cell leukemia (HCL). No pharmaceutical companies were interested in selling the drug because HCL was an orphan disease, so Beutler’s lab synthesized and packaged it and supplied it to the hospital pharmacy; the lab also developed a test to monitor blood levels. This was the first treatment that led to prolonged remission of HCL, which was previously untreatable.[17]:14–15

In February 1991 Scripps began a collaboration with Johnson & Johnson to bring intravenous cladribine to market and by December of that year J&J had filed an NDA; cladrabine was approved by the FDA in 1993 for HCL as an orphan drug,[18] and was approved in Europe later that year.[19]:2

The subcutaneous formulation was developed in Switzerland in the early 1990s and it was commercialized by Lipomed GmbH in the 2000s.[19]:2[20]

Multiple sclerosis

In the mid-1990s Beutler, in collaboration with Jack Sipe, a neurologist at Scripps, ran several clinical trials exploring the utility of cladribine in multiple sclerosis, based on the drug’s immunosuppressive effects. Sipe’s insight into MS, and Beutler’s interest in MS due to his sister’s having had it, led a very productive collaboration.[17]:17[21] Ortho-Clinical, a subsidiary of J&J, filed an NDA for cladribine for MS in 1997 but withdrew it in the late 1990s after discussion with the FDA proved that more clinical data would be needed.[22][23]

Ivax acquired the rights for oral administration of cladribine to treat MS from Scripps in 2000,[24] and partnered with Serono in 2002.[23] Ivax was acquired by Teva in 2006,[25][26] and Merck KGaA acquired control of Serono’s drug business in 2006.[27]

An oral formulation of the drug with cyclodextrin was developed[28]:16 and Ivax and Serono, and then Merck KGaA conducted several clinical studies. Merck KGaA submitted an application to the European Medicines Agency in 2009, which was rejected in 2010, and an appeal was denied in 2011.[28]:4–5 Likewise Merck KGaA’s NDA with the FDA rejected in 2011.[29] The concerns were that several cases of cancer had arisen, and the ratio of benefit to harm was not clear to regulators.[28]:54–55 The failures with the FDA and the EMA were a blow to Merck KGaA and were one of a series of events that led to a reorganization, layoffs, and closing the Swiss facility where Serono had arisen.[30][31] However, several MS clinical trials were still ongoing at the time of the rejections, and Merck KGaA committed to completing them.[29] A meta-analysis of data from clinical trials showed that cladiribine did not increase the risk of cancer at the doses used in the clinical trials.[32]

In 2015 Merck KGaA announced it would again seek regulatory approval with data from the completed clinical trials in hand,[30] and in 2016 the EMA accepted its application for review.[33] On June 22, 2017, the EMA’s Committee for Medicinal Products for Human Use (CHMP) adopted a positive opinion, recommending the granting of a marketing authorisation for the treatment of relapsing forms of multiple sclerosis.[34]

Finally, after all these problems it was approved in Europe on August 2017 for highly active RRMS.[35]

Efficacy

Cladribine is an effective treatment for relapsing remitting MS, with a reduction in the annual rate of relapses of 54.5%.[11] These effects may be sustained up to 4 years after initial treatment, even if no further doses are given.[36] Thus, cladribine is considered to be a highly effective immune reconstitution therapy in MS. Similar to alemtuzumab, cladribine is given as two courses approximately one year apart. Each course consists of 4-5 tablets given over a week in the first month, followed by a second dosing of another 4-5 tablets the following month[37] During this time and after the final dose patients are monitored for adverse effects and signs of relapse.

https://www.merckneurology.co.uk/wp-content/uploads/2017/08/mavenclad-table-1.jpg

Safety

Compared to alemtuzumab, cladribine is associated with a lower rate of severe lymphopenia. It also appears to have a lower rate of common adverse events, especially mild to moderate infections[11][36] As cladribine is not a recombinant biological therapy, it is not associated with the development of antibodies against the drug, which might reduce the effectiveness of future doses. Also, unlike alemtuzumab, cladribine is not associated with secondary autoimmunity.[38]

This is probably due to the fact cladribine more selectively targets B cells. Unlike alemtuzumab, cladribine is not associated with a rapid repopulation of the peripheral blood B cell pool, which then ´overshoots´ the original number by up to 30%.[39] Instead, B cells repopulate more slowly, reaching near normal total B cells numbers at 1 year. This phenomenon and the relative sparing of T cells, some of which might be important in regulating the system against other autoimmune reactions, is thought to explain the lack of secondary autoimmunity.

Use in clinical practice

The decision to start cladribine in MS depends on the degree of disease activity (as measured by number of relapses in the past year and T1 gadolinium-enhancing lesions on MRI), the failure of previous disease-modifying therapies, the potential risks and benefits and patient choice.

In the UK, the National Institute for Clinical Excellence (NICE) recommends cladribine for treating highly active RRMS in adults if the persons has:

rapidly evolving severe relapsing–remitting multiple sclerosis, that is, at least 2 relapses in the previous year and at least 1 T1 gadolinium-enhancing lesion at baseline MRI or

relapsing–remitting multiple sclerosis that has responded inadequately to treatment with disease-modifying therapy, defined as 1 relapse in the previous year and MRI evidence of disease activity.[40]

People with MS require counselling on the intended benefits of cladribine in reducing the risk of relapse and disease progression, versus the risk of adverse effects such as headaches, nausea and mild to moderate infections. Women of childbearing age also require counselling that they should not conceive while taking cladribine, due to the risk of harm to the fetus.

Cladribine, as the 10 mg oral preparation Mavenclad, is administered as two courses of tablets approximately one year apart. Each course consists of four to five treatment days in the first month, followed by an additional four to five treatment days in the second month. The recommended dose of Mavenclad is 3.5 mg/kg over 2 years, given in two treatment courses of 1.75 mg/kg/year. Therefore, the number of tablets administered on each treatment day depends on the person’s weight. A full guide to the dosing strategy can be found below:

https://www.merckneurology.co.uk/mavenclad/mavenclad-efficacy/

After treatment, people with MS are monitored with regular blood tests, looking specifically at the white cell count and liver function. Patients should be followed up regularly by their treating neurologist to assess efficacy, and should be able to contact their MS service in the case of adverse effects or relapse. After the first two years of active treatment no further therapy may need to be given, as cladribine has been shown to be efficacious for up to last least four years after treatment. However, if patients fail to respond, options include switching to other highly effective disease-modifying therapies such as alemtuzumab, fingolimod or natalizumab.

Research directions

Cladribine has been studied as part of a multi-drug chemotherapy regimen for drug-resistant T-cell prolymphocytic leukemia.[41]

REF

A universal biocatalyst for the preparation of base- and sugar-modified nucleosides via an enzymatic transglycosylation
Helv Chim Acta 2002, 85(7): 1901

Synthesis of 2-chloro-2′-deoxyadenosine by microbiological transglycosylation
Nucleosides Nucleotides 1993, 12(3-4): 417

Synthesis of 2-chloro-2′-deoxyadenosine by washed cells of E. coli
Biotechnol Lett 1992, 14(8): 669

Efficient syntheses of 2-chloro-2′-deoxyadenosine (cladribine) from 2′-deoxyguanosine
J Org Chem 2003, 68(3): 989

WO 2004028462

Synthesis of 2′-deoxytubercidin, 2′-deoxyadenosine, and related 2′-deoxynucleosides via a novel direct stereospecific sodium salt glycosylation procedure
J Am Chem Soc 1984, 106(21): 6379

WO 2011113476

A stereoselective process for the manufacture of a 2′-deoxy-beta-D-ribonucleoside using the vorbruggen glycosylation
Org Process Res Dev 2013, 17(11): 1419

A new synthesis of 2-chloro-2′-deoxyadenosine (Cladribine), CdA)
Nucleosides Nucleotides Nucleic Acids 2011, 30(5): 353

A dramatic concentration effect on the stereoselectivity of N-glycosylation for the synthesis of 2′-deoxy-beta-ribonucleosides
Chem Commun (London) 2012, 48(56): 7097

CN 105367616

PATENT

https://patents.google.com/patent/EP2891660A1/en

Previously Robins and Robins (Robins, M. J. and Robins, R. K., J. Am. Chem. Soc. 1965, 87, 4934-4940) reported that acid-catalyzed fusion of 1,3,5-tri-O-acety-2-deoxy-D-ribofuranose and 2,6-dichloropurine gave a 65% yield of an anomeric mixture 2,6-dichloro-9-(3′,5′-di-O-acetyl-2′-deoxy-α-,β-D-ribofuranosyl)-purines from which the α-anomer was obtained as a pure crystalline product by fractional crystallization from ethanol in 32% yield and the equivalent β-anomer remained in the mother liquor (see Scheme 1). The β-anomer, which could have been used to synthesize cladribine, wasn’t isolated further. The α-anomer was treated with methanolic ammonia which resulted in simultaneous deacetylation and amination to give 6-amino-2-chloro-9-(2′-deoxy-α-D-ribofuranosyl)-purine, which is a diastereomer of cladribine.

Figure imgb0001

[0004]

Broom et al. (Christensen, L. F., Broom, A. D., Robins, M. J., and Bloch, A., J. Med. Chem. 1972, 15, 735-739) adapted Robins et al.’s method by treating the acetylated mixture (viz., 2,6-dichloro-9-(3′,5′-di-O-acety-2′-deoxy-α,β-D-ribofuranosyl)-purine) with liquid ammonia and reacylating the resulting 2′-deoxy-α-and –β-adenosines with p-toluoyl chloride (see Scheme 2). The desired 2-chloro-9-(3′,5′-di-Op-toluoyl-2′-deoxy-β-D-ribofuranosyl)-adenine was then separated by chromatography and removal of the p-toluoyl group resulted in cladribine in 9% overall yield based on the fusion of 1,3,5-tri-O-acety-2-deoxy-D-ribofuranose and 2,6-dichloropurine.

Figure imgb0002
[0005]

To increase the stereoselectivity in favour of the β-anomer, Robins et al.(Robins, R. L. et al., J. Am. Chem. Soc. 1984, 106, 6379-6382US4760137 EP0173059 ) provided an improved method in which the sodium salt of 2,6-dichloropurine was coupled with 1-chloro-2-deoxy-3,5-di-Op-toluoyl-α-D-ribofuranose in acetonitrile (MeCN) to give the protected β-nucleoside in 59% isolated yield, following chromatography and crystallisation, in addition to 13% of the undesired N-7 regioisomer (see Scheme 3). The apparently higher selectivity in this coupling reaction is attributed to it being a direct SN2 displacement of the chloride ion by the purine sodium salt. The protected N-9 2′-deoxy-β-nucleoside was treated with methanolic ammonia at 100°C to give cladribine in an overall 42% yield. The drawback of this process is that the nucleophilic 7- position nitrogen competes in the SN2 reaction against the nucleophilic 9- position, leading to a mixture of the N-7 and N-9 glycosyl isomers as well as the need for chromatography and crystallisation to obtain the pure desired isomer.

Figure imgb0003
[0006]

Gerszberg and Alonso (Gerszberg S. and Alonso, D. WO0064918 , and US20020052491 ) also utilised an SN2 approach with 1-chloro-2-deoxy-3,5-di-Op-toluoyl-α-D-ribofuranose but instead coupled it with the sodium salt of 2-chloroadenine in acetone giving the desired β-anomer of the protected cladribine in 60% yield following crystallisation from ethanol (see Scheme 4). After the deprotection step using ammonia in methanol (MeOH), the β-anomer of cladribine was isolated in an overall 42% yield based on the 1-chlorosugar, and 30% if calculated based on the sodium salt since this was used in a 2.3 molar excess.

Figure imgb0004
[0007]

To increase the regioselectivity towards glycosylation of the N-9 position, Gupta and Munk recently ( Gupta, P. K. and Munk, S. A., US20040039190 WO2004018490 and CA2493724 ) conducted an SN2 reaction using the anomerically pure α-anomer, 1-chloro-2-deoxy-3,5-di-Op-toluoyl-α-D-ribofuranose but coupling it with the potassium salt of a 6-heptanoylamido modified purine (see Scheme 5). The bulky alkyl group probably imparted steric hindrance around the N-7 position, resulting in the reported improved regioselectivity. Despite this, following deprotection, the overall yield of cladribine based on the 1-chlorosugar was 43%, showing no large improvement in overall yield on related methods. Moreover 2-chloroadenine required prior acylation with heptanoic anhydride at high temperature (130°C) in 72% yield, and the coupling required cryogenic cooling (-30°C) and the use of the strong base potassium hexamethyldisilazide and was followed by column chromatography to purify the product protected cladribine.

Figure imgb0005
[0008]

More recently Robins et al. (Robins, M. J. et al., J. Org. Chem. 2006, 71, 7773-7779US20080207891 ) published a procedure for synthesis of cladribine that purports to achieve almost quantitative yields in the N-9-regioselective glycosylation of 6-(substituted-imidazol-1-yl)-purine sodium salts with 1-chloro-2-deoxy-3,5-di-Op-toluoyl-α-D-ribofuranose in MeCN/dichloromethane (DCM) mixtures to give small or no detectable amounts of the undesired α-anomer (see Scheme 6). In actuality this was only demonstrated on the multi-milligram to several grams scale, and whilst the actual coupling yield following chromatography of the desired N-9-β-anomer was high (83% to quantitative), the protected 6-(substituted-imidazol-1-yl)-products were obtained in 55% to 76% yield after recrystallisation. Following this, toxic benzyl iodide was used to activate the 6-(imidazole-1-yl) groups which were then subsequently displaced by ammonia at 60-80°C in methanolic ammonia to give cladribine in 59-70% yield following ion exchange chromatography and multiple crystallisations, or following extraction with DCM and crystallisation. Although high anomeric and regioselective glycosylation was demonstrated the procedure is longer than the prior arts, atom uneconomic and not readily applicable to industrial synthesis of cladribine such as due to the reliance on chromatography and the requirement for a pressure vessel in the substitution of the 6-(substituted-imidazole-1-yl) groups.

Figure imgb0006
[0009]
Therefore, there is a need for a more direct, less laborious process, which will produce cladribine in good yield and high purity that is applicable to industrial scales.

EXAMPLE 1 Preparation of 2-chloro-6-trimethylsilylamino-9-[3,5-di-O-(4-chlorobenzoyl)-2-deoxy-β-D-ribofuranosyl]-purine

  • [0052]
    2-Chloroadenine (75 g, 0.44 mol, 1.0 eq.), MeCN (900 mL, 12 P), and BSTFA (343.5 g, 1.33 mol, 3.0 eq.) were stirred and heated under reflux until the mixture was almost turned clear. The mixture was cooled to 60°C and TfOH (7.9 mL, 0.089 mol, 0.2 eq.) and then 1-O-acetyl-3,5-di-O-(4-chlorobenzoyl)-2-deoxy-D-ribofuranose (III; 200.6 g, 1.0 eq.) were added into the mixture, and then the mixture was stirred at 60°C. After 1 hour, some solid precipitated from the solution and the mixture was heated for at least a further 10 hours. The mixture was cooled to r.t. and stirred for 2 hours. The solid was filtered and dried in vacuo at 60°C to give 180.6 g in 64% yield of a mixture of 2-chloro-6-trimethylsilylamino-9-[3,5-di-O-(4-chlorobenzoyl)-2-deoxy-β-D-ribofurano syl]-purine (IVa) with 95.4% HPLC purity and its non-silylated derivative 2-chloro-6-amino-9-[3,5-di-O-(4-chlorobenzoyl)-2′-deoxy-β-D-ribofuranosyl]-purine (IVb) with 1.1 % HPLC purity.

EXAMPLE 2 Preparation of 2-chloro-6-trimethylsilylamino-9-[3,5-di-O-(4-chlorobenzoyl)-2-deoxy-β-D-ribofuranosyl]-purine by isomerisation of a mixture of 2-chloro-6-amino-9-[3,5-di-O-(4-chlorobenzoyl)-2-deoxy-α,β-D-ribofuranosyl]-purine mixture

  • [0053]
    50.0 g of 2-chloro-6-amino-9-[3,5-di-O-(4-chlorobenzoyl)-2-deoxy-α,β-D-ribofuranosyl]-purine as a 0.6:1.0 mixture of the β-anomer IVb and α-anomer Vb(83.16 mmol, assay of α-anomer was 58.6% (52.06 mmol) and β-anomer was 34.3% (31.10 mmol, 17.15 g)), 68.6 g BSTFA (266.5 mmol) and 180 mL of MeCN (3.6 P) were charged into a dried 4-necked flask. The mixture was heated to 60°C under N2 for about 3 h and then 2.67 g of TfOH (17.8 mmol) was added. The mixture was stirred at 60°C for 15 h and was then cooled to about 25°C and stirred for a further 2 h, and then filtered. The filter cake was washed twice with MeCN (20 mL each) and dried at 60°C in vacuo for 6 h to give 24 g of off-white solid (the assay of 2-chloro-6-amino-9-[3,5-di-O-(4-chlorobenzoyl)-2-deoxy-α-D-ribofuranosyl]-purine was 1.4% (0.60 mmol, 0.34 g),
    2-chloro-6-amino-9-[3,5-di-O-(4-chlorobenzoyl)-2-deoxy-β-D-ribofuranosyl]-purine was 8.4% (3.18 mmol, 2.02 g) and
    2-chloro-6-trimethylsilylamino-9-[3,5-di-O-(4-chlorobenzoyl)-2-deoxy-β-D-ribofuranosyl]-purine was 86.6% (32.73 mmol, 20.78 g)).
    Analysis of the 274.8 g of the mother liquor by assay showed that it in addition to the α-anomer it contained 0.5% (1.37 g, 2.43 mmol) of
    2-chloro-6-amino-9-[3,5-di-O-(4-chlorobenzoyl)-2-deoxy-β-D-ribofuranosyl]-purine and 0.01% (0.027 g, 0.05 mmol) of
    2-chloro-6-trimethylsilylamino-9-[3,5-di-O-(4-chlorobenzoyl)-2-deoxy-β-D-ribofuranosyl]-purine.

EXAMPLE 3 Preparation of 2-chloro-2′-deoxy-adenosine (cladribine)

  • [0054]
    To the above prepared mixture of 2-chloro-6-trimethylsilylamino-9-[3,5-di-O-(4-chlorobenzoyl)-2-deoxy-β-D-ribofurano syl]- purine (IVa) and 2-chloro-6-amino-9-[3,5-di-O-(4-chlorobenzoyl)-2′-deoxy-β-D-ribofuranosyl]-purine (IVb) (179 g, >95.4% HPLC purity) in MeOH (895 mL, 5 P) was added 29% MeONa/MeOH solution (5.25 g, 0.1 eq.) at 20-30°C. The mixture was stirred at 20-30°C for 6 hours, the solid was filtered, washed with MeOH (60 mL, 0.34 P) and then dried in vacuo at 50°C for 6 hour to give 72 g white to off-white crude cladribine with 98.9% HPLC purity in ca. 93% yield.

EXAMPLE 4 Recrystallisation

  • [0055]
    Crude cladribine (70 g), H2O (350 mL, 5 P), MeOH (350 mL, 5 P) and 29% MeONa/MeOH solution (0.17 g) were stirred and heated under reflux until the mixture turned clear. The mixture was stirred for 3 hour and was then filtered to remove the precipitates at 74-78°C. The mixture was stirred and heated under reflux until the mixture turned clear and was then cooled. Crystals started to form at ca. 45°C. The slurry was stirred for 2 hour at the cloudy point. The slurry was cooled slowly at a rate of 5°C/0.5 hour. The slurry was stirred at 10-20°C for 4-8 hours and then filtered. The filter cake was washed three times with MeOH (50 mL each) and dried at 50°C in vacuo for 6 hours to give 62.7 g of 99.9% HPLC pure cladribine in ca. 90% yield.

EXAMPLE 5 Preparation of 2-chloro-6-trimethylsilylamino-9-[3,5-di-O-(4-chlorobenzoyl)-2-deoxy-β-D-ribofuranosyl]-purine

  • [0056]
    2-Chloroadenine (2.2 Kg, 13.0 mol, 1.0 eq.), MeCN (20.7 Kg, 12 P), and BSTFA (10.0 Kg, 38.9 mol, 3.0 eq.) were stirred and heated under reflux for 3 hours and then filtered through celite and was cooled to about 60°C. TfOH (0.40 Kg, 2.6 mol, 0.2 eq.) and 1-O-acetyl-3,5-di-O-(4-chlorobenzoyl)-2-deoxy-D-ribofuranose (III; 5.87 Kg, 13.0 mol, 1.0 eq.) were added into the filtrate and the mixture was stirred at about 60°C for 29.5 hours. The slurry was cooled to about 20°C and stirred for 2 hours. The solids were filtered and washed with MeCN (2.8 Kg) twice and dried in vacuo at 60°C to give 5.17 Kg with a 96.5% HPLC purity in 62% yield of a mixture of 2-chloro-6-trimethylsilylamino-9-[3,5-di-O-(4-chlorobenzoyl)-2-deoxy-β-D-ribofurano syl]-purine (IVa), and non-silylated derivative 2-chloro-6-amino-9-[3,5-di-O-(4-chlorobenzoyl)-2′-deoxy-β-D-ribofuranosyl]-purine (IVb).

EXAMPLE 6 Preparation of 2-chloro-2′-deoxy-adenosine (cladribine)

  • [0057]
    To a mixture of 25% sodium methoxide in MeOH (0.11 Kg, 0.5 mol, 0.1 eq.) and MeOH (14.8 Kg, 5 P) at about at 25°C was added 2-chloro-6-trimethylsilylamino-9-[3,5-di-O-(4-chlorobenzoyl)-2-deoxy-β-D-ribofurano syl]-purine (IVa) and non-silylated derivative 2-chloro-6-amino-9-[3,5-di-O-(4-chlorobenzoyl)-2′-deoxy-β-D-ribofuranosyl]-purine (IVb) (3.70 Kg, combined HPLC purity of >96.3%) and the mixture was agitated at about 25°C for 2 hours. The solids were filtered, washed with MeOH (1.11 Kg, 0.4 P) and then dried in vacuo at 60°C for 4 hours to give 1.43 Kg of a crude cladribine with 97.8% HPLC purity in ca. 87% yield.

EXAMPLE 7 Recrystallisation of crude cladribine

  • [0058]
    A mixture of crude cladribine (1.94 Kg, >96.0% HPLC purity), MeOH (7.77 Kg, 5 P), process purified water (9.67 Kg, 5 P) and 25% sodium methoxide in MeOH (32 g, 0.15 mol) were stirred and heated under reflux until the solids dissolved. The solution was cooled to about 70°C and treated with activated carbon (0.16 Kg) and celite for 1 hour at about 70°C, rinsed with a mixture of preheated MeOH and process purified water (W/W = 1:1.25, 1.75 Kg). The filtrate was cooled to about 45°C and maintained at this temperature for 1 hours, and then cooled to about 15°C and agitated at this temperature for 2 hours. The solids were filtered and washed with MeOH (1.0 Kg, 0.7 P) three times and were then dried in vacuo at 60°C for 4 hours giving API grade cladribine (1.5 Kg, 5.2 mol) in 80% yield with 99.84% HPLC purity.

EXAMPLE 8 Recrystallisation of crude cladribine

  • [0059]
    A mixture of crude cladribine (1.92 Kg, >95.7% HPLC purity), MeOH (7.76 Kg, 5 P), process purified water (9.67 Kg, 5 P) and 25% sodium methoxide in MeOH (36 g, 0.17 mol) were stirred and heated under reflux until the solids dissolved. The solution was cooled to about 70°C and treated with activated carbon (0.15 Kg) and celite for 1 hour at about 70°C, rinsed with a mixture of preheated MeOH and process purified water (1:1.25, 1.74 Kg). The filtrate was cooled to about 45°C and maintained at this temperature for 1 hour, and then cooled to about 15°C and agitated at this temperature for 2 hours. The solids were filtered and washed with MeOH (1.0 Kg, 0.7 P) three times and were giving damp cladribine (1.83 Kg). A mixture of this cladribine (1.83 Kg), MeOH (7.33 Kg, 5 P) and process purified water (9.11 Kg, 5 P) were stirred and heated under reflux until the solids dissolved and was then cooled to about 45°C and maintained at this temperature for 1 hours. The slurry was further cooled to about 15°C and agitated at this temperature for 2 hours. The solids were filtered and washed with MeOH (0.9 Kg, 0.7 P) three times and were then dried in vacuo at 60°C for 4 hours giving API grade cladribine (1.38 Kg, 4.8 mol) in 75% yield with 99.86% HPLC purity.

SYN

Image result for cladribine

Cladribine can be got from 2-Deoxy-D-ribose. The detail is as follows:

Production of Cladribine

SYN

https://www.tandfonline.com/doi/abs/10.1080/15257770.2015.1071848?journalCode=lncn20

clip
FDA approves new oral treatment for multiple sclerosis, Mavenclad (cladribine)
The U.S. Food and Drug Administration today approved Mavenclad (cladribine) tablets to treat
relapsing forms of multiple sclerosis (MS) in adults, to include relapsing-remitting disease and active secondary progressive disease. Mavenclad is not recommended for MS patients with clinically isolated syndrome. Because of its safety profile, the use of Mavenclad is generally recommended for patients who have had an inadequate response to…

March 29, 2019

Release

The U.S. Food and Drug Administration today approved Mavenclad (cladribine) tablets to treat relapsing forms of multiple sclerosis (MS) in adults, to include relapsing-remitting disease and active secondary progressive disease. Mavenclad is not recommended for MS patients with clinically isolated syndrome. Because of its safety profile, the use of Mavenclad is generally recommended for patients who have had an inadequate response to, or are unable to tolerate, an alternate drug indicated for the treatment of MS.

