AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

Amneal Pharma’s, 4,5-Dihydro-1H-pyrazolo[3,4-d]pyrimidine containing phenothiazines as antitubercular agents

 Uncategorized  Comments Off on Amneal Pharma’s, 4,5-Dihydro-1H-pyrazolo[3,4-d]pyrimidine containing phenothiazines as antitubercular agents
Jun 152016
 

STR1

Cas 1580464-40-9

MW458.97, C24 H19 Cl N6 S,

1H-​Pyrazolo[3,​4-​d]​pyrimidin-​6-​amine, 4-​(4-​chlorophenyl)​-​4,​7-​dihydro-​3-​methyl-​1-​(10H-​phenothiazin-​2-​yl)​-

4-(4-chlorophenyl)-3-methyl-1-(10H-phenothiazin-2-yl)-4,5-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-amine

4-(4-chlorophenyl)-3-methyl-1-(10H-Phenothiazin-2-yl)-4,5-dihydro-1H-pyrazolo[3,4-d] pyrimidin-6-amine

STR1

Yield 79%, m.p.: 186-188 ºC.

IR (KBr): 3328 (NH), 1648 (C-N), 640 (C-S-C). 1H NMR (300 MHz, CDCl3): d 2.32 (s, 3H, CH3), 4.95 (s, 1H, CH), 7.36-7.38 (dd, 2H, J=8.10 Hz), 7.84-7.87 (dd, 2H, J=7.80 Hz), 7.90-8.05 (m, 7H, Ar-H), 8.46 (s, 1H, NH), 8.56 (s, 2H, NH2), 9.11 (s, 1H, NH):

13C NMR (75 MHz, CDCl3): d 26.1, 41.2, 52.5, 59.8, 103.6, 104.2, 105.3, 114.2, 116.6, 122.7, 127.1, 127.9, 128.2, 128.6, 129.2, 132.5, 134.6, 142.4, 143.7, 155.3, 162.5. Mass (m/z): 459. Anal. (%) for C24H19ClN6S, Calcd. C, 62.81; H, 4.17; N, 18.31. Found: C, 62.75; H, 4.15; N, 18.26.

Mass spectrum of 4g

 STR1

1H NMR spectrum of 4g

STR1

 

 

 

 

Tuberculosis (TB) is a highly infectious airborne disease caused by the pathogenic bacterium Mycobacterium tuberculosis (Mtb). 1According to the latest World Health Organization (WHO) report, an estimated 8.6 million people developed TB and 1.3 million died from the disease (including 320,000 deaths among HIV-positive people) in 2012. The majority of cases worldwide in 2012 were in the South-East Asia (29%), African (27%) and western Pacific (19%) regions. India and China alone accounted for 26% and 12% of total cases, respectively.2 The standard antitubercular treatment regimen, termed DOTS (Directly Observed Therapy, Short-course), is based on the co-administration of age-old drugs like isoniazid (INH), rifampin (RMP), ethambutol (EMB), and pyrazinamide (PZA) for the first two months, followed by a prolonged treatment with INH and RMP for additional 4–7 months with no guarantee of complete sterilization from the infection. 4 and 5 Furthermore, emergence of new virulent forms of TB such as multi drug resistant (MDR-TB) and extremely drug resistant (XDR-TB), and its synergy with human immunodeficiency virus (HIV) has fuelled its epidemic nature.  These reasons make a compelling case for an urgent need for new and effective antitubercular agents with improved properties such as enhanced activity against MDR strains, reduced toxicity, rapid mycobactericidal mechanism of action and the ability to penetrate host cells and exert antimycobacterial effects in the intracellular environment.

Phenothiazines are important classes of compounds which have increasingly attracted attention, owing to their remarkable biological and pharmacological properties, such as antibacterial, antifungal, anticancer, antiviral, anti-inflammatory, antimalarial, antifilarial, trypanocidal, anticonvulsant, analgesic, immunosuppressive and multidrug resistance reversal. These activities are the results of the actions exerted by phenothiazines on biological systems via the interaction of the pharmacophoric substituent (in some cases of strict length), via the interaction multicyclic ring system (π–π interaction, intercalation in DNA) and via the lipophilic character permitting the penetration through the biological membranes to reach its site of action. Further, Phenothiazines have been shown to exhibit in vitro and in vivo activity against Mtb and multidrug-resistant Mtb. Some of the phenothiazine derived antipsychotic agents such as chlorpromazine, trifluoperazine (TPZ) and thioridazine are found to be effective inhibitors of Mtb.Phenothiazines are predicted to target the genetically validated respiratory chain component type II NADH:quinone oxidoreductase (Ndh)

