Quantcast
Channel: Preclinical drugs – New Drug Approvals
Viewing all 87 articles
Browse latest View live

SEN 826

$
0
0

figure

SEN 826
CAS 1160833-51-1
C25 H31 N5 O, 417.55
Methanone, [1-[3-(1-methyl-1H-benzimidazol-2-yl)phenyl]-4-piperidinyl](4-methyl-1-piperazinyl)-
CAS HBr SALT 1612250-71-1

WO2009074300 product patent

Russell John Thomas, Mohr Gal.La Pericot, Giacomo Minetto, Annette Cornelia Bekker, Pietro Ferruzzi
Applicant Siena Biotech S.P.A.
Image result for Siena Biotech S.P.A.
Siena Biotech S.p.A. operates as a drug discovery and development company which develops a portfolio of disease modifying small molecule therapeutics for oncology and neurodegenerative diseases. Its products include blood-brain barrier penetrant compounds, which are in pipeline, for the treatment of brain cancers and peripheral tumors capable of metastasizing to the brain; clinical candidates for Alzheimer’s disease; and SEN196, a Sirtuin 1 inhibitor against Huntington disease. The company also provides contract research services, drug discovery, integrated chemistry, in-vitro technologies, and preclinical technologies. Siena Biotech S.p.A. has a strategic partnership with Aptuit Inc. The company was founded in 2000 and is based in Siena, Italy. Siena Biotech S.p.A operates as a subsidiary of THERAMetrics holding AG
Russell Thomas

Russell Thomas

https://www.linkedin.com/in/russell-thomas-0317464/

PLEASE MAIL ME AT amcrasto@gmail.com if picture is a mistake or cal +919323115463

The SMO receptor mediates Hedgehog (Hh) signaling critical to development, differentiation, growth, and cell migration. In normal conditions, activation of the pathway is induced by binding of specific endogenous ligands (i.e., Sonic Hh) to its receptor Patched (Ptch), which in turns reverts the Ptch inhibitory effect on SMO. SMO activation ultimately determines specific target genes activation through a family of three transcription factors, Gli1, Gli2 and Gli3.
Although Hh signaling is significantly curtailed in adults, it retains functional roles in stem cell maintenance, and aberrant Hh signaling has been described in a range of tumours.
Mutational inactivation of the inhibitory pathway components results in a constitutive ligand-independent activation seen in tumours such as basal cell carcinoma (BCC) and medulloblastoma. Ligand-dependent activation is seen in tumours such as prostate cancer, pancreatic cancer, gastrointestinal malignancies, melanoma, gliomas, breast cancer, ovarian cancer, leukemia, and B-cell lymphomas. A significant body of evidence supports the conclusion that SMO receptor antagonism will block the downstream signaling events.
As part of a program to address unmet medical need with regard to tumours in the CNS, Siena Biotech has designed and investigated selective antagonists of the SMO receptor. The newly designed API development candidate SEN826 1  is part of a group of potent antagonists of the Hedgehog pathway.
SYNTHESIS

PATENT

WO 2009074300

Figure imgf000025_0001

Figure imgf000019_0002

Figure

The synthesis starts with the formation of the 2-arylbenzimidazole derivative 6 which can be carried out starting from N-methylphenylenediamine 2 (Method A; blue path in Scheme 1) or employing o-phenylenediamine 4 in the ring closure reaction followed by N-methylation (Method B; orange path in Scheme 1). Sodium hydrogen sulfite is used to promote the condensation of the corresponding o-phenylenediamine with the Br-aromatic aldehyde 3.(6b) The next step is the coupling of the aryl bromide with isonipecotic ethyl ester in Buchwald conditions. After acidic hydrolysis with HCl under microwave irradiation, the final amide 1 was synthesized with CDI as coupling agent.

PAPER

A Scalable Route to the SMO Receptor Antagonist SEN826: Benzimidazole Synthesis via Enhanced in Situ Formation of the Bisulfite–Aldehyde Complex

Process Chemistry Unit, Siena Biotech SpA, 53100 Siena, Italy
Compound Management & Analysis Unit, Siena Biotech SpA, 53100 Siena, Italy
Org. Process Res. Dev., 2014, 18 (6), pp 699–708
Abstract Image

A practical and scalable route to the SMO antagonist SEN826 1 is described herein, including the discussion of an alternative approach to the synthesis of the target molecule. The optimized route consists of five chemical steps. A new and efficient access to the key intermediate 6 via the bisulfite–aldehyde complex was developed, significantly enhancing the yields and reducing costs. As a result, a synthetic procedure for preparation of multihundred gram quantities of the final product has been developed.

1 as hydrobromide salt. Yield: 71%.
UPLC–MS: tR = 1.24 min; m/z = 418 [M + 1]+.
HRMS calcd for C25H33N5O [M + 1]+ 418.26069, found 418.26075.
HPLC: tR = 5.99 min; purity 99.1%.
1H NMR (400 MHz DMSO-d6): δ 9.80 (broad, 1H), 7.89 (m, 1H), 7.77 (m, 1H), 7.55–7.45 (m, 3H), 7.38 (s, 1H), 7.24 (m, 2H), 4.48–4.15 (m, 2H), 3.96 (s, 3H), 3.86 (m, 2H), 3.55–3.15 (m, 3H), 3.10–2.82 (m, 6H), 2.81 (s, 3H), 1.76–1.57 (m, 4H).
13C NMR (100 MHz DMSO-d6): δ 173.5, 152.3, 151.5, 135.1, 135.0, 130.5, 126.2, 125.6, 125.3, 119.9, 119.1, 117.1, 116.5, 113.0, 53.2, 48.2, 42.7, 38.8, 37.4, 33.1, 28.2.
Water content (KF): 3.5 wt %.
Pd content (ICP-MS): 128 ppm.
Bromine content (ionic exchange LC): 20 wt % (1.2 equiv).
str1 str2
/////////////////

Filed under: Preclinical drugs, Uncategorized

Debio-1452

$
0
0

Image result for Debio-1452

Debio-1452, AFN 1252

AFN-1252; UNII-T3O718IKKM; API-1252; CAS 620175-39-5; CHEMBL1652621; (E)-N-methyl-N-((3-methylbenzofuran-2-yl)methyl)-3-(7-oxo-5,6,7,8-tetrahydro-1,8-naphthyridin-3-yl)acrylamide

  • MFC22 H21 N3 O3
  • 2-Propenamide, N-methyl-N-[(3-methyl-2-benzofuranyl)methyl]-3-(5,6,7,8-tetrahydro-7-oxo-1,8-naphthyridin-3-yl)-, (2E)-
  •  MW375.42
  • Phase 2, clinical trials for the oral treatment of staphylococcal infections, including hospital and community-acquired MRSA and acute bacterial skin and skin structure infections
  • Qualified Infectious Disease Product designation

GlaxoSmithKline plc INNOVATOR

Image result

Debiopharm SA,

Image result for DEBIOPHARM

Image result for Affinium

Melioidosis, Enoyl ACP reductase Fabl inhibitor

Debio-1452, a novel class fatty acid biosynthesis (FAS) II pathway inhibitor, was studied in phase II clinical trials for the oral treatment of staphylococcal infections, including hospital and community-acquired MRSA and acute bacterial skin and skin structure infections. Debiopharm is developing oral and IV formulations of a prodrug of Debio-1452, Debio-1450.

Infections caused by or related to bacteria are a major cause of human illness worldwide. Unfortunately, the frequency of resistance to standard antibacterials has risen dramatically over the last decade, especially in relation to Staphylococcus aureus. For example, such resistant S. aureus includes MRSA, resistant to methicillin, vancomycin, linezolid and many other classes of antibiotics, or the newly discovered New Delhi metallo-beta-lactamase- 1 (NDM-1) type resistance that has shown to afford bacterial resistant to most known antibacterials, including penicillins, cephalosporins, carbapenems, quinolones and fluoroquinolones, macrolides, etc. Hence, there exists an urgent, unmet, medical need for new agents acting against bacterial targets..

In recent years, inhibitors of Fabl, a bacterial target involved in bacterial fatty acid synthesis, have been developed and many have been promising in regard to their potency and tolerability in humans, including a very promising Fabl inhibitor, (E)-N-methyl-N-((3-methylbenzofuran-2-yl)methyl)-3-(7-oxo-5,6,7,8-tetrahydro-l,8-naphthyridin-3-yl)acrylamide. This compound, however, has been found to be difficult or impracticable to formulate into acceptable oral and parenteral (e.g., intravenous or subcutaneous) formulations, and has marked insolubility, poor solution stability, and oral bioavailability. Much effort, over a decade or more, has been expended to design and synthesize an alternative compound that retains the significant inhibition of Fabl upon administration, but has improved physical and chemical characteristics that finally allow for practical oral and parenteral formulations. Up to now, no such compound has been identified that has adequate stability in the solid state, in aqueous solutions, together with excellent oral bioavailability that is necessary for oral and/or a parenteral administration, and is capable of being formulated into an oral and/or intravenous or intramuscular drug product using practical and commonly utilized methods of sterile formulation manufacture.

Debio-1452 is expected to have high potency against all drug-resistant phenotypes of staphylococci, including hospital and community-acquired MRSA.

Affinium obtained Debio-1452, also known as API-1252, through a licensing deal with GlaxoSmithKline. In 2014, Debiopharm acquired the product from Affinium.

In 2013, Qualified Infectious Disease Product designation was assigned to the compound for the treatment of acute bacterial skin and skin structure infections (ABSSSI).

Image result for Debio-1452

Image result for Debio-1452

AFN-1252.png

SYNTHESIS

Heck coupling of 6-bromo-3,4-dihydro-1,8-naphthyridin-2-one with t-butyl acrylate in the presence of Pd(OAc)2, DIEA and P(o-tol)3  in propionitrile/DMF or acetonitrile/DMF affords naphthyridinyl-acrylate,

Whose t-butyl ester group is then cleaved using TFA in CH2Cl2 to furnish, after treatment with HCl in dioxane, 3-(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-3-yl)acrylic acid hydrochloride

SEE BELOW………

Finally, coupling of acid with N-methyl-N-(3-methylbenzofuran-2-ylmethyl)amine using EDC, HOBt and DIEA in DMF provides the target AFN-1252

Preparation of N-methyl-N-(3-methylbenzofuran-2-ylmethyl)amine :

Chlorination of 3-methylbenzofuran-2-carboxylic acid  with (COCl)2 and catalytic DMF, followed by condensation with CH3NH2 in CH2Cl2 yields the corresponding benzofuran-2-carboxamide,

Which is then reduced with LiAlH4 in THF to furnish N-methyl-N-(3-methylbenzofuran-2-ylmethyl)amine.

CONTD……..

Reduction of 2-aminonicotinic acid  with LiAlH4 in THF gives (2-amino-3-pyridinyl)methanol ,

which upon bromination with Br2 in AcOH yields (2-amino-5-bromo-3-pyridinyl)methanol hydrobromide.

Substitution of alcohol  with aqueous HBr at reflux provides the corresponding bromide,

which undergoes cyclocondensation with dimethyl malonate  in the presence of NaH in DMF/THF to furnish methyl 6-bromo-2-oxo-1,2,3,4-tetrahydro-1,8-naphthyridine-3-carboxylate.

Hydrolysis of ester with NaOH in refluxing MeOH, followed by decarboxylation in refluxing HCl leads to 6-bromo-3,4-dihydro-1,8-naphthyridin-2-one

PATENT

US-20170088822

Image result for Aurigene Discovery Technologies Ltd

Aurigene Discovery Technologies Ltd

Novel co-crystalline polymorphic form of a binary enoyl-acyl carrier protein reductase (FabI) and FabI inhibitor ie AFN-1252. The FabI was isolated from Burkholderia pseudomallei (Bpm). The co-crystal is useful for identifying an inhibitor of FabI, which is useful for treating BpmFabI associated disease ie melioidosis. Appears to be the first patenting to be seen from Aurigene Discovery Technologies or its parent Dr Reddy’s that focuses on BpmFabI crystal; however, see WO2015071780, claiming alkylidine substituted heterocyclyl derivatives as FabI inhibitors, useful for treating bacterial infections. Aurigene was investigating FabI inhibitors, for treating infectious diseases, including bacterial infections such as MRSA infection, but its development had been presumed to have been discontinued since December 2015; however, publication of this application would suggest otherwise.

WO2015071780

PATENTS

US 20060142265

http://www.google.co.in/patents/US20060142265

PATENT

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2013190384&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=PCTDescription

Patent ID Patent Title Submitted Date Granted Date
US8901105 Prodrug derivatives of (E)-N-methyl-N-((3-M ethylbenzofuran-2-yl)methyl)-3-(7-oxo-5, 6, 7, 8-tetrahydro-1, 8-naphthyridin-3-yl)acrylamide 2013-08-26 2014-12-02
US2015065415 PRODRUG DERIVATIVES OF (E)-N-METHYL-N-((3-METHYLBENZOFURAN-2-YL)METHYL)-3-(7-OXO-5, 6, 7, 8-TETRAHYDRO-1, 8-NAPHTHYRIDIN-3-YL)ACRYLAMIDE 2014-11-06 2015-03-05
Patent ID Patent Title Submitted Date Granted Date
US7049310 Fab I inhibitors 2004-07-29 2006-05-23
US7250424 Fab I inhibitors 2006-06-01 2007-07-31
US7879872 Compositions comprising multiple bioactive agents, and methods of using the same 2006-06-29 2011-02-01
US2009042927 Salts, Prodrugs and Polymorphs of Fab I Inhibitors 2009-02-12
US7741339 Fab I Inhibitors 2009-09-03 2010-06-22
US8153652 Fab I Inhibitors 2011-04-28 2012-04-10
US2012010127 Compositions Comprising Multiple Bioactive Agents, and Methods of Using the Same 2012-01-12
US2013281442 Compounds for Treatment of Bovine Mastitis 2011-06-13 2013-10-24
US2013150400 SALTS, PRODRUGS AND POLYMORPHS OF FAB I INHIBITORS 2012-08-09 2013-06-13
US2014309191 SALTS, PRODRUGS AND POLYMORPHS OF FAB I INHIBITORS 2013-11-08 2014-10-16

////////////Debio-1452, AFN 1252,AFN-1252, UNII-T3O718IKKM, API-1252, 620175-39-5, PRECLINICAL, Phase 2, Qualified Infectious Disease Product designation

CC1=C(OC2=CC=CC=C12)CN(C)C(=O)C=CC3=CC4=C(NC(=O)CC4)N=C3


Filed under: Phase2 drugs, Preclinical drugs, QIDP, Uncategorized Tagged: 620175-39-5, AFN 1252, API-1252, Debio-1452, phase 2, preclinical, Qualified Infectious Disease Product designation, UNII-T3O718IKKM

Process Development and Good Manufacturing Practice Production of a Tyrosinase Inhibitor via Titanium-Mediated Coupling between Unprotected Resorcinols and Ketones

$
0
0

(S)-4-(2,4-Dihydroxyphenyl)-N-(1-phenylethyl)piperidine-1-carboxamide (1)

In a………………….. to yield crude 1 (3.51 kg, 77%, 97.7 A% purity). Recrystallization: In a 100 L double jacketed reactor were charged crude 1 (3.51 kg, 10.31 mol, 1.0 equiv), iPrOH (27.0 L, 7.5 vol), AcOH (74.1 g), and water (27.0 L, 7.5 vol). The suspension was warmed to reflux and turned to a solution after 30 min of reflux. Heating was stopped, and the reaction medium was allowed to cool to 23 °C over 20 h. The suspension was filtered through a 25 μm filter medium; the cake was washed with a mixture of water (3.6 L) and AcOH (7.3 g) and the solid collected and dried under vacuum at 45 °C for 48 h to yield 1 (2.86 kg, 81%, 98.5 A% purity).
1H NMR (400 MHz, DMSO-d6): δ 9.11 (s, 1H), 8.96 (s, 1H), 7.30–7.31 (m, 4), 7.19–7.20 (m, 1H), 6.79 (d, J = 8.3 Hz, 2H), 6.7 (d, J = 7.9 Hz, 2H), 6.28 (d, J = 2.4 Hz, 1H), 6.16 (dd, J = 8.3, 2.4 Hz, 1H), 4.85–4.87 (m, 1 H), 4.13 (d, J = 12.9 Hz, 2H), 2.85 (t, J = 11.9 Hz, 1H), 2.70 (t, J = 12.7 Hz, 2H), 1.64 (d, J = 12.1 Hz, 2H), 1.40–1.41 (m, 5H).
13C NMR (101 MHz, DMSO-d6) δ 156.6, 156.0, 155.2, 146.3, 127.9, 126.7, 126.1, 125.9, 122.5, 106.0, 102.4, 49.3, 44.4, 34.7, 31.8, 31.7, 22.9;
mp: 200–201 °C;
HRMS (m/z, ES+) for C20H25N2O3 (M + H)+ calcd. 341.1865, measd. 341.1859.

Process Development and Good Manufacturing Practice Production of a Tyrosinase Inhibitor via Titanium-Mediated Coupling between Unprotected Resorcinols and Ketones

Nestlé Skin Health R&D, 2400 Route des colles BP 87, 06902 Sophia-Antipolis Cedex, France
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.7b00036

ACS Editors’ Choice – This is an open access article published under an ACS AuthorChoice License, which permits copying and redistribution of the article or any adaptations for non-commercial purposes.

Thibaud Gerfaud

Thibaud Gerfaud

Team Leader Process Chemistry

Nestlé Skin Health Logo

Boiteau Jean-Guy

Boiteau Jean-Guy

Head of Process Research & Development

Nestlé Skin Health

Nestlé Skin Health Logo

Abstract

Abstract Image

A concise and economically attractive process for the synthesis of a novel tyrosinase inhibitor has been developed and implemented on a multikilogram scale under GMP. A major achievement to the success of the process is the development of a direct coupling between free resorcinol and ketone. First developed under basic conditions, this coupling has been turned to a novel titanium(IV) mediated process allowing good selectivity, easy isolation, and high atom efficiency. Other key steps feature an alkene reduction by palladium catalyzed transfer hydrogenation and a urea formation using N,N′-disuccinimidyl carbonate as the carbonyl source. This route allowed us to produce kilogram batches of the candidate to support preclinical and clinical studies.

Figure

Boiteau, J.-G.; Bouquet, K.; Talano, S.; Millois-Barbuis, C. Patent WO 2010/063774 A1, 2010.

More………………

str1

Cas 1228342-28-6
MF C20 H24 N2 O3,
MW  340.42
1-Piperidinecarboxamide, 4-(2,4-dihydroxyphenyl)-N-[(1S)-1-phenylethyl]-
  • 4-(2,4-Dihydroxyphenyl)-N-[(1S)-1-phenylethyl]-1-piperidinecarboxamide
  • 4-(2,4-Dihydroxyphenyl)piperidine-1-carboxylic acid N-((S)-1-phenylethyl)amide
Inventors Jean-Guy Boiteau , Karine Bouquet , Sandrine Talano , Barbuis Corinne Millois
Applicant Galderma Research & Development

Hyperpigmentation disorders such as melasma are characterized by an increase in melanin synthesis which accumulates in the epidermis and is responsible for a darkening of the skin. Melanogenesis occurs in the basal layer of the epidermis into specific organelles of the melanocytes called melanosomes.

A detailed analysis of the biosynthetic pathway reveals that tyrosinase is a key enzyme in melanogenesis and is responsible for the oxidation of tyrosine into DOPA (3,4-dihydroxyphenylalanine) and DOPA quinone.

It is a melanogenesis inhibitor working through the inhibition of tyrosinase (IC50 = 0.1 μM on normal human epidermal melanocytes) currently under development at Nestlé Skin Health R&D for the topical treatment of hyperpigmentation disorders. REF 1-5

WO 2010063774

Novel 4- (azacycloalkyl)benzene-l ,3-diol compounds as tyrosinase inhibitors, process for the preparation thereof and use thereof in human medicine and in cosmetics

The invention relates to novel 4- (azacycloalkyl) benzene-1, 3-diol compounds as industrial and useful products. It also relates to the process for the preparation thereof and to the use thereof, as tyrosinase inhibitors, in pharmaceutical or cosmetic compositions for use in the treatment or prevention of pigmentary disorders.

Skin pigmentation, in particular human skin pigmentation, is the result of melanin synthesis by dendritic cells, melanocytes. Melanocytes contain organelles called melanosomes which transfer melanin into the upper layers of keratinocytes which are then transported to the surface of the skin through differentiation of the epidermis (Gilchrest BA, Park HY, Eller MS, Yaar M, Mechanisms of ultraviolet light-induced pigmentation. Photochem Photobiol 1996; 63: 1-10; Hearing VJ, Tsukamoto K, Enzymatic control of pigmentation in mammals. FASEB J 1991; 5: 2902-2909) .

Among the enzymes of melanogenesis, tyrosinase is a key enzyme which catalyses the first two steps of melanin synthesis. Homozygous mutations of tyrosinase cause oculocutaneous albinism type I characterized by a complete lack of melanin synthesis (Toyofuku K, Wada I, Spritz RA, Hearing VJ, The molecular basis of oculocutaneous albinism type 1 (OCAl) : sorting failure and degradation of mutant tyrosinases results in a lack of pigmentation. Biochem J 2001; 355: 259-269) .

In order to treat pigmentation disorders resulting from an increase in melanin production, for which there is no treatment that meets all the expectations of patients and dermatologists, it is important to develop new therapeutic approaches.

Most of the skin-lightening compounds that are already known are phenols or hydroquinone derivatives.

These compounds inhibit tyrosinase, but the majority of them are cytotoxic to melanocytes owing to the formation of quinones. There is a risk of this toxic effect causing a permanent depigmentation of the skin. The obtaining of compounds that can inhibit melanogenesis while at the same time being very weakly cytotoxic or devoid of toxicity to melanocytes is most particularly sought.

Among the compounds already described in the literature, patent application WO 99/15148 discloses the use of 4-cycloalkyl resorcinols as depigmenting agents .

Patent FR2704428 discloses the use of 4-halo-resorcinols as depigmenting agents.

Patent applications WO 2006/097224 and WO 2006/097223 disclose the use of 4-cycloalkylmethyl resorcinols as depigmenting agents.

Patent application WO 2005/085169 discloses the use of alkyl 3- (2, 4-dihydroxyphenyl) propionate as a depigmenting agent.

Patent application WO 2004/017936 discloses the use of 3- (2, 4-dihydroxyphenyl) acrylamide as a depigmenting agent.

Patent application WO 2004/052330 discloses the use of 4- [ 1, 3] dithian-2-ylresorcinols as depigmenting agents .

More particularly, patent EP0341664 discloses the use of 4-alkyl resorcinols as depigmenting agents, among which 4-n-butyl resorcinol, also known as rucinol, is part of the composition of a depigmenting cream sold under the name Iklen®.

The applicant has now discovered, unexpectedly and surprisingly, that novel compounds of 4- (azacycloalkyl) benzene-1, 3-diol structure have a very good tyrosinase enzyme-inhibiting activity and a very low cytotoxicity. Furthermore, these compounds have a tyrosinase enzyme-inhibiting activity that is greater than that of rucinol while at the same time being less cytotoxic with respect to melanocytes than rucinol.

These compounds find uses in human medicine, in particular in dermatology, and in the cosmetics field.

FR 2939135

References

  1. Briganti, S.; Camera, E.; Picardo, M. Pigm. Cell Res. 2003, 16, 101, DOI: 10.1034/j.1600-0749.2003.00029.x

  2. 2.

    Brenner, M.; Hearing, V. J. Photochem. Photobiol. 2008, 84, 539, DOI: 10.1111/j.1751-1097.2007.00226.x

  3. 3.

    (a) Schallreuter, K. U.; Kothari, S.; Chavan, B.; Spencer, J. D. Exp. Dermatol. 2008, 17, 395, DOI: 10.1111/j.1600-0625.2007.00675.x

    (b) Cooksey, C. J.; Garratt, P. J.;Land, E. J.; Pavel, S.; Ramsden, C. A.; Riley, P. A.; Smit, N. P.J. Biol. Chem. 1997, 272, 26226, DOI: 10.1074/jbc.272.42.26226

    (c) Stratford, M. R. L.; Ramsden, C. A.; Riley, P. A.Bioorg. Med. Chem. 2013, 21, 1166, DOI: 10.1016/j.bmc.2012.12.031

  4. 4.

    Chang, T. S. Int. J. Mol. Sci. 2009, 10, 2440, DOI: 10.3390/ijms10062440

  5. 5.

    Hypopigmentation effect have already been demonstrated for resorcinols; see:

    (a) Kim, D. S.; Kim, S. Y.;Park, S. H.; Choi, Y. G.; Kwon, S. B.; Kim, M. K.; Na, J. I.; Youn, S. W.; Park, K. C. Biol. Pharm. Bull. 2005,28, 2216, DOI: 10.1248/bpb.28.2216

    (b) Khemis, A.; Kaiafa, A.;Queille-Roussel, C.; Duteil, L.; Ortonne, J. P. Br. J. Dermatol.2007, 156, 997, DOI: 10.1111/j.1365-2133.2007.07814.x

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

O=C(N[C@@H](C)c1ccccc1)N2CCC(CC2)c3ccc(O)cc3O


Filed under: Preclinical drugs, Uncategorized Tagged: Thibaud Gerfaud

BMS 986158

$
0
0

SCHEMBL16861831.png

str1

BMS 986158

MF C30H33N5O2, MW495.627 g/mol

CAS 1800340-40-2

5H-Pyrido[3,2-b]indole-7-methanol, 3-(1,4-dimethyl-1H-1,2,3-triazol-5-yl)-α,α-dimethyl-5-[(S)-phenyl(tetrahydro-2H-pyran-4-yl)methyl]-

MOA:Bromodomain and extraterminal domain protein inhibitor

Indication:Solid tumoursStatus:

Phase II :Bristol-Myers Squibb (Originator)

Phase I/IISolid tumours

  • Originator Bristol-Myers Squibb
  • Class Antineoplastics; Small molecules
  • Mechanism of Action Bromodomain and extraterminal domain protein inhibitors
  • 01 Jun 2015 Phase-I/II clinical trials for Solid tumours (Late-stage disease, Metastatic disease) in Canada (NCT02419417)
  • 02 Apr 2015 Bristol-Myers Squibb plans a phase I/IIa trial for Solid tumours (Late-stage disease) in USA, Australia and Canada (NCT02419417)

The genomes of eukaryotic organisms are highly organized within the nucleus of the cell. The long strands of duplex DNA are wrapped around an octomer of histone proteins to form a nucleosome. This basic unit is then further compressed by the aggregation and folding of nucleosomes to form a highly condensed chromatin structure. A range of different states of condensation are possible, and the tightness of this structure varies during the cell cycle, being most compact during the process of cell division. There has been appreciation recently that chromatin templates form a fundamentally important set of gene control mechanisms referred to as epigenetic regulation. By conferring a wide range of specific chemical modifications to histones and DNA (such as acetylation, methylation, phosphorylation, ubiquitinylation and SUMOylation) epigenetic regulators modulate the structure, function and accessibility of our genome, thereby exerting a huge impact in gene expression.

Histone acetylation is most usually associated with the activation of gene transcription, as the modification loosens the interaction of the DNA and the histone octomer by changing the electrostatics. In addition to this physical change, specific proteins bind to acetylated lysine residues within histones to read the epigenetic code. Bromodomains are small (-110 amino acid) distinct domains within proteins that bind to acetylated lysine residues commonly but not exclusively in the context of histones. There is a family of around 50 proteins known to contain bromodomains, and they have a range of functions within the cell. The BET family of bromodomain containing proteins

comprises 4 proteins (BRD2, BRD3, BRD4 and BRD-T) which contain tandem bromodomains capable of binding to two acetylated lysine residues in close proximity, increasing the specificity of the interaction.

BRD2 and BRD3 are reported to associate with histones along actively

transcribed genes and may be involved in facilitating transcriptional elongation (Leroy et al, Mol. Cell. 2008 30(1):51-60), while BRD4 appears to be involved in the recruitment of the pTEF-I3 complex to inducible genes, resulting in phosphorylation of RNA polymerase and increased transcriptional output (Hargreaves et al, Cell, 2009 138(1): 1294145). All family members have been reported to have some function in controlling or executing aspects of the cell cycle, and have been shown to remain in complex with chromosomes during cell division – suggesting a role in the maintenance of epigenetic memory. In addition some viruses make use of these proteins to tether their genomes to the host cell chromatin, as part of the process of viral replication (You et al., Cell, 2004 117(3):349-60).

Recent articles relating to this target include Prinjha et al., Trends in

Pharmacological Sciences, March 2012, Vol. 33, No. 3, pp. 146-153; Conway, ACS Med. Chem. Lett., 2012, 3, 691-694 and Hewings et al, J. Med. Chem., 2012, 55, 9393-9413.

Small molecule BET inhibitors that are reported to be in development include GSK-525762A, OTX-015, TEN-010 as well as others from the University of Oxford and Constellation Pharmaceuticals Inc.

Hundreds of epigenetic effectors have been identified, many of which are chromatin-binding proteins or chromatin-modifying enzymes. These proteins have been associated with a variety of disorders such as neurodegenerative disorders, metabolic diseases, inflammation and cancer. Thus, these compounds which inhibit the binding of a bromodomain with its cognate acetylated proteins, promise new approaches in the treatment of a range of autoimmune and inflammatory diseases or conditions and in the treatment of various types of cancer.

 
Inventors Derek J. Norris, George V. Delucca, Ashvinikumar V. Gavai, Claude A. Quesnelle, Patrice Gill, Daniel O’MALLEY, Wayne Vaccaro, Francis Y. Lee, Mikkel V. DEBENEDETTO, Andrew P. Degnan, Haiquan Fang, Matthew D. Hill, Hong Huang, William D. Schmitz, JR John E. STARRETT, Wen-Ching Han, John S. Tokarski, Sunil Kumar MANDAL
Applicant Bristol-Myers Squibb Company

PATENT

WO 2015100282

Examples 54 & 55

2-[3-(Dimethyl-lH-l,2,3-triazol-5-yl)-5-[oxan-4-yl(phenyl)methyl]-5H-pyrido[3,2- b] indol-7-yl] pr opan-2-ol

Enantiomer A, Example 54 Enantiomer B, Example 55

Step 1 : 2-C hloro-5-(l ,4-dimethyl- 1H- 1 ,2,3-triazol-5-yl)pyridin-3-amine

To a 100 mL round bottom flask containing 5-bromo-2-chloropyridin-3-amine (2.90 g, 14.0 mmol), l,4-dimethyl-5-(tributylstannyl)-lH-l,2,3-triazole (2.70 g, 6.99 mmol) [Seefeld, M.A. et al. PCT Int. AppL, 2008, WO2008098104] and Pd(PPh3)4 (0.61 g, 0.52 mmol) in DMF (20 mL) was added cuprous iodide (0.20 g, 1.05 mmol) and Et3N (1.9 mL, 14.0 mmol). The reaction mixture was purged with N2 for 3 min and then heated at 100 °C for 1 h. After cooling to room temperature, the mixture was diluted withl0% LiCl solution and extracted with EtOAc (2x). The combined organics were washed with sat. NaCl, dried over MgS04, filtered and concentrated. CH2C12 was added, and the resulting precipitate was collected by filtration. The mother liquor was concentrated and purified using ISCO silica gel chromatography (40 g column, gradient from 0% to 100% EtOAc/CH2Cl2). The resulting solid was combined with the precipitate and triturated with cold EtOAc to give the title compound (740 mg, 47%) as a light tan solid. LCMS (M+H) = 224.1; HPLC RT = 1.03 min (Column: Chromolith ODS S5 4.6 x 50 mm; Mobile Phase A: 10:90 MeOH: water with 0.1% TFA; Mobile Phase B: 90: 10 MeOH:water with 0.1% TFA; Temperature: 40 °C; Gradient: 0-100% B over 4 min; Flow: 4 mL/min).

Step 2: Methyl 3-((2-chloro-5-(l,4-dimethyl-lH-l,2,3-triazol-5-yl)pyridin-3-yl)amino)benzoate

Following a procedure analogous to that described in Step 2 of Example 1, 2-chloro-5-(l ,4-dimethyl-lH-l,2,3-triazol-5-yl)pyridin-3-amine (740 mg, 3.31 mmol) was converted to the title compound (644 mg, 54%). 1H NMR (400 MHz, CDC13) δ 7.94 (t, J=1.9 Hz, 1H), 7.88 (d, J=2.1 Hz, 1H), 7.83 (dt, J=7.8, 1.3 Hz, 1H), 7.49 (t, J=7.9 Hz, 1H), 7.40 (d, J=2.1 Hz, 1H), 7.36 (ddd, J=8.0, 2.3, 0.9 Hz, 1H), 6.38 (s, 1H), 3.99 (s, 3H), 3.93 (s, 3H), 2.34 (s, 3H); LCMS (M+H) = 358.2; HPLC RT = 2.34 min (Column:

Chromolith ODS S5 4.6 x 50 mm; Mobile Phase A: 10:90 MeOH:water with 0.1% TFA; Mobile Phase B: 90: 10 MeOH:water with 0.1% TFA; Temperature: 40 °C; Gradient: 0-100% B over 4 min; Flow: 4 mL/min).

Step 3: Methyl 3-(l,4-dimethyl-lH-l,2,3-triazol-5-yl)-5H-pyrido[3,2-6]indole-7-carboxylate

Following a procedure analogous to that described in Step 3 of Example 1 , methyl 3-((2-chloro-5-(l,4-dimethyl-lH-l,2,3-triazol-5-yl)pyridin-3-yl)amino)benzoate (2.82 g, 7.88 mmol) was converted to the title compound (1.58 g, 62%). 1H NMR (500 MHz, DMSO-de) δ 11.93 (s, 1H), 8.62 (d, J=1.8 Hz, 1H), 8.36 (dd, J=8.2, 0.6 Hz, 1H), 8.29 -8.22 (m, 1H), 8.16 (d, J=1.8 Hz, 1H), 7.91 (dd, J=8.2, 1.4 Hz, 1H), 4.02 (s, 3H), 3.94 (s, 3H), 2.31 (s, 3H); LCMS (M+H) = 322.3; HPLC RT = 1.98 min (Column: Chromolith ODS S5 4.6 x 50 mm; Mobile Phase A: 10:90 MeOH:water with 0.1% TFA; Mobile Phase B: 90: 10 MeOH:water with 0.1% TFA; Temperature: 40 °C; Gradient: 0-100% B over 4 min; Flow: 4 mL/min).

Alternate synthesis of Methyl 3-(l,4-dimethyl-lH-l,2,3-triazol-5-yl)-5H-pyrido[3,2-b] indole-7-carboxylate

A mixture of methyl 3-bromo-5H-pyrido[3,2-b]indole-7-carboxylate (Step 2 of Example 40, 3.000 g, 9.83 mmol), l,4-dimethyl-5-(tributylstannyl)-lH-l,2,3-triazole (4.18 g, 10.82 mmol), copper (I) iodide (0.281 g, 1.475 mmol), Pd(Ph3P)4 (0.738 g, 0.639 mmol) and triethylamine (2.74 mL, 19.66 mmol) in DMF (25 mL) was purged under a nitrogen stream and then heated in a heating block at 95 °C for 2 hours. After cooling to room temperature the reaction mixture was diluted with water and extracted into ethyl acetate. Washed with water, NH4OH, brine and concentrated. The residue was triturated with 100 mL CHC13, filtered off the solid and rinsed with CHC13 to give. 1.6 g of product. The filtrate was loaded unto the ISCO column (330 g column, A: DCM; B:

10%MeOH/DCM, 0 to 100% gradient) and chromatographed to give an additional 0.7 g. of methyl 3 -( 1 ,4-dimethyl- 1 H- 1 ,2,3 -triazol-5 -yl)-5H-pyrido [3 ,2-b]indole-7-carboxylate (2.30 g total, 7.16 mmol, 72.8 % yield).

Step 4: Methyl 3-(l,4-dimethyl-lH-l,2,3-triazol-5-yl)-5-(phenyl(tetrahydro-2H-pyran-4-yl)methyl)-5H-pyrido[3,2-b]indole-7-carboxylate

Following a procedure analogous to that described in Step 4 of Example 1 , methyl 3-(l,4-dimethyl-lH-l,2,3-triazol-5-yl)-5H-pyrido[3,2-¾]indole-7-carboxylate (80 mg, 0.25 mmol) was converted to the title compound (65 mg, 53%) after purification by prep HPLC (Column: Phen Luna C 18, 30 x 100 mm, 5 μιη particles; Mobile Phase A: 5:95 acetonitrile: water with 0.1% TFA; Mobile Phase B : 95 : 5 acetonitrile: water with 0.1% TFA; Gradient: 10-100% B over 14 min, then a 2-min hold at 100% B; Flow: 40 mL/min). 1H NMR (400 MHz, CDC13) δ 8.51 (d, J=1.8 Hz, 1H), 8.50 (s, 1H), 8.47 (d, J=8.1 Hz, 1H), 8.10 (dd, J=8.1, 1.1 Hz, 1H), 7.63 (d, J=1.8 Hz, 1H), 7.46 (d, J=7.3 Hz, 2H), 7.40 – 7.30 (m, 3H), 5.62 (d, J=10.6 Hz, 1H), 4.11 – 4.03 (m, 4H), 3.92 – 3.83 (m, 4H), 3.56 (td, J=l 1.9, 1.8 Hz, 1H), 3.35 (td, J=l 1.9, 1.9 Hz, 1H), 3.18 – 3.05 (m, 1H), 2.30 (s, 3H), 2.04 (d, J=13.0 Hz, 1H), 1.71 – 1.58 (m, 1H), 1.50 – 1.37 (m, 1H), 1.09 (d, J=12.8 Hz, 1H); LCMS (M+H) = 496.3; HPLC RT = 2.93 min (Column: Chromolith ODS S5 4.6 x 50 mm; Mobile Phase A: 10:90 MeOH:water with 0.1% TFA; Mobile Phase B: 90: 10 MeOH:water with 0.1% TFA; Temperature: 40 °C; Gradient: 0-100% B over 4 min; Flow: 4 mL/min).

Step 5 : 2- [3-(Dimethyl- lH-1 ,2,3-triazol-5-yl)-5- [oxan-4-yl(phenyl)methyl] -5H-pyrido [3,2-6] indol-7-yl] pr opan-2-ol,

Following a procedure analogous to that described in Step 5 of Example 1 , methyl 3-(l ,4-dimethyl- IH- 1 ,2,3-triazol-5-yl)-5-(phenyl(tetrahydro-2H-pyran-4-yl)methyl)-5H-pyrido[3,2-b]indole-7-carboxylate (65 mg, 0.13 mmol) was converted to racemic 2-[3-(dimethyl-lH-l,2,3-triazol-5-yl)-5-[oxan-4-yl(phenyl)methyl]-5H-pyrido[3,2-¾]indol-7-yl]propan-2-ol, which was separated by chiral prep SFC (Column: Chiralpak IB 25 x 2 cm, 5 μιη; Mobile Phase: 70/30 C02/MeOH; Flow: 50 mL/min);to give Enantiomer A (24 mg, 36%) and Enantiomer B (26 mg, 38%). Enantiomer A: 1H NMR (500 MHz, CDC13) 5 8.44 (d, J=1.8 Hz, IH), 8.36 (d, J=8.2 Hz, IH), 7.98 (s, IH), 7.56 (d, J=1.7 Hz, IH), 7.47 – 7.41 (m, 3H), 7.37 – 7.32 (m, 2H), 7.31 – 7.28 (m, IH), 5.59 (d, J=10.5 Hz, IH), 4.06 (dd, J=11.8, 2.8 Hz, IH), 3.90 – 3.84 (m, 4H), 3.55 (td, J=11.9, 2.0 Hz, IH), 3.35 (td, J=11.9, 2.0 Hz, IH), 3.15 – 3.04 (m, IH), 2.30 (s, 3H), 2.04 (d, J=13.6 Hz, IH), 1.92 (s, IH), 1.75 (s, 6H), 1.69 – 1.58 (m, IH), 1.47 – 1.38 (m, IH), 1.12 (d, J=13.4 Hz, IH); LCMS (M+H) = 496.4; HPLC RT = 2.46 min (Column: Chromolith ODS S5 4.6 x 50 mm; Mobile Phase A: 10:90 MeOH:water with 0.1% TFA; Mobile Phase B: 90: 10 MeOH:water with 0.1% TFA; Temperature: 40 °C; Gradient: 0- 100% B over 4 min; Flow: 4 mL/min). SFC RT = 5.50 min (Column: Chiralpak IB 250 x 4.6 mm, 5 μιη; Mobile Phase: 70/30 C02/MeOH; Flow: 2 mL/min); SFC RT = 1.06 min (Column:

Chiralcel OD-H 250 x 4.6 mm, 5 μιη; Mobile Phase: 50/50 C02/(1 : 1 MeOH/CH3CN); Flow: 2 mL/min); [a]D2° = -117.23 (c = 0.08, CHC13). Enantiomer B: 1H NMR (500 MHz, CDC13) δ 8.44 (d, J=l .8 Hz, IH), 8.36 (d, J=8.2 Hz, IH), 7.98 (s, IH), 7.56 (d, J=1.7 Hz, IH), 7.47 – 7.41 (m, 3H), 7.37 – 7.32 (m, 2H), 7.31 – 7.28 (m, IH), 5.59 (d, J=10.5 Hz, IH), 4.06 (dd, J=11.8, 2.8 Hz, IH), 3.90 – 3.84 (m, 4H), 3.55 (td, J=11.9, 2.0 Hz, IH), 3.35 (td, J=l 1.9, 2.0 Hz, IH), 3.15 – 3.04 (m, IH), 2.30 (s, 3H), 2.04 (d, J=13.6 Hz, IH), 1.92 (s, IH), 1.75 (s, 6H), 1.69 – 1.58 (m, IH), 1.47 – 1.38 (m, IH), 1.12 (d, J=13.4 Hz, IH); LCMS (M+H) = 496.4; HPLC RT = 2.46 min (Column: Chromolith ODS S5 4.6 x 50 mm; Mobile Phase A: 10:90 MeOH:water with 0.1% TFA; Mobile Phase B: 90: 10 MeOH:water with 0.1% TFA; Temperature: 40 °C; Gradient: 0-100% B over 4 min; Flow: 4 mL/min). SFC RT = 8.30 min (Column: Chiralpak IB 250 x 4.6 mm, 5 μιη; Mobile Phase: 70/30 C02/MeOH; Flow: 2 mL/min); SFC RT = 2.83 min (Column: Chiralcel OD-H 250 x 4.6 mm, 5 μιη; Mobile Phase: 50/50 C02/(1 : 1 MeOH/CH3CN); Flow: 2 mL/min); [a]D2° = +88.78 (c = 0.10, CHC13).

Alternate Synthesis of Examples 54

2-[3-(Dimethyl-lH-l,2,3-triazol-5-yl)-5-[oxan-4-yl(phenyl)methyl]-5H-pyrido[3,2- b] indol-7-yl] propan-2-ol.

Enantiomer A, Example 54

Step 1: (S)-methyl 3-(l,4-dimethyl-lH-l,2,3-triazol-5-yl)-5-(phenyl(tetrahydro-2H-pyran-4-yl)methyl)-5H-pyrido[3,2-b]indole-7-carboxylate

The enantiomers of phenyl(tetrahydro-2H-pyran-4-yl)methanol ( 2.0 g, 10.4 mmol) [Orjales, A. et al. J. Med. Chem. 2003, 46, 5512-5532], were separated on preperative SFC. (Column: Chiralpak AD 5 x 25 cm, 5 μιη; Mobile Phase: 74/26

C02/MeOH; Flow: 270 mL/min; Temperature 30°C). The separated peaks were concentrated and dried under vacuum to give white solids. Enantiomer A: (S)-phenyl(tetrahydro-2H-pyran-4-yl)methanol: (0.91 g, 45.5%) SFC RT = 2.32 min

(Column: Chiralpac AD 250 x 4.6 mm, 5 μιη; Mobile Phase: 70/30 C02/MeOH; Flow: 3 mL/min); Temperature 40°C. Enantiomer B: (R)-phenyl(tetrahydro-2H-pyran-4-yl)methanol. (0.92 g, 46%) SFC RT = 3.09 min (Column: Chiralpac AD 250 x 4.6 mm, 5 μιη; Mobile Phase: 70/30 C02/MeOH; Flow: 3 mL/min); Temperature 40°C.

Following a procedure analogous to that described in Step 4 of Example 1 except using toluene (120mL) as the solvent, methyl 3-(l ,4-dimethyl-lH-l,2,3-triazol-5-yl)-5H-pyrido[3,2-b]indole-7-carboxylate (4 g, 12.45 mmol) and (R)-phenyl(tetrahydro-2H-pyran-4-yl)methanol (Enantiomer B above, 5.86 g, 30.5 mmol) was converted to the title compound (5.0 g, 81%). HPLC RT = 2.91 min (Column: Chromolith ODS S5 4.6 x 50 mm; Mobile Phase A: 10:90 MeOFLwater with 0.1% TFA; Mobile Phase B: 90: 10 MeOFLwater with 0.1% TFA; Temperature: 40 °C; Gradient: 0- 100% B over 4 min; Flow: 4 mL/min).

Step 2. (S)-2-[3-(Dimethyl-lH-l,2,3-triazol-5-yl)-5-[oxan-4-yl(phenyl)methyl]-5H-pyrido [3,2-b] indol-7-yl] propan-2-ol

A 500 mL round bottom flask containing (S)-methyl 3-(l,4-dimethyl-lH-l,2,3-triazol-5-yl)-5-(phenyl(tetrahydro-2H-pyran-4-yl)methyl)-5H-pyrido[3,2-b]indole-7-carboxylate (5.0 g, 10.09 mmol) in THF (150 mL) was cooled in an ice/MeOH bath. MeMgBr, (3M in Et20, 17.0 mL, 51.0 mmol) was added slowly over 4 min. The resulting solution was stirred for 2 h and then quenched carefully with sat. NH4C1. The reaction mixture was diluted with 10% LiCl solution extracted with EtOAc. The organic layer was dried over MgS04, filtered and concentrated. The crude material was purified using ISCO silica gel chromatography (120 g column, gradient from 0%> to 6%>

MeOH/CH2Cl2). The product was collected and concentrated then dissolved in hot MeOH(35mL). To the mixture was added 15mL water and the mixture was cooled to room temperature. The resulting white precipitate was collected by filtration with 2: 1 MeOH/water rinse then dried under vacuum to give the title compound (3.2 g, 62%>). 1H

NMR (500 MHz, CDC13) δ 8.40 (d, J=1.8 Hz, 1H), 8.33 (d, J=8.2 Hz, 1H), 7.93 (s, 1H), 7.53 (d, J=l .8 Hz, 1H), 7.46 (d, J=7.3 Hz, 2H), 7.42 (dd, J=8.2, 1.4 Hz, 1H), 7.37 – 7.31 (m, 2H), 7.30 – 7.28 (m, 1H), 5.56 (d, J=10.5 Hz, 1H), 4.06 (d, J=8.9 Hz, 1H), 3.89 – 3.83 (m, 1H), 3.55 (td, J=11.9, 2.1 Hz, 1H), 3.35 (td, J=11.9, 2.1 Hz, 1H), 3.10 (q, J=10.8 Hz, 1H), 2.39 (s, 3H), 2.23 (s, 3H), 2.03 (d, J=14.2 Hz, 1H), 1.89 (s, 1H), 1.74 (s, 6H), 1.68 -1.59 (m, 1H), 1.46 – 1.36 (m, 1H), 1.12 (d, J=12.2 Hz, 1H); LCMS (M+H) = 496.3; HPLC RT = 2.44 min (Column: Chromolith ODS S5 4.6 x 50 mm; Mobile Phase A: 10:90 MeOH: water with 0.1% TFA; Mobile Phase B: 90: 10 MeOH: water with 0.1%

TFA; Temperature: 40 °C; Gradient: 0-100% B over 4 min; Flow: 4 mL/min); SFC RT = 2.01 min (Column: Chiralcel OD-H 250 x 4.6 mm, 5 μιη; Mobile Phase: 60/40 C02/(1 : 1 MeOH/CH3CN); Flow: 2 mL/min). SFC RT = 1.06 min (Column: Chiralcel OD-H 250 x 4.6 mm, 5 μιη; Mobile Phase: 50/50 C02/(1 : 1 MeOH/CH3CN); Flow: 2 mL/min).

1 to 1 of 1
Patent ID Patent Title Submitted Date Granted Date
US9458156 Tricyclic compounds as anticancer agents 2014-12-23 2016-10-04

3rd speaker at 1st time disclosures is Ashvin Gavai of @bmsnews talking about an oral BET inhibitor to treat cancer

str0

//////////

CC(C)(O)c2cc3n(c1cc(cnc1c3cc2)c4c(C)nnn4C)[C@@H](C5CCOCC5)c6ccccc6


Filed under: Preclinical drugs, Uncategorized Tagged: BMS 986158

ABBV 2222

$
0
0

str1

ABBV 2222

Benzoic acid, 4-[(2R,4R)-4-[[[1-(2,2-difluoro-1,3-benzodioxol-5-yl)cyclopropyl]carbonyl]amino]-7-(difluoromethoxy)-3,4-dihydro-2H-1-benzopyran-2-yl]-

4-[(2R,4R)-4-({[1-(2,2-difluoro-1,3-benzodioxol-5-yl)cyclopropyl]carbonyl}- amino)-7-(difluoromethoxy)-3,4-dihydro-2H-chromen-2-yl]benzoic acid

CAS  1918143-53-9

MF C28 H21 F4 N O7
MW 559.46
1H NMR (400 MHz, CDCl.sub.3) .delta. 8.17-8.03 (m, 2H), 7.49 (d, J=8.2 Hz, 2H), 7.16-6.99 (m, 4H), 6.73-6.67 (m, 2H), 6.38 (d, J=73.6 Hz, 1H), 5.48 (td, J=10.4, 6.1 Hz, 1H), 5.36 (d, J=8.8 Hz, 1H), 5.31-5.21 (m, 1H), 2.52 (ddd, J=13.3, 6.0, 2.2 Hz, 1H), 1.86-1.71 (m, 2H), 1.68-1.60 (m, 1H), 1.10 (q, J=3.7, 2.4 Hz, 2H);
 
MS (ESI-) m/z=558 (M-H).sup.-.