“We are committed to supporting the development of safe and effective treatments for patients with multiple sclerosis,” said Billy Dunn, M.D., director of the Division of Neurology Products in the FDA’s Center for Drug Evaluation and Research. “The approval of Mavenclad represents an additional option for patients who have tried another treatment without success.”

MS is a chronic, inflammatory, autoimmune disease of the central nervous system that disrupts communications between the brain and other parts of the body. Most people experience their first symptoms of MS between the ages of 20 and 40. MS is among the most common causes of neurological disability in young adults and occurs more frequently in women than in men.

For most people, MS starts with a relapsing-remitting course, in which episodes of worsening function (relapses) are followed by recovery periods (remissions). These remissions may not be complete and may leave patients with some degree of residual disability. Many, but not all, patients with MS experience some degree of persistent disability that gradually worsens over time. In some patients, disability may progress independent of relapses, a process termed secondary progressive multiple sclerosis (SPMS). In the first few years of this process, many patients continue to experience relapses, a phase of the disease described as active SPMS. Active SPMS is one of the relapsing forms of MS, and drugs approved for the treatment of relapsing forms of MS can be used to treat active SPMS.

The efficacy of Mavenclad was shown in a clinical trial in 1,326 patients with relapsing forms of MS who had least one relapse in the previous 12 months. Mavenclad significantly decreased the number of relapses experienced by these patients compared to placebo. Mavenclad also reduced the progression of disability compared to placebo.

Mavenclad must be dispensed with a patient Medication Guide that describes important information about the drug’s uses and risks. Mavenclad has a Boxed Warning for an increased risk of malignancy and fetal harm. Mavenclad is not to be used in patients with current malignancy. In patients with prior malignancy or with increased risk of malignancy, health care professionals should evaluate the benefits and risks of the use of Mavenclad on an individual patient basis. Health care professionals should follow standard cancer screening guidelines in patients treated with Mavenclad. The drug should not be used in pregnant women and in women and men of reproductive potential who do not plan to use effective contraception during treatment and for six months after the course of therapy because of the potential for fetal harm. Mavenclad should be stopped if the patient becomes pregnant.

Other warnings include the risk of decreased lymphocyte (white blood cell) counts; lymphocyte counts should be monitored before, during and after treatment. Mavenclad may increase the risk of infections; health care professionals should screen patients for infections and treatment with Mavenclad should be delayed if necessary. Mavenclad may cause hematologic toxicity and bone marrow suppression so health care professionals should measure a patient’s complete blood counts before, during and after therapy. The drug has been associated with graft-versus-host-disease following blood transfusions with non-irradiated blood. Mavenclad may cause liver injury and treatment should be interrupted or discontinued, as appropriate, if clinically significant liver injury is suspected.

The most common adverse reactions reported by patients receiving Mavenclad in the clinical trials include upper respiratory tract infections, headache and decreased lymphocyte counts.

The FDA granted approval of Mavenclad to EMD Serono, Inc.

References

  1. ^ Drugs.com International trade names for Cladribine Page accessed Jan 14, 2015
  2. Jump up to:a b c d “PRODUCT INFORMATION LITAK© 2 mg/mL solution for injection” (PDF)TGA eBusiness Services. St Leonards, Australia: Orphan Australia Pty. Ltd. 10 May 2010. Retrieved 27 November 2014.
  3. ^ Liliemark, Jan (1997). “The Clinical Pharmacokinetics of Cladribine”. Clinical Pharmacokinetics32 (2): 120–131. doi:10.2165/00003088-199732020-00003PMID 9068927.
  4. Jump up to:a b “European Medicines Agency – – Litak”http://www.ema.europa.eu.
  5. Jump up to:a b “Leustat Injection. – Summary of Product Characteristics (SPC) – (eMC)”http://www.medicines.org.uk.
  6. ^ Leist, TP; Weissert, R (2010). “Cladribine: mode of action and implications for treatment of multiple sclerosis”. Clinical Neuropharmacology34 (1): 28–35. doi:10.1097/wnf.0b013e318204cd90PMID 21242742.
  7. Jump up to:a b c d Cladribine label, last updated July 2012. Page accessed January 14, 2015
  8. ^ https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm634837.htm
  9. ^ Histiocytosis Association Erdheim-Chester Disease Page accessed Aug 20, 2016
  10. ^ Aricò M (2016). “Langerhans cell histiocytosis in children: from the bench to bedside for an updated therapy”. Br J Haematol173 (5): 663–70. doi:10.1111/bjh.13955PMID 26913480The combination of cytarabine and cladribine is the current standard for second-line therapy of refractory cases with vital organ dysfunction.
  11. Jump up to:a b c Giovannoni, G; Comi, G; Cook, S; Rammohan, K; Rieckmann, P; Soelberg Sørensen, P; Vermersch, P; Chang, P; Hamlett, A; Musch, B; Greenberg, SJ; CLARITY Study, Group. (4 February 2010). “A placebo-controlled trial of oral cladribine for relapsing multiple sclerosis”. The New England Journal of Medicine362 (5): 416–26. doi:10.1056/NEJMoa0902533PMID 20089960.
  12. ^ Johnston, JB (June 2011). “Mechanism of action of pentostatin and cladribine in hairy cell leukemia”. Leukemia & Lymphoma. 52 Suppl 2: 43–5. doi:10.3109/10428194.2011.570394PMID 21463108.
  13. ^ Beutler, E; Piro, LD; Saven, A; Kay, AC; McMillan, R; Longmire, R; Carrera, CJ; Morin, P; Carson, DA (1991). “2-Chlorodeoxyadenosine (2-CdA): A Potent Chemotherapeutic and Immunosuppressive Nucleoside”. Leukemia & Lymphoma5 (1): 1–8. doi:10.3109/10428199109068099PMID 27463204.
  14. ^ Baker, D; Marta, M; Pryce, G; Giovannoni, G; Schmierer, K (February 2017). “Memory B Cells are Major Targets for Effective Immunotherapy in Relapsing Multiple Sclerosis”EBioMedicine16: 41–50. doi:10.1016/j.ebiom.2017.01.042PMC 5474520PMID 28161400.
  15. ^ Baker, D; Herrod, SS; Alvarez-Gonzalez, C; Zalewski, L; Albor, C; Schmierer, K (July 2017). “Both cladribine and alemtuzumab may effect MS via B-cell depletion”Neurology: Neuroimmunology & Neuroinflammation4 (4): e360. doi:10.1212/NXI.0000000000000360PMC 5459792PMID 28626781.
  16. Jump up to:a b Ceronie, B; Jacobs, BM; Baker, D; Dubuisson, N; Mao, Z; Ammoscato, F; Lock, H; Longhurst, HJ; Giovannoni, G; Schmierer, K (May 2018). “Cladribine treatment of multiple sclerosis is associated with depletion of memory B cells”Journal of Neurology265 (5): 1199–1209. doi:10.1007/s00415-018-8830-yPMC 5937883PMID 29550884.
  17. Jump up to:a b Marshall A. Lichtman Biographical Memoir: Ernest Beutler 1928–2008 National Academy of Sciences, 2012
  18. ^ Staff, The Pink Sheet Mar 8, 1993 Ortho Biotech’s Leustatin For Hairy Cell Leukemia
  19. Jump up to:a b EMA 2004 Litak EMA package: Scientific Discussion
  20. ^ EMA 2004 Litak: Background Information one the Procedure
  21. ^ Eric Sauter and Mika Ono for Scripps News and Views. Vol 9. Issue 18. June 1, 2009 A Potential New MS Treatment’s Long and Winding Road
  22. ^ Tortorella C, Rovaris M, Filippi M (2001). “Cladribine. Ortho Biotech Inc”. Curr Opin Investig Drugs2 (12): 1751–6. PMID 11892941.
  23. Jump up to:a b Carey Sargent for Dow Jones Newswires in the Wall Street Journal. Oct. 31, 2002 Serono Purchases Rights To Experimental MS Drug
  24. ^ Reuters. Dec 4, 2000. Ivax to Develop Cladribine for Multiple Sclerosis
  25. ^ Jennifer Bayot for the New York Times. July 26, 2005 Teva to Acquire Ivax, Another Maker of Generic Drugs
  26. ^ Teva Press Release, 2006. Teva Completes Acquisition of Ivax
  27. ^ Staff, First Word Pharma. Sept 21, 2006 Merck KGaA to acquire Serono
  28. Jump up to:a b c EMA. 2011 Withdrawal Assessment Report for Movectro Procedure No. EMEA/H/C/001197
  29. Jump up to:a b John Gever for MedPage Today June 22, 2011 06.22.2011 0 Merck KGaA Throws in Towel on Cladribine for MS
  30. Jump up to:a b John Carroll for FierceBiotech Sep 11, 2015 Four years after a transatlantic slapdown, Merck KGaA will once again seek cladribine OK
  31. ^ Connolly, Allison (24 April 2012). “Merck KGaA to Close Merck Serono Site in Geneva, Cut Jobs”Bloomberg.
  32. ^ Pakpoor, J; et al. (December 2015). “No evidence for higher risk of cancer in patients with multiple sclerosis taking cladribine”Neurology: Neuroimmunology & Neuroinflammation2 (6): e158. doi:10.1212/nxi.0000000000000158PMC 4592538PMID 26468472.
  33. ^ Press release
  34. ^ Merck. “Cladribine Tablets Receives Positive CHMP Opinion for Treatment of Relapsing Forms of Multiple Sclerosis”http://www.prnewswire.co.uk. Retrieved 2017-08-22.
  35. ^ Cladribine approved in Europe, Press Release
  36. Jump up to:a b Giovannoni, G; Soelberg Sorensen, P; Cook, S; Rammohan, K; Rieckmann, P; Comi, G; Dangond, F; Adeniji, AK; Vermersch, P (1 August 2017). “Safety and efficacy of cladribine tablets in patients with relapsing-remitting multiple sclerosis: Results from the randomized extension trial of the CLARITY study”. Multiple Sclerosis (Houndmills, Basingstoke, England): 1352458517727603. doi:10.1177/1352458517727603PMID 28870107.
  37. ^ “Sustained Efficacy – Merck Neurology”Merck Neurology. Retrieved 28 September2018.
  38. ^ Guarnera, C; Bramanti, P; Mazzon, E (2017). “Alemtuzumab: a review of efficacy and risks in the treatment of relapsing remitting multiple sclerosis”Therapeutics and Clinical Risk Management13: 871–879. doi:10.2147/TCRM.S134398PMC 5522829PMID 28761351.
  39. ^ Baker, D; Herrod, SS; Alvarez-Gonzalez, C; Giovannoni, G; Schmierer, K (1 August 2017). “Interpreting Lymphocyte Reconstitution Data From the Pivotal Phase 3 Trials of Alemtuzumab”JAMA Neurology74 (8): 961–969. doi:10.1001/jamaneurol.2017.0676PMC 5710323PMID 28604916.
  40. ^ “Cladribine tablets for treating relapsing–remitting multiple sclerosis”National Institute for Clinical Excellence. Retrieved 23 September 2018.
  41. ^ Hasanali, Zainul S.; Saroya, Bikramajit Singh; Stuart, August; Shimko, Sara; Evans, Juanita; Shah, Mithun Vinod; Sharma, Kamal; Leshchenko, Violetta V.; Parekh, Samir (24 June 2015). “Epigenetic therapy overcomes treatment resistance in T cell prolymphocytic leukemia”Science Translational Medicine7 (293): 293ra102. doi:10.1126/scitranslmed.aaa5079ISSN 1946-6234PMC 4807901PMID 26109102.
Cladribine
Cladribine.svg
Clinical data
Trade names Leustatin, others[1]
AHFS/Drugs.com Monograph
MedlinePlus a693015
License data
Pregnancy
category
  • AU:D
  • US:D (Evidence of risk)
Routes of
administration
Intravenoussubcutaneous(liquid)
ATC code
Legal status
Legal status
  • AU:S4 (Prescription only)
  • CA℞-only
  • UK:POM (Prescription only)
Pharmacokinetic data
Bioavailability 100% (i.v.); 37 to 51% (orally)[3]
Protein binding 25% (range 5-50%)[2]
Metabolism Mostly via intracellularkinases; 15-18% is excreted unchanged[2]
Elimination half-life Terminal elimination half-life: Approximately 10 hours after both intravenous infusion an subcutaneous bolus injection[2]
Excretion Urinary[2]
Identifiers
CAS Number
PubChemCID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEBI
ChEMBL
ECHA InfoCard 100.164.726Edit this at Wikidata
Chemical and physical data
Formula C10H12ClN5O3
Molar mass 285.687 g/mol g·mol−1
3D model (JSmol)
Cladribine
CAS Registry Number: 4291-63-8
CAS Name: 2-Chloro-2¢-deoxyadenosine
Additional Names: 2-chloro-6-amino-9-(2-deoxy-b-D-erythro-pentofuranosyl)purine; 2-chlorodeoxyadenosine; 2-CdA; CldAdo
Manufacturers’ Codes: NSC-105014-F
Trademarks: Leustatin (Ortho Biotech)
Molecular Formula: C10H12ClN5O3
Molecular Weight: 285.69
Percent Composition: C 42.04%, H 4.23%, Cl 12.41%, N 24.51%, O 16.80%
Literature References: Substituted purine nucleoside with antileukemic activity. Prepn as intermediate in synthesis of 2-deoxynucleosides: H. Venner, Ber. 93, 140 (1960); M. Ikehara, H. Tada, J. Am. Chem. Soc. 85, 2344 (1963); eidem, ibid. 87, 606 (1965). Synthesis and biological activity: L. F. Christensen et al., J. Med. Chem. 15, 735 (1972). Stereospecific synthesis: Z. Kazimierczuk et al., J. Am. Chem. Soc. 106, 6379 (1984); R. K. Robins, G. R. Revankar, EP 173059eidem, US 4760137 (1986, 1988 both to Brigham Young Univ.). Specific toxicity to lymphocytes: D. A. Carson et al., Proc. Natl. Acad. Sci. USA 77, 6865 (1980); eidem, Blood 62, 737 (1983). Mechanism of action: S. Seto et al., J. Clin. Invest. 75, 377 (1985). Clinical evaluation in chronic lymphocytic leukemia: L. D. Piro et al., Blood 72, 1069 (1988); in hairy cell leukemia: eidem, N. Engl. J. Med. 322, 1117 (1990).
Properties: Crystals from water, softens at 210-215°, solidifies and turns brown (Christensen). Also reported as crystals from ethanol, mp 220° (softens), resolidifies, turns brown and does not melt below 300° (Kazimierczuk). [a]D25 -18.8° (c = 1 in DMF). uv max in 0.1N NaOH: 265 nm; in 0.1N HCl: 265 nm.
Melting point: mp 220° (softens), resolidifies, turns brown and does not melt below 300°
Optical Rotation: [a]D25 -18.8° (c = 1 in DMF)
Absorption maximum: uv max in 0.1N NaOH: 265 nm; in 0.1N HCl: 265 nm
Therap-Cat: Antineoplastic.
Keywords: Antineoplastic; Antimetabolites; Purine Analogs.
////////////fda 2019, Mavenclad, cladribine, multiple sclerosis, EMD Serono, クラドリビン , Leustatin, クラドリビン , orphan drug designation
NC1=C2N=CN([C@H]3C[C@H](O)[C@@H](CO)O3)C2=NC(Cl)=N1

RISDIPLAM , リスジプラム

$
0
0

Risdiplam.svg

Image result for RISDIPLAM

RISDIPLAM

RG-7916, RO-7034067, リスジプラム

Formula
C22H23N7O
Cas
1825352-65-5
Mol weight
401.4643
US9969754

7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[1,2-b]pyridazin-6-yl)pyrido[1,2-a]pyrimidin-4-one

WHO 10614

RG-7916

HY-109101

RO7034067

CS-0039501

EX-A2074

RG7916

The compound was originally claimed in WO2015173181 , for treating spinal muscular atrophy (SMA). Roche , under license from PTC Therapeutics , and Chugai , are developing risdiplam (RO-7034067; RG-7916), a small-molecule survival motor neuron (SMN)2 gene splicing modulator and a lead from an SMN2 gene modulator program initiated by PTC Therapeutics in collaboration with the SMA Foundation , for the oral treatment of spinal muscular atrophy

The product was granted orphan drug designation in the U.S., E.U. and in Japan for the treatment of spinal muscular atrophy. In 2018, it also received PRIME designation in the E.U. for the same indication.

Risdiplam (RG7916RO7034067) is a highly potent, selective and orally active small molecule experimental drug being developed by F. Hoffmann-La RochePTC Therapeutics and SMA Foundation to treat spinal muscular atrophy (SMA). It is a pyridazine derivative that works by increasing the amount of functional survival of motor neuron protein produced by the SMN2 gene through modifying its splicing pattern.[1][2]

As of September 2018, risdiplam is undergoing late-stage clinical trials across the spectrum of spinal muscular atrophy[3][4][5] where it has shown promising preliminary results.[6][7]

PATENT

WO2015173181

https://patentscope.wipo.int/search/en/detail.jsf;jsessionid=B8D897794EC02E2BBFD5D2280B3E1883.wapp1nC?docId=WO2015173181&recNum=9&office=&queryString=&prevFilter=%26fq%3DOF%3AKR%26fq%3DICF_M%3A%22C07D%22%26fq%3DPAF_M%3A%22F.+HOFFMANN-LA+ROCHE+AG%22&sortOption=Pub+Date+Desc&maxRec=912

Example 20

7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6- yl)pyrido[l,2-a]pyrimidin-4-one

In a sealed tube, 2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-7-fluoro-pyrido[l,2-a]pyrimidin-4-one (Intermediate 2; 50 mg, 0.162 mmol), DIPEA (0.22 mL, 1.29 mmol, 4 eq.) and 4,7-diazaspiro[2.5]octane dihydrochloride (32 mg, 0.320 mmol, 3.0 eq.) were stirred in

DMSO (2 mL) at 130°C for 48 hours. The solvent was removed under high vacuum. The residue was taken up in CH2CI2 and washed with an aqueous saturated solution of NaHC03. The organic layer was separated and dried over Na2S04 and concentrated in vacuo. The crude was purified by column chromatography (Si02, CH2Cl2/MeOH=98/2 to 95/5) to afford the title product (12 mg, 18%) as a light yellow solid. MS m/z 402.3 [M+H+].

PATENT

WO-2019057740

Process for the preparation of risdiplam and its derivatives.

Scheme 1:

Scheme 3:

Scheme 4:

xample 1: tert-Butyl 7-(6-chloro-3-pyridyl)-4,7-diazaspiro[2.5]octane-4-carboxylate

5-Bromo-2-chloropyridine (85.0 g, 442 mmol), tert-butyl 4,7-diazaspiro[2.5]octane-4-carboxylate (102 g, 442 mmol) and Me-THF (722 g) were charged into a reaction vessel. After 10 minutes stirring, most of the solids were dissolved and [Pd(Xantphos)Cl2] (3.34 g) was added followed after 5 minutes by a solution of sodium tert-butanolate (56.3 g, 574 mmol) in Me-THF (173 g). The reaction mixture was stirred at 70 °C for 1.25 hours, cooled to room temperature and water (595 g) and 1-propylacetate (378 g) were added. After vigorous stirring, the phases were separated, the organic phase was washed with a second portion of water (425 g) and with a mixture of water (425 g) and brine (25 mL). The organic phase was treated with active charcoal (6.8 g), filtered and concentrated under reduced pressure to afford a brown oil, which was dissolved in tert-amyl-methyl-ether (347 g) at reflux. The solution was cooled slowly to room temperature. After stirring 18 hours at room temperature, n-heptane (205 g) was added and the suspension was further cooled to -10 °C. The precipitate was filtered off and dried under high vacuum to afford tert-butyl 7-(6-chloro-3-pyridyl)-4,7-diazaspiro[2.5]octane-4-carboxylate (110.9 g, 77.5%) as a beige solid.

Ή-ΝΜΡν (CDC13, 600 MHz): 7.95 (d, 1H); 7.18 – 7.14 (m, 1H); 7.13 – 7.09 (m, 1H); 3.79 – 3.63 (m, 2H); 3.24 – 3.12 (m, 2H); 2.96 (s, 2H); 1.47 (s, 9H); 1.11 – 1.04 (m, 2H); 0.90 -0.79 (m, 2H); LCMS: 324.15, 326.15 (M+H+)

Example 2: tert-butyl 7-(6-amino-3-pyridyl)-4,7-diazaspiro[2.5]octane-4-carboxylate

An autoclave equipped with an ascending pipe was filled with ammonia (78.7 g, 15 eq; 10 eq are sufficient) at -70 °C. Another autoclave was charged with tert-butyl 7-(6-chloro-3-pyridyl)-4,7-diazaspiro[2.5]octane-4-carboxylate (100 g, 309 mmol), sodium tert-butanolate (32.6 g, 340 mmol) and dioxane (800 mL). After 10 minutes stirring at room temperature under Ar, a solution of Pd2(dba)3 (1.41 g, 1.54 mmol) and tBuBrettPhos (1.50 g, 3.09 mmol) in dioxane (180 mL) was added. Thereafter, the connected ammonia vessel was warmed with a warm water bath and the connecting valve was opened. The autoclave was warmed to 30 °C and the reaction mixture stirred 5 hours at this temperature. The ammonia vessel was closed and disconnected. The excess ammonia was washed out of the autoclave with Argon. The reaction solution was poured into a separating funnel, the autoclave washed with ethyl acetate (300 mL) and water (100 mL) and these two solvent portions were added to the separating funnel. The biphasic mixture was further diluted with ethyl acetate (900 mL) and water (1000 mL). After vigorous stirring, the phases were separated. The organic phase was washed with a mixture of water (500 mL) and brine (10 mL). The combined aqueous phases were extracted twice with ethyl acetate (500 mL). The combined organic phases were treated with active charcoal (3.70 g, 309 mmol), filtered and the filtrate was concentrated under reduced pressure to afford a thick brown oil. This oil was dissolved in 1 -propyl acetate (160 mL) at 45-50°C and n-heptane (940 mL) was added drop wise within 1.5 hours. The suspension was cooled slowly to -5°C, stirred 4 hours at -5 °C and filtered. The precipitate was washed with cold n-heptane and dried under high vacuum at 50°C to afford tert-butyl 7-(6-amino-3-pyridyl)-4,7-diazaspiro[2.5]octane-4-carboxylate (81.4 g, 86.5%) as a beige solid.

Ή-ΝΜΡν (CDCb, 600 MHz): 7.71 (d, 1H); 7.12 (dd, 1H); 6.47 (d, 1H); 4.18 (br s, 2H); 3.74 – 3.58 (m, 2H); 3.09 – 2.94 (m, 2H); 2.81 (s, 2H); 1.52 – 1.39 (m, 9H); 1.17 – 0.98 (m, 2H); 0.92 – 0.75 (m, 2H); LCMS: 305.20 (M+H+)

Example 3: tert-butyl 7-(6-amino-3-pyridyl)-4,7-diazaspiro[2.5]octane-4-carboxylate

An autoclave was charged with tert-butyl 7-(6-chloro-3-pyridyl)-4,7-diazaspiro[2.5]octane-4-carboxylate (339 mg, 1 mmol), sodium tert-butanolate (109 mg, 1.1 mmol) and dioxane (5 mL). After 5 minutes stirring at room temperature under Argon [Pd(allyl)(tBuBrettPhos)]OTf (4 mg, 5 μιηοΐ) was added. Thereafter, the autoclave was closed and connected to an ammonia tank, the valve was open and ammonia (230 mg, 13.5 mmol) was introduced into the autoclave. The valve was closed and the autoclave disconnected. The autoclave was warmed to 30 °C and the reaction mixture stirred 4 hours at this temperature. Then the autoclave was opened and the excess ammonia was washed out of the autoclave with Argon. The reaction solution was poured into a flask and taken to dryness under reduced pressure. The residue was purified by chromatography over silica gel (eluent: dichloromethane/ethyl acetate to dichloromethane/methanol). After evaporation of the solvents tert-butyl 7-(6-amino-3-pyridyl)-4,7-diazaspiro[2.5]octane-4-carboxylate (283 mg, 93%) was isolated as a brown oil containing 4% dichloromethane and 3% ethyl acetate.

Example 4: tert-butyl 7-(6-nitro-3-pyridyl)-4,7-diazaspiro[2.5]octane-4-carboxylate

tert-Butyl 4,7-diazaspiro[2.5]octane-4-carboxylate oxalate salt (2.46 kg, 8.13 mol), 5-bromo-2-nitro-pyridine (1.50 kg, 7.39 mol) and dimethyl sulfoxide (7.80 L) were char; into a reaction vessel pre-heated to 35 °C. With stirring, and keeping the temperature below 40°C, lithium chloride (1.25 kg, 25.6 mol) was added portion- wise followed by tetramethylguanidine (2.98 kg, 25.9 mol). Dimethyl sulfoxide (450 mL) was used to rinse the feed line. The reaction mixture was stirred at 79 °C for 8 hours, cooled to 70°C and water (2.48 L) was added within 2 hours. After stirring at 70 °C for an additional 1 hour, the precipitate was filtered off and washed with water (4.5 L) three times. The precipitate was dissolved in ethyl acetate (15 L) and water (7.5 L) at reflux temperature. The phases were separated at 60°C and n-heptane (7.5 L) was added to the organic layer at 60°C within 30 minutes. The solution was cooled to 0°C in 2 hours and further stirred at 0°C for 1 hour. The precipitate was filtered off, washed with a mixture of ethyl acetate (750 mL)/n-heptane (375 mL) twice and dried under reduced pressure to afford 1.89 kg (76.4%) of tert-butyl 7-(6-nitro-3-pyridyl)-4,7-diazaspiro[2.5]octane-4-carboxylate as a yellow to light brown solid.