 

 

 

Paper

Volume 24, Issue 6, 15 March 2014, Pages 1493–1495

4,5-Dihydro-1H-pyrazolo[3,4-d]pyrimidine containing phenothiazines as antitubercular agents

  • a Amneal Pharmaceuticals India Pvt Ltd, 882/1-871, Village Rajoda, Tal.: Bavla Dist.: Ahmedabad 382220, Gujarat, India
  • b Division of Medicinal Chemistry, Department of Chemistry (DST-FIST Sponsored), Mahatma Gandhi Campus, Maharaja Krishnakumarsinhji Bhavnagar University, Bhavnagar 364002, Gujarat, India
  • c Department of Chemistry, Saurashtra University, Kalawad Road, Rajkot 360005, Gujarat, India

A series of novel dihydropyrazolo[3,4-d]pyrimidine derivatives bearing a phenothiazine nucleus were synthesized in excellent yields via a modified Biginelli multicomponent reaction. The newly synthesized compounds were characterized by IR, 1H NMR, 13C NMR, Mass spectra and elemental analysis followed by antimycobacterial screening. Among all the screened compounds, compound 4g showed most pronounced activity against Mycobacterium tuberculosis (Mtb) with minimum inhibitory concentration (MIC) of 0.02 μg/mL, making it more potent than first line antitubercular drug isoniazid.

Synthetic protocol of title compounds 4a–k. Reagents and conditions: (a) NH2NH2, ...

Scheme 1.

Synthetic protocol of title compounds 4ak. Reagents and conditions: (a) NH2NH2, reflux; (b) ethyl acetoacetate, sodium ethoxide, reflux; (c) guanidine hydrochloride, aldehyde (R-CHO), P2O5, EtOH, reflux.

 

 

 

Amneal Pharmaceuticals’ co-CEO Chirag Patel

 

Chirag Patel and Chintu Patel

Chintu Patel, owner of Amneal Pharmaceuticals,

///////Dihydropyrazolo[3,4-d]pyrimidine, Phenothiazines, Biginelli multicomponent reaction, Cytotoxicity, Antitubercular activity, 4,5-Dihydro-1H-pyrazolo[3,4-d]pyrimidine,  phenothiazines, antitubercular agents, amneal, 1580464-40-9

Clc1ccc(cc1)C2N=C(N)Nc3c2c(C)nn3c4cc5Nc6ccccc6Sc5cc4

Share

GSK 1070916 For Advanced solid tumor

 phase 1, Uncategorized  Comments Off on GSK 1070916 For Advanced solid tumor
Jun 152016
 

 

GSK 1070916

NMI-900 , GSK-1070916, GSK-1070916A

4-[3-(4-N,N-Dimethylcarbamylaminophenyl)-1-ethyl-1H-pyrazol-4-yl]-2-[3-(dimethylaminomethyl)phenyl]-1H-pyrrolo[2,3-b]pyridine

N’-[4-[4-[2-[3-[(Dimethylamino)methyl]phenyl]-1H-pyrrolo[2,3-b]pyridin-4-yl]-1-ethyl-1H-pyrazol-3-yl]phenyl]-N,N-dimethylurea

CAS 942918-07-2,

MFC30H33N7O,

MW507.63

PHASE 1/II , Advanced solid tumor, Cancer Research Technology,

off-white solid.

1H NMR (400 MHz, DMSO-d6) δ ppm 12.14 (d, J = 1.8 Hz, 1H), 8.31 (s, 1H), 8.27 (s, 1 H), 8.07 (d, J = 4.8 Hz, 1H), 7.78 (d, J = 8.1 Hz, 1H), 7.77 (s, 1H), 7.43 (d, J = 8.6 Hz, 2H), 7.39 (d, J = 8.1 Hz, 1H), 7.27 (d, J = 8.6 Hz, 2H), 7.27 (dd, 1H), 6.79 (d, J = 5.1 Hz, 1H), 6.76 (d, J = 2.0 Hz, 1H), 4.27 (q, J = 7.3 Hz, 2H), 3.43 (s, 2H), 2.91 (s, 6H), 2.18 (s, 6H), 1.51 (t, J = 7.2 Hz, 3H).

MS m/z 508.4 [M + H]+. Anal. (C30H33N7O·1.0H2O) C, H, N.

GSK1070916 is a reversible and ATP-competitive inhibitor of Aurora B/C with IC50 of 3.5 nM/6.5 nM; displays >100-fold selectivity against the closely related Aurora A-TPX2 complex(IC50=490 nM).