Image result

DESCRIPTION

Cystic fibrosis (CF), one of the most common autosomal recessive genetic diseases in the Caucasian population, is caused by loss of function mutations in the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) gene, which is located on chromosome 7 (http://www.cff.org/AboutCF/; Rowe S. M et al. (2005); N Eng J Med. (352), 1992-2001). Approximately 1:3500 and 1:3000 infants born in the United States and in Europe, respectively, are affected by CF, resulting in ˜75,000 cases worldwide, ˜30,000 of which are in the United State. Approximately 1,000 new cases of CF are diagnosed each year, with more than 75% of patients being diagnosed by 2 years of age. Nearly half the CF population is currently 18 years of age and older. The CFTR protein (Gregory, R. J. et al. (1990) Nature 347:382-386; Rich, D. P. et al. (1990) Nature 347:358-362; Riordan, J. R. et al. (1989) Science 245:1066-1073) is a cAMP/ATP-mediated ion channel expressed in a variety of cell types, including secretory and absorptive epithelial cells. CFTR regulates chloride and bicarbonate anion flux across the cell membrane, maintaining electro neutrality and osmolarity across the epithelial membrane (Quinton, P. M. (1990), FASEB J. 4: 2709-2727). CFTR is also responsible for regulating the activity of other ion channels and proteins (Guggino, W. B. et al. (2006), Nat Revs Molecular Cell Biology 7, 426-436).

Aberrations in CFTR function result in imbalance of the airway surface liquid, leading to mucus dehydration, inflammation, recurrent bacterial infection and irreversible lung damage, which lead to premature death in affected patients. Besides respiratory disease, CF patients suffer from gastrointestinal problems and pancreatic insufficiency. The majority of males (95%) with cystic fibrosis are infertile as a result of azoospermia caused by altered vas deferens; which may be absent, atrophic, or fibrotic. Fertility is also decreased among females with cystic fibrosis due to abnormal cervical mucus.

The F508del mutation, the most common of the approximately 1900 identified polymorphisms in CFTR, results in defective processing of CFTR in the endoplasmic reticulum (ER) (http://www.cftr2.org/index.php). Approximately 90% of the CF patients carry at least one copy of the F508del mutation (deletion of a phenylalanine on position 508), and 50%-60% of the patients are homozygous for this mutation. The defective processing of CFTR results in early CFTR degradation, which leads to reduced trafficking or absence of the protein on the membrane. As there have been over 100 CF disease-causing mutations identified, they have been classified according to their phenotypic consequences and belong to synthesis, maturation, regulation, conductance, reduced number due to quantity and reduced number due to stability classifications.

Current CF drug discovery efforts focus upon developing two classes of compounds to modulate CFTR. One class, called Correctors, helps to overcome the folding defects of the mutated CFTR protein to promote its maturation resulting in higher cell surface expression. The other classes of compounds, called Potentiators, help overcome the defective regulation and/or conductance of the protein by increasing the probability of channel opening on the membrane surface.

In addition, as the modulation of CFTR protein mutations to promote proper protein folding is beneficial for CF, there are other diseases mediated by CFTR. For example, Sjögren’s Syndrome (SS), an autoimmune disorder that results in symptoms of xerostomia (dry mouth) and keratoconjunctivitis sicca (KCS, dry eyes) may result from dysregulation of moisture producing glands throughout the body. Chronic obstructive lung disease (COLD), or chronic obstructive airway disease (COAD), which is a progressive and irreversible airflow limitation in the airways is result of several physiologic abnormalities, including mucus hyper secretion and impaired mucociliary secretion. Increasing the anion secretion by CFTR potentiators have been suggested to overcome these phenotypic complexities with Sjögren’s Syndrome by increasing the corneal hydration and by overcoming the impaired mucociliary secretion in COAD (Bhowmik A, et al. (2009) Vol. 103(4), 496-502; Sloane P, et al. PLOS One (2012) Vol 7(6), 239809 (1-13)).

STEP 1

(R)-methyl 4-(7-hydroxy-4-oxochroman-2-yl)benzoate

RXN……….By reacting  7-hydroxy-4H-chromen-4-one AND  (4-(methoxycarbonyl)phenyl)boronic acid

STEP 2

(R)-methyl 4-(7-hydroxy-4-(methoxyimino)chroman-2-yl)benzoate

Reacting ABOVE compd  and O-methylhydroxylamine,

STEP 3

Methyl 4-((2R,4R)-4-amino-7-hydroxychroman-2-yl)benzoate

reacting ABOVE  compd with 5% platinum (0.05 equivalent) on carbon in acetic acid. The reaction was stirred at room temperature under hydrogen

THEN STEP 4

Methyl 4-((2R,4R)-4-amino-7-hydroxychroman-2-yl)benzoate isolated AS  trifluroroacetic acid salt

STEP 5
methyl 4-((2R,4R)-4-(1-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)cyclopropanec- arboxamido)-7-hydroxychroman-2-yl)benzoate

by reacting  1-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)cyclopropanecarboxylic acid  and HATU (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate, the ABOVE compound AND  N-ethyl-N-isopropylpropan-2-amine

STEP 6

Methyl 4-((2R,4R)-4-(1-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)cyclopropanec- arboxamido)-7-(difluoromethoxy)chroman-2-yl)benzoate

by reacting ABOVE compound  and diethyl(bromodifluoromethyl)phosphonate

AND FINAL STEP7  is ESTER HYDROLYSIS USING lithium hydroxide to get ABBV 2222

PATENT
US 20160120841

str1

Example 122

4-[(2R,4R)-4-({[1-(2,2-difluoro-1,3-benzodioxol-5-yl)cyclopropyl]carbonyl}- amino)-7-(difluoromethoxy)-3,4-dihydro-2H-chromen-2-yl]benzoic acid

[1880] To Example 123E (130 mg, 0.227 mmol) in methanol (2 mL) and water (0.5 mL) was added lithium hydroxide (32.6 mg, 1.360 mmol). The mixture was stirred at 35.degree. C. for 4 hours, LC/MS showed the conversion was complete. Solvent was removed under reduced pressure and water (2 mL) was added. The pH of the mixture was adjusted to pH 1-2 with the addition of 2 M HCl. The precipitated white solid was collected by filtration, and dried to provide the title compound (110 mg, 0.197 mmol, 87% yield). .sup.1H NMR (400 MHz, CDCl.sub.3) .delta. 8.17-8.03 (m, 2H), 7.49 (d, J=8.2 Hz, 2H), 7.16-6.99 (m, 4H), 6.73-6.67 (m, 2H), 6.38 (d, J=73.6 Hz, 1H), 5.48 (td, J=10.4, 6.1 Hz, 1H), 5.36 (d, J=8.8 Hz, 1H), 5.31-5.21 (m, 1H), 2.52 (ddd, J=13.3, 6.0, 2.2 Hz, 1H), 1.86-1.71 (m, 2H), 1.68-1.60 (m, 1H), 1.10 (q, J=3.7, 2.4 Hz, 2H); MS (ESI-) m/z=558 (M-H).sup.-.

Example 123

methyl 4-[(2R,4R)-4-({[1-(2,2-difluoro-1,3-benzodioxol-5-yl)cyclopropyl]ca- rbonyl}amino)-7-(difluoromethoxy)-3,4-dihydro-2H-chromen-2-yl]benzoate

Example 123A

(R)-methyl 4-(7-hydroxy-4-oxochroman-2-yl)benzoate

[1881] A mixture of bis(2,2,2-trifluoroacetoxy)palladium (271 mg, 0.816 mmol), (S)-4-(tert-butyl)-2-(pyridin-2-yl)-4,5-dihydrooxazole (200 mg, 0.979 mmol), ammonium hexafluorophosphate(V) (798 mg, 4.90 mmol), (4-(methoxycarbonyl)phenyl)boronic acid (2203 mg, 12.24 mmol) and dichloroethane (8 mL) in a 20 mL vial was stirred for 5 minutes at room temperature, followed by the addition of 7-hydroxy-4H-chromen-4-one (CAS 59887-89-7, MFCD00209371, 1323 mg, 8.16 mmol) and water (256 mg, 14.19 mmol). The vial was capped and the mixture was stirred at 60.degree. C. overnight. The reaction gradually turned black, with Pd plated out on the sides of the vial. The mixture was filtered through a plug of celite and eluted with ethyl acetate to give a red solution which was washed with brine. The solvent was removed in vacuo and the crude material was chromatographed using a 100 g silica gel cartridge and eluted with a gradient of 5-40% ethyl acetate in heptane to provide the title compound (1.62 g, 66.6% yield). .sup.1H NMR (400 MHz, CDCl.sub.3) .delta. 8.15-8.04 (m, 2H), 7.87 (d, J=8.7 Hz, 1H), 7.60-7.49 (m, 2H), 6.62-6.45 (m, 2H), 5.87 (s, 1H), 5.53 (dd, J=12.8, 3.2 Hz, 1H), 3.94 (s, 3H), 3.07-2.80 (m, 2H); MS (ESI+) m/z=299 (M+H).sup.+.

Example 123B

(R)-methyl 4-(7-hydroxy-4-(methoxyimino)chroman-2-yl)benzoate

[1882] The mixture of Example 123A (960 mg, 3.22 mmol), sodium acetate (528 mg, 6.44 mmol) and O-methylhydroxylamine, hydrochloric acid (538 mg, 6.44 mmol) in methanol (10 mL) was stirred at 60.degree. C. overnight. Solvent was removed under reduced pressure. The residue was dissolved in ethyl acetate and washed with water. The organic layers was dried over MgSO.sub.4, filtered, and concentrated. The residue was washed with ether to provide the title compound (810 mg, 2.475 mmol, 77% yield). .sup.1H NMR (400 MHz, CDCl.sub.3) .delta. 8.15-8.03 (m, 2H), 7.81 (d, J=8.7 Hz, 1H), 7.58-7.43 (m, 2H), 6.50 (dd, J=8.6, 2.5 Hz, 1H), 6.45 (d, J=2.5 Hz, 1H), 5.21 (d, J=3.0 Hz, 1H), 5.12 (dd, J=12.2, 3.2 Hz, 1H), 3.95 (s, 3H), 3.93 (s, 3H), 3.45 (dd, J=17.2, 3.2 Hz, 1H), 2.63 (dd, J=17.2, 12.2 Hz, 1H); MS (ESI+) m/z 328 (M+H).sup.+.

Example 123C

Methyl 4-((2R,4R)-4-amino-7-hydroxychroman-2-yl)benzoate

[1883] A mixture of Example 123B (570 mg, 1.741 mmol) was treated with 5% platinum (0.05 equivalent) on carbon in acetic acid (5 mL). The reaction was stirred at room temperature under hydrogen (1 atmosphere) for 24 hours, LC/MS showed conversion over 95%. The mixture was filtered through a celite pad and solvent removed under reduced pressure. The residue was purified by preparative LC method TFA2 to provide the trifluroroacetic acid salt of the title compound (300 mg, 44% yield). LC/MS m/z 283 (M-NH.sub.2).sup.+.

Example 123D

methyl 4-((2R,4R)-4-(1-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)cyclopropanec- arboxamido)-7-hydroxychroman-2-yl)benzoate

[1884] A mixture of 1-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)cyclopropanecarboxylic acid (162 mg, 0.668 mmol) and HATU (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate, 380 mg, 1.0 mmol) in DMF (2 mL) was stirred for 5 minutes at room temperature, followed by the addition of Example 123C (200 mg, 0.334 mmol) and N-ethyl-N-isopropylpropan-2-amine (0.466 ml, 2.67 mmol). The mixture was stirred at room temperature for 2 hours, LC/MS showed reaction complete. The mixture was loaded on to a 25 g silica gel cartridge eluting with 5-50% ethyl acetate in heptane provide the title compound (204 mg, 58.3% yield). .sup.1H NMR (400 MHz, CDCl.sub.3) .delta. 8.11-7.90 (m, 2H), 7.42 (d, J=8.0 Hz, 2H), 7.16-7.02 (m, 2H), 6.94 (dd, J=37.7, 8.3 Hz, 2H), 6.49-6.32 (m, 2H), 5.67 (s, 1H), 5.36 (dt, J=15.3, 8.7 Hz, 2H), 5.18 (d, J=10.7 Hz, 1H), 3.93 (s, 3H), 2.56-2.36 (m, 1H), 1.80-1.70 (m, 2H), 1.26 (d, J=2.2 Hz, 1H), 1.10-1.04 (m, 2H); MS (ESI-) m/z=521.9 (M-H).sup.-.

Example 123E

Methyl 4-((2R,4R)-4-(1-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)cyclopropanec- arboxamido)-7-(difluoromethoxy)chroman-2-yl)benzoate

[1885] To Example 123D (190 mg, 0.363 mmol) and diethyl(bromodifluoromethyl)phosphonate (0.129 ml, 0.726 mmol) in a mixture of acetonitrile (2 mL) and water (1 mL) was added 50% aqueous potassium hydroxide (244 mg, 2.178 mmol) drop wise via syringe while stirring vigorously. After the addition was completed, LC/MS showed conversion was complete with a small by-product peak. Additional water was added to the mixture and the mixture was extracted with ethyl acetate (3.times.20 mL). The combined organic extracts were washed with 1 M HCl (5 mL) and water, dried over MgSO.sub.4, filtered, and concentrated. The residue was purified by preparative LC method TFA2 to provide the title compound (150 mg, 72% yield). .sup.1H NMR (400 MHz, CDCl.sub.3) .delta. 8.09-8.00 (m, 2H), 7.49-7.41 (m, 2H), 7.15-6.99 (m, 4H), 6.75-6.66 (m, 2H), 5.50-5.40 (m, 1H), 5.33 (d, J=8.9 Hz, 1H), 5.25 (dd, J=11.3, 2.0 Hz, 1H), 3.93 (s, 3H), 2.50 (ddd, J=13.4, 6.1, 2.1 Hz, 1H), 1.84-1.71 (m, 2H), 1.65 (d, J=2.8 Hz, 1H), 1.11-1.06 (m, 2H); MS (ESI-) m/z=572 (M-H).sup.-.

REFERENCE

Next up is Xueqing Wang of @abbvie speaking about a collaboration with @GalapagosNV on a different cystic fibrosis treatment

str0

///////////ABBV 2222

O=C(O)c1ccc(cc1)[C@@H]3Oc2cc(OC(F)F)ccc2C(C3)NC(=O)C4(CC4)c5ccc6OC(F)(F)Oc6c5


Filed under: Preclinical drugs, Uncategorized Tagged: ABBV 2222

PF 06821497

$
0
0

str1

PF 06821497

Cas 1844849-11-1

Designed to treat lymphoma

1(2H)-Isoquinolinone, 5,8-dichloro-2-[(1,2-dihydro-4-methoxy-6-methyl-2-oxo-3-pyridinyl)methyl]-3,4-dihydro-7-[(S)-methoxy-3-oxetanylmethyl]-

MF C22 H24 Cl2 N2 O5, 

MW 467.34

ChemSpider 2D Image | 5,8-Dichloro-2-[(4-methoxy-6-methyl-2-oxo-1,2-dihydro-3-pyridinyl)methyl]-7-[methoxy(3-oxetanyl)methyl]-3,4-dihydro-1(2H)-isoquinolinone | C22H24Cl2N2O5PF 06821497

5,8-Dichloro-2-[(4-methoxy-6-methyl-2-oxo-1,2-dihydro-3-pyridinyl)methyl]-7-[methoxy(3-oxetanyl)methyl]-3,4-dihydro-1(2H)-isoquinolinone

1(2H)-Isoquinolinone, 5,8-dichloro-2-[(1,2-dihydro-4-methoxy-6-methyl-2-oxo-3-pyridinyl)methyl]-3,4-dihydro-7-(methoxy-3-oxetanylmethyl)-

  • Molecular Formula C22H24Cl2N2O5
  • Average mass 467.342 Da

SCHEMBL17330377.pngPF 06821497

5,8-dichloro-2-[(4-methoxy-6-methyl-2-oxo-1H-pyridin-3-yl)methyl]-7-[(S)-methoxy(oxetan-3-yl)methyl]-3,4-dihydroisoquinolin-1-one

US2015361067

Inventors Michael Raymond Collins, Robert Steven Kania, Robert Arnold Kumpf, Pei-Pei Kung, Daniel Tyler Richter, Scott Channing Sutton, Martin James Wythes
Original Assignee Pfizer Inc.Image result
  • Epigenetic alterations play an important role in the regulation of cellular processes, including cell proliferation, cell differentiation and cell survival. The epigenetic silencing of tumor suppressor genes and activation of oncogenes may occur through alteration of CpG island methylation patterns, histone modification, and dysregulation of DNA binding protein. Polycomb genes are a set of epigenetic effectors. EZH2 (enhancer of zeste homolog 2) is the catalytic component of the Polycomb Repressor Complex 2 (PRC2), a conserved multi-subunit complex that represses gene transcription by methylating lysine 27 on Histone H3 (H3K27). EZH2 plans a key role in regulating gene expression patterns that regulate cell fate decisions, such as differentiation and self-renewal. EZH2 is overexpressed in certain cancer cells, where it has been linked to cell proliferation, cell invasion, chemoresistance and metastasis.
  • High EZH2 expression has been correlated with poor prognosis, high grade, and high stage in several cancer types, including breast, colorectal, endometrial, gastric, liver, kidney, lung, melanoma, ovarian, pancreatic, prostate, and bladder cancers. See Crea et al., Crit. Rev. Oncol. Hematol. 2012, 83:184-193, and references cited therein; see also Kleer et al., Proc. Natl. Acad. Sci. USA 2003, 100:11606-11; Mimori et al., Eur. J. Surg. Oncol. 2005, 31:376-80; Bachmann et al., J. Clin. Oncol. 2006, 24:268-273; Matsukawa et al., Cancer Sci. 2006, 97:484-491; Sasaki et al. Lab. Invest. 2008, 88:873-882; Sudo et al., Br. J. Cancer 2005, 92(9):1754-1758; Breuer et al., Neoplasia 2004, 6:736-43; Lu et al., Cancer Res. 2007, 67:1757-1768; Ougolkov et al., Clin. Cancer Res. 2008, 14:6790-6796; Varambally et al., Nature 2002, 419:624-629; Wagener et al., Int. J. Cancer 2008, 123:1545-1550; and Weikert et al., Int. J. Mol. Med. 2005, 16:349-353.
    Recurring somatic mutations in EZH2 have been identified in diffuse large B-cell lymphoma (DLBCL) and follicular lymphomas (FL). Mutations altering EZH2 tyrosine 641 (e.g., Y641C, Y641F, Y641N, Y641S, and Y641H) were reportedly observed in up to 22% of germinal center B-cell DLBCL and 7% of FL. Morin et al. Nat. Genetics 2010 February; 42(2):181-185. Mutations of alanine 677 (A677) and alanine 687 (A687) have also been reported. McCabe et al., Proc. Natl. Acad. Sci. USA 2012, 109:2989-2994; Majer et al. FEBS Letters 2012, 586:3448-3451. EZH2 activating mutations have been suggested to alter substrate specificity resulting in elevated levels of trimethylated H3K27 (H3K27me3).
    Accordingly, compounds that inhibit the activity of wild type and/or mutant forms of EZH2 may be of interest for the treatment of cancer.

SYNTHESIS

Steps

1 COUPLING, Ag2CO3

2 Alkylation, K2CO3

3 LiAlH4 REDUCTION

4 THIONYL CHLORIDE

5 N-Alkylation of Amides, t-BuOK

6 A GRIGNARD REACTION

7 AN ALKYLATION , METHYL IODIDE, t-BuOK

8 HYDROGENATION, DE BENZYLATION,  PLATINUM OXIDE

9 LAST STEP separation by chiral preparative, SFC on (R,R) Whelk O1 column, TO GET PF 06821497

PATENT

US 20150361067

///////////////PF 06821497, 1844849-11-1, PFIZER, lymphoma, Pei-Pei Kung,  @pfizer, #ACSSanFran, Michael Raymond Collins, Robert Steven Kania, Robert Arnold Kumpf, Pei-Pei Kung, Daniel Tyler Richter, Scott Channing Sutton, Martin James Wythes

Next up in #MEDI 1st time disclosures Pei-Pei Kung from @pfizer presenting a molecule designed to treat lymphoma #ACSSanFran

str0

CO[C@H](c2cc(Cl)c3CCN(CC1=C(OC)C=C(C)NC1=O)C(=O)c3c2Cl)C4COC4

CC1=CC(=C(C(=O)N1)CN2CCC3=C(C=C(C(=C3C2=O)Cl)C(C4COC4)OC)Cl)OC

Filed under: Preclinical drugs, Uncategorized Tagged: #ACSSanFran, 1844849-11-1, Daniel Tyler Richter, lymphoma, Martin James Wythes, Michael Raymond Collins, Pei-Pei Kung, PF 06821497, PFIZER, Robert Arnold Kumpf, Robert Steven Kania, Scott Channing Sutton

Takeda’s Peripherally selective noradrenaline reuptake inhibitor

$
0
0

str1

SCHEMBL1279856.png

ChemSpider 2D Image | 1-{[(6S,7R)-7-(4-Chloro-3-fluorophenyl)-1,4-oxazepan-6-yl]methyl}-2-oxo-1,2-dihydro-3-pyridinecarboxylic acid | C18H18ClFN2O4

1-{[(6S,7R)-7-(4-Chloro-3-fluorophenyl)-1,4-oxazepan-6-yl]methyl}-2-oxo-1,2-dihydro-3-pyridinecarboxylic acid

  • Molecular Formula C18H18ClFN2O4
  • Average mass 380.798 Da

CAS 1372185-97-1

CAS 1372180-09-0 hydrochloride

Peripherally selective noradrenaline reuptake inhibitor

Image result for takeda pharmaceuticals1-([(6S,7R)-7-(4-Chloro-3-fluorophenyl)-1,4-oxazepan-6-yl]methyl]-2-oxo-1,2-dihydropyridine-3-carboxylic acid monohydrochloride

3-Pyridinecarboxylic acid, 1-[[(6S,7R)-7-(4-chloro-3-fluorophenyl)hexahydro-1,4-oxazepin-6-yl]methyl]-1,2-dihydro-2-oxo-, hydrochloride (1:1)

1-{[(6S,7R)-7-(4-Chloro-3-fluorophenyl)-1,4-oxazepan-6-yl]methyl}-2-oxo-1,2-dihydropyridine-3-carboxylic Acid Hydrochloride (1:1) (1·HCl)

TAKEDA PHARMACEUTICAL COMPANY LIMITED [JP/JP]; 1-1, Doshomachi 4-chome, Chuo-ku, Osaka-shi, Osaka 5410045 (JP)

ISHICHI, Yuji; (JP).
YAMADA, Masami; (US).
KAMEI, Taku; (JP).
FUJIMORI, Ikuo; (US).
NAKADA, Yoshihisa; (JP).
YUKAWA, Tomoya; (JP).
SAKAUCHI, Nobuki; (JP).
OHBA, Yusuke; (JP).
TSUKAMOTO, Tetsuya; (JP)

Paper

Development of a Practical Synthesis of a Peripherally Selective Noradrenaline Reuptake Inhibitor Possessing a Chiral 6,7-trans-Disubstituted-1,4-oxazepane as a Scaffold

Process Chemistry, Pharmaceutical Sciences, Takeda Pharmaceutical Company Limited, 17-85, Jusohonmachi 2-Chome, Yodogawa-ku, Osaka 532-8686, Japan
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.7b00313

Abstract

Abstract Image

A practical synthesis of a peripherally selective noradrenaline reuptake inhibitor that has a chiral 6,7-trans-disubstituted-1,4-oxazepane as a new class of scaffold is described. The amino alcohol possessing the desired stereochemistry was obtained with excellent dr and ee, starting from a commercially available aldehyde via a Morita–Baylis–Hillman reaction, Michael addition, isolation as maleic acid salt, reduction, and diastereomeric salt formation with (+)-10-camphorsulfonic acid. The desired single stereoisomer obtained at an early stage of the synthesis was used for seven-membered ring formation in fully telescoped processes, providing the chiral 6,7-trans-disubstituted-1,4-oxazepane efficiently. In addition to controls of dr and ee of the chiral 1,4-oxazepane, and control of N,O-selectivity in SN2 reaction of the intermediate mesylate with a pyridone derivative, finding appropriate intermediates that were amenable to isolation and upgrade of purity enabled a practical chiral HPLC separation-free, column chromatograph-free synthesis of the drug candidate with excellent chemical and optical purities in a higher overall yield.

Mp 261–262 °C;
1H NMR (600 MHz, DMSO-d6) δ 3.09–3.18 (m, 1H), 3.20–3.43 (m, 4H), 3.77–3.88 (m, 1H), 3.96 (br dd, J = 13.2, 5.7 Hz, 1H), 4.04 (dt, J = 13.8, 4.2 Hz, 1H), 4.17 (br dd, J = 13.6, 7.6 Hz, 1H), 4.59 (br d, J = 9.1 Hz, 1H), 6.66 (t, J = 7.0 Hz, 1H), 7.27 (br dd, J = 8.3, 1.1 Hz, 1H), 7.47 (br dd, J = 10.4, 1.3 Hz, 1H), 7.54 (br t, J = 8.1 Hz, 1H), 8.10 (dd, J = 6.4, 1.9 Hz, 1H), 8.26 (dd, J = 7.2, 1.9 Hz, 1H), 9.59 (br s, 2H), 14.2 (br s, 1H);
 13C NMR (151 MHz, DMSO-d6) δ 40.5, 44.9, 46.5, 50.0, 63.9, 82.1, 108.4, 116.0 (2JCF = 21.1 Hz), 116.7, 119.3 (2JCF = 18.1 Hz), 125.1 (3JCF = 4.5 Hz), 130.4, 140.9 (3JCF = 7.6 Hz), 145.1, 145.2, 156.8 (1JCF = 247.6 Hz), 163.6, 164.4;
IR (ATR) 2925, 2693, 1725, 1625, 1563, 1484, 1445, 1379, 1293, 1206, 1126, 1097, 1064, 1003, 934, 868, 856, 820, 783, 771, 627, 538, 521, 459, 411 cm–1;
HRMS (ESI): [M + H]+ calcd for C18H19ClFN2O4 (1), 381.1017; found, 381.1009.

PATENT

https://www.google.com/patents/WO2012046882A1?cl=zh

PAPER

Volume 24, Issue 16, 15 August 2016, Pages 3716–3726

http://www.sciencedirect.com/science/article/pii/S0968089616304382

Abstract

Peripheral-selective inhibition of noradrenaline reuptake is a novel mechanism for the treatment of stress urinary incontinence to overcome adverse effects associated with central action. Here, we describe our medicinal chemistry approach to discover a novel series of highly potent, peripheral-selective, and orally available noradrenaline reuptake inhibitors with a low multidrug resistance protein 1 (MDR1) efflux ratio by cyclization of an amide moiety and introduction of an acidic group. We observed that the MDR1 efflux ratio was correlated with the pKa value of the acidic moiety. The resulting compound 9exhibited favorable PK profiles, probably because of the effect of intramolecular hydrogen bond, which was supported by a its single-crystal structure. The compound 9, 1-{[(6S,7R)-7-(4-chloro-3-fluorophenyl)-1,4-oxazepan-6-yl]methyl}-2-oxo-1,2-dihydropyridine-3-carboxylic acid hydrochloride, which exhibited peripheral NET-selective inhibition at tested doses in rats by oral administration, increased urethral resistance in a dose-dependent manner.


Graphical abstract

Image for unlabelled figure

REFERNCES

(a) IshichiY.YamadaM.KameiT.FujimoriI.NakadaY.YukawaT.SakauchiN.OhbaY.TsukamotoT. WO 2012/046882 A1, Apr 12, 2012.

(b) FujimoriI.YukawaT.KameiT.NakadaY.SakauchiN.YamadaM.OhbaY.TakiguchiM.KunoM.KamoI.NakagawaH.HamadaT.IgariT.OkudaT.YamamotoS.TsukamotoT.IshichiY.UenoH. Bioorg. Med. Chem. 2015235000– 5014 DOI: 10.1016/j.bmc.2015.05.017

(c) YukawaT.FujimoriI.KameiT.NakadaY.SakauchiN.YamadaM.OhbaY.UenoH.TakiguchiM.KunoM.KamoI.NakagawaH.FujiokaY.IgariT.IshichiY.TsukamotoT. Bioorg. Med. Chem. 2016243207– 3217 DOI: 10.1016/j.bmc.2016.05.038

(d) YukawaT.NakadaY.SakauchiN.KameiT.YamadaM.OhbaY.FujimoriI.UenoH.TakiguchiM.KunoM.KamoI.NakagawaH.FujiokaY.IgariT.IshichiY.TsukamotoT. Bioorg. Med. Chem. 2016243716– 3726 DOI: 10.1016/j.bmc.2016.06.014

//////////////////1372185-97-1, 1372180-09-0, Peripherally selective,  noradrenaline reuptake inhibitor,  TAKEDA

O=C(O)C3=CC=CN(C[C@@H]1CNCCO[C@H]1c2ccc(Cl)c(F)c2)C3=O

“NEW DRUG APPROVALS” 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
Follow amcrasto on Twitter

The synthesis, biological evaluation and structure–activity relationship of 2-phenylaminomethylene-cyclohexane-1,3-diones as specific anti-tuberculosis agents

$
0
0

ST50238235.png

str1

CAS  74102-02-6

Molecular Formula: C15H17NO3
Molecular Weight: 259.305 g/mol

2-(((2-hydroxyphenyl)amino)methylene)-5,5-dimethylcyclohexane-1,3-dione (39): White solid; m.p. 249 o C; TLC Rf value, 0.48 (in EtOAc:Hexane,60:40);

IR (neat) 2980, 2950, 1678, 1040 cm-1;

1 H NMR (400 MHz, CD3OD) δ 9.86 (1H, bs), 8.66 (1H, d, J = 16.0 Hz), 7.46- 7.34 (1H, m), 7.07-6.84 (3H, m), 2.46 (2H, s), 2.41 (2H, s), 1.10 (3H, s), 1.09 (3H, s);

13C NMR (101 MHz, CDCl3) δ 199.8, 197.2, 149.6, 149.3, 147.8, 127.2, 126.6, 120.6, 120.3, 108.

The synthesis, biological evaluation and structure–activity relationship of 2-phenylaminomethylene-cyclohexane-1,3-diones as specific anti-tuberculosis agents

 Author affiliations

Abstract

The present study utilised whole cell based phenotypic screening of thousands of diverse small molecules against Mycobacterium tuberculosis H37Rv (M. tuberculosis) and identified the cyclohexane-1,3-dione-based structures 5 and 6 as hits. The selected hit molecules were used for further synthesis and a library of 37 compounds under four families was synthesized for lead generation. Evaluation of the library against M. tuberculosis lead to the identification of three lead antituberculosis agents (3739 and 41). The most potential compound, 2-(((2-hydroxyphenyl)amino)methylene)-5,5-dimethylcyclohexane-1,3-dione (39) showed an MIC of 2.5 μg mL−1, which falls in the range of MICs values found for the known antituberculosis drugs ethambutol, streptomycin and levofloxacin. Additionally, this compound proved to be non-toxic (<20% inhibition at 50 μM concentration) against four human cell lines. Like first line antituberculosis drugs (isoniazid, rifampicin and pyrazinamide) this compound lacks activity against general Gram positive and Gram negative bacteria and even against M. smegmatis; thereby reflecting its highly specific antituberculosis activity.

Graphical abstract: The synthesis, biological evaluation and structure–activity relationship of 2-phenylaminomethylene-cyclohexane-1,3-diones as specific anti-tuberculosis agents
http://pubs.rsc.org/en/Content/ArticleLanding/2017/MD/C7MD00350A?utm_source=feedburner&utm_medium=feed&utm_campaign=Feed%3A+rss%2FMD+%28RSC+-+Med.+Chem.+Commun.+latest+articles%29#!divAbstract
Background Image

Muzafar Ahmad Rather

Ph.D Research Scholar

CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Srinagar

Clinical Microbiology and PK/PD Division, Clinical Microbiology PK/PD/Laboratory, CSIR-Indian Institute of Integrative Medicine, Sanatnagar, Srinagar, India-190005

Image result for Zahoor Ahmad CSIR

CSIR-Indian Institute of Integrative Medicine

(Council of Scientific & Industrial Research)

Dr. Zahoor Ahmad Parry

Clinical Microbiology Division
CSIR – Indian Institute of Integrative Medicine,Canal Road, Jammu – 180001
Email: zahoorap@iiim.ac.in
Positions Held
Position Held Date Organization
Sr. Scientist   2010 – Present CSIR-IIIM

Dr. Bilal Ahmad Bhat

Medicinal Chemistry Division
CSIR – Indian Institute of Integrative Medicine,Canal Road, Jammu – 180001
Email: bilal@iiim.ac.in
Positions Held
Position Held Date Organization
Scientist 2010 – Present CSIR-IIIM

Image result for Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Sanatnagar, Srinagar,

Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Sanatnagar, Srinagar,

A small Drug Research Laboratory working under the Government of Jammu & Kashmir was taken over by CSIR in 1957 and named as Regional Research Laboratory, Jammu. Col. Sir Ram Nath Chopra, who is acclaimed the father of modern Pharmacology in India, was the Director of Drug Research Laboratory, continued as the first Director of Regional Research Laboratory. Having significant expertise in the area of medicinal & aromatic plants, Col. Chopra started its related R&D activities such as collection of plants from north & north-west and study the chemistry & pharmacology of the plant extracts and the new molecules isolated from these plants. Thus the initial mandate of this laboratory was mainly focused on screening the flora of north India for new molecules and to study the biological activity of these molecules. Gradually the activities of the institute increased, many more disciplines were introduced, that were important for the exploitation of regional resources such as mineral technology division, paper & pulp, fur technology division, sericulture, food technology division and mycology division. The main stream department such as chemistry, botany and pharmacology were strengthened by the introduction of a small animal house, instrumentation and chemical engineering & design division. The activity of the institute gradually increased which showed up in its publications and technology developments.

With the progress of time, the institute developed high quality expertise and infrastructure for working in the area of plant based products & drugs to explore new botanicals for new molecules and new activity. The institute specialized for working in the area of chemistry of natural products, synthesis of new & nature like molecules. These were studied for their use on various indication such as Oncology, hepatoprotection, anti-bacterial, bio-enhancers, anti-diabetes, anti-inflammation, aphrodisiac, hypertension, immunomodulation, anti-oxidants, oral care and beauty care. Some of the areas which did not progress to the satisfaction level gradually became redundant and were dropped.

Keeping in view the expertise developed in the area of natural products and revised mandate of the institute to explore and exploit natural, nature like and synthetic products with modern scientific tools to reduce the burden of disease, the institute became more focused towards integrative medicine hence was renamed as Indian Institute of Integrative Medicine in 2007 by the governing body of CSIR

////////////////// synthesis, biological evaluation, structure–activity relationship, 2-phenylaminomethylene-cyclohexane-1,3-diones, anti-tuberculosis agents

O=C2CC(C)(C)CC(=O)/C2=C\Nc1ccccc1O

 

DISCLAIMER

“NEW DRUG APPROVALS ” 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

Design, synthesis and biological evaluation of novel 5-hydroxy-2-methyl-4H-pyran-4-one derivatives as antiglioma agents

$
0
0

Med. Chem. Commun., 2018, Advance Article
DOI: 10.1039/C7MD00551B, Research Article
Yi-Bin Li, Wen Hou, Hui Lin, Ping-Hua Sun, Jing Lin, Wei-Min Chen
Two series of 5-hydroxy-2-methyl-4H-pyran-4-one derivatives were synthesized and their antiglioma activities were evaluated.

Design, synthesis and biological evaluation of novel 5-hydroxy-2-methyl-4H-pyran-4-one derivatives as antiglioma agents

Author affiliations

Abstract

D-2-Hydroxyglutarate (D-2HG) is frequently found in human brain cancers. Approximately 50–80% of grade II glioma patients have a high level of D-2HG production, which can lead to cancer initiation. In this study, a series of novel 5-hydroxy-2-methyl-4H-pyran-4-one derivatives were designed and synthesized as antiglioma agents, and their related structure–activity relationships are discussed. Among these novel compounds, 4a exhibited promising anti-proliferative activity against glioma HT1080 cells and U87 cells with an IC50 of 1.43 μM and 4.6 μM, respectively. Further studies found that the most active compound (4a) shows an 86.3% inhibitory rate against the intracellular production of D-2HG at 1 μM, and dramatic inhibitory effects, even at 1 μM on the colony formation and migration of U87 and HT1080 cells.

STR1 STR2 str3 str4
6,6′-((4-(Benzyloxy)phenyl)methylene)bis(5-hydroxy-2-methyl-4H-pyran-4- one) (4a) The reaction was performed according to the general procedure C, using 1 (1.00 g, 7.90 mmol) and 4-(benzyloxy)benzaldehyde (0.84 g, 3.95 mmol).2 The crude product was recrystallized from isopropanol affording a white powder 4a (1.53 g, 87%): mp 261.4-262.1oC; 1HNMR (300 MHz, DMSO-d6)  2.22 (s, 6H, CH3), 5.08 (s, 3H, OCH2- Ph), 5.96 (s, 1H, CH-Ar), 6.25 (s, 2H, C=CH), , 7.01 (d, J = 9.0 Hz, 2H, Ar-H3’/H5’), 7.22 (d, J = 9.0 Hz, 2H, Ar-H2’/H6’), 7.31-7.45 (m, 5H, Ph-H); 13CNMR (75 MHz, DMSO-d6)  173.95, 165.08, 158.12, 151.20, 147.68, 142.19, 140.77, 137.42, 129.87, 128.91, 128.16, 127.69, 115.46, 114.97, 111.74, 69.69, 19.63; ESI-MS m/z: 447.1 [M+H]+ ; ESI-HRMS m/z: 447.1438 [M+H]+ , calcd for C26H23O7 447.1438.

Pfizer’s Monobactam PF-?

$
0
0

STR1

Pfizer’s monobactam PF-?

1380110-34-8, C20 H24 N8 O12 S2, 632.58

Propanoic acid, 2-​[[(Z)​-​[1-​(2-​amino-​4-​thiazolyl)​-​2-​[[(2R,​3S)​-​2-​[[[[[(1,​4-​dihydro-​1,​5-​dihydroxy-​4-​oxo-​2-​pyridinyl)​methyl]​amino]​carbonyl]​amino]​methyl]​-​4-​oxo-​1-​sulfo-​3-​azetidinyl]​amino]​-​2-​oxoethylidene]​amino]​oxy]​-​2-​methyl-

2-((Z)-1-(2-Aminothiazol-4-yl)-2-((2R,3S)-2-((((1,5-dihydroxy-4-oxo-1,4-dihydropyridin-2-yl)methoxy)carbonylamino)methyl)-4-oxo-1-sulfoazetidin-3-ylamino)-2-oxoethylideneaminooxy)-2-methylpropanoic Acid

2-[[(Z)-[1-(2-Amino-4-thiazolyl)-2-[[(2R,3S)-2-[[[[[(1,4-dihydro-1,5-dihydroxy-4-oxo-2-pyridinyl)methyl]amino]carbonyl]amino]methyl]-4-oxo-1-sulfo-3-azetidinyl]amino]-2-oxoethylidene]amino]oxy]-2-methylpropanoic acid

Monobactams are a class of antibacterial agents which contain a monocyclic beta-lactam ring as opposed to a beta-lactam fused to an additional ring which is found in other beta-lactam classes, such as cephalosporins, carbapenems and penicillins. The drug Aztreonam is an example of a marketed monobactam; Carumonam is another example. The early studies in this area were conducted by workers at the Squibb Institute for Medical Research, Cimarusti, C. M. & R.B. Sykes: Monocyclic β-lactam antibiotics. Med. Res. Rev. 1984, 4, 1 -24. Despite the fact that selected

monobacatams were discovered over 25 years ago, there remains a continuing need for new antibiotics to counter the growing number of resistant organisms.

Although not limiting to the present invention, it is believed that monobactams of the present invention exploit the iron uptake mechanism in bacteria through the use of siderophore-monobactam conjugates. For background information, see: M. J. Miller, et al. BioMetals (2009), 22(1 ), 61-75.

The mechanism of action of beta-lactam antibiotics, including monobactams, is generally known to those skilled in the art and involves inhibition of one or more penicillin binding proteins (PBPs), although the present invention is not bound or limited by any theory. PBPs are involved in the synthesis of peptidoglycan, which is a major component of bacterial cell walls.

WO 2012073138

https://www.google.com/patents/WO2012073138A1?cl=en

Inventors Matthew Frank BrownSeungil HanManjinder LallMark. J. Mitton-FryMark Stephen PlummerHud Lawrence RisleyVeerabahu ShanmugasundaramJeremy T. Starr
Applicant Pfizer Inc.

Example 4, Route 1

2-({[(1Z)-1 -(2-amino-1 ,3-thiazol-4-yl)-2-({(2f?,3S)-2-[({[(1 ,5-dihydroxy-4-oxo-1 ,4- dihydropyridin-2-yl)methyl]carbamoyl}amino)methyl]-4-oxo-1 -sulfoazetidin-3- yl}amino)-2-oxoethylidene]amino}oxy)-2-methylpropanoic acid, bis sodium salt

(C92-Bis Na Salt).

Figure imgf000080_0001

C92-bis Na salt

Step 1 : Preparation of C90. A solution of C26 (16.2 g, 43.0 mmol) in tetrahydrofuran (900 mL) was treated with 1 , 1 ‘-carbonyldiimidazole (8.0 g, 47.7 mmol). After 5 minutes, the reaction mixture was treated with a solution of C9 (15 g, 25.0 mmol) in anhydrous tetrahydrofuran (600 mL) at room temperature. After 15 hours, the solvent was removed and the residue was treated with ethyl acetate (500 mL) and water (500 mL). The layers were separated and the aqueous layer was back extracted with additional ethyl acetate (300 mL). The organic layers were combined, washed with brine solution (500 mL), dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified via chromatography on silica gel (ethyl acetate / 2-propanol) to yield C90 as a yellow foam. Yield: 17.44 g, 19.62 mmol, 78%. LCMS m/z 889.5 (M+1 ). 1H NMR (400 MHz, DMSO-d6) 1 1 .90 (br s, 1 H), 9.25 (d, J=8.7 Hz, 1 H), 8.40 (br s, 1 H), 7.98 (s, 1 H), 7.50-7.54 (m, 2H), 7.32-7.47 (m, 8H), 7.28 (s, 1 H), 6.65 (br s, 1 H), 6.28 (br s, 1 H), 5.97 (s, 1 H), 5.25 (s, 2H), 5.18 (dd, J=8.8, 5 Hz, 1 H), 4.99 (s, 2H), 4.16-4.28 (m, 2H), 3.74-3.80 (m, 1 H), 3.29-3.41 (m, 1 H), 3.13-3.23 (m, 1 H), 1.42 (s, 9H), 1.41 (s, 3H), 1.39 (br s, 12H).

Step 2: Preparation of C91. A solution of C90 (8.5 g, 9.6 mmol) in anhydrous N,N- dimethylformamide (100 mL) was treated sulfur trioxide /V,/V-dimethylformamide complex (15.0 g, 98.0 mmol). The reaction was allowed to stir at room temperature for 20 minutes then quenched with water (300 mL). The resulting solid was collected by filtration and dried to yield C91 as a white solid. Yield: 8.1 g, 8.3 mmol, 87%. LCMS m/z 967.6 (M-1 ). 1H NMR (400 MHz, DMSO-d6) δ 1 1.62 (br s, 1 H), 9.29 (d, J=8.8 Hz, 1 H), 9.02 (s, 1 H), 7.58-7.61 (m, 2H), 7.38-7.53 (m, 9H), 7.27 (s, 1 H), 7.07 (s, 1 H), 6.40 (br d, J=8 Hz, 1 H), 5.55 (s, 2H), 5.25 (s, 2H), 5.20 (dd, J=8.8, 5.6 Hz, 1 H), 4.46 (br dd, half of ABX pattern, J=17, 5 Hz, 1 H), 4.38 (br dd, half of ABX pattern, J=17, 6 Hz, 1 H), 3.92-3.98 (m, 1 H), 3.79-3.87 (m, 1 H), 3.07-3.17 (m, 1 H), 1.40 (s, 9H), 1 .39 (s, 3H), 1 .38 (s, 12H).

Step 3: Preparation of C92. A solution of C91 (8.1 g, 8.3 mmol) in anhydrous dichloromethane (200 mL) was treated with 1 M boron trichloride in p-xylenes (58.4 mL, 58.4 mmol) and allowed to stir at room temperature for 15 minutes. The reaction mixture was cooled in an ice bath, quenched with 2,2,2-trifluoroethanol (61 mL), and the solvent was removed in vacuo. A portion of the crude product (1 g) was purified via reverse phase chromatography (C-18 column; acetonitrile / water gradient with 0.1 % formic acid modifier) to yield C92 as a white solid. Yield: 486 mg, 0.77 mmol. LCMS m/z 633.3 (M+1 ). 1H NMR (400 MHz, DMSO-d6) δ 9.22 (d, J=8.7 Hz, 1 H), 8.15 (s, 1 H), 7.26-7.42 (br s, 2H), 7.18-7.25 (m, 1 H), 6.99 (s, 1 H), 6.74 (s, 1 H), 6.32-6.37 (m, 1 H), 5.18 (dd, J=8.7, 5.7 Hz, 1 H), 4.33 (br d, J=4.6 Hz, 2H), 3.94-4.00 (m, 1 H), 3.60-3.68 (m, 1 H), 3.19-3.27 (m, 1 H), 1.40 (s, 3H), 1.39 (s, 3H).

Step 4: Preparation of C92-Bis Na Salt. A flask was charged with C92 (388 mg, 0.61 mmol) and water (5.0 mL). The mixture was cooled in an ice bath and treated dropwise with a solution of sodium bicarbonate (103 mg, 1.52 mmol) in water (5.0 mL). The sample was lyophilized to yield C92-Bis Na Salt as a white solid. Yield: 415 mg, 0.61 mmol, quantitative. LCMS m/z 633.5 (M+1 ). 1H NMR (400 MHz, D20) δ 7.80 (s, 1 H), 6.93 (s, 1 H), 6.76 (s, 1 H), 5.33 (d, J=5.7 Hz, 1 H), 4.44 (ddd, J=6.0, 6.0, 5.7 Hz, 1 H), 4.34 (AB quartet, JAB=17.7 Hz, ΔνΑΒ=10.9 Hz, 2H), 3.69 (dd, half of ABX pattern, J=14.7, 5.8 Hz, 1 H), 3.58 (dd, half of ABX pattern, J=14.7, 6.2 Hz, 1 H), 1.44 (s, 3H), 1.43 (s, 3H).

Alternate preparation of C92

Figure imgf000082_0001

Step 1 : Preparation of C93. An Atlantis pressure reactor was charged with 10% palladium hydroxide on carbon (0.375 g, John Matthey catalyst type A402028-10), C91 (0.75 g, 0.77 mmol) and treated with ethanol (35 mL). The reactor was flushed with nitrogen and pressurized with hydrogen (20 psi) for 20 hours at 20 °C. The reaction mixture was filtered under vacuum and the filtrate was concentrated using the rotary evaporator to yield C93 as a tan solid. Yield: 0.49 g, 0.62 mmol, 80%. LCMS m/z 787.6 (M-1 ). 1H NMR (400 MHz, DMSO-d6) δ 1 1.57 (br s, 1 H), 9.27 (d, J=8.5 Hz, 1 H), 8.16 (s, 1 H), 7.36 (br s, 1 H), 7.26 (s, 1 H), 7.00 (s, 1 H), 6.40 (br s, 1 H), 5.18 (m, 1 H), 4.35 (m, 2H), 3.83 (m, 1 H), 3.41 (m, 1 H), 3.10 (m, 1 H), 1.41 (s, 6H), 1.36 (s, 18H).

Step 2: Preparation of C92. A solution of C93 (6.0 g, 7.6 mmol) in anhydrous dichloromethane (45 mL) at 0 °C was treated with trifluoroacetic acid (35.0 mL, 456 mmol). The mixture was warmed to room temperature and stirred for 2 hours. The reaction mixture was cannulated into a solution of methyl ferf-butyl ether (100 mL) and heptane (200 mL). The solid was collected by filtration and washed with a mixture of methyl ferf-butyl ether (100 mL) and heptane (200 mL) then dried under vacuum. The crude product (~5 g) was purified via reverse phase chromatography (C-18 column; acetonitrile / water gradient with 0.1 % formic acid modifier) and lyophilized to yield C92 as a pink solid. Yield: 1.45 g, 2.29 mmol. LCMS m/z 631.0 (M-1). 1H NMR (400 MHz, DMSO-de) δ 9.20 (d, J=8.7 Hz, 1H), 8.13 (s, 1H), 7.24-7.40 (br s, 2H), 7.16-7.23 (m, 1H), 6.97 (s, 1H), 6.71 (s, 1H), 6.31-6.35 (m, 1H), 5.15 (dd, J=8.7, 5.7 Hz, 1H), 4.31 (br d, J=4.6 Hz, 2H), 3.92-3.98 (m, 1H), 3.58-3.67 (m, 1H), 3.17-3.25 (m, 1H), 1.37 (s, 3H), 1.36 (s, 3H).