!H-NMR (CDCls, 600 MHz): 8.16 (d, 1H); 8.07 (d, 1H); 7.15 (dd, 1H); 3.80 – 3.72 (m, 2H); 3.49 – 3.41 (m, 2H); 3.23 (s, 2H); 1.48 (s, 9H); 1.16 – 1.08 (m, 2H); 0.92 – 0.85 (m, 2H); LCMS: 335.17 (M+H+)

Example 5: tert-butyl 7-(2-hydroxy-4-oxo-pyrido[l,2-a]pyrimidin-7-yl)-4,7-diazaspiro[2.5]octane-4-carboxylate

tert-Butyl 7-(6-amino-3-pyridyl)-4,7-diazaspiro[2.5]octane-4-carboxylate (80.0 g, 263 mmol) was dissolved in anisole (800 mL) and di-tert-butyl malonate (71.1 g, 315 mmol) was added. The solution was stirred 3.5 hours at 145 °C then cooled to room temperature. The precipitate was filtered off, washed with toluene (in portions, 320 mL in total) and dried under high vacuum at 50°C to afford tert-butyl 7-(2-hydroxy-4-oxo-pyrido[l,2-a]pyrimidin-7-yl)-4,7-diazaspiro[2.5]octane-4-carboxylate (65.6 g, 67%) as a light pink powder.

Ή-ΝΜΡν (CDCI3, 600 MHz): 8.46 (d, 1H); 7.74 (dd, 1H); 7.52 (d, 1H); 5.37 (s, 2H); 3.83 – 3.69 (m, 2H); 3.23 (t, 2H); 3.01 (s, 2H); 1.48 (s, 9H); 1.17 – 1.03 (m, 2H); 0.95 – 0.75 (m, 2H); LCMS: 373.19 (M+H+)

Example 6: tert-butyl 7-(2-hydroxy-4-oxo-pyrido[l,2-a]pyrimidin-7-yl)-4,7-diazaspiro[2.5]octane-4-carboxylate

tert-Butyl 7-(6-nitro-3-pyridyl)-4,7-diazaspiro[2.5]octane-4-carboxylate (950 g, 2.84 mol), Pt 1%, V 2% on active charcoal (95.1 g, 2 mmol) and ethyl acetate (9.5 L) were charged into an autoclave that was pressurized with hydrogen gas to 3 bar. The reaction mixture was stirred at room temperature for 6 hours. The excess hydrogen was vented. The reaction mixture was filtered, the catalyst was washed with ethyl acetate (0.95 L) three times. The filtrate was concentrated under reduced pressure and the solvent exchanged to anisole (add two portions of 2.85 L and 5.18 L) by distillation. Di tert-butyl malonate (921.7 g, 4.26 mol) was added and the charging line was rinsed with anisole (618 mL) and the reaction mixture was stirred at 125-135 °C for 8 hours. It may be necessary to distill off the by-product tert-butanol to reach this temperature. The progress of the reaction was followed eg.by HPLC. If the reaction stalls, the temperature is increased to 135-145°C and checked for progress after 1 hour. When the reaction was complete, the batch was cooled to room temperature and stirred at room temperature for 4 hours. The precipitate was filtered off, washed with toluene (3.55 L) and dried under vacuum at 60°C to afford tert-butyl 7-(2-hydroxy-4-oxo-pyrido[l,2-a]pyrimidin-7-yl)-4,7-diazaspiro[2.5]octane-4-carboxylate (861.0 g, 81.4%) as a yellow to light brown solid.

Example 7: tert-butyl 7-[4-oxo-2-(p-tolylsulfonyloxy)pyrido[l,2-a]pyrimidin-7-yl]-4,7-diazaspiro[2.5]octane-4-carboxylate

A reactor was charged with tert-butyl 7-(2-hydroxy-4-oxo-pyrido[l,2-a]pyrimidin-7-yl)-4,7-diazaspiro[2.5]octane-4-carboxylate (920 g, 2.47 mol) and then triethylamine (325 g, 3.21 mol), followed by tosyl chloride (527.1 g, 2.77 mol) and dichloromethane (4.6 L). The reaction mixture was stirred at 20-25 °C for at least three hours. Upon complete reaction, the organic solution was washed with a prepared solution of HC1 (32%, 247.8 mL) and water (4.6 L), followed by a prepared solution of sodium hydroxide (432.3 mL of a 30% stock solution) and water (3.9 L) in that order. The organic phase was finally washed with water (4.8 L) and then dichloromethane was nearly completely distilled off under reduced pressure at 50-55°C. Ethyl acetate (920 mL) was added and distilled twice at this temperature under reduced pressure, and then ethyl acetate (4.8 L) was added and the suspension cooled to 20-25 °C over two hours. n-Heptane (944.4 mL) was added and the mixture was cooled to 0-5 °C and then stirred for an additional 3 hours. The precipitate was filtered off, washed with a prepared solution of ethyl acetate (772.8 mL) and n-heptane (147.2 mL), and then twice with n-heptane (2.6 L). The solid was dried under vacuum at 45-50°C to afford 1122.6 g (86.3%) tert-butyl 7-[4-oxo-2-(p-tolylsulfonyloxy)pyrido[l,2-a]pyrimidin-7-yl]-4,7-diazaspiro[2.5]octane-4-carboxylate as yellow crystals.

!H-NMR (CDCls, 600 MHz): 8.32 (d, 1H); 8.00 – 7.89 (m, 2H); 7.66 (dd, 1H); 7.50 (d, 1H); 7.36 (d, 2H); 6.04 (s, 1H); 3.80 – 3.68 (m, 2H); 3.23 (t, 2H); 3.01 (s, 2H); 1.48 (s, 9H); 1.15 – 1.04 (m, 2H); 0.92 – 0.82 (m, 2H); LCMS: 527.20 (M+H+)

Example 8: 2,8-dimethyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)imidazo[l,2-b]pyridazine

6-Chloro-2,8-dimethylimidazo[l,2-b]pyridazine (40.0 g, 220 mmol), bis pinacol diborane (69.9 g, 275 mmol) and potassium acetate (43.2 g, 440 mmol) were suspended in acetonitrile (440 mL). The suspension was heated to reflux and stirred 30 minutes at reflux, then a suspension of PdCl2(dppf) (4.03 g, 5.51 mmol) and dppf (610 mg, 1.1 mmol) in acetonitrile (40 mL) was added. The vessel was rinsed with acetonitrile (20 mL), which were also poured into the reaction mixture. The orange suspension was further stirred at reflux, whereby acetonitrile (50 mL) were distilled off. After 4 hours, the reaction mixture was filtered off, the filter was washed with several portions of acetonitrile (in total 150 mL). The filtrate was diluted to obtain a volume of 700 mL. The 314 mmolar solution of 2,8-dimethyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)imidazo[l,2-b]pyridazine in acetonitrile was used as such in the next step.

Example 9: 2,8-dimethyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)imidazo[l,2-b]pyridazine

6-chloro-2,8-dimethylimidazo[l,2-b]pyridazine (29.0 g, 22.8 mmol), bis pinacol diborane (44.6, 25.1 mmol) and potassium acetate (31.3 g, 45.6 mmol) were suspended in 1-propyl acetate (365 mL). The suspension was heated to 80°C and a solution of

tricyclohexylphosphine (448 mg, 0.23 mmol) and Pd(OAc)2 (179 mg, 0.11 mmol) in 1-propyl acetate (37 mL) was added within 20 minutes. After 2.5 hours further stirring at 80°C, the suspension was cooled to 40°C and filtered at this temperature. The precipitate was washed with 1-propyl acetate (200 mL). The filtrate corresponds to 516.4 g of a 8.5% solution of 2,8-dimethyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)imidazo[l,2-b]pyridazine in 1 -propyl acetate.

Example 10: Isolation of 2,8-dimethyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)imidazo[ 1 ,2-b]pyridazine

In another experiment, the above solution obtained was cooled to 0-5 °C within 3 hours. The precipitate was filtered off, washed with cold 1 -propyl acetate and dried under high vacuum at 60°C to afford 2,8-dimethyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)imidazo[l,2-b]pyridazine (24. Og, 55%) as a colourless solid.

lH NMR (CDCls, 600 MHz, ) δ ppm 7.86 (d, J=0.7 Hz, 1 H), 7.20 (d, J=1.0 Hz, 1 H), 2.63 (d, J=1.0 Hz, 3 H), 2.51 (d, J=0.7 Hz, 3 H), 1.33 – 1.49 (m, 12 H)

Example 11: (step 6) tert-butyl 7-[2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-4-oxo-pyrido[l,2-a]pyrimidin-7-yl]-4,7-diazaspiro[2.5]octane-4-carboxylate

tert-Butyl 7-[4-oxo-2-(p-tolylsulfonyloxy)pyrido[l,2-a]pyrimidin-7-yl]-4,7-diazaspiro[2.5] octane-4-carboxylate (25 g, 47.5 mmol), 2,8-dimethyl-6-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)imidazo[l,2-b]pyridazine (314 mM in acetonitrile, 191 mL, 59.8 mmol), PdCi2(dppf) (868 mg, 1.19 mmol) and aqueous potassium carbonate 4.07 M (17.1 mL, 69.8 mmol) were charged into a reaction vessel. The reaction mixture was stirred at reflux for 3 hours, cooled overnight to room temperature and filtered. The precipitate was washed with several portions of acetonitrile (146 mL in total), then suspended in methyl-THF (750 mL) and methanol (75 mL). Aqueous sodium hydrogen carbonate 5% (250 mL) was added, the mixture was vigorously stirred at 35°C. The phases were separated, the organic phase was washed again with aqueous sodium hydrogen carbonate 5% (250 mL). The organic phase was treated with active charcoal for 1 hour at room temperature, filtered and the filtrate was concentrated under reduced pressure at 60 °C to a volume of 225 mL, heated to reflux then cooled to room temperature, stirred at room temperature for 16 hours, then cooled to 0°C and stirred at 0°C for 3 hours. The precipitate was filtered off, washed with n-heptane (60 mL) and dried under high vacuum at 55°C to afford tert-butyl 7-[2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-4-oxo-pyrido[l,2-a]pyrimidin-7-yl]-4,7-diazaspiro[2.5]octane-4-carboxylate (20.13 g, 84.5%) as a yellow solid.

This solid could be recrystallized in the following manner: 15 g of the above solid was dissolved at reflux in toluene (135 mL) and ethanol (15 mL). The solution was slowly cooled to room temperature, stirred 16 hours at room temperature, then cooled to 0°C and stirred at 0°C for 4 hours. The precipitate was filtered off, washed with cold toluene and dried under high vacuum at 55°C to afford tert-butyl 7-[2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-4-oxo-pyrido[l,2-a]pyrimidin-7-yl]-4,7-diazaspiro[2.5]octane-4-carboxylate (11.92 g, 79.5%) as a yellow-green solid.

!H-NMR (CDCls, 600 MHz): 8.44 (d, 1H); 7.93 (d, 1H); 7.96 – 7.89 (m, 1H); 7.80 (d, 1H); 7.76 – 7.72 (m, 1H); 7.70 – 7.63 (m, 1H); 7.38 (s, 1H); 3.85 – 3.69 (m, 2H); 3.28 (t, 2H); 3.07 (s, 2H); 2.74 (d, 3H); 2.55 (s, 3H); 1.49 (s, 9H); 1.16 – 1.09 (m, 2H); 0.93 – 0.86 (m, 2H); LCMS: 502.26 (M+H+)

Example 12: tert-butyl 7-[2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-4-oxo-pyrido[l,2-a]pyrimidin-7-yl]-4,7-diazaspiro[2.5]octane-4-carboxylate

6-chloro-2,8-dimethylimidazo[l,2-b]pyridazine (4.14 g, 22.8 mmol), bis pinacol diborane (6.37g, 25.1 mmol) and potassium acetate (4.47 g, 45.6 mmol) were suspended in 1-propyl acetate (59 mL). The suspension was heated to 80°C and a solution of

tricyclohexylphosphine (63.9 mg, 0.23 mmol) and Pd(OAc)2 (25.6 mg, 0.11 mmol) in 1-propyl acetate (6 mL) was added within 20 minutes. After 2.5 hours further stirring at 80°C, the suspension was cooled to 40°C and filtered at this temperature. The precipitate was washed with 1-propyl acetate (32 mL). The filtrate corresponds to 74.6 g of a 8.5% solution of 2,8-dimethyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)imidazo[l,2-b]pyridazine in 1-propyl acetate.

A reaction vessel was charged with tert-butyl 7-[4-oxo-2-(p-tolylsulfonyloxy)pyrido[l,2-a]pyrimidin-7-yl]-4,7-diazaspiro[2.5]octane-4-carboxylate (10.0 g, 19.0 mmol), tricyclohexylphosphine (58.6 mg, 0.21 mmol) and Pd(OAc)2 (21.3 mg, 0.10 mmol) and 1-propyl acetate (42 mL) and a solution of potassium carbonate (5.25 g, 38.0 mmol) in water (19.0 mL) was added. The suspension was heated to 70°C and the solution of 2,8-dimethyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)imidazo[l,2-b]pyridazine in 1-propyl acetate was added within 30 minutes. The mixture was stirred for 2 hours at 70-75°C. The suspension was cooled to 40°C, water (10 mL) was added. The suspension was aged for 30 minutes. The crude product was filtered off and rinsed with 1-propyl acetate (41 mL). The crude product was taken up in toluene (100 mL), 5% aqueous NaHC03-solution (30 mL) and 1-propanol (20.0 mL). The mixture was heated to 60-65 °C, the phases were separated and the organic phase was washed with 2 more portions of water (30.0 mL). The organic phase was filtered on active charcoal, the filter washed with toluene (60.0 mL). The filtrate was concentrated under reduced pressure to a volume of ca. 120 mL, heated to reflux and 1-propanol (0.8 mL) was added to obtain a solution. The solution was cooled to 0-5°C within 4-6 hours, stirred at 0-5°C for 1 hour. The precipitate was filtered off, washed with toluene (30 mL) and dried under reduced pressure at 70-80°C to afford tert-butyl 7-[2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-4-oxo-pyrido[l,2-a]pyrimidin-7-yl]-4,7-diazaspiro[2.5]octane-4-carboxylate (7.7 g, 80.8%) as a yellowish solid.

Example 13: 7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one di-hydrochloride salt

To prepare a solution of HC1 in in 1-propyl acetate/ 1-propanol, acetyl chloride (15.8 g, 199 mmol) was slowly added to a mixture of 1-propyl acetate (60 mL) and 1-propanol (30 mL) at 0°C, and stirring was pursued for an additional 2 hours at room temperature.

tert-Butyl 7-[2-(2,8-dimethylimidazo[ 1 ,2-b]pyridazin-6-yl)-4-oxo-pyrido[ 1 ,2-a]pyrimidin-7-yl]-4,7-diazaspiro[2.5]octane-4-carboxylate (20 g, 39.9 mmol) was suspended in 1-propyl acetate (60 mL) and 1-propanol (30 mL) at room temperature and the HC1 solution in 1-propyl acetate and 1-propanol was added. The reaction mixture was heated within 3 hours to 70°C and stirred 16 hours at this temperature, then cooled to 20°C. The precipitate was filtered off, washed with 1-propyl acetate (50 mL) in several portions and dried under vacuum at 55 °C to afford 7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one hydrochloride salt (18.8 g, 99%) as yellow crystals.

^-NMR (CDCls, 600 MHz): 8.34 (s, 1H); 8.22(s, 1H); 8.05 (s, 1H); 8.01 (dd, 1H); 7.80 (d, 1H); 7.16 (s, 1H); 3.71 – 3.67 (m, 2H); 3.64 – 3.59 (m, 2H); 3.52 (s, 2H); 2.69 (s, 3H); 2.54 (s, 3H); 1.23- 1.20 (m, 2H); 1.14 – 1.08 (m, 2H); LCMS: 402.20 (M+H+)

Example 14: 7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one

To a suspension of tert-butyl 7-[2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-4-oxo-pyrido[l,2-a]pyrimidin-7-yl]-4,7-diazaspiro[2.5]octane-4-carboxylate (25 g, 50 mmol) in 1-propyl acetate (375 mL) was added a solution of HC1 in 1-propanol (prepared by adding slowly at 5°C acetyl chloride (18.0 mL) to 1-propanol (37.6 mL) and stirring 1 hour at room temperature). The stirred suspension was heated to 75°C within 10 hours and stirred a further 5 hours at 75 °C. Water (160.0 mL) was added and the phases were separated at 75°C. Aqueous sodium hydroxide 32% (27.8 mL) was added to the aqueous phase. The suspension obtained was cooled to room temperature within 5 hours and stirred one hour at room temperature. The precipitate was filtered off, washed with water (100.0 mL) and dried under reduced pressure at 50°C for 18 hours to afford 7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one (19.7 g, 98.3%) as yellow crystals.

!H-NMR (CDCb, 600 MHz): 8. 45 (d, 1H); 7.92 (d, 1H); 7.80 (s, 1H); 7.75 – 7.71 (m, 1H); 7.71 – 7.67 (m, 1H); 7.37 (s, 1H); 3.31 – 3.24 (m, 2H); 3.22 – 3.16 (m, 2H); 3.09 (s, 2H); 2.73 (s, 3H); 2.55 (s, 3H); 0.82- 0.76 (m, 2H); 0.71 – 0.63 (m, 2H); LCMS: 402.20

(M+H+)

Example 15: 7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)pyrido[ 1 ,2-a]pyrimidin-4-one

A suspension of tert-butyl 7-[2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)-4-oxo-pyrido[l,2-a]pyrimidin-7-yl]-4,7-diazaspiro[2.5]octane-4-carboxylate (13.5 g, 26.9

in toluene (237.0 g) was stirred at 75°C and a 21.9% solution of HCl in 1-propanol (21.4 g, 134.5 mmol) was added within 2.5 hours. The reaction mixture was stirred further at 75 °C until complete conversion. The reaction mixture was cooled to 20-25°C. Water (70 g) was added. The biphasic mixture was stirred another 10 minutes at 20-25 °C and the phases were separated. The organic phase was extracted with water (17 g) twice and the combined aqueous phases were added into mixture of aqueous sodium hydroxide 28% (15.0 g) and water (45.0 g). The suspension obtained was cooled to 20°C. The precipitate was filtered off , washed with water (25 g) three times and dried under reduced pressure at 60°C to afford 7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[l,2-b]pyridazin-6-yl)pyrido[l,2-a]pyrimidin-4-one (9.5 g, 95.1%) as yellow crystals.

Example 16: 4-bromo-6-chloro-pyridazin-3-amine

3-amino-6-chloropyridazine (20 g, 154 mmol), sodium bicarbonate (25.9 g, 309 mmol) and methanol (158 g) were charged in a reaction vessel and cooled to 0-10°C. Bromine (34.5 g, 216 mmol) was added drop wise and the reaction mixture was stirred 3 days at room temperature. 10% Aqueous sodium sulfate was added. The suspension was filtered off. The filtrate was washed with ethyl acetate (300 mL) twice. The combined organic layers were dried and evaporated. A suspension of the residue in methanol (50 mL) was heated to reflux, water (120 mL) was added and the suspension was stirred 16 hours at room temperature. The precipitate was filtered off and dried. The residue was suspended in n-heptane (50 mL), stirred 2 hours at room temperature, filtered off and dried to afford 4-bromo-6-chloro-pyridazin-3-amine (14.5 g, 46.2%) as a light brown solid.

!H-NMR (CDCls, 600 MHz): 7.55 (s, 1H); 5.83-4.89 (m, 2H); LCMS: 209.93 (M+H+)

Example 17: 4-bromo-6-chloro-pyridazin-3-amine

3-amino-6-chloropyridazine (50 g, 360 mmol), acetic acid (5.8 g, 96.5 mmol), sodium acetate (28.7 g, 289.5 mmol) and methanol (395 g) were charged in a reaction vessel and heated to 25-35°C. Dibromodimethylhydatoin (66.0 g, 231.6 mmol) was added in several portions and the reaction mixture was stirred 3 hours at 30°C. Completion is checked by IPC and if the conversion is incomplete, dibromodimethylhydantoin is added (5.5g). At reaction completion, 38% aqueous sodium sulfate (77.2 mmol NaHS03) was added slowly. The suspension was concentrated under reduced pressure and water (500 g) was added slowly at 45°C, then 30% aqueous sodium hydroxide (31.5 g, 231.6 mmol NaOH) was added at 20°C to adjust pH to 7-8. The precipitate was filtered off, washed with water and dried under reduced pressure to afford 4-bromo-6-chloro-pyridazin-3-amine (50.2 g, 62.5%) as a grey solid.

Example 18: 6-chloro-4-methyl-pyridazin-3-amine

4-bromo-6-chloro-pyridazin-3-amine (3.0 g, 14.4 mmol) and

tetrakis(triphenylphosphine)palladium (1666 mg, 144 μιηοΐ) were suspended in THF (13.2 g) and a solution of zinc chloride in Me-THF (2.0 M, 9 mL, 18 mmol) was added. The reaction mixture was cooled to -5°C and methyllithium in diethoxymethane (3.1 M, 11.6 mL, 36 mmol) was added. The reaction mixture was stirred at 45°C for 4 hours. Sodium sulfate decahydrate (11.7 g, 36 mmol) was added at room temperature, the mixture was stirred 1.5 hours at 60°C, diluted with water (100 mL) and after 30 minutes the precipitate was filtered off. The precipitate was dissolved in aqueous HC1 2M (100 mL) and ethyl acetate (140 mL). The biphasic system was filtered, the phases were separated and the pH of the water layer adjusted to 7 with aqueous NaOH 32% (18 mL). The precipitate was filtered and dried. The solid obtained was digested twice in methanol (20 mL) at room temperature. The two filtrates were combined, evaporated and dried under high vacuum to afford 6-chloro-4-methyl-pyridazin-3-amine (1.2 g, 58.1%) as a red solid.

Ή-ΝΜΡν (CDCb, 600 MHz): 7.09 (d, 1H); 4.90 (br s, 2H), 2.17 (d, 3H)

Example 19: 6-chloro-4-methyl-pyridazin-3-amine

4-bromo-6-chloro-pyridazin-3-amine (30.02 g, 143 mmol) and THF (180 mL) were charged into a reaction vessel. Methylmagnesium chloride (22% in THF, 50.0 mL, 1.03 eq.) was added at 20°C over 60 minutes, followed by zinc chloride in Me-THF (25%, 37 mL, 0.50 eq.) and palladium tetrakis(triphenyphosphine) (1.66 g, lmol%). The reaction mixture was heated to 50°C and methylmagnesium chloride (22% in THF, 81 mL, 1.7 eq.) was added slowly. The reaction mixture was stirred at 50°C until complete conversion, then at 10°C for 14.5 hours and poured into a mixture of water (90 g), aqueous HCl 33% (52.5 g) and toluene (150 mL) maintained at 20-30°C. The aqueous phase was separated and the organic phase was extracted with a solution of aqueous HCl 33% (2.0 g) and water (45 g). The aqueous layers were combined and washed with toluene (30 mL) twice and the pH was adjusted by addition of 25% aqueous ammonia solution. When a pH of 2.4 was reached, seeding crystals were added, the mixture was stirred further for 15 minutes and thereafter the pH was brought to 4.0. The suspension was stirred at 20°C for 2 hours, the precipitate was filtered off, washed with water (20 mL) three times to afford crude 6-chloro-4-methyl-pyridazin-3-amine (29 g) as a brown solid.

29 g crude product was transferred to a reaction vessel and methanol (20 mL) was added. The mixture was refluxed for 30 minutes and 12 g water was added. The solution was cooled to 0°C and stirred for 2 hours at this temperature. The precipitate was filtered off, washed with water three times and dried under reduced pressure at 40°C to afford purified 6-chloro-4-methyl-pyridazin-3-amine (13.8 g, 66%) as a light brown solid.

Alternative purification:

50 g crude 6-chloro-4-methyl-pyridazin-3-amine were dissolved in methanol (250 mL) and active charcoal (4.0 g) and diatomaceous earth (2.5 g) were added. The suspension was stirred at 45°C for 1 hour, cooled to 30°C and potassium hydrogenophosphate (2.1 g) was added. The suspension was stirred at 30°C for another 90 minutes, filtered and the precipitate washed with methanol (100 mL). The filtrate was concentrated to a residual volume of 175 mL and water (120 mL) was added. The resulting suspension was heated

to reflux affording a solution which was cooled to 20°C resulting in a suspension. The precipitate was filtered off, washed with water (90 mL) and dried under reduced pressure to afford pure 6-chloro-4-methyl-pyridazin-3-amine (38 g, 76%) as a light yellow solid.