NMI-900, an Aurora B/C kinase inhibitor, is under development at Cancer Research Technology in phase I/II clinical studies for the treatment of advanced and/or metastatic solid tumors. Other phase I clinical trials for the treatment of solid tumors had been previously completed, in a collaboration between GlaxoSmithKline and Cancer Research Technology, under the Cancer Research UK’s Clinical Development Partnerships (CDP) program.

The drug was originated by GlaxoSmithKline. The rights of the product were acquired by Cancer Research Technology from GlaxoSmithKline after the company elected not to take the program forward. In December 2015, the product was licensed by Cancer Research Technology to Nemucore Medical Innovations for the exclusive worldwide development and commercialization for the treatment of difficult-to-treat cancers.

GSK-1070916

PATENT

US 20070149561

https://www.google.com/patents/US20070149561

PAPER

Journal of Medicinal Chemistry (2010), 53 (10), 3973-4001

http://pubs.acs.org/doi/abs/10.1021/jm901870q

Discovery of GSK1070916, a Potent and Selective Inhibitor of Aurora B/C Kinase

Cancer Research, Oncology R&D
Molecular Discovery Research
GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426
§ Tsukuba Research Laboratories, Japan
J. Med. Chem., 2010, 53 (10), pp 3973–4001
DOI: 10.1021/jm901870q
Abstract Image

The Aurora kinases play critical roles in the regulation of mitosis and are frequently overexpressed or amplified in human tumors. Selective inhibitors may provide a new therapy for the treatment of tumors with Aurora kinase amplification. Herein we describe our lead optimization efforts within a 7-azaindole-based series culminating in the identification of GSK1070916 (17k). Key to the advancement of the series was the introduction of a 2-aryl group containing a basic amine onto the azaindole leading to significantly improved cellular activity. Compound 17k is a potent and selective ATP-competitive inhibitor of Aurora B and C with Ki* values of 0.38 ± 0.29 and 1.5 ± 0.4 nM, respectively, and is >250-fold selective over Aurora A. Biochemical characterization revealed that compound 17k has an extremely slow dissociation half-life from Aurora B (>480 min), distinguishing it from clinical compounds 1 and 2. In vitro treatment of A549 human lung cancer cells with compound 17k results in a potent antiproliferative effect (EC50 = 7 nM). Intraperitoneal administration of 17k in mice bearing human tumor xenografts leads to inhibition of histone H3 phosphorylation at serine 10 in human colon cancer (Colo205) and tumor regression in human leukemia (HL-60). Compound 17k is being progressed to human clinical trials.

http://pubs.acs.org/doi/pdf/10.1021/jm901870q………..PDF FILE

STR1

 

PAPER

Molecules 2014, 19(12), 19935-19979; doi:10.3390/molecules191219935

http://www.mdpi.com/1420-3049/19/12/19935/htm

http://www.mdpi.com/1420-3049/19/12/19935/htm

 

Biological Activity of GSK-1070916

GSK1070916 is a reversible and ATP-competitive inhibitor of Aurora B/C with IC50 of 3.5 nM/6.5 nM; displays >100-fold selectivity against the closely related Aurora A-TPX2 complex(IC50=490 nM).
IC50 Value: 3.5 nM(Aurora B); 6.5 nM(Aurora C)
Target: Aurora B/C
in vitro: GSK1070916 selectively inhibits Aurora B and Aurora C with Ki of 0.38 nM and 1.5 nM over Aurora A with Ki of 490 nM. Inhibition of Aurora B and Aurora C is time-dependent, with an enzyme-inhibitor dissociation half-life of >480 min and 270 min respectively. In addition, GSK1070916 is also a competitive inhibitor with respect to ATP. Human tumor cells treated with GSK1070916 shows dose-dependent inhibition of phosphorylation on serine 10 of Histone H3, a substrate specific for Aurora B. Moreover, GSK1070916 inhibits the proliferation of tumor cells with EC50 values of <10 nM in over 100 cell lines spanning a broad range of tumor types, with a median EC50 of 8 nM. Although GSK1070916 has potent activity against proliferating cells, a dramatic shift in potency is observed in primary, nondividing, normal human vein endothelial cells. Furthermore, GSK1070916-treated cells do not arrest in mitosis but instead fails to divide and become polyploid, ultimately leading to apoptosis. In another study, it is also reported high chromosome number associated with resistance to the inhibition of Aurora B and C suggests cells with a mechanism to bypass the high ploidy checkpoint are resistant to GSK1070916.
in vivo: GSK1070916 (25, 50, or 100 mg/kg) shows dose-dependent inhibition of phosphorylation of an Aurora B–specific substrate in mice and consistent with its broad cellular activity, has antitumor effects in 10 human tumor xenograft models including breast, colon, lung, and two leukemia models.