Example 4, route 2

2-({[(1Z)-1-(2-amino-1,3-thiazol-4-yl)-2-({(2 ?,3S)-2-[({[(1,5-dihydroxy-4-oxo-^ dihydropyridin-2-yl)methyl]carbamoyl}amino)methyl]-4-oxo-1-sulfoazetidin-3- yl}amino)-2-oxoethylidene]amino}oxy)-2-methylpropanoic acid (C92).

lt

Figure imgf000083_0001

single

enantiomer

Figure imgf000083_0002

Step 1. Preparation of C95. A solution of C94 (50.0 g, 189.9 mmol) in

dichloromethane (100 mL) was treated with trifluoroacetic acid (50.0 mL, 661.3 mmol). The reaction mixture was stirred at room temperature for 24 hours. The dichloromethane and trifluoroacetic acid was displaced with toluene (4 x 150 mL) using vacuum, to a final volume of 120 mL. The solution was added to heptane (250 mL) and the solid was collected by filtration. The solid was washed with a mixture of toluene and heptane (1 : 3, 60 mL), followed by heptane (2 x 80 mL) and dried under vacuum at 50 °C for 19 hours to afford C95 as a solid. Yield: 30.0 g, 158 mmol, 84%. 1H NMR (400 MHz, CDCI3) δ 9.66 (s, 1 H), 7.86 – 7.93 (m, 2H), 7.73 – 7.80 (m, 2H), 4.57 (s, 2H). HPLC retention time 5.1 minutes; column: Agilent Extended C-18 column (75 mm x 3 mm, 3.5 μηη); column temperature 45 °C; flow rate 1.0 mL / minute; detection UV 230 nm; mobile phase: solvent A = acetonitrile (100%), solvent B = acetonitrile (5%) in 10 mM ammonium acetate; gradient elusion: 0-1.5 minutes solvent B (100%), 1.5-10.0 minutes solvent B (5%), 10.0-13.0 minutes solvent B (100%); total run time 13.0 minutes.

Step 2: Preparation of C96-racemic. A solution of C95 (32.75 g; 173.1 mmol) in dichloromethane (550 mL) under nitrogen was cooled to 2 °C. The solution was treated with 2,4-dimethoxybenzylamine (28.94 g, 173.1 mmol) added dropwise over 25 minutes, maintaining the temperature below 10 °C. The solution was stirred for 10 minutes at 2 °C and then treated with molecular sieves (58.36 g, UOP Type 3A). The cold bath was removed and the reaction slurry was stirred for 3 hours at room temperature. The slurry was filtered through a pad of Celite (34.5 g) and the filter cake was rinsed with dichloromethane (135 mL). The dichloromethane filtrate (imine solution) was used directly in the following procedure.

A solution of A/-(ferf-butoxycarbonyl)glycine (60.6 g, 346.1 mmol) in

tetrahydrofuran (622 mL) under nitrogen was cooled to -45 °C and treated with triethylamine (38.5 g, 380.8 mmol). The mixture was stirred for 15 minutes at -45 °C and then treated with ethyl chloroformate (48.8 g, 450 mmol) over 15 minutes. The reaction mixture was stirred at -50 °C for 7 hours. The previously prepared imine solution was added via an addition funnel over 25 minutes while maintaining the reaction mixture temperature below -40 °C. The slurry was treated with triethylamine (17.5 g, 173 mmol) and the reaction mixture was slowly warmed to room temperature over 5 hours and stirred for an additional 12 hours. The reaction slurry was charged with water (150 mL) and the volatiles removed using a rotary evaporator. The reaction mixture was charged with additional water (393 mL) and the volatiles removed using a rotary evaporator. The mixture was treated with methyl ferf-butyl ether (393 mL) and vigorously stirred for 1 hour. The solid was collected by vacuum filtration and the filter cake was rinsed with a mixture of methyl ferf-butyl ether and water (1 : 1 , 400 mL). The solid was collected and dried in a vacuum oven at 50 °C for 16 hours to afford C96- racemic. Yield: 55.8 g, 1 13 mmol, 65%. 1H-NMR (400 MHz, DMSO-d6) δ 7.85 (s, NH), 7.80 (s, 4H), 6.78 (d, J=7.8 Hz, 1 H), 6.25 (m, 1 H), 6.10 (m, 1 H), 4.83 (m, 1 H), 4.38 (d, J=9.5 Hz, 1 H), 3.77-3.95 (m, 3H), 3.62 (s, 3H), 3.45 (m, 1 H), 3.40 (s, 3H), 1.38 (s, 9H). HPLC retention time 6.05 minutes; XBridge C8 column (4.6 x 75 mm, 3.5 μηη); column temperature 45 °C; flow rate 2.0 mL/minute; detection UV 210 nm, 230 nm, and 254 nm; mobile phase: solvent A = methanesulfonic acid (5%) in 10 mmol sodium octylsulfonate, solvent B = acetonitrile (100%); gradient elusion: 0-1.5 minutes solvent A (95%) and solvent B (5%), 1.5-8.5 minutes solvent A (5%) and solvent B (95%), 8.5- 10.0 minutes solvent A (5%) and solvent B (95%), 10.01 -12.0 minutes solvent A (95%) and solvent B (5%); total run time 12.0 minutes.

Step 3: Preparation of C97-racemic. A solution of C96-racemic (15.0 g, 30.3 mmol) in ethyl acetate (150 mL) under nitrogen was treated with ethanolamine (27.3 mL, 454.1 mmol). The reaction mixture was heated at 90 °C for 3 hours and then cooled to room temperature. The mixture was charged with water (150 mL) and the layers separated. The aqueous layer was extracted with ethyl acetate (75 mL) and the combined organic layers washed with water (2 x 150 mL) followed by saturated aqueous sodium chloride (75 mL). The organic layer was dried over magnesium sulfate, filtered and the filtrate concentrated to a volume of 38 mL. The filtrate was treated with heptane (152 mL) and the solid was collected by filtration. The solid was washed with heptane and dried at 50 °C in a vacuum oven overnight to yield C97-racemic as a solid. Yield: 9.68 g, 26.5 mmol, 88%. LCMS m/z 967.6 (M-1 ). 1H NMR (400 MHz, DMSO-d6) δ 7.64 (d, J=9.4 Hz, 1 H), 7.14 (d, J=8.2 Hz, 1 H), 6.56 (s, 1 H), 6.49 (dd, J=8.20, 2.3 Hz, 1 H), 4.78 (dd, J=9.37, 5.1 Hz, 1 H), 4.30 (d, J=14.8 Hz, 1 H), 4.14 (d, J=14.8 Hz, 1 H), 3.77 (s, 3H), 3.75 (s, 3H), 3.45 – 3.53 (m, 1 H), 2.65 – 2.75 (m, 1 H), 2.56 – 2.64 (m, 1 H), 1.38 (s, 9H), 1.30 – 1.35 (m, 2H). HPLC retention time 5.1 minutes; column: Agilent Extended C-18 column (75 mm x 3 mm, 3.5 μΐη); column temperature 45 °C; flow rate 1.0 mL / minute;

detection UV 230 nm; mobile phase: solvent A = acetonitrile (100%), solvent B = acetonitrile (5%) in 10 mM ammonium acetate; gradient elusion: 0-1 .5 minutes solvent B (100%), 1 .5-10.0 minutes solvent B (5%), 10.0-13.0 minutes solvent B (100%); total run time 13.0 minutes. Step 4: Preparation of C97-(2R,3S) enantiomer. A solution of C97-racemic (20.0 g, 54.7 mmol) in ethyl acetate (450 mL) was treated with diatomaceous earth (5.0 g) and filtered through a funnel charged with diatomaceous earth. The filter cake was washed with ethyl acetate (150 mL). The filtrate was charged with diatomaceous earth (20.0 g) and treated with (-)-L-dibenzoyltartaric acid (19.6 g, 54.7 mmol). The slurry was heated at 60 °C for 1.5 hours and then cooled to room temperature. The slurry was filtered and the solid washed with ethyl acetate (90 mL). The solid was collected and dried at 50 °C in a vacuum oven for 17 hours to yield C97-(2R,3S) enantiomer as a solid (mixed with diatomaceous earth). Yield: 17.3 g, 23.9 mmol, 43.6%, 97.6% ee. 1H NMR (400 MHz, DMSO-de) δ 7.89 – 7.91 (m, 4H), 7.59 – 7.65 (m, 3H), 7.44 – 7.49 (m, 4H), 7.09 (d, J=8.3 Hz, 1 H), 6.53 (d, J=2.3 Hz, 1 H), 6.49 (dd, J=8.3, 2.3 Hz, 1 H), 5.65 (s, 2H), 4.85 (dd, J=9.3, 4.9 Hz, 1 H), 4.30 (d, J=15.3 Hz, 1 H), 4.10 (d, J=15.3 Hz, 1 H), 3.74 (s, 3H), 3.72 (s, 3H), 3.68 – 3.70 (m, 1 H), 2.92 – 2.96 (dd, J=13.6, 5.4 Hz, 1 H), 2.85 – 2.90 (dd, J=13.6, 6.3 Hz, 1 H), 1.36 (s, 9H). HPLC retention time 5.1 minutes; column: Agilent Extended C-18 column (75 mm x 3 mm, 3.5 μηη); column temperature 45 °C; flow rate 1.0 mL / minute; detection UV 230 nm; mobile phase: solvent A = acetonitrile (100%), solvent B = acetonitrile (5%) in 10 mM ammonium acetate; gradient elusion: 0-1 .5 minutes solvent B (100%), 1.5-10.0 minutes solvent B (5%), 10.0-13.0 minutes solvent B (100%); total run time 13.0 minutes. Chiral HPLC retention time 9.1 minutes; column: Chiralcel OD-H column (250 mm x 4.6 mm); column temperature 40 °C; flow rate 1 .0 mL / minute; detection UV 208 nm; mobile phase: solvent A = ethanol (18%), solvent B = heptane (85%); isocratic elusion; total run time 20.0 minutes.

Step 5: Preparation of C98-(2R,3S) enantiomer. A solution of C97-(2R,3S) enantiomer. (16.7 g, 23.1 mmol) in ethyl acetate (301 mL) was treated with diatomaceous earth (18.3 g) and 5% aqueous potassium phosphate tribasic (182 mL). The slurry was stirred for 30 minutes at room temperature, then filtered under vacuum and the filter cake washed with ethyl acetate (2 x 67 mL). The filtrate was washed with 5% aqueous potassium phosphate tribasic (18 mL) and the organic layer dried over magnesium sulfate. The solid was filtered and the filter cake washed with ethyl acetate (33 mL). The filtrate was concentrated to a volume of 42 mL and slowly added to heptane (251 mL) and the resulting solid was collected by filtration. The solid was washed with heptane and dried at 50 °C in a vacuum oven for 19 hours to yield C98- (2R,3S) enantiomer as a solid. Yield: 6.4 g, 17.5 mmol, 76%, 98.8% ee. 1H NMR (400 MHz, DMSO-de) δ 7.64 (d, J=9.4 Hz, 1 H), 7.14 (d, J=8.2 Hz, 1 H), 6.56 (s, 1 H), 6.49 (dd, J=8.20, 2.3 Hz, 1 H), 4.78 (dd, J=9.37, 5.1 Hz, 1 H), 4.30 (d, J=14.8 Hz, 1 H), 4.14 (d, J=14.8 Hz, 1 H), 3.77 (s, 3H), 3.75 (s, 3H), 3.45 – 3.53 (m, 1 H), 2.65 – 2.75 (m, 1 H), 2.56 – 2.64 (m, 1 H), 1.38 (s, 9H), 1.30 – 1.35 (m, 2H). HPLC retention time 5.2 minutes; column: Agilent Extended C-18 column (75 mm x 3 mm, 3.5 μηη); column temperature 45 °C; flow rate 1.0 mL / minute; detection UV 230 nm; mobile phase: solvent A = acetonitrile (100%), solvent B = acetonitrile (5%) in 10 mM ammonium acetate; gradient elusion: 0-1 .5 minutes solvent B (100%), 1.5-10.0 minutes solvent B (5%), 10.0-13.0 minutes solvent B (100%); total run time 13.0 minutes. Chiral HPLC retention time 8.7 minutes; column: Chiralcel OD-H column (250 mm x 4.6 mm); column temperature 40 °C; flow rate 1.0 mL / minute; detection UV 208 nm; mobile phase: solvent A = ethanol (18%), solvent B = heptane (85%); isocratic elusion; total run time 20.0 minutes.

Step 6: Preparation of C99. A solution of potassium phosphate tribasic N-hydrate (8.71 g, 41 .05 mmol) in water (32.0 mL) at 22 °C was treated with a slurry of C26- mesylate salt (12.1 g, 27.4 mmol, q-NMR potency 98%) in dichloromethane (100.00 mL). The slurry was stirred for 1 hour at 22 °C. The reaction mixture was transferred to a separatory funnel and the layers separated. The aqueous layer was back extracted with dichloromethane (50.0 mL). The organic layers were combined, dried over magnesium sulfate, filtered under vacuum and the filter cake washed with

dichloromethane (2 x 16 mL). The filtrate (-190 mL, amine solution) was used directly in the next step.

A solution of 1 ,1 ‘-carbonyldiimidazole (6.66 g, 41 .0 mmol) in dichloromethane (100 mL) at 22 °C under nitrogen was treated with the previously prepared amine solution (-190 mL) added dropwise using an addition funnel over 3 hour at 22 °C with stirring. After the addition, the mixture was stirred for 1 hour at 22 °C, then treated with C98-(2R,3S) enantiomer. (10.0 g, 27.4 mmol) followed by /V,/V-dimethylformamide (23.00 mL). The reaction mixture was stirred at 22 °C for 3 hours and then heated at 40 °C for 12 hours. The solution was cooled to room temperature and the dichloromethane was removed using the rotary evaporator. The reaction mixture was diluted with ethyl acetate (216.0 mL) and washed with 10% aqueous citric acid (216.0 mL), 5% aqueous sodium chloride (2 x 216.0 mL), dried over magnesium sulfate and filtered under vacuum. The filter cake was washed with ethyl acetate (3 x 13 mL) and the ethyl acetate solution was concentrated on the rotary evaporator to a volume of (-1 10.00 mL) providing a suspension. The suspension (~1 10.00 mL) was warmed to 40 °C and transferred into a stirred solution of heptane (22 °C) over 1 hour, to give a slurry. The slurry was stirred for 1 hour and filtered under vacuum. The filter cake was washed with heptane (3 x 30 mL) and dried under vacuum at 50 °C for 12 hours to afford C99 as a solid. Yield: 18.1 g, 24.9 mmol, 92%. LCMS m/z 728.4 (M+1 ). 1H NMR (400 MHz, DMSO-d6) δ 8.09 (s, 1 H), 7.62 (d, J=9.4 Hz, 1 H), 7.33-7.52 (m, 10H), 7.07 (d, J=8.3 Hz, 1 H), 6.51 (d, J=2.3 Hz, 1 H), 6.50 (m, 1 H), 6.44 (dd, J=8.3, 2.3 Hz, 1 H), 6.12 (m, 1 H), 6.07 (s, 1 H), 5.27 (s, 2H), 5.00 (s, 2H), 4.73 (dd, J=9.4, 5.2 Hz, 1 H), 4.38 (d, J=15.0 Hz, 1 H), 4.19 (m, 2H), 3.99 (d, J=15.0 Hz, 1 H), 3.72 (s, 3H), 3.71 (s, 3H), 3.48 (m, 1 H), 3.28 (m, 1 H), 3.12 (m, 1 H), 1 .37 (s, 9H).

Step 7: Preparation of C100. A solution of C99 (46.5 g, 63.9 mmol) in acetonitrile (697 mL and water (372 mL) was treated with potassium persulfate (69.1 g, 255.6 mmol) and potassium phosphate dibasic (50.1 g, 287.5 mmol). The biphasic mixture was heated to 75 °C and vigorously stirred for 1.5 hours. The pH was maintained between 6.0-6.5 by potassium phosphate dibasic addition (-12 g). The mixture was cooled to 20 °C, the suspension was filtered and washed with acetonitrile (50 mL). The filtrate was concentrated using the rotary evaporator and treated with water (50 mL) followed by ethyl acetate (200 mL). The slurry was stirred for 2 hours at room temperature, filtered and the solid dried under vacuum at 40 °C overnight. The solid was slurried in a mixture of ethyl acetate and water (6 : 1 , 390.7 mL) at 20 °C for 1 hour then collected by filtration. The solid was dried in a vacuum oven to yield C100. Yield: 22.1 g, 38.3 mmol, 60%. 1H NMR (400 MHz, DMSO-d6) δ 8.17 (br s, 1 H), 7.96 (s, 1 H), 7.58 (d, J=9.6 Hz, 1 H), 7.29-7.50 (m, 10H), 6.49 (dd, J=8.0, 6.0 Hz, 1 H), 6.08 (dd, J=5.6, 5.2 Hz, 1 H), 5.93 (s, 1 H), 5.22 (s, 2H), 4.96 (s, 2H), 4.77 (dd, J=9.6, 5.0 Hz, 1 H), 4.16 (m, 2H), 3.61 (m, 1 H), 3.1 1 (m, 2H), 1.36 (s, 9H). HPLC retention time 6.17 minutes; XBridge C8 column (4.6 x 75 mm, 3.5 μηη); column temperature 45 °C; flow rate 2.0 mL/minute; detection UV 210 nm, 230 nm, and 254 nm; mobile phase: solvent A = methanesulfonic acid (5%) in 10 mmol sodium octylsulfonate, solvent B = acetonitrile (100%); gradient elusion: 0-1 .5 minutes solvent A (95%) and solvent B (5%), 1.5-8.5 minutes solvent A (5%) and solvent B (95%), 8.5-10.0 minutes solvent A (5%) and solvent B (95%), 10.01- 12.0 minutes solvent A (95%) and solvent B (5%); total run time 12.0 minutes.

Step 8: Preparation of C101. A solution of trifluoroacetic acid (120 mL, 1550 mmol) under nitrogen was treated with methoxybenzene (30 mL, 269 mmol) and cooled to -5 °C. Solid C100 (17.9 g, 31.0 mmol) was charged in one portion at -5 °C and the resulting mixture stirred for 3 hours. The reaction mixture was cannulated with nitrogen pressure over 15 minutes to a stirred mixture of Celite (40.98 g) and methyl ferf-butyl ether (550 mL) at 10 °C. The slurry was stirred at 16 °C for 30 minutes, then filtered under vacuum. The filter cake was rinsed with methyl ferf-butyl ether (2 x 100 mL). The solid was collected and slurried in methyl ferf-butyl ether (550 mL) with vigorous stirring for 25 minutes. The slurry was filtered by vacuum filtration and washed with methyl ferf-butyl ether (2 x 250 mL). The solid was collected and dried in a vacuum oven at 60 °C for 18 hours to afford C101 on Celite. Yield: 57.6 g total = C101 + Celite; 16.61 g C101 , 28.1 mmol, 91%. 1H NMR (400 MHz, DMSO-d6) δ 8.75-8.95 (br s, 2H), 8.65 (s, 1 H), 8.21 (s, 1 H), 7.30-7.58 (m, 10H), 6.83 (br s, 1 H), 6.65 (br s, 1 H), 6.17 (s, 1 H), 5.30 (s, 2H), 5.03 (s, 2H), 4.45 (br s, 1 H), 4.22 (br s, 2H), 3.77 (m, 1 H), 3.36 (m, 1 H), 3.22 (m, 1 H). 19F NMR (376 MHz, DMSO-d6) δ -76.0 (s, 3F). HPLC retention time 5.81 minutes; XBridge C8 column (4.6 x 75 mm, 3.5 μηη); column temperature 45 °C; flow rate 2.0 mL/minute; detection UV 210 nm, 230 nm, and 254 nm; mobile phase: solvent A = methanesulfonic acid (5%) in 10 mmol sodium octylsulfonate, solvent B = acetonitrile (100%); gradient elusion: 0-1.5 minutes solvent A (95%) and solvent B (5%), 1.5-8.5 minutes solvent A (5%) and solvent B (95%), 8.5-10.0 minutes solvent A (5%) and solvent B (95%), 10.01-12.0 minutes solvent A (95%) and solvent B (5%); total run time 12.0 minutes.

Step 9: Preparation of C90. A suspension of C101 (67.0 g, 30% activity on Celite = 33.9 mmol) in acetonitrile (281 .4 mL) was treated with molecular sieves 4AE (40.2 g), C5 (17.9 g, 33.9 mmol), 4-dimethylaminopyridine (10.4 g, 84.9 mmol) and the mixture was stirred at 40°C for 16 hours. The reaction mixture was cooled to 20 °C, filtered under vacuum and the filter cake washed with acetonitrile (2 x 100 mL). The filtrate was concentrated under vacuum to a volume of -50 mL. The solution was diluted with ethyl acetate (268.0 mL) and washed with 10% aqueous citric acid (3 x 134 mL) followed by 5% aqueous sodium chloride (67.0 mL). The organic layer was dried over magnesium sulfate and filtered under vacuum. The filter cake was washed with ethyl acetate (2 x 50 mL) and the filtrate was concentrated to a volume of -60 mL. The filtrate was added slowly to heptane (268 mL) with stirring and the slurry was stirred at 20 °C for 1 hour. The slurry was filtered under vacuum and the filter cake washed with a mixture of heptane and ethyl acetate (4: 1 , 2 x 27 mL). The solid was collected and dried under vacuum for 12 hours at 50 °C to afford a solid. The crude product was purified via chromatography on silica gel (ethyl acetate / 2-propanol), product bearing fractions were combined and the volume was reduced to -60 mL. The solution was added dropwise to heptane (268 mL) with stirring. The slurry was stirred at room temperature for 3 hours, filtered and washed with heptane and ethyl acetate (4: 1 , 2 x 27 mL). The solid was collected and dried under vacuum for 12 hours at 50 °C to afford C90 as a solid. Yield: 16.8 g, 18.9 mmol, 58%. LCMS m/z 889.4 (M+1 ). 1H NMR (400 MHz, DMSO-cfe) 1 1.90 (br s, 1 H), 9.25 (d, J=8.7 Hz, 1 H), 8.40 (br s, 1 H), 7.98 (s, 1 H), 7.50-7.54 (m, 2H), 7.32- 7.47 (m, 8H), 7.28 (s, 1 H), 6.65 (br s, 1 H), 6.28 (br s, 1 H), 5.97 (s, 1 H), 5.25 (s, 2H), 5.18 (dd, J=8.8, 5 Hz, 1 H), 4.99 (s, 2H), 4.16-4.28 (m, 2H), 3.74-3.80 (m, 1 H), 3.29-3.41 (m, 1 H), 3.13-3.23 (m, 1 H), 1 .42 (s, 9H), 1 .41 (s, 3H), 1.39 (br s, 12H).

Step 10: Preparation of C91. A solution of C90 (14.5 g, 16.3 mmol) in anhydrous N,N- dimethylformamide (145.0 mL) was treated with sulfur trioxide /V,/V-dimethylformamide complex (25.0 g, 163.0 mmol). The reaction mixture was stirred at room temperature for 45 minutes, then transferred to a stirred mixture of 5% aqueous sodium chloride (290 mL) and ethyl acetate (435 mL) at 0 °C. The mixture was warmed to 18 °C and the layers separated. The aqueous layer was extracted with ethyl acetate (145 mL) and the combined organic layers washed with 5% aqueous sodium chloride (3 x 290 mL) followed by saturated aqueous sodium chloride (145 mL). The organic layer was dried over magnesium sulfate, filtered through diatomaceous earth and the filter cake washed with ethyl acetate (72 mL). The filtrate was concentrated to a volume of 36 mL and treated with methyl ferf-butyl ether (290 mL), the resulting slurry was stirred at room temperature for 1 hour. The solid was collected by filtration, washed with methyl ferf- butyl ether (58 mL) and dried at 50 °C for 2 hours followed by 20 °C for 65 hours in a vacuum oven to yield C91 as a solid. Yield: 15.0 g, 15.4 mmol, 95%. LCMS m/z 967.6 (M-1 ). 1H NMR (400 MHz, DMSO-d6) δ 1 1.62 (br s, 1 H), 9.29 (d, J=8.8 Hz, 1 H), 9.02 (s, 1 H), 7.58-7.61 (m, 2H), 7.38-7.53 (m, 9H), 7.27 (s, 1 H), 7.07 (s, 1 H), 6.40 (br d, J=8.0 Hz, 1 H), 5.55 (s, 2H), 5.25 (s, 2H), 5.20 (dd, J=8.8, 5.6 Hz, 1 H), 4.46 (br dd, half of ABX pattern, J=17.0, 5.0 Hz, 1 H), 4.38 (br dd, half of ABX pattern, J=17.0, 6.0 Hz, 1 H), 3.92- 3.98 (m, 1 H), 3.79-3.87 (m, 1 H), 3.07-3.17 (m, 1 H), 1.40 (s, 9H), 1.39 (s, 3H), 1.38 (s, 12H).

Step 11 : Preparation of C92. A solution of C91 (20.0 g, 20.6 mmol) in

dichloromethane (400 mL) was concentrated under reduced pressure (420 mmHg) at 45 °C to a volume of 200 mL. The solution was cooled to -5 °C and treated with 1 M boron trichloride in dichloromethane (206.0 mL, 206.0 mmol) added dropwise over 40 minutes. The reaction mixture was warmed to 15 °C over 1 hour with stirring. The slurry was cooled to -15 °C and treated with a mixture of 2,2,2-trifluoroethanol (69.2 mL) and methyl ferf-butyl ether (400 mL), maintaining the temperature at -15 °C. The reaction mixture was warmed to 0 °C over 1 hour. The suspension was filtered using nitrogen pressure and the solid washed with methyl ferf-butyl ether (2 x 200 mL).

Nitrogen was passed over the solid for 2 hours. The solid was collected and suspended in methyl ferf-butyl ether (400 mL) for 1 hour with stirring at 18 °C. The suspension was filtered using nitrogen pressure and the solid washed with methyl ferf-butyl ether (2 x 200 mL). Nitrogen was passed over the resulting solid for 12 hours. A portion of the crude product was neutralized with 1 M aqueous ammonium formate to pH 5.5 with minimal addition of /V,/V-dimethylformamide to prevent foaming. The feed solution was filtered and purified via reverse phase chromatography (C-18 column; acetonitrile / water gradient with 0.2% formic acid modifier). The product bearing fractions were combined and concentrated to remove acetonitrile. The solution was captured on a GC-161 M column, washed with deionized water and blown dry with nitrogen pressure. The product was released using a mixture of methanol / water (10: 1 ) and the product bearing fractions were added to a solution of ethyl acetate (6 volumes). The solid was collected by filtration to afford C92 as a solid. Yield: 5.87 g, 9.28 mmol. LCMS m/z 633.3 (M+1 ). 1H NMR (400 MHz, DMSO-d6) δ 9.22 (d, J=8.7 Hz, 1 H), 8.15 (s, 1 H), 7.26-7.42 (br s, 2H), 7.18-7.25 (m, 1 H), 6.99 (s, 1 H), 6.74 (s, 1 H), 6.32-6.37 (m, 1 H), 5.18 (dd, J=8.7, 5.7 Hz, 1 H), 4.33 (br d, J=4.6 Hz, 2H), 3.94-4.00 (m, 1 H), 3.60-3.68 (m, 1 H), 3.19-3.27 (m, 1 H), 1.40 (s, 3H), 1.39 (s, 3H).

PAPER

Journal of Medicinal Chemistry (2014), 57(9), 3845-3855

Siderophore Receptor-Mediated Uptake of Lactivicin Analogues in Gram-Negative Bacteria

Medicinal Chemistry, Computational Chemistry, §Antibacterials Research Unit, and Structural Biology, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
J. Med. Chem.201457 (9), pp 3845–3855
DOI: 10.1021/jm500219c
Publication Date (Web): April 2, 2014
Copyright © 2014 American Chemical Society
*Phone: (860)-686-1788. E-mail: seungil.han@pfizer.com.

Abstract

Abstract Image

Multidrug-resistant Gram-negative pathogens are an emerging threat to human health, and addressing this challenge will require development of new antibacterial agents. This can be achieved through an improved molecular understanding of drug–target interactions combined with enhanced delivery of these agents to the site of action. Herein we describe the first application of siderophore receptor-mediated drug uptake of lactivicin analogues as a strategy that enables the development of novel antibacterial agents against clinically relevant Gram-negative bacteria. We report the first crystal structures of several sideromimic conjugated compounds bound to penicillin binding proteins PBP3 and PBP1a from Pseudomonas aeruginosa and characterize the reactivity of lactivicin and β-lactam core structures. Results from drug sensitivity studies with β-lactamase enzymes are presented, as well as a structure-based hypothesis to reduce susceptibility to this enzyme class. Finally, mechanistic studies demonstrating that sideromimic modification alters the drug uptake process are discussed.

PAPER

Pyridone-Conjugated Monobactam Antibiotics with Gram-Negative Activity

Worldwide Medicinal Chemistry, Computational Chemistry, §Antibacterials Research Unit, Pharmacokinetics, Dynamics & Metabolism, Structural Biology, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
J. Med. Chem.201356 (13), pp 5541–5552
DOI: 10.1021/jm400560z
Publication Date (Web): June 11, 2013
Copyright © 2013 American Chemical Society
*Phone: 860-441-3522. E-mail: matthew.f.brown@pfizer.com.
Abstract Image

Herein we describe the structure-aided design and synthesis of a series of pyridone-conjugated monobactam analogues with in vitro antibacterial activity against clinically relevant Gram-negative species including Pseudomonas aeruginosaKlebsiella pneumoniae, and Escherichia coli. Rat pharmacokinetic studies with compound 17 demonstrate low clearance and low plasma protein binding. In addition, evidence is provided for a number of analogues suggesting that the siderophore receptors PiuA and PirA play a role in drug uptake in P. aeruginosa strain PAO1.

STR1

17 as a solid. Yield: 5.87 g, 9.28 mmol. LCMS m/z 633.3 (M+1). 1H NMR (400 MHz, DMSOd6) δ 9.22 (d, J=8.7 Hz, 1H), 8.15 (s, 1H), 7.26-7.42 (br s, 2H), 7.18-7.25 (m, 1H), 6.99 (s, 1H), 6.74 (s, 1H), 6.32-6.37 (m, 1H), 5.18 (dd, J=8.7, 5.7 Hz, 1H), 4.33 (br d, J=4.6 Hz, 2H), 3.94-4.00 (m, 1H), 3.60-3.68 (m, 1H), 3.19-3.27 (m, 1H), 1.40 (s, 3H), 1.39 (s, 3H).

Nc1nc(cs1)\C(=N\OC(C)(C)C(=O)O)C(=O)N[C@@H]3C(=O)N([C@@H]3CNC(=O)NCC2=CC(=O)C(O)=CN2O)S(=O)(=O)O

PAPER

Process Development for the Synthesis of Monocyclic β-Lactam Core 17

Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.7b00359
Publication Date (Web): January 4, 2018
Copyright © 2018 American Chemical Society
Abstract Image

Process development and multikilogram synthesis of the monocyclic β-lactam core 17 for a novel pyridone-conjugated monobactam antibiotic is described. Starting with commercially available 2-(2,2-diethoxyethyl)isoindoline-1,3-dione, the five-step synthesis features several telescoped operations and direct isolations to provide significant improvement in throughput and reduced solvent usage over initial scale-up campaigns. A particular highlight in this effort includes the development of an efficient Staudinger ketene–imine [2 + 2] cycloaddition reaction of N-Boc-glycine ketene 12 and imine 9 to form racemic β-lactam 13 in good isolated yield (66%) and purity (97%). Another key feature in the synthesis involves a classical resolution of racemic amine 15 to afford single enantiomer salt 17 in excellent isolated yield (45%) with high enantiomeric excess (98%).

Figure

https://pubs.acs.org/doi/suppl/10.1021/acs.oprd.7b00359/suppl_file/op7b00359_si_001.pdf

Nc1nc(cs1)\C(=N\OC(C)(C)C(=O)O)C(=O)N[C@@H]3C(=O)N([C@@H]3CNC(=O)NCC2=CC(=O)C(O)=CN2O)S(=O)(=O)O

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

J. Med. Chem.201356 (13), pp 5541–5552
DOI: 10.1021/jm400560z

OXYGEN ANALOGUE…………..

STR2
 1380110-45-1, C20 H23 N7 O13 S2, 633.57
Propanoic acid, 2-​[[(Z)​-​[1-​(2-​amino-​4-​thiazolyl)​-​2-​[[(2R,​3S)​-​2-​[[[[(1,​4-​dihydro-​1,​5-​dihydroxy-​4-​oxo-​2-​pyridinyl)​methoxy]​carbonyl]​amino]​methyl]​-​4-​oxo-​1-​sulfo-​3-​azetidinyl]​amino]​-​2-​oxoethylidene]​amino]​oxy]​-​2-​methyl-
2-[[(Z)-[1-(2-Amino-4-thiazolyl)-2-[[(2R,3S)-2-[[[[(1,4-dihydro-1,5-dihydroxy-4-oxo-2-pyridinyl)methoxy]carbonyl]amino]methyl]-4-oxo-1-sulfo-3-azetidinyl]amino]-2-oxoethylidene]amino]oxy]-2-methylpropanoic acid

STR2

18 as a light yellow solid. Yield: 43 mg, 0.068 mmol, 51%. LCMS m/z 634.4 (M+1). 1H NMR (400 MHz, DMSO-d6), characteristic peaks: δ 9.29 (d, J=8.5 Hz, 1H), 8.10 (s, 1H), 7.04-7.10 (m, 1H), 7.00 (s, 1H), 6.75 (s, 1H), 5.05-5.30 (m, 3H), 4.00-4.07 (m, 1H), 1.42 (s, 3H), 1.41 (s, 3H).

Nc1nc(cs1)\C(=N\OC(C)(C)C(=O)O)C(=O)N[C@@H]3C(=O)N([C@@H]3CNC(=O)OCC2=CC(=O)C(O)=CN2O)S(=O)(=O)O

Step 4: Preparation of 18-Bis Na salt. A suspension of 5 (212 mg, 0.33 mmol) in water (10 mL) was cooled to 0 oC and treated with a solution of sodium bicarbonate (56.4 mg, 0.67 mmol) in water (2 mL), added dropwise. The reaction mixture was cooled to -70 oC (frozen) and lyophilized to afford 18-Bis Na salt as a white solid. Yield: 210 mg, 0.31 mmol, 93%. LCMS m/z 632.5 (M-1). 1H NMR (400 MHz, D2O) δ 7.87 (s, 1H), 6.94 (s, 1H), 6.92 (s, 1H), 5.35 (d, J=5 Hz, 1H), 5.16 (s, 2H), 4.46-4.52 (m, 1H), 3.71 (dd, half of ABX pattern, J=14.5, 6 Hz, 1H), 3.55 (dd, half of ABX pattern, J=14.5, 6 Hz, 1H), 1.43 (s, 3H), 1.42 (s, 3H).

WO 2012073138

Inventors Matthew Frank BrownSeungil HanManjinder LallMark. J. Mitton-FryMark Stephen PlummerHud Lawrence RisleyVeerabahu ShanmugasundaramJeremy T. Starr
Applicant Pfizer Inc.

Example 5

disodium 2-({[(1Z)-1 -(2-amino-1 ,3-thiazol-4-yl)-2-({(2R,3S)-2-[({[(1 ,5-dihydroxy-4- oxo-1 ,4-dihydropyridin-2-yl)methoxy]carbonyl}amino)methyl]-4-oxo-1 – sulfonatoazetidin-3-yl}amino)-2-oxoethylidene]amino}oxy)-2-methylpropanoate

(C104-Bis Na salt).

Figure imgf000092_0001

Step 1 : Preparation of C102. A solution of C28 (300 mg, 0.755 mmol) in

tetrahydrofuran (10 mL) was treated with 1 , 1 ‘-carbonyldiimidazole (379 mg, 2.26 mmol) at room temperature and stirred for 20 hours. The yellow reaction mixture was treated with a solution of C9 (286 mg, 0.543 mmol) in tetrahydrofuran (25 mL). The mixture was stirred for 6 hours at room temperature, then treated with water (20 mL) and extracted with ethyl acetate (3 x 25 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated in vacuo. The crude material was purified via chromatography on silica gel (heptane / ethyl acetate / 2-propanol) to afford C102 as a light yellow solid. Yield: 362 mg, 0.381 mmol, 62%. LCMS m/z 950.4 (M+1 ). 1H NMR (400 MHz, DMSO-de), characteristic peaks: δ 9.31 (d, J=8.4 Hz, 1 H), 8.38 (s, 1 H), 8.00 (s, 1 H), 7.41 (br d, J=8.2 Hz, 2H), 7.36 (br d, J=8.8 Hz, 2H), 7.26 (s, 1 H), 6.10 (s, 1 H), 5.20 (s, 2H), 4.92 (br s, 4H), 3.77 (s, 3H), 3.76 (s, 3H), 1.45 (s, 9H), 1.38 (s, 9H). Step 2: Preparation of C103. A solution of C102 (181 mg, 0.191 mmol) in anhydrous /V,/V-dimethylformamide (2.0 mL) was treated with sulfur trioxide pyridine complex (302 mg, 1.91 mmol). The reaction mixture was allowed to stir at room temperature for 6 hours, then cooled to 0 °C and quenched with water. The resulting solid was collected by filtration and dried in vacuo to yield C103 as a white solid. Yield: 145 mg, 0.14 mmol, 74%. APCI m/z 1028.5 (M-1 ). 1H NMR (400 MHz, DMSO-d6), characteristic peaks: δ 1 1.65 (br s, 1 H), 9.37 (d, J=8.6 Hz, 1 H), 8.87 (s, 1 H), 7.49 (br d, J=8.6 Hz, 2H), 7.43 (br d, J=8.6 Hz, 2H), 7.26 (s, 1 H), 7.01 (br d, J=8.9 Hz, 2H), 7.00 (br d, J=8.8 Hz, 2H), 5.43 (s, 2H), 5.20 (dd, J=8.4, 6 Hz, 1 H), 4.01-4.07 (m, 1 H), 3.78 (s, 3H), 3.77 (s, 3H), 3.50- 3.58 (m, 1 H), 3.29-3.37 (m, 1 H), 1.44 (s, 9H), 1.37 (s, 9H). Step 3: Preparation of C104. A solution of C103 (136 mg, 0.132 mmol) in anhydrous dichloromethane (5 mL) was treated with 1 M boron trichloride in p-xylenes (0.92 mL, 0.92 mmol) and allowed to stir at room temperature for 40 minutes. The reaction mixture was cooled in an ice bath, quenched with water (0.4 mL), and transferred into a solution of methyl ferf-butyl ether: heptane (1 :2, 12 mL). The solvent was removed in vacuo and the crude product was purified via reverse phase chromatography (C-18 column; acetonitrile / water gradient with 0.1 % formic acid modifier) to yield C104 as a light yellow solid. Yield: 43 mg, 0.068 mmol, 51 %. LCMS m/z 634.4 (M+1 ). 1H NMR (400 MHz, DMSO-de), characteristic peaks: δ 9.29 (d, J=8.5 Hz, 1 H), 8.10 (s, 1 H), 7.04- 7.10 (m, 1 H), 7.00 (s, 1 H), 6.75 (s, 1 H), 5.05-5.30 (m, 3H), 4.00-4.07 (m, 1 H), 1 .42 (s, 3H), 1 .41 (s, 3H).

Step 4: Preparation of C104-Bis Na salt. A suspension of C104 (212 mg, 0.33 mmol) in water (10 mL) was cooled to 0 °C and treated with a solution of sodium bicarbonate (56.4 mg, 0.67 mmol) in water (2 mL), added dropwise. The reaction mixture was cooled to -70 °C (frozen) and lyophilized to afford C104-Bis Na salt as a white solid. Yield: 210 mg, 0.31 mmol, 93%. LCMS m/z 632.5 (M-1 ). 1H NMR (400 MHz, D20) δ 7.87 (s, 1 H), 6.94 (s, 1 H), 6.92 (s, 1 H), 5.35 (d, J=5 Hz, 1 H), 5.16 (s, 2H), 4.46-4.52 (m, 1 H), 3.71 (dd, half of ABX pattern, J=14.5, 6 Hz, 1 H), 3.55 (dd, half of ABX pattern, J=14.5, 6 Hz, 1 H), 1.43 (s, 3H), 1 .42 (s, 3H).

////////////Pfizer,  monobactam,  PF-?, 1380110-34-8, pfizer, pf, 1380110-45-1, WO 2012073138, Matthew Frank BrownSeungil HanManjinder LallMark. J. Mitton-FryMark Stephen PlummerHud Lawrence RisleyVeerabahu ShanmugasundaramJeremy T. Starr, preclinical

LY 3104607

$
0
0
imgChemSpider 2D Image | LY3104607 | C27H25N3O3
FDIWCHYTKOPHPS-QFIPXVFZSA-N.png
 LY3104607
(3S)-3-[4-[[2-(2,6-Dimethylphenyl)-[l,2,4]triazolo[l,5-a]pyridin-6-yl]methoxy]phenyl]hex-4-ynoic Acid
(3S)-3-[4-[[2-(2,6-dimethylphenyl)-[1,2,4]triazolo[1,5-a]pyridin-6-yl]methoxy]phenyl]hex-4-ynoic acid
CAS: 1795232-22-2
Chemical Formula: C27H25N3O3
Molecular Weight: 439.515
(3S)-3-(4-{[2-(2,6-Dimethylphenyl)[1,2,4]triazolo[1,5-a]pyridin-6-yl]methoxy}phenyl)-4-hexinsäure
Benzenepropanoic acid, 4-[[2-(2,6-dimethylphenyl)[1,2,4]triazolo[1,5-a]pyridin-6-yl]methoxy]-β-1-propyn-1-yl-, (βS)-

[+]Enlarge

Structure of LY3104607.
Credit: Tien Nguyen/C&EN

Presented by: Chafiq Hamdouchi, founder at Hamdouchi Pharmaceutical Consulting

Target: G-protein-coupled receptor 40 (GPR40), a receptor that modulates insulin secretion in cells

Disease: Type 2 diabetes

Reporter’s notes: Developed by Eli Lilly, LY3104607 joins the handful of GPR40 agonists recently offered by the company. The compound is not exactly a first disclosure as its structure was revealed in a January publication that describes its discovery and pharmacokinetic properties (J. Med. Chem. 2018, DOI: 10.1021/acs.jmedchem.7b01411). Hamdouchi, who worked on the molecule while at Eli Lilly, presented what the team learned about GPR40 and suggested that allosteric binding, binding which happens at a location other than the active site, may be a viable mode of action for GPR40 agonists.

Image result for LY3104607

Chafiq Hamdouchi

Image result

STR OF TITLE LY 3104607

str1 str2 str3

Paper

Discovery of LY3104607: A Potent and Selective G Protein-Coupled Receptor 40 (GPR40) Agonist with Optimized Pharmacokinetic Properties to Support Once Daily Oral Treatment in Patients with Type 2 Diabetes Mellitus

 Lilly Research Laboratories, A Division of Eli Lilly and Company, Lilly Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
 Jubilant Biosys Research Center, Bangalore, India
J. Med. Chem.201861 (3), pp 934–945
DOI: 10.1021/acs.jmedchem.7b01411
Publication Date (Web): December 13, 2017
Copyright © 2017 American Chemical Society
*E-mail: hamdouchi_chafiq@lilly.comchafiq.hamdouchi@gmail.com. Phone: 317-797-4751.

Abstract

Abstract Image

As a part of our program to identify potent GPR40 agonists capable of being dosed orally once daily in humans, we incorporated fused heterocycles into our recently disclosed spiropiperidine and tetrahydroquinoline acid derivatives 12, and 3 with the intention of lowering clearance and improving the maximum absorbable dose (Dabs). Hypothesis-driven structural modifications focused on moving away from the zwitterion-like structure. and mitigating the N-dealkylation and O-dealkylation issues led to triazolopyridine acid derivatives with unique pharmacology and superior pharmacokinetic properties. Compound 4 (LY3104607) demonstrated functional potency and glucose-dependent insulin secretion (GDIS) in primary islets from rats. Potent, efficacious, and durable dose-dependent reductions in glucose levels were seen during glucose tolerance test (GTT) studies. Low clearance, volume of distribution, and high oral bioavailability were observed in all species. The combination of enhanced pharmacology and pharmacokinetic properties supported further development of this compound as a potential glucose-lowering drug candidate.

(3S)-3-[4-[[2-(2,6-Dimethylphenyl)-[l,2,4]triazolo[l,5-a]pyridin-6-yl]methoxy]phenyl]hex-4-ynoic Acid (4)

Compound 4 (LY3104607)

title compound as white solid (35.76 kg, 91%). LCMS m/z [M + H]+: calcd, 439.5; found, 439.2.
1H NMR (399.80 MHz, DMSO, δ): 12.22 (s, 1H), 9.13 (dd, J = 0.8, 1.5 Hz, 1H), 7.88 (dd, J = 0.8, 9.2 Hz, 1H), 7.75 (dd, J = 1.7, 9.2 Hz, 1H), 7.29–7.24 (m, 3H), 7.14–7.12 (m, 2H), 7.01–6.99 (m, 2H), 5.18 (s, 2H), 3.96–3.91 (m, 1H), 2.58 (d, J = 7.7 Hz, 2H), 2.06 (s, 6H), 1.75 (d, J = 2.4 Hz, 3H).
PATENT
WO 2015088868
Applicants: ELI LILLY AND COMPANY [US/US]; Lilly Corporate Center Indianapolis, Indiana 46285 (US)
Inventors: HAMDOUCHI, Chafiq; (US)

A Novel Triazolo-Pyridine Compound

This invention relates to triazolo-pyridine compounds or pharmaceutically acceptable salts thereof, and for use of compounds in therapy. Triazolo-pyridine compounds of this invention are activators of GPR-40.

GPR-40, also known as Free Fatty Acid Receptor 1 (FFA1 or FFAR1), is reported as predominately expressed at high levels in rodent pancreatic beta cells, insulinoma cell lines, and human islets. The glucose modulation of insulin secretion is an important feature of activating GPR-40. Compounds that effectuate GPR-40 activation are associated with stimulation of insulin secretion in a patient with type II diabetes (T2D). Compounds that are GPR-40 activators are desired for use in treatment of GPR-40 mediated conditions.

WO2004/041266 discloses GPR-40 receptor function regulators comprising a compound having an aromatic ring and a group capable of releasing a cation.

The present invention rovides compounds of the Formula la below:

la

Example 1

(3S)-3-[4-[[2-(2,6-Dimethylphenyl)-[l,2,4]triazolo[l,5-a]pyridin-6- yl]methoxy]phenyl]hex-4-ynoic acid

To a solution of ethyl (3S)-3-[4-[[2-(2,6-dimethylphenyl)-[l,2,4]triazolo[l,5-a]pyridin-6-yl]methoxy]phenyl]hex-4-ynoate (0.22 g, 0.47 mmol) in EtOH (20 mL) is added 5 N NaOH (0.3 mL) and the reaction mixture is stirred at 80 °C in a microwave instrument for 30 minutes. The reaction mixture is evaporated to dryness, diluted with water, and acidified with 6 N HC1 solution to pH ~ 3. The precipitated solid is filtered, washed with n-pentane, and dried to give the title compound as a white solid (0.155 g, 75%). LCMS m/z 440 (M+H)+.

Alternate Preparation, Example 1

To a solution of ethyl (3S)-3-[4-[[2-(2,6-dimethylphenyl)-[l,2,4]triazolo[l,5-a]pyridin-6-yl]methoxy]phenyl]hex-4-ynoate (16 g, 34.22 mmol) in ethanol (160 mL) is added aqueous 5 N NaOH (2.73 g, 68.44 mmol in 16 mL water) drop wise at room temperature and the reaction mixture is stirred for 16 hours. The reaction mixture is evaporated to dryness, the residue is dissolved in water (300 mL), washed with diethyl ether (2 χ 200 mL), and the organic extract is discarded. The aqueous layer is cooled to 10 °C- 15 °C, acidified with saturated citric acid solution to pH~5, and extracted with DCM (2 x 300 mL). The combined organic extracts are washed with water (2 x 500 mL), brine solution (500 mL), dried over Na2S04, filtered, and evaporated to dryness to give the title compound as an off-white solid (14 g, 93%). LCMS m/z 440 (M+H)+.

The products from other batches, prepared as in Alternate Preparation of Example 1, are mixed with the product from Alternate Preparation Example 1 DCM (5 L) and warmed to 40 °C to get a clear solution. Then the solvent is evaporated to give an off-white solid. The possibility of trapped DCM is a concern, thus EtOAc (7.5 L) is charged and the resulting mixture is warmed to 65 °C to get a clear solution (-30 minutes). The solvent is evaporated and the resulting solid is dried under vacuum at 50 °C to obtain the desired product as an off-white solid. LCMS m/z 440 (M+H)+.

Form II Seed Crystal, Example 1

A saturated ethanol solution of (3S)-3-[4-[[2-(2,6-Dimethylphenyl)-[l,2,4]triazolo[l,5-a]pyridin-6-yl]methoxy]phenyl]hex-4-ynoic acid is filtered through 0.22 μιη nylon syringe filter into a clean vessel. Slow solvent evaporation at 25°C results in Form II seed crystals of Example 1.