Example 20: 6-chloro-2,8-dimethyl-imidazo[l,2-b]pyridazine

6-chloro-4-methyl-pyridazin-3-amine (70.95 kg, 494.2 mol), sodium bromide (35 kg, 345.9 mol), isopropyl acetate (611 kg), isopropanol (28 kg and water (35 kg) were charged into a reaction vessel. The reaction mixture was stirred at 80-85 °C for 8 hours. Isopropyl acetate (310 kg) and water (420 kg) were added. 30% Aqueous NaOH was added at 45-55 °C and the system was stirred for 2 hours. The phases were separated at 25-35 °C. The organic layer was washed with water (370 kg), filtered on diatomite (7 kg) and the filter washed with isopropyl acetate (35 kg). The organic phase was extracted with two portions of 5.4% aqueous sulfuric acid (910 kg followed by 579 kg). The combined aqueous phases were basified with 30% aqueous NaOH (158 kg). The suspension was stirred 2 hours at 15-25 °C. The precipitate was isolated by centrifugation in three portions, each washed with water (31 kg). The wet solid was dissolved in isopropyl acetate (980 kg) at 25-35 °C, the solution washed with water (210 kg), three times. The organic phase was treated with active charcoal for 12 hours at 45-50 °C, concentrated to ca. 300 kg and heated to 70-80 °C to obtain a clear solution. This solution was cooled to 50-60 °C, stirred at this temperature for 1 hour, n-heptane (378 kg) was added and stirring was pursued for 1 hour. The mixture was cooled to -10- -5°C and stirred for another 3 hours. The precipitate was isolated by centrifuging, washed with n-heptane (33 kg) and dried under reduced pressure at 30-50 °C for 15 hours to afford 67.4 kg (76%) 6-chloro-2,8-dimethyl-imidazo[l,2-b]pyridazine as an off-white solid.

XH-NMR (CDCls, 600 MHz): 7.67 (s, 1H); 6.86 (s, 1H); 2.65 (s, 3H), 2.50 (s, 3H)

Paper

https://pubs.acs.org/doi/pdf/10.1021/acs.jmedchem.8b00741

Abstract Image

SMA is an inherited disease that leads to loss of motor function and ambulation and a reduced life expectancy. We have been working to develop orally administrated, systemically distributed small molecules to increase levels of functional SMN protein. Compound 2 was the first SMN2 splicing modifier tested in clinical trials in healthy volunteers and SMA patients. It was safe and well tolerated and increased SMN protein levels up to 2-fold in patients. Nevertheless, its development was stopped as a precautionary measure because retinal toxicity was observed in cynomolgus monkeys after chronic daily oral dosing (39 weeks) at exposures in excess of those investigated in patients. Herein, we describe the discovery of 1 (risdiplam, RG7916, RO7034067) that focused on thorough pharmacology, DMPK and safety characterization and optimization. This compound is undergoing pivotal clinical trials and is a promising medicine for the treatment of patients in all ages and stages with SMA.

 7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[1,2-b]pyridazin-6-yl)pyrido[1,2-a]pyrimidin-4-one 1 (12 mg, 18%) as a pale yellow solid. 1H NMR (600 MHz,CDCl3) δ ppm 8.45 (d, J = 2.4 Hz, 1H), 7.92 (d, J = 1.0 Hz, 1H), 7.73 (d, J = 9.6 Hz, 1H) 7.80 (s, 1H), 7.70 (dd, J = 9.7, 2.5 Hz, 1H), 7.38 (s, 1H), 3.31–3.22 (m, 2H), 3.20–3.16 (m, 2H), 3.08 (s, 2H), 2.74 (d, J = 0.9 Hz, 3H) 2.55 (s, 3H), 1.68 (br s, 1H), 0.77–0.75 (m, 2H), 0.67–0.64 (m, 2 H);

13C NMR (151 MHz,CDCl3) δ ppm 158.2, 156.3, 148.5, 147.2, 144.1, 142.2, 140.0, 135.6, 131.2, 126.7, 114.9, 114.7, 110.1, 99.3, 56.7, 49.9, 44.5, 36.5, 16.9, 15.0, 13.0. LC–HRMS: m/z = 402.2051 [(M + H)+ calcd for C22H24N7O, 402.2042; Diff 0.9 mDa].

References

  1. ^ Maria Joao Almeida (2016-09-08). “RG7916”. BioNews Services. Retrieved 2017-10-08.
  2. ^ Zhao, Xin; Feng, Zhihua; Ling, Karen K. Y; Mollin, Anna; Sheedy, Josephine; Yeh, Shirley; Petruska, Janet; Narasimhan, Jana; Dakka, Amal; Welch, Ellen M; Karp, Gary; Chen, Karen S; Metzger, Friedrich; Ratni, Hasane; Lotti, Francesco; Tisdale, Sarah; Naryshkin, Nikolai A; Pellizzoni, Livio; Paushkin, Sergey; Ko, Chien-Ping; Weetall, Marla (2016). “Pharmacokinetics, pharmacodynamics, and efficacy of a small-molecule SMN2 splicing modifier in mouse models of spinal muscular atrophy”Human Molecular Genetics25 (10): 1885. doi:10.1093/hmg/ddw062PMC 5062580PMID 26931466.
  3. ^ “Genentech/Roche Releases Clinical Trial Update for RG7916”. CureSMA. 2017-09-15. Retrieved 2017-10-08.
  4. ^ “A Study to Investigate the Safety, Tolerability, Pharmacokinetics, Pharmacodynamics and Efficacy of RO7034067 in Infants With Type1 Spinal Muscular Atrophy (Firefish)”.
  5. ^ “A Study to Investigate the Safety, Tolerability, Pharmacokinetics, Pharmacodynamics and Efficacy of RO7034067 in Type 2 and 3 Spinal Muscular Atrophy Participants (Sunfish)”.
  6. ^ “Updated Preliminary Data from SMA FIREFISH Program in Type 1 Babies Presented at the CureSMA Conference”http://www.prnewswire.com. Retrieved 2018-09-11.
Risdiplam
Risdiplam.svg
Clinical data
Synonyms RG7916; RO7034067
Identifiers
CAS Number
PubChem CID
UNII
KEGG
Chemical and physical data
Formula C22H23N7O
Molar mass 401.474 g/mol g·mol−1
3D model (JSmol)

///////////RISDIPLAM, RG-7916, RO-7034067, リスジプラム , PHASE 3, PRIME designation, ORPHAN DRUG

76RS4S2ET1 (UNII code)

CC1=CC(=NN2C1=NC(=C2)C)C3=CC(=O)N4C=C(C=CC4=N3)N5CCNC6(C5)CC6

CMX-8521, CMX-521

$
0
0

str1

PMQFVTNOZQVIOK-HTVVRFAVSA-N.png

CMX-8521, CMX-521

MF C13 H17 N5 O5,  MW 323.30

CAS Number 2077178-99-3

7H-Pyrrolo[2,3-d]pyrimidine-5-carboxamide, 4-amino-2-methyl-7-β-D-ribofuranosyl-

Nucleoside analogs (oral, norovirus infection), Chimerix

Image result for chimerix

4-amino-7-((2R,3R,4S,5R)-3,4-dihydroxy-5-(hydroxymethyl)tetrahydrofuran-2-yl)-2-methyl-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide

4-amino-7-[(2R,3R,4S,5R)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-2-methylpyrrolo[2,3-d]pyrimidine-5-carboxamide

CMX8521 is a nucleoside analog that inhibits the norovirus RNA polymerase. CMX8521 has in vitro activity against mouse and human norovirus.Where possible, Chimerix uses its lipid conjugate technology to build nucleoside-analog antivirals that are orally absorbed and have favorable tissue penetration.

CMX-8521 (presumed to be CMX-521) being developed by Chimerix for treating norovirus infection. In June 2018, a phase II efficacy trial was planned in 2019.

In January 2016, preclinical data were presented at the 34th Annual JP Morgan Healthcare Conference in San Francisco, CA. CMX-8521 had in vitro activity against mouse and human norovirus (EC50 = 2.1; CC50 = 114 microM). A 7-day non GLP toxicology/toxicokinetic study was completed in-life with no clinical or gross post mortem signs of toxicity. No off-target pharmacology was observed in vitro when screened against a panel of 87 receptors, transporters and enzymes associated with adverse pharmacology

PATENT

WO2017024310

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017024310

 Scheme 1: General Synthesis of Compounds of the Invention

Figure imgf000052_0001

Scheme 2: General Synthesis of Compounds of the Invention

Figure imgf000053_0001

Example 7– Synthesis of Compound 1

Figure imgf000149_0001

[00315] Step 1 (Protocol #1): To a 100-L jacketed reactor were charged 4-amino-6- bromo-2-methyl-7H-pyrrolo[2,3-d]pyrimidine-5-carbonitrile (3.00 kg), (3R,4R,5R)-2-acetoxy-5- ((benzoyloxy)methyl)tetrahydrofuran-3,4-diyl dibenzoate (6.60 kg) and DCE (18.89 kg). Stirring was started and DBU (3.61) kg was added. Over a period of 03 h and 14 min, TMSOTf (8.01 kg) was added between 30.6 °C and 37.3 °C. IPC after 01 h and 30 min at approx.32 °C showed 4% of 4-amino-6-bromo-2-methyl-7H-pyrrolo[2,3-d]pyrimidine-5-carbonitrile (3.00 kg),

(3R,4R,5R)-2-acetoxy-5-((benzoyloxy)methyl)tetrahydrofuran-3,4-diyl dibenzoate remaining. IPC after 03h and 16 min at approx.32 °C showed 2% 4-amino-6-bromo-2-methyl-7H- pyrrolo[2,3-d]pyrimidine-5-carbonitrile (3.00 kg), (3R,4R,5R)-2-acetoxy-5- ((benzoyloxy)methyl)tetrahydrofuran-3,4-diyl dibenzoate remaining (spec:≤3%). The reaction mixture was diluted with DCM (39.81 kg) and quenched with potable water (15.02 kg) over an 11 min period between 9.5 °C and 15.6 °C. The extractive work-up (at approx.22 °C) was completed by a back extraction of the aqueous phase with DCM (19.90 kg), a wash with sat NaHCO3 (1.3 kg NaHCO3 in 14.9 kg potable water), a back extraction of the bicarbonate phase with DCM (19.71 kg) and a wash with brine (4.5 kg NaCl in 14.9 kg potable water). Note: the reactor was cleaned with potable water, acetone and DCM after each wash/back extraction.

[00316] The drummed organic phase containing the product was charged to the 100-L jacketed reactor through an in-line filter followed by a DCM rinse of the drum and filter with DCM (2.48 kg). The contents of the reactor were distilled to 31 L with the aid of vacuum over a period of 06 h and 04 min with a maximum temperature of 50.1 °C. At this point a thick suspension had formed. Next, over a period of 39 min, IPAc (41.88 kg) was added between 44.5 °C and 49.5 °C and the contents of the reactor were heated to 76.9 °C over a period of 01 h and 25 min. Next, the contents of the reactor were cooled to 9.9 °C over a period of 04 h and 21 min and stirred for 12 h and 26 min with a minimum temperature of 1.6 °C.

[00317] Step 1 (Protocol # 2): To a 100-L jacketed reactor were charged 4-amino-6- bromo-2-methyl-7H-pyrrolo[2,3-d]pyrimidine-5-carbonitrile (3.00 kg), (3R,4R,5R)-2-acetoxy-5- ((benzoyloxy)methyl)tetrahydrofuran-3,4-diyl dibenzoate (6.60 kg) and DCE (18.80 kg). Stirring was started and DBU (3.59) kg was added. Over a period of 01 h and 46 min, TMSOTf (7.90 kg) was added between 30.4 °C and 34.2 °C. IPC after 02 h and 49 min at approx.34 °C showed 1% of 4-amino-6-bromo-2-methyl-7H-pyrrolo[2,3-d]pyrimidine-5-carbonitrile remaining (spec: ≤3%). The reaction mixture was diluted with DCM (40/70 kg) and quenched with potable water (14.97 kg) over an 04 min period between 9.9 °C and 18.0 °C. The extractive work-up (at approx.22 °C) was completed by a back extraction of the aqueous phase with DCM (20.34 kg), a wash with sat NaHCO3 (1.30 kg NaHCO3 in 14.90 kg potable water), a back extraction of the bicarbonate phase with DCM (20.65 kg) and a wash with brine (4.50 kg NaCl in 14.96 kg potable water). Note: the reactor was cleaned with potable water, acetone and DCM after each wash/back extraction.

[00318] The drummed organic phase containing the product was charged to the 100-L jacketed reactor through an in-line filter followed by a DCM rinse of the drum and filter with DCM (1.49 kg). The contents of the reactor were distilled to with the aid of vacuum over a period of 04 h and 49 min with a maximum temperature of 45.6 °C. At this point a thick suspension had formed. Next, over a period of 27 min, IPAc (41.70 kg) was added between 45.6 °C and 48.2 °C and the contents of the reactor were heated to 75.7 °C over a period of 01 h and 20 min. Next, the contents of the reactor were cooled to 9.4 °C over a period of 04 h and 15 min and stirred overnight with a minimum temperature of 2.3 °C.

[00319] Step 2: To the reactor were charged (2R,3R,4R,5R)-2-(4-amino-6-bromo-5- cyano-2-methyl-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-5-((benzoyloxy)methyl)tetrahydrofuran-3,4- diyl dibenzoate (10.0 kg), 10% Pd on C (Degussa, Type E101NE/W), trimethylamine (7.3 kg) and THF (44.5 kg). Hydrogen was submitted to the reactor and the mixture was stirred for 03 h and 54 min between 24.7 °C and 19.6 °C at approx.30.8 psig. IPC (HPLC) showed that

(2R,3R,4R,5R)-2-(4-amino-6-bromo-5-cyano-2-methyl-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-5- ((benzoyloxy)methyl)tetrahydrofuran-3,4-diyl dibenzoate could no longer be detected.

[00320] The reaction mixture was filtered over Celite (7.2 kg) and a polish filter and the filter residue was washed with THF (5.2 kg). The combined filtrate and wash was transferred to a 100-L jacketed reactor with the aid of a THF wash (2.12 kg). The contents of the reactor were vacuum distilled with a maximum batch temperature of 30.0 °C over a period of 05 h and 38 min to a final volume of 27 L. IPA (31.48 kg) was charged over a 40 min period to the reactor between 39.7 °C and 53.2 °C. The contents of the reactor were vacuum distilled with a maximum batch temperature of 53.2 °C over a period of 03 h and 02 min to a final volume of 33 L. IPA (48.99 kg) was charged over a 43 min period to the reactor between 53.1 °C and 57.1 °C. The contents of the reactor were heated to 60.2 °C, agitated for 12 min and cooled over a period of 04 and 28 min to 5.4 °C. Cold stirring was continued for a period of 08 h and 55 min with a minimum temperature of 1.1 °C. The slurry was filtered and washed with IPA (9.41 kg, at approx.4.5 °C). The residue was dried under vacuum with a nitrogen bleed for a period of 11 h and 44 min at a maximum temperature of 44.0 °C to provide an LOD of 0.36%. Yield: 6.58 kg (73.9 %).1H NMR confirms structure. Purity: 97.78 % (HPLC, AUC).

[00321] Step 3:

Figure imgf000152_0001

1100 g NaOH dissolved in potable water to a total volume of 1 L; 2 Diluted 500 mL conc. HCl in 2 L total with potable water [00322] A solution of (2R,3R,4R,5R)-2-(4-amino-5-cyano-2-methyl-7H-pyrrolo[2,3- d]pyrimidin-7-yl)-5-((benzoyloxy)methyl)tetrahydrofuran-3,4-diyl dibenzoate and THF was heated to 54 °C and the addition of 2.5 M NaOH was started. The initial addition gave a biphasic mixture and endothermic response (the temperature dropped to 50 °C) but as the addition continued a single phased, clear solution formed which was accompanied by a fast exotherm to 61 °C; the reaction temperature was maintained at 60 °C to 61 °C during the rest of the addition and for an additional 2 ½ h. IPC showed that no (2R,3R,4R,5R)-2-(4-amino-5-cyano-2-methyl- 7H-pyrrolo[2,3-d]pyrimidin-7-yl)-5-((benzoyloxy)methyl)tetrahydrofuran-3,4-diyl dibenzoate was left.

[00323] The reaction mixture was cooled to 21 °C and neutralized with 3 N HCl with external cooling to pH = 7.06 (Denver Instrument UB-10 pH meter equipped with a Sartorius P- P11 pH electrode, the electrode was checked with buffer solutions of pH = 4.00 and pH = 7.00); the mixture continued to cool to 8°C. The resulting neutralized mixture was distilled under vacuum with a pot temperature of 45 °C to 50 °C until the emergence of solids were observed in the pot. The suspension was cooled and stirred for 2 h at 2 °C. The beige suspension was filtered to afford a dark filtrate; the off-white residue was washed once with cold water (500 mL, 5 °C). A first LOD after 16 h gave a value of 18.73 %. HPLC) of the drying material showed the presence of 1.6% benzoate.

[00324] A brief rework study for compound 1, (containing 1.6% benzoic acid per AUC, HPLC) was executed in 10 vol of water (1 g in 10 mL):

● 3 h slurry at ambient

● 3h slurry at 50 °C

● 24 h slurry at ambient

[00325] All three experiments gave compound 1 with less than 0.1 % benzoic acid (UAC, HPLC). The slurries were fluid, were easily stirred and filtration was fast. Short term drying on the filter gave a powder-like solid indicating that a displacement wash with an organic solvent is not needed. Without wishing to be bound by theory, a loss of NMT than 1% is expected

(solubility 1 mg/mL).HPLC data for compound 1 were obtained with a method suitable for polar compounds using a Zorbax Eclipse Plus C18 column (water / ACN / TFA, 97.5 / 2.5 / 0.05). This is the same column used for steps 1 and 2.

[00326] The cold product suspension was filtered and the reactor and residue were washed with cold IPAc (approx.7.5 °C, 13.16 kg and 13.62 kg) until a colorless filtrate had been obtained. The residue was dried under vacuum and a nitrogen bleed≤ 45 °C for a period of 65 h and 19 min to an LOD of 0 %. Yield: 5.87 kg (70.7 %), 1H NMR confirmed identity; HPLC purity 98.84% (AUC). EQUIVALENTS

[0001] The disclosure can be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting on the disclosure described herein. Scope of the disclosure is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.

PATENT

WO-2019060692

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019060692&tab=PCTDESCRIPTION&maxRec=1000

Novel crystalline forms of 4-amino-7-((2R,3R,4S,5R)-3,4-dihydroxy-5-(hydroxymethyl) tetrahydrofuran-2-yl)-2-methyl-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide and their stable hemihydrate crystalline forms (designated as Form A-G), processes for their preparation and compositions comprising them are claimed. Also claimed is their use for treating viral infection.

Viral infections can have serious adverse effects on individuals and society as a whole. In addition to fatal viral infections such as Ebola, even non-fatal infections can have serious societal and economic consequences. For example, human noroviruses (NV) are the most common cause of epidemic acute gastroenteritis worldwide with an estimated 19-21 million cases each year in the United States including 56,000-71,000 hospitalizations and 570-800 deaths (Hall et al., Emerg.Infect.Dis. 2013 Aug; 19(8): 1198-205).

[0004] 4-amino-7-((2R,3R,4S,5R)-3,4-dihydroxy-5-(hydroxymethyl) tetrahydrofuran-2-yl)-2-methyl-7H-pyrrolo [2,3-d]pyrimidine-5-carboxamide (Compound 1) is an antiviral drug.

Formula 1

[0065] As used herein, “Formula I” is understood to encompass all diastereomers of 4-amino-7-(3,4-dihydroxy-5-(hydroxymethyl)tetrahydrofuran-2-yl)-2-methyl-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide, and pharmaceutically acceptable salts and solvates thereof. The structure of Formula I is shown below:


(Formula I).

[0066] In some embodiments, a compound of Formula I can be 4-amino-7-((2R,3R,4S,5R)-3,4-dihydroxy-5-(hydroxymethyl)tetrahydrofuran-2-yl)-2-methyl-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide (“Compound 1”), or a pharmaceutically acceptable salt solvate, or isomers (e.g., enantiomers and diastereomers) thereof. The structure of Compound 1 is shown below:

atent ID Title Submitted Date Granted Date
US9701706 Pyrrolopyrimidine nucleosides and analogs thereof 2016-11-22 2017-07-11
US9708359 PYRROLOPYRIMIDINE NUCLEOSIDES AND ANALOGS THEREOF 2016-08-08
US2017253628 PYRROLOPYRIMIDINE NUCLEOSIDES AND ANALOGS THEREOF 2017-05-18

///////////CMX-8521, CMX 8521, CMX-521, PHASE 1

NC(=O)c2cn([C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O)c3nc(C)nc(N)c23

DESLORATADINE, デスロラタジン

$
0
0

Desloratadine.svg

Desloratadine

  • Molecular FormulaC19H19ClN2
  • Average mass310.821 Da
100643-71-8 [RN]
5H-Benzo[5,6]cyclohepta[1,2-b]pyridine, 8-chloro-6,11-dihydro-11-(4-piperidinylidene)-
7817
Desloratadine, Descarboethoxyloratadine, Sch-34117, DCL, Denosin, Clarinex RediTabs, Allex, Desalex, Opulis, Clarinex, Neoclarityn, Aerius, MK-4117

Desloratadine (trade name Clarinex and Aerius) is a tricyclic H1-antihistamine that is used to treat allergies. It is an active metaboliteof loratadine.

It was patented in 1984 and came into medical use in 2001.[1]

Medical uses

Desloratadine is used to treat allergic rhinitisnasal congestion and chronic idiopathic urticaria (hives).[2] It is the major metabolite of loratadine and the two drugs are similar in safety and effectiveness.[2] Desloratadine is available in many dosage forms and under many trade names worldwide.[3]

An emerging indication for desloratadine is in the treatment of acne, as an inexpensive adjuvant to isotretinoin and possibly as maintenance therapy or monotherapy.[4][5]

Side effects

The most common side-effects are fatiguedry mouth, and headache.[2]

Interactions

A number of drugs and other substances that are prone to interactions, such as ketoconazoleerythromycin and grapefruit juice, have shown no influence on desloratadine concentrations in the body. Desloratadine is judged to have a low potential for interactions.[6]

Pharmacology

Pharmacodynamics

Desloratadine is a selective H1antihistamine which functions as an inverse agonist at the histamine H1 receptor.[7]

At very high doses, is also an antagonist at various subtypes of the muscarinic acetylcholine receptors. This effect is not relevant for the drug’s action at therapeutic doses.[8]

Pharmacokinetics

Desloratadine is well absorbed from the gut and reaches highest blood plasma concentrations after about three hours. In the bloodstream, 83 to 87% of the substance are bound to plasma proteins.[6]

Desloratadine is metabolized to 3-hydroxydesloratadine in a three-step sequence in normal metabolizers. First, n-glucuronidation of desloratadine by UGT2B10; then, 3-hydroxylation of desloratadine N-glucuronide by CYP2C8; and finally, a non-enzymatic deconjugation of 3-hydroxydesloratadine N-glucuronide.[9] Both desloratadine and 3-hydroxydesloratadine are eliminated via urine and feces with a half-life of 27 hours in normal metabolizers.[6][10]

3-Hydroxydesloratadine, the main metabolite

It exhibits only peripheral activity since it does not readily cross the blood-brain barrier; hence, it does not normally cause drowsiness because it does not readily enter the central nervous system.[11]

Desloratadine does not have a strong effect on a number of tested enzymes in the cytochrome P450 system. It was found to weakly inhibit CYP2B6CYP2D6, and CYP3A4/CYP3A5, and not to inhibit CYP1A2CYP2C8CYP2C9, or CYP2C19. Desloratadine was found to be a potent and relatively selective inhibitor of UGT2B10, a weak to moderate inhibitor of UGT2B17UGT1A10, and UGT2B4, and not to inhibit UGT1A1UGT1A3UGT1A4UGT1A6UGT1A9UGT2B7UGT2B15UGT1A7, and UGT1A8.[9]

Pharmacogenomics

2% of Caucasian people and 18% of people from African descent are desloratadine poor metabolizers. In these people, the drug reaches threefold highest plasma concentrations six to seven hours after intake, and has a half-life of about 89 hours. However, the safety profile for these subjects is not worse than for extensive (normal) metabolizers.[6][10]

Clip

https://www.beilstein-journals.org/bjoc/articles/9/265

The value of substituted 3-picoline precursors is illustrated in the synthesis of clarinex (1.22, Desloratadine, Scheme 5), a dual antagonist of platelet activating factor (PAF) and of histamine used in the treatment of allergies. This compound consists of a highly functional tricyclic core with an unsaturated linkage to a pendant piperidine ring. The picoline derivative 1.23 is first treated with two equivalents of n-butyllithium (n-BuLi) followed by alkylation with benzyl chloride to give the chain elongated adduct [27]. The tert-butylamide 1.24 is then dehydrated with phosphorous oxychloride at elevated temperatures to yield the nitrile derivative 1.25. Introduction of the piperidine ring is achieved by utilisation of the appropriately substituted Grignard reagent 1.26. A Friedel–Crafts type acylation promoted by either triflic acid or polyphosphoric acid (PPA) furnishes the tricyclic structure 1.28 which upon N-demethylation affords clarinex (1.22).