Clinical Information of GSK-1070916

Product Name Sponsor Only Condition Start Date End Date Phase Last Change Date
GSK-1070916 Cancer Research UK Advanced solid tumor 31-MAR-10 31-MAR-13 Phase 1 17-JUN-13

References on GSK-1070916

[1]. Anderson K, et al. Biochemical characterization of GSK1070916, a potent and selective inhibitor of Aurora B and Aurora C kinases with an extremely long residence time1. Biochem J. 2009 May 13;420(2):259-65.
Abstract


[2]. Hardwicke, Mary Ann; Oleykowski, Catherine A.; Plant, Ramona; GSK1070916, a potent Aurora B/C kinase inhibitor with broad antitumor activity in tissue culture cells and human tumor xenograft models. Molecular Cancer Therapeutics (2009), 8(7), 1808-1817.

[3]. Moy C, Oleykowski CA, Plant R, Greshock J, Jing J, Bachman K, Hardwicke MA, Wooster R, Degenhardt Y.High chromosome number in hematological cancer cell lines is a negative predictor of response to the inhibition of Aurora B and C by GSK1070916.J Transl Med. 2011 Jul 15;9:110.

[4]. Adams ND, Adams JL, Burgess JL, Chaudhari AM, Copeland RA, Donatelli CA, Drewry DH, Fisher KE, Hamajima T, Hardwicke MA, Huffman WF, Koretke-Brown KK, Lai ZV, McDonald OB, Nakamura H, Newlander KA, Oleykowski CA, Parrish CA, Patrick DR, Plant R, Sarpong MA, Sasaki K, Schmidt SJ, Silva DJ, Sutton D, Tang J, Thompson CS, Tummino PJ, Wang JC, Xiang H, Yang J, Dhanak D.Discovery of GSK1070916, a potent and selective inhibitor of Aurora B/C kinase.J Med Chem. 2010 May 27;53(10):3973-4001.

[5]. Medina JR, Grant SW, Axten JM, Miller WH, Donatelli CA, Hardwicke MA, Oleykowski CA, Liao Q, Plant R, Xiang H.Discovery of a new series of Aurora inhibitors through truncation of GSK1070916.Bioorg Med Chem Lett. 2010 Apr 15;20(8):2552-5. Epub 2010 Mar 1.

http://www.ingentaconnect.com/content/ben/lddd/2014/00000012/00000001/art00003?crawler=true

/////////////GSK1070916, GSK-1070916,  942918-07-2 GSK, phase1, Advanced solid tumor, NMI-900 , GSK-1070916, GSK-1070916A

Share

AMG 900, An aurora kinase (ARK) inhibitor potentially for the treatment of leukemia and solid tumours

 phase 1, Uncategorized  Comments Off on AMG 900, An aurora kinase (ARK) inhibitor potentially for the treatment of leukemia and solid tumours
Jun 152016
 

AMG-900

N-(4-((3-(2-aminopyrimidin-4-yl)pyridin-2-yl)oxy)phenyl)-4-(4-methylthiophen-2-yl)phthalazin-1-amine.

N-(4-(3-(2-Aminopyrimidin-4-yl)pyridin-2-yloxy)phenyl)-4-(4-methylthiophen-2-yl)phthalazin-1-amine

Phase I

Amgen Inc. INNOVATOR

Inventors Victor J. Cee, Holly L. Deak, Bingfan Du,Stephanie D. Geuns-Meyer, Brian L. Hodous,Hanh Nho Nguyen, Philip R. Olivieri, Vinod F. Patel, Karina Romero, Laurie Schenkel,Less «
Applicant Amgen Inc.

An aurora kinase (ARK) inhibitor potentially for the treatment of leukemia and solid tumours.

CAS No. 945595-80-2

In 2014, orphan drug designation was assigned in the U.S. for the treatment of ovarian cancer

Molecular Formula: C28H21N7OS
Molecular Weight: 503.57764 g/mo
AMG 900; AMG-900; 945595-80-2; AMG900; UNII-9R2G075611; N-(4-((3-(2-aminopyrimidin-4-yl)pyridin-2-yl)oxy)phenyl)-4-(4-methylthiophen-2-yl)phthalazin-1-amine;

AMG 900 is a small-molecule inhibitor of Aurora kinases A, B and C with potential antineoplastic activity. Aurora kinase inhibitor AMG 900 selectively binds to and inhibits the activities of Aurora kinases A, B and C, which may result in inhibition of cellular division and proliferation in tumor cells that overexpress these kinases. Aurora kinases are serine-threonine kinases that play essential roles in mitotic checkpoint control during mitosis and are overexpressed by a wide variety of cancer cell types. Check for active clinical trials or closed clinical trials using this agent