Crystalline Form II (3S)-3-[4-[[2-(2,6-Dimethylphenyl)-[l,2,4]triazolo[l,5-a]pyridin- 6-yl] methoxy] phenyl] hex-4-ynoic acid

(3S)-3-[4-[[2-(2,6-Dimethylphenyl)-[l,2,4]triazolo[l,5-a]pyridin-6-yl]methoxy]phenyl]hex-4-ynoic acid can be prepared as a crystalline anhydrous Form II by dissolving (3S)-3-[4-[[2-(2,6-Dimethylphenyl)-[l,2,4]triazolo[l,5-a]pyridin-6-yl]methoxy]phenyl]hex-4-ynoic acid (580 mg, 132 mmol) in EtOH (1.2 mL) while stirring the mixture at 80 °C for 10 minutes. The solution is filtered and cooled to 70 °C at which point seeds of Form II are introduced. The mixture is then slowly cooled to ambient temperature while stirring overnight. The resulting solid plug is loosened with the addition of heptane (600 μΐ.) and the solids are recovered by vacuum filtration and dried under vacuum at 60 °C to give the crystalline title product (438 mg, 75.5%).

Patent ID Patent Title Submitted Date Granted Date
US9120793 Triazolo-pyridine compound
2014-12-04
2015-09-01
US2015166535 NOVELTRIAZOLO-PYRIDINE COMPOUND
2014-12-04
2015-06-18

/////////LY3104607, LY-3104607, LY 3104607, PRECLINICAL

CC#C[C@H](C1=CC=C(OCC2=CN3C(C=C2)=NC(C4=C(C)C=CC=C4C)=N3)C=C1)CC(O)=O

BMS-986118, for treatment for type 2 diabetes( GPR40 agonists with a dual mechanism of action, promoting both glucose-dependent insulin and incretin secretion)

$
0
0
str1img
str2
str1
str1
GKIUHMMLGAMMOO-OITFXXTJSA-N.png
BMS-986118
BMS compd for treatment for type 2 diabetes( GPR40 agonists with a dual mechanism of action, promoting both glucose-dependent insulin and incretin secretion)
Cas 1610562-74-7
1H-Pyrazole-5-acetic acid, 1-[4-[[(3R,4R)-1-(5-chloro-2-methoxy-4-pyridinyl)-3-methyl-4-piperidinyl]oxy]phenyl]-4,5-dihydro-4-methyl-3-(trifluoromethyl)-, (4S,5S)-
Molecular Weight, 540.96, C25 H28 Cl F3 N4 O4

2-((4S,5S)-1-(4-(((3R,4R)-1-(5-Chloro-2-methoxypyridin-4-yl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-1H-pyrazol-5-yl)acetic acid

(-)-[(4S,5S)-1-(4-[[(3R,4R)-1-(5-Chloro-2-methoxypyridin-4-yl)-3-methylpiperidin-4-yl]oxy]phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-1H-pyrazol-5-yl]acetic acid

  • (4S,5S)-1-[4-[[(3R,4R)-1-(5-Chloro-2-methoxy-4-pyridinyl)-3-methyl-4-piperidinyl]oxy]phenyl]-4,5-dihydro-4-methyl-3-(trifluoromethyl)-1H-pyrazole-5-acetic acid
  • 2-[(4S,5S)-1-[4-[[1-(5-Chloro-2-methoxypyridin-4-yl)-3-methylpiperidin-4-yl]oxy]phenyl]-4-methyl-3-(trifluoromethyl)-4,5-dihydro-1H-pyrazol-5-yl]acetic acid isomer 2

BMS-986118 is a GPR40 full agonist. GPR40 is a G-protein-coupled receptor expressed primarily in pancreatic islets and intestinal L-cells that has been a target of significant recent therapeutic interest for type II diabetes. Activation of GPR40 by partial agonists elicits insulin secretion only in the presence of elevated blood glucose levels, minimizing the risk of hypoglycemia

Image result for bms

NOTE CAS OF , 1H-Pyrazole-5-acetic acid, 1-[4-[[(3S,4S)-1-(5-chloro-2-methoxy-4-pyridinyl)-3-methyl-4-piperidinyl]oxy]phenyl]-4,5-dihydro-4-methyl-3-(trifluoromethyl)-, (4S,5S)- IS 1610562-73-6

str1

Image result for BMS-986118,

SYNTHESIS

WO 2014078610

PAPER

https://pubs.acs.org/doi/10.1021/acs.jmedchem.7b00982

Discovery of Potent and Orally Bioavailable Dihydropyrazole GPR40 Agonists

Abstract

Abstract Image

G protein-coupled receptor 40 (GPR40) has become an attractive target for the treatment of diabetes since it was shown clinically to promote glucose-stimulated insulin secretion. Herein, we report our efforts to develop highly selective and potent GPR40 agonists with a dual mechanism of action, promoting both glucose-dependent insulin and incretin secretion. Employing strategies to increase polarity and the ratio of sp3/sp2 character of the chemotype, we identified BMS-986118 (compound 4), which showed potent and selective GPR40 agonist activity in vitroIn vivo, compound 4 demonstrated insulinotropic efficacy and GLP-1 secretory effects resulting in improved glucose control in acute animal models.

Compound 4

2-((4S,5S)-1-(4-(((3R,4R)-1-(5-Chloro-2-methoxypyridin-4-yl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-1H-pyrazol-5-yl)acetic acid (4)

To a stirred solution of methyl 2-((4S,5S)-1-(4-(((3R,4R)-1-(5-chloro-2-methoxypyridin-4-yl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-1H-pyrazol-5-yl)acetate (5.5 g, 9.9 mmol) in THF (90 mL) and water (9 mL) at room temperature was added 2 N LiOH solution (12 mL, 24 mmol). The reaction mixture was stirred at room temperature for 16 h, and 1 N HCl (25 mL, 25 mmol) was added at 0 °C to pH = 4–5. The solvent was evaporated, and the residue was extracted three times with EtOAc. The organic extracts were dried over Na2SO4; the solution was filtered and concentrated. The residue was recrystallized from isopropanol to give 4(neutral form) as white solid (4.3 g, 7.7 mmol, 78% yield).
1H NMR (500 MHz, DMSO-d6) δ ppm 12.52 (br s, 1H), 8.01 (s, 1H), 7.05 (d, J = 9.1 Hz, 2H), 6.96 (d, J = 9.1 Hz, 2H), 6.40 (s, 1H), 4.49–4.33 (m, 1H), 4.02 (td, J = 8.8, 4.1 Hz, 1H), 3.80 (s, 3H), 3.56–3.39 (m, 2H), 3.37–3.29 (m, 1H), 2.94–2.85 (m, 1H), 2.72–2.66 (m, 1H), 2.64 (dd, J = 16.1, 2.9 Hz, 1H), 2.49–2.41 (m, 1H), 2.22–2.05 (m, 1H), 2.01–1.86 (m, 1H), 1.68–1.50 (m, 1H), 1.25 (d, J = 7.2 Hz, 3H), 1.03 (d, J = 6.9 Hz, 3H).
 
13C NMR (126 MHz, DMSO-d6) δ 171.5, 163.7, 157.1, 152.5, 146.3, 139.7 (q, J = 34.7 Hz), 136.2, 121.7 (q, J = 269.3 Hz), 117.3, 117.2, 116.0, 100.4, 78.9, 65.6, 54.2, 53.4, 47.8, 44.2, 36.0, 34.9, 29.5, 17.4, 15.3. 19F NMR (471 MHz, DMSO-d6) δ −61.94 (s, 3F).
 
Optical rotation: [α]D(20)−11.44 (c 2.01, MeOH).
 
HRMS (ESI/HESI) m/z: [M + H]+ Calcd for C25H29ClF3N4O4 541.1824; Found 541.1813. HPLC (Orthogonal method, 30% Solvent B start): RT = 11.9 min, HI: 97%. m/zobs 541.0 [M + H]+.
 
Melting point = 185.5 °C.
PAPER

Palladium-Catalyzed C–O Coupling of a Sterically Hindered Secondary Alcohol with an Aryl Bromide and Significant Purity Upgrade in the API Step

Chemical and Synthetic DevelopmentBristol-Myers Squibb CompanyOne Squibb Drive, New Brunswick, New Jersey08903, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.8b00022

Abstract

Abstract Image

The final two steps used to prepare greater than 1 kg of a compound evaluated as a treatment for type 2 diabetes are reported. The application of a palladium-catalyzed C–O coupling presented significant challenges due to the nature of the reactants, impurities produced, and noncrystalline coupling intermediate. Process development was able to address these limitations and enable production of kilogram quantities of the active pharmaceutical ingredient (API) in greater efficiency than a Mitsunobu reaction for formation of the key bond. The development of a sequence that telescopes the coupling with the subsequent ester hydrolysis to yield the API and the workup and final product crystallization necessary to produce high-quality drug substance without the need of column chromatography are discussed.

Bruce Ellsworth

Bruce Ellsworth, Director, Head of Fibrosis Discovery Chemistry at Bristol-Myers Squibb

Rick EwingRick Ewing, Head, External Partnerships, Discovery Chemistry and Molecular Technologies at Bristol-Myers Squibb
str1 str2
PATENT
WO 2014078610
Original Assignee Bristol-Myers Squibb Company
Patent
Patent ID Patent Title Submitted Date Granted Date
US9133163 DIHYDROPYRAZOLE GPR40 MODULATORS
2013-11-15
2014-05-22
US9604964 Dihydropyrazole GPR40 modulators
2013-11-15
2017-03-28
REF
1: Li Z, Qiu Q, Geng X, Yang J, Huang W, Qian H. Free fatty acid receptor
agonists for the treatment of type 2 diabetes: drugs in preclinical to phase II
clinical development. Expert Opin Investig Drugs. 2016 Aug;25(8):871-90. doi:
10.1080/13543784.2016.1189530. PubMed PMID: 27171154.
2
Discovery of BMS-986118, a dual MOA GPR40 agonist that produces glucose-dependent insulin and GLP-1 secretion
248th Am Chem Soc (ACS) Natl Meet (August 10-14, San Francisco) 2014, Abst MEDI 31
MEDI John Macor Sunday, August 10, 2014
Oral Session
General Oral Session – PM Session
Organizers: John Macor
Presiders: John Macor
Duration: 1:30 pm – 5:15 pm
1:55 pm 31 Discovery of BMS-986118, a dual MOA GPR40 agonist that produces glucose-dependent insulin and GLP-1 secretion
Bruce A Ellsworth, Jun Shi, Elizabeth A Jurica, Laura L Nielsen, Ximao Wu, Andres H Hernandez, Zhenghua Wang, Zhengxiang Gu, Kristin N Williams, Bin Chen, Emily C Cherney, Xiang-Yang Ye, Ying Wang, Min Zhou, Gary Cao, Chunshan Xie, Jason J Wilkes, Heng Liu, Lori K Kunselman, Arun Kumar Gupta, Ramya Jayarama, Thangeswaran Ramar, J. Prasada Rao, Bradley A Zinker, Qin Sun, Elizabeth A Dierks, Kimberly A Foster, Tao Wang, Mary Ellen Cvijic, Jean M Whaley, Jeffrey A Robl, William R Ewing.

///////////BMS-986118, Preclinical, BMS, Bruce A. Ellsworth,  Jun Shi,  William R. Ewing,  Elizabeth A. Jurica,  Andres S. Hernandez,  Ximao Wu, DIABETES,

COc1cc(c(Cl)cn1)N4CCC(Oc2ccc(cc2)N3N=C([C@@H](C)C3CC(=O)O)C(F)(F)F)[C@H](C)C4

COc1cc(c(Cl)cn1)N4CC[C@@H](Oc2ccc(cc2)N3N=C([C@H](C)[C@@H]3CC(=O)O)C(F)(F)F)[C@@H](C)C4

COc1cc(c(Cl)cn1)N4CC[C@@H](Oc2ccc(cc2)N3N=C([C@@H](C)[C@@H]3CC(=O)O)C(F)(F)F)[C@H](C)C4

BMS-986169

$
0
0

imgUNVYDSCXINFREZ-BHDDXSALSA-N.pngBDBM198728.png

BMS-986169

CAS 1801151-08-5 Related CAS : 1801151-09-6   1801151-08-5
Chemical Formula: C23H27FN2O2
Molecular Weight: 382.4794
Elemental Analysis: C, 72.23; H, 7.12; F, 4.97; N, 7.32; O, 8.37

(R)-3-((3S,4S)-3-fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one

(3R)-3-[(3S,4S)-3-fluoro-4-(4-hydroxyphenyl)piperidin-1-yl]-1-[(4-methylphenyl)methyl]pyrrolidin-2-one

Preclinical

BMS-986169 is a Novel, Intravenous, Glutamate N-Methyl-d-Aspartate 2B Receptor Negative Allosteric Modulator with Potential in Major Depressive Disorder. BMS-986169 showed high binding affinity for the GluN2B subunit allosteric modulatory site (Ki = 4.03-6.3 nM) and selectively inhibited GluN2B receptor function in Xenopus oocytes expressing human N-methyl-d-aspartate receptor subtypes (IC50 = 24.1 nM). BMS-986169 weakly inhibited human ether-a-go-go-related gene channel activity (IC50 = 28.4 μM) and had negligible activity in an assay panel containing 40 additional pharmacological targets.

Chemical structures of BMS-986169 and the phosphate prodrug BMS-986163.

Chemical structures of BMS-986169 and the phosphate prodrug BMS-986163. 
Image result for BMS-986169

 

PAPER

Evolution of a Scale-Up Synthesis to a Potent GluN2B Inhibitor and Its Prodrug

 Discovery Chemistry and Molecular TechnologiesBristol-Myers Squibb Research and Development, Princeton, New Jersey 08540, United States
 Drug Product Science & Technology, Materials Science & EngineeringBristol-Myers Squibb Research and Development, Princeton, New Jersey 08540, United States
§ Department of Discovery SynthesisBiocon Bristol-Myers Squibb Research Center (BBRC), Bangalore 560099, India
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.8b00120
Abstract Image

This paper describes the efficient scale-up synthesis of the potent negative allosteric glutamate N2B (GluN2B) inhibitor 1 (BMS-986169), which relies upon a stereospecific SN2 alkylation strategy and a robust process for the preparation of its phosphate prodrug 28 (BMS-986163) from parent 1 using POCl3. A deoxyfluorination reaction employing bis(2-methoxyethyl)aminosulfur trifluoride (Deoxo-Fluor) is also used to stereospecifically introduce a fluorine substituent. The optimized routes have been demonstrated to provide APIs suitable for toxicological studies in vivo.

https://pubs.acs.org/doi/suppl/10.1021/acs.oprd.8b00120/suppl_file/op8b00120_si_001.pdf

PAPER

https://pubs.acs.org/doi/abs/10.1021/acsmedchemlett.8b00080

BMS-986163, a Negative Allosteric Modulator of GluN2B with Potential Utility in Major Depressive Disorder

 Bristol-Myers Squibb Research and Development5 Research Parkway, Wallingford, Connecticut 06492, United States
 Biocon Bristol-Myers Squibb Research Center, Bangalore, India
§ Bristol-Myers Squibb Research and Development3551 Lawrenceville Road, Princeton, New Jersey 08648, United States
ACS Med. Chem. Lett.20189 (5), pp 472–477
DOI: 10.1021/acsmedchemlett.8b00080
*Phone 203-677-6701. E-mail: lawrence.marcin@bms.com.

 

Abstract Image

There is a significant unmet medical need for more efficacious and rapidly acting antidepressants. Toward this end, negative allosteric modulators of the N-methyl-d-aspartate receptor subtype GluN2B have demonstrated encouraging therapeutic potential. We report herein the discovery and preclinical profile of a water-soluble intravenous prodrug BMS-986163 (6) and its active parent molecule BMS-986169 (5), which demonstrated high binding affinity for the GluN2B allosteric site (Ki = 4.0 nM) and selective inhibition of GluN2B receptor function (IC50 = 24 nM) in cells. The conversion of prodrug 6 to parent 5 was rapid in vitro and in vivo across preclinical species. After intravenous administration, compounds 5 and 6 have exhibited robust levels of ex vivo GluN2B target engagement in rodents and antidepressant-like activity in mice. No significant off-target activity was observed for 56, or the major circulating metabolites met-1 and met-2. The prodrug BMS-986163 (6) has demonstrated an acceptable safety and toxicology profile and was selected as a preclinical candidate for further evaluation in major depressive disorder.

Image result for BMS-986169

Image result for BMS-986169

 

 

(S)-3-((3S,4S)-3-fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-
methylbenzyl)pyrrolidin-2-one (compound 23) and (R)-3-((3S,4S)-3-fluoro-4-(4-
hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one (BMS-986169, compound
5)……https://pubs.acs.org/doi/suppl/10.1021/acsmedchemlett.8b00080/suppl_file/ml8b00080_si_001.pdf

Analytical data for BMS-986169 (compound 5): LCMS (C23H27FN2O2, MW 382.2, ESAPI),
observed 383.2 m/z (M+H)+; []D20 = +6.09 (c = 1.15, MeOH); Anal. Calcd for
C23H27FN2O2 (382.21): C, 72.22; H, 7.12; N, 7.32. Found: C, 72.26; H, 7.05; N, 7.31; HRMS
(ESI) Calcd for C23H27N2O2, 383.2118. Found, 383.2129;

13C NMR (126 MHz, chloroformd)
172.4, 155.0, 137.5, 133.0, 132.8, 129.4, 128.6, 128.2, 115.6, 91.6 (d, J=173.5 Hz),
65.0, 54.5 (d, J=25.4 Hz), 48.3, 47.7 (d, J=17.3 Hz), 46.7, 43.6, 31.5, 21.1, 19.2;(500 MHz, chloroform-d) 7.23 – 7.11 (m, 5H), 6.92 (d, J=8.5 Hz, 2H), 6.18 (br. s., 1H),
4.79 – 4.55 (m, 1H), 4.57 – 4.33 (m, 2H), 3.72 (t, J=8.7 Hz, 1H), 3.46 – 3.30 (m, 1H), 3.30 –
3.09 (m, 2H), 2.82 (d, J=8.5 Hz, 1H), 2.73 – 2.56 (m, 2H), 2.49 (d, J=2.5 Hz, 1H), 2.36 (s,
3H), 2.21 – 1.98 (m, 2H), 1.87 (br. s., 2H). The corresponding 1H NMR spectrum for
compound 5 is shown below

1H NMR

 

PATENT

https://patents.google.com/patent/US9221796B2/und

InventorDalton KingLorin A. Thompson, IIIJianliang ShiSrinivasan ThangathirupathyJayakumar Sankara WarrierImadul IslamJohn E. Macor

Current Assignee Bristol-Myers Squibb Co

https://patents.google.com/patent/WO2015105772A1/und

N-Methyl-D-aspartate (NMDA) receptors are ion channels which are gated by the binding of glutamate, an excitatory neurotransmitter in the central nervous system. They are thought to play a key role in the development of a number of neurological diseases, including depression, neuropathic pain, Alzheimer’s disease, and Parkinson’s disease. Functional NMDA receptors are tetrameric structures primarily composed of two NRl and two NR2 subunits. The NR2 subunit is further subdivided into four individual subtypes: NR2A, NR2B, NR2C, and NR2D, which are differentially distributed throughout the brain. Antagonists or allosteric modulators of NMDA receptors, in particular NR2B subunit-containing channels, have been investigated as therapeutic agents for the treatment of major depressive disorder (G. Sanacora, 2008, Nature Rev. Drug Disc. 7: 426-437).

The NR2B receptor contains additional ligand binding sites in additon to that for glutamate. Non-selective NMDA antagonists such as Ketamine are pore blockers, interfering with the transport of Ca++ through the channel. Ketamine has demonstrated rapid and enduring antidepressant properties in human clinical trials as an i.v. drug. Additionally, efficacy was maintained with repeated, intermittent infusions of Ketamine (Zarate et al., 2006, Arch. Gen. Psychiatry 63: 856-864). This class of drugs, though, has limited therapeutic value because of its CNS side effects, including dissociative effects.

An allosteric, non-competitive binding site has also been identified in the N-terminal domain of NR2B. Agents which bind selectively at this site, such as

Traxoprodil, exhibited a sustained antidepressant response and improved side effect profile in human clinical trials as an i.v. drug (Preskorn et al., 2008, J. Clin.

PsychopharmacoL, 28: 631-637, and F. S. Menniti, et al, 1998, CNS Drug Reviews, 4, 4, 307-322). However, development of drugs from this class has been hindered by low bioavailability, poor pharmacokinetics, and lack of selectivity against other pharmacological targets including the hERG ion channel. Blockade of the hERG ion channel can lead to cardiac arrythmias, including the potentially fatal Torsades de pointe, thus selectivity against this channel is critical. Thus, in the treatment of major depressive disorder, there remains an unmet clinical need for the development of effective NR2B-selective negative allosteric modulators which have a favorable tolerability profile.

NR2B receptor antagonists have been disclosed in PCT publication WO 2009/006437.

The invention provides technical advantages, for example, the compounds are novel and are ligands for the NR2B receptor and may be useful for the treatment of various disorders of the central nervous system. Additionally, the compounds provide advantages for pharmaceutical uses, for example, with regard to one or more of their mechanism of action, binding, inhibition efficacy, target selectivity, solubility, safety profiles, or bioavailability.

Synthetic Scheme 1

The l-phenyl/benzyl-3-bromo-pyrrolidinones/piperidinones V may be reacted with (4-oxy-phenyl)cyclic amines VI in the presence of base to produce protected products VII, which may be subjected to cleavage conditions appropriate for the protecting group (PGi) to generate final products I, which may be separated into individual enantiomers/diastereomers I*, as shown in synthetic scheme 2.

Synthetic Scheme 2

I I*

Compounds la may be prepared by condensing l-phenyl/benzyl-3-bromo-pyrroli-dinones/piperidinones V with substituted 4(4-oxyphenyl)piperidines Vllla-c to generate protected intermediates IX, which may be subjected to cleavage conditions appropriate for the protecting group (PGi) to generate final products la, which may be separated into individual enantiomers/diastereomers la*, as shown in synthetic scheme 3.

Synthetic Scheme 3

The 4(4-oxyphenyl)piperidines Vllla-c may be synthesized in turn by a sequence starting with a protected tetrahydropiperidine X, which can be hydroxylated via hydroboration/oxidation to give the protected hydroxypiperidine XI, which may be either directly transformed into the protected fluoropiperidine XII by treatment with DAST or oxidized into the protected 3-oxopiperidine XIII, which may be further transformed into protected 3,3-difluoropiperidines XIV via treatment with DAST. XI, XII, and XIV may be transformed into Villa, Vlllb, and VIIIc, respectively, by employing cleaving conditions appropriate for the protecting group (PG2), as shown in synthetic scheme 3 a.

S nthetic scheme 3 a

Chiral

Cleavage Individual enantiomers/

G2P-N diastereomers

separation

conditions

OH 
Villa*

Villa

XI

Chiral

Cleavage Individual enantiomers/

HN

G2P-N diastereomers

%_\J> PQ separation

PG1 conditions

R F

F Vlllb*

Vlllb

XII

Chiral

Individual enantiomers/

G2P- diastereomers separation

Vlllc*

For tetrahydropyridines X which are not commercially available may be synthesized by coupling protected bromophenols XV with protected unsaturated

piperidineboronic acids XVI, as shown in synthetic scheme 4a.

Synthetic scheme 4a:

For tetrahydropyridines X which are not commercially available may be synthesized by adding the anion generated from protected bromophenols XV to a protected 4-piperidinone XVII to yield 4-phenyl-4-piperidinol XVIII, which may be dehydrated under acid conditions to yield the desired X, as shown in synthetic scheme 4b.

Synthetic scheme 4b:

l-Phenyl/benzyl-3-bromo-pyrroli-dinones/piperidinones V may be condensed with isolated individual enantiomers VIIIa-c*, which results in diastereomers 1- phenyl/benzyl-3-bromo-pyrroli-dinones/piperidinones IX*, which may be deprotected and separated to give final products la*, as shown in scheme 5.

Alternatively, the backbone scaffold may be synthesized by condensing 1- phenyl/benzyl-3-bromo-pyrroli-dinones/piperidinones V with hydroxypiperidines Villa to yield the protected 3-fluoropiperidines IXa, which may themselves be converted to the protected 3-fluoropiperidines IXb or oxidized to the ketones XIX, which may be converted to the 3,3-difluoropiperidines Ixc, as shown in scheme 6. The final compounds can then be isolated after the deprotection of IXa-c.

Scheme 6

Example 46, P-1 Example 46, P-2

(S)-3-((3S,4S)-3-Fluoro-4-(4-hydroxyphenyl)piperidin-l-yl)-l-(4-methylbenzyl)pyrrolidin-2-one and (R)-3-((3S,4S)-3-fluoro-4-(4-hydroxyphenyl)piperidin-l-yl)-l-(4-methylbenzyl)pyrrolidin-2-one.

Example 46, P-3 Example 46, P-4

Step A. (±)-rel-(3S,4S)- 1 -benzyl-4-(4-methoxyphenyl)piperidin-3-ol.

To a suspension of sodium tetrahydroborate (2.7 g, 72 mmol) in THF (200 mL) at 0 °C under a nitrogen atmosphere was added dropwise boron trifluoride etherate (8.8 mL, 70 mmol) and the resulting mixture was stirred for 30 minutes. Then 1-benzyl- 4-(4-methoxyphenyl)-l,2,3,6-tetrahydropyridine (10 g, 36 mmol, from S. Halazy et al WO 97/28140 (8/7/97)) dissolved in 100 mL of tetrahydrofuran was added. The mixture was allowed to warm to rt and stirred for 2 h. The reaction was then quenched by the dropwise addition of 100 mL of water. Next were added

sequentially 100 mL of ethanol, 100 mL of a 10% aqueous sodium hydroxide solution, and 30%> hydrogen peroxide (18 mL, 180 mmol) and the mixture was stirred at reflux temperature overnight. The reaction mixture was then allowed to cool, diluted with saturated aqueous ammonium chloride (200 mL), and extracted with ethyl acetate (500 mL). The organic layer was dried over Na2S04, filtered, and evaporated under reduced pressure to give (±)-rel-(3S,4S)- 1 -benzyl-4-(4-methoxyphenyl)piperidin-3-ol (8.5 g, 24.6 mmol, 69%> yield) which was used without further purification. LCMS (Method K) RT 1.99 min; m/z 298.0 (M+H+).

Step B. (±)-re -(3S,4S)-4-(4-methoxyphenyl)piperidin-3-ol.

To a solution of (±)-re/-(35′,45)-l-benzyl-4-(4-methoxyphenyl)piperidin-3-ol (9 g, 30 mmol) in methanol (150 mL) was added 10 % Pd/C (4.8 g) and the reaction mixture was stirred overnight under a hydrogen atmosphere. The catalyst was then removed by filtration through Celite and the solvent was evaporated under reduced pressure to give (±)-re/-(3S,4S)-4-(4-methoxyphenyl)piperidin-3-ol (5.1 g, 24.6 mmol, 81% yield) which was used without further purification. 1H NMR (400 MHz, DMSO-de) δ ppm 7.10 – 7.15 (m, 2 H) 6.80 – 6.86 (m, 2 H) 4.30 (d, J=5.27 Hz, 1 H) 3.37 – 3.43 (m, 1 H) 3.04 (dd, J=11.58, 4.36 Hz, 1 H) 2.86 (d, J=12.17 Hz, 1 H) 2.43 (td, J=12.09, 2.67 Hz, 1 H) 2.22 – 2.35 (m, 2 H) 1.57 – 1.63 (m, 1 H) 1.43 – 1.54 (m, 1 H).

To a solution of (±)-re/-(3S,4S)-4-(4-methoxyphenyl)piperidin-3-ol (4.5 g, 21.7 mmol) in DCM (150 mL) at -10°C under nitrogen was added a 1 M solution of boron tribromide in DCM (109 mL, 109 mmol). The reaction mixture was allowed to warm to rt, stired for 2 h, and then rechilled to 0 °C and quenched by the addition of a saturated aqueous sodium bicarbonate solution (300 mL). The aqueous layer was washed with 250 mL of DCM and then to it was added 200 mL 10% aqueous NaOH, followed by 9.5 g (43.5 mmol) of di-t-butyl dicarbonate and the resulting mixture was stirred for an additional 2 h. The mixture was then extracted with 200 mL ethyl acetate and the organic layer was separated, dried over Na2S04,filtered, and evaporated under reduced pressure to (±)-re/-(35′,45)-tert-butyl 4-(4-(tert-butoxycarbonyloxy)phenyl)-3-hydroxypiperidine-l-carboxylate (6.5 g, 12 mmol, 56 % yield) which was used without further purification. LCMS (Method K) RT 2.33 min, m/z 282 (M+H+ -2 t-butyl), 370; 1H NMR (400 MHz, DMSO-d6) δ ppm 7.27 (d, J=8.66 Hz, 2 H) 7.08 (d, J=8.66 Hz, 2 H) 4.85 (d, J=5.65 Hz, 1 H) 4.13 (d, J=8.41 Hz, 1 H) 3.97 (d, J=10.48 Hz, 1 H) 3.45 (tt, J=10.27, 5.19 Hz, 1 H) 1.67 (d, J=3.39 Hz, 1 H) 1.50 – 1.59 (m, 1 H) 1.49 (s, 11 H).

Step D. (±)-re/-(35′,45)-tert-butyl 3-hydroxy-4-(4-hydroxyphenyl)piperidine-l-carboxylate.

To a solution of (±)-re/-(35′,45)-tert-butyl 4-(4-(tert-butoxycarbonyloxy)phenyl)-3-hydroxypiperidine-l-carboxylate (6.5 g, 16.5 mmol) in 100 mL of methanol was added 11.42 g of potassium carbonate (83 mmol) and the reaction mixture was stirred at rt for 5 h. The organic solvent was removed under reduced pressure and the residue was partitioned between IN HC1 (300 mL) and ethyl acetate (300 mL). The layers were separated and the organic layer was dried over Na2S04 and evaporated under reduced pressure to give (±)-re/-(35′,45)-tert-butyl 3-hydroxy-4-(4-hydroxyphenyl)piperidine-l-carboxylate (5 g, 15 mmol, 92 % yield) which was used without further purification. LCMS (method F) RT 1.85 min, m/z 238 (M+H+ – 1-butyl), 279 (M+H+ – t-butyl+CH3CN), 1H NMR (400 MHz, DMSO-d6) δ ppm 7.01 (d, J=8.53 Hz, 2 H) 6.66 (d, J=8.53 Hz, 2 H) 4.70 (d, J=5.02 Hz, 1 H) 4.09 (br. s., 1 H) 3.94 (d, J=11.55 Hz, 1 H) 3.35 – 3.41 (m, 1 H) 2.66 – 2.77 (m, 1 H) 2.29 – 2.39 (m, 1 H) 1.63 (dd, J=13.30, 3.26 Hz, 1 H) 1.44 – 1.52 (m, 1 H) 1.42 (s, 9 H).

Step E. (3S,4S)-tert-Butyl 3 -hydroxy-4-(4-hydroxyphenyl)piperidine-l -carboxylate and (3R, -tert-butyl 3-hydroxy-4-(4-hydroxyphenyl)piperidine-l-carboxylate.

E-1 E-2

(±)-rel-(3S,4S)-tert-Butyl 3-hydroxy-4-(4-hydroxyphenyl)piperidine- 1 -carboxylate (5 g, 17 mmol, from step D) was subjected to chiral SFC separation (method C-5) to yield enantiomers E-1 (1.9 g, 6.48 mmol, 38.0 % yield) and E-2 (2.4 g, 8.18 mmol, 48.0 % yield). Data for E-1 : chiral HPLC (method A5 ) retention time 3.42 min. Data for E-2: chiral HPLC (method A5) retention time 4.2 min.

Step F. (3R,4R)-tert-Butyl 4-(4-(benzyloxy)phenyl)-3-hydroxypiperidine-l-carboxylate.

A mixture of (3R,4R)-tert-butyl 3-hydroxy-4-(4-hydroxyphenyl)piperidine-l-carboxylate (620 mg, 2.1 mmol, E-2 from step E), potassium carbonate (584 mg, 4.2 mmol), and benzyl bromide (0.25 mL, 2.1 mmol) in DMF (5 mL) was stirred at rt for 16 h. The solvent was removed by evaporation and the residue was treated with 50 mL of water. The aqueous mixture was then extracted 4 times with 50 mL of chloroform. The combined organic phases were dried over anhydous Na2S04, filtered, and evaporated to yield 750 mg of (3R,4R)-tert-butyl 4-(4-(benzyloxy)phenyl)-3-hydroxypiperidine-l -carboxylate which was used without further purification. LCMS (method F) RT 2.28 min, m/z = 310 (M+H+ – t-butyl -water), 328 (M+H+ -t-butyl).

Step G. (3i?,4i?)-4-(4-(Benzyloxy)phenyl)piperidin-3-ol hydrochloride.

A mixture of (3R,4R)-tert-butyl 4-(4-(benzyloxy)phenyl)-3-hydroxypiperidine-1-carboxylate (750 mg, 2 mmol), dioxane (4 mL) and 4.9 mL of 4 M HCI in dioxane was stirred at rt for 2h. The reaction was then evaporated to dryness to yield 550 mg of (3i?,4i?)-4-(4-(Benzyloxy)phenyl)piperidin-3-ol hydrochloride which was used without further purification. LCMS (method J) RT 0.70 min, m/z 284 (M+H+).

Step H. 3-((3i?,4i?)-4-(4-(Benzyloxy)phenyl)-3-hydroxypiperidin-l -yl)- 1 -(4-methylbenzyl)pyrrolidin-2-one .

A mixture of 3-bromo-l-(4-methylbenzyl)pyrrolidin-2-one (Intermediate 2, 220 mg, 0.82 mmol), (3i?,4i?)-4-(4-(benzyloxy)phenyl)piperidin-3-ol hydrochloride (262 mg, 0.82 mmol, from step G) and triethylamine (11 mL, 8.2 mmol) was stirred at 60 °C for lh, 80 °C for 1 h, 100 °C for 1 h and 120 °C for 1 h. The reaction mixture was then allowed to cool, diluted with 40 mL of water and extracted four times with 50 mL of chloroform. The combined organic layers were washed with 60 mL brine, dried over anhydrous sodium sulfate, filtered, and evaporated to yield 382 mg of 3-((3 ?,4i?)-4-(4-(benzyloxy)phenyl)-3-hydroxypiperidin- 1 -yl)- 1 -(4-methylbenzyl)pyrrolidin-2-one which was used without further purification. LCMS (method J) (main component of a mixture) RT 2.23 min, m/z 471 (M+H+).

Step I. 3-((3R, 4R)-4-(4-(Benzyloxy)phenyl)-3-fluoropiperidin- 1 -yl)- 1 -(4-methylbenzyl)pyrrolidin-2-one .

A solution of 3-(-4-(4-(benzyloxy)phenyl)-3-hydroxypiperidin-l-yl)-l-(4-methylbenzyl)pyrrolidin-2-one (382 mg, 0.81 mmol) in DCM (5 mL) cooled to 0 °C was treated dropwise with DAST (0.32 mL, 2.4 mmol) over 3 min. The reaction mixture was then allowed to warm to rt and was stirred for 2 h. The reaction was then quenched with 50 mL of 10% aqueous sodium bicarbonate solution and extracted 4 times with 40 mL of DCM. The combined organic layers were washed with 50 mL of brine, dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum to yield 382 mg of 3-((3i?,4i?)-4-(4-(benzyloxy)phenyl)-3-fluoropiperidin-l-yl)-l-(4-methylbenzyl)pyrrolidin-2-one as a mixture of two diastereomers and rearrangement products which was used without further purification. LCMS (method J) (main component of a mixture) RT 0.9 min, m/z 473 (M+H+).

Step J. 3-((3i?,4i?)-3-Fluoro-4-(4-hydroxyphenyl)piperidin- 1 -yl)-l -(4-methylbenzyl)pyrrolidin-2-one .

A mixture of 3-((Ji?,4i?)-(4-(4-(benzyloxy)phenyl)-3-fluoropiperidin-l-yl)-l-(4-methylbenzyl)pyrrolidin-2-one (382 mg, 0.81 mmol) and methanol (4 mL) was flushed with nitrogen, followed by the addition of 172 mg of 10% Pd/C. Then the mixture was stirred at rt overnight under 25-99 psi hydrogen pressure. The reaction was then transferred to a 100 mL autoclave and stirred at 7 kg/cm2 hydrogen pressure for 4 days. The catalyst was removed by filtration through Celite and the solvent was evaporated off. The crude product was subjected to HPLC purification (method B) to yield 77.3 mg 3-((Ji?,4i?)-3-fluoro-4-(4-hydroxyphenyl)-piperidin-l-yl)-l-(4-methylbenzyl)pyrrolidin-2-one (diastereomeric pair) LCMS (method Q) RT 1.15 min, m/z 383.0 (M+H+).

Step K. (5)-3-((3i?,4i?)-3-Fluoro-4-(4-hydroxyphenyl)piperidin- 1 -yl>

methylbenzyl)pyrrolidin-2-one and (i?)-3-((3i?,4i?)-3-fluoro-4-(4-hydroxyphenyl)piperidin- 1 -yl)- 1 -(4-methylbenzyl)pyrrolidin-2-one.

The diastereomeric mixture from step J was separated by SFC method C-7 to yield homochiral Examples 46 P-l (29.3 mg) and P-2 (32.8 mg). Data for P-l (S)-3-((3R, 4R)-3 -fluoro-4-(4-hydroxyphenyl)piperidin- 1 -yl)- 1 -(4-methylbenzyl)pyrrolidin-2-one: LCMS (method F) RT 2.10 min, m/z 383.2 (M+H+), 405.2 (M+Na+); HPLC (method B) RT 8.24 min (98.8% AP); HPLC (method C) RT 6.52 min (99.1% AP); Chiral HPLC (method C-6) RT 4.1 min; 1H NMR (400 MHz, methanol-d4) δ ppm 1.76 – 1.86 (m, 2 H) 2.07 (d, J=8.53 Hz, 1 H) 2.13 – 2.21 (m, 1 H) 2.34 (s, 3 H) 2.43 (s, 0 H) 2.55 – 2.60 (m, 1 H) 2.65 – 2.70 (m, 1 H) 2.75 (br. s., 1 H) 3.20 – 3.30 (m, 2 H) 3.38 – 3.45 (m, 1 H) 3.70 (t, J=8.78 Hz, 1 H) 4.44 (t, J=79.81 Hz, 3 H) 4.63 – 4.71 (m, 1 H) 6.70 – 6.80 (m, 2 H) 7.07 – 7.15 (m, 2 H) 7.07 – 7.12 (m, 1 H) 7.13 – 7.22 (m, 4 H); 19F NMR δ ppm -184.171. Data for P-2: (R)-3-((3R,4R)-3-fluoro-4-(4-hydroxyphenyl)piperidin- 1 -yl)- 1 -(4-methylbenzyl)pyrrolidin-2-one: LCMS (method F) RT 2.10 min, m/z 383.2 (M+H+), 405.2 (M+Na+); HPLC (method B) RT 8.29 min (99.7% AP); HPLC (method C) RT 6.52 min (99.8% AP); Chiral HPLC (method C-6) RT 6.92 min; 1H NMR (400 MHz, methanol-d4) δ ppm 1.80 – 1.90 (m, 2 H) 2.07 (d, J=8.03 Hz, 1 H) 2.19 (s, 1 H) 2.34 (s, 3 H) 2.41 – 2.48 (m, 1 H) 2.66 (d, J=4.52 Hz, 2 H) 2.95 – 3.03 (m, 1 H) 3.10 – 3.18 (m, 1 H) 3.20 – 3.30 (m, 2 H) 3.68 – 3.78 (m, 1 H) 4.38 (s, 1 H) 4.51 (d, J=14.56 Hz, 2 H) 6.70 – 6.80 (m, 2 H) 7.05 – 7.13 (m, 2 H) 7.13 – 7.22 (m, 4 H); 19F NMR δ ppm -184.311.

(3S,4S)-tert-Butyl 3-fluoro-4-(4-hydroxyphenyl)piperidine-l-carboxylate.

To a solution of (3S,4S)-tert-butyl 3-hydroxy-4-(4-hydroxyphenyl)piperidine-l-carboxylate (400 mg, 1.36 mmol, the first eluting enantiomer E-l from step E) in DCM (5 mL) cooled to 0 °C was added dropwise DAST (0.54 mL, 4.1 mmol) over 10 min. The mixture was allowed to warm up to rt and was stirred for 2h. The reaction was slowly quenched with 50 mL of a 10%> aqueous sodium bicarbonate solution and extracted four times with 50 mL of DCM. The combined organic layerss were washed with 75 mL of brine, dried, and concentrated under vacuum to yield 390 mg of {3S,4S)-tert-bvXy\ 3-fluoro-4-(4-hydroxyphenyl)piperidine-l-

carboxylate which was used without further purification. LCMS (Method Q) RT 0.92 min, m z 240.1(M+H+).

Step M. 4-((3S’,4S)-3-Fluoropi ridin-4-yl)phenol hydrochloride.

A mixture of (3S,4S)-tert-butyl 3-fluoro-4-(4-hydroxyphenyl)piperidine-l-carboxylate (390 mg, 1.3 mmol) and 4M HC1 in dioxane (3.3 mL, 13.2 mmol) in dioxane (4 mL) was stirred at rt for 2 hr. It was then concentrated to dryness, washed with 10 mL of 5% DCM/diethyl ether mixture and the solid was isolated by filtration. Yield: 260 mg of 4-((J£4S)-3-fluoropiperidin-4-yl)phenol hydrochloride; LCMS

(method Q) RT 0.46 min, mz 196.1(M+H+) 1H NMR (400 MHz, DMSO-d6) δ = 9.57 (br. s., 4 H), 8.92 – 8.68 (m, 1 H), 7.14 (d, J= 8.5 Hz, 1 H), 7.06 (d, J= 8.5 Hz, 2 H), 6.82 – 6.73 (m, 2 H), 5.07 – 4.85 (m, 1 H), 3.77 – 3.36 (m, 9 H), 3.32 – 3.22 (m, 2 H), 3.13 – 2.85 (m, 5 H), 2.06 – 1.88 (m, H).

Step N. 3-((3S,4S)-3-Fluoro-4-(4-hydroxyphenyl)piperidin- 1 -yl)- 1 -(4-methylbenzyl)pyrrolidin-2-one.

A mixture of 3-bromo-l-(4-methylbenzyl)pyrrolidin-2-one (200 mg, 0.75 mmol), triethylamine (0.52 mL, 3.7 mmol) and 4-((3S,4S)-3-fluoropiperidin-4-yl)phenol hydrochloride (173 mg, 0.75 mmol) in DMF (3 mL) was heated to 120 °C in a microwave reactor for 1.5 h. The mixture was allowed to cool and was then mixed with 60 mL water and extracted 5 times with 40 mL of DCM. The combined organic extracts were washed with 80 mL of brine, dried over anhydrous sodium sulfate, filtered, and evaporated to give 265 mg of 3-((3 4S)-3-fluoro-4-(4-hydroxy-phenyl)piperidin-l-yl)-l-(4-methylbenzyl)pyrrolidin-2-one as a mixture of 2 diastereoisomers. LCMS (method P) RT 0.92 min m/z 383.4 (M+H+).

Step O. (5)-3-((3lS,45)-3-Fluoro-4-(4-hydroxyphenyl)piperidin- 1 -yl)- 1 -(4-methylbenzyl)pyrrolidin-2-one and (i?)-3-((35,,45)-3-fluoro-4-(4-hydroxyphenyl)piperidin- 1 -yl)- 1 -(4-methylbenzyl)pyrrolidin-2-one.

A portion of the diasteromer mixture from step N (130 mg) was subjected to chiral purification via SFC (method C-7) to give homochiral Examples 46 P-3 (37.7 mg) and P-4 (60.7 mg). Data for P-3 (S)-3-((3S,4S)-3-fluoro-4-(4-hydroxyphenyl)piperidin- 1 -yl)- 1 -(4-methylbenzyl)pyrrolidin-2-one: LCMS (Method F) RT = 2.10 min, m/z 383.2 (M+H+); HPLC (Method C) RT 6.54 min, (Method D) RT 8.20 min; chiral HPLC (method C-6) RT 3.42 min;1H NMR (400 MHz, methanol-d4) δ ppm 1.76 – 1.86 (m, 2 H) 2.06 (d, J=8.53 Hz, 1 H) 2.10 – 2.21 (m, 1 H) 2.34 (s, 3 H) 2.40 – 2.48 (m, 1 H) 2.53 – 2.60 (m, 1 H) 2.61 – 2.70 (m, 2 H) 2.95 -3.01 (m, 1 H) 3.01 (s, 2 H) 3.10 – 3.16 (m, 1 H) 3.18 – 3.28 (m, 2 H) 3.72 (s, 1 H) 4.35 – 4.41 (m, 1 H) 4.46 – 4.70 (m, 2 H) 6.72 – 6.80 (m, 2 H) 7.05 – 7.23 (m, 6 H). Data for P-4 (R)-3-((3S,4S)-3-fiuoro-4-(4-hydroxyphenyl)piperidin-l-yl)-l-(4-methylbenzyl)pyrrolidin-2-one: LCMS (Method F) RT 2.11 min, m/z 383.2 (M+H+);; HPLC (Method C) RT 6.50 min, (Method D) RT 8.21 min; chiral HPLC (method C-6) RT 6.31 min; 1H NMR (400 MHz, methanol-d4) δ ppm 1.81 (dd, J=7.28, 2.76 Hz, 2 H) 2.06 (d, J=9.04 Hz, 2 H) 2.33 (s, 3 H) 2.43 (s, 1 H) 2.55 (br s, 1 H) 2.66 (d, J=40.16 Hz, 2 H) 2.75 – 2.80 (m, 1 H) 2.96 – 3.10 (m, 2 H) 3.20 – 3.28 (m, 2 H) 3.41 (d, J=5.52 Hz, 1 H) 3.66 – 3.75 (m, 1 H) 4.31 – 4.41 (m, 1 H) 4.46 – 4.71 (m, 2 H) 6.76 (d, J=8.53 Hz, 2 H) 7.05 – 7.23 (m, 6 H).

PATENT

https://patentscope.wipo.int/search/en/detail.jsf?docId=US215055421&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=PCTDescription

Example 46 (Peak-1, Peak-2, Peak-3, Peak-4)

(S)-3-((3R,4R)-3-Fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one and (R)-3-((3R,4R)-3-fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one


(S)-3-((3S,4S)-3-Fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one and (R)-3-((3S,4S)-3-fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one


Step A. (±)-rel-(3S,4S)-1-benzyl-4-(4-methoxyphenyl)piperidin-3-ol

      To a suspension of sodium tetrahydroborate (2.7 g, 72 mmol) in THF (200 mL) at 0° C. under a nitrogen atmosphere was added dropwise boron trifluoride etherate (8.8 mL, 70 mmol) and the resulting mixture was stirred for 30 minutes. Then 1-benzyl-4-(4-methoxyphenyl)-1,2,3,6-tetrahydropyridine (10 g, 36 mmol, from S. Halazy et al WO 97/28140 (8/7/97)) dissolved in 100 mL of tetrahydrofuran was added. The mixture was allowed to warm to rt and stirred for 2 h. The reaction was then quenched by the dropwise addition of 100 mL of water. Next were added sequentially 100 mL of ethanol, 100 mL of a 10% aqueous sodium hydroxide solution, and 30% hydrogen peroxide (18 mL, 180 mmol) and the mixture was stirred at reflux temperature overnight. The reaction mixture was then allowed to cool, diluted with saturated aqueous ammonium chloride (200 mL), and extracted with ethyl acetate (500 mL). The organic layer was dried over Na2SO4, filtered, and evaporated under reduced pressure to give (±)-rel-(3S,4S)-1-benzyl-4-(4-methoxyphenyl)piperidin-3-ol (8.5 g, 24.6 mmol, 69% yield) which was used without further purification. LCMS (Method K) RT 1.99 min; m/z 298.0 (M+H+).

Step B. (±)-rel-(3S,4S)-4-(4-methoxyphenyl)piperidin-3-ol


      To a solution of (±)-rel-(3S,4S)-1-benzyl-4-(4-methoxyphenyl)piperidin-3-ol (9 g, 30 mmol) in methanol (150 mL) was added 10% Pd/C (4.8 g) and the reaction mixture was stirred overnight under a hydrogen atmosphere. The catalyst was then removed by filtration through Celite and the solvent was evaporated under reduced pressure to give (±)-rel-(3S,4S)-4-(4-methoxyphenyl)piperidin-3-ol (5.1 g, 24.6 mmol, 81% yield) which was used without further purification. 1H NMR (400 MHz, DMSO-d6) δ ppm 7.10-7.15 (m, 2H) 6.80-6.86 (m, 2H) 4.30 (d, J=5.27 Hz, 1H) 3.37-3.43 (m, 1H) 3.04 (dd, J=11.58, 4.36 Hz, 1H) 2.86 (d, J=12.17 Hz, 1H) 2.43 (td, J=12.09, 2.67 Hz, 1H) 2.22-2.35 (m, 2H) 1.57-1.63 (m, 1H) 1.43-1.54 (m, 1H).