CLIP

Image result for desloratadine

FTIR

Image result for desloratadine

SYN

Alcoholysis of 3-methylpyridine-2-carbonitrile (I) with hot tert-butanol and H2SO4 gives the N-tert-butylcarboxamide (II), which is alkylated with 3-chlorobenzyl chloride (III) and BuLi in THF, yielding N-tert-butyl-3-[2-(3-chlorophenyl)ethyl]pyridine-2-carboxamide (IV). The reaction of (IV) with refluxing POCl3 and then with NaOH affords the corresponding nitrile (V), which is condensed with 1-methylpiperidin-4-ylmagnesium chloride (VI) in THF to give the ketone (VII). Cyclization of (VII) by means of either BF3 in HF or trifluoromethanesulfonic acid yields 8-chloro-11-(1-methylpiperidin-4-ylidene)-6,11-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridine (VIII), which is reacted with cyanogen bromide in benzene to give the N-cyano compound (IX). Finally, this compound is treated with HCl in refluxing acetic acid/water. Alternatively, 8-chloro-11-(1-methylpiperidin-4-ylidene)-6,11-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridine (VIII) is treated with ethyl chloroformate in hot toluene, affording the carbamate (X) (2), which is finally decarboxylated with KOH or NaOH in refluxing ethanol/water.

SYN

Condensation of ethyl nicotinate (XI) with 3-chlorophenylacetonitrile (XII) by means of sodium ethoxide in ethanol gives 2-(3-chlorophenyl)-3-oxo-3-(3-pyridyl)propionitrile (XIII), which by refluxing with concentrated HBr yields 2-(3-chlorophenyl)-1-(3-pyridyl)ethanone (XIV). The reduction of (XIV) with hydrazine hydrate and NaOH in diethylene glycol at 235-40 C affords 3-(2-phenylethyl) pyridine (XV), which is oxidized with H2O2 in hot acetic acid to provide the corresponding N-oxide (XVI). Reaction of (XVI) with NaCN and dimethyl sulfate in water affords the previously described 3-(2-phenylethyl)pyridine-2-carbonitrile (V), which can be worked up as previously described or cyclized with polyphosphoric acid (PPA) at 180 C to give 8-chloro-6,11-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridin-11-one (XVII). The condensation of (XVII) with 1-methylpiperidin-4-ylmagnesium chloride (VI) in THF yields the corresponding carbinol (XVIII), which is dehydrated with PPA at 170 C to afford the previously reported 8-chloro-11-(1-methylpiperidin-4-ylidene)-6,11-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridine (VIII).

SYN

Condensation of ethyl nicotinate (XI) with 3-chlorophenylacetonitrile (XII) by means of sodium ethoxide in ethanol gives 2-(3-chlorophenyl)-3-oxo-3-(3-pyridyl)propionitrile (XIII), which by refluxing with concentrated HBr yields 2-(3-chlorophenyl)-1-(3-pyridyl)ethanone (XIV). The reduction of (XIV) with hydrazine hydrate and NaOH in diethylene glycol at 235-40 C affords 3-(2-phenylethyl) pyridine (XV), which is oxidized with H2O2 in hot acetic acid to provide the corresponding N-oxide (XVI). Reaction of (XVI) with NaCN and dimethyl sulfate in water affords the previously described 3-(2-phenylethyl)pyridine-2-carbonitrile (V), which can be worked up as previously described or cyclized with polyphosphoric acid (PPA) at 180 C to give 8-chloro-6,11-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridin-11-one (XVII). The condensation of (XVII) with 1-methylpiperidin-4-ylmagnesium chloride (VI) in THF yields the corresponding carbinol (XVIII), which is dehydrated with PPA at 170 C to afford the previously reported 8-chloro-11-(1-methylpiperidin-4-ylidene)-6,11-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridine (VIII).

Syn

2) By reaction of 8-chloro-6,11-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridin-11-one (III) with the Grignard reagent (IV) to give the tertiary carbinol (V), which is dehydrated with 85% H2SO4 affording 8-chloro-11-piperidinylidene derivative (VI). Finally, cornpound (VI) is treated with ethyl chloroformate (II) in toluene.

SYN

1) By carboxylation of 8-chloro-6,11-dihydro-11-(4-piperidylidene)-5H-benzo[5,6]cyctohepta[1,2-b]pyridine (I) with ethyl chloroformate (II) in refluxing benzene.

SYN

The condensation of S-methylisothiourea (I) with trans-4-(aminomethyl)cyclohexanecarboxylic acid (II) by means of NaOH in water gives trans-4-(guanidinomethyl)cyclohexanecarboxylic acid (III) (I), which is esterified with benzyl salicylate (IV) by means of dicyclohexylcarbodiimide (DCC) or SOCl2 yielding 2-benzyloxycarbonylphenyl trans-4-(guanidinomethyl)cyclohexanecarboxylate (V). Finally, this compound is treated with cyclodextrin in aqueous solution to afford the corresponding complex.

SPECTROSCOPY

Figure CN103755682AD00061

[0052] Table 1, desloratadine sample IH-NMR data of the DMS0_d6

Figure CN103755682AD00062

[0055] The desloratadine 1H spectra of the samples were assigned:
[0056] (I) 1H spectra show that there are 10 groups of hydrogen from low field to high field integral hydrogen ratio was 1: 1: 1: 1: 1: 1: 2: 4:
2: 4, and desloratadine structure match.
[0057] (2) δ 8.334 处 hydrogen as a set of double doublet, number of protons is I, attributed to two hydrogen;
[0058] (3) δ 7.560 处 hydrogen as a set of double doublet, number of protons is I, attributed to four hydrogen;
[0059] (4) δ 7.282 处 doublet hydrogen as a group, the number of protons is I, 12 attributed to hydrogen.
[0060] (5) δ 7.198 处 hydrogen as a set of double doublet, number of protons is I, 14 attributed to hydrogen;
[0061] (6) δ 7.174 处 hydrogen as a set of double doublet, number of protons is I, attributed to three hydrogen;
[0062] (7) δ 7.064 处 doublet hydrogen as a group, the number of protons is I, 15 attributed to hydrogen;
[0063] (8) δ 3.314 处 hydrogen as a group multiplet, 2 protons attributable to 10 hydrogen;
[0064] (9) δ 2.831,2.554 hydrogen groups at multiplet, protons of 4, 18, 20, the home position is hydrogen;
[0065] (10) δ 2.819 处 hydrogen as a group multiplet, 2 protons attributable to 11 hydrogen;
[0066] (11) δ 2.108 处 hydrogen as a single peak, the number of protons is I, home to 19 active hydrogen;
[0067] (12) δ 2.205, 2.002 处 two hydrogen multiplet, protons of 4, 17, 21 bits attributed to hydrogen; [0068] From the foregoing, 1H-NMR spectrum data and the resulting product in this embodiment is of he will be loratadine same structure as the target product.

 http://www.google.com/patents/CN103755682A?cl=en

References

  1. ^ Fischer, Jnos; Ganellin, C. Robin (2006). Analogue-based Drug Discovery. John Wiley & Sons. p. 549. ISBN 9783527607495.
  2. Jump up to:a b c See S (2003). “Desloratadine for allergic rhinitis”Am Fam Physician68 (10): 2015–6. PMID 14655812.
  3. ^ Drugs.com Desloratadine entry at drugs.com international Page accessed May 4, 2015
  4. ^ Lee HE, Chang IK, Lee Y, Kim CD, Seo YJ, Lee JH, Im M (2014). “Effect of antihistamine as an adjuvant treatment of isotretinoin in acne: a randomized, controlled comparative study”. J Eur Acad Dermatol Venereol28 (12): 1654–60. doi:10.1111/jdv.12403PMID 25081735.
  5. ^ Layton AM (2016). “Top Ten List of Clinical Pearls in the Treatment of Acne Vulgaris”. Dermatol Clin34 (2): 147–57. doi:10.1016/j.det.2015.11.008PMID 27015774.
  6. Jump up to:a b c d “Aerius: EPAR – Product Information” (PDF)European Medicines Agency. 2017-06-07.
  7. ^ Canonica GW, Blaiss M (2011). “Antihistaminic, anti-inflammatory, and antiallergic properties of the nonsedating second-generation antihistamine desloratadine: a review of the evidence”World Allergy Organ J4 (2): 47–53. doi:10.1097/WOX.0b013e3182093e19PMC 3500039PMID 23268457.
Desloratadine
Desloratadine.svg
Desloratadine 3D ball-and-stick.png
Clinical data
Trade names Clarinex (US), Aerius, Dasselta, Deslordis (EU), others
AHFS/Drugs.com Monograph
MedlinePlus a602002
License data
Pregnancy
category
  • AU: B1
  • US: C (Risk not ruled out)
Routes of
administration
Oral (tablets, solution)
ATC code
Legal status
Legal status
Pharmacokinetic data
Bioavailability Rapidly absorbed
Protein binding 83 to 87%
Metabolism UGT2B10CYP2C8
Metabolites 3-Hydroxydesloratadine
Onset of action within 1 hour
Elimination half-life 27 hours
Duration of action up to 24 hours
Excretion 40% as conjugated metabolites into urine
Similar amount into the feces
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEBI
ChEMBL
ECHA InfoCard 100.166.554 Edit this at Wikidata
Chemical and physical data
Formula C19H19ClN2
Molar mass 310.82 g/mol g·mol−1
3D model (JSmol)

//////////Desloratadine, Descarboethoxyloratadine, Sch-34117, DCL, Denosin, Clarinex RediTabs, Allex, Desalex, Opulis, Clarinex, Neoclarityn, Aerius, MK-4117

Solriamfetol hydrochloride, ソルリアムフェトル塩酸塩 , солриамфетол , سولريامفيتول , 索安非托 ,

$
0
0

2D chemical structure of 178429-65-7

Solriamfetol hydrochloride

FDA APPROVED 2019/3/20, Sunosi

ソルリアムフェトル塩酸塩; R228060, R 228060

Formula
C10H14N2O2. HCl
CAS
178429-65-7 HCL
Mol weight
230.6913
(2R)-2-Amino-3-phenylpropyl carbamate
(2R)-2-Amino-3-phenylpropylcarbamat
10117
178429-62-4 [RN] FREE FORM
Benzenepropanol, β-amino-, carbamate (ester), (βR)- [
солриамфетол [Russian] [INN]
سولريامفيتول [Arabic] [INN]
索安非托 [Chinese] [INN]
JZP-110
Originator SK Holdings
  • Developer Jazz Pharmaceuticals plc; SK biopharmaceuticals
  • Class Carbamates; Sleep disorder therapies; Small molecules
  • Mechanism of Action Adrenergic uptake inhibitors; Dopamine uptake inhibitors
  • Orphan Drug Status Yes – Narcolepsy
  • Registered Hypersomnia
  • Discontinued Depressive disorders
  • 26 Mar 2019 Discontinued – Phase-I for Depressive disorders (Adjunctive treatment) in USA (PO) (Jazz Pharmaceuticals pipeline, March 2019)
  • 20 Mar 2019 Registered for Hypersomnia (excessive daytime sleepiness) in patients with obstructive sleep apnoea and narcolepsy in USA (PO) – First global approval
  • 20 Mar 2019 US FDA approves solriamfetol to improve wakefulness in adult patients with excessive daytime sleepiness associated with narcolepsy or obstructive sleep apnoea(OSA)
  • New Drug Application (NDA): 211230
    Company: JAZZ PHARMA IRELAND LTD

Solriamfetol, sold under the brand name Sunosi, is a medication used for the treatment of excessive sleepiness associated with narcolepsy and sleep apnea.[1]

Common side effects include headache, nausea, anxiety, and trouble sleeping.[1] It is a norepinephrine–dopamine reuptake inhibitor(NDRI). It is derived from phenylalanine and its chemical name is (R)-2-amino-3-phenylpropylcarbamate hydrochloride.[2]

The drug was discovered by a subsidiary of SK Group, which licensed rights outside of 11 countries in Asia to Aerial Pharma in 2011.[3]

History

The drug was discovered by a subsidiary of SK Group, which licensed rights outside of 11 countries in Asia to Aerial Pharma in 2011.[3]Aerial ran two Phase II trials of the drug in narcolepsy[4] before selling the license to solriamfetol to Jazz in 2014; Jazz Pharmaceuticalspaid Aerial $125 million up front and will pay Aerial and SK up to $272 million in milestone payments, and will pay double digit royalties to SK.[3][5]

In March 2019 the FDA accepted SK’s and Jazz’ NDA for use of solriamfetol to treat excessive sleepiness in people with narcolepsy or obstructuve sleep apnea; the drug has an orphan designation for narcolepsy.[3][6]

Names

During development it has been called SKL-N05, ADX-N05, ARL-N05, and JZP-110.[6]

Research

Solriamfetol had also been tested in animal models of depression, but as of 2017 that work had not been advanced to clinical trials.[7]

PATENT

WO 9607637

https://patents.google.com/patent/WO1996007637A1/e

Organic alkyl carbamates have been effectively used for controlling various central nervous system (CNS) disorders. For example, U.S. Pat. Nos . 2,884,444, 2,937,119 and 3,313,697 disclose function of carbamate in CNS disorders, especially as antiepileptic and centrally acting muscle relaxant.
Phenylethylamine derivatives, one important class of therapeutical medicines useful for managing CNS diseases, have been used mainly to treat obesity, narcolepsy, minimal brain dysfunction and mild depression.
Recent design of pharmacologically useful compounds has been based on amino acids or the derivatives thereof, which is mainly attributable to the fact that many of the compounds found in biological systems come from amino acids or the derivatives thereof. In addition, in most cases, the function of a pharmaceutically useful compound is effected after it binds to an enzyme or receptor, which may trigger the regulatory mechanisms of the enzyme or receptor.

REACTION SCHEME I

REACTION SCHEME II

REACTION SCHEME III

EXAMPLE I
Preparation of N-Benzyloxycarbonyl-D-phenylalaninol

In a 500 mL RB flask equipped with a mechanical stirrer and a dropping funnel, D-phenylalaninol (45.4 g, 300 mmol) was dissolved in 220 mL of distilled water, and cooled in an ice-bath. The pH of the solution was adjusted with 50 % sodium hydroxide to 14. Benzyl chloroformate (49.3 mL, 345 mmol) was charged into the dropping funnel and added slowly to the well stirred solution over 0.5 hr. After the completion of the addition, the reaction mixture was stirred for 1 hr. at 0 *C. The product precipitated from the reaction mixture as a white solid. It was collected by filtration and washed completely with distilled water. After being dried in vacuo, the solid thus obtained weighed 104 grams without any further purification: 99.8% Yield.
Melting point = 90 – 92 *C
[α]D20 = + 43.4 (c = 1.0, EtOH)
Analysis calc: C, 71.56; H, 6.71; N,4.91
Found: C, 71.35; H, 6.71; N,4.91

EXAMPLE II
Preparation of N-Benzyloxycarbonyl-D-phenylalaninol
carbamate

In a 500 mL RB flask, N-benzyloxycarbonyl-D- phenylalaninol (13.56 g, 50 mmol) was charged with antipyrine (11.29 g, 60 mmol) in 250 mL of dry THF under a nitrogen atmosphere. The reaction mixture was cooled in an ice-bath and phosgene (30.3 mL of 1.93 M solution in toluene, 58.5 mmol) was added quickly while vigorously stirring. After stirring for 1 hr. , the formation of a corresponding chloroformate from the starting material was monitored by TLC. The chloroformate solution thus prepared, was slowly added to a well stirred and ice-chilled aqueous ammonium hydroxide solution (75 mL, 28-30 %, 1,190 mmol) via cannula over 0.5 hr. The resulting reaction mixture was stirred for an extra 0.5 hr. The organic phase separated was collected. The aqueous phase was extracted twice with methylene chloride (100 mL). The combined organic phase was washed with brine (50 mL), dried over sodium sulfate, and concentrated to yield 17.8 g (113%) of foamy solid. It was purified a flash column chromatography to give 14.8 g of the title compound, white solid: 94% Yield.
Melting point = 121 – 125 *C
[α]D20 = + 28.6 (c = 2.0, EtOH)
Analysis calc. : C, 65.84; H, 6.14; N, 8.53
Found: C, 66.68; H, 6.21; N, 7.80

EXAMPLE III
Preparation of D-Phenylalaninol carbamate hydrochloric
acid salt In a 160 mL Parr reactor, N-benzyloxycarbonyl-D-phenylalaninol carbamate (9.43 g) was added with 75 mL of anhydrous methanol and 10 % palladium on charcoal (0.32 g). Then, the reactor was closed and purged with hydrogen for 1 in. The reaction was completed in 2 hrs . under 40 psi pressure of hydrogen at 45 #C. The catalyst was filtered off. Thereafter, the organic layer was concentrated into 5.97 g (102 %) of pale yellow thick liquid. The liquid was poured in 50 mL of anhydrous THF and cooled to 0 “C. Anhydrous hydrogen chloride gas was then purged through the solution with slowly stirring for

0.5 hr. 50 mL of anhydrous ether was added, to give a precipitate. Filtration with THF-ether (1:1) mixture provided 6.1 g of the title compound as a white solid: 88 % Yield.
Melting point = 172 – 174 “C
[α]D20 = – 12.9 (c = 2.0, H20)
Analysis calc. : C, 52.60; H, 6.55; N, 12.14; Cl, 15.37
Found: C, 51.90; H, 6.60; N, 12.15; Cl ,

15.52

EXAMPLE IV
Preparation of N-benzyloxγcarbonyl-L-Phenγlalaninol

The title compound was prepared in the same manner as that of Example I, except that (L)-phenylalaninol was used as the starting material.
Melting point = 90 – 92 *C
[α]D20 = – 42.0 (c = 1.0, EtOH)
Analysis calc. : C, 71.56; H, 6.71; N,4.91
Found: C, 70.98; H, 6.67; N,4.95

EXAMPLE V
Preparation of -N-benzyloxycarbonyl-L-Phenylalaninol
carbamate

The title compound was prepared in the same manner as that of Example II, except that N-benzyloxycarbonyl-L-phenylalaninol was used as the starting material.
Melting point = 121 – 128 ‘C
[α]D20 = – 28.9 (c = 2.0, EtOH)
Analysis calc: C, 65.84; H, 6.14; N, 8.53
Found: C, 65.45; H, 6.15; N, 8.32

EXAMPLE VI
Preparation of L-Phenylalaninol carbamate hydrochloric
acid salt

The title compound was prepared in the same manner as that of Example III, except that N-benzyloxycarbonyl-L-phenylalaninol carbamate was used as the starting material.
Melting point = 175 – 177 *C [α]D20 = + 13.1 (c = 1.0, H20)
Analysis calc : C, 52.60; H, 6.55; N, 12.14; Cl, 15.37
Found: C, 51.95; H, 6.58; N, 12.09; Cl , 15.37

EXAMPLE VII
Preparation of N-benzyloxycarbonyl-D,L-Phenylalaninol

The title compound was prepared in the same manner as that of Example I, except that (D,L)-phenylalaninol was used as the starting material.
Melting point = 72 – 75 #C
Analysis calc: C, 71.56; H, 6.71; N,4.91
Found: C, 71.37; H, 6.74; N,4.84

EXAMPLE VIII
Preparation of N-benzyloxycarbonyl-D,L-Phenylalaninol
carbamate

The title compound was prepared in the same manner as that of Example II, except that N-benzyloxycarbonyl-D,L-phenylalaninol was used as the starting material.
Melting point = 130 – 133 *C
Analysis calc: C, 65.84; H, 6.14; N, 8.53
Found: C, 65.85; H, 6.14; N, 8.49 EXAMPLE IX
Preparation of D,L-Phenylalaninol carbamate hydrochloric
acid salt

The title compound was prepared in the same manner as that of Example III, except that N-benzyloxycarbonyl-D,L-phenylalaninol carbamate was used as the starting material.
Melting point = 163 – 165 *C
Analysis calc: C, 52.60; H, 6.55; N, 12.14; Cl, 15.37
Found: C, 51.92; H, 6.56; N, 11.95; Cl , 15.82

PATENT

US 20050080268

PATENT

WO 2018133703

https://patents.google.com/patent/WO2018133703A1/en

Excessive daytime sleepiness (Excessive Daytime Sleepiness, EDS) or pathological somnolence refers to excessive daytime sleep and wakefulness associated with various sleep disorders. These disorders can be the basis for a sleep disorder or sleep have side effects caused by some other medical conditions. Excessive daytime sleep, also known as narcolepsy, sleep clinics is seen mainly in patients with disease that affects 12% of the general population. EDS patients may be manifested as mental distress, poor work or school performance, increasing the risk of accidents, the impact of EDS can debilitating, even life-threatening.

R228060, also known JZP-110, is a selective dopamine and norepinephrine reuptake inhibitor, originally developed by R & D, SK biopharmaceutical, 2014 Sir ownership of the pharmaceutical compound. R228060 has the potential to treat narcolepsy and sleep apnea syndrome, in three multi-center study in two global reached the primary endpoint, and achieved positive results, significantly improved adult obstructive sleep apnea patients excessive sleepiness in patients with narcolepsy and excessive sleep problems.

R228060 chemical name is O- carbamoyl – (D) – phenylalaninol, as shown in the structural formula of formula (I):

Figure PCTCN2018071889-appb-000001

Solid Form different chemicals, can cause varying their solubility and stability, and thus affects the absorption and bioavailability of the drug, and can lead to differences in clinical efficacy. Improve the candidate compound has a solubility by salt way become an important means of drug development. Compared to the free form of the drug, suitable pharmaceutically acceptable salts can improve the solubility of the drug type, increased physical and chemical stability, and also to improve the drug-salt having a melting point, hygroscopicity, crystal type and other physical properties, further development of the pharmaceutical dosage form It plays an important role. Patent Document WO1996007637A1 discloses R228060 hydrochloride and its preparation method, and other characteristics of the obtained having a melting point of 172-174 deg.] C as a white solid, the solid was not given in the text data. Further, the present inventors found no other relevant R228060 hydrochloride polymorph or patent literature. Accordingly, the present need in the art to develop a comprehensive system R228060 hydrochloride polymorph, found to be suitable to the development of crystalline form. The present inventors after many experiments, found that polymorph CS1 R228060 hydrochloride CS2 and a melting point polymorph, Form CS1 and CS2 is Form 183 ℃, much higher than the melting point disclosed in prior art solid. It provides a better alternative preparation of pharmaceutical preparations containing R228060 is, has very important implications for drug development.

PATENT

WO 2019027941

https://patentscope2.wipo.int/search/en/detail.jsf;jsessionid=15B8F200BCC820C3761C600EA64A2018?docId=WO2019027941&recNum=4220&office=&queryString=&prevFilter=%26fq%3DOF%3AWO&sortOption=Pub+Date+Desc&maxRec=3471866

(i?)-2-amino-3-phenylpropyl carbamate (APC) is a phenylalanine analog that has been demonstrated to be useful in the treatment of a variety of disorders, including excessive daytime sleepiness, cataplexy, narcolepsy, fatigue, depression, bipolar disorder, fibromyalgia, and others. See, for example, US Patent Nos. 8,232,315; 8,440,715; 8,552,060; 8,623,913; 8,729,120; 8,741,950; 8,895,609; 8,927,602; 9,226,910; and 9,359,290; and U.S. Publication Nos. 2012/0004300 and 2015/0018414. Methods for producing APC (which also has other names) and related compounds can be found in US Patent Nos. 5,955,499; 5,705,640; 6,140,532 and 5,756,817. All of the above patents and applications are hereby incorporated by reference in their entireties for all purposes.

EXAMPLE 1

Synthesis of Compounds

Compound 8 (110CR002)

1 B 110CR002

[0083] tert- utyl (if)-(l-(Carbamothioyloxy)-3-phenylpropan-2-yl)carbamate (IB): A

60% dispersion of sodium hydride (0.36 g, 4.78 mmol, 1.2 equiv) in mineral oil was added in portions to compound 1A (1.0 g. 3.98 mmol, 1 equiv) in THF (20 mL) at 0 °C. After stirring for 1 hour, carbon disulfide (0.191 g, 4.78 mmol, 1.2 equiv) was added at 0 °C. After an additional hour of stirring, methyl iodide (0.3 mL, 4.78 mmol, 1.2 equiv) was added and the reaction was warmed to room temperature. After stirring two additional hours, concentrated ammonium hydroxide (1.6 mL, 7.98 mmol, 2 equiv) was added and the reaction was stirred overnight at room temperature. The reaction was diluted with water (50 mL) and extracted with dichloromethane (3 x 50 mL). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure to give crude compound IB. The solid was triturated in diethyl ether (20 mL) to give compound IB (0.17 g, 14% yield) as a light yellow solid.

[0084] (R)-0-(2-Amino-3-phenylpropyl) carbamothioate dihydrochloride (110CR002):

4M HCI in dioxane (0.68 mL, 2.74 mmol, 5 equiv) was added to neat compound IB (0.17 g, 0.548 mmol, 1 equiv) and the reaction was stirred overnight. The solution was diluted with diethyl ether (20 mL) and the resulting suspension was filtered. The solid was triturated in diethyl ether (20 mL) and the filtered solid was dried under vacuum at room temperature for two hours to give compound 110CR003 (140 mg, 93% yield, 96.9% purity) as a white solid.