AMG 900 is a potent and highly selective pan-Aurora kinases inhibitor for Aurora A/B/C with IC50 of 5 nM/4 nM /1 nM;  >10-fold selective for Aurora kinases than p38α, Tyk2, JNK2, Met and Tie2.
IC50 Value: 5 nM(Aurora A); 4 nM(Aurora B); 1 nM(Aurora C)
Target: pan-Aurora
in vitro: AMG 900 is a novel class of ATP-competitive phthalazinamine small molecule inhibitors of aurora kinases. In HeLa cells, AMG 900 inhibits autophosphorylation of aurora-A and -B as well as phosphorylation of histone H3 on Ser, a proximal substrate of aurora-B. The predominant cellular response of tumor cells to AMG 900 treatment is aborted cell division without a prolonged mitotic arrest, which ultimately results in cell death. AMG 900 inhibits the proliferation of 26 tumor cell lines, including cell lines resistant to the antimitotic drug paclitaxel and to other aurora kinase inhibitors (AZD1152, MK-0457, and PHA-739358), at low nanomolar concentrations (about 2- 3 nM). Furthermore, AMG 900 is active in an AZD1152-resistant HCT116 variant cell line that harbors an aurora-B mutation (W221L) [1].
in vivo: Oral administration of AMG 900 blocks the phosphorylation of histone H3 in a dose-dependent manner and significantly inhibited the growth of HCT116 tumor xenografts. AMG 900 is broadly active in multiple xenograft models, including 3 multidrugresistant xenograft models, representing 5 tumor types [1]. AMG 900 exhibits a low-to-moderate clearance and a small volume of distribution. Its terminal elimination half-life ranged from 0.6 to 2.4 hours. AMG 900 is well-absorbed in fasted animals with an oral bioavailability of 31% to 107%. Food intake has an effect on rate (rats) or extent (dogs) of AMG 900 oral absorption. The clearance and volume of distribution at steady state in humans are predicted to be 27.3 mL/h/kg and 93.9 mL/kg, respectively. AMG 900 exhibits acceptable PK properties in preclinical species and is predicted to have low clearance in humans [2].

In mammalian cells, the aurora kinases (aurora-A, -B, and -C) play essential roles in regulating cell division. The expression of aurora-A and -B is elevated in a variety of human cancers and is associated with high proliferation rates and poor prognosis, making them attractive targets for anticancer therapy. AMG 900 is an orally bioavailable, potent, and highly selective pan-aurora kinase inhibitor that is active in taxane-resistant tumor cell lines. In tumor cells, AMG 900 inhibited autophosphorylation of aurora-A and -B as well as phosphorylation of histone H3 on Ser(10), a proximal substrate of aurora-B. The predominant cellular response of tumor cells to AMG 900 treatment was aborted cell division without a prolonged mitotic arrest, which ultimately resulted in cell death. AMG 900 inhibited the proliferation of 26 tumor cell lines, including cell lines resistant to the antimitotic drug paclitaxel and to other aurora kinase inhibitors (AZD1152, MK-0457, and PHA-739358), at low nanomolar concentrations. Furthermore, AMG 900 was active in an AZD1152-resistant HCT116 variant cell line that harbors an aurora-B mutation (W221L). Oral administration of AMG 900 blocked the phosphorylation of histone H3 in a dose-dependent manner and significantly inhibited the growth of HCT116 tumor xenografts. Importantly, AMG 900 was broadly active in multiple xenograft models, including 3 multidrug-resistant xenograft models, representing 5 tumor types. AMG 900 has entered clinical evaluation in adult patients with advanced cancers and has the potential to treat tumors refractory to anticancer drugs such as the taxanes.

MG 900 is an orally bioavailable, potent, and highly selective pan-aurora kinase inhibitor that is active in taxane-resistant tumor cell lines. In tumor cells, AMG 900 inhibited autophosphorylation of aurora-A and -B as well as phosphorylation of histone H3 on Ser10, a proximal substrate of aurora-B. The predominant cellular response of tumor cells to AMG 900 treatment was aborted cell division without a prolonged mitotic arrest, which ultimately resulted in cell death. AMG 900 inhibited the proliferation of 26 tumor cell lines, including cell lines resistant to the antimitotic drug paclitaxel and to other aurora kinase inhibitors (AZD1152, MK-0457, and PHA-739358), at low nanomolar concentrations. Furthermore, AMG 900 was active in an AZD1152-resistant HCT116 variant cell line that harbors an aurora-B mutation (W221L). Oral administration of AMG 900 blocked the phosphorylation of histone H3 in a dose-dependent manner and significantly inhibited the growth of HCT116 tumor xenografts. Importantly, AMG 900 was broadly active in multiple xenograft models, including 3 multidrug-resistant xenograft models, representing 5 tumor types. AMG 900 has entered clinical evaluation in adult patients with advanced cancers and has the potential to treat tumors refractory to anticancer drugs such as the taxanes. (Source: Cancer Res; 70(23); 9846–54.)