Step C. (±)-rel-(3S,4S)-tert-butyl 4-(4-(tert-butoxycarbonyloxy)phenyl)-3-hydroxypiperidine-1-carboxylate


 (
      To a solution of (±)-rel-(3S,4S)-4-(4-methoxyphenyl)piperidin-3-ol (4.5 g, 21.7 mmol) in DCM (150 mL) at −10° C. under nitrogen was added a 1 M solution of boron tribromide in DCM (109 mL, 109 mmol). The reaction mixture was allowed to warm to rt, stirred for 2 h, and then rechilled to 0° C. and quenched by the addition of a saturated aqueous sodium bicarbonate solution (300 mL). The aqueous layer was washed with 250 mL of DCM and then to it was added 200 mL 10% aqueous NaOH, followed by 9.5 g (43.5 mmol) of di-t-butyl dicarbonate and the resulting mixture was stirred for an additional 2 h. The mixture was then extracted with 200 mL ethyl acetate and the organic layer was separated, dried over Na2SO4, filtered, and evaporated under reduced pressure to (±)-rel-(3S,4S)-tert-butyl 4-(4-(tert-butoxycarbonyloxy)phenyl)-3-hydroxypiperidine-1-carboxylate (6.5 g, 12 mmol, 56% yield) which was used without further purification. LCMS (Method K) RT 2.33 min, m/z 282 (M+H+-2 t-butyl), 370; 1H NMR (400 MHz, DMSO-d6) δ ppm 7.27 (d, J=8.66 Hz, 2H) 7.08 (d, J=8.66 Hz, 2H) 4.85 (d, J=5.65 Hz, 1H) 4.13 (d, J=8.41 Hz, 1H) 3.97 (d, J=10.48 Hz, 1H) 3.45 (tt, J=10.27, 5.19 Hz, 1H) 1.67 (d, J=3.39 Hz, 1H) 1.50-1.59 (m, 1H) 1.49 (s, 11H).

Step D. (±)-rel-(3S,4S)-tert-butyl 3-hydroxy-4-(4-hydroxyphenyl)piperidine-1-carboxylate


      To a solution of (±)-rel-(3S,4S)-tert-butyl 4-(4-(tert-butoxycarbonyloxy)phenyl)-3-hydroxypiperidine-1-carboxylate (6.5 g, 16.5 mmol) in 100 mL of methanol was added 11.42 g of potassium carbonate (83 mmol) and the reaction mixture was stirred at rt for 5 h. The organic solvent was removed under reduced pressure and the residue was partitioned between 1N HCl (300 mL) and ethyl acetate (300 mL). The layers were separated and the organic layer was dried over Na2SOand evaporated under reduced pressure to give (±)-rel-(3S,4S)-tert-butyl 3-hydroxy-4-(4-hydroxyphenyl)piperidine-1-carboxylate (5 g, 15 mmol, 92% yield) which was used without further purification. LCMS (method F) RT 1.85 min, m/z 238 (M+H+-t-butyl), 279 (M+H+-t-butyl+CH3CN), 1H NMR (400 MHz, DMSO-d6) δ ppm 7.01 (d, J=8.53 Hz, 2H) 6.66 (d, J=8.53 Hz, 2H) 4.70 (d, J=5.02 Hz, 1H) 4.09 (br. s., 1H) 3.94 (d, J=11.55 Hz, 1H) 3.35-3.41 (m, 1H) 2.66-2.77 (m, 1H) 2.29-2.39 (m, 1H) 1.63 (dd, J=13.30, 3.26 Hz, 1H) 1.44-1.52 (m, 1H) 1.42 (s, 9H).

Step E. (3S,4S)-tert-Butyl 3-hydroxy-4-(4-hydroxyphenyl)piperidine-1-carboxylate and (3R,4R)-tert-butyl 3-hydroxy-4-(4-hydroxyphenyl)piperidine-1-carboxylate


      (±)-rel-(3S,4S)-tert-Butyl 3-hydroxy-4-(4-hydroxyphenyl)piperidine-1-carboxylate (5 g, 17 mmol, from step D) was subjected to chiral SFC separation (method C-5) to yield enantiomers E-1 (1.9 g, 6.48 mmol, 38.0% yield) and E-2 (2.4 g, 8.18 mmol, 48.0% yield). Data for E-1: chiral HPLC (method A5) retention time 3.42 min. Data for E-2: chiral HPLC (method A5) retention time 4.2 min.

Step F. (3R,4R)-tert-Butyl 4-(4-(benzyloxy)phenyl)-3-hydroxypiperidine-1-carboxylate


      A mixture of (3R,4R)-tert-butyl 3-hydroxy-4-(4-hydroxyphenyl)piperidine-1-carboxylate (620 mg, 2.1 mmol, E-2 from step E), potassium carbonate (584 mg, 4.2 mmol), and benzyl bromide (0.25 mL, 2.1 mmol) in DMF (5 mL) was stirred at rt for 16 h. The solvent was removed by evaporation and the residue was treated with 50 mL of water. The aqueous mixture was then extracted 4 times with 50 mL of chloroform. The combined organic phases were dried over anhydrous Na2SO4, filtered, and evaporated to yield 750 mg of (3R,4R)-tert-butyl 4-(4-(benzyloxy)phenyl)-3-hydroxypiperidine-1-carboxylate which was used without further purification. LCMS (method F) RT 2.28 min, m/z=310 (M+H+-t-butyl -water), 328 (M+H+-t-butyl).

Step G. (3R,4R)-4-(4-(Benzyloxy)phenyl)piperidin-3-ol hydrochloride


      A mixture of (3R,4R)-tert-butyl 4-(4-(benzyloxy)phenyl)-3-hydroxypiperidine-1-carboxylate (750 mg, 2 mmol), dioxane (4 mL) and 4.9 mL of 4 M HCl in dioxane was stirred at rt for 2 h. The reaction was then evaporated to dryness to yield 550 mg of (3R,4R)-4-(4-(Benzyloxy)phenyl)piperidin-3-ol hydrochloride which was used without further purification. LCMS (method J) RT 0.70 min, m/z 284 (M+H+).

Step H. 3-((3R,4R)-4-(4-(Benzyloxy)phenyl)-3-hydroxypiperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one


      A mixture of 3-bromo-1-(4-methylbenzyl)pyrrolidin-2-one (Intermediate 2, 220 mg, 0.82 mmol), (3R,4R)-4-(4-(benzyloxy)phenyl)piperidin-3-ol hydrochloride (262 mg, 0.82 mmol, from step G) and triethylamine (11 mL, 8.2 mmol) was stirred at 60° C. for 1 h, 80° C. for 1 h, 100° C. for 1 h and 120° C. for 1 h. The reaction mixture was then allowed to cool, diluted with 40 mL of water and extracted four times with 50 mL of chloroform. The combined organic layers were washed with 60 mL brine, dried over anhydrous sodium sulfate, filtered, and evaporated to yield 382 mg of 3-((3R,4R)-4-(4-(benzyloxy)phenyl)-3-hydroxypiperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one which was used without further purification. LCMS (method J) (main component of a mixture) RT 2.23 min, m/z 471 (M+H+).

Step I. 3-((3R,4R)-4-(4-(Benzyloxy)phenyl)-3-fluoropiperidin-1l-yl)-1-(4-methylbenzyl)pyrrolidin-2-one


      A solution of 3-(-4-(4-(benzyloxy)phenyl)-3-hydroxypiperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one (382 mg, 0.81 mmol) in DCM (5 mL) cooled to 0° C. was treated dropwise with DAST (0.32 mL, 2.4 mmol) over 3 min. The reaction mixture was then allowed to warm to rt and was stirred for 2 h. The reaction was then quenched with 50 mL of 10% aqueous sodium bicarbonate solution and extracted 4 times with 40 mL of DCM. The combined organic layers were washed with 50 mL of brine, dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum to yield 382 mg of 3-((3R,4R)-4-(4-(benzyloxy)phenyl)-3-fluoropiperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one as a mixture of two diastereomers and rearrangement products which was used without further purification. LCMS (method J) (main component of a mixture) RT 0.9 min, m/z 473 (M+H+).

Step J. 3-((3R,4R)-3-Fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one


      A mixture of 3-((3R,4R)-(4-(4-(benzyloxy)phenyl)-3-fluoropiperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one (382 mg, 0.81 mmol) and methanol (4 mL) was flushed with nitrogen, followed by the addition of 172 mg of 10% Pd/C. Then the mixture was stirred at rt overnight under 25-99 psi hydrogen pressure. The reaction was then transferred to a 100 mL autoclave and stirred at 7 kg/cmhydrogen pressure for 4 days. The catalyst was removed by filtration through Celite and the solvent was evaporated off. The crude product was subjected to HPLC purification (method B) to yield 77.3 mg 3-((3R,4R)-3-fluoro-4-(4-hydroxyphenyl)-piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one (diastereomeric pair) LCMS (method Q) RT 1.15 min, m/z 383.0 (M+H+).

Step K. (S)-3-((3R,4R)-3-Fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one and (R)-3-((3R,4R)-3-fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one

      The diastereomeric mixture from step J was separated by SFC method C-7 to yield homochiral Examples 46 P-1 (29.3 mg) and P-2 (32.8 mg). Data for P-1 (S)-3-((3R,4R)-3-fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one: LCMS (method F) RT 2.10 min, m/z 383.2 (M+H+), 405.2 (M+Na+); HPLC (method B) RT 8.24 min (98.8% AP); HPLC (method C) RT 6.52 min (99.1% AP); Chiral HPLC (method C-6) RT 4.1 min; 1H NMR (400 MHz, methanol-d4) δ ppm 1.76-1.86 (m, 2H) 2.07 (d, J=8.53 Hz, 1H) 2.13-2.21 (m, 1H) 2.34 (s, 3H) 2.43 (s, 0H) 2.55-2.60 (m, 1H) 2.65-2.70 (m, 1H) 2.75 (br. s., 1H) 3.20-3.30 (m, 2H) 3.38-3.45 (m, 1H) 3.70 (t, J=8.78 Hz, 1H) 4.44 (t, J=79.81 Hz, 3H) 4.63-4.71 (m, 1H) 6.70-6.80 (m, 2H) 7.07-7.15 (m, 2H) 7.07-7.12 (m, 1H) 7.13-7.22 (m, 4H); 19F NMR δ ppm −184.171. Data for P-2: (R)-3-((3R,4R)-3-fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one: LCMS (method F) RT 2.10 min, m/z 383.2 (M+H+), 405.2 (M+Na+); HPLC (method B) RT 8.29 min (99.7% AP); HPLC (method C) RT 6.52 min (99.8% AP); Chiral HPLC (method C-6) RT 6.92 min; 1H NMR (400 MHz, methanol-d4) δ ppm 1.80-1.90 (m, 2H) 2.07 (d, J=8.03 Hz, 1H) 2.19 (s, 1H) 2.34 (s, 3H) 2.41-2.48 (m, 1H) 2.66 (d, J=4.52 Hz, 2H) 2.95-3.03 (m, 1H) 3.10-3.18 (m, 1H) 3.20-3.30 (m, 2H) 3.68-3.78 (m, 1H) 4.38 (s, 1H) 4.51 (d, J=14.56 Hz, 2H) 6.70-6.80 (m, 2H) 7.05-7.13 (m, 2H) 7.13-7.22 (m, 4H); 19F NMR δ ppm −184.311.

Step L. (3S,4S)-tert-Butyl 3-fluoro-4-(4-hydroxyphenyl)piperidine-1-carboxylate


      To a solution of (3S,4S)-tert-butyl 3-hydroxy-4-(4-hydroxyphenyl)piperidine-1-carboxylate (400 mg, 1.36 mmol, the first eluting enantiomer E-1 from step E) in DCM (5 mL) cooled to 0° C. was added dropwise DAST (0.54 mL, 4.1 mmol) over 10 min. The mixture was allowed to warm up to rt and was stirred for 2 h. The reaction was slowly quenched with 50 mL of a 10% aqueous sodium bicarbonate solution and extracted four times with 50 mL of DCM. The combined organic layers were washed with 75 mL of brine, dried, and concentrated under vacuum to yield 390 mg of (3S,4S)-tert-butyl 3-fluoro-4-(4-hydroxyphenyl)piperidine-1-carboxylate which was used without further purification. LCMS (Method Q) RT 0.92 min, m/z 240.1 (M+H+).

Step M. 4-((3S,4S)-3-Fluoropiperidin-4-yl)phenol hydrochloride


      A mixture of (3S,4S)-tert-butyl 3-fluoro-4-(4-hydroxyphenyl)piperidine-1-carboxylate (390 mg, 1.3 mmol) and 4M HCl in dioxane (3.3 mL, 13.2 mmol) in dioxane (4 mL) was stirred at rt for 2 hr. It was then concentrated to dryness, washed with 10 mL of 5% DCM/diethyl ether mixture and the solid was isolated by filtration. Yield: 260 mg of 4-((3S,4S)-3-fluoropiperidin-4-yl)phenol hydrochloride; LCMS (method Q) RT 0.46 min, mz 196.1 (M+H+)1H NMR (400 MHz, DMSO-d6) δ=9.57 (br. s., 4H), 8.92-8.68 (m, 1H), 7.14 (d, J=8.5 Hz, 1H), 7.06 (d, J=8.5 Hz, 2H), 6.82-6.73 (m, 2H), 5.07-4.85 (m, 1H), 3.77-3.36 (m, 9H), 3.32-3.22 (m, 2H), 3.13-2.85 (m, 5H), 2.06-1.88 (m, H).

Step N. 3-((3S,4S)-3-Fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one


      A mixture of 3-bromo-1-(4-methylbenzyl)pyrrolidin-2-one (200 mg, 0.75 mmol), triethylamine (0.52 mL, 3.7 mmol) and 4-((3S,4S)-3-fluoropiperidin-4-yl)phenol hydrochloride (173 mg, 0.75 mmol) in DMF (3 mL) was heated to 120° C. in a microwave reactor for 1.5 h. The mixture was allowed to cool and was then mixed with 60 mL water and extracted 5 times with 40 mL of DCM. The combined organic extracts were washed with 80 mL of brine, dried over anhydrous sodium sulfate, filtered, and evaporated to give 265 mg of 3-((3S,4S)-3-fluoro-4-(4-hydroxy-phenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one as a mixture of 2 diastereoisomers. LCMS (method P) RT 0.92 min m/z 383.4 (M+H+).

Step O. (S)-3-((3S,4S)-3-Fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one and (R)-3-((3S,4S)-3-fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one

      A portion of the diastereomer mixture from step N (130 mg) was subjected to chiral purification via SFC (method C-7) to give homochiral Examples 46 P-3 (37.7 mg) and P-4 (60.7 mg). Data for P-3 (S)-3-((3S,4S)-3-fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one: LCMS (Method F) RT=2.10 min, m/z 383.2 (M+H+); HPLC (Method C) RT 6.54 min, (Method D) RT 8.20 min; chiral HPLC (method C-6) RT 3.42 min; 1H NMR (400 MHz, methanol-d4) δ ppm 1.76-1.86 (m, 2H) 2.06 (d, J=8.53 Hz, 1H) 2.10-2.21 (m, 1H) 2.34 (s, 3H) 2.40-2.48 (m, 1H) 2.53-2.60 (m, 1H) 2.61-2.70 (m, 2H) 2.95-3.01 (m, 1H) 3.01 (s, 2H) 3.10-3.16 (m, 1H) 3.18-3.28 (m, 2H) 3.72 (s, 1H) 4.35-4.41 (m, 1H) 4.46-4.70 (m, 2H) 6.72-6.80 (m, 2H) 7.05-7.23 (m, 6H). Data for P-4 (R)-3-((3S,4S)-3-fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one: LCMS (Method F) RT 2.11 min, m/z 383.2 (M+H+); HPLC (Method C) RT 6.50 min, (Method D) RT 8.21 min; chiral HPLC (method C-6) RT 6.31 min; 1H NMR (400 MHz, methanol-d4) δ ppm 1.81 (dd, J=7.28, 2.76 Hz, 2H) 2.06 (d, J=9.04 Hz, 2H) 2.33 (s, 3H) 2.43 (s, 1H) 2.55 (br s, 1H) 2.66 (d, J=40.16 Hz, 2H) 2.75-2.80 (m, 1H) 2.96-3.10 (m, 2H) 3.20-3.28 (m, 2H) 3.41 (d, J=5.52 Hz, 1H) 3.66-3.75 (m, 1H) 4.31-4.41 (m, 1H) 4.46-4.71 (m, 2H) 6.76 (d, J=8.53 Hz, 2H) 7.05-7.23 (m, 6H).

ADDITIONAL INFORMATION

Intravenous administration of BMS-986169 or BMS-986163 dose-dependently increased GluN2B receptor occupancy and inhibited in vivo [3H](+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine ([3H]MK-801) binding, confirming target engagement and effective cleavage of the prodrug. BMS-986169 reduced immobility in the mouse forced swim test, an effect similar to intravenous ketamine treatment. Decreased novelty suppressed feeding latency, and increased ex vivo hippocampal long-term potentiation was also seen 24 hours after acute BMS-986163 or BMS-986169 administration. BMS-986169 did not produce ketamine-like hyperlocomotion or abnormal behaviors in mice or cynomolgus monkeys but did produce a transient working memory impairment in monkeys that was closely related to plasma exposure. Finally, BMS-986163 produced robust changes in the quantitative electroencephalogram power band distribution, a translational measure that can be used to assess pharmacodynamic activity in healthy humans. Due to the poor aqueous solubility of BMS-986169, BMS-986163 was selected as the lead GluN2B NAM candidate for further evaluation as a novel intravenous agent for TRD.

ADDITIONAL INFORMATION

Intravenous administration of BMS-986169 or BMS-986163 dose-dependently increased GluN2B receptor occupancy and inhibited in vivo [3H](+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine ([3H]MK-801) binding, confirming target engagement and effective cleavage of the prodrug. BMS-986169 reduced immobility in the mouse forced swim test, an effect similar to intravenous ketamine treatment. Decreased novelty suppressed feeding latency, and increased ex vivo hippocampal long-term potentiation was also seen 24 hours after acute BMS-986163 or BMS-986169 administration. BMS-986169 did not produce ketamine-like hyperlocomotion or abnormal behaviors in mice or cynomolgus monkeys but did produce a transient working memory impairment in monkeys that was closely related to plasma exposure. Finally, BMS-986163 produced robust changes in the quantitative electroencephalogram power band distribution, a translational measure that can be used to assess pharmacodynamic activity in healthy humans. Due to the poor aqueous solubility of BMS-986169, BMS-986163 was selected as the lead GluN2B NAM candidate for further evaluation as a novel intravenous agent for TRD.

 

REFERENCES

1: Bristow LJ, Gulia J, Weed MR, Srikumar BN, Li YW, Graef JD, Naidu PS, Sanmathi
C, Aher J, Bastia T, Paschapur M, Kalidindi N, Kumar KV, Molski T, Pieschl R,
Fernandes A, Brown JM, Sivarao DV, Newberry K, Bookbinder M, Polino J, Keavy D,
Newton A, Shields E, Simmermacher J, Kempson J, Li J, Zhang H, Mathur A, Kallem
RR, Sinha M, Ramarao M, Vikramadithyan RK, Thangathirupathy S, Warrier J, Islam
I, Bronson JJ, Olson RE, Macor JE, Albright CF, King D, Thompson LA, Marcin LR,
Sinz M. Preclinical Characterization of
(R)-3-((3S,4S)-3-fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrr
olidin-2-one (BMS-986169), a Novel, Intravenous, Glutamate N-Methyl-d-Aspartate
2B Receptor Negative Allosteric Modulator with Potential in Major Depressive
Disorder. J Pharmacol Exp Ther. 2017 Dec;363(3):377-393. doi:
10.1124/jpet.117.242784. Epub 2017 Sep 27. PubMed PMID: 28954811.

2. BMS-986163, a Negative Allosteric Modulator of GluN2B with Potential Utility in Major Depressive Disorder
Lawrence R. Marcin, Jayakumar Warrier, Srinivasan Thangathirupathy, Jianliang Shi, George N. Karageorge, Bradley C. Pearce, Alicia Ng, Hyunsoo Park, James Kempson, Jianqing Li, Huiping Zhang, Arvind Mathur, Aliphedi B. Reddy, G. Nagaraju, Gopikishan Tonukunuru, Grandhi V. R. K. M. Gupta, Manjunatha Kamble, Raju Mannoori, Srinivas Cheruku, Srinivas Jogi, Jyoti Gulia, Tanmaya Bastia, Charulatha Sanmathi, Jayant Aher, Rajareddy Kallem, Bettadapura N. Srikumar, Kumar Kuchibhotla Vijaya, Pattipati S. Naidu, Mahesh Paschapur, Narasimharaju Kalidindi, Reeba Vikramadithyan, Manjunath Ramarao, Rex Denton, Thaddeus Molski, Eric Shields, Murali Subramanian, Xiaoliang Zhuo, Michelle Nophsker, Jean Simmermacher, Michael Sinz, Charlie Albright, Linda J. Bristow, Imadul Islam, Joanne J. Bronson, Richard E. Olson, Dalton King, Lorin A. Thompson, and John E. Macor
Publication Date (Web): April 13, 2018 (Letter)
DOI: 10.1021/acsmedchemlett.8b00080

Patent ID Patent Title Submitted Date Granted Date
US9221796 Selective NR2B antagonists
2015-01-05
2015-12-29

//////////////////BMS-986169, BMS-986169, BMS 986169, BMS986169

 O=C1N(CC2=CC=C(C)C=C2)CC[C@H]1N3C[C@@H](F)[C@H](C4=CC=C(O)C=C4)CC3

K-8986

$
0
0

Figure

YNRQDEGURLSOGN-BTJKTKAUSA-N.png

K-8986

(Z)-but-2-enedioic acid;7-[3-[4-[[1-(2-ethoxyethyl)benzimidazol-2-yl]methyl]piperazin-1-yl]propoxy]-4H-1,4-benzothiazin-3-one

cas 1335112-55-4 mono maleate

cas 1335112-57-6  di maleate

cas 219741-69-2 free form

C27 H35 N5 O3 S . C4 H4 O4
2H-1,4-Benzothiazin-3(4H)-one, 7-[3-[4-[[1-(2-ethoxyethyl)-1H-benzimidazol-2-yl]methyl]-1-piperazinyl]propoxy]-, (2Z)-2-butenedioate (1:1)
7-[3-[4-[[1-(2-Ethoxyethyl)benzimidazol-2-yl]methyl]-1-piperazinyl]propoxy]-3,4-dihydro-2H-1,4-benzothiazin-3-one monomaleate
KOWA CO., LTD.
福田 友昭 FUKUDA, Tomoaki; JP
纐纈 章泰 KOKETSU, Akiyasu; JP
金児 佳生 KANEKO, Yoshio; JP
芦川 由香 ASHIKAWA, Yuka; JP

Image result for KOWA CO., LTD.

Mono maleate

1H NMR (396 MHz, DMSO-d6) δ 1.03 (t, J = 7.0 Hz, 3H), 2.04–2.08 (m, 2H), 3.10 (br, 8H), 3.18 (br, 2H), 3.38 (t, J = 7.0 Hz, 2H), 3.42 (s, 2H), 3.71 (t, J = 7.9 Hz, 2H), 3.95 (s, 2H), 4.01 (t, J = 5.9 Hz, 2H), 4.51 (t, J = 5.2 Hz, 2H), 6.06 (s, 2H), 6.79 (dd, J = 9.1, 2.7 Hz, 1H), 6.90–6.92 (m, 2H), 7.17–7.26 (m, 2H), 7.58–7.61 (m, 2H), 10.43 (s, 1H);

13C NMR (100 MHz, DMSO-d6) δ 15.0, 23.8, 29.0, 43.5, 49.7 (×2), 51.3 (×2), 53.2, 53.4, 65.3, 65.7, 68.7, 110.7, 112.8, 113.9, 118.2, 118.8, 120.3, 121.6, 122.2, 131.3, 135.6, 135.8 (×2), 141.8, 150.6, 153.7, 164.7, 167.3 (×2);

HRMS (FD) calcd for C27H36N5O3S [(MH – maleic acid)+] 510.2539, found 510.2558.

Allergic conjunctivitis, which can be classified into seasonal allergic conjunctivitis and perennial allergic conjunctivitis, is a type I hypersensitivity to allergens. Symptoms such as itching, redness, eyelid swelling, and chemosis are common among afflicted patients and are caused by the release of chemical mediators such as histamine from activated mast cells through cross-linking of antigen-specific immunoglobulin E. The binding of histamine to its receptors plays a central role in the induction of allergic symptoms. K-8986 (1), a histamine H1-receptor antagonist, was developed as a potential therapeutic for treatment of allergic conjunctivitis

SYN

Clip

Development of a Synthetic Process for K-8986, an H1-Receptor Antagonist

Tomoaki Fukuda* Takeaki HaraShinji InaTetsuhiro Nemoto , and Takeshi Oshima*

 Tokyo New Drug Research Laboratories, Pharmaceutical DivisionKowa Company, Ltd.2-17-43, Noguchicho, Higashimurayama, Tokyo 189-0022, Japan
 Graduate School of Pharmaceutical SciencesChiba University1-8-1, Inohana, Chuo-ku, Chiba 260-8675, Japan
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.8b00380
This article is part of the Japanese Society for Process Chemistry special issue.
Abstract Image

This article describes the development of a robust and scalable synthetic process for K-8986 (1). To solve the problems in terms of the physicochemical properties of 6 (a free base unit of 1), we have screened the suitable salt forms of the target. The monomaleate salt was the most suitable form for the API. To overcome challenges regarding the unremovable impurity Imp B caused by the carryover of piperazine in the medicinal chemistry route, we designed and developed a novel synthetic route. This route furnished more opportunities to purify the synthetic intermediates after introduction of the piperazine unit. Both impurities and co-products in each step of the revised synthesis could be easily removed via filtration, leveraging the low solubility of benzothiazine derivatives. The newly established process was applied to the synthesis of 1 (the monomaleate salt of 6) on a practical scale, achieving high purity and reproducibility.

1H NMR (396 MHz, DMSO-d6) δ 1.03 (t, J = 7.0 Hz, 3H), 2.04–2.08 (m, 2H), 3.10 (br, 8H), 3.18 (br, 2H), 3.38 (t, J = 7.0 Hz, 2H), 3.42 (s, 2H), 3.71 (t, J = 7.9 Hz, 2H), 3.95 (s, 2H), 4.01 (t, J = 5.9 Hz, 2H), 4.51 (t, J = 5.2 Hz, 2H), 6.06 (s, 2H), 6.79 (dd, J = 9.1, 2.7 Hz, 1H), 6.90–6.92 (m, 2H), 7.17–7.26 (m, 2H), 7.58–7.61 (m, 2H), 10.43 (s, 1H);

13C NMR (100 MHz, DMSO-d6) δ 15.0, 23.8, 29.0, 43.5, 49.7 (×2), 51.3 (×2), 53.2, 53.4, 65.3, 65.7, 68.7, 110.7, 112.8, 113.9, 118.2, 118.8, 120.3, 121.6, 122.2, 131.3, 135.6, 135.8 (×2), 141.8, 150.6, 153.7, 164.7, 167.3 (×2);

PATENT

WO2011115173

https://patentscope.wipo.int/search/en/detail.jsf;jsessionid=CB6FAC725A85FC9DDE6D08A63CD4B038.wapp1nB?docId=WO2011115173&tab=FULLTEXT&queryString=ALL%3A%28%25E7%2582%258E%25E7%2597%2587%25E6%2580%25A7%25E8%2585%25B8%25E7%2596%25BE%25E6%2582%25A3%29&recNum=236&maxRec=6346

Example 1-1 Production of 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4- dihydro-2H- 1,4-benzothiazin- Production of On (1a) (Manufacture of Free Body)

[Chemical Formula 5]

 a) 65 g (359 mmol) of 7-hydroxy-3,4-dihydro-2H-1,4-benzothiazin-3-one obtained by the method described in JP-A-60-4176 and JP-A-59-70675, Was suspended in tetrahydrofuran (194 mL) under an argon atmosphere, 104 g (397 mmol) of triphenylphosphine and 32 mL (379 mmol) of 3-chloropropanol were added and the mixture was cooled to 0 ° C. Next, 78 mL (396 mmol) of azodicarboxylic acid diisopropyl ester was added dropwise to the obtained reaction solution at 30 ° C. or less, and the mixture was stirred at room temperature for 1 hour. The solvent was distilled off from the resulting solution under reduced pressure, methanol (390 mL) was added thereto, and the mixture was stirred at room temperature for 1 hour. The precipitated crystals were collected by filtration and then dried under reduced pressure at 50 ° C. for 5 hours to obtain 59 g (yield 64%) of 7- (3-chloropropoxy) -3,4-dihydro-2H-1,4-benzothiazin- ) As blue-white crystals.
[Chemical Formula 6]
1 H-NMR (400 MHz, DMSO-d 6 ) δ: 2.12 (2H, quint, J = 6.2 Hz), 3.28 (2H, s), 3.76 (2H, t, J = (2H, t, J = 5.8 Hz), 6.78 (1 H, dd, J = 2.8, 8.8 Hz), 6.88 (1 H, d, J = 8.8 Hz ), 6.90 (1 H, d, J = 2.8 Hz), 10.38 (1 H, s)
 57 g (221 mmol) of 7- (3-chloropropoxy) -3,4-dihydro-2H-1,4-benzothiazin-3-one was suspended in dimethylformamide (172 mL), 49 g (355 mmol) of potassium carbonate, 40 g (241 mmol) of potassium iodide and 43 g (231 mmol) of Nt-butoxycarbonylpiperazine were added and the mixture was heated to 100 ° C. and stirred for 4 hours. Water (344 mL) was added to the reaction solution, and the mixture was cooled to 0 ° C. and further stirred at the same temperature for 1 hour. The precipitated crystals were collected by filtration and then dried under reduced pressure at 50 ° C. for 5 hours to give 7- [3- (Nt-butoxycarbonylpiperazinyl) propoxy] -3,4-dihydro-2H-1,4-benzothiazine -3-one (89% yield) as bluish-white crystals.
[Chemical Formula 7]
1 H-NMR (400 MHz, DMSO-d 6 ) δ: 1.39 (9 H, s), 1.83 (2 H, quint, J = 6.8 Hz), 2.31 (4 H, t, J = 4. 3.30 (2H, t, J = 4.6 Hz), 3.41 (2H, s), 3.95 (2H, t, J = 6.4 Hz), 6.78 (1 H, dd, J = 2.8, 8.8 Hz), 6.88 (1 H, d, J = 8.8 Hz), 6.89 (1 H, s) 10.38 (1 H, s)
 c) 87 g (214 mmol) of 7- {3- (Nt-butoxycarbonylpiperazinyl) propoxy} -3,4-dihydro-2H- 1,4-benzothiazin-3-one was suspended in ethanol (174 mL) , 6N hydrochloric acid aqueous solution (174 mL) was added dropwise at 50 ° C., and the mixture was stirred at the same temperature for 1 hour. Ethanol (522 mL) was added to the reaction solution, followed by cooling to 0 ° C. and further stirring at the same temperature for 1 hour. The precipitated crystals were collected by filtration and then dried under reduced pressure at 50 ° C. for 5 hours to give 7- {3- (piperazin-1-yl) propoxy} -3,4-dihydro-2H-1,4-benzothiazin- · Hydrochloride salt 75 g (yield 92%) was obtained as blue-white crystals.
[Chemical Formula 8]
1 H-NMR (400 MHz, D 2 O) [delta]: 2.13 (2H, td, J = 5.9,15.6Hz), 3.34 (2H, s), 3.35 (2H, t, J = 8.0 Hz), 3.44-3.64 (8H, m), 4.02 (2H, t, J = 5.6 Hz), 6.74 (1H, dd, J = 2.4, 6.85 (1 H, d, J = 8.8 Hz), 6.90 (1 H, d, J = 2.4 Hz)
 d) 1- (2-ethoxyethyl) -2-chloromethyl-1H-benzimidazole obtained by the method described in Journal of Heterocyclic Chemistry (1987), 24 (1), 31-37 was dissolved in tetrahydrofuran (293 mL) and Was dissolved in a mixture of water (147 mL), and 7- {3- (Nt-butoxycarbonylpiperazinyl) propoxy} -3,4-dihydro-2H- 73 g (192 mmol) of 1,4-benzothiazin-3-one was added. Then, 117 mL (673 mmol) of diisopropylethylamine and 35 g (211 mmol) of potassium iodide were added, and the mixture was stirred at room temperature for 15 hours. Ethyl acetate (293 mL) and water (147 mL) were added to the reaction solution and extracted, and the organic layer was washed with 20% brine (147 mL). The organic layer was concentrated under reduced pressure to give 115 g (2 steps, quantitative) of the title compound (1a) as a brown oil.
1 H-NMR (400 MHz, CDCl 3 ) δ: 1.13 (3H, t, J = 7.0 Hz), 1.93 (2H, quint, J = 6.9 Hz), 2.40-2.70 (2H, s), 3.42 (2H, q, J = 6.8 Hz), 3.76 (2H, t, J = 7.2 Hz), 2.51 5. 2 (t, J = 6.0 Hz), 3.88 (2H, s), 3.97 (2H, t, J = 6.2 Hz), 4.51 (2H, t, J = 5.8 Hz), J = 8.8 Hz), 6.85 (1 H, d, J = 2.4 Hz), 7.24 (1 H, d, -7.28 (2H, m), 7.39 (1 H, ddd, J = 1.2, 6, 6.8 Hz), 7.73 (1 H, ddd, J = 1.2, 6.0 , 6.8 Hz) 8.35 (1H, s)
Example 1-2: 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4- dihydro-2H-1,4-benzothiazin- Production of On Monomaleate (2a) (Production of Seed Crystal)
[Chemical Formula 9]
 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4- dihydro-2H- 1,4-benzothiazin-3-one (1a) 0 g (1.96 mmol) was dissolved in ethanol (8 mL) and warmed to 60 ° C. After adding 211 mg (1.80 mmol) of maleic acid and stirring at 50 ° C. for 1 hour, the mixture was stirred at room temperature for 16 hours and further stirred at 0 ° C. for 3 hours. The precipitated crystals were collected by filtration and then dried under reduced pressure at 50 ° C. for 5 hours to obtain 1.02 g (yield 91%) of the monomaleate (2a) as bluish white crystals (melting point: 148 ° -151 ° C.).
Examples 1-3: 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4- dihydro-2H- 1,4-benzothiazin- Preparation of on-monomaleate (2a)
 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4- dihydro-2H- 1,4-benzothiazin-3-one (1a). After dissolving 0 g (13.7 mmol) in ethanol (56 mL) and heating to 60 ° C., 1.46 g (12.6 mmol) of maleic acid was added and the mixture was cooled to 50 ° C. to obtain 0.035 g (0.056 mmol) of seed crystals was added. The reaction solution was stirred at 50 ° C. for 1 hour, then stirred at room temperature for 1 hour, and further stirred at 0 ° C. for 3 hours. The precipitated crystals were collected by filtration and then dried under reduced pressure at 50 ° C. for 5 hours to obtain 7.08 g (yield 90%) of monomaleate (2a) as bluish-white crystals.
1 H-NMR (400 MHz, DMSO-d 6 ) δ: 1.02 (3H, t, J = 7.2 Hz), 2.00-2.07 (2H, m), 2.80-3.61 J = 5.2 Hz), 3.93 (2H, q, J = 6.9 Hz), 3.42 (2H, s), 3.71 (2H, (2H, t, J = 5.2 Hz), 6.03 (2H, s), 6.78 (1 H, dd, J = 2.4, 8.8 Hz), 6.88 (1 H, s), 6.91 (1 H, dd, J = 2.4, 2.4 Hz), 7.18 (1 H, ddd, J = 1 (2H, d, J = 8.4 Hz), 7.24 (1H, ddd, J = 1.4, 7.5, 7.5 Hz), 7.59 10.40 (1 H, s)
 Elementary analysis value of the  monomaleate (2a) obtained in Example 1-3: C 31 H 39 N 5 O 7 S
: theoretical value: C 59.50%; H 6.28%; N 11.19 %
Found: C 59.33%; H 6.29%; N 11.10%
 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4- dihydro-2H- 1,4-benzothiazine obtained in Example 1-3 -3-one monomaleate (2a) was subjected to thermal analysis measurement. In the thermal analysis measurement, approximately 5 mg of a sample was accurately weighed in an aluminum pan for thermal analysis, Al 2 O 3 was used as a reference substance , and the temperature was raised at a heating rate of 10 ° C./min in the presence of an atmosphere of N 2 gas (150 mL / min) (DTA) and thermogravimetry (TG) using a Thermo Plus 2 system (manufactured by Rigaku) as a thermal analyzer. The results of the thermal analysis measurement are shown in FIG. The melting point of the monomaleate (2a) was 147-150 ° C. (B – 545, manufactured by BUCHI).
 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4- dihydro-2H- 1,4-benzothiazine obtained in Example 1-3 -3-one monomaleate (2a) by infrared spectrophotometer (manufactured by Thermo Nicolet Co., Ltd., AVATAR 370; ATR method) shows the pattern shown in FIG. 2, and it is in the vicinity of 1669 cm -1 , 1492Cm -1 around, 1231Cm -1 around, 1208Cm -1around, 868Cm -1 and around 754Cm -1 had an absorption peak specific to the vicinity.
 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4- dihydro-2H- 1,4-benzothiazine obtained in Example 1-3 -3-one monomaleate (2a) was measured by powder X-ray diffraction (Miniflex manufactured by Rigaku Denki Kogyo Co., Ltd.). Measurement of powder X-ray crystal diffraction was carried out by filling the sample in the sample holder part of the silicon non-reflecting sample plate for X-ray diffraction and measuring with a desktop X-ray diffractometer: MiniFlex (Rigaku) a scanning range of diffraction angle 2θ; 3.00 ° to 40.00 °, sampling width: 0.02 °, and scanning speed: 2.00 ° / min. The obtained diffraction pattern is shown in FIG. 3. The monomaleate (2a) had specific diffraction angles and relative intensities shown in Table 1
[table 1]
Examples 1-4: 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4- dihydro-2H- 1,4-benzothiazin-3- Preparation of On Monomaleate (2a) (Study of Reproducibility on Large Scale)
 (1a) (115 g) was added to a solution of 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4-dihydro-2H-1,4-benzothiazin- 226 mmol) was dissolved in ethanol (293 mL), activated charcoal 5.5 g was added, and the mixture was stirred at room temperature for 1 hour, then filtered through celite and washed with ethanol (147 mL) and washed. Ethanol (147 mL) was added to the filtrate, and after heating to 60 ° C., 18.9 g (163 mmol) of maleic acid was added and cooled to 50 ° C. 0.58 g (0.93 mmol) of the seed crystals of the monomaleate (2a) obtained in Example 1-3 was added and stirred at 50 ° C. for 1 hour, followed by stirring at room temperature for 15 hours and further at 0 ° C. And the mixture was stirred for 3 hours. The precipitated crystals were collected by filtration and dried under reduced pressure at 50 ° C. for 5 hours to obtain 75.2 g (yield 63%) of monomaleate (2a) as white crystals (melting point: 147 ° -149 ° C.).
 Elementary analysis value of the  monomaleate (2a) obtained in Examples 1-4: C 31 H 39 N 5 O 7 S
: theoretical value: C 59.50%; H 6.28%; N 11.19 %
Found: C 59.41%; H 6.29%; N 11.08%
Comparative Example 1 Synthesis of 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4- dihydro-2H- 1,4-benzothiazin- Preparation of dimaleate
 15. 9 g (31 (3-ethoxyethylbenzoimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4-dihydro-2H-1,4- benzothiazin- . 1 mmol) was dissolved in 70 mL of ethanol, the solution was heated to 60 ° C., 8.0 g (68.9 mmol) of maleic acid was added, and the mixture was stirred at room temperature for 15 hours. The precipitated crystals were collected by filtration and then dried under reduced pressure at 50 ° C. for 5 hours to give 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4- 13.3 g of dihydro-2H-1,4-benzothiazin-3-one / dimaleate was obtained. The obtained compound was dissolved in methanol (13 mL), heated to 60 ° C., THF (52 mL) was added, and the mixture was stirred at room temperature for 20 hours. The obtained crystals were collected by filtration and dried under reduced pressure at 50 ° C. for 5 hours to give 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4 -Dihydro-2H-1,4-benzothiazin-3-one · dimaleate was obtained as blueish white crystals.
1 H-NMR (400 MHz, DMSO-d 6 ) δ: 1.01 (3H, t, J = 7.0 Hz), 2.00-2.07 (2H, m), 3.00 (4H, m) , 3.20 (2H, m), 3.37 (2H, q, J = 6.9 Hz), 3.41-3.47 (4H, m), 3.70 (2H, t, J = 5. (2H, t, J = 5.8 Hz), 4.50 (2H, t, J = 5.0 Hz), 6.14 (4H, s), 3.95 (2H, s) , 6.76 (1 H, dd, J = 2.4, 8.8 Hz), 6.88 (1 H, s), 6.90 (1 H, m), 7.19 – 7.27 (2 H, m) , 7.60 (2H, d, J = 7.6 Hz), 10.40 (1 H, s)
Comparative Example 2 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4- dihydro-2H- 1,4-benzothiazin-3-one Production of monofumarate
 6.81 g of 13- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) – 1 – piperazinyl} propoxy] -3,4- dihydro-2H-1,4-benzothiazin- . 3 mmol) was dissolved in a mixed solvent of ethanol (60 mL) and (water 6 mL), and the mixture was heated to 60 ° C. To the mixed solution was added a mixed solution of ethanol (14 mL) containing 1.55 g (13.3 mmol) of fumaric acid and water (1.5 mL), the mixture was stirred at 40 ° C. for 30 minutes, and further stirred at room temperature for 20 hours . The precipitated crystals were collected by filtration and dried under reduced pressure at 40 ° C. for 53.5 hours to give 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] 6.16 g (yield: 74%) of 4-dihydro-2H-1,4-benzothiazin-3-one monofumarate was obtained as slightly yellow crystals.
1 H-NMR (400 MHz, DMSO-d 6 ) δ: 1.01 (3H, t, J = 7.0 Hz), 1.81 (2H, quint, J = 6.6 Hz), 2.40-2. J = 5.6 Hz), 3.78 (2H, s), 3.93 (2H, m), 3.72 (2H, J = 6.4 Hz), 4.47 (2H, t, J = 5.2 Hz), 6.60 (2H, s), 6.75 (1 H, dd, J = 3.0, 9.0 Hz , 6.87 (1 H, d, J = 8.8 Hz), 6.89 (1 H, s), 7.15 (1 H, t, J = 7.6 Hz), 7.20 (1 H, t, J = 7.4 Hz), 7.54 (2H, t, J = 7.6 Hz), 10.36 (1 H, s)
Comparative Example 3 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4- dihydro-2H- 1,4-benzothiazin-3-one Production of disulfate

 8.28 g (16 parts) of 7- [3- {4- (N-ethoxyethylbenzoimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4-dihydro-2H- 1,4-benzothiazin- . 2 mmol) was dissolved in a mixed solvent of ethanol (104 mL) and water (11 mL) and cooled to 0 ° C. A solution of 3.19 g (16.2 mmol) of sulfuric acid in water (11 mL) was added dropwise and the mixture was stirred at 40 ° C. for 30 minutes, and further stirred at room temperature for 20 hours. The precipitated crystals were collected by filtration and dried under reduced pressure at 40 ° C. for 53.5 hours to give 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] (86% yield) of 4-dihydro-2H-1,4-benzothiazin-3-one disulfate as slightly yellow crystals.

1 H-NMR (400 MHz, DMSO-d 6 ) δ: 1.02 (3H, t, J = 6.8 Hz), 2.03 (2H, m), 2.65 (2H, m), 3.00 (4H, m), 3.26 (2H, m), 3.37 (2H, q, J = 6.8 Hz), 3.41-3.47 (4H, m), 3.75 , J = 5.0 Hz), 4.01 (2H, t, J = 5.8 Hz), 4.21 (2H, brs), 4.65 (2H, t, J = 5.0 Hz), 6.78 J = 8.8 Hz), 6.90 (1 H, d, J = 3.2 Hz), 7.50 – (1 H, d, J = 2.8, 9.2 Hz), 6.89 7.55 (2H, m), 7.79 (1H, d, J = 8.4 Hz), 7.91 (1H, d, J = 6.0 Hz), 10.41 (1H, s)

Presence or Absence of Crystallization of Each Product]

 The monomaleate (2a) obtained in Example 1-3 and the comparative compound obtained in Comparative Examples 1 to 3 (the dimaleate of the title compound (1a) , Monofumarate, disulfate) were obtained as crystals as described above. On the other hand, salts of hydrochloric acid, boric acid, phosphoric acid and citric acid were prepared as a comparative example using the title compound (1a) in the same manner as in Comparative Example 2, and crystallization of each compound was attempted. Upon crystallization of each product, methanol or ethanol was used as a crystallization solvent. The results are shown in Table 2.

[Table 2]

 Crystallization studies gave crystalline salts for sulfuric acid, hydrochloric acid, maleic acid and fumaric acid. On the other hand, the borate, phosphate and citrate of the title compound (1a) did not crystallize, the monoborate was an oily substance and the monophosphate and the monocitrate were amorphous. For the maleate, hydrochloride and sulfate of the title compound (1a), a double salt was obtained in addition to the 1-fold salt. The hydrochloride salt of the title compound (1a) showed clear deliquescence for both monohydrochloride salt and dihydrochloride salt.
[Comparison of Purification Efficiency of Monomeric Acid Salt and Dimaleate Salt of
Title Compound (1a) ] Monomaleate and dimaleate of the title compound (1a) were synthesized under the same conditions using the same means to give crystals Was obtained. Means of synthesis of each product is shown below.
(A) Synthesis of
Monomeric Salt of Title Compound (1a) 7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4- -1,4-benzothiazin-3-one (1a) (8.26 g, 16.2 mmol) was added to 71.74 g of ethanol and heated to 60 ° C., 1.79 g (15.40 mmol) of maleic acid was added , Cooled to 50 ° C. and 40 mg (0.064 mmol) of seed crystals was added. The reaction solution was stirred at 50 ° C. for 1 hour and then stirred overnight at room temperature. Subsequently, the reaction solution was stirred at 3 ° C. or less for 5 hours. After completion of the stirring, the precipitated crystals were collected by filtration to obtain 6.26 g (yield 62%) of the monomaleic acid salt of the title compound (1a).
(B) Synthesis of Dimaleate of Title Compound (1a)
7- [3- {4- (N-ethoxyethylbenzimidazol-2-ylmethyl) -1-piperazinyl} propoxy] -3,4- dihydro-2H- 8.26 g (16.2 mmol) of 1,4-benzothiazin-3-one (1a) was added to 71.74 g of ethanol and heated to 60 ° C., and 4.7 g (40.48 mmol) of maleic acid was added. After confirming that the maleic acid was completely dissolved in the solution, it was stirred overnight at room temperature. Subsequently, the reaction solution was stirred at 3 ° C. or less for 5 hours. After completion of the stirring, the precipitated crystals were collected by filtration to obtain 8.04 g (yield 67%) of the dimaleic acid salt of the title compound (1a).

[0114]
 Crystals of the monomaleate and dimaleate obtained by means (a) and (b) above were each dissolved in a small amount of solvent and the purity of each substance was measured by high performance liquid chromatography (HPLC). The HPLC conditions are as follows and charts showing the HPLC measurement results are shown in FIGS. 4 and 5. Table 3 summarizes the HPLC measurement results.
 Column: A stainless steel tube having an inner diameter of 4.6 mm and a length of 5 cm was
charged
with 3.5 μm of phenylhexylsilylated silica gel for liquid chromatography (HPLC) .
(  B%) 20% → <10 minutes> → 60% (10 minutes) → <10 minutes>
Column temperature: constant temperature around 40 ° C.
Gradient condition (B%) 20% → 85% (10 min)
A solution: 0.01 mol / L phosphate buffer, pH 6.0
B: methanol
flow rate: 1.0 mL / min
area measurement range: 40 minutes
injection volume: 3 [mu] L
sample concentration: 1 mg / mL

PATENT

 JP 2013035773

JP 2013049632

1.(a) Fukuda, T.Koketsu, A.Kaneko, Y.Ashikawa, Y. Monomaleate of Benzothiazine CompoundWO20111151732011.

(b) Fukuda, T.Koketsu, A. Method for Producing Benzothiazine CompoundWO20111151502011.
(b) Fukuda, T.Koketsu, A. Method for Producing Benzothiazine CompoundWO20111151502011.

//////////K-8986, K 8986, 

O=C(O)/C=C\C(=O)O.CCOCCn4c5ccccc5nc4CN1CCN(CC1)CCCOc2ccc3NC(=O)CSc3c2

SRT 1720

$
0
0

img

SRT-1720 diHCl

CAY10559

CAS: 1001645-58-4 (di HCl) , 925434-55-5 (free base)   1001645-58-4 (HCl)
Chemical Formula: C25H25Cl2N7OS
Molecular Weight: 542.483
Elemental Analysis: C, 55.35; H, 4.65; Cl, 13.07; N, 18.07; O, 2.95; S, 5.91

SRT-1720 HCl, SRT-1720 hudrochloride; SRT1720; SRT-1720; SRT 1720; CAY10559; CAY-10559; CAY 10559; SIRT-1933; SIRT 1933; SIRT1933.