Compound 9 (110CR003)

Scheme 2

2A 2B 110CR003

[0085] (R)-2-((ter^Butoxycarbonyl)amino)-3-phenylpropyl sulfamate (2B): A solution of sulfamoyl chloride (1.15 g, 9.95 mmol, 2.5 equiv) in acetonitrile (2 mL) was added dropwise to a solution of compound 2 A (1.0 g, 3.98 mmol, 1 equiv) and triethylamine (2.1 mL, 14.95 mmol, 3.75 equiv) in N,N-dimethylacetamide (20 mL) at 0 °C. After stirring at room temperature for 4 hours, additional triethylamine (2.1 mL, 14.95 mmol, 3.75 equiv) and sulfamoyl chloride (1.15 g, 9.95 mmol, 2.5 equiv) in acetonitrile (2 mL) was added at 0 °C. The reaction was stirred at room temperature overnight, at which point LCMS indicated a 3 :2 mixture of product to starting material. Additional triethylamine (2.1 mL, 14.95 mmol, 3.75 equiv) and sulfamoyl chloride (1.15 g, 9.95 mmol, 2.5 equiv) in acetonitrile (2 mL) was added at 0 °C and the reaction was stirred at room temperature for an additional 6 hours. LCMS indicated a 4: 1 mixture of product to starting material. The reaction was quenched with saturated sodium bicarbonate (5 mL) and stirred for an additional hour at room temperature. The reaction was diluted with saturated sodium bicarbonate (25 mL) and extracted with ethyl acetate (3 x 50 mL). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure. The product still contained unreacted starting material which could not be easily separated. Sulfamoyl chloride (1.15 g, 9.95 mmol, 2.5 equiv) in acetonitrile (2 mL) was added dropwise to a solution of crude compound 2B (0.9 g) and triethylamine (2.1 mL, 14.95 mmol, 3.75 equiv) in N,N-dimethylacetamide (20 mL) at 0 °C. After stirring at room temperature for two hours, the reaction was quenched with saturated sodium bicarbonate (5 mL) and the reaction was stirred for an additional hour at room temperature. The reaction was diluted with saturated sodium bicarbonate (25 mL) and extracted with ethyl acetate (3 x 50 mL). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure. The residue was purified on an AnaLogix automated system (Redisep 24 g silica gel column), eluting with a gradient of 25 to 50% ethyl acetate in heptanes, to give compound 2B (0.37 g, 28% yield) as a white solid.

[0086] (R)-2-Amino-3-phenylpropyl sulfamate hydrochloride (110CR003): 4M HC1 in dioxane (1.4 mL, 5.6 mmol, 5 equiv) was added to neat compound 2B (0.37 g, 1.12 mmol, 1 equiv) and the reaction was stirred overnight. The solution was diluted with diethyl ether (20 mL) and the resulting suspension was filtered. The solid was triturated in diethyl ether (20 mL) and the filtered solid was dried under a vacuum at room temperature for two hours to give compound 110CR003 (250 mg, 84% yield, 97.8% purity) as a white solid.

Com ound 3 (110CR007)

[0087] (Benzyl (R)-(l-phenyl-3-ureidopropan-2-yl)carbamate) (3B): Concentrated hydrochloric acid (0.06 mL, 0.68 mmol, 0.12 equiv) was added to a solution of benzyl (ft)-(l -amino-3-phenylpropan-2-yl)carbamate ( 1.5 g, 5.28 mmol, 1 equiv) and urea (1.26 g, 21.21 mmol, 4 equiv) in toluene (150 mL) under nitrogen. After refluxing overnight, LCMS indicated the reaction was complete. The reaction was concentrated under reduced pressure, diluted with water (150 mL) and stirred for 30 minutes. The resulting solid was filtered and washed with water (25 mL) to give crude compound 3B (1.4 g, 4.27 mmol, 80% yield) as a white solid, which was used sequentially.

[0088] ((R)-l-(2-mino-3-phenylpropyl)urea) (3C): Compound 3B (0.5 g, 1.5 mmol, 1 equiv) and 10% palladium on carbon (0.09 g) in methanol (60 mL) was hydrogenated at 30 psi for 1 hour at which time LC-MS determined that the reaction was incomplete. The solution was filtered and fresh catalyst (0.09 g) was added. The solution was hydrogenated at 30 psi for an additional 45 minutes resulting in complete conversion. Two identical scale reactions were run for 105 minutes each, both resulting in complete conversion. The three runs were combined and filtered through celite, which was washed with methanol (50 mL). The filtrate was concentrated under reduced pressure to give crude compound 3C (0.9 g), which was used sequentially.

[0089] (R)-l-(2-Amino-3-phenylpropyI)urea hydrochloride (110CR007): Compound 3C (0.88 g, 4.58 mmol, 1 equiv) was dissolved diethyl ether (10 mL) and 4 N HCl in dioxane (2.31 mL, 9.27 mmol, 2 equiv) was added. The reaction was stirred overnight and then concentrated under reduced pressure to give crude 110CR007 as a white solid. The material was twice recrystallized from 10% methanol in ethanol (30 mL) to give 110CR007 (0.163 g, 16 % yield, 93.7 % purity) as a white solid.

Compound 4 (110CR009)

Scheme 4

[0090] Ethyl (R^)-4-((tert-butoxycarbonyI)amino)-5-phenylpent-2-enoate (4B): A solution of compound 4A (4.0 g, 16.1 mmol, 1 equiv) and ethyl (triphenylphos-phoranylidene)acetate (5.6 g, 16.1 mmol, 1 equiv) in dichloromethane (40 mL) was stirred at room temperature overnight. The reaction was concentrated under reduce pressure to remove the organic solvent and the resulting residue was purified on an AnaLogix automated system (40 g Sorbtech silica gel column), eluting with gradient of 50 to 100% ethyl acetate in heptanes, to give compound 4B (4.8 g, 94% yield) as a white solid.

[0091] (R^E)-4-((te *i-ButoxycarbonyI)amino)-5-phenylpent-2-enoic acid (4C): Lithium hydroxide (1.4 g, 60 mmol, 4 equiv) in water (15 mL) was added to compound 4B (4.8 g, 15 mmol, 1 equiv) in THF (60 mL) at room temperature and the reaction was stirred overnight. After 16 hours, the reaction was adjusted to pH 4 with IN hydrochloric acid. The organic layer was removed and the aqueous layer was extracted with ethyl acetate (2 x 50 mL). The combined organic layers was washed with saturated brine (50 mL), dried over sodium sulfate and concentrated under reduced pressure to give compound 4C (4.2 g, 97% yield) as a light cream solid, which was used subsequently.

[0092] Methyl (R E)-4-((½ -i-butoxycarbonyl)amino)-5-phenylpent-2-enoate (4D1):

Isobutyl chloro formate (1.3 mL, 10 mmol, 1 equiv) in THF (4 mL) was added dropwise to a solution of compound 4C (3.0 g, 10 mmol, 1 equiv) and N-methyl-morpholine (1.1 mL, 10 mmol, 1 equiv) in THF (12 mL) at -15 °C. After 30 minutes of stirring, LCMS indicated complete conversion to the anhydride intermediate. 2M Ammonia in methanol (5 mL, 10 mmol, 1 equiv) was added dropwise over 20 minutes, keeping the internal temperature between -25 to -15 °C. After 30 minutes of stirring, the reaction was warmed to room

temperature and stirred overnight. The reaction mixture was concentrated at reduced pressure to remove the organic solvent. The resulting residue was dissolved in ethyl acetate (50 mL) and washed with water (100 mL). The aqueous layer was extracted with ethyl acetate (2 x 50 mL). The combined organic layers were washed with saturated brine (50 mL), dried over sodium sulfate and concentrated under reduced pressure. The residue was purified on an AnaLogix automated system (80 g Sorbtech silica gel column), eluting with a gradient of 25 to 50% ethyl acetate in heptanes, to give compound 4D1 (1.1 g, 35 % yield) as a white solid.

[0093] Methyl (S)-4-((te^-butoxycarbonyl)amino)-5-phenylpentanoate (4D2): A mixture of compound 4D1 (1.1 g, 3.6 mmol, 1 equiv) and 10% palladium on carbon (0.33 g, 50% wet) in methanol (40 mL) was hydrogenated at 40 psi at room temperature for 4 hours. The mixture was filtered through celite, which was washed with methanol (100 mL). The filtrate was concentrated under reduced pressure to give compound 4D2 (1.1 g, 99% yield) as a white solid.

[0094] (S)-4-((ii? i-Butoxycarbonyl)amino)-5-phenylpentanoic acid (4D3): Lithium hydroxide (73 mg, 3 mmol, 1.5 equiv) in water (1 mL) was added to compound 4B (0.6 g, 2 mmol, 1 equiv) in THF (9 mL) at room temperature. After stirring overnight, the reaction was adjusted to pH 4 with IN hydrochloric acid. The organic layer was removed and the aqueous layer was extracted with ethyl acetate (3 x 25 mL). The combined organic layers was washed with saturated brine (25 mL), dried over sodium sulfate and concentrated under reduced pressure to give compound 4D3 (0.56 g, 98% yield) as a white solid, which was used subsequently.

[0095] tert-Butyl (S)-(5-amino-5-oxo-l-phenylpentan-2-yl)carbamate (4E): Isobutyl chloroformate (0.23 mL, 1.8 mmol, 1 equiv) in THF (0.5 mL) was added drop-wise to a solution of compound 4C (0.54 g, 1.8 mmol, 1 equiv) and N-methylmorpholine (0.2 mL, 1.8 mmol, 1 equiv) in THF (1 mL) at -15 °C. After 20 minutes of stirring, LCMS indicated complete conversion to the anhydride intermediate. 0.4M Ammonia in THF (9 mL, 3.6 mmol, 2 equiv) was added drop-wise over 20 minutes, keeping the internal temperature between -25 to -15 °C. After 30 minutes of stirring the reaction was warmed to room temperature and stirred overnight. The reaction mixture was concentrated under reduced pressure to remove the organic solvent. The resulting residue was dissolved in ethyl acetate (25 mL) and washed with water (25 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (2 x 25 mL). The combined organic layers were washed with saturated brine (50 mL), dried over sodium sulfate and concentrated under

reduced pressure to give compound 4E (0.5 g, 93% yield) as a white solid, which was used subsequently.

[0096] (S)-4-Amino-5-phenylpentanamide hydrochloride (110CR009): 4M HC1 in dioxane (6 mL, 25 mmol, 10 equiv) was added to compound 4E (0.73 g, 1.12 mmol, 1 equiv) After stirring overnight at room temperature, the reaction was diluted with diethyl ether (20 mL) and stirred for 6 hours. The resulting suspension was filtered and the solid was washed with diethyl ether (20 mL). The filtered solid was dried under vacuum at room temperature for two hours to give compound 110CR009 (340 mg, 60% yield, 97.9 % purity) as a white solid.

Compound 10 (110CR012)

[0097] tert-Butyl (R)-(l-(carbamoylthio)-3-phenyIpropan-2-yI)carbamate (5B):

Compound 5 A (0.15 g, 0.56 mmol, 1 equiv) was dissolved in THF (8 mL) and sparged with nitrogen for 15 minutes. Trichloroacetyl isocyanate (0.1 mL, 0.84 mmol, 1.5 equiv) was added and the solution stirred for 3 hours, at which point TLC (30% ethyl acetate in heptane) indicated absence of starting material. The reaction was cooled to 0°C and concentrated ammonium hydroxide (0.15 mL) was added. After stirring overnight at room temperature, TLC indicated that the reaction was complete. The reaction was washed with a 10% ammonium hydroxide (10 mL). The organic layer was concentrated under reduced pressure. The residue was purified on an AnaLogix automated system (12 g silica gel column), eluting with a gradient of 0 to 30% ethyl acetate in heptane, to give compound 5B. This reaction was repeated an additional two times 0.15 g and 0.18 g). The products were to give compound 5B (0.35 g, 1.12 mmol, 62.2% yield) as a white solid.

[0098] (R)-S-(2-Amino-3-phenylpropyl) carbamothioate hydrochloride (110CR012):

Compound 5B (0.35 g, 1.12 mmol, 1 equiv) was dissolved in 4N HCI in dioxane (2 mL). The reaction was stirred for two hours and then concentrated under reduced pressure to give crude 110CR012 as a white solid. The material was triturated in diethyl ether (15 mL) to give 110CR012 (0.215 g, 78 % yield, 98.0 % purity) as a white solid.

References

  1. Jump up to:a b “SUNOSI™ (solriamfetol) Tablets, for Oral Use. Full Prescribing Information” (PDF). Jazz Pharmaceuticals. 2019. Retrieved 21 March2019.
  2. ^ Abad, VC; Guilleminault, C (2017). “New developments in the management of narcolepsy”Nature and Science of Sleep9: 39–57. doi:10.2147/NSS.S103467PMC 5344488PMID 28424564.
  3. Jump up to:a b c d Ji-young, Sohn (5 March 2018). “SK Biopharmaceuticals’ narcolepsy drug on track to hitting US market”The Korea Herald.
  4. ^ Sullivan, SS; Guilleminault, C (2015). “Emerging drugs for common conditions of sleepiness: obstructive sleep apnea and narcolepsy”. Expert Opinion on Emerging Drugs20 (4): 571–82. doi:10.1517/14728214.2015.1115480PMID 26558298.
  5. ^ Garde, Damian (January 14, 2014). “Jazz bets up to $397M on Aerial’s narcolepsy drug”FierceBiotech.
  6. Jump up to:a b “Solriamfetol – Jazz Pharmaceuticals/SK Biopharmaceuticals”. AdisInsight. Retrieved 15 April 2018.
  7. ^ de Biase, S; Nilo, A; Gigli, GL; Valente, M (August 2017). “Investigational therapies for the treatment of narcolepsy”. Expert Opinion on Investigational Drugs26 (8): 953–963. doi:10.1080/13543784.2017.1356819PMID 28726523.
Solriamfetol
Solriamfetol.svg
Clinical data
Trade names Sunosi
Synonyms SKL-N05, ADX-N05, ARL-N05, and JZP-110; (R)-2-amino-3-phenylpropylcarbamate hydrochloride
Routes of
administration
By mouth
ATC code
Pharmacokinetic data
Bioavailability ~95%
Protein binding 13.3–19.4%
Metabolism negligible
Elimination half-life ~7.1 h
Excretion urine (95% unchanged)
Identifiers
CAS Number
PubChem CID
ChemSpider
UNII
KEGG
Chemical and physical data
Formula C10H14N2O2
Molar mass 194.234 g/mol g·mol−1
3D model (JSmol)

///////////Solriamfetol hydrochloride, Solriamfetol, ソルリアムフェトル塩酸塩; солриамфетол , سولريامفيتول 索安非托 JZP-110, Orphan Drug, fda 2019, R228060, R 228060


AB 680

$
0
0

str1

MFYLCAMJNGIULC-KCVUFLITSA-N.png

SCHEMBL19100484.png

20190404lnp1-ab680.jpg

AB 680

C20H24ClFN4O9P2, 580.827 g/mol

Cas 2105904-82-1

1H-Pyrazolo[3,4-b]pyridin-4-amine, 6-chloro-N-[(1S)-1-(2-fluorophenyl)ethyl]-1-[5-O-[hydroxy(phosphonomethyl)phosphinyl]-β-D-ribofuranosyl]-

[[(2R,3S,4R,5R)-5-[6-chloro-4-[[(1S)-1-(2-fluorophenyl)ethyl]amino]pyrazolo[3,4-b]pyridin-1-yl]-3,4-dihydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]methylphosphonic acid

[({[(2R,3S,4R,5R)-5-(6-chloro-4-{[(1S)-1-(2-fluorophenyl)ethyl]amino}-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-3,4-dihydroxyoxolan-2-yl]methoxy}(hydroxy)phosphoryl)methyl]phosphonic Acid

  • Originator C
  • Class Antineoplastics; Small molecules
  • Mechanism of Action 5-nucleotidase inhibitors; Adenosine A2 receptor antagonists
  • Phase I Cancer
  • 19 Nov 2018 Arcus Biosciences plans to initiate a clinical trial in Cancer in first half of 2019
  • 16 Oct 2018 Phase-I clinical trials in Cancer (In volunteers) in Australia (IV) (NCT03677973)
  • 30 Sep 2018 Preclinical pharmacodynamics data in Cancer presented at 4th CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference (CRI-CIMT-EATI-AACR – 2018)

Clip

https://cen.acs.org/pharmaceuticals/drug-discovery/Drug-structures-displayed-first-time-in-Orlando/97/web/2019/04?utm_source=Facebook&utm_medium=Social&utm_campaign=CEN

Credit: Tien Nguyen/C&EN

Presenter: Kenneth V. Lawson, senior scientist at Arcus Biosciences

Target: Ecto-5’-nucleotidase (CD73)

Disease: Cancer

Reporter’s notes: In the first talk of the day, Lawson introduced the idea of cancer drugs that target the host’s immune system. “Checkpoint inhibitors changed the way we think of treating cancer,” he said. These drugs successfully disrupt the binding interaction between a protein and a checkpoint protein that stops immune T cells from killing cancer cells. As a result, these drugs turn immune cells loose to attack tumor cells. But the drugs work only in about 30-40% of patients—an issue pharmaceutical companies like Arcus hope to address with new immunotherapies that can be taken in combination with checkpoint inhibitors.

Lawson’s team set out to inhibit an enzyme commonly found in tumors called CD73, the second of two enzymes which break down extracellular adenosine trisphosphate (ATP) to adenosine. Adenosine then binds to immunosuppressive receptors on immune cells and shuts them down. Yet developing a small molecule inhibitor of CD73 proved challenging, Lawson said. After striking out with high-throughput screening, the team turned to CD73’s natural substrate for inspiration. However, the molecule possessed more than one phosphate group, which is notoriously a liability for drug molecules because small molecules with such negative changes struggle to cross cell membranes. The team’s goal was to remove the phosphate groups, Lawson says, but things didn’t exactly go according to plan. After showing the audience a series of compounds from structure-activity relationship (SAR) studies—slides no medicinal chemistry talk would be complete without—Lawson revealed the structure of their final clinical compound AB680 as the sound of people flipping notebook sheets rippled across the room. Synthesized in 34% overall yield, the candidate ultimately included two phosphate groups—a feature that surprised audience members.

Tests revealed that AB680 can be given intravenously but the compound also showed moderate oral bioavailability. Lawson suggested a possible route for how the molecule might pass from the digestive tract to the bloodstream, a paracellular mechanism by which molecules cross the epithelium by passing through the space between cells. AB680 showed “extraordinary potency,” inhibiting CD73 in human T-cells at a concentration of 0.008 nM. The compound has a 4 day half-life, which means it could be dosed every two weeks, coinciding with the dosing schedule for patients who receive a checkpoint inhibitor. AB680 is currently in Phase 1 clinical trials with healthy patients.

str1

PATENT

US2017267710

https://patentscope.wipo.int/search/en/detail.jsf?docId=US204141996&tab=PCTDESCRIPTION&maxRec=1000

Purinergic signaling, a type of extracellular signaling mediated by purine nucleotides and nucleosides such as ATP and adenosine, involves the activation of purinergic receptors in the cell and/or in nearby cells, resulting in the regulation of cellular functions. Most cells have the ability to release nucleotides, which generally occurs via regulated exocytosis (see Praetorius, H. A.; Leipziger, J. (1 Mar. 2010) Ann Rev Physiology 72(1): 377-393). The released nucleotides can then be hydrolyzed extracellularly by a variety of cellular membrane-bound enzymes referred to as ectonucleotidases.
      Ectonucleotides catalyze the conversion of ATP to adenosine, an endogenous modulator that impacts multiple systems, including the immune system, the cardiovascular system, the central nervous system, and the respiratory system. Adenosine also promotes fibrosis in a variety of tissues. In the first step of the production of adenosine, ectonucleoside triphosphate diphosphohydrolase 1 (ENTPD1), also known as CD39 (Cluster of Differentiation 39), hydrolyzes ATP to ADP, and then ADP to AMP. In the next step, AMP is converted to adenosine by 5′-nucleotidase, ecto (NT5E or 5NT), also known as CD73 (Cluster of Differentiation 73).
      The enzymatic activities of CD39 and CD73 play strategic roles in calibrating the duration, magnitude, and chemical nature of purinergic signals delivered to various cells (e.g., immune cells). Alteration of these enzymatic activities can change the course or dictate the outcome of several pathophysiological events, including cancer, autoimmune diseases, infections, atherosclerosis, and ischemia-reperfusion injury, suggesting that these ecto-enzymes represent novel therapeutic targets for managing a variety of disorders.
      CD73 inhibition with monoclonal antibodies, siRNA, or small molecules delays tumor growth and metastasis (Stagg, J. (2010) PNAS U.S.A. 107:1547-52). For example, anti-CD73 antibody therapy was shown to inhibit breast tumor growth and metastasis in animal models (Stagg, J. (26 Jan. 2010) PNAS U.S.A, 107(4):1547-52). In addition, the use of antibodies that specifically bind CD73 has been evaluated for the treatment of bleeding disorders (e.g., hemophilia) (U.S. Pat. No. 9,090,697). Recently, there have been several efforts to develop therapeutically useful CD73 small molecule inhibitors. For example, Bhattarai et al. ((2015) J Med Chem 58:6248-63) have studied derivatives and analogs of α,β-Methylene-ADP (AOPCP), one of the most metabolically stable, potent and selective CD73 inhibitors known, and purine CD73 derivatives have been reported in the patent literature (WO 2015/164573). However, the development of small molecules has been hampered due to, for example, less than ideal metabolic stability.
      In view of the role played by CD73 in cancer, as well as a diverse array of other diseases, disorders and conditions, and the current lack of CD73 inhibitors available to medical practitioners, new CD73 inhibitors, and compositions and methods associated therewith, are needed.

Example 92

Synthesis of [({[(2R,3S,4R,5R)-5-(6-chloro-4-{[(1S)-1-(2-fluorophenyl)ethyl]amino}-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-3,4-dihydroxyoxolan-2-yl]methoxy}(hydroxy)phosphoryl)methyl]phosphonic Acid


      The title compound was synthesized in similar fashion to Example 87. 1H NMR (400 MHz, DMSO-d 6) δ 9.28-9.15 (m, 1H), 8.33 (dd, J=1.5, 0.7 Hz, 1H), 7.43 (t, J=7.8 Hz, 1H), 7.29 (dd, J=7.8, 5.6 Hz, 1H), 7.23-7.08 (m, 2H), 6.00 (d, J=4.2 Hz, 1H), 5.65-5.51 (m, 1H), 4.48 (t, J=4.9 Hz, 1H), 4.26 (t, J=4.5 Hz, 1H), 4.05 (dq, J=10.1, 5.9, 5.2 Hz, 2H), 3.88 (dt, J=11.3, 6.0 Hz, 1H), 2.29-2.08 (t, J=20.4 Hz, 2H), 1.53 (d, J=6.8 Hz, 3H). ESI MS [M+H] + for C 19H24ClFN 592, calcd 582.1. found 582.1.

PATENT

WO 2017120508

////////////////ARCUS, AB 680, AB680, AB-680, PHASE 1

https://www.arcusbio.com/wp-content/uploads/2018/04/AACR_AB680_1756_final_90x42-abstract-4886.pdf

https://cen.acs.org/pharmaceuticals/drug-discovery/Drug-structures-displayed-first-time-in-Orlando/97/web/2019/04?utm_source=Facebook&utm_medium=Social&utm_campaign=CEN

Fc1ccccc1[C@H](C)Nc4cc(Cl)nc3c4cnn3[C@@H]2O[C@H](COP(=O)(O)CP(=O)(O)O)[C@@H](O)[C@H]2O

CC(C1=CC=CC=C1F)NC2=CC(=NC3=C2C=NN3C4C(C(C(O4)COP(=O)(CP(=O)(O)O)O)O)O)Cl

LHC 165

$
0
0

SDLWKRZBLTZSEL-UHFFFAOYSA-N.png

str1

LHC165

3-[5-amino-2-[2-[4-[2-(3,3-difluoro-3-phosphonopropoxy)ethoxy]-2-methylphenyl]ethyl]benzo[f][1,7]naphthyridin-8-yl]propanoic acid

C29H32F2N3O7P, 603.56 g/mol

CAS  1258595-14-0

5-Amino-2-[2-[4-[2-(3,3-difluoro-3-phosphonopropoxy)ethoxy]-2-methylphenyl]ethyl]benzo[f][1,7]naphthyridine-8-propanoic acid

Benzo[f][1,7]naphthyridine-8-propanoic acid, 5-amino-2-[2-[4-[2-(3,3-difluoro-3-phosphonopropoxy)ethoxy]-2-methylphenyl]ethyl]-

  • Originator Novartis
  • Class Antineoplastics
  • Mechanism of Action
  • Undefined mechanism
  • Phase I Solid tumours
  • 31 Jan 2018 Phase-I clinical trials in Solid tumours (Combination therapy, Inoperable/Unresectable, Late-stage disease, Metastatic disease, Second-line therapy or greater) in USA, Belgium, Italy, Japan (Intratumoural) (NCT03301896)
  • 31 Jan 2018 Phase-I clinical trials in Solid tumours (Inoperable/Unresectable, Late-stage disease, Metastatic disease, Monotherapy, Second-line therapy or greater) in USA, Japan, Italy, Belgium (Intratumoural) (NCT03301896)
  • 10 Oct 2017 Novartis plans a phase I trial for Solid tumours (Monotherapy, Combination therapy, Inoperable/Unresectable, Late-stage disease, Metastatic disease, Second-line therapy or greater) in USA, Belgium, Canada, France, Germany, Italy, South Korea and Spain in November 2017 (Intratumoural) (NCT03301896)

PATENT

WO 2010144734

PATENT

US 20110053893

PATENT

WO 2011130379

PATENT

WO 2011027222

 

Scheme (III)

Scheme (IV)

Scheme (V)

Example 19 (Table 1: Compound 19): Synthesis of 3-(5-amino-2-(4-(2-(3,3-difluoro-3-phosphonopropoxy)ethoxy)-2-methylphenethyl)benzo[f][ 1, 7]naphthyridin-8-yl)propanoic acid (19)

Scheme 6

Step 1: (E)-ethyl 3-(3-(tert-butoxycarbonylamino)-4-chlorophenyl)acrylate (6-3)

[517] To a solution of tert-butyl 5-bromo-2-chlorophenylcarbamate (6-1) (1.0 equiv.) in acetonitrile (0.3 M) and EtOH (0.5 M) was added K2C03 (2.0 equiv.). The reaction was degassed and flushed with N , then added (E)-ethyl 3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)acrylate (6-2) (1.2 equiv.) and Pd(PPh3)4 (0.1 equiv.). The reaction was flushed again with N2 and stirred at 100 °C overnight. After cooling to room temperature, hexane was added, and the mixture was filtered through a pad of silica, eluting with EA/Hex (1 : 1) until the product was completely eluted. The filtrate was concentrated and purified on Combiflash, eluting with 0-15% EA in Hex to give (E)-ethyl 3-(3-(tert-butoxycarbonylamino)-4-chlorophenyl)acrylate (6-3) as a white solid.