Clinical Information of AMG 900

Product Name Sponsor Only Condition Start Date End Date Phase Last Change Date
AMG 900 Amgen Inc Leukemia 31-JUL-11 31-JUL-14 Phase 1 14-SEP-13
Amgen Inc Advanced solid tumor 30-APR-09 30-JUN-13 Phase 1 10-SEP-13

AMG 900.png

PATENT

WO 2007087276

http://www.google.co.in/patents/WO2007087276A1?cl=en

PATENT

WO 2015084649

https://google.com/patents/WO2015084649A1?cl=en

The compound, N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)- 4-(4-methyl-2-thienyl)-l-phthalazinamine, also chemically named as 4-((3-(2-amino- pyrimidin-4-yl)-pyridin-2-yl)oxy)phenyl-(4-(4-methyl-thiophen-2-yl)-phthalazin-l- yl)amine, and is referred to herein as “AMG 900” has a chemical structure of

AMG 900 is an ATP competitive small molecule Aurora kinase inhibitor that is highly potent and selective for Aurora kinases A, B and C. AMG 900 is disclosed in US patent publication no. 20070185111, which published on August 9, 2007 and issued as U.S. Patent No. 7,560,551. AMG 900 is further disclosed in US patent publication no.

20090163501, now US patent no 8,022,221. Various uses and applications of AMG 900 are described in patent publications US20120028917 and WO2013149026. AMG 900 is being clinically evaluated primarily for its safety, tolerability and pharmacokinetic (PK) profile in human phase I trials for (1) advanced solid tumors (US Clinical Trial Id No. NCT00858377), and (2) for acute leukemias (US Clinical Trial Id No. NCT1380756).

Different solid state forms of a given compound are typically investigated to determine whether or not a particular form possesses and/or exhibits desirable properties allowing that compound to be clinically and/or commercially developed. Such beneficial and advantageous properties, by way of example, include without limitation, crystallinity, improved thermodynamic stability, non-hygroscopicity, high purity, minimal to total absence of moisture and/or residual solvents, chemical stability, high yielding synthetic process and/or manufacturability and reproducibility, desirable biopharmaceutical properties including improved dissolution characteristics and increased bioavailability, absence or reduced toxicities due to reduced or limited exposure, rate of exposure or release, or related to counter ions, good bulk and formulation properties including good flow, bulk density, desirable particle size and the like, or a combination of the aforementioned characteristic attributes.

Generally when a compound, also referred to herein as drug substance (DS), has been identified as a developmental candidate, the DS is screened to identify potentially beneficial polymorphic, crystalline or solid state forms of the compound and/or a pharmaceutically acceptable salt thereof. X-ray diffraction, Raman, solid state NMR and a melting point temperature and/or a melting point temperature range have been typically used to monitor or screen and identify the different polymorphic form of the DS.

Different polymorphic forms of a given DS can have an impact on that compound’s solubility, stability and bioavailability. Also, it is important to monitor possible changes in polymorphic forms of the DS during stability studies.

AMG 900 was previously isolated and identified as a free base compound. This compound exhibited rather lack-luster pharmacokinetic (PK) and/or pharmacodynamic (PD) properties, including poor aqueous solubility, poor bioavailability, poor absorption, poor target exposure and overall, a not-so-attractive in-vivo efficacy profile. Thus, there is a need to address and solve the technical problem of identifying alternative forms of AMG 900 to achieve substantially the same effect or an improved effect, including improved PK and PD profiles, as that of AMG 900 known in the art.