 N-(2-(3-(piperazin-1-ylmethyl)imidazo[2,1-b]thiazol-6-yl)phenyl)quinoxaline-2-carboxamide dihydrochloride

SRT1720.svg

  • Molecular FormulaC25H23N7OS
  • Average mass469.561 Da

SRT-1720, also known as CAY10559 and is a drug developed by Sirtris Pharmaceuticals intended as a small-molecule activator of the sirtuin subtype SIRT1. It has similar activity in the body to the known SIRT1 activator resveratrol, but is 1000x more potent. In animal studies it was found to improve insulin sensitivity and lower plasma glucose levels in fat, muscle and liver tissue, and increased mitochondrial and metabolic function. A study of SRT1720 conducted by the National Institute on Aging found that the drug may extend the lifespan of obese mice by 44% .

SRT1720 is an experimental drug that was studied by Sirtris Pharmaceuticals intended as a small-molecule activator of the sirtuinsubtype SIRT1. The compound has been studied in animals, but safety and efficacy in humans have not been established.

Animal research

In animal models of obesity and diabetes SRT1720 was found to improve insulin sensitivity and lower plasma glucose levels in fat, muscle and liver tissue, and increase mitochondrial and metabolic function.[1] In mice rendered obese and diabetic by feeding a high-fat, high-sugar diet, a study performed at the National Institute of Aging found that feeding chow infused with the highest dose of SRT1720 beginning at one year of age increased mean lifespan by 18%, and maximum lifespan by 5%, as compared to other short-lived obese, diabetic mice; however, treated animals still lived substantially shorter lives than normal-weight mice fed normal chow with no drug.[2] In a later study, SRT1720 increased mean lifespan of obese, diabetic mice by 21.7%, similar to the earlier study, but there was no effect on maximum lifespan in this study.[3] In normal-weight mice fed a standard rodent diet, SRT1720 increased mean lifespan by just 8.8%, and again had no effect on maximum lifespan.[3]

Since the discovery of SRT1720, the claim that this compound is a SIRT1 activator has been questioned[4][5][6] and further defended.[7][8]

Although SRT1720 is not currently undergoing clinical development, a related compound, SRT2104, is currently in clinical development for metabolic diseases.[9]

PAPER

Letters in Drug Design & Discovery, 10(9), 793-797; 2013

The Identification of the SIRT1 Activator SRT2104 as a Clinical Candidate

Author(s): Pui Yee Ng, Jean E. Bemis, Jeremy S. Disch, Chi B. Vu, Christopher J. Oalmann, Amy V. Lynch,David P. Carney, Thomas V. Riera, Jeffrey Song, Jesse J. Smith, Siva Lavu, Angela Tornblom, Meghan Duncan, Marie Yeager, Kristina Kriksciukaite, Akanksha Gupta, Vipin Suri, Peter J. Elliot, Jill C. Milne, Joseph J. Nunes, Michael R. Jirousek, George P. Vlasuk, James L. Ellis, Robert B. Perni.

Journal Name: Letters in Drug Design & Discovery

Volume 10 , Issue 9 , 2013

Paper

Milne, J.C.; Lambert, P.D.; Schenk, S.; Carney, D.P.; Smith, J.J.; Gagne, D.J.; Jin, L.; Boss, O.; Perni, R.B.; Vu, C.B.; Bemis, J.E.; Xie, R.; Disch, J.S.; Ng, P.Y.; Nunes, J.J.; Lynch, A.V.; Yang, H.; Galonek, H.; Israelian, K.; Choy, W.; Iffland, A.; Lavu, S.; Medvedik, O.; Sinclair, D.A.; Olefsky, J.M.; Jirousek, M.R.; Elliott, P.J.; Westphal, C.H.
Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes
Nature 2007, 450(7170): 712

PATENT

WO 2007019417

WO 2007019416

WO 2007019345

WO 2007019344

WO 2007019346

WO 2008115518

PAPER

Vu, Chi B.; Journal of Medicinal Chemistry 2009, VOL 52(5), PG 1275-1283 

https://pubs.acs.org/doi/abs/10.1021/jm8012954

Abstract Image

A series of imidazo[1,2-b]thiazole derivatives is shown to activate the NAD+-dependent deacetylase SIRT1, a potential new therapeutic target to treat various metabolic disorders. This series of compounds was derived from a high throughput screening hit bearing an oxazolopyridine core. Water-solubilizing groups could be installed conveniently at either the C-2 or C-3 position of the imidazo[1,2-b]thiazole ring. The SIRT1 enzyme activity could be adjusted by modifying the amide portion of these imidazo[1,2-b]thiazole derivatives. The most potent analogue within this series, namely, compound 29, has demonstrated oral antidiabetic activity in the ob/ob mouse model, the diet-induced obesity (DIO) mouse model, and the Zucker fa/fa rat model.

Discovery of Imidazo[1,2-b]thiazole Derivatives as Novel SIRT1 Activators

Sirtris Pharmaceuticals, 200 Technology Square, Cambridge, Massachusetts 02139
J. Med. Chem.200952 (5), pp 1275–1283
DOI: 10.1021/jm8012954

* To whom correspondence should be addressed. Phone: (617)-252-6920, extension 2129. Fax: (617)-252-6924. E-mail: cvu@sirtrispharma.com., †

Present address: Department of Medicine, Division of Endocrinology and Metabolism, University of California—San Diego, 9500 Gilman Drive, La Jolla, CA 92093.

Preparation of N-(2-(3-(Piperazin-1-ylmethyl)imidazo[2,1-b]thiazol-6-yl)phenyl)quinoxaline-2-carboxamide (29)

Essentially the same procedure as detailed in the preparation of 3,4,5-trimethoxy-N-(2-(3-(piperazin-1-ylmethyl)imidazo[2,1-b]thiazol-6-yl)phenyl)benzamide was employed except that 2-quinoxaloyl chloride was used.
Mp: dec (HCl salt), 221.4 °C (freebase).
 1H NMR (300 MHz, DMSO-d6) δ 9.60 (br s, 1 H), 8.88 (d, 1 H, J = 8 Hz), 8.60 (br s, 1 H), 8.50 (s, 1 H), 8.0−8.30 (m, 5 H), 7.78 (d, 1 H, J = 8 Hz), 7.10−7.33 (m, 4 H), 3.90 (br s, 2 H), 3.00−3.10 (m, 4H), 2.60−2.80 (m, 4 H).
13C NMR (100 MHz, DMSO-d6): δ 47.49, 49.88, 111.45, 120.47, 121.84, 124.02, 127.04, 128.10, 129.20, 129.23, 131.39, 132.15, 135.39, 139.54, 143.03, 143.80, 144.36, 144.62, 147.76, 161.57.
High resolution MS, calcd for C25H23N7OS [M + H]+ 470.1763; found, 470.1753.

References

  1. ^ Milne JC; Lambert PD; Schenk S; Carney DP; Smith JJ; Gagne DJ; Jin L; Boss O; Perni RB; Vu CB; Bemis JE; Xie R; Disch JS; Ng PY; Nunes JJ; Lynch AV; Yang H; Galonek H; Israelian K; Choy W; Iffland A; Lavu S; Medvedik O; Sinclair DA; Olefsky JM; Jirousek MR; Elliott PJ; Westphal CH (November 2007). “Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes”Nature450(7170): 712–6. doi:10.1038/nature06261PMC 2753457PMID 18046409.
  2. ^ Minor RK; Baur JA; Gomes AP; Ward TM; Csiszar A; Mercken EM; Abdelmohsen K; Shin YK; Canto C; Scheibye-Knudsen M; Krawczyk M; Irusta PM; Martín-Montalvo A; Hubbard BP; Zhang Y; Lehrmann E; White AA; Price NL; Swindell WR; Pearson KJ; Becker KG; Bohr VA; Gorospe M; Egan JM; Talan MI; Auwerx J; Westphal CH; Ellis JL; Ungvari Z; Vlasuk GP; Elliott PJ; Sinclair DA; de Cabo R (Aug 2011). “SRT1720 improves survival and healthspan of obese mice”Scientific Reports1 (70): 70. doi:10.1038/srep00070PMC 3216557PMID 22355589. Retrieved 1 March 2014.
  3. Jump up to:a b Mitchell SJ; Martin-Montalvo A; Mercken EM; et al. (Feb 2014). “The SIRT1 Activator SRT1720 Extends Lifespan and Improves Health of Mice Fed a Standard Diet”Cell Reports6 (4): 836–43. doi:10.1016/j.celrep.2014.01.031PMC 4010117PMID 24582957. Retrieved 1 March 2014.
  4. ^ Pacholec M; Chrunyk BA; Cunningham D; Flynn D; Griffith DA; Griffor M; Loulakis P; Pabst B; Qiu X; Stockman B; Thanabal V; Varghese A; Ward J; Withka J; Ahn K (January 2010). “SRT1720, SRT2183, SRT1460, and resveratrol are not direct activators of SIRT1”J Biol Chem285 (11): 8340–8351. doi:10.1074/jbc.M109.088682PMC 2832984PMID 20061378.
  5. ^ Beher D; Wu J; Cumine S; Kim KW; Lu SC; Atangan L; Wang M (December 2009). “Resveratrol is not a direct activator of SIRT1 enzyme activity”. Chem Biol Drug Des74 (6): 619–24. doi:10.1111/j.1747-0285.2009.00901.xPMID 19843076.
  6. ^ Zarse, K.; Schmeisser, S.; Birringer, M.; Falk, E.; Schmoll, D.; Ristow, M. (2010). “Differential Effects of Resveratrol and SRT1720 on Lifespan of AdultCaenorhabditis elegans”. Hormone and Metabolic Research42 (12): 837–839. doi:10.1055/s-0030-1265225PMID 20925017.
  7. ^ Callaway E (2010-08-16). “GlaxoSmithKline strikes back over anti-ageing pills: Drugs do work as thought, says pharmaceutical giant”Naturedoi:10.1038/news.2010.412.
  8. ^ Dai H; Kustigian L; Carney D; Case A; Considine T; Hubbard BP; Perni RB; Riera TV; Szczepankiewicz B; Vlasuk GP; Stein RL (August 2010). “SIRT1 activation by small molecules – kinetic and biophysical evidence for direct interaction of enzyme and activator”J Biol Chem285 (43): 32695–32703. doi:10.1074/jbc.M110.133892PMC 2963390PMID 20702418.
  9. ^ “Sirtuin Pipeline”Sirtris Pharmaceuticals.
SRT1720
SRT1720.svg
Identifiers
PubChem CID
IUPHAR/BPS
ChemSpider
CompTox Dashboard(EPA)
Chemical and physical data
Formula C25H23N7OS
Molar mass 469.560 g/mol g·mol−1
3D model (JSmol)

////////////SRT-1720 DI HCl, obesity, diabetes, SRT 1720,  Sirtris Pharmaceuticals,  CAY10559,  CAY 10559, Preclinical

O=C(NC1=CC=CC=C1C2=CN3C(SC=C3CN4CCNCC4)=N2)C5=NC6=CC=CC=C6N=C5.[H]Cl.[H]Cl


BI-882370

$
0
0

img

AEJACXAFHXBVHF-UHFFFAOYSA-N.png

BI-882370

XP-102

N-(3-(5-((1-ethylpiperidin-4-yl)(methyl)amino)-3-(pyrimidin-5-yl)-1H-pyrrolo[3,2-b]pyridin-1-yl)-2,4-difluorophenyl)propane-1-sulfonamide

CAS 1392429-79-6
Chemical Formula: C28H33F2N7O2S
Molecular Weight: 569.68
Elemental Analysis: C, 59.03; H, 5.84; F, 6.67; N, 17.21; O, 5.62; S, 5.63

N-(3-(5-((1-ethylpiperidin-4-yl)(methyl)amino)-3-(pyrimidin-5-yl)-1H-pyrrolo[3,2-b]pyridin-1-yl)-2,4-difluorophenyl)propane-1-sulfonamide

N-(3-{5-[(1-Ethylpiperidin-4-Yl)(Methyl)amino]-3-(Pyrimidin-5-Yl)-1h-Pyrrolo[3,2-B]pyridin-1-Yl}-2,4-Difluorophenyl)propane-1-Sulfonamide

N-[3-[5-[(1-ethylpiperidin-4-yl)-methylamino]-3-pyrimidin-5-ylpyrrolo[3,2-b]pyridin-1-yl]-2,4-difluorophenyl]propane-1-sulfonamide

BI 882370 is a highly potent and selective RAF inhibitor that binds to the DFG-out (inactive) conformation of the BRAF kinase. BI 882370 inhibits proliferation of human BRAF-mutant melanoma cells with 100× higher potency (1-10 nmol/L) than vemurafenib.

Xynomic, under license from Boehringer Ingelheim , is investigating for treating BRAF mutant cancers, including colorectal cancer and melanoma; in October 2017, preclinical data were reported in the melanoma and colorectal cancer settings.

  • Originator Boehringer Ingelheim
  • Developer Boehringer Ingelheim; Xynomic Pharmaceuticals
  • Class Antineoplastics; Piperidines; Pyridines; Pyrimidines; Pyrroles; Small molecules
  • Mechanism of Action Proto oncogene protein b raf inhibitors
  • Preclinical Colorectal cancer; Malignant melanoma
  • 20 Dec 2018 Xynomic Pharma plans a phase Ib trial for Colorectal cancer (in combination with BI 860585) in third quarter of 2019
  • 01 Jun 2018 Xynomic Pharmaceuticals plans a phase I trial for Colorectal cancer and Malignant melanoma in 2018 or 2019
  • 06 Nov 2017 Chemical structure information added
  • US8889684

PATENT

WO2012104388

PATENT

WO-2019084459

Novel crystalline salts (monosuccinate salt), designated as Form A, of BI-882370 and their substantially anhydrous and non-solvated, processes for their preparation and compositions comprising them. Also claimed are their use as a RAF kinase Inhibitor, for the treatment of cancers and other diseases, such as infections, inflammations and autoimmune diseases.

The compound N-(3-(5-((l -ethylpiperidin-4-yl)(methyl)andno)-3-(pyrimidin-5-yl)-lH-pyrrolo [3, 2-Z>]pyri din- l-yl)-2,4-difluorophenyl)propane-l -sulfonamide (BI 882370), having Formula I:

I

is a RAF kinase inhibitor useful in the treatment of various diseases including cancer. The compound of Formula I, as well as its preparation and use, have been described in

WO/2012/104388, which is incorporated herein by reference in its entirety.

The RAS-RAF-MAPK (mitogen-activated protein kinase) signaling pathway plays a critical role in transmitting proliferation signals generated by the cell surface receptors and cytoplasmic signaling elements to the nucleus. Constitutive activation of this pathway is involved in malignant transformation by several oncogenes. Activating mutations in RAS

occur in approximately 15 % of cancers, and recent data has shown that B-RAF is mutated in about 7% of cancers (Wellbrock et al, “The RAF proteins take centre stage”, Nature Rev. Mol. Cell Biol., 2004, 5, 875-885), identifying it as another important oncogene in this pathway. In mammals, the RAF family of serine/threonine kinases comprises three members: A-RAF, B-RAF and C-RAF. However, activating mutations have so far been only identified in B-RAF underlining the importance of this isoform. It is believed that B-RAF is the main isoform that couples RAS to MEK, and that C-RAF and A-RAF signal to ERK only to fine-tune cellular responses (Wellbrock et al. Nature Rev. Mol. Cell Biol, 2004, 5, 875-885). The most common cancer mutation in B-RAF results in a valine to glutamic acid exchange at position 600 of the protein (V600E), which dramatically enhances B-RAF activity, presumably because its negative charge mimics activation loop phosphorylation (Wan et al , “Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF”, Cell, 2004, 116, 855-867). The highest incidence of B-RAF V600 mutations occurs in malignant melanoma (39%), thyroid cancer (46%), colorectal cancer (10%), biliary tract cancer (10%), prostate cancer (4%), ovary cancer (3%) and non-small cell lung cancer (2%), but they also occur at a low frequency in a wide variety of other cancers (frequencies of mutations according to COSMIC (Catalogue Of Somatic Mutations In Cancer; Wellcome Trust Sanger Institute) release v.53, 15th May 2011 ;

http://www.sanger.ac.uk/genetics/CGP/cosmic/). Literature supported the hypothesis that B-RA 600E mutated tumor cells seem to rely heavily on the continued activation of this pathway – a phenomenon termed “oncogene addiction” – whereas normal B-RAFwt cells use a broader range of signals. This provides an Achilles’ heel that can be exploited

therapeutically by treating patients with somatically mutated B-RAFV600E using orally available B-RAF inhibitors.

The key role of B-RAF V600E in aberrant ERK signaling and consequently oncogenesis has been demonstrated in several independent experimental approaches such as

overexpression of oncogenic/mutated B-RAF in vitro and in vivo (Wan et al., Cell, 2004, 116, 855-867; Wellbrock et al, Cancer Res. 2004, 64: 2338-2342), siRNA knock-down in vitro (Karasarides et al., Oncogene, “V599EB-RAF is an oncogene in melanocytes”, 2004, 23, 6292-6298) or in inducible short-hairpin RNA xenograft models where gain-of-function B-RAF signaling was found to be strongly associated with in vivo tumorigenicity (Hoeflich et al, “Oncogenic BRAF is required for tumor growth and maintenance in melanoma models”, Cancer Res., 2006, 66, 999-1006).

Treatment of B-RAFV600E mutated melanoma or colon carcinoma cells induces a B-RAF inhibition phenotype (e.g. reduction of phospho-MEK and phospho-ERK levels, reduction of cyclin D expression and induction of p27 expression). Consequently, these cells are locked in the Gl -phase of the cell cycle and do not proliferate.

Clinical proof of mechanism and proof of concept has been established for treating in cancer in B-RAFV600E mutated melanoma patients treated with Zelboraf®, B-RAF inhibitor (PLX-4032, vemurafenib, from Plexxikon/Daiichi Sankyo/Roche. Bollag et al., “Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma”, Nature, 2010, 467(7315), 596-9.; Flaherty et al, New Engl. J. Med., “Inhibition of Mutated, Activated BRAF in Metastatic Melanoma”, 2010, 363, 809-819; Chapman et al. “Improved Survival with Vemurafenib in Melanoma with BRAF V600E Mutation”, New Engl. J. Med, 2011, 364:2507-2516. Favorable response rates were observed in both Phase I and Phase III clinical trials. It was reported, that melanoma patients carrying a B-RAFV600K mutation also do respond to therapy (Rubinstein et al, “Incidence of the V600K mutation among melanoma patients with BRAF mutations, and potential therapeutic response to the specific BRAF inhibitor PLX4032”, J. Transl. Med , 2010, 8, 67).

The most frequent B-RAF mutation is the exchange at amino acid position 600 from valine to glutamate with more than 90% frequency of all B-RAF mutations (Wellbrock et al. Nature Rev. Mol. Cell Biol, 2004, 5, 875-885), the second most frequent mutation is an alteration from valine to lysine, other mutations were found with lower frequency at that position (Wellbrock et al. Nature Rev. Mol. Cell Biol, 2004, 5, 875-885 and frequencies of mutations according to COSMIC (Catalogue Of Somatic Mutations In Cancer; Wellcome Trust Sanger Institute) release v53, 15th May 2011 ;

http://www.sanger.ac.uk/genetics/CGP/cosmic/). Additional mutations were found at e.g. the glycine rich loop (Wellbrock et al. Nature Rev. Mol. Cell Biol, 2004, 5, 875-885). Not all of these rather rare mutations seem to lead to direct activation of B-RAF (Wan et al. ,

“Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF”, Cell, 2004, 116, 855-867).

The compound of Formula I is a highly potent and selective RAF inhibitor that binds to the DFG-out (inactive) conformation of the B-RAF kinase. The compound inhibited proliferation of human B-RAF-mutant melanoma cells with 100 times higher potency (1-10 nmol/L) than vemurafenib, whereas wild-type cells were not affected at 1,000 nmol/L. A solution of the compound administered orally was efficacious in mouse models of B-RAF-mutant melanomas and colorectal carcinomas, and at 25 mg/kg twice daily showed superior efficacy compared with vemurafenib, dabrafenib, or trametinib. The compound was also active in A375 melanoma-bearing mice that were resistant to vemurafenib, particularly when dosed in combination with trametinib. Mice treated with the compound did not show any body weight loss or clinical signs of intolerability, and no pathologic changes were observed in several major organs investigated, including skin. Furthermore, in a pilot study in rats (up to 60 mg/kg daily for 2 weeks), the compound lacked toxicity in terms of clinical chemistry, hematology, pathology, and toxicogenomics. These results are described in Waizenegger et al., Mol. Cancer Ther., 2016, 75(3); 354-65, which is incorporated herein by reference in its entirety.

For the manufacture, purification, and formulation of a drug, it may be advantageous to employ a form of the drug having superior stability or other desirable formulation property exhibited by, for example, one or more salt or crystalline forms of the drug. Formation of salts of basic or acidic drugs can sometimes provide forms of the drug that have

advantageous properties such as solubility, non-hygroscopicity, crystallinity, and other physical properties that advantageous for formulating the drug. On the other hand, discovering a suitable salt or other crystalline form that is suitable for formulation is difficult, since there are numerous variables in the formation of a salt or crystalline form. These include the existence of numerous possible acids and bases that might be used as a counter-ion, various stoichiometric ratios that may be possible for combining a given basic or acid drug with an acid or base counter-ion, a wide variety of solvents and solvent systems

(including combinations of solvents) that potentially can be used to attempt to form salts or crystalline forms, and a variety of conditions (such as temperature or heating or cooling conditions) under which salts or crystalline forms may be generated. All of these variables of which may affect the properties of the salts or crystalline forms that might be obtained. Salts or solid forms may also have a variety of properties that render them unsuitable for drug development and formulation such as lack of crystallinity (amorphous forms), the presence or formation of multiple crystalline forms, which may interconvert and/or have different properties (polymorphism), lack of aqueous solubility, hygroscopicity, or stickiness of the solid. Furthermore, the formation of salts and crystalline forms and their properties are generally very unpredictable.

Accordingly, the crystalline salt forms of the compound of Formula I provided herein help satisfy the ongoing need for the development of a RAF kinase inhibitor for the treatment of serious diseases.

Preparation of A^-(3-(5-((l-ethylpiperidin-4-yl)(methyl)amino)-3-(pyrimidin-5-yl)-lH-pyrrolo[3,2-Z>]pyridin-l- amide (BI 882370)

Step 1. 4-(6-Methyl-5-nitro-pyridin-2-yl)-piperazine-l-carboxylic acid tert-butyi ester

(3)

1 2 3

DIPEA (62.82 mL, 0.435 mol) is added to the solution of 6-chloro-3-nitro-2-methylpyridine (1) (50 g, 290 mmol) and N-Boc-piperazine (2) (53.95 g, 290 mmol) in dry MeCN (200 mL) and stirred for 4 h at 50 °C. After the reaction is finished the reaction mixture is diluted with MeCN and water and stirred for 30 min. The precipitated product is collected by filtration, washed with water and the solid is dried in vacuo.

Step 2. 4- [6-((£’)-2-Dimethylamino-vinyl)-5-nitro-pyridin-2-yl] -piperazine- 1-carboxylic acid

To a stirred solution of 4-(6-methyl-5-nitro-pyridin-2-yl)-piperazine- 1-carboxylic acid tert-butyl ester (3) (13 g, 40.3 mmol) in DMF (35 mL) is added N,N-dimethylformamide dimethylacetal (14.47 g, 121 mmol) and stirred in argon atmosphere for 36 h at 90 °C.

Additional 1.5 eq. of N^V-dimethylformamide dimethylacetal is added and stirred for 12 h at 90 °C. The reaction mixture is poured into water and extracted with DCM. The combined organic layers are washed with water, dried over anhydrous Na2S04 and concentrated in vacuo. The residue is used without further purification for the next step.

Step -(lH-pyrrolo[3,2-Z>]pyridin-5-yl)piperazine-l-carboxylic acid tert-butyl ester (5)

4 5

4-[6-((i?)-2-Dimethylairdno-vinyl)-5-nitro-pyridin-2-yl]-piperazine-l-carboxylic acid tert-butyl ester (36.4 g, 96 mmol) is taken up in MeOH, Pd/C (0.56 g, 10 %) is added and the mixture is hydrogenated in an autoclave at 60 psi for 16 h. The reaction mixture is filtered and concentrated under reduced pressure. The residue is purified by column chromatography viaNP MPLC. The product containing fractions of compound (5) (HPLC-MS method B: tRet. = 1.55 min.; MS (M+H)+ = 303) are combined and evaporated in vacuo.

Step 4. N- -Amino-2,6-difluorophenyl)acetamide (7)

6 7

Compound (6) (55.0 g, 254 mmol) is taken-up in MeOH (1.0 L). Pd/C (10.0 g, 10 %) is added and the mixture is hydrogenated in an autoclave at 200 psi for 3 h. The reaction mixture is filtered and concentrated under reduced pressure. The residue is purified by NP-MPLC on silica gel using DCM/MeOH (96:4) as eluent. The product containing fractions of the aniline intermediate (HPLC-MS method B: tRet. = 0.25 min.; MS (M-H) = 185) are combined and evaporated.

Step 5. N- -Difluoro-3-(propylsulfonamido)phenyl)acetamide (9)

To the aniline intermediate (35.0 g, 188 mmol) in DCM (100 mL) pyridine (6.6 mL, 75 mmol) and ^-propane sulfonyl chloride (8) (29.5 mL, 263 mmol) are added and the mixture is stirred at rt for 16 h. The reaction mixture is diluted with EtOAc (200 mL), washed with H2O and HC1 (aq., 1 N) and the layers are separated, dried over MgS04 and evaporated to yield the sulfonamide (9) which was used without further purification.

Step 6. N-

9 10

The sulfonylated aniline (9) (38.0 g, 130 mmol) is taken-up in EtOH (250 mL), H2O (200 mL) and concentrated hydrochloric acid (200 mL) and heated to 80 °C for 2 h. The reaction mixture is concentrated under reduced pressure, aqueous NaOH (4 N) is added until pH = 6 is reached and the mixture is extracted 2 x with DCM. The combined organic layer is washed with brine, dried over MgS04, filtered and evaporated to yield the deacylated aniline (10) (HPLC-MS method B: tRet. = 0.22 min.; MS (M-H) = 249) as a hydrochloride which was used without further purification.

Step 7. N-(2 -Difluoro-3-iodophenyl)propane-l-sulfonamide (11)

10 11

The hydrochloride of compound (10) is taken-up in DCM and extracted with NaHCCb solution. The organic layer is dried over MgSCn, filtered and evaporated. To the free base (10) (3.55 g, 14.21 mmol) in TFA (80 mL) at 0 °C is added NaNC (1.96 g, 28.4 mmol) in small portions and the mixture is stirred for 30 min. KI (23.83 g, 142 mmol) is added and stirring is continued for additional 15 min. The reaction mixture is diluted with Et^O and stirred for 1 h. Na2S203 solution (semiconc.) is added and the mixture is extracted 3 x with Et20. The combined organic layer is dried over MgSCn, filtered and concentrated in vacuo. The residue is purified by column chromatography via NP-MPLC. The product containing fractions of compound (11) (HPLC-MS method A: tRet. = 1.58 min.; MS (M-H) = 360) are combined and evaporated in vacuo.

Step 8. 4-((l-(2,6-Difluoro-3-(propylsulfonamido)phenyl)-lH-pyrrolo [3,2-b] pyridin-5-yl)

12

The lH-pyrrolo [3,2-*] pyridine (5) (10.0 g, 30.27 mmol), sulfonamide (11) (16.4 g,

45.4 mmol), Cul (576 mg, 3.03 mmol), ^^-(l ^^^-^N’-bismethyl-l^-cyclohexandiamine

(1.91 mL, 12.1 mmol) and CS2CO3 (29.6 g, 90.85 mmol) are taken-up in dry toluene (3 mL) and the resulting mixture is flushed with argon and stirred for 16 h at 120 °C. After the addition of further Cul (576 mg, 3.03 mmol), trans-(\R,2R)-N,N’-bismet y 1-1,2-cyclohexandiamine (1.91 mL, 12.1 mmol) and CS2CO3 (20.0 g, 60.0 mmol) the reaction mixture is stirred for further 24 h. The solvent is removed in vacuo, the residue is taken up in DCM and extracted with NaHCC solution (semiconc). The organic layer is dried over MgS04, filtered, the solvent is removed in vacuo and the residue is purified viaNP-MPLC. The product containing fractions of (12) (HPLC-MS method C: teet. = 1.62 mia; MS (M+H)+ = 564) are combined and the solvent is removed in vacuo.

Step 9. 4-((l-(2,6-Difluoro-3-(propylsulfonamido)phenyl)-3-iodo-lH-pyrrolo[3,2-b]pyridin-5 3)

To a solution of sulfonamide (12) (1.078 g, 1.9 mmol) in DMF (4 mL)/THF (100 μί) is added NIS (474 mg, 2.1 mmol) and the mixture is stirred for 1 h at rt. The reaction mixture is diluted with 30 mL DCM and extracted with NaHCCb solution (semiconc). The combined organic layer is dried over MgSCn, filtered and concentrated under reduced pressure. The residue is purified by column chromatography via RP HPLC. The product containing fractions of (13) (HPLC-MS method B: tRet. = 2.035 mia; MS (M+H)+ = 688) are freeze dried.

Step 10. 4-((l-(2,6-Difluoro-3-(propylsulfonamido)phenyl)-3-(pyrimidin-5-yl)-lH-pyrrolo[3,2-b]pyridin-5-yl)(methyl)amino)piperidine-l-carboxylic acid tert-butyi ester (15)

13 15

Sulfonamide (13) (770 mg, 1.12 mmol), pyrimidin-5-yl-boronic acid (14) (194 mg, 1.57 mmol), Pd(dppf)Cl2 (82 mg, 0.11 mmol), LiCl (142 mg, 3.35 mmol) and Na2C03 (294 mg, 2.8 mmol) are taken-up in dioxane/LhO (2: 1 mixture, 12 mL), and the resulting mixture is flushed with argon and stirred for 1 h at 100 °C. The reaction mixture is diluted with DCM and extracted with NaHCCb solution (semi-concentrated). The organic layer is dried over MgS04, filtered, Isolute® is added, the solvent is removed in vacuo and the residue is purified via RP HPLC. The product containing fractions of (15) (HPLC-MS method C: tRet. = 2.149 min.; MS (M+H)+ = 642) are freeze dried.

Step 11. N-(2,4-Difluoro-3-(5-(methyl(piperidin-4-yl)amino)-3-(pyrimidin-5-yl)- 1H-pyrrolo[3,2-b]pyridin-l-yl)phenyl)propane-l-sulfonamide

15 16

To a solution of example compound (15) (154 mg, 0.24 mmol) in DCM/MeOH (1 : 1, 4 mL) is added HC1 (in dioxane, 4 N, 2 mL) and the mixture is stirred for 3 h at rt. The solvent is removed in vacuo. Obtained compound (16) (HPLC-MS method B: tRet. = 1.02 min.; MS (M+H)+ = 542) is used without further purification.

Step 12. ^-(3-(5-((l-Ethylpiperidin-4-yl)(methyl)amino)-3-(pyrimidin-5-yl)-lH-pyrrolo [3,2-Z>] pyridin- l-yl)-2,4-diflu

Compound I was obtained from compound (16) by reductive alkylation with acetaldehyde (40% in iPrOH) in the presence of 1.5 eq. sodium acetoxyborohydride in iPrOH. The crude product was recrystallized from ethanol to obtain the title compound in 84% yield.

Scale-Up Synthesis of A/-(3-(5-((l-ethylpiperidin-4-yl)(methyl)amino)-3-(pyrimidin-5-yl)-lH-pyrrolo[3,2-Z>]pyridin-l-yl)-2,4-difluorophenyl)propane- 1-sulfonamide (BI 882370)

Step 1. N-(2,4-Difluoro-3-(5-(methyl(piperidin-4-yl)amino)-3-(pyrimidin-5-yl)-lH-pyrrolo[

15 16

Isopropanol (8.83 kg) and compound (15) (1.80 kg, 2.8 mol) were added into a reactor, and the mixture was stirred and heated to 55-60 °C. Concentrated hydrochloric acid (2.76 kg, 28 mol) was dropped into the reactor over than 20 min. at 60-65 °C. Then, the reaction mass was heated to 60-70 °C and held for 1 h. The conversion was monitored by HPLC, and reached about 99.5% after about 1 h.

The reaction mass was cooled and the isopropanol was removed by distillation under reduced pressure at not more than 50 °C. A brown oil was obtained, dissolved into water (6.75 kg) and washed by extraction with ethyl acetate (2.02 kg) at 20-30 °C. The water-phase was cooled to 15-20 °C. The pH was adjusted to 8.0-8.5 with 10% aqueous NaOH solution (-8.0 kg) at 20-30°C. The mixture was stirred for 3-4h at 20-30°C with the pH adjusted to 8.0-8.5 by addition of 10% NaOH solution every half-hour. The product was isolated by filtration and the cake washed with water (3.6 kg). The solid was dried under vacuum at 45-50 until the water content was not more than 5.5%. This provided about 1.64 kg of crude compound (16) (yield 108% of theoretical; the crude product containing water and NaCl detected). The crude product was used directly).

Step 12. ^-(3-(5-((l-Ethylpiperidin-4-yl)(methyl)amino)-3-(pyrimidin-5-yl)-lH-pyrr -Z>] pyridin- l-yl)-2,4-difluorophenyl)propane- 1-sulfonamide (I)

Bl 878426 Bl 882370 

Process:

Dichloromethane (19.88 kg) and compound (16) (1.5kg, 2.77mol) were added into a reactor, and the mixture was stirred and cooled to 0-10°C under a nitrogen atmosphere. Sodium triacetoxyborohydride (95%, 0.93 kg, 4.16 mol) was added into the mixture at 0-10°C. The mixture was stirred for 20-30 min. at 0- 10°C. Acetaldehyde in DCM (40%,

1.07 kg, 9.71 mol) added into the mixture slowly over 2 h at 0-10 °C. The reaction mixture was stirred at 0-10 °C under a nitrogen atmosphere for 0.5-lh. The conversion was monitored by HPLC, and reached about 99.5% after about 0.5-1 h.

Water (15 kg) was added into the reaction mass at a temperature below 15 °C. The mixture was stirred at 15-30 °C for 20-30 min. Aqueous ammonia (25%, 1.13 kg, 16.61 mol) was added into the mixture and the mixture was then stirred for 0.5 h. The organic phase was separated and then washed by extraction with water (15 kg) at 20-25 °C. Activated charcoal (0.15 kg) was added into the organic phase. The mixture was stirred for 1 h and then filtered. The filtrate was concentrated under reduced pressure at not more than 40°C, and compound (I) (1.58 kg, 100% yield) was obtained as a foamy solid.

Investigation of the Crystallinity of iV-(3-(5-((l-Ethylpiperidin-4-yl)(methyl)amino)-3-(pyrimidin-5-yl)- lH-pyrrolo [3,2-Z>] pyridin- l-yl)-2,4-difluorophenyl)propane- 1-sulfonamide Free Base

Investigation of the crystallinity of N-(3-(5-((l-ethylpiperidin-4-yl)(methyl)amino)-3-(py rimidin-5-y 1)- lH-pyrrolo[3 ,2-b] pyridin- 1 -y l)-2,4-difluoropheny l)propane- 1 -sulfonamide free base, obtained by recrystallization from aqueous ethanol, which was used as a starting material to investigate salt formation showed that the compound had low crystallinity, as seen in FIG. 1.

Investigation of Salt forms of iV-(3-(5-((l-Ethylpiperidin-4-yl)(methyl)amino)-3-(pyrimidin-5-yl)- lH-pyrrolo [3,2-Z>] pyridin- l-yl)-2,4-difluorophenyl)propane- 1-sulfonamide

The compound N-(3-(5-((l-ethylpiperidin-4-yl)(methyl)andno)-3-(pyrimidin-5-yl)-lH-pyrrolo [3 ,2-Z>]pyri din- l-yl)-2,4-difluorophenyl)propane-l -sulfonamide was combined with various acids in various solvent systems.

A 96-well master plate was charged by dosing compound in MeOH (stock solution) with a concentration of approx. 40 mg/mL. This plate was placed in a vacuum oven for liquid removal to obtain the same amount of solid material in each well. Subsequently different solvents/solvent mixtures and the acids were added to the solid material in each well (approx. 500μί) and the whole plate was heated up to 50 °C for 2 hours while stirring (using a small stirring bar added to each well).

The acids used were as shown in Table 1. The solvents used were as shown in Table 2. Crystallinity of salts obtained either by the slurry experiment or crystallization by evaporation.

To investigate crystal formation by a slurry experiment, the plate was allowed to cool and the crystallinity of the resulting salts was investigated by XRPD. An image of the master plate showing the salts obtained is shown in FIG. 2A and images of XRPD performed on the salt from each of the master plate wells, showing the crystallinity of the salts formed, is shown in FIG. 2B.

To investigate crystal formation by an evaporation experiment, after the heating period, the solutions were filtered at the same temperature (50 °C) using a preheated filter plate to ensure that no non-dissolved material can be transferred into the other crystallization plates. The filtrate was dispensed into an evaporation plate (approx.. 200μί). The solvents were allowed to evaporate, and the crystallinity of the resulting salts was investigated by XRPD. An image of the master plate showing the salts obtained is shown in FIG. 3A and images of XRPD performed on the salt from each of the evaporation plate wells, showing the crystallinity of the salts formed, is shown in FIG. 3B.

Table 1. Salts Used for Salt Form Investigation

Table 2. Solvents Used for Salt Form Investigation

REFERENCES

1: Waizenegger IC, Baum A, Steurer S, Stadtmüller H, Bader G, Schaaf O, Garin-Chesa P, Schlattl A, Schweifer N, Haslinger C, Colbatzky F, Mousa S, Kalkuhl A, Kraut N, Adolf GR. A Novel RAF Kinase Inhibitor with DFG-Out-Binding Mode: High Efficacy in BRAF-Mutant Tumor Xenograft Models in the Absence of Normal Tissue Hyperproliferation. Mol Cancer Ther. 2016 Mar;15(3):354-65. doi: 10.1158/1535-7163.MCT-15-0617. Epub 2016 Feb 25. PubMed PMID: 26916115.

/////////////// BI-882370,  BI 882370,  BI882370, XP-102, Boehringer Ingelheim, Xynomic Pharmaceuticals, Preclinical,  Colorectal cancer, Malignant melanoma

CCN1CCC(CC1)N(C)c3ccc4n(cc(c2cncnc2)c4n3)c5c(F)ccc(NS(=O)(=O)CCC)c5F

GFH 018

$
0
0

(E)-3-[6-[2-(6-Methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl]-[1,2,4]triazolo[1,5-a]pyridin-5-yl]prop-2-enamide.png

GFH-018

CAS 2169299-67-4

C21 H19 N7 O, 385.42
(E)-3-[6-[2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl]-[1,2,4]triazolo[1,5-a]pyridin-5-yl]prop-2-enamide
2-Propenamide, 3-[6-[5,6-dihydro-2-(6-methyl-2-pyridinyl)-4H-pyrrolo[1,2-b]pyrazol-3-yl][1,2,4]triazolo[1,5-a]pyridin-5-yl]-, (2E)-

GenFleet Therapeutics

Advanced solid tumor; Cancer

TGF-beta Receptor Type-1 (TGFBR1; ALK5; SKR4; TbetaR-I) Inhibitors

Signal Transduction Modulators

GFH-018 , a TGFBR1 inhibitor, being investigated by GenFleet as an oral tablet formulation, for the treatment of cancer, including advanced solid tumors and hepatocellular carcinoma,  in March 2019, the company was developing GFH-018 as a class 1 chemical drug in China, with a clinical trial expected to begin in the second half of 2019.

Transforming growth factor-β (TGF-β) is a multifunctional growth factor superfamily with extensive biological activity, involved in early embryonic development, cartilage and bone formation, extracellular matrix synthesis, inflammation, Interstitial fibrosis, regulation of immune and endocrine functions, tumor formation and development.
The TGF-β superfamily consists of a class of structural and functionally related polypeptide growth factors, including TGF-βs (ie, narrowly defined TGF-β), activins (axivins), inhibins, and bone morphogenetic proteins (BMPs). Müllerian inhibitors (mullerian), etc., TGF-β is one of the important members of this family. In mammals, TGF-β mainly exists in three forms of TGF-β1, TGF-β2 and TGF-β3, which are located on different chromosomes, and TGF-β1 accounts for the highest proportion (>90%) in somatic cells. It has the strongest activity, the most functions, and the widest distribution. The newly synthesized TGF-β appears as an inactive precursor consisting of a signal peptide, a latent-associated polypeptide (LAP) and a mature TGF-β. After enzymatic hydrolysis, it forms active TGF-β, and then Receptor binding exerts a biological effect.
TGF-[beta] signaling molecules signal through a transmembrane receptor complex. TGF-β receptor is a transmembrane protein present on the cell surface and is divided into type I receptor (TGF-βRI), type II receptor (TGF-βRII) and type III receptor (TGF-βRIII), of which TGF- βRI is also known as activin receptor-like kinase 5 (ALK5). TGF-βRIII lacks intrinsic activity and is primarily involved in the storage of TGF-β. TGF-βRI and TGF-βRII belong to the serine/threonine kinase family. Type II receptors bind to TGF-β ligands with higher affinity and form heterologous receptor complexes with type I receptors. Phosphorylation of a region rich in glycine and serine residues (GS domain) of the proximal membrane of the receptor initiates an intracellular signal cascade reaction.
Smads are important TGF-β signal transduction and regulatory molecules in cells, which can directly transduce TGF-β signaling from the cell membrane, such as the nucleus. TGF-β/Smads signaling pathway plays an important role in the occurrence and development of tumors. . In TGF-β/Smads signal transduction, activated TGF-β first binds to TGF-βRII on the cell membrane surface to form a heterodimeric complex, and TGF-βRI recognizes and binds to the binary complex.
TGF-βRII phosphorylates serine/threonine in the GS domain of the cytoplasmic domain of TGF-βRI, thereby activating TGF-βRI; activated TGF-βRI further phosphorylates R-Smads (Smad2/Smad3) protein, which in turn Co-Smad (Smad4) binds to a heterotrimeric complex that enters the nucleus and acts synergistically with other co-activators and co-inhibitors to regulate transcription of target genes. . Any change in any part of the TGF-β/Smads signaling pathway leads to abnormalities in the signal transduction pathway.
Current research indicates that in tumor cells, TGF-β can directly affect tumor growth (non-inherent effects of TGF-β signaling), or by inducing epithelial-mesenchymal transition, blocking anti-tumor immune responses, and increasing tumor-associated fibrosis And enhanced angiogenesis indirectly affects tumor growth (the intrinsic effect of TGF-β). At the same time, TGF-β has a strong fibrotic induction, which is an activator of tumor-associated fibroblasts. These fibroblasts are a major source of collagen type I and other fibrotic factors. Induction products of fibroblasts and other fibrotic factors may continue to develop a microenvironment that reduces immune responses, increases drug resistance, and enhances tumor angiogenesis. In addition, TGF-β affects blood vessels during individual development and tumor growth. Raw regeneration. For example, TGF-βRI-deficient mouse embryos show severe vascular development defects, demonstrating that the TGF-β signaling pathway is a key regulator in vascular endothelium and smooth muscle cell development.
In 2013, the FDA awarded Lilly’s small molecule TGF-βRI inhibitor LY2157299 (WO 2002/094833) for the treatment of glioma and liver cancer. LY2157299 is an orphan drug under research, named Galunisertib. Galunisertib inhibits tumor cell invasion and metastasis while inhibiting the infiltration of tumor cells into blood vessels. In the phase 2 clinical trial of patients with liver cancer, about 23% of patients treated with Galunisertib had a decrease in serum alpha-fetoprotein (AFP) levels of more than 20%. Compared with patients without AFP response, these patients had slower tumor progression and longer survival, and increased expression of cadherin in epithelial cells was also observed in these patients, suggesting that Galunisertib can be regulated by inhibiting TGF-β signaling pathway. EMT, thereby inhibiting the progression of liver cancer, the structure of Galunisertib (LY2157299) is shown in formula (II):
Background research and development materials refer to the following documents:
WO2009/009059; WO2007/076127; WO2004/026306; WO2004/072033; WO2002/094833.
Synthesis
WO2017215506

PATENT

WO2017215506

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017215506

Example 1
Preparation of intermediates 1-6:
Step A: Ethyl acetate (291.41 ml, 2.98 mol) was dissolved in toluene (750.00 ml), and then sodium ethoxide (135.06 g, 1.98 mol) was added portionwise at room temperature, and the mixture was stirred at room temperature for 1 hour. Methyl 6-methylpyridine-2-carboxylate (150.00 g, 992.33 mmol) was added to the above reaction solution at 25 ° C, then heated to 95 ° C and stirred for 15 hours. The reaction mixture was cooled to 30 ° C, the pH was adjusted to 7 with acetic acid, diluted with water (500 ml), and ethyl acetate (500 ml). The organic phase was dried with anhydrous sodium s The residue was purified with EtOAc EtOAc EtOAc (EtOAc:EtOAc Rate: 58.35%).
Step B: Ethyl 3-(6-methyl-2-pyridine)-3-oxo-propanoate (120.00 g, 579.07 mmol) was dissolved in pyridine (300 mL) then 1-aminopyrrolidine- 2-keto-p-toluenesulfonate (172.01 g, 631.66 mmol). The reaction mixture was stirred at 25 ° C for 16 hours and then concentrated under reduced pressure to remove solvent. The residue was diluted with water (300 ml) and then extracted with ethyl acetate (300 ml). The combined organic phases were dried with anhydrous sodium s , yield: 90.28%).
Step C: Dissolving 3-(6-methyl-2-pyridine)-3-(2-carbonyl-pyrrolidine)imino-propionic acid ethyl ester (155.00 g, 535.72 mmol) in toluene and then adding ethanol Sodium (72.91 g, 1.07 mol). The reaction mixture was heated to 100 ° C and stirred for 16 hours, then cooled to room temperature. It was slowly diluted with water (1.5 liters), adjusted to pH 4 with concentrated hydrochloric acid, and extracted with dichloromethane / isopropyl alcohol (10/1) (1 liter x 7). The combined organic layers were dried with anhydrous sodium s The residue was triturated with petroleum ether / ethyl acetate = 10/1 (200 mL). The solid was dried under reduced pressure to give 2-(6-methyl-2-pyridine)-5,6-dihydro-4H-pyrrole[1,2-b]pyrazole-3-carboxylic acid (52.80 g, yield : 40.52%).
Step D: Dissolving 2-(6-methyl-2-pyridyl)-5,6-dihydro-4H-pyrrole[1,2-b]pyrazole-3-carboxylic acid (45.00 g, 184.99 mmol) In N,N-dimethylformamide (650.00 ml), then NBS (49.09 g, 258.99 mmol). The reaction mixture was stirred at 30-40 ° C for 60 hours, then diluted with water (600 mL) and extracted with dichloromethane / isopropyl alcohol (10/1) (500 mL × 3). The combined organic phases were washed with EtOAc (EtOAc m. The resulting solid was slurried with EtOAc/EtOAc =EtOAc (EtOAc). The solid was dried under reduced pressure to give 3-bromo-2-(6-methyl-2-pyridine)-5,6-dihydro-4H-pyrrole[1,2-b]pyrazole (33.00 g, yield: 64.13%).
Step E: 3-Bromo-2-(6-methyl-2-pyridyl)-5,6-dihydro-4H-pyrrole [1,2-b]pyrazole (1.00 g, 3.60 mmol) and boric acid Triisopropyl ester (1.79 g, 9.54 mmol) was dissolved in tetrahydrofuran (20.00 mL). The reaction mixture was cooled to minus 70 ° C, then n-butyllithium (2.5 M, 3.74 mL) was then added dropwise. After completion of the dropwise addition, the reaction mixture was stirred at 25 ° C for 1 hour, and then the pH was adjusted to 7 with aqueous hydrochloric acid (0.5 mol / liter). The tetrahydrofuran was then concentrated under reduced pressure and cooled to 15 °C. The mixture was filtered, and the filtered cake was purified with EtOAc EtOAc EtOAc (EtOAc) 5,6-Dihydro-4H-pyrrole[1,2-b]pyrazol-3-yl]boronic acid (750 mg, yield: 85.71%).
Preparation of Example 1:
Step A: 6-Iodo-[1,2,4]triazolo[1,5-a]pyridine (16.00 g, 65.30 mmol) was dissolved in tetrahydrofuran (800.00 mL) and cooled to below 60-70 ° C. Thereafter, lithium hexamethyldisilazide (1 mol/liter, 130.60 ml, 65.30 mmol) was added dropwise. The reaction mixture was stirred at minus 60-70 ° C for 15 minutes and N,N-dimethylformamide (14.32 g, 195.90 mmol, 15.07 mL). Stirring was then continued at minus 60 to 70 degrees C for 15 minutes and then quenched with saturated aqueous ammonium chloride (500 mL). The reaction mixture was warmed to room temperature and then extracted with ethyl acetate (500 ml). The combined organic layers were washed with EtOAc EtOAc m. The residue was purified with a silica gel column (eluent: methylene chloride / ethyl acetate = 10/1) to afford 6-iodo-[1,2,4]triazolo[1,5-a]pyridine-5- Formaldehyde (6.40 g, yield: 35.90%). . 1H NMR (400 MHz, DMSO-d6) 10.46 (S, IH), 8.62 (S, IH), 8.16 (D, J = 9.3Hz, IH), 7.88 (D, J = 9.3Hz, IH).
Step B: To a 500 ml three-necked flask equipped with a thermometer and a nitrogen balloon, 2-diethoxyphosphorylacetonitrile (3.83 g, 21.61 mmol, 3.48 ml) and tetrahydrofuran (80 ml) were added. The mixture was cooled to 0.degree. C. and then potassium tert-butoxide (2.42 g, 21.61 mmol). The reaction mixture was stirred at 0 ° C for 15 minutes and then added dropwise to another suspension through a dropping funnel (dispersing 6-iodo-[1,2,4]triazolo[1,5-a]pyridine-5-carbaldehyde In tetrahydrofuran (120 ml) and cooled to 0 ° C). The reaction mixture was stirred at 0<0>C for 15 min then EtOAc (EtOAc)EtOAc. The combined organic layers were washed with EtOAc EtOAc m. The residue was purified with a silica gel column (eluent: methylene chloride / ethyl acetate = 200/1 to 10/1) to afford (E)-3-(6-iodo-[1,2,4]triazole. [1,5-a]pyridin-5-yl)prop-2-enenitrile (4.2 g, yield: 65.66%). . 1 H NMR (400 MHz, CHLOROFORM-D) [delta] 8.42 (S, IH), 8.03 (D, J = 9.3Hz, IH), 7.98-7.91 (m, IH), 7.85-7.78 (m, IH), 7.60 (d, J = 9.2 Hz, 1H).
Step C: (E)-3-(6-Iodo-[1,2,4]triazolo[1,5-a]pyridin-5-yl)prop-2-enenitrile (4.50 g, 15.20 m Mole), [2-(6-methyl-2-pyridyl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl]boronic acid (4.43 g, 18.24 m Mole), sodium carbonate (4.83 g, 45.60 mmol), [1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride (556.07 mg, 759.96 μmol), 2-dicyclohexylphosphine- 2′,6′-dimethoxybiphenyl (311.98 mg, 759.96 μmol) and [2-(2-aminophenyl)phenyl]-chloro-palladium-cyclohexyl-[2-(2,6- Dimethoxyphenyl)phenyl]phosphine (547.64 mg, 759.96 μmol) was added to a mixed solvent of dioxane (100 ml) and water (20 ml). It was replaced with nitrogen 3 times and then heated to 90-100 ° C and stirred for 2 hours. The reaction mixture was poured into water (200 ml) and evaporated and evaporated. The combined organic layers were washed with EtOAc EtOAc m. The residue was purified with EtOAc mjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjj The solid was concentrated and dried under reduced pressure to give (E)-3-[6-[2-(6-methyl-2-pyridyl)-5,6-dihydro-4H-pyrrolo[1,2-b] Pyrazol-3-yl]-[1,2,4]triazolo[1,5-a]pyridin-5-yl]prop-2-enenitrile (5.37 g, yield: 96.16%). . 1 H NMR (400 MHz, CHLOROFORM-D) [delta] 8.49 (S, IH), 7.82-7.74 (m, 2H), 7.59-7.46 (m, 4H), 6.99 (dd, J = 2.6,6.1Hz, IH) , 4.39 (d, J = 6.3 Hz, 2H), 2.90 – 2.70 (m, 4H), 2.20 (s, 3H).
Step D: (E)-3-[6-[2-(6-Methyl-2-pyridyl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole-3 -yl]-[1,2,4]triazolo[1,5-a]pyridin-5-yl]prop-2-enenitrile (5.37 g, 14.62 mmol) dissolved in dichloromethane (20 mL) , a mixed solvent of dimethyl sulfoxide (70 ml) and water (20 ml), then separately added hydrogen peroxide (8.29 g 73.10 mmol, 7.02 ml, 30%) and sodium hydroxide (2 mol / liter, 14.62 ml) ). The mixture was stirred at 15-20 degrees Celsius for 12 hours. The mixture was poured into water (200 ml), and extracted with a mixture solvent of dichloromethane/isopropanol (3/1) (200 ml × 1). The organic layer was washed with EtOAc EtOAc m. The residue was purified by preparative high performance liquid chromatography (column: Phenomenex Gemini C18 250 x 50 mm x 10 μm; mobile phase: [water (0.05% ammonia v/v)-acetonitrile]; gradient: 5%-32%, 33 80% min) Example 1 (3.6 g, yield: 63.82%) was obtained. . 1 H NMR (400 MHz, CHLOROFORM-D) [delta] 8.45 (S, IH), 8.09 (D, J = 15.6Hz, IH), 7.85 (D, J = 15.6Hz, IH), 7.69 (D, J = 9.2 Hz, 1H), 7.55-7.45 (m, 2H), 7.37 (d, J = 7.8 Hz, 1H), 6.99 (d, J = 7.7 Hz, 1H), 5.93-5.65 (m, 2H), 4.35 (br .s., 2H), 2.99-2.64 (m, 4H), 2.33 (s, 3H).