Step 2: ethyl 3-(3-(tert-butoxycarbonylamino)-4-chlorophenyl)propanoate (6-4)

[518] To a solution of (E)-ethyl 3-(3-(tert-butoxycarbonylamino)-4-chlorophenyl)acrylate (6-3) (1.0 equiv.) in ethyl acetate/ethanol (1 : 1 , 0.3 M) was added Wilkinson’s catalyst (0.10 equiv.).

Hydrogen gas was introduced via a ballon, and the reaction was stirred at room temperature for 24 hours. The mixture was filtered through a pad of celite, washing with dichloromethane. The filtrate was concentrated in vacuo and purified by Combiflash using 0-10% ethyl acetate in hexane to give ethyl 3-(3-(tert-butoxycarbonylamino)-4-chlorophenyl)propanoate (6-4) as a solid.

Step 3: ethyl 3-(3-(tert-butoxycarbonylamino)-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)propanoate (6-5)

[519] A solution of ethyl 3-(3-(tert-butoxycarbonylamino)-4-chlorophenyl)propanoate (6-4) (1 .0 equiv.), 4,4,4,,4′,5,5,5′,5′-octamethyl-2,2′-bi(l ,3,2-dioxaborolane) (2.0 equiv.), tris(dibenzylideneacetone)dipalladium(0) (0.05 equiv.), 2-dicyclohexylphosphino-2′,4′,6′-triisopropylbiphenyl (0.20 equiv.), and potassium acetate (2.0 equiv.) in 1 ,4-dioxane (0.2 M) was degassed and stirred at 100 °C overnight. After cooling to ambient temperature, the reaction content was concentrated in vacuo. The crude material was purified by Combiflash using 0-50% ethyl acetate in hexane to afford ethyl 3-(3-(tert-butoxycarbonylamino)-4-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)phenyl)propanoate (6-5) as a brown oil. The product was stored at -20°C and used within a month of synthesis.

Step 4: l-bromo-4-(methoxymethoxy)-2-methylbenzene (6-7)

[520] To a solution of 4-bromo-3-methylphenol (6-6) (1.0 equiv.) in DMF (0.5 M) at 0 °C was added portionwise 60% wt NaH (1.5 equiv.). The addition was controlled such that internal reaction temperature never went above 10 °C. The reaction was stirred at room temperature for 45 minutes, then a solution of chloro(methoxy)methane (1.2 equiv.) in DMF (3 M) was added dropwise via additional funnel. The reaction was stirred at room temperature for 3.5 hours, and then quenched by pouring into ice. The resulting mixture was stirred at room temperature for 1 hour. Ether was added, and the two layers were separated. The aqueous layer was extracted (lx) with ether. The combined organic layers were washed with water (2x), brine, dried over MgS04, and concentrated to give 1 -bromo-4-(methoxymethoxy)-2-methylbenzene (6-7) as a colorless oil. The crude material was used in the next step without further purification.

Step 5: triethylf (4-(methoxymethoxy)-2-methylphenyl)ethynyl)silane

[521] A solution of l -bromo-4-(methoxymethoxy)-2-methylbenzene (1.0 equiv.), triethylamine (5.0 equiv.) in DMF (0.5 M) was degassed and flushed with nitrogen. To the reaction was added TES-acetylene (1.05 equiv.), Cul (0.098 equiv.), and Pd(PPh3)2Cl2 (0.098 equiv.). The reaction was heated to 60 °C and stirred overnight. After cooling to room temperature, water and ether were added. The layers were separated, and the organic layer was washed with water (2x). The organic layer was separated and passed through a pad of silica (packed with hexane). The silica was eluted with 10% EA in Hex. The fractions were combined and concentrated to give triethyl((4-(methoxymethoxy)-2-methylphenyl)ethynyl)silane as a black oil. The crude material was used in the next step without further purification.

Step 6: l-ethynyl-4-(methoxymethoxy)-2-methylbenzene (6-8)

[522] To a solution of triethyl((4-(methoxymethoxy)-2-methylphenyl)ethynyl)silane (1.0 equiv.) at

0 °C was slowly added tetrabutylammonium fluoride (1M solution in THF, 0.20 equiv.). At this

point, the ice-bath was removed and the reaction mixture was allowed to stir at room temperature for 45 minutes. The reaction mixture was then passed through a pad of silica (packed with hexane) and eluted with 20% EtOAc in Hexanes to remove insoluble salts. The crude product was then purified by Combiflash using 0-10% EtOAc in Hexanes to give 1 -ethynyl-4-(methoxymethoxy)-2-methylbenzene (6-8) as a slightly brown liquid.

Step 7: 3-chloro-5-((4-(methoxymethoxy)-2-methylphenyl)ethynyl)picolinonitrile (6-10)

[523] A solution of l -ethynyl-4-(methoxymethoxy)-2-methylbenzene (6-8) (1 .0 equiv.), 3,5-dichloropicolinonitrile (6-9) (0.90 equiv.), Cul (0.10 equiv.), and Pd(PPh3)2CI2 (0.10 equiv.), and triethylamine (5.0 equiv.) in DMF (0.25 M) was degassed and flushed with nitrogen. The reaction mixture was then heated to 60 °C and stirred overnight. After cooling to room temperature, water was added. The mixture was extracted with EA (2x). The combined organic layers were washed with 10% aq NH4OH (2x), brine, and concentrated. The crude material was filtered through a pad of silica (wetted with hexane). The silica was eluted with 10% EA in Hex. The fractions were combined and concentrated. The resulting solids were washed in hot ether and filtered to give a yellow solid, which was used in the next step without further purification. The filtrate was concentrated and purified by Combiflash using 0- 10% EtOAc in Hexanes to give 3-chloro-5-((4-(methoxymethoxy)-2-methylphenyl)ethynyl)picolinonitrile (6-10) as a yellow solid.

Step 8: ethyl 3-(5-amino-2-((4-(methoxymethoxy)-2-methylphenyl)ethynyl)-ben∑o fJfl, 7J

naphthyridin-8-yl)propanoate (6-11)

[524] A solution of 3-chloro-5-((4-(methoxymethoxy)-2-methylphenyl)ethynyl)picolinonitrile (6-10) (1 .0 equiv.), ethyl 3-(3-(tert-butoxycarbonylamino)-4-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2-yl)phenyl)propanoate (6-5) (1.25 equiv.), tris(dibenzylideneacetone)dipalladium(0) (0.10 equiv.), dicyclohexyl(2′,6′-dimethoxybiphenyl-2-yl)phosphine (0.20 equiv.), and sodium bicarbonate (3.0 equiv.) in «-butanol /H20 (5: 1 , 0.2 M) was degassed and stirred at 100 °C overnight. After cooling to ambient temperature, the reaction content was diluted with ethyl acetate and water. The two phases were separated, and the aqueous layer was extracted twice with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous MgS04, and concentrated in vacuo. The crude material was purified by flash chromatography on a COMBIFLASH® system (1SCO) using 0-40% ethyl acetate in DCM first to remove the impurity, then 0-4% MeOH in DCM to give ethyl 3-(5-amino-2-((4-(methoxymethoxy)-2-methylphenyl)ethynyl)-benzo[f][l ,7]naphthyridin-8-yl) propanoate (6-11). Further purification was accomplished by precipitating and washing in hot ether.

Step 9: ethyl 3-(5-amino-2-(4-(methoxymethoxy)-2-methylphenethyl)benzo[fl[l ]naphthyridin-8-yl)propanoate (6-12)

[525] A solution of ethyl 3-(5-amino-2-((4-(methoxymethoxy)-2-methylphenyl)ethynyl)-benzo[f][l ,7]naphthyridin-8-yl)propanoate (6-11) (1.0 equiv.) in EtOH/THF (3: 1 , 0.16 M) was flushed with nitrogen. Then, 10% wt Pd/C (0.20 equiv. by weight) was added. The reaction was flushed with hydrogen (2x) and stirred under a hydrogen balloon. After 24 hours, the reaction was filtered through a pad of celite, washing with 5%MeOH in DCM. The filtrate was checked for the presence of starting material using LCMS. The hydrogenation reaction was repeated until no more

of the alkyne starting material or alkene intermediate was detected. The crude product was purified by Combiflash using 0-4% eOH in DCM to give ethyl 3-(5-amino-2-(4-(methoxymethoxy)-2-methylphenethyl)benzo[f][l ,7]naphthyridin-8-yl)propanoate (6-12) as a white solid.

Step 10: ethyl 3-(5-amino-2-(4-hydroxy-2-methylphenethyl)benzo[fl[l ]naphthyridin-8-yl)propanoate (6-13)

[526] Ethyl 3-(5-amino-2-(4-(methoxymethoxy)-2-methylphenethyl)benzo[fJ[l ,7]naphthyridin-8-yl)propanoate (6-12) (1 .0 equiv.) was dissolved in EtOH (0.2 M), then added a solution of 4M HC1 in dioxane (0.2 M). The product precipitated out as a yellow salt. After stirring for 3 hours, the reaction was poured into a stirring solution of ether. The mixture was stirred for 10 minutes, then filtered and washed with ether. Ethyl 3-(5-amino-2-(4-hydroxy-2-methylphenethyl)benzo[fJ[l ,7]naphthyridin-8-yl)propanoate (6-13) was obtained as a yellow solid which was dried on vacuum overnight (bis-HCl salt). Alternatively, the crude product was purified by Combiflash using 0-5% MeOH in DCM to give the free base.

Step 11: ethyl 3-(5-amino-2-(4-(2-(3-(diethoxyphosphoryl)-3,3-difluoropropoxy)ethoxy)-2-methylphenethyl)benzo[f] [1 , 7]naphthyridin-8-yl)propanoate ( 6-15)

[527] To a solution of ethyl 3-(5-amino-2-(4-hydroxy-2-methylphenethyl)benzo[fJ [ l ,7]naphthyridin-8-yl)propanoate (6-13) (1.0 equiv.) dissolved in DMF (0.14 M) was added a solution of diethyl 3-(2-bromoethoxy)-l ,l -difluoropropylphosphonate (6-14: described in Example 7 – Step 1) (1 .3 equiv.) in DMF (0.7 M) and cesium carbonate (4 equiv.). The reaction was stirred at 60 °C. After 1.5 hours (or until reaction is complete by LCMS), DCM (2 volume equivalent) was added to the reaction. The solids (inorganic) were filtered, and the filtrate was concentration. The crude product was purified by Combiflash using 0-5%MeOH in DCM to give ethyl 3-(5-amino-2-(4-(2-(3-(diethoxyphosphoryl)-3,3-difluoropropoxy)ethoxy)-2-methylphenethyl)benzo[fJ

[1 ,7]naphthyridin-8-yl)propanoate (6-15) as an oil which upon standing became a white solid.

Step 12: 3-(5-amino-2-(4-(2-(3,3-difluoro-3-phosphompropoxy)ethoxy)-2-methylphenethyl)be o[f]

[1, 7]naphthyridin-8-yl)propanoic acid (19)

[528] To a solution of ethyl 3-(5-amino-2-(4-(2-(3-(diethoxyphosphoryl)-3,3-difluoropropoxy)ethoxy)-2-methylphenethyl)benzo[f][l ,7]naphthyridin-8-yl)propanoate (6-15) (1.0 equiv.) in DCM (0.16 M) at 0 °C was added slowly TMSBr (10 equiv.). The reaction was stirred at room temperature overnight. Additional TMSBr (5.0 equiv.) was added at 0 °C, and the reaction was again stirred at room temperature overnight. The solvent was removed by evaporation and the crude orange solids dried on hi-vac briefly. The solids were suspended in EtOH (0.5 M) and added 2.5 N

NaOH (10.0 equiv.). The reaction was stirred at 80 °C for 3 hours. After cooling to room temperature, the mixture was adjusted to pH 9 to 10 and directly purified on RP-HPLC using a CI 8 column, eluting with 10-40% 95:5 (MeCN/5mM NH4OAc) in l OmM NH4OAc (pH 9) gradient. The fractions containing the product were combined and concentrated in vacuo. The resulting white gel was dissolved in refluxing 1 :1 EtOH/water (0.04 M) with the addition of a few drops of ammonium hydroxide. While hot, the mixture was slowly poured into a stirring hot solution of acetone (0.009

M) preheated at 50 °C. The acetone suspension was slowly cooled to room temperature for 15 minutes with continued stirring, and then sat in an ice bath for 10 minutes. The solids were filtered and washed successively with acetone (2x) and ether (2x). The solids were dried on hi-vac overnight to give the 3-(5-amino-2-(4-(2-(3,3-difluoro-3-phosphonopropoxy)ethoxy)-2-methylphenethyl)benzo [fj[l ,7]naphthyridin-8-yl)propanoic acid (19) as a solid. Ή NMR (Dimethylsulfoxide-d6): δ 9.02 (s, 1 H), 8.82 (s, 1H), 8.55 (d, 1H, J = 8.4 Hz), 7.58 (s, 1H), 7.48 (d, 1 H, J = 8.4 Hz), 7.07 (d, 1H, J = 8.4 Hz), 6.75 (s, 1 H), 6.68 (d, 1H, J = 8.4 Hz), 4.03-4.00 (m, 2H), 3.72-3.68 (m, 4H), 3.16-3.12 (m, 2H), 3.03-2.96 (m, 4H), 2.67-2.64 (m, 2H), 2.33-2.32 (m, 2H), 2.26 (s, 3H). LRMS [M+H] = 604.2

PATENT

US 20120237546

PATENT

WO 2012031140

PATENT

WO 2018211453

Toll-like receptors (TLRs) are pattern recognition receptors which play an essential role in the innate immunity, by recognizing invasion of microbial pathogens and initiating intracellular signal transduction pathways to trigger expression of genes, the products of which can control innate immune responses. Specifically, Toll like receptor (TLR) agonists activate innate immune cells through the TLR-MyD88-NFk and IRF3/7 pathways. TLR7, TLR8, and TLR9 belong to a subfamily of TLRs based on their genomic structure, sequence similarities, and homology. TLR7, TLR8, and TLR9 are located in intracellular endolysosomal compartments and show a unique pattern of cell type-specific expression that is thought to be responsible for different pathogen response profiles.

Small molecule agonists of TLR7 and/or TLR8 have been reported and shown to activate innate immune responses by inducing selected cytokine biosynthesis, the induction of co-stimulatory molecules, and by increased antigen-presenting capacity. Such compounds include imidazoquinoline amine derivatives (U.S. Patent No. 4689338), imidazopyridine amine derivative (U.S. Patent No. 5446153), imidazonaphthyridine derivative (U.S. Patent No.

6194425), oxazoloquinoline amine derivatives (U.S. Patent No. 61 10929); thiazoloquinoline amine derivatives (U.S. Patent No. 61 10929), selenazoloquinoline amine derivatives (U.S. Patent No. 61 10929), pyrazolopyridine derivatives (U.S. Patent No. 9145410), and

benzonaphthyridine amine derivatives (U.S. Patent Nos. 8466167 and 9045470).

The synthetic TLR7 agonist, Imiquimod (1 -(2-methylpropyl)-1 H-imidazo[ 4,5-c]quinolin-4-amine) is FDA-approved in a cream formulation for the topical treatment of cutaneous basal cell carcinoma, actinic keratosis and genital warts, and has limited activity against cutaneous melanoma and breast tumors (J. Immunol. 2014, 193(9) : 4722^1-731 ). Systemic administration of Imiquimod, and structurally similar Resiquimod, is limited by cytokine- mediated adverse effects including severe flu-like symptoms (Expert Opin. Emerging Drugs (2010), 15:544-555). Consequently, Imiquimod is used exclusively in topical applications and is not used to treat deep, non-cutaneous tumors such as melanoma or solid tumors.

An injectable lipid modified imidazoquinoline (TLR7/8 dual agonist) that forms a tissue depot with gradual, sustained release which allows for local TLR triggering activity without systemic cytokine release has been reported (J. Immunol. 2014, 193(9): 4722^731 ). However, this compound was shown to be ineffective for large tumors and in addition the serum concentration of this compound 24 hours post subcutaneous administration decreased by approximately 50% (Journal for ImmunoTherapy of Cancer, 2014, 2:12). Therefore, there remains a need for intratumor administration of a TLR7 agonist with prolonged sustained release, which may benefit the treatment of large tumors.

clip

https://cen.acs.org/pharmaceuticals/drug-discovery/Drug-structures-displayed-first-time-in-Orlando/97/web/2019/04?utm_source=Facebook&utm_medium=Social&utm_campaign=CEN

Candidate: LHC165

20190404lnp1-lhc165.jpg

Credit: Tien Nguyen/C&EN

Presenter: Alex Cortez, senior Investigator I at the Genomics Institute of the Novartis Research Foundation

Target: Toll-like receptor 7 (TLR7)

Disease: Solid tumors

Reporter’s notes: Cortez shared another story in the realm of immuno-oncology, although the program that yielded this compound actually started in the world of vaccines. Cortez’s team had been focusing on vaccine adjuvants, small molecules that turn on the immune system to enhance a vaccine’s effect. They developed one such class of compound that activates toll-like receptor 7 (TLR7), a protein in the immune system that recognizes dangerous-looking molecules and can trigger the release of infection-clearing proteins. After observing TLR7 agonists’ ability to induce an immune response with vaccines, the researchers wondered whether the molecules could also be effective in immuno-oncology.

They found that LHC165 adsorbed to aluminum hydroxide reduced tumor growth in mice and, intriguingly, showed signs of an abscopal effect, in which untreated tumors shrink concurrently with treated tumors. The implication is that if the immune system recognizes one tumor site, it can recognize others. As with several of the candidates presented throughout the day, LHC165 bears a phosphate group and is injected into the tumor. It’s currently in Phase I trials in patients with advanced malignancies, which means they’ve already tried second and third line therapies, as a single agent and in combination with the checkpoint inhibitor PDR001.

US9618508FLOW CYTOMETRY ANALYSIS OF MATERIALS ADSORBED TO METAL SALTS2011-12-142013-12-12
US2014112950COMBINATION VACCINES WITH LOWER DOSES OF ANTIGEN AND/OR ADJUVANT2012-03-022014-04-24
Patent ID Title Submitted Date Granted Date
US9597326 BENZONAPTHYRIDINE COMPOSITIONS AND USES THEREOF 2011-04-13 2013-05-16
US9950062 COMPOUNDS AND COMPOSITIONS AS TLR ACTIVITY MODULATORS 2010-09-01 2012-09-20
US9517263 BENZONAPHTHYRIDINE-CONTAINING VACCINES 2010-06-10 2012-10-18
US2015225432 COMPOUNDS AND COMPOSITIONS AS TLR ACTIVITY MODULATORS 2015-04-24 2015-08-13
US9315530 ADSORPTION OF IMMUNOPOTENTIATORS TO INSOLUBLE METAL SALTS 2011-09-01
Patent ID Title Submitted Date Granted Date
US2016213776 ADSORPTION OF IMMUNOPOTENTIATORS TO INSOLUBLE METAL SALTS 2016-04-07 2016-07-28
US2012177681 Formulation of immunopotentiators 2011-09-01 2012-07-12
US9045470 COMPOUNDS AND COMPOSITIONS AS TLR ACTIVITY MODULATORS 2011-03-03
US2018169204 COMBINATION VACCINES WITH LOWER DOSES OF ANTIGEN AND/OR ADJUVANT 2018-02-02
US9375471 ADJUVANTED FORMULATIONS OF BOOSTER VACCINES 2013-03-08 2013-09-12

//////LHC165, LHC 165, LHC -165, Phase I,  Solid tumours, novartis

O=P(O)(O)C(F)(F)CCOCCOc4ccc(CCc1cc2c3ccc(CCC(=O)O)cc3nc(N)c2nc1)c(C)c4

CC1=C(C=CC(=C1)OCCOCCC(F)(F)P(=O)(O)O)CCC2=CN=C3C(=C2)C4=C(C=C(C=C4)CCC(=O)O)N=C3N

https://cen.acs.org/pharmaceuticals/drug-discovery/Drug-structures-displayed-first-time-in-Orlando/97/web/2019/04?utm_source=Facebook&utm_medium=Social&utm_campaign=CEN

Acefylline

$
0
0

Acefylline

Skeletal formula of acefylline

Acefylline

  • Molecular FormulaC9H10N4O4
  • Average mass238.200 Da
(1,3-Dimethyl-2,6-dioxo-1,2,3,6-tetrahydro-7H-purin-7-yl)acetic acid
1,2,3,6-Tetrahydro-1,3-dimethyl-2,6-dioxo-7H-purine-7-acetic Acid
1,3-Dimethylxanthine-7-acetic acid
211-490-2 [EINECS]
652-37-9 [RN]
7-(Carboxymethyl)theophylline
7H-Purine-7-acetic acid, 1,2,3,6-tetrahydro-1,3-dimethyl-2,6-dioxo-
CAS Registry Number: 652-37-9
CAS Name: 1,2,3,6-Tetrahydro-1,3-dimethyl-2,6-dioxopurine-7-acetic acid
Additional Names: carboxymethyltheophylline; 7-theophyllineacetic acid
Molecular Formula: C9H10N4O4
Molecular Weight: 238.20
Percent Composition: C 45.38%, H 4.23%, N 23.52%, O 26.87%
Literature References: Prepn: DE 352980 (1922 to E. Merck); Frdl. 14, 1320; S. M. Ride et al., Pharmazie 32, 672 (1977). Prepn of salts: J. Baisse, Bull. Soc. Chim. Fr. 1949, 769; M. Milletti, F. Virgili, Chimica 6, 394 (1951), C.A. 46, 8615h (1952). GC determn in urine: J. Zuidema, H. Hilbers, J. Chromatogr. 182, 445 (1980). HPLC determn in serum and pharmacokinetics: S. Sved et al.,Biopharm. Drug Dispos. 2, 177 (1981).
Properties: Crystals from water, mp 271°.
Melting point: mp 271°
Derivative Type: Sodium salt
CAS Registry Number: 837-27-4
Molecular Formula: C9H9N4NaO4
Molecular Weight: 260.18
Percent Composition: C 41.55%, H 3.49%, N 21.53%, Na 8.84%, O 24.60%
Properties: Silky needles, mp >300°.
Melting point: mp >300°
Derivative Type: Compd with piperazine
Additional Names: Acefylline piperazine; acepifylline
Trademarks: Dynaphylline (Welcker-Lyster); Etaphylline (Delalande); Etafillina (Delalande)
Properties: Undefined mixture of the 1:1 and 2:1 salts; contains 75-78% theophylline acetic acid and 22-25% anhydrous piperazine.
Therap-Cat: Bronchodilator.
Keywords: Bronchodilator; Xanthine Derivatives.

Acefylline (INN),[1] also known as acetyloxytheophylline, is a stimulant drug of the xanthine chemical class. It acts as an adenosine receptor antagonist. It is combined with diphenhydramine in the pharmaceutical preparation etanautine to help offset diphenhydramine induced drowsiness.[2]

Synthesis

DE 352980 (1922 to E. Merck); Frdl. 14, 1320; S. M. Ride et al., Pharmazie 32, 672 (1977).