 

Example 1

Synthesis of N-(4-((3-(2-amino-4-pyrimidinylN)-2-pyridinylN)oxyN)phenylN)-4-(4-methyl-2-thienvD-l-phthalazinamine (AMG 900)

Step 1 : 4-(2-chloropyridin-3-yl)pyrimidin-2 -amine

In an argon purged 500 mL round bottom flask placed in an isopropanol bath, was added sodium metal (3.40g, 148mmol) slowly to methanol (180mL). The mixture was stirred at room temperature (RT) for about 30 minutes. To this was added guanidine hydrochloride (12.0 mL, 182 mmol) and the mixture was stirred at RT for 30 minutes, followed by addition of (E)-l-(2-chloropyridin-3-yl)-3-(dimethylamino)prop-2-en-l-one (12.0 g, 57.0 mmol), attached air condenser, moved reaction to an oil bath, where it was heated to about 50 °C for 24 hr. Approximately half of the methanol was evaporated under reduced pressure and the solids were filtered under vacuum, then washed with saturated sodium bicarbonate (NaHCO and H^O, air dried to yield 4-(2-chloropyridin-3-yl)pyrimidin-2-amine as off white solid. MS m/z = 207 [M+l]+. Calc’d for C9H7C1N4: 206.63.

Step 2: 4-(2-(4-aminophenoxy)pyridin-3-yl)pyrimidin-2-amine

To a resealable tube was added 4-aminophenol (1.3 g, 12 mmol), cesium carbonate (7.8 g, 24 mmol), and DMSO (16 ml, 0.75 M). The mixture was heated to 100 °C for 5 minutes, and then 4-(2-chloropyridin-3-yl)pyrimidin-2 -amine (2.5 g, 12 mmol) was added, and the reaction mixture was heated to 130 °C overnight. Upon completion, as judged by LCMS, the reaction mixture was allowed to cool to RT and diluted with water. The resulting precipitate was filtered, and the solid washed with water and diethyl ether. The solid was then taken up in 9: 1 CH2Cl2:MeOH and passed through a pad of silica gel with 9:1 CH2Cl2:MeOH as eluent. The solvent was concentrated in vacuo to provide the desired product, 4-(2-(4-aminophenoxy)pyridin-3-yl)pyrimidin-2-amine. MS m/z = 280

[M+l]+. Calc’d for Ci5H13N50: 279.30.

Step 3: l-Chloro-4-(4-methylthiophen-2-yl)phthalazine

1 ,4-Dichlorophthalazine (1.40 g, 7.03 mmol), 4-methyltmophen-2-ylboronic acid (999 mg, 7.03 mmol), and PdCl2(DPPF) (721 mg, 985 μιηοΐ) were added into a sealed tube. The tube was purged with Argon. Then sodium carbonate (2.0 M in water) (7.74 ml, 15.5 mmol) and 1,4-dioxane (35.2 ml, 7.03 mmol) were added. The tube was sealed, stirred at RT for 5 min, and placed in a preheated oil bath at 110 °C. After 1 hr, LC-MS showed product and byproduct (double coupling), and starting material

dichlorophthalazme. The reaction was cooled to RT, filtered through a pad of celite with an aid of ethyl acetate (EtOAc), concentrated, and loaded onto column. The product was purified by column chromatography using Hex to remove the top spot, then 80:20 hexanes:EtOAc to collect the product. The product, 1 -chloro-4-(4-methylthiophen-2-yl)phthalazine was obtained as yellow solid. LC-MS showed that the product was contaminated with a small amount of dichlorophthalazme and biscoupling byproduct. MS m/z = 261 [M+l]+. Calcd for Ci3H9ClN2S: 260.12.

Step 4: N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)- 1 -phthalazinamine

To 4-(2-(4-aminophenoxy)pyridin-3-yl)pyrimidin-2 -amine and l-chloro-4-(4-methyl-2-thienyl)phthalazine was added tBuOH. The resulting mixture was heated at 100 °C in a sealed tube for 16 hours. The rection was diluted with diethyl ether and saturated sodium carbonate and vigorously shaken. The resulting solids were filtered and washed with water, diethyl ether and air dried to yield N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)-l -phthalazinamine as an off-white solid. MS m/z = 504 [M+H]+. Calc’d for C28 H21 N7 O S: 503.58.

Example 2

Alternative Synthesis of N-(4-((3-(2-amino-4-pyrimidinylN)-2-pyridinylN)oxyN)phenylN)-4-(4-methyl-2-thienvD-l-phthalazinamine (AMG 900)

Step 1 : 4-(2-chloropyridin-3-yl)pyrimidin-2 -amine

In an argon purged 500 mL round bottom flask placed in an isopropanol bath, was added sodium metal (3.40g, 148mmol) slowly to methanol (180mL). The mixture was stirred at room temperature (RT) for about 30 minutes. To this was added guanidine hydrochloride (12.0 mL, 182 mmol) and the mixture was stirred at RT for 30 minutes, followed by addition of (E)-l-(2-chloropyridin-3-yl)-3-(dimethylamino)prop-2-en-l-one (12.0 g, 57.0 mmol), attached air condenser, moved reaction to an oil bath, where it was heated to about 50 °C for 24 hr. Approximately half of the methanol was evaporated under reduced pressure and the solids were filtered under vacuum, then washed with saturated sodium bicarbonate (NaHCO and H^O, air dried to yield 4-(2-chloropyridin-3-yl)pyrimidin-2-amine as off white solid. MS m/z = 207 [M+l]+. Calc’d for C9H7C1N4: 206.63.