PATENT

WO-2019114792

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019114792&tab=FULLTEXT&maxRec=1000

Novel crystalline and salt (hydrochloride, sulfate and mesylate) forms of a TGF-βRI inhibitor, designated as Forms A and B, processes for their preparation and compositions comprising them are claimed for treating cancers. The compound was originally claimed in WO2017215506 , assigned to Medshine Discovery Inc alone.

Example 1 Preparation of a compound of formula (I)
Preparation of intermediates 1-6:
Step A: Ethyl acetate (291.41 ml, 2.98 mol) was dissolved in toluene (750.00 ml), and then sodium ethoxide (135.06 g, 1.98 mol) was added portionwise at room temperature, and the mixture was stirred at room temperature for 1 hour. 1-1 (150.00 g, 992.33 mmol) was added to the above reaction liquid at 25 ° C, and then heated to 95 ° C and stirred for 15 hours. The reaction mixture was cooled to about 30 ° C, and the pH was adjusted to 7 with acetic acid, diluted with water (500 ml), and ethyl acetate (500 ml). The organic phase was dried with anhydrous sodium s The residue was purified with a silica gel column (eluent: petroleum ether/ethyl acetate v/v = 50/1) to afford 1-2.
Step B: Dissolve 1-2 (120.00 g, 579.07 mmol) in pyridine (300 mL), then add 1-aminopyrrolidin-2-one p-toluenesulfonate (172.01 g, 631.66 mmol) ). The reaction mixture was stirred at 25 ° C for 16 hours and then concentrated under reduced vacuo. The residue was diluted with water (300 ml) and then extracted with ethyl acetate (300 ml). The combined organic layers were dried with anhydrous sodium s
Step C: 1-3 (155.00 g, 535.72 mmol) was dissolved in toluene then sodium ethoxide (72.91 g, 1.07 mol). The reaction mixture was heated to 100 ° C and stirred for 16 hours, then cooled to room temperature. It was slowly diluted with water (1.5 liters), adjusted to pH 4 with concentrated hydrochloric acid, and extracted with dichloromethane/isopropanol (v/v = 10/1, 1 liter x 7). The combined organic layers were dried with anhydrous sodium s The residue was triturated with petroleum ether / ethyl acetate (v/v = 10/1, 200 mL). The solid was dried under reduced pressure to give 1-4.
Step D: 1-4 (45.00 g, 184.99 mmol) was dissolved in N,N-dimethylformamide (650.00 ml), then NBS (49.09 g, 258.99 mmol). The reaction mixture was stirred at 30 to 40 ° C for 60 hours, then diluted with water (600 ml), and extracted with dichloromethane / isopropyl alcohol (v / v = 10 / 1,500 ml × 3). The combined organic phases were washed with EtOAc (EtOAc m. The resulting solid was slurried with EtOAc/EtOAc (EtOAc/EtOAc) The solid was dried under reduced pressure to give 1-5.
Step E: 1-5 (1.00 g, 3.60 mmol) and triisopropyl borate (1.79 g, 9.54 mmol) were dissolved in tetrahydrofuran (20.00 mL). The reaction mixture was cooled to minus 70 ° C, then n-butyllithium (2.5 M, 3.74 mL) was added dropwise. After completion of the dropwise addition, the reaction mixture was stirred at 25 ° C for 1 hour, and then the pH was adjusted to 7 with aqueous hydrochloric acid (0.5 mol / liter). It was then concentrated under reduced pressure to remove tetrahydrofuran and cooled to 15 °C. The mixture was filtered, and the EtOAc EtOAc m.
Preparation of the compound of formula (I):
Step A: 1-7 (16.00 g, 65.30 mmol) was dissolved in tetrahydrofuran (800.00 ml), cooled to minus 60-70 ° C, and lithium hexamethyldisilazide (1 mol/L, 130.60) was added dropwise. ML, 65.30 mmol). The reaction mixture was stirred at -60 to 70 ° C for 15 minutes, and N,N-dimethylformamide (14.32 g, 195.90 mmol, 15.07 ml) was added. Stirring was then continued at minus 60-70 ° C for 15 minutes and then quenched with saturated aqueous ammonium chloride (500 mL). The reaction mixture was warmed to room temperature and then extracted with ethyl acetate (500 ml). The combined organic layers were washed with EtOAc EtOAc m. The residue was purified with a silica gel column (eluent: methylene chloride / ethyl acetate v/v = 10/1) to afford 1-8. . 1 H NMR (400 MHz, DMSO-d6) 10.46 (S, IH), 8.62 (S, IH), 8.16 (D, J = 9.3Hz, IH), 7.88 (D, J = 9.3Hz, IH).
Step B: To a 500 ml three-necked flask equipped with a thermometer and a nitrogen balloon, 2-diethoxyphosphorylacetonitrile (3.83 g, 21.61 mmol, 3.48 ml) and tetrahydrofuran (80 ml) were added. The mixture was cooled to 0 ° C then potassium tert-butoxide (2.42 g, 21.61 mmol). The reaction mixture was stirred at 0<0>C for 15 min then added dropwise to a further suspension (1~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~ The reaction mixture was stirred at 0<0>C for 15 min then EtOAc (EtOAc)EtOAc. The combined organic layers were washed with EtOAc EtOAc m. The residue was purified with a silica gel column (eluent: methylene chloride/ethyl acetate v/v = 200/1 to 10/1) to afford 1-9. . 1 H NMR (400 MHz, CDCl3 . 3 ) [delta] 8.42 (S, IH), 8.03 (D, J = 9.3Hz, IH), 7.98-7.91 (m, IH), 7.85-7.78 (m, IH), 7.60 ( d, J = 9.2 Hz, 1H).

Step C: 1-9 (4.50 g, 15.20 mmol), 1-6 (4.43 g, 18.24 mmol), sodium carbonate (4.83 g, 45.60 mmol), [1,1′-bis (diphenyl) Phosphine) ferrocene] palladium dichloride (556.07 mg, 759.96 μmol), 2-biscyclohexylphosphine-2′, 6′-dimethoxybiphenyl (311.98 mg, 759.96 μmol) and [2-( 2-Aminophenyl)phenyl]-chloro-palladium-cyclohexyl-[2-(2,6-dimethoxyphenyl)phenyl]phosphine (547.64 mg, 759.96 μmol) was added to the dioxane (100 ml) and water (20 ml) in a mixed solvent. It was replaced with nitrogen three times and then heated to 90 to 100 ° C and stirred for 2 hours. The reaction mixture was poured into water (200 ml) and evaporated and evaporated. The combined organic layers were washed with EtOAc EtOAc m. The residue was purified on a silica gel column (eluent: methylene chloride/methanol, v/v=30/1) to afford crude crude product in petroleum ether/ethyl acetate (v/v=5/1) After stirring for 12 hours, the solid was collected by filtration, and the solid was concentrated and dried under reduced pressure to give 1-10. . 1 H NMR (400 MHz, CDCl3 . 3 ) [delta] 8.49 (S, IH), 7.82-7.74 (m, 2H), 7.59-7.46 (m, 4H), 6.99 (dd, J = 2.6,6.1Hz, IH), 4.39 (d, J = 6.3 Hz, 2H), 2.90 – 2.70 (m, 4H), 2.20 (s, 3H).

Step D: 1-10 (5.37 g, 14.62 mmol) was dissolved in a mixed solvent of dichloromethane (20 ml), dimethyl sulfoxide (70 ml) and water (20 ml), and then hydrogen peroxide ( 8.29 g 73.10 mmol, 7.02 mL, 30%) and sodium hydroxide (2 mol/L, 14.62 mL). The mixture was stirred at 15 to 20 ° C for 12 hours. The mixture was poured into water (200 ml), and extracted with a mixture solvent of dichloromethane/isopropanol (3/1) (200 ml × 1). The organic layer was washed with EtOAc EtOAc m. The residue was purified by preparative high performance liquid chromatography (column: Phenomenex Gemini C18 250 x 50 mm x 10 μm; mobile phase: [water (0.05% ammonia v/v)-acetonitrile]; gradient: 5%-32%, 33 80% minute) to give a compound of formula (I). . 1 H NMR (400 MHz, CDCl3 . 3 ) [delta] 8.45 (S, IH), 8.09 (D, J = 15.6Hz, IH), 7.85 (D, J = 15.6Hz, IH), 7.69 (D, J = 9.2Hz , 1H), 7.55-7.45 (m, 2H), 7.37 (d, J = 7.8 Hz, 1H), 6.99 (d, J = 7.7 Hz, 1H), 5.93-5.65 (m, 2H), 4.35 (br. s., 2H), 2.99-2.64 (m, 4H), 2.33 (s, 3H).
Example 2 Preparation of a compound of formula (II)
115 mg of the compound of formula (I) was added to an 8 ml glass vial, 4 ml of tetrahydrofuran was added, and the solution was sonicated by ultrasonication; then 1.05 equivalent of p-toluenesulfonic acid monohydrate was slowly added. The suspension sample was placed on a magnetic stirrer (40 ° C) and stirred for 16 hours. The sample solution was centrifuged, and the solid was taken out and dried in a vacuum oven at 35 ° C for 16 hours to obtain a compound of the formula (II). 1 H NMR (400 MHz, CD 3 OD) δ 8.61 (s, 1H), 8.14 (t, J = 8.0 Hz, 1H), 8.05 (d, J = 15.6 Hz, 1H), 7.90 (d, J = 8.8 Hz, 1H), 7.70 (dd, J=8.4, 15.6 Hz, 4H), 7.54 (d, J = 15.6 Hz, 1H), 7.39 (d, J = 8.0 Hz, 1H), 7.20 (d, J = 7.6) Hz, 2H), 4.42 (m, 2H), 3.05-2.87 (m, 2H), 2.82 (s, 3H), 2.81-2.74 (m, 2H), 2.35 (s, 3H).
Example 3 Preparation of a compound of formula (IV)
115 mg of the compound of formula (I) was added to an 8 ml glass vial, 4 ml of tetrahydrofuran was added, and the solution was sonicated by ultrasonication; then 1.05 equivalent of hydrochloric acid was slowly added. The suspension sample was placed on a magnetic stirrer (40 ° C) and stirred for 16 hours. The sample solution was centrifuged, and the solid was taken out and dried in a vacuum oven at 35 ° C for 16 hours. The obtained solid was added to an appropriate amount of acetone to prepare a suspension and stirred at 40 ° C, and the supernatant was discarded by centrifugation, and the solid sample was drained with an oil pump at room temperature to obtain a compound of the formula (IV).
Example 4 Preparation of a compound of formula (V)
115 mg of the compound of formula (I) was added to an 8 ml glass vial, 4 ml of tetrahydrofuran was added, and the solution was sonicated by ultrasonication; then 1.05 equivalent of sulfuric acid was slowly added. The suspension sample was placed on a magnetic stirrer (40 ° C) and stirred for 16 hours. The sample solution was centrifuged, and the solid was taken out and dried in a vacuum oven at 35 ° C for 16 hours to obtain a compound of the formula (V).
Example 5 Preparation of a compound of formula (VI)
115 mg of the compound of formula (I) was added to an 8 ml glass vial, 4 ml of tetrahydrofuran was added, and the solution was sonicated by ultrasonication; then 1.05 equivalent of methanesulfonic acid was slowly added. The suspension sample was placed on a magnetic stirrer (40 ° C) and stirred for 16 hours. The sample solution was centrifuged, and the solid was taken out and dried in a vacuum oven at 35 ° C for 16 hours to obtain a compound of the formula (VI).
Example 6 Preparation of Form A of Compound of Formula (I)
10 g of the compound of the formula (I) was placed in a mixed solvent of ethanol (80 ml) and water (40 ml), heated to 70-75 ° C and stirred until clarified, and filtered while hot, and the filtrate was distilled under reduced pressure to a volume of the remaining solution. 50 ml, followed by cooling to stand for crystallisation, filtration, and the resulting filter cake was dried under reduced pressure to give a solid of the compound of formula (I).
Example 7 Preparation of Form B of Compound of Formula (II)

192 mg of the compound of formula (I) was weighed into a glass bottle. 10 ml of a tetrahydrofuran:acetic acid (v/v=9/1) mixed solvent was added, and after ultrasonic assisted for 30 minutes, the sample was dissolved into a clear solution. Stir on a magnetic stirrer (40 ° C). After 1.05 equivalents of p-toluenesulfonic acid monohydrate was slowly added, the sample was stirred overnight. After naturally cooling to room temperature, the supernatant was discarded by centrifugation, stirred for 10 hours by adding 10 ml of tetrahydrofuran, and the supernatant was discarded by centrifugation, and the same procedure was repeated twice more. The obtained solid was dried in a vacuum oven at 40 ° C for 1 hour, and after milling, it was further dried in a vacuum oven at 30 ° C for 16 hours to obtain a crystal form B of the compound of the formula (II).

.///////////////////GFH-018, GFH 018, GenFleet Therapeutics, Advanced solid tumor,  Cancer, PRECLINICAL

NC(=O)/C=C/c4n5ncnc5ccc4c2c3CCCn3nc2c1cccc(C)n1

HM04 or H0900

$
0
0

str2

3-[(1R)-1-(2,3-Dichloro-4-pyrazin-2-ylphenyl)-2,2,2-trifluoroethyl]-1-methyl-1-(1-methylpiperidin-4-yl)urea.png

HM04 or H0900

Cas 1808913-24-7

MF C20 H22 Cl2 F3 N5 O
MW 476.32
Urea, N‘-[(1R)-1-[2,3-dichloro-4-(2-pyrazinyl)phenyl]-2,2,2-trifluoroethyl]-N-methyl-N-(1-methyl-4-piperidinyl)-

(R)-3-(1-(2,3-dichloro-4-(pyrazin-2-yl)phenyl)-2,2,2-trifluoroethyl)-1-methyl-1-(1-methylpiperidin-4-yl) urea

The compound was disclosed in WO2015134839 . Helsinn under license from Novo Nordisk , is investigating ghrelin antagonists for treating obesity, Prader-Willi syndrome and other metabolic disorders; in May 2015, the program was listed as being in preclinical development

Helps reducing ghrelin signaling activity and treating disorder associated with an increase in ghrelin level (eg food abuse, alcohol addiction, and Prader-Willi syndrome).

Ghrelin, a growth hormone-releasing peptide produced by ghrelinergic cells in the gastrointestinal tract, is understood to function as a neuropeptide that regulates energy metabolism by stimulating appetite. The modulation, for example inhibition, of ghrelin signaling, through the ghrelin/growth hormone secretagogue receptor (GHS-Rla), is an attractive target for pharmacological treatment of disorders associated with high ghrelin level. Potential disorders for treatment using ghrelin modulators include food abuse (such as binge eating, obesity, hyperphagia (or uncontrollable appetite), post-dieting body weight rebound (including post-dieting hyperphagia), alcohol addiction, and genetic diseases associated with increased ghrelin level (e.g., Prader-Willi syndrome (PWS)).

PATENT

US 20150252021

PATENT

WO2015134839

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2015134839

Example 1

nthesis of Intermediate lk

Intermediate k

Step 1:

To a solution of la (100 g, 0.62 mol) in DMF (1.2 L) was added N-bromosuccinimide (110 g, 0.62 mol) at 0 °C. The mixture was stirred at room temperature for 4 h, then water (800 mL) was added and the resulting mixture was extracted with EtOAc (3 x 500 mL). The combined organic layers were dried over anhydrous Na2S04 and concentrated under reduced pressure. The residue was triturated with petroleum ether to provide lb (133.7 g, 89% yield) as a brown solid. !H-NMR (CDC13, 300 MHz): δ= 7.30 (d, 1 H), 6.59 (d, 1 H), 4.22 (br, 2 H). LC-MS: 241 [M+l]+.

Step 2:

To a solution of lb (133.7 g, 0.55 mol) in dry CH2C12 (1.5 L) was added acetic anhydride (110 g, 0.62 mol) dropwise over a period of 20 minutes at room temperature. The mixture was stirred at room temperature overnight, then diluted with CH2C12 (300 mL) and washed with water (150 mL) and brine (200 mL). The organic layer was separated, dried over anhydrous Na2SC>4 and concentrated under reduced pressure. The residue was triturated with petroleum ether (300 mL) to provide compound lc (143.0 g, 91% yield) as a white solid. ¾-NMR (CDC13, 400 MHz): δ= 8.26 (d, 1 H), 7.63 (br, 1 H), 7.54 (d, 1 H), 2.26 (s, 3 H). LC-MS: 280 [M-l].

Step 3:

A mixture of compound lc (50.0 g, 0.18 mol), butyl vinyl ether (Id, 89.0 g, 0.89 mol), bis(l,3-diphenylphosphino)propane (DPPP, 22.0 g, 0.053 mol), TEA (100 mL, 0.71 mol) and Pd(OAc)2 (6.4 g, 0.027 mol) in DMSO (1.2 L) was heated at 130 °C under N2 overnight. After the reaction was completed, the mixture was cooled to 0 °C and 2N HC1 (480 mL) was added dropwise over a period of 30 minutes. Then, the mixture was extracted with EtOAc (3 x 100 mL). The combined organic layers were dried over anhydrous a2S04 and concentrated under reduced pressure. The residue was purified by column chromatography (silica, EtOAc: PE=1 : 10) to provide le (19.5 g, 45% yield) as a yellow solid. 1H-NMR (CDC13, 400 MHz): 3= 8.46 (d, 1 H), 7.82 (br, 1 H), 7.51 (d, 1 H), 2.63 (s, 3 H), 2.29 (s, 3 H). LC-MS: 244 [M-l].

Step 4:

To a solution of le (21.9 g, 89.4 mmol) in MeOH (350 mL) was added 2N NaOH solution (350 mL) at room temperature. The mixture was heated at 50 °C overnight, then cooled and concentrated under reduced pressure. The resulting solid was triturated with water (100 mL) for 30 min and filtered to provide If (18.0 g, 98% yield) as a brown solid. ¾-NMR (CDC13, 400 MHz): 3= 7.48 (d, 1 H), 6.68 (d, 1 H), 4.56 (br, 2 H), 2.62 (s, 3 H). LC-MS: 202[M-1]\

Step 5:

To a mixture of compound If (18.0 g, 89.2 mmol) and ice (360 g) in cone. HC1 (180 mL) was added a solution of NaN02 (9.2 g, 133.7 mmol) in water (20 mL) dropwise over a period of 30 minutes, and the resulting mixture stirred in an ice bath for 30 min. A solution of KI (74.0 g, 446 mmol) in water (360 mL) was added dropwise over 45 min at 0 °C. The mixture was stirred for 30 min and then extracted with EtOAc (3 x 100 mL). The combined organic layers were dried over anhydrous Na2SC>4 and concentrated under reduced pressure. The residue was purified by column chromatography (silica, EtOAc: PE=1 :40) to provide lg (23.9 g, 86% yield) as a yellow solid. 1H-NMR (CDC13, 400 MHz): 3= 7.6 (d, 1 H), 7.06 (d, 1 H), 2.62 (s, 3 H).

Step 6:

To a solution of lg (23.9 g, 76.1 mmol) in MeOH (100 mL)/THF (100 mL) was slowly added NaB¾ (2.9 g, 76.1 mmol) at 0 °C. The mixture was stirred at room temperature for 5 min, and then quenched with water (100 mL). The mixture was extracted with EtOAc (3 x 100 mL). The combined organic layers were dried over anhydrous Na2SC>4 and concentrated under reduced pressure. The residue was purified by column chromatography (silica, EtOAc: PE=1 : 10) to provide lh (22.4 g, 93% yield) as a white solid. 1H-NMR (CDC13, 400 MHz): 3= 7.81 (d, 1 H), 7.26 (d, 1 H), 5.23 (q, 1 H), 2.17 (br, 1 H), 1.47 (d, 3 H).

Step 7:

To a mixture of lh (22.4 g, 70.9 mmol), phthalimide (12.5 g, 85.0 mmol) and PPh3 (22.3 g, 85.0 mmol) in dry THF (450 mL) was added DIAD (21.5 g, 106.3 mmol) at room temperature under N2 protection. The mixture was stirred at room temperature overnight and then concentrated under reduced pressure. The residue was purified by column chromatography (silica, EtOAc: PE=1 : 15) to provide li (18.5 g, 58% yield) as a white solid. 1H-NMR (CDC13, 400 MHz): 3= 7.78-7.84 (m, 3 H), 7.70-7.73 (m, 2 H), 7.41-7.43 (d, 1 H), 5.76-5.81 (q, 1 H), 1.84 (d, 3 H).

Step 8:

A solution of li (7.2 g, 16.2 mmol) and hydrazine hydrate (98%, 4.0 g, 80.9 mmol) in MeOH (150 mL) was heated under reflux for 2 h, then cooled and concentrated under reduced pressure. The residue was diluted with water (100 mL) and extracted with CH2C12 (3 x 100 mL). The combined organic layers were dried over anhydrous Na2SC>4 and concentrated under reduced pressure to give lj (3.8 g, 75% yield) as a white solid. 1H-NMR (CDC13, 400 MHz): 3= 7.81 (d, 1 H), 7.25 (d, 1 H), 4.55 (q, 1 H), 1.36-1.38 (d, 3 H). LC-MS: 316 [M+l]+.

Step 9:

To a solution of lj (41. Og, 0.13 mol) in methyl tert-butyl ether (750 mL) was added slowly a solution of D-mandelic acid (7.8 g, 0.052 mol) in methyl tert-butyl ether (1 10 mL) at 45°C. The mixture was stirred at this temperature for 30 min then cooled and filtered. White solid obtained was partitioned between 5% NaOH solution (300 mL) and methyl tert-butyl ether (300 mL). The bi -phases were separated and the aqueous phase was extracted with methyl tert-butyl ether (300 mL). The combined organic layer was concentrated to provide Intermediate lk (12 g, 58.5% yield) as a white solid (ee%=98.0%, Chiralpak AD-H, 5 μπι, 4.6*250mm, mobile phase: Hex: EtOH : DEA=80 : 20 : 0.2), retention time = 6.408 min).

Example 2

Synthesis of Compoun

A suspension of N-methyl-4-piperidone 2a (13.3 g, 58.6 mmol), NH2Me (30% in MeOH, 100 mL) and Pd/C (0.66 g) in MeOH (200 mL) was heated at 60 °C under H2 atmosphere (50 psi) overnight, then cooled and filtered. The filtrate was concentrated under reduced pressure and the residue was dissolved in HC1 in dioxane (3N, 100 mL) and stirred for 30 min. The precipitate was filtered and washed with EtOAc (50 mL) to provide 2b (7.7g, 54% yield) as white powder. 1H-NMR (DMSO, 400 MHz): δ= 9.50 (br, 2 H), 3.48 (d, 2 H), 3.15-3.16 (m, 1 H), 2.96-3.01 (m, 2 H), 2.70 (s, 3 H), 2.51 (s, 3 H), 2.22-2.28 (m, 2 H), 1.94-2.02 (m, 2 H), LC-MS: 129 [M+l]+ .

Example 20

Synthesis of H0900

Step 1:

To a mixture of 16d (32 g, 120 mmol) in dry CH2CI2 (800 mL) was added Dess-Martin peroxide reagent (76 g, 180 mmol) portion- wise at 0 °C. The mixture was stirred at room temperature for 1 h, then diluted with DCM (800 mL), washed with aqueous NaHC03 solution (300 mL) and brine (300 mL). The organic phase was separated, dried over anhydrous Na2S04 and

concentrated under reduced pressure to afford crude 18a (31.4 g) which was used directly in the next step without further purification.

Step 2:

To a solution of 18a (12 g, 40 mmol) and 3b (22.2 g, 60 mmol) in DME (560 mL) were added Pd(PPh3)4 (9.25 g, 8 mmol) and Cul (1.52 g, 8 mmol) at room temperature. The mixture was stirred at 90 °C overnight, then concentrated under reduced pressure. The residue was purified with silica gel column chromatography (silica, EA : PE = 1 :5) to provide 18b (8.0 g, 79.3%) as a white solid. LC-MS: 253 [M+l]+.

Step 3:

To a solution of 18b (7 g, 27.7 mmol) and (¾)-tert-butylsulfinamide (7.27 g, 30.56 mmol) in dry THF (200 mL) was added Ti(i-OPr)4 (15.7 g, 55.4 mmol) dropwise at room temperature. The mixture was stirred at 80 °C overnight, and then cooled. Ethyl acetate (40 mL) was added, the resulting mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was purified with silica gel column chromatography (silica, EA:PE =1 :5) to provide 18c (6.8 g, 69%) as a yellow solid. 1H-NMR (CDC13, 400 MHz): 3= 9.10 (s, 1H), 8.97 (s, 1H), 8.72 (s, 1H), 8.64 (d, 1H),8.12 (d, 1H), 7.59 (d, 1H), 1.30 (s, 9H).LC-MS: 356 [M+l]+.

Step 4:

To a stirred solution of 18c (6.8 g, 19 mmol) and Tetrabutylammonium difluorotnphenylsilicate (15.8 g, 29 mmol) in dry THF (250 mL) was added a solution of TMSCF3 (11 g, 77 mmol) in anhydrous THF (50 mL) at -65 °C. The mixture was then stirred at -65 °C for 2 h, and at that point aqueous NH4CI solution (250 mL) was added. The mixture was diluted with ethyl acetate (250 mL), washed with brine (250 mL), dried over anhydrous Na2SC>4 and concentrated under reduced pressure. The residue was purified with silica gel column chromatography (silica, EA : PE=1 :2) to provide 18d (4.3 g, 52%) as a yellow solid. LC-MS: 426 [M+l]+.

Step 5:

To a stirred solution of 18d (4.3 g, 10.1 mmol) in MeOH (40 mL) was added a solution of HCl/MeOH (4N, 40 mL) at room temperature. The mixture was stirred for 1 h, then concentrated under reduced pressure. The residue was triturated with ethyl acetate (40 mL) to afford crude 18e (4.3g) which was directly in the next step without further purification. LC-MS: 322 [M+l]+.

Step 6:

To a solution of 18e (2.7 g, 7.1 mmol), 2b (3.4 g, 21.3 mmol) and TEA (80 mL) in DCM (220 mL) was added thiphosgene (3.15 g, 10.6 mmol) in DCM (40 mL) dropwise at 0 °C. The solution was warmed to ambient temperature and stirred for 1 h, then diluted with DCM ( 100 mL) and washed with aqueous Na2C03 solution (100 mL) and brine (100 mL). The organic layer was separated, dried over anhydrous Na2SC>4 and concentrated. The residue was purified with silica gel column chromatography (silica, DCM : CH3OH=10 : 1) to provide crude H0900 (2.13 g, ee%=92.5%) which was further purified through chiral separation to afford H0900 (1.6 g, 49% yield) as a white solid. (ee%=98.5%, Chiralpak IC 5um, 4.6*250mm, Phase: Hex: EtOH:

DEA=90: 10:0.2), retention tine =12.829 min. 1H-NMR (CDC13, 400 MHz): δ= 8.86 (d, 1H), 8.63 (dd, 1H), 8.55 (d, 1H), 7.47 (d, 1H), 7.40 (d, 1H), 6.28 (m, 1H), 5.18 (d, 1H), 4.12 (m, 1H), 2.88 (t, 2H), 2.77 (s, 3H), 2.22 (s, 3H), 2.05 (m, 2H), 2.48 (m, 2H), 1.52 (m, 2H), 1.73-1.49 (m, 4H). LC-MS: 476 [M+l]+.

PATENT

WO-2019118298

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019118298&tab=PCTDESCRIPTION&maxRec=1000

Novel crystalline fumarate salt forms of (R)-3-(1-(2,3-dichloro-4-(pyrazin-2-yl)phenyl)-2,2,2-trifluoroethyl)-1-methyl-1-(1-methylpiperidin-4-yl) urea (also referred to as HM04 or H0900; designated as Forms 1-4), process for their preparation and compositions comprising them are claimed.

PWS occurs in approximately 1 in 10,000 births and is associated with deletion or lack of expression of region 15ql 1.2 of the paternal chromosome 15.

Characteristics of PWS include short stature, low muscle tone, and hyperphagia. Growth hormone replacement is frequently used to treat growth deficiencies and hypotonia. However, treatment for the insatiable appetite is lacking and PWS children can mature into adults suffering from obesity and type 2 diabetes. Levels of ghrelin are generally elevated in PWS; however, the relationship with ghrelin signaling and food intake in PWS remains unclear. See Purtell L., et ah, In adults with Prader-Willi syndrome, elevated ghrelin levels are more consistent with hyperphagia than high PYY and GLP-l levels. Neuropeptides. 201 l;45(4):30l-7; Cummings D.E., et ah, Elevated plasma ghrelin levels in Prader Willi syndrome. Nature Medicine . 2002;8(7):643-4; DelParigi A., et ah, High circulating ghrelin: a potential cause for hyperphagia and obesity in Prader-Willi syndrome. The Journal of Clinical Endocrinology and Metabolism. 2002;87(l2):546l-4.

[005] Accordingly, it is desirable to find treatments that effectively inhibit GHSRla, that are tolerable to the patient, and that do not interfere with other functions of the growth hormones. GHSRla modulators, including inhibitors such as (R)-3-(l-(2,3-dichloro-4-(pyrazin-2-yl)phenyl)-2, 2, 2-trifluoroethyl)-l -methyl- l-(l-m ethylpiperidin-4-yl) urea (HM04, H0900) depicted below, are reported in LT.S. Patent No. 9,546,157.

Step 1 : Synthesis of compound 2A

[00106] 2,2,6,6-tetramethylpiperidine (7.20 kg, 51.1 mol, 3.0 eq.,

KF=0.30%) was added into a 100 L reactor equipped with a temperature probe and overhead stirrer and mixed at RT under nitrogen protection. THF (50 L) was added into the reactor and stirred. The vessel was purged with nitrogen three times and cooled to 0 °C. n-BuLi (20.4 L, 3.0 eq.; 2.5 M hexane solution) was added to the mixture dropwise while keeping the temperature at about 0 °C to about 5 °C for over one hour. The color of the solution turned yellow. The mixture was stirred at about 0 °C to about 5 °C for 30 minutes. The mixture was cooled to about -78 °C to about -70 °C to form Solution A.

[00107] Compound 1 (3.25 kg, 17.0 mol. 1.0 eq., KF=0.03%) was dissolved in 15 L of THF to form Solution B.

[00108] Solution B was added to solution A dropwise at a temperature of about -70 °C to about -78 °C over one hour and then stirred for 30 minutes to form solution C. Tri-isopropyl borate ((i-PrO)3B) (3.52 kg, 18.7 mol., 1.1 eq.) was added dropwise into solution C over 10 minutes. The reaction mixture was stirred at a

temperature of about -70 °C to about -78 °C for one hour. HC1 (40 L, 3M, 7.0 eq.) was added over 30 minutes to quench the reaction. A 10 degree rise in temperature was noted.

[00109] The resulting aqueous layer was separated and extracted with EtOAc (40 L). The aqueous layer was separated and extracted twice again with EtOAc (35 L, 30 L). The organic layers were combined resulting in about 160 L of liquid. The combined organic layer was washed twice with 50 L of a 1M aqueous HC1 solution saturated with NaCl. The organic layer was concentrated to about 5 L in a 50 L rotavapor at a temperature of about 50 °C to about 55 °C under 30-40 mmHg for about 8 hours.

[00110] The residual EtOAc was swapped with DME for 3 times (10 L x 3). The organic layer was concentrated in the 50 L rotavapor at a temperature of about 50 °C to about 55 °C under 30-40 mmHg for about 6 hours. Each time about 5 L of residual remained. DME (20 L) was added to the residual to obtain a deep brown solution of 14.2% compound 2A (3.55 kg in 25 kg of solution; 88.8% yield; 97.4% purity (AETC by HPLC, retention time = 1.6 minutes); 0.24% residual ethyl acetate). 1H-NMR (400 MHz, DMSO): 5=8.55 (s, 2H), 7.36 (d, 1H), 7.69 (d, 1H). A second batch of compound 2A was prepared by the same method to produce 3.29 kg (95.4% purity, 82.3% yield, 0.11% residual ethyl acetate).

[00111] Step 2: Synthesis of Compound 3A

C! , N


M

K2CO3 (I .O equiv)

2A OH

DME/H20 3:1 (20 vol), 50 e C 3A N

[00112] Compound 2 A (2.91 kg in 20.5 kg solution) was added into a 100

L reactor at room temperature under nitrogen. DME (45 mL), 2-chloropyrazin (1.42 kg,

12.4 mol., 1.0 eq.), and Pd(dppf)Cl2 (10% w/w, 291 g) were added sequentially, and each

mixed at room temperature under nitrogen. Nitrogen was bubbled into the mixture for 20

minutes and the resulting mixture was purged and filled with nitrogen (3 times). The

mixture was heated to 48-52°C over 60 minutes. K2CO3 (2.57 kg, 18.6 mol, 1.5 eq.) was

added to 22 L of water in another reactor at room temperature and then added dropwise to

the compound 2 A mixture over 10 minutes. The mixture was stirred at 48-52°C for 16

hours and then cooled to room temperature. This procedure was repeated twice and all

three batches were combined.

[00113] An aqueous solution of K2CO3 (1.0 kg) was dissolved in 22 L of

water and added to the combined mixture to adjust the pH to 9. TBME (50 L) was added

into the mixture and filtered (PET filter, 3-5 pm, 205g/m2) to remove about 50 g of

sticky, brown solid material (catalyst analog). The aqueous layer was twice separated and

extracted with TBME (40 L, 40L).

[00114] The aqueous layer was combined with the aqueous layer of a

fourth batch prepared according to the above method. The pH of the combined aqueous layers was adjusted to pH<3 with HC1 (2N, 48 L). The solid precipitated out slowly as

the mixture was stirred at room temperature for 1 hour. The mixture was filtered (PET

filter, 3-5 pm, 205g/m2) over 30 minutes to obtain 20 kg of wet product. ACN (40 L) was

added into a 100 L reactor equipped with an overhead stirrer at room temperature. The 20

kg of wet product was added into the reactor and the reaction mixture heated to reflux

and stirred at reflux for 4 hours. The reaction mixture was cooled to room temperature

over 3 hours (around 15 °C/hour) and filtered to obtain 8.5 kg of wet solid. The wet solid

was dried under vacuum (20-30 mmHg) at 50-55 °C for 15 hours to obtain compound 3 A

as a pale white solid (6.1 kg; 97.4% purity (AUC by HPLC, retention time = 3.7

minutes); 83.8% yield). 1H-NMR (400 MHz, DMSO): 5=7.67 (d, 1H), 7.82 (d, 1H), 8.75

(d, 1H), 8.82 (t, 1H), 8.98 (d, 1H), 13.89 (bs, 1H).

[00115] Step 3: Synthesis of compound 6A

3A 6A N

N

[00116] Compound 3 A (6.1 kg, 22.7 mol, 1.0 eq.) was added into a 100 L

reactor equipped with a temperature probe, overhead stirrer, and condenser. Methanol

(92 L) was added into the reactor at room temperature. The mixture was cooled to

0-10 °C and added with SOCk (5.4 kg, 45.3 mol, 2.0 eq.) dropwise at 0-10 °C over 30

minutes. The reaction mixture was heated to reflux (65 °C) and stirred at reflux for 15

hours. A suspension was formed. Most of the solvent and SOCk was removed under

vacuum distillation until about 30 L remained. The mixture was concentrated under

vacuum (30-40 mmHg) at 50-55 °C for about 6 hours. Water (10 L) was added to the residual at -5 to 15 °C. The pH was adjusted to 8-9 with an aqueous solution of K2CO3 (200 g, dissolved in 2L of water) at -5 to 15 °C. The resulting aqueous layer was extracted twice with isopropyl acetate (25 L, 25 L). The combination of organic layers (about 50 kg) was washed with 20 L of NaHCCb aqueous layer. The organic layer was separated and washed with 10 L of of an aqueous solution of NaHCCb. All the aqueous layers were combined (55.8 kg). The organic layer was filtered through a silica pad (30 cm) and the pad washed with extra isopropyl acetate until the compound 6 A was filtered from the silica gel (about 3 hours). The organic layer was concentrated to about 5 L. THF (10 L) was added to the residual and concentrated to about 5 L (3 times) under vacuum (30-40 mmHg) at 50-55 °C for about 3 hours. Another 10 L of THF was added to the residual concentrate, giving a concentrated solution of compound 6A (15.8 kg; 32.83%,

5.19 kg compound 6A in solution; 97.9% purity (AUC by HPLC, retention time = 8.5 min); 80.8% yield). 1H-NMR (400 MHz, DMSO): 5=3.98 (s, 3H), 7.54 (d, 1H), 7.78 (d, 1H), 8.63 (d, 1H), 8.72 (t, 1H), 8.94 (d, 1H).

[00117] Step 4: Synthesis of compound 6B

[00118] THF (26 L) was added into a 100 L reactor equipped with a temperature probe and overhead stirrer under nitrogen. DIBAL-H (26 kg, 46 mol, 5.0 eq.) was added and the system purged and filled with nitrogen three times. The mixture was cooled to -78 to -70 °C to form solution A. A room temperature solution of compound 6A (2.6 kg, 9.2 mol, 1.0 eq.) in 52 L of THF was added dropwise at -78 to -70 °C over 30 minutes under nitrogen. The mixture was warmed to -30 °C over about 5-6 hours. The reaction mixture was stirred at -40 to -30 °C for 30 minutes. The mixture was slowly added to 42 L of 2N HCL over 1 hour reaching a maximum temperature of 35 °C. The mixture was extracted with 26 L of isopropyl acetate. The organic layer was separated and washed with 30 L of brine. This procedure was repeated and both batches of organic layer were combined and concentrated from about 100 L to about 5-10 L under vacuum.

A solid slowly formed during concentration. The mixture was cooled to 5-15 °C and stirred for 1 hour. The mixture was filtered (30-50 pm) over 30 minutes. The solid was dried under vacuum at 50 °C for 6 hours to obtain compound 6B as a brown solid (2.1 kg; 97.5% purity (AUC by HPLC, retention time = 8.6 min); 45.7% yield). 1H-NMR (400 MHz, DMSO): d = 4.65 (d, 2H), 5.68 (t, 1H), 7.62 (d, 1H), 7.68 (d, 1H), 8.72 (d, 1H),

8.80 (t, 1H), 8.94 (d, 1H).

[00119] Step 5: Synthesis of compound 7

[00120] DMSO (10 L) was added to a 50 L flask equipped with a temperature probe and overhead stirrer under nitrogen at room temperature. Compound 6B (2.05 kg, 8.04 mol, 1.0 eq.) was added under nitrogen at room temperature. Et3N (8 L) was added under nitrogen at RT and the mixture was then cooled to 15-20 °C.

SCb. pyridine (5.1 kg, 32.08 mol, 4.0 eq.) was dissolved into 10 L of DMSO at 5-15 °C in a separate flask and added to the mixture dropwise over 3.5 hours at about 20 °C. The reaction mixture was transferred to 70 L of ice-water. The suspension mixture was stirred at 0-10 °C for 1 hour and filtered (PET, 3-5 pm, 205 g/m2) by centrifuge over 1.5 hours to obtain compound 7 as a brown solid. The solid was dissolved in 35 L of DCM at room temperature. The resulting DCM layer was washed with 5 L of brine. The organic layer was separated and concentrated under vacuum at 40-45 °C to dryness to obtain compound 7 as a brown solid (2.33 kg; 96.3% purity (AEiC by HPLC, retention time = 9.2 minutes); 93.5% yield). 1H-NMR (400 MHz, DMSO): d = 7.67 (d, 1H), 7.99 (d, 1H), 8.67 (d, 1H), 8.75 (s, 1H), 8.99 (d, 1H), 10.56 (s, 1H).

[00121] Step 6: Synthesis of compound 8

[00122] THF (23 L) was added to a 50 L flask equipped with a temperature probe and overhead stirrer under nitrogen at room temperature. Compound 7 (2.3 kg, 9.1 mol, 1.0 eq.) and (S)-2-methylpropane-2-sulfmamide (1.21 kg, 10 mol, 1.1 eq.) were added sequentially to the flask under nitrogen. Ti(OEt)4 (6.22 kg, 27.3 mol, 3.0 eq.) was added dropwise to the flask over 1 hour at 30-35 °C under nitrogen. The system was purged with nitrogen three times and then the mixture was stirred at room temperature for 2 hours. Isopropyl acetate (40 L) was added to the reaction mixture. The entire reaction mixture was then charged to 20 L of brine while stirring slowly at RT. A lot of solid was formed and no heat release was observed. The solid (about 18 kg) was filtered using centrifuge, and then the solid was slurried with 20 L of isopropyl acetate again for 20 minutes, and filtered again, resulting is slightly less solid (17.3 kg). The filtrates were then combined and washed with 20 L of brine. The organic layer was separated and concentrated in a rotavapor under vacuum (30-40 mmHg) at 40-50 °C for about 4 hours to remove the solvents and obtain a brown oil (compound 8). The oil was dissolved in DMF to obtain a black solution (7.36 kg; 40.1%; 3.0 kg compound 8 in solution; 92.1% purity (AUC by HPLC, retention time = 9.7 minutes); >100% yield). 1H-NMR (400 MHz, CDCb): d = 1.30 (s, 9H), 7.59 (d, 1H), 8.11 (d, 1H), 8.64 (s, 1H), 8.73 (m, 1H), 8.97 (s, 1H), 9.10 (s, 1H).

[00123] Step 7: Synthesis of compound 11

O

S

10 s C

8

11 N

[00124] DMF (26 L, 10 v/w) was added to a 50 L flask equipped with a temperature probe and overhead stirrer under nitrogen at 15 °C. Compound 8 (7.3 kg of

DMF solution, containing 2.9 kg, 8.1 mol, 1.0 eq.) and TBAA (2.44 kg, 8.1 mol, 1.0 eq.) were added sequentially to the flask under nitrogen. The mixture was cooled to 0-10 °C.

TMSCF3 (2.88 kg, 20.3 mol, 2.5 eq.) was then added to the flask over 60 min at 0-10 °C.

The reaction mixture was stirred at 0-5 °C under nitrogen protection for 3 hours.

Isopropyl acetate (60 L) was added to the mixture, followed by the addition of 45 L of

NaHCCb under stirring at 5-25 °C. The organic layer was separated, washed three times with NaHC03 (30 L x 3), and concentrated from 60 kg to 2.5 kg of brown oil. The oil product was dissolved in 20 L of TBME and filtered through a pad of silica gel (about 40 cm high, 30 cm diameter) over 2 hours to obtain 2.14 kg of compound 1 1 in TBME solution. The solution was concentrated at 45-50 °C to dryness to obtain compound 1 1 as a black oil (1.85 kg; 85.2% purity (AETC by HPLC, retention time = 9.1 minutes, 9.6 minutes for diastereoisomer); 53.6% yield). 1H-NMR (400 MHz, CDCh): d = 1.33 (s, 9H), 3.82-3.85 (d, 1H), 5.61-5.66 (m, 1H), 7.53-7.60 (m, 2H), 8.63-8.64 (d, 1H), 8.71-8.72 (m, 1H), 8.95 (s, 1H).

[00125] Step 8: Synthesis of compound 12 (free base)

[00126] Compound 1 1 (1.8 kg, 4.23 mol, 1.0 eq., crude) was added to a 50 L reactor equipped with a temperature probe and overhead stirrer under nitrogen at 25 °C. Anhydrous MeOH (18 L) was added to dissolve compound 1 1. Then MeOH/HCl (18 L, 1 N) was added dropwise at 25-30 °C over 10 minutes and the mixture was stirred at 25-30 °C for 1 hour. Water (15 L) was added to the reaction and the mixture concentrated in a rotavapor under vacuum (30-40 mmHg) at 45-50 °C for about 4 hours to remove the solvent. The pH of the mixture was adjusted to 10 with 5 L of K2CO3 solution. 20 L of EtOAc was then added to the mixture and the organic layer was separated and the aqueous layer extracted twice with EtOAc (15 L x 2). The organic layers were combined and washed with 10 L of brine. The combined organic layers contained 996 g of

compound 12 in 40 kg of EtOAc solution (84% purity (AUC by HPLC, retention time =

2.8 minutes). The organic layers were concentrated in a rotavapor under vacuum (30-40 mmHg) at 45-50 °C for about 3 hours to a 7.5 kg volume of compound 12 in EtOAc solution (83% purity (AETC by HPLC, retention time = 2.7 minutes).

[00127] In a separate 50 L reactor equipped with a temperature probe and overhead stirrer, D-CSA was added (930 g, 4.0 mol, 1.0 eq. to 1.26 kg compound 12) and stirred at room temperature under nitrogen. EtOAc (10 L) and then the EtOAc solution of compound 12 (1.26 kg, 3.9 mol, 1.0 eq.) were each sequentially added to the reactor. The mixture was stirred at room temperature for 1 hour and slowly became a suspension. The mixture was filtered by centrifuge and washed with EtOAc to produce 2.3 kg of compound 12 as an off-white solid (96.0% purity).

[00128] The solid product, 20 L of EtOAc, and 10 L of 10% aqueous K2CO3 were added sequentially to a 50 L flask and stirred at room temperature until no solid remained (pH = 9-10). The organic layer was separated and the aqueous layer extracted twice with EtOAc (10 L x 2). The organic layers were combined (about 32 kg) and washed with 10 L of brine. The organic layer contained 716 g of compound 12 in

31.8 kg of solution.