File:Acefylline synthesis.svg

Acefylline

  • Use:cardiotonic, diuretic, antispasmodic, bronchodilator
  • Chemical name:1,2,3,6-tetrahydro-1,3-dimethyl-2,6-dioxo-7H-purine-7-acetic acid
  • Formula:C9H10N4O4
  • MW:238.20 g/mol
  • CAS-RN:652-37-9
  • EINECS:211-490-2
  • LD50:1180 mg/kg (M, i.p.); 2733 mg/kg (M, p.o.)
Acepifylline
  • Use:
  • Chemical name:1,2,3,6-tetrahydro-1,3-dimethyl-2,6-dioxo-7H-purine-7-acetic acid compd. with piperazine
  • Formula:C9H10N4O4 • xC4H10N2
  • MW:unspecified
  • CAS-RN:18833-13-1
  • EINECS:242-614-3
Acefylline heptaminol
  • Use:
  • Chemical name:1,2,3,6-tetrahydro-1,3-dimethyl-2,6-dioxo-7H-purine-7-acetic acid compd. with 6-amino-2-methyl-2-heptaminol (1:1)
  • Formula:C9H10N4O3 • C8H19NO
  • MW:367.45 g/mol
  • CAS-RN:59989-20-7
  • EINECS:262-012-4
References
  1. ^ “International Nonproprietary Names for Pharmaceutical Substances (INN). Recommended International Nonproprietary Names (Rec. INN): List 21” (PDF). World Health Organization. Retrieved 29 December 2016.
  2. ^ Zuidema, Jan. (1978). “Biofarmaceutische en farmacokinetische aspecten van theofylline en acefylline”. Thesis (doctoral)–Universiteit van Amsterdam. References
Baisse, J.: Bull. Soc. Chim. Fr. (BSCFAS) 1949, 769.
DE 352 980 (E. Merck; 1922).
Acefylline
Skeletal formula of acefylline
Ball-and-stick model of the acefylline molecule
Clinical data
ATC code
Identifiers
CAS Number
PubChemCID
ChemSpider
UNII
ChEMBL
ECHA InfoCard 100.010.447 Edit this at Wikidata
Chemical and physical data
Formula C9H10N4O4
Molar mass 238.20 g/mol g·mol−1
3D model (JSmol)

////////Acefylline

VNRX-7145

$
0
0

str1

str1

CAS 1842399-68-1

MF C19 H26 B N O7

MW 391.22

2H-1,2-Benzoxaborin-8-carboxylic acid, 3,4-dihydro-2-hydroxy-3-[(1-oxopropyl)amino]-, (2-ethyl-1-oxobutoxy)methyl ester, (3R)-

The VNRX-7145 combination is now in Phase I studies to treat resistant urinary tract infections.

str1

VNRX-7145

PATENT

WO 2015191907

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2015191907

ntibiotics are the most effective drugs for curing bacteria-infectious diseases clinically. They have a wide market due to their advantages of good antibacterial effect with limited side effects. Among them, the beta-lactam class of antibiotics (for example, penicillins, cephalosporins, and carbapenems) is widely used because they have a strong bactericidal effect and low toxicity.

[0005] To counter the efficacy of the various beta-lactams, bacteria have evolved to produce variants of beta-lactam deactivating enzymes called beta-lactamases, and in the ability to share this tool inter- and intra-species. These beta-lactamases are categorized as“serine” or“metallo” based, respectively, on presence of a key serine or zinc in the enzyme active site. The rapid spread of this mechanism of bacterial resistance can severely limit beta-lactam treatment options in the hospital and in the community.

SCHEME 1

SCHEME 2

SCHEME 3

[00390] Alternatively, (II) can be obtained by treatment of (I) with hydrochloric acid (around 3-5 Molar in dioxane) in an alcohol solvent such as methanol, ethanol, or n-butanol at a temperature between room temperature and 120 ºC (SCHEME 4).

SCHEME 4

SCHEME 5

EXAMPLE 62: (R)-2-Hydroxy-3-propionylamino-3,4-dihydro-2H-benzo[e][1,2]oxaborinine-8-carboxylic acid

Step 1. Synthesis of 2-Methoxy-3-[2-propionylamino-2-(2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethyl]-benzoic acid tert-butyl ester.

[00540] Prepared from [(1S)-2-(3-tert-butoxycarbonyl-2-methoxy-phenyl)-1-chloro-ethyl]boronic acid (+) pinanediol ester and propionic acid following the procedure in Step 2 of Example 1. The crude product was purified by flash chromatography on silica gel (25-100% EtOAc/Hexane). ESI-MS m/z 486 (MH)+.

Step 2. Synthesis of (R)-2-Hydroxy-3-propionylamino-3,4-dihydro-2H-benzo[e][1,2]oxaborinine-8-carboxylic acid.

[00541] Prepared from 2-Methoxy-3-[2-propionylamino-2-(2,9,9-trimethyl-3,5-dioxa-4-bora-tricyclo[6.1.1.02,6]dec-4-yl)-ethyl]-benzoic acid tert-butyl ester following the procedure described in Step 3 of Example 1. The crude product was purified by reverse phase preparative HPLC and dried using lyophilization. ESI-MS m/z 264 (MH)+

CLIP

https://cen.acs.org/pharmaceuticals/drug-discovery/Drug-structures-displayed-first-time-in-Orlando/97/web/2019/04?utm_source=Facebook&utm_medium=Social&utm_campaign=CEN

Candidate: VNRX-7145

20190404lnp1-vnrx7145.jpg

Credit: Tien Nguyen/C&EN

Presenter: Christopher John Burns, president and chief executive officer of VenatoRx Pharmaceuticals

Target: β-lactamases

Disease: Resistant urinary tract infections

Reporter’s notes: Having unveiled an antibacterial candidate at last spring’s first time disclosures session, Burns was back with another, this time the molecule can be taken orally. Both VenatoRx (pronounced Ven-a-tor-ix) compounds resuscitate the activity of β-lactam drugs, which make up more than 60% of all antibiotics prescribed. Unfortunately, many bacteria have grown resistant to these antibiotics. The new compounds rescue the old antibacterials by inhibiting β-lactamases, enzymes that chew up the antibiotics. To test the activity of new β-lactamase-targeting compounds, the researchers settled on several “sentinel” bacteria strains. Then to find a candidate with oral bioavailability, the team focused on molecules with low polarity and low molecular weight. They found VNRX-7145, developed as a prodrug in which esterases in the liver clip off the tips of the molecule to reveal the active drug. VNRX-5133, disclosed at last year’s meeting, had to be delivered intravenously along with another IV-antibiotic Cefepime, and targeted serine and metallo β-lactamases. The new oral candidate VNRX-7145 inhibits serine β-lactamases with Ceftibuten as its partner. The VNRX-7145 combination is now in Phase I studies to treat resistant urinary tract infections.

////////////VNRX-7145, VNRX7145, VNRX 7145, Phase I, VenatoRx

CCC(CC)C(=O)OCOC(=O)c1cccc2C[C@H](NC(=O)CC)B(O)Oc12

CCC(CC)C(=O)OCOC(=O)c1cccc2C[C@H](NC(=O)CC)B(O)Oc12

BIIB-095

$
0
0

str1

GCZUIPVRHLYYOG-BEFAXECRSA-N.png

BIIB-095

ROTATION +

1493790-64-9 CAS free form,

1493772-48-7 cas Hcl salt

1493790-65-0, 1496563-32-6 ,SULPHATE

1496563-31-5  SULFATE1;1

1496563-32-6 SULFATE HYDRAT 1;1;1

(2R,5S)-7-methyl-2-[4-methyl-6-[4-(trifluoromethyl)-phenyl]pyrimidin-2-yl]-1 ,7-diazaspiro[4.4]nonan-6-one

1,7-Diazaspiro[4.4]nonan-6-one, 7-methyl-2-[4-methyl-6-[4-(trifluoromethyl)phenyl]-2-pyrimidinyl]-, (2R,5S)-

C20 H21 F3 N4 O, 390.40

  • Originator Biogen
  • Class Analgesics
  • Mechanism of Action Nav1.7 voltage-gated sodium channel inhibitors
  • Phase I Neuropathic pain
  • 29 Mar 2018 Phase-I clinical trials in Neuropathic pain (In volunteers) in United Kingdom (PO) (NCT03454126)
  • 05 Mar 2018 Biogen plans a phase I trial for Pain, including Neuropathic pain (In volunteers) in USA (PO) (NCT03454126)
  • 05 Mar 2018 Preclinical trials in Neuropathic Pain in USA (PO), before March 2018

In March 2018, a randomized, double blind, placebo controlled, single and multiple-ascending dose, dose-escalation phase I study ( NCT03454126; 255HV101; 2017-003982-90) was initiated in the UK in healthy subjects (expected n = 80) to evaluate the safety, tolerability and pharmacokinetics of BIIB-095. At that time, the trial was expected to complete in December 2018

Biogen is developing BIIB-095, a voltage-gated sodium channel 1.7 inhibitor, for the potential oral treatment of neuropathic pain [2027279], [2027426]. In March 2018, a phase I trial was initiated in healthy subjects

Biogen is developing oral agent BIIB-095 for the treatment of chronic pain, including neuropathic pain. A phase I clinical trial is under way in healthy volunteers.

The compound was first claimed in WO2013175205 , for treating schizophrenia, assigned to subsidiary Convergence Pharmaceuticals Limited , naming some of the inventors. This might present the structure of BIIB-095 , a voltage-gated sodium channel 1.7 inhibitor, being developed by Biogen for the oral treatment of neuropathic pain; in March 2018, a phase I trial was initiated in healthy subjects.

PATENT

WO2013175205

WO 2013175206

US 20150119404

https://patents.google.com/patent/US20150119404

Patent

WO-2019067961

https://patentscope2.wipo.int/search/en/detail.jsf;jsessionid=4E8EDA900F4ACD794E922F827F6F20D5?docId=WO2019067961&tab=PCTDESCRIPTION&office=&prevFilter=&sortOption=Pub+Date+Desc&queryString=&recNum=7931&maxRec=74545645

Novel salts (citrate, mesylate, hydrosulfate, saccharinate and oxalate) forms of 7-methyl-2-[4-methyl-6-[4-(trifluoromethyl)-phenyl]pyrimidin-2-yl]-1,7-diazaspiro[4.4]nonan-6-one, processes for their preparation and compositions comprising them are claimed. Also claimed are their use for treating diseases and conditions mediated by modulation of voltage-gated sodium channels.

Voltage-gated sodium channels are responsible for the initial phase of the action potential, which is a wave of electrical depolarisation usually initiated at the soma of the neuron and propagated along the axon to the terminals. At the terminals, the action potential triggers the influx of calcium and the release of neurotransmitter. Drugs, such as lidocaine, that block voltage-gated sodium channels are used as local anaesthetics. Other sodium channel blockers, such as lamotrigine and carbamazepine are used to treat epilepsy. In the latter case, partial inhibition of voltage-gated sodium channels reduces neuronal excitability and reduces seizure propagation. In the case of local anaesthetics, regional block of sodium channels on sensory neurons prevents the conduction of painful stimuli. A key feature of these drugs is their state-dependent mechanism of action. The drugs are thought to stabilise an inactivated conformation of the channel that is adopted rapidly after the channel opens. This inactivated state provides a refractory period before the channel returns to its resting (closed) state ready to be reactivated. As a result, state-dependent sodium channel blockers inhibit the firing of neurons at high frequency, for example in response to painful stimuli, and will help to prevent repetitive firing during periods of prolonged neuronal depolarisation that might occur, for example, during a seizure. Action potentials triggered at lower frequencies, for example in the heart, will not be significantly affected by these drugs, although the safety margin differs in each case, since at high enough concentrations each of these drugs is capable of blocking the resting or open states of the channels.

The voltage-gated sodium channel family is made up of 9 subtypes, four of which are found in the brain, NaV1.1 , 1.2, 1.3 and 1.6. Of the other subtypes, NaV1.4 is found only in skeletal muscle, NaV1.5 is specific to cardiac muscle, and NaV1.7, 1.8, and 1.9 are found

predominantly in sensory neurons. The hypothesised binding site for state-dependent sodium channel blockers is the local anaesthetic (LA) binding site in the inner vestibule of the pore on transmembrane S6 of domain IV. Critical residues are located in a highly conserved region among the different subtypes, thus presenting a challenge for the design of new subtype selective drugs. Drugs such as lidocaine, lamotrigine and carbamazepine do not distinguish between the subtypes. However, selectivity can be achieved, and can be further enhanced functionally, as a result of the different frequencies at which the channels operate.

Drugs that block voltage-gated sodium channels in a state-dependent manner are also used in the treatment of bipolar disorder, either to reduce symptoms of mania or depression, or as mood stabilisers to prevent the emergence of mood episodes. Clinical and preclinical evidence also suggests that state-dependent sodium channel blockers may help to reduce the symptoms of schizophrenia. For example, lamotrigine has been shown to reduce symptoms of psychosis induced by ketamine in healthy human volunteers, and furthermore, studies in patients suggest that the drug can augment the antipsychotic efficacy of some atypical antipsychotic drugs, such as clozapine or olanzapine. It is hypothesised that efficacy in these psychiatric disorders may result in part from a reduction of excessive glutamate release. The reduction in glutamate release is thought to be a consequence of sodium channel inhibition in key brain areas, such as the frontal cortex. However, interaction with voltage-gated calcium channels may also contribute to the efficacy of these drugs.

WO 2013/175205 (Convergence Pharmaceuticals Limited) describes (2R,5S)-7-methyl-2-[4-methyl-6-[4-(trifluoromethyl)-phenyl]pyrimidin-2-yl]-1 ,7-diazaspiro[4.4]nonan-6-one hydrochloride, sulfuric acid salt and sulfuric acid salt hydrate which are claimed to be modulators of voltage-gated sodium channels. The object of the invention is to identify alternative salts of said compound which have advantageous properties.

Example 1

(2R,5S)-7-Methyl-2-[4-methyl-6-[4-(trifluoromethyl)-phenyl]pyrimidin-2-yl]-1,7-diazaspiro[4.4]nonan-6-

To a solution of (2R,5S)-7-methyl-2-[4-methyl-6-[4-(trifluoromethyl)-phenyl]pyrimidin-2-yl]-1 ,7-diazaspiro[4.4]nonan-6-one (which may be prepared in accordance with the procedure described in Example 1 of WO 2013/175205) (4.45g, 0.0114 mol) dissolved in absolute ethanol (66.82 ml, 15 vol) at 45 °C was added a solution of citric acid in ethanol (1 M, 1.05 equiv. 12 ml) over a period of 2-3 minutes. The solution was aged at 45 °C for a period of 1 hour. After 30 minutes a seed of citrate salt (0.1 wt%) was added and the mixture allowed to cool over approximately 2 hours and mature for 18 hours at ambient temperature (approximately 10-15 °C). Following maturation the salt was noted to be a very thick suspension (white) that required mobilisation with 20 ml additional ethanol and a further maturation period of 2 hours at ambient temperature. Filtration was carried out under vacuum and the vessel and cake rinsed with 15 ml ethanol. The de-liquored cake was dried further in a vacuum oven at 50 °C to provide 6.0 g of crystalline white solid (91 % yield).

H NMR (400MHz, DMSO-D6): δΗ 1.90-2.05 (2H, m), 2.10-2.20 (2H, m,), 2.20-2.30 (1 H, m), -2.50 (1 H, m, partially masked by solvent)), 2.55-2.68 (4H, m), 2.56 (3H, s), 2.79 (3H, s),

3.28-3.40 (2H, m), 4.79 (1 H, t, J= 8.0 Hz), 7.92 (2H, d, J = 8.4 Hz), 8.03 (1 H, s), 8.45 (2H, d, J= 8.8Hz) ppm, (exchangeables not reported)

Characterisation of Example 1

The XRPD of Example 1 is presented in FIG. 1 and the DSC/TGA of Example 1 is presented in FIG. 2. The citrate salt of Example 1 displayed the following characteristics:

1 endotherm onset: 171.82°C

peak maximum: 174.55°C

There was an endotherm post the main endotherm.

There was no weight reduction until ca 168°C had been reached. The weight reduction commenced with the start of the main endotherm and coincided with the endotherm post the main endotherm which indicated that this thermal event was the onset of compound decomposition and loss of citric acid. Thermal events >220°C were due to compound decomposition.

The XPRD data in FIG. 1 demonstrated that under different extremes of humidity indicate a stable crystalline form of the citrate salt of Example 1 with no tendency to form hydrates. This is supported by DSC/TGA data in FIG. 2 which show clear transitions and no evidence of solvates.

Aqueous solubility of the citrate salt (Example 1) = 22mg/ml (25°C).

Example 2

(2R,5S)-7-Methyl-2-[4-methyl-6-[4-(trifluoromethyl)-phenyl]pyrimidin-2-yl]-1,7-diazaspiro[4.4]nonan-6-one ) salt (E2)

To a solution of (2R,5S)-7-methyl-2-[4-methyl-6-[4-(trifluoromethyl)-phenyl]pyrimidin-2-yl]-1 ,7-diazaspiro[4.4]nonan-6-one (which may be prepared in accordance with the procedure described in Example 1 of WO 2013/175205) (4.45g, 0.0114 mol) dissolved in absolute ethanol (66.82 ml, 15 vol) at 45 °C was added a solution of methanesulfonic acid in ethanol (1 M, 1.05 equiv. 12 ml) over a period of 2-3 minutes. The solution was aged at 45 °C for a period of 1 hour. After 10 minutes nucleation and gradual crystallisation was noted to afford a thick mixture. Additional ethanol was added (10 ml) to mobilise the suspension that was then allowed to cool over approximately 2 hours and mature for 18 hours at ambient temperature (approximately 10-15 °C). Following maturation the salt was noted to be a thin, mobile suspension (white) that was filtered under vacuum and the vessel and cake rinsed with 15 ml ethanol. The de-liquored cake was dried further in a vacuum oven at 50 °C to provide 4.0 g of crystalline white solid (72% yield).

H NMR (400MHz, DMSO-D6): δΗ 2.1-2.45 (4H, m), 2.27 (3H, s), 2.50-2.75 (2H, m), 2.61 (3H, s), 2.86 (3H, s), 3.35-3.50 (2H, m), 5.20 (1 H, t, J = 8 Hz), 7.96 (2H, d, J = 8.8 Hz), 8.17 (1 H, s), 8.51 (2H, d, J = 8.4Hz), 9.45 (1 H, br), 10.16 (1 H, br) ppm.

Characterisation of Example 2

The XRPD of Example 2 is presented in FIG. 3 and the DSC/TGA of Example 2 is presented in FIG. 4. The DSC thermograph of the methanesulfonate (mesylate) (Example 2) displayed the following characteristics:

One distinct endotherm onset: 247.34°C

peak maximum: 250.34°C

The TGA thermograph showed no weight reduction until ca 250°C had been reached. The weight reduction commenced with the start of the main endotherm and indicated that this thermal event was the onset of compound decomposition. There is no evidence of entrapped solvents or water.

The XPRD data in FIG. 3 demonstrated that under different extremes of humidity indicate a stable crystalline form of the mesylate salt of Example 2 with no tendency to form hydrates. This is supported by DSC/TGA data in FIG. 4 which show clear transitions and no evidence of solvates.

Aqueous solubility of the mesylate salt (Example 2) = 65mg/ml (25°C).

Example 3

Preparation of (2R,5S)-7-methyl-2-(4-methyl-6-(4-(trifluoromethyl)phenyl)pyrimidin-2-yl)-1,7-diazaspiro[4.4]nonan-6-one hydrosulfate single crystals: 25.0 mg of (2R,5S)-7-Methyl-2-(4-methyl-6-(4-(trifluorome

one hydrosulfate was added to 4 mL vial. 1.000 mL of anhydrous EtOH was added, and the sample was filtered. Anhydrous hexanes were added dropwise until the solution neared the precipitation point. The vial was sealed and left undisturbed for 24 hr, after which time a crop of single crystals was evident. The sample was sent for single crystal analysis and confirmed as the anhydrous (2R,5S)-7-Methyl-2-(4-methyl-6-(4-(trifluoromethyl)phenyl)pyrimidin-2-yl)-1 ,7-diazaspiro[4.4]nonan-6-one hydrosulfate form (FIGs. 5A-5B).

Example 4

Preparation of (2R,5S)-7-methyl-2-(4-methyl-6-(4-(trifluoromethyl)phenyl)pyrimidin-2-yl)-1,7-diazaspiro[4.4]nonan-6-one freebase: 8.00 g of (2R,5S)-7-Methyl-2-(4-methyl-6-(4-(trifluoromethyl)phenyl)pyrimidin-2-yl)-1 ,7-diazaspiro[4.4]nonan-6-one hydrosulfate (JM Lot R-2017-4323 D 301) was added to a 1 L Erlenmeyer flask and suspended and stirred vigorously in 400 mL of THF. 20% K2C03 (250 mL) was added and dissolved. The mixture was transferred to 1 L sep. funnel. 100 mL EtOAc was added and the aqueous and organic layers were separated. The aqueous layer was re-extracted with 50 mL of EtOAc and the combined organics were back-extracted with brine (100 mL) and water (100 mL). Due to fairly poor separation, a significant quantity of MgSCU was required to dry the solution. The solution was reduced via Rotavap (45 °C) to -50 mL, transferred to a 100 mL RB flask, reduced down to -10 mL, transferred to 20 mL scintillation vial and continued to be reduced to a thick oil. The oil was left on the Rotavap for another hour and a “wet” solid was obtained. Loosened solids on the bottom of the vial were left on the Rotavap for 1 hr with no heat applied to obtain a chunky solid. The contents was transferred to a mortar and pestle, ground to powder and fine granules, placed back in a 20 mL scintillation vial and left on a Rotavap overnight to obtain a dry solid (5.1 g). The XRPD pattern of (2R,5S)-7-Methyl-2-(4-methyl-6-(4-(trifluoromethyl)phenyl)pyrimidin-2-yl)-1 ,7-diazaspiro[4.4]nonan-6-one freebase is shown in FIG. 6.

Example 5

Preparation of (2R,5S)-7-methyl-2-(4-methyl-6-(4-(trifluoromethyl)phenyl)pyrimidin-2-yl)-1,7-diazaspiro[4.4]nonan-6-one saccharinate: 199.7 mg of (2R,5S)-7-Methyl-2-(4-

methyl-6-(4-(trifluoromethyl)phenyl)pyrirnidin-2-yl)-1 ,7-diazaspiro[4.4]nonan-6-one free base (0.5115 mmol) was dissolved in 4.2 mL of 2-Me-THF. 98.1 mg of saccharin (0.5106 mmol) was dissolved in 4.2 mL of 2-Me-THF. Saccharin was added to the freebase, and after 15 seconds the mixture began to precipitate and solidify. 10 mL of 2-Me-THF was added and stirred at max rpm as to provide a thick white suspension in 10 min. The suspension was filtered, air dried under vacuum for 10 min on frit, then dried under a stream of nitrogen for 30 min resulting in 215 mg of white solid product. The XRPD pattern for (2R,5S)-7-Methyl-2-(4-methyl-6-(4-(trifluoromethyl)phenyl)pyrimidin-2-yl)-1 ,7-diazaspiro[4.4]nonan-6-one saccharinate is shown in FIG. 7.

Example 6

Preparation of (2R,5S)-7-methyl-2-(4-methyl-6-(4-(trifluoromethyl)phenyl)pyrimidin-2-yl)-1,7-diazaspiro[4.4]nonan-6-one oxalate: 403 mg of (2R,5S)-7-Methyl-2-(4-methyl-6-(4-(trifluoromethyl)phenyl)pyrimidin-2-yl)-1 ,7-diazaspiro[4.4]nonan-6-one freebase was dissolved in 4.03 mL EtOH. 1.000 mL of this solution was added to a 4 mL vial. 23.8 mg of oxalic acid was dissolved in 1.000 mL of EtOH and added dropwise to the stirring (2R,5S)-7-Methyl-2-(4-methyl-6-(4-(trifluoromethyl)phenyl)pyrimidin-2-yl)-1 ,7-diazaspiro[4.4]nonan-6-one freebase solution. After 5 min, a white precipitate was evident and 2.000 mL of EtOH was added to the slurry to aid stirring. The resulting suspension was stirred overnight. The following day the suspension was filtered and dried on a frit under vacuum for 10 min yielding 106 mg of white solid. The XRPD pattern for (2R,5S)-7-Methyl-2-(4-methyl-6-(4-(trifluoromethyl)phenyl)pyrimidin-2-yl)-1 ,7-diazaspiro[4.4]nonan-6-one oxalate is shown in FIG. 8.

Example 7

The single crystal structural information and refinement parameters for (2R,5S)-7-Methyl-2-(4-methyl-6-(4-(trifluoromethyl)phenyl)pyrimidin-2-yl)-1 ,7-diazaspiro[4.4]nonan-6-one hydrosulfate are shown in Table 1.

Table 1.

Largest peak, hole / e A-3 0.363, -0.264

The most prominent XRPD diffraction peaks were (2Θ): 7.8±0.2°, 8.1±0.2°, 12.6±0.2°, 14.3±0.2°, 16.5±0.2°, 18.5±0.2°, 19.6±0.2°, 24.8±0.2° and 25.3±0.2°.

PATENTS

US2018360833NOVEL PYRIMIDINYL-DIAZOSPIRO COMPOUNDS2018-06-27

Patent ID Title Submitted Date Granted Date
US2017304303 Novel Pyrimidinyl-DiazoSpiro Compounds 2017-07-11
US9737536 Novel Pyrimidinyl-DiazoSpiro Compounds 2016-05-25 2016-09-15
US2016184306 Novel Pyrimidinyl-DiazoSpiro Compounds 2016-02-15 2016-06-30
US9309254 NOVEL COMPOUNDS 2013-05-22 2015-04-30
US9376445 NOVEL COMPOUNDS 2013-05-22 2015-06-18

////////////////BIIB-095, BIIB095, BIIB 095, PHASE 1

CC1=NC(=NC(=C1)C2=CC=C(C=C2)C(F)(F)F)C3CCC4(N3)CCN(C4=O)C

Viewing all 2909 articles
Browse latest View live


<script src="https://jsc.adskeeper.com/r/s/rssing.com.1596347.js" async> </script>