[00129] The organic layer was concentrated under vacuum at 45-50 °C to about 8 L. Activated carbon (200 g) was added to the organic layer and the mixture stirred at 60-70 °C for 1 hour, cooled to room temperature, and filtered using a Buchner funnel and filter paper (pore size: 30-50 pm) over 30 minutes to remove the activated carbon. The mixture was concentrated in a rotavapor under vacuum (30-40 mmHg) at 45-50 °C for about 3 hours to yield 710 g of compound 12 as a yellow solid (99.4% purity). [00130] D-CSA (410 g, 1.77 mol, 1.0 eq. to 680 g compound 12), 3.4 L iPrOH, and 68 mL of water were added sequentially to a 10 L reactor equipped with a temperature probe and overhead stirrer and stirred at room temperature under nitrogen. The mixture was heated to reflux (84 °C) to form solution A after 1 hour. Compound 12 (680 g) was dissolved in 3.4 L of iPrOH and added into solution A for one partition. A clear solution was formed and the temperature decreased to 65 °C. The mixture was stirred at 65 °C for about 15 minutes after which a solid appeared. The mixture was cooled to 10 °C over 2 hours, stirred at 10 °C for an additional 30 minutes, and filtered through a Buchner funnel and filter paper (pore size: 30-50 pm) over 30 minutes to collect the 1.1 kg of white solid.

[00131] EtOAc (10 L), 1.1 kg of white solid product, and 5 L of 10% K2CO3 were added sequentially to a 20 L flask and mixed for 5 minutes. The solid dissolved (pH = 9-10). The EtOAc layer was separated and the aqueous layer extracted twice with EtOAc (5 L each). The organic layers were combined (about 20 L), washed with 5 L of brine, and concentrated in a rotavapor under vacuum (30-40 mmHg) at 45-55 °C for about 3 hours to remove most of the solution and until the residue weight reached 1 kg. Heptanes (1 L) was added to the mixture and stirred at room temperature for 30 minutes. The mixture was filtered using a Buchner funnel and filter paper (pore size: 30-50 pm) over 30 minutes to obtain 419 g of compound 12 base as a white solid (99.7% purity). The filtrate was concentrated to 135 g of compound 12 as a yellow solid (98.7% purity). 1H-NMR (400 MHz, CDCh): d = 1.85 (bs, 2H), 5.17 (m, 1H), 7.56 (d, 1H), 7.68 (d, 1H), 8.62 (d, 1H), 8.70-8.71 (m, 1H), 8.93 (s, 1H). Combined, the products resulted in a 40.7% yield of compound 12.

[00132] Step 9: Synthesis of compound 10

10A 10

[00133] Pd/C (40 g, 5% w/w) was added into a 10 L autoclave reactor at room temperature under nitrogen. THF (2 L), 2 L of methylamine (27%-30% alcoholic solution, 2.1 eq.), and 800 g of compound 10A (7 mol, 1.0 eq.) were sequentially added into the reactor. The system was purged with hydrogen three times. The mixture was stirred at hydrogen pressure (50 psi) at 70-75 °C overnight and was then filtered using a Biichner funnel and filter paper (pore size: 30-50 pm) over 10 minutes to remove the Pd/C. The filtrate was concentrated in a rotavapor under vacuum (30-40 mmHg) at 45-50 °C for about 3 hours to obtain 933 g of yellow oil. The mixture was distilled without a column at atmospheric pressure and the 140-170 °C portion was collected to obtain 763 g of compound 10 as a colorless oil (98.6% purity (AUC by HPLC, retention time = 4.8 minutes); 84.2% yield; 8000 ppm residual ethanol). A portion of the oil (563 g) was distilled using a 3 cm column at atmospheric pressure and the 140-170 °C portion was collected to obtain 510 g of compound 10 (75.8% yield; 134 ppm residual ethanol). 1H-NMR (400 MHz, CDCb): d = 0.82 (bs, 1H), 1.10-1.12 (q, 2H), 1.66 (d, 2H), 1.73-1.81 (t, 2H), 2.05 (s, 3H), 2.08-2.19 (m, 1H), 2.22 (s, 3H), 2.60 (d, 2H).

[00134] Step 10: Synthesis of HM04 fumarate salt

[00135] DCM (1L), 200 g CDI (1.23 mol, 2.0 eq.), and 35 g DABCO (0.31 mol, 0.5 eq.) were sequentially added into a 3 L reactor equipped with a temperature probe and overhead stirrer, and stirred at room temperature under nitrogen. The mixture was cooled to -10 to -5 °C. Compound 12 (200 g) was dissolved in 1 L of DCM and added into the mixture dropwise over 1 hour, followed by stirring for 16 hours at -10 to -5 °C. Compound 10 (159 g, 1.24 mol, 2.0 eq.) was added at -10 to 0 °C over 10 minutes. The mixture was then warmed to 0 to 5 °C and held for 2 hours. The mixture was concentrated under vacuum at 40-45 °C to about 1 L. HC1 (1 L of 1 N) was added to the residual and concentrated in a rotavapor under vacuum (30-40 mmHg) at 45-50 °C for about 2 hours to remove the DCM. Another 3 L of 1N HC1 was added to the residual and extracted three times with TBME (4 L, 2 L, 2 L). The aqueous layer was slowly adjusted to pH = 9-10 with 20% aqueous K2CO3 (about 1.5 L) and extracted with DCM (2 L x 3). The organic layers were combined (about 4 L) and washed three times with 0.25 N KH2PO4 (1.2 L x 3). The organic layer was washed with 2 L of brine to bring the pH to neutral and concentrated in a rotavapor under vacuum (30-40 mmHg) at 45-50 °C for about 2 hours to 450 g (335 mL). MTBE (1.5 L) was added to the residual and distilled until 500 mL of liquid was collected. This step was repeated four times with the addition of 500 mL of TBME and collection of 500 mL of distillate, with the exception that 330 mL of liquid was collected at the final distillation. About 1 to 1.2 L of residual remained in the flask. The residual was slowly cooled to room temperature and stirred at room temperature overnight. The mixture was filtered, washed twice with TBME (400 mL x 2), and dried to obtain 192 g of HM04 free base a light yellow solid (99.3% purity (AUC by HPLC, retention time = 11.0 minutes). The product on the wall was dissolved in DCM and concentrated under vacuum to obtain 22 g of HM04 free base as a brown sticky oil (97.6% purity). The filtrate was concentrated under vacuum to obtain 22.5 g of yellow solid (94.0% purity).

[00136] HM04 free base (187 g, 0.39 mol, 1.0 eq., 99.3% purity) and 1.9 L of ACN were sequentially added to a 3 L flask equipped with a temperature probe and overhead stirrer and stirred at 15 °C under nitrogen to obtain a light-yellow suspension. Fumaric acid (45.6 g, 0.39 mol, 1.0 eq.) was added to the flask and generated a white suspension after 1 minute. The reaction suspension was stirred overnight at room temperature, filtered (15-20 pm, ash<0.l5), washed twice with ACN (50 mL x 2), and dried under vacuum at 50 °C for 6 hours to obtain 207 g of HM04 fumarate salt as a light yellow solid (99.4% purity (AUC by HPLC, retention time = 11.1 minutes); 57.8% yield; 3100 ppm residual ACN). The filtrate was concentrated under vacuum to obtain 20.1 g of HM04 fumarate salt as a light yellow solid (97.3% purity).

[00137] A portion of the product (117 g) was further dried in a vacuum oven (20-40 mmHg) to lower the residual acetonitrile content. After drying at 60 °C for 6 hours, 15 hours, and 72 hours; and at 65 °C for 18 hours, the residual acetonitrile content was measured as 3100 ppm, 2570 ppm, 1300 ppm, and 256 ppm, respectively. After the drying process, 98 g of HM04 fumarate salt was isolated (99.4% purity (AUC by HPLC, retention time = 11.0 minutes); 1H-NMR (400 MHz, DMSO): d = 1.49-1.58 (m, 2H),

1.81-1.92 (m, 2H), 2.44-2.53 (m, 5H), 2.78 (s, 3H), 3.12 (m, 2H), 4.06-4.13 (m, 1H), 6.36-6.41 (m, 1H), 6.55 (s, 2H), 7.47 (d, 1H), 7.73 (d, 1H), 8.11 (d, 1H), 8.75 (d, 1H),

8.81-8.82 (m, 1H), 8.99 (d, 1H). The yield of 98g of HM04 fumarate salt isolated after drying the partial batch was extrapolated over the whole batch to calculate an

approximate yield of 48% for step 10.

[00138] XRPD analysis of HM04 fumarate salt products obtained after drying at 60 °C for 6 hours, 15 hours, and 72 hours; and at 65 °C for 18 hours was performed (see Figures 6-9, respectively). The XRPD profile showed that the HM04 fumarate salt product was consistent with Form 1.

Example 6. Streamlined Synthesis of HM04 Fumarate Salt Form 1

[00139] The overall yield of HM04 fumarate salt produced using Step 10 of Example 5 was calculated as approximately 48%. In order to increase the overall yield, a streamlined synthesis was investigated that eliminated the step of isolating HM04 free base. In particular, step 10 of the method of Example 5 shown in Figure 5 was changed. An overview of the streamlined synthesis beginning after step 9 of Example 5 is shown in Figure 10.

[00140] Streamlined HM04 Fumarate Salt Trial 1 : PCM (121.4 g). CPI (20.0 g, 123 mmol, 2 eq.) and DABCO (3.5 g, 31 mmol) were sequentially added into an inertized 1 L reactor. The mixture was cooled to -10 °C. Separately, a solution of DCM (132.5 g) and compound 12 (20.0 g, 62.1 mmol) were charged into a vessel and stirred until a solution was obtained. This solution was dropped into the 1 L reactor over 33 minutes by keeping the internal temperature at -10 to -5 °C. At the end of the addition, the vessel was rinsed with DCM (7.0 g), which was then added to the reaction mixture.

After stirring overnight (19 hours) and positive IPC, compound 10 (15.9 g, 124 mmol, 2 eq.) was added over 15 minutes and the vessel rinsed with DCM (9.0 g). After heating at 0 °C, 1 hour of stirring, positive IPC, and a further 1.5 hours of stirring, the mixture was heated at room temperature and charged with water (200.1 g). The aqueous layer was separated and the organic layer extracted twice with 1 N HC1 (201, 200 g). The combined aqueous layers containing the product were washed with TBME (148 g). After removal of the organic layer, the aqueous layer was charged with DCM (265.0 g) and 50% K2CO3 solution (about 240 ml) until reaching pH 9.61.

[00141] Meanwhile, a solution of KH2PO4 (8.2 g) in water (240 g) was prepared. The organic layer containing the product was charged with the KH2PO4 solution until reaching pH 7.12 (142.2 g). After separation of the aqueous layer, the organic layer was washed with water (200 g). After separation of the aqueous layer, the organic layer was evaporated at 50 °C. ACN (314.4 g) was added and the solvent distilled again at 70-75 °C under vacuum. ACN (235.8 g) was added and the solvent distilled again under vacuum. ACN (141.5 g) was added, the resulting solution polish filtered and the filter washed with ACN (16 g). After heating at 60 °C, fumaric acid (7.2 g, 62 mmol) was added to the solution, causing a white precipitate. After cooling to 20 °C over 1 hour, the suspension was filtered and washed twice with TBME (2 x 30 g). After drying on the filter with nitrogen flow, 70.7 g of wet raw product was obtained. This was slurried with TBME (177.0 g) for 1 hour, filtered, and washed with TBME (70 g). After drying on the filter under nitrogen flow, 33.0 g of wet product was obtained. Heating at 50 °C under vacuum afforded the dry product as a white powder of HM04 fumarate salt (21.1 g;

Patent ID Title Submitted Date Granted Date
US9926337 SUBSTITUTED ASYMMETRIC UREAS AND MEDICAL USES THEREOF 2016-12-02
US9546157 p-Substituted Asymmetric Ureas and Medical Uses Thereof 2015-03-06 2015-09-10

////////////HM04, H0900, Helsinn,  Novo Nordisk, PRECLINICAL, obesity, Prader-Willi syndrome, ghrelin

CN(C1CCN(C)CC1)C(=O)N[C@H](c3ccc(c2cnccn2)c(Cl)c3Cl)C(F)(F)F

TL 487

$
0
0

str1

TL-487

CAS  1469746-55-1
2-Butenamide, N-[3-cyano-7-ethoxy-4-[(4-phenoxyphenyl)amino]-6-quinolinyl]-4-(dimethylamino)-, (2E)-
Molecular Weight, 507.58, MF C30 H29 N5 O3

Teligene Inc(2E)-N-[3-Cyano-7-ethoxy-4-[(4-phenoxyphenyl)amino]-6-quinolinyl]-4-(dimethylamino)-2-butenamide

(E)-N-(3-cyano-7-ethoxy-4-((4-phenoxyphenyl)amino)quinolin-6-yl)-4-(dimethylamino)but-2-enamide

Maleate in anhydrous or monohydrate CAS, 2326561-36-6, AND 2326561-38-8 form are BTK and HER-2 kinase inhibitor useful for treating cancer

Useful for treating breast cancer, ovary cancer and colon cancer. are BTK and HER-2 kinase inhibitor useful for treating cancer.

Anticancer protein kinase inhibitor

The compound was originally claimed in WO2013152135 , and may provide the structure of TL-487 , a small molecule inhibitor to HERs, being investigated by Teligene for the treatment of breast cancer; in July 2016, the company intended to develop the product as a class 1.1 chemical drug in China.

PATENT

US 20150057312

PATENT

WO2013152135

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2013152135&tab=PCTDESCRIPTION&queryString=%28ET%2Fkinase%29+&recNum=8&maxRec=4574

PATENT

WO-2019096327

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019096327&redirectedID=true

Novel crystalline maleate salt of (E)-N-(3-cyano-7-ethoxy-4-((4-phenoxyphenyl)amino)quinolin-6-yl)-4-(dimethylamino)but-2-enamide (first disclosed in WO2013152135) and its hydrates (monohydrate) and anhydrates, process for its preparation, composition comprising it and its use for treating cancers such as breast cancer, ovary cancer, colon cancer, prostate cancer, kidney cancer, bladder cancer, stomach cancer, lung cancer, mantle cell lymphoma and multiple myeloma are claimed. The compound is disclosed to be an irreversible inhibitor to BTK and Her-2 (also known as Erb-2 or neu).

(E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide is mentioned in WO2013152135 and corresponds to the compound of the Formula I:
Formula I
Compounds derived from 3-cyanoquinoline have been shown to have anti-tumor activity, which may make them useful as chemotherapeutic agents in treating various cancers, including but not limited to, pancreatic cancer, melanoma, lymphatic cancer, parotid tumors, Barrett’s esophagus, esophageal carcinomas, head and neck tumors, ovarian cancer, breast cancer, epidermoid tumors, cancers of major organs, such as kidney, bladder, larynx, stomach, and lung, colonic polyps and colorectal cancer and prostate cancer. Examples of compounds derived from 3-cyanoquinoline are disclosed and shown to possess anti-tumor activity in many literatures. One limitation of certain 3-cyanoquinoline compounds is that they are not water soluble in a free base form.
The crystalline form of a particular drug as a salt, a hydrate and/or any polymorph thereof is often one important determinant of the drug’s ease of preparation, stability, water solubility, storage stability, ease of formulation and in-vivo pharmacology. It is possible that one crystalline form is preferable over another where certain aspects such as ease of preparation, stability, water solubility and/or superior pharmacokinetics are deemed to be critical. Crystalline forms of (E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide salts that possess a higher degree of water solubility than the free base but are stable fulfill an unmet need for stable, crystalline, water-solubl
Example 1. (E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide sulfate
95%ethanol (4.0 ml) was added to (E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide (500 mg, 0.99 mmol, 1.0 eq) , followed sulfuric acid (101.9 mg, 1.04 mmol, 1.05 eq) in 95%ethanol (1.0 ml) was added dropwise to the reaction mixture. Then an amount of precipitate was founded. Another 95% (60 ml) was added to the reaction mixture and the reaction mixture was heated to 70℃. Filtered and the filtrate was heated to 70℃ again. Then the reaction mixture was cooled to room temperature and The reaction mixture was crystallized at -10℃ for 41.5h. Filtered the precipitated solid and dried at 40℃ under vacuum for 1 hour to get the title compound (260 mg) as a yellow solid.
X-ray detection shows an amorphous structure to the compound as FIG. 9.
Example 2. Synthesis of (E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide hydrochloride
95%ethanol (5.0 ml) was added to (E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide (500 mg, 0.99 mmol, 1.0 eq) , followed hydrochloric acid (38.0 mg, 1.04 mmol, 1.05 eq) in 95%ethanol (1.0 ml) was added dropwise to the reaction mixture. The reaction mixture was heated to 70℃. Filtered and the filtrate was crystallized under -10℃ for 44.5h. Filtered the precipitated solid and dried at 40℃ under vacuum for 1 hour to get the title compound (96 mg) as a yellow solid.
X-ray detection shows an amorphous structure to the compound in FIG. 6.
Example 3. Synthesis of (E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide malate
(E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide (500 mg, 0.99 mmol, 1.0 eq) , L-malic acid (139.4 mg, 1.04 mmol, 1.05 eq) and 95%ethanol (5.0 ml) was added to a 50 ml round-bottom flask. The reaction mixture was heated to 70℃. Filtered and the filtrate was crystallized under -10℃ for 45.5h. A little of precipitate was founded and then the reaction mixture was evaporated under vacuum at 40℃ to give the target (370 mg) as a yellow solid.
X-ray detection shows an amorphous structure to the compound in FIG. 8
Example 4: synthesis of (E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide citrate
To a solution of (E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide (500 mg, 0.99 mmol, 1.0 eq) , citric acid (198.8 mg, 1.04 mmol, 1.05 eq) and 95%ethanol (5.0 ml) . The reaction mixture was heated to 70℃. Filtered and the filtrate was crystallized under -10℃ for 45h. A little of precipitate was founded and then the reaction mixture was evaporated under vacuum at 40℃ to give the target compound (610 mg) as a yellow solid.
X-ray detection shows an crystalline structure to the compound in FIG. 7.
Example 5: Preparation of (E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide maleate monohydrate.
(E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide free base (0.091 kg) is rinsed with a 10%solution of USP purified water in n-propanol (0.082 kg, 0.10 L) followed by the addition of water: n-propanol solution (0.74 kg, 0.90 L) . Maleic acid is added (1.01 equiv) and the mixture is rinsed with 10%water: n-propanol (0.082 kg, 0.10 L) . The mixture is quickly heated to 50-60 ℃ and held for a minimum of 15 min. until a solution is obtained. The hot solution is clarified through a pre-heated 50-60 ℃, 0.2 Mm filter cartridge and the filtrates are collected in a preheated 45-55℃, 2 L multi-neck flask. The filter cartridge is rinsed through with 10%water: n-propanol pre-heated to 45-55 ℃ (0.082 kg, 0.10 L) . The solution is cooled over at least one hour to 40 ℃ and held at that temperature for 12 hours then cooled to room temperature (25 ℃) over a minimum of four hours and held at that temperature for at least two hours. The mixture is filtered on a 12.5 cm diameter Buchner funnel for 5 min., then rinsed and washed with prefiltered10%water: n-propanol solution (2 x 0.12 kg, 2 x 0.15 L) . The cake is dammed and suction maintained until dripping essentially stops, about 1 h.
PXRD is shown in FIG. 1.
Example 6: The product from Example 1 is dried (50 ℃, 10 mm Hg, 24 h) to give crystalline, anhydrous (E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide maleate.
PXRD is shown in FIG. 3.
Example 7: Preparation of (E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide maleate monohydrate.
To a solution of (E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide (38.0 g, 75.0 mmol, 1.0 eq) and n-propanol/H 2O (380 ml, V: V=9: 1) . maleic acid (8.7 g, 75.0 mmol, 1.0 eq) in n-propanol/H 2O (76 ml, V: V=9: 1) was added to the reaction mixture. An amount of precipitate was founded, then the reaction mixturewas heated to 65 ℃. The solid was dissolved completely, then the reaction mixture was cooled to room temperature and stand for 20 hours. Filtered and filtrate was evaporated under vacuum to get the crude product.
The crude product (14.0 g) was recrystallized in n-propanol/H 2O (240 ml, V: V=9: 1) at 70℃. The solid was dissolved completely, then the reaction mixture was cooled to room temperature and stand for 20.5 hours. Filtered and wash the cake with n-propanol/H 2O (20 ml, V: V=9: 1) to get target product (12.9 g, wet) .
PXRD as FIG. 1.
Example 8: crystalline, anhydrous (E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide maleate.
To a solution of (E) -N- (3-cyano-7-ethoxy-4- ( (4-phenoxyphenyl) amino) quinolin-6-yl) -4- (dimethylamino) but-2-enamide (21.5 g, 42.4 mmol, 1.0 eq) and ethanol (300 ml) . maleic acid (5.2 g, 44.8 mmol, 1.05 eq) was added to the reaction mixture. An amount of precipitate was founded, then the reaction mixture was heated to 70 ℃. Another ethanol (1980 ml) was added to the reaction mixture in several times and the reaction temperature was keep at 70 ℃. Filtered and filtrate was cooled to room temperature, stop stirring and stand for 16-20 hours. Filtered and the solid was dried at room temperature for 24 hours to get the title compound.

///////////////TL-487, PRECLINICAL, CHINA, breast cancer, ovary cancer, olon cancer,  BTK, HER-2 kinase inhibitor,

CN(C)C\C=C\C(=O)Nc3cc4c(Nc2ccc(Oc1ccccc1)cc2)c(cnc4cc3OCC)C#N

CS 3001

$
0
0

str1

CS-3001

BB 7, VX 033

CAS 2159116-56-8
Propanoic acid, 2-[[5-bromo-4-(3-cyclopropyl-5,5-difluoro-4,5,6,7-tetrahydrobenzo[c]thien-1-yl)-4H-1,2,4-triazol-3-yl]thio]-2-methyl-
Molecular Weight, 478.37

C17 H18 Br F2 N3 O2 S2

CStone Pharmaceuticals Co Ltd, JUNE 2018 IND FILED CHINA

URAT1 inhibitor – useful for treating hyperuricemia and gout.

The compound was originally claimed in WO2017202291 , covering thiophene derivative URAT1 inhibitors, useful for treating hyperuricemia and gouty arthritis, assigned to Medshine Discovery Inc , but naming the inventors.and has been reported in some instances to be a URAT1 modulator. In June 2018, an IND application was filed in

Uric acid is a product of the metabolism of terpenoids in animals. For humans, due to the lack of uric acid enzymes that continue to oxidatively degrade uric acid, uric acid is excreted in the human body as the final product of sputum metabolism through the intestines and kidneys. Renal excretion is the main pathway for uric acid excretion in humans. The upper limit of the normal range of uric acid concentration in the human body is: male 400 μmol/L (6.8 mg/dL) and female 360 μmol/L (6 mg/dL). Abnormal uric acid levels in the human body are often due to an increase in uric acid production or a decrease in uric acid excretion. Conditions associated with abnormal levels of uric acid include hyperuricemia, gout, and the like.
Hyperuricemia refers to a disorder in which the metabolism of substances in the human body is disordered, resulting in an increase or decrease in the synthesis of uric acid in the human body, and an abnormally high level of uric acid in the blood. Gouty arthritis refers to the fact that when uric acid is more than 7 mg/dL in human blood, uric acid is deposited as a monosodium salt in the joints, cartilage and kidneys, causing excessive reaction (sensitivity) to the body’s immune system and causing painful inflammation. The general site of attack is the big toe joint, ankle joint, knee joint and so on. Red, swollen, hot, and severe pain in the site of acute gout attacks, usually in the midnight episode, can make people wake up from sleep. In the early stages of gout, the attack is more common in the joints of the lower extremities. Hyperuricemia is the pathological basis of gouty arthritis. The use of drugs to lower blood uric acid concentration is one of the commonly used methods to prevent gouty arthritis.
In Europe and the United States, the onset of hyperuricemia and gout disease is on the rise. Epidemiological studies have shown that the incidence of gouty arthritis accounts for 1-2% of the total population and is the most important type of arthritis in adult males. Bloomberg estimates that there will be 17.7 million gout patients in 2021. In China, the survey showed that among the population aged 20 to 74, 25.3% of the population had a high blood uric acid content and 0.36% had gout disease. At present, clinical treatment drugs mainly include 1) inhibition of uric acid-producing drugs, such as xanthine oxidase inhibitor allopurinol and febuxostat; 2) uric acid excretion drugs, such as probenecid and benzbromarone; 3) Inflammation inhibitors, such as colchicine. These drugs have certain defects in treatment, poor efficacy, large side effects, and high cost are some of the main bottlenecks in their clinical application. It has been reported that 40%-70% of patients with serum uric acid levels do not meet the expected therapeutic goals (<6mg/dL) after receiving standard treatment.
As a uric acid excretion agent, its mechanism of action is to reduce the reabsorption of uric acid by inhibiting the URAT1 transporter on the brush-like edge membrane of the proximal convoluted tubule. Uric acid is a metabolite of sputum in the body. It is mainly filtered by glomerulus in the original form, reabsorbed and re-secreted by the renal tubules, and finally excreted through the urine. Very few parts can be secreted into the intestinal lumen by mesenteric cells. The S1 segment of the proximal convoluted tubule is a site of uric acid reabsorption, and 98% to 100% of the filtered uric acid enters the epithelial cells through the uric acid transporter URAT1 and the organic anion transporter OAT4 on the brush epithelial cell border of the tubular epithelial cells. The uric acid entering the epithelial cells is reabsorbed into the capillaries around the tubules via the renal tubular basement membrane. The S2 segment of the proximal convoluted tubule is the site of re-secretion of uric acid, and the amount secreted is about 50% of the excess of the small filter. The uric acid in the renal interstitial enters the epithelial cells first through the anion transporters OAT1 and OAT3 on the basal membrane of the tubular epithelial cells. The uric acid entering the epithelial cells passes through another anion transporter MRP4 on the brush border membrane and is discharged into the small lumen. The S3 segment of the proximal convoluted tubule may be a reabsorption site after uric acid secretion, and the amount of reabsorption is about 40% of the excess of the microsphere filtration, and similar to the first step of reabsorption, URAT1 may be a key reabsorption transporter. Therefore, if the urate transporter URAT1 can be significantly inhibited, it will enhance the excretion of uric acid in the body, thereby lowering blood uric acid level and reducing the possibility of gout attack.
In December 2015, the US FDA approved the first URAT1 inhibitor, Zurampic (Leinurad). The 200 mg dose was approved in combination with xanthine oxidase inhibitor XOI (such as Febuxostat, etc.) for the treatment of hyperuricemia and gouty arthritis, but the combination was compared with the xanthine oxidase inhibitor alone. The effect is not very significant. The Zurampic 400 mg dose was not approved due to significant toxic side effects at high doses (the incidence of renal-related adverse events, especially the incidence of kidney stones). Therefore, the FDA requires the Zurampic label to be filled with a black box warning to warn medical staff Zulampic of the risk of acute kidney failure, especially if it is not used in conjunction with XOI. If the over-approved dose uses Zurampic, the risk of kidney failure is even greater. high. At the same time, after the FDA asked for the listing of Zurampic, AstraZeneca continued its investigation of kidney and cardiovascular safety. Therefore, the development of a new type of safe blood-supplemented uric acid drug has become a strong demand in this field.
WO2009070740 discloses Leinurad, which has the following structure:
SYN
PATENT

WO-2019101058

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019101058&tab=FULLTEXT&maxRec=1000

Novel crystalline forms of URAT1 inhibitor (designated as Forms A and B) are claimed. The compounds are disclosed to be useful for treating hyperuricemia and gouty arthritis.

Novel crystalline forms of a URAT1 inhibitor, designated as Forms A and B, and their preparation.

Example 1: Preparation of a compound of formula (I)
synthetic route:
Step 1: Synthesis of Compound 2
In a three-necked flask (10 L), 4.5 L of dimethyl sulfoxide was added, and potassium t-butoxide (836.66 g, 7.46 mol, 2 eq) was added with stirring, and stirring was continued for 10 minutes until the dissolution was clear, and then cooled to an ice water bath. The internal temperature of the reaction solution was 20-25 °C. To the above solution, a solution of Compound 1 (500.05 g, 3.73 mol, 1 eq) in dimethyl sulfoxide (500 mL) was added dropwise, and the mixture was stirred for 30 minutes, and then carbon disulfide (283.86 g, 3.73 mol, 1 eq) was added dropwise thereto. ), after the completion of the dropwise addition, the reaction was stirred for 30 minutes. Further, ethyl bromoacetate (1250 g, 7.46 mol, 2 eq) was added dropwise thereto, and the mixture was stirred for further 2 hours. Finally, potassium carbonate (515.52 g, 7.46 mol, 1 eq) was added, and the temperature was raised to an internal temperature of 65 ° C, and the reaction was further stirred for 8 hours. After the reaction was completed, the reaction solution was cooled to room temperature. The reaction solution was diluted with ethyl acetate (10 L), and then 1M hydrochloric acid (2 L) and water (2 L) were added and stirred for 10 minutes, and the mixture was allowed to stand. The aqueous layer was separated and the organic phase was washed with water (2L×3). The combined aqueous layers were extracted with ethyl acetate (3L). All organic phases were combined and washed with saturated brine (2 L×2). The organic phase was dried over an appropriate amount of anhydrous sodium sulfate, and then filtered, and then evaporated. On the same scale, 6 batches were fed in parallel, and the combined black and red oily products were obtained. After the crude product was allowed to stand for 72 hours, a large amount of solid was precipitated, ethanol (2 L) was added thereto, stirred for 30 minutes, filtered, and the cake was collected and dried in vacuo to give Compound 2. . 1 H NMR (400 MHz, CDCl3 . 3 ) [delta]: 4.32 (Q, J = 7.2 Hz, 2H), 4.19 (Q, J = 7.2 Hz, 2H), 3.56 (S, 2H), 3.25 (T, J = 6.8Hz , 2H), 3.19 (t, J = 14.4 Hz, 2H), 2.26-2.17 (m, 2H), 1.37 (t, J = 7.2 Hz, 3H), 1.27 (t, J = 7.2 Hz, 3H); MS m/z = 364.8 [M+H] + .
Step 2: Synthesis of Compound 3
Compound 2 (241.00 g, 0.66 mol) was dissolved in ethanol (1 L) and placed in an autoclave (5 L), and Raney nickel (120 g) was added under argon atmosphere, followed by the addition of ethanol (2 L). The autoclave was charged and replaced with argon three times, then replaced with hydrogen three times, hydrogen was charged to a pressure of 2.0 MP in the autoclave, stirred and heated to an internal temperature of 85 ° C for 28 hours. The reaction was stopped, the reaction system was cooled to room temperature, the reaction solution was filtered, and the filter cake was washed three times with ethanol, 0.5 L each time. The filtrates were combined and then dried to give compound 3. . 1 H NMR (400 MHz, CDCl3 . 3 ) [delta]: 7.09 (S, IH), 4.26 (Q, J = 7.2 Hz, 2H), 3.20 (T, J = 6.8Hz, 2H), 3.12 (T, J = 14.4Hz , 2H), 2.20-2.10 (m, 2H), 1.30 (t, J = 6.8 Hz, 3H); MS m/z = 247.0 [M+H] + .
Step 3: Synthesis of Compound 4
Compound 3 (406.2 g, 1.65 mol, 1 eq) was dissolved in acetonitrile (6 L), then N-bromosuccinimide (1484.2 g, 6.60 mol, 4 eq) was slowly added, and the obtained reaction mixture was at 23 to 25 ° C. The reaction was stirred for 12 hours. After the reaction was completed, the reaction liquid was concentrated to about 1.0 L. The solid was removed by filtration, and a saturated solution of sodium hydrogensulfite (1 L) was added to the filtrate and stirred for 10 min. Add acid ethyl ester and extract three times, 2L each time. The organic phases were combined and dried over anhydrous sodium sulfate. The desiccant was removed by filtration, and the filtrate was concentrated under reduced pressure. Petroleum ether (3 L) was added to the residue, and the mixture was stirred at 30 ° C for 30 minutes. After filtration, the filter cake was washed 5 times with petroleum ether, 200 mL each time, until no product remained in the filter cake. Combine all the organic phases and spin dry to obtain a crude product. Petroleum ether (100 mL) was added to the crude product, stirred well, filtered, and filtered, and then dried in vacuo. . 1 H NMR (400 MHz, CDCl3 . 3) [delta]: 4.24 (Q, J = 7.2 Hz, 2H), 3.19 (T, J = 6.8Hz, 2H), 2.95 (T, J = 14.4Hz, 2H), 2.17-2.07 (m, 2H), 1.29 (t, J = 7.2 Hz, 3H).
Step 4: Synthesis of Compound 5
Compound 4 (340.21 g, 1.05 mol), cyclopropylboronic acid (108.12 g, 1.26 mol), anhydrous potassium phosphate (444.98 g, 2.10 mol), palladium acetate (12.03 g, 53.58 mmol) and 2-dicyclohexyl Phospho-2′,4′,6′-triisopropylbiphenyl (23.86 g, 50.05 mmol) was added to a mixed solvent of toluene and water (10:1, 3.4 L/340 mL), and the reaction flask was replaced with nitrogen. After that, place it in an oil bath. The reaction solution was heated at an internal temperature of 80 ° C, and the reaction was stirred at this temperature for 16 hours. After completion of the reaction, the reaction solution was cooled to room temperature, and tris-thiocyanic acid (6.51 g, suspended in ethanol (34 mL)) was added to the reaction mixture and stirred for 0.5 hour. On a similar scale (300.00 g of compound 4), 5 batches were fed in parallel and combined. After filtration, the organic phase was separated and the aqueous phase was extracted with ethyl acetate (250mL). The organic phases were combined and dried over anhydrous sodium sulfate. The desiccant was removed by filtration, and the filtrate was concentrated under reduced pressure to yield crude crude oil. After the crude product was allowed to stand for 20 hours, a yellow solid was precipitated, and petroleum ether (3 L) was added thereto and stirred for 1 hour. Filtration and drying of the filter cake in vacuo gave compound 5. . 1 H NMR (400 MHz, CDCl3 . 3 ) [delta]: 4.29 (Q, J = 7.2 Hz, 2H), 3.23 (T, J = 6.4Hz, 2H), 3.16 (T, J = 14.8 Hz, 2H), 2.24-2.18 (m, 2H), 1.95-1.85 (m, 1H), 1.35 (t, J = 6.8 Hz, 3H), 1.09-1.07 (m, 2H), 0.77-0.75 (m, 2H).
Step 5: Synthesis of Compound 6
Compound 5 (619.27 g, 2.16 mol) was added to a mixed solution of ethanol and water (3 L/3 L) of sodium hydroxide (173.55 g, 4.33 mol), and the reaction liquid was heated to an internal temperature of 60 ° C to stir the reaction 3 hour. After the reaction was completed, the reaction solution was cooled to room temperature. On a similar scale (750.17 g of compound 5), 1 batch was fed in parallel and combined. The combined reaction solution was extracted with petroleum ether (4 L). The organic phase was separated and the organic phase was backwashed twice with water (1.5L x 2). The aqueous phases were combined and concentrated under reduced pressure to remove ethanol. Water was added to the aqueous phase to dilute to 13 L, and then slowly added with dilute hydrochloric acid (3 M) to adjust to pH = 2, and a large amount of pale yellow solid precipitated. Filter and filter cake with water (3.0L x 2). After draining, the filter cake was collected and dried under vacuum at 60 ° C to give Compound 6. . 1 H NMR (400 MHz, DMSO-D . 6 ) [delta]: 12.79 (brs, IH), 3.23 (T, J = 14.8 Hz, 2H), 3.07 (T, J = 6.8Hz, 2H), 2.27-2.20 (m, 2H), 2.19-2.02 (m, 1H), 1.09-1.04 (m, 2H), 0.68-0.66 (m, 2H).
Step 6: Synthesis of Compound 7
Compound 6 (641.27 g, 2.48 mol), triethylamine (754.07 g, 7.45 mol) and diphenyl azide (1025.34 g, 3.73 mol) were added to t-butanol (6.5 L) with stirring. The reaction solution was heated in a 100 ° C oil bath for 16 hours. After the reaction was completed, it was cooled to room temperature. On a similar scale (650.00 g of compound 6), 4 batches were fed in parallel and combined. The reaction mixture was combined and concentrated under reduced pressure to remove t-butyl alcohol. The remaining black residue was dissolved with ethyl acetate (10L). Dry with an appropriate amount of anhydrous sodium sulfate. The desiccant was removed by filtration, and the filtrate was concentrated under reduced pressure to give a crude brown solid. Petroleum ether (8 L) was added to the crude product and stirred for 2 hours. After filtration, the filter cake was rinsed with petroleum ether (1 L) in portions, and the filter cake was vacuum dried in a vacuum oven at 60 ° C to obtain Compound 7. . 1 H NMR (400 MHz, CDCl3 . 3 ) [delta]: 6.31 (brs, IH), 3.11 (T, J = 14.8 Hz, 2H), 2.66 (T, J = 6.8Hz, 2H), 2.23-2.15 (m, 2H) , 1.82-1.75 (m, 1H), 1.51 (s, 9H), 0.94-0.90 (m, 2H), 0.68-0.65 (m, 2H).
Step 7: Synthesis of Compound 8
Compound 7 (1199.17 g, 3.64 mol) was added to ethyl acetate (2 L), and then stirred and then ethyl acetate (4L, 16. The reaction solution was reacted at 15 ° C for 2.5 hours, and then placed in a 40 ° C warm water bath to continue the reaction for 2 hours. After the reaction was completed, a large amount of dark red solid precipitated. Filter and filter cake was rinsed with ethyl acetate (2.0 L). The filter cake was dried under vacuum in a vacuum oven at 60 ° C to give compound 8. . 1 H NMR (400 MHz, DMSO-D . 6 ) [delta]: 3.17 (T, J = 14.8 Hz, 2H), 2.82 (T, J = 6.8Hz, 2H), 2.25-2.15 (m, 2H), 2.00-1.94 ( m, 1H), 0.99-0.95 (m, 2H), 0.58-0.54 (m, 2H); MS m/z = 229.8 [M+H-HCl] + .
Step 8: Synthesis of Compound 9
In a 3 L three-necked flask, Compound 8 (301.25 g) was added to tetrahydrofuran (600 mL), and the mixture was cooled to an internal temperature of 0 to 10 ° C under ice-cooling. Diisopropylethylamine (635.72 g) was added dropwise, and after completion of the dropwise addition, the ice water bath was removed, and the mixture was stirred at an internal temperature of 10 to 15 ° C for about 10 minutes. Filter and filter cake was washed with tetrahydrofuran (100 mL x 2). The filtrates were combined to give a solution A for use.
Tetrahydrofuran (2 L) was added to a 5 L reaction flask containing thiophosgene (257.48 g). The mixture was stirred and cooled to an internal temperature of 0 to 10 ° C in an ice water bath, and the solution A was slowly added dropwise thereto, and the dropwise addition was completed within about 5.5 hours, and stirring was continued for 10 minutes. After the reaction was completed, it was filtered, and the filter cake was washed with tetrahydrofuran (150 mL × 2). The filtrate was combined and concentrated under reduced pressure to remove solvent. Tetrahydrofuran (400 mL) was added to the residue, which was dissolved to give a solution B.
The hydrazine hydrate (112.94 g) was added to tetrahydrofuran (2.5 L), and the mixture was cooled to an internal temperature of 5 to 10 ° C under ice-cooling. Solution B was added dropwise, and the addition was completed for about 2 hours, and stirring was continued for 10 minutes. After the reaction was completed, the reaction was stopped. The ice water bath was removed, N,N-dimethylformamide dimethyl acetal (333.45 g) was added, and the mixture was heated to an internal temperature of 60 to 65 ° C, and the reaction was stopped after the heat retention reaction for 3 hours.
The reaction solution was dried to dryness, and ethyl acetate (2 L) and purified water (1L) were added to the residue, and the mixture was stirred. The pH was adjusted to 5-6 with 10% hydrobromic acid, stirring was continued for 5 minutes, and allowed to stand for 10 minutes. Dispense and separate the aqueous phase. The organic phase was washed with pure water (500 mL x 2). The combined aqueous phases were extracted with EtOAc (1 mL). The desiccant was removed by filtration, and the filtrate was concentrated to dryness to dryness. n-Heptane (2.0 L) and tert-butyl methyl ether (150 mL) were added to the crude product, and the mixture was stirred ( stirring speed 550 rpm) for 18 hours. Filter and filter cake was washed with n-heptane (150 mL). The filter cake was collected and the filter cake was dried under vacuum at 60 ° C to give compound 9. . 1 H NMR (400 MHz, CDCl3 . 3 ) [delta]: 7.82 (S, IH), 3.20 (T, J = 14.8 Hz, 2H), 2.74 (T, J = 6.8Hz, 2H), 2.28-2.10 (m, 2H) , 1.98-1.82 (m, 1H), 1.06-1.02 (m, 2H), 0.75-0.71 (m, 2H); MS m/z = 313.9 [M+H] + .
Step 9: Synthesis of Compound 10
Acetonitrile (3 L) was placed in a 5 L three-necked flask. Compound 9 (303.25 g) and potassium carbonate (261.83 g) were added first with stirring. Further, methyl 2-bromoisobutyrate (203.85 g) was added, and the reaction system was replaced with nitrogen, and then heated to an internal temperature of 60 to 65 ° C, and the reaction was kept for about 2 hours. After the completion of the reaction, the heating was stopped, and the mixture was naturally cooled to 15 to 20 ° C under stirring. Filter and filter cake was washed with ethyl acetate (100 mL x 3). The filtrate was combined and concentrated under reduced pressure to dryness. The crude product was purified by column chromatography (mobile phase: ethyl acetate / n-heptane = 1:5 to 2:1). . 1 H NMR (400 MHz, CDCl3 . 3 ) [delta]: 8.20 (S, IH), 3.68 (S, 3H), 3.19 (T, J = 14.4Hz, 2H), 2.57 (T, J = 6.8Hz, 2H), 2.22 -2.12 (m, 2H), 1.93-1.83 (m, 1H), 1.67 (s, 6H), 1.08-1.03 (m, 2H), 0.73-0.69 (m, 2H); MS m/z = 414.0 [M +H] + .
Step 10: Synthesis of Compound 11
Acetonitrile (3.17 L) was placed in a 5 L three-necked flask. Under stirring, compound 10 (317.22 g) and thiocarbonyldiimidazole (26.94 g) were added, and the mixture was stirred at 16 to 20 ° C for 5 minutes. N-bromosuccinimide (158.60 g) was added and stirred for about 30 minutes with heat. After the reaction was over, the reaction was stopped. Filtration and concentration of the filtrate under reduced pressure afforded crude crude. The crude product was purified by column chromatography (EtOAc:EtOAc:EtOAc This crude product was dissolved in ethyl acetate (3.50 L) and washed with purified water (700 mL×4). The organic phase was separated and the organic phase was dried over anhydrous sodium sulfate. The desiccant was removed by filtration, and the filtrate was concentrated to dryness to give Compound 11. . 1 H NMR (400 MHz, CDCl3 . 3 ) [delta]: 3.73 (S, 3H), 3.22 (T, J = 14.4Hz, 2H), 2.53 (T, J = 6.8Hz, 2H), 2.24-2.14 (m, 2H) , 1.95-1.91 (m, 1H), 1.71 (d, J = 4.4 Hz, 6H), 1.11-1.07 (m, 2H), 0.78-0.74 (m, 2H); MS m/z = 491.7 [M+H ] + ,493.7[M+H+2] + .
Step 11: Synthesis of a compound of formula (I)
Tetrahydrofuran (1.2 L) was added to a 5 L reaction flask, and Compound 11 (243.03 g) was added with stirring. After the solution was dissolved, pure water (1.2 L) was added, and then lithium hydroxide monohydrate (125.46 g) was added, and the mixture was stirred at 20 to 25 ° C for about 2.5 hours. After the reaction was completed, the reaction was stopped. The reaction solution was concentrated under reduced pressure at 40 ° C to remove organic solvent. Pure water (1 L) was added to the residue, and the mixture was extracted with t-butyl methyl ether (300 mL). The aqueous phase was placed in a 10 L three-necked flask and cooled to 5 to 10 ° C in an ice bath. The pH was adjusted to 2 to 3 with a 40% hydrobromic acid solution, and a large amount of a pale yellow solid precipitated. Stirring was continued for 30 minutes, and the pH was again measured to be 2-3. Stirring was continued for 20 minutes and filtered. The filter cake was washed with pure water (150 mL x 3). The filter cake was collected, pure water (1500 mL) was added, and the mixture was beaten at room temperature for 1 hour. After filtration, the filter cake was washed with pure water (150 mL × 2), and the filter cake was collected and dried under vacuum at 40 ° C for 3 hours to obtain a compound of the formula (I). . 1 H NMR (400 MHz, the CD . 3 the OD) [delta]: 3.27 (T, J = 15.6Hz, 2H), 2.60-2.47 (m, 2H), 2.27-2.17 (m, 2H), 2.10-2.03 (m, IH) , 1.68 (d, J = 1.2 Hz, 6H), 1.15.10.10 (m, 2H), 0.80-0.71 (m, 2H); MS m/z = 477.99 [M+H] + , 480.1 [M+H+ 2] + .
Example 2: Preparation of Form A of Compound of Formula (I)
The compound of the formula (I) (50 mg) was added to a glass bottle, and methanol (0.4 mL) was added thereto, followed by stirring to a suspension or a solution. The suspension sample was placed in a thermomixer (40 ° C), shaken at 40 ° C for 60 hours, and then centrifuged to collect a sample. The above-mentioned lysed sample was volatilized at room temperature, centrifuged, and the sample was collected. The above sample was dried in a vacuum oven (40 ° C) overnight, and its crystalline form was examined by XRPD to obtain a crystal form of the final product having a crystalline form of the compound of the formula (I).
The compound of the formula (I) (50 mg) was added to a glass bottle, and ethyl acetate (0.4 mL) was added and stirred to a suspension or a solution. The suspension sample was placed in a thermomixer (40 ° C), shaken at 40 ° C for 60 hours, and then centrifuged to collect a sample. The above-mentioned lysed sample was volatilized at room temperature, centrifuged, and the sample was collected. The above sample was dried in a vacuum oven (40 ° C) overnight, and its crystalline form was examined by XRPD to obtain a crystal form of the final product having a crystalline form of the compound of the formula (I).
Example 3: Preparation of Form B of Compound of Formula (I)
The compound of the formula (I) (50 mg) was added to a glass bottle, tetrahydrofuran (0.4 mL) was added, and the mixture was stirred to dissolve. The above-mentioned lysed sample was volatilized at room temperature, centrifuged, and the sample was collected. The collected sample was dried in a vacuum oven (40 ° C) overnight, and its crystalline form was examined by XRPD to obtain a crystalline form of the final product in the form of Form B of the compound of formula (I).
Example 4: Solubility test of Form A of the compound of formula (I)
1. Preparation of diluent and mobile phase
Diluent: Accurately measure 300mL of pure water and 100mL of pure acetonitrile, mix in a 1L glass bottle, ultrasonic degassing for 10 minutes and then set aside.
Mobile phase A: 0.1% phosphoric acid aqueous solution

For example, remove 2.0 mL of phosphoric acid into 2000 mL of water, sonicate for 10 minutes, mix, and let cool to room temperature as mobile phase A.

Mobile phase B: acetonitrile.
2. Preparation of the reference solution (using the A crystal form itself as a control sample)
Accurately weigh 5 mg of Form A, place it in a sample vial, add 10 mL of diluent, sonicate for 5 minutes, then cool to room temperature and mix well, and mark it as working reference solution STD-1.
Accurately weigh 5 mg of Form A, place it in a sample vial, add 10 mL of diluent, sonicate for 5 minutes, then cool to room temperature and mix well, and mark it as working reference solution STD-2.
3. Preparation of linear solution
The above working reference solution STD-1 was diluted 1 time, 10 times, 100 times, 1000 times and 2000 times, and recorded as linear solutions L1, L2, L3, L4 and L5.
4. Solubility test
Accurately weigh 6mg of A crystal form into 8mL glass bottle, then accurately add 3mL different solvent (0.1N hydrochloric acid solution, 0.01N hydrochloric acid solution, purified water, pH3.8 buffer solution, pH4.5 buffer solution, pH5 .5 buffer solution, pH 6.0 buffer solution, pH 7.4 buffer solution, pH 6.8 buffer solution), made into a suspension. A stir bar was added to the above suspension, and the mixture was thoroughly stirred at 37 ° C in the dark. After stirring, the solids in the pH 7.4 buffer solution and the pH 6.8 buffer solution were all dissolved, and 6 mg of the A crystal form was accurately weighed, added to the buffer solution, and thoroughly stirred again to prepare a suspension. After stirring for 4 hours and 24 hours, the sample was centrifuged, and the solution was filtered through a filter and the concentration thereof was measured by HPLC. The HPLC analysis method is shown in Table 3.
Table 3: HPLC analysis methods

////////////CS-3001, BB 7, VX 033, CHINA, PRECLINICAL, CStone Pharmaceuticals, URAT1 inhibitor,  hyperuricemia, gout

O=C(O)C(C)(C)Sc4nnc(Br)n4c2sc(c1CC(F)(F)CCc12)C3CC3

Viewing all 87 articles
Browse latest View live