Step 2: 4-(2-(4-aminophenoxy)pyridin-3-yl)pyrimidin-2-amine

To a reaction vessel at ambient temperature was added 4-aminophenol (571 g, 5.25 mol, 1.05 equiv) followed by 4-(2-chloropyridin-3-yl)pyrimidin-2-amine (1064g, 97 wt%, 5.00 mol, 1.0 equiv) and DMSO (7110 ml, 7820 g, 7x the volume of 4-(2-chloropyridin-3-yl)pyrimidin-2 -amine). The reaction mixture was agitated and sparged with nitrogen gas for at least 10 minutes. Then a 50 weight % aqueous KOH solution (593 g, 5.25 mol, 1.05 equiv.) was added to the mixture while keeping the reaction

mixture temperature below about 40°C. The mixture was sparged with nitrogen gas for more than 5 minutes, the sparging tube was removed, and the reaction mixture was heated to 110 +/- 10 °C for at least 1.5 hrs. Upon completion, as judged by HPLC, the reaction mixture was allowed to cool to RT and diluted with 6N HC1 (42 mL, 0.25 mol, 0.05 equiv). The desired product, 4-(2-(4-aminophenoxy)pyridin-3-yl)pyrimidin-2 -amine was not isolated. Rather, it was formed in-situ and combined with the product of step 3 below, in step 4 to form the desired product.

Step 3: l-Chloro-4-(4-methylthiophen-2-yl)phthalazine

A separate reaction vessel was fitted with a reflux condenser and an addition funnel, and 4-(4-methylthiophen-2-yl)phthalazin-l(2H)-one (1,537 mg, 6.34 mol, 1.0 equivalent) was added to the reaction vessel. Acetonitrile (7540 mL, 5859 g, 5 V), was added and the reaction vessel was agitated to allow the starting material to dissolve. The vessel was then charged with phosphorus oxychloride (709 ml, 1166 g, 7.44 mol, 1.2 equivalents) and the reaction was heated to about 75 +/- 5 °C for a least 4 hrs. The reaction was cooled to about about 25 +/- 5 °C and held there for more than 24 hrs. N,N-diisopropylethylamine (3046 g, 4100 mL, 3.8 equivalents) was added to the reaction vessel and the temperature was maintained at <30°C. Pyridine (97g, 1.24 mol, 0.2 equiv) was added in a single portion followed by water (4100 g, 2.7V) over more than 30 minutes. The reaction mixture was stirred at ambient temperature ofr about 24 hrs. the mixture was filtered through a <25uM polypropylene filter and the rsulting mother liquor was diluted with 1 : 1 ACN:water (9000 mL total) and stirred for a minimum of 2 minutes. Filter off product solids as they precipitate. Collect mother liquor and washes to obtain additional product. Dry the filter cake, and additional product crops, under a constant stream of nitrogen gas for at least 14 hrs. Unlike the previous method, the present method avoids contamination of impurities, such as dichlorophthalazine and biscoupling byproduct, as seen via LC-MS. Yield: 1537 g (97.2 weight %). MS m/z = 261 [M+l]+. Calcd for Ci3H9ClN2S: 260.12.

Step 4: N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)- 1 -phthalazinamine

To the reaction mixture was added l-chloro-4(4-methylthiophen-2-yl)phthalazine

(1450g, 97.2 wt%, 5.40 mol, 1.08 equiv) rinding the addition port with DMSO (520 ml, 572 g, 0.5x the volume of 4-(2-chloropyridin-3-yl)pyrimidin-2-amine). The reaction mixture was again agitated and sparged with nitrogen gas for at least 10 minutes. The sparging tube was removed, and the reaction mixture was heated to 80 +/- 20 °C for at least 2 hrs. Upon completion, as judged by HPLC, the reaction mixture was allowed to cool to RT and N,N-diisopropylethylamine (776 g, 1045 mL, 6.0 mol, 1.2 equiv) was added and the mixture was kept at about 80 +/- 10°C. Filter the mixture at about 80oC into a separate reactor vessel rinsing with DMSO (1030 mL, 1133 g, 1 V). Then adjust the raction mixture temperature to about 70+/-5 °C and add 2-propanol (13200 mL, 10360 g, 12.75 V) over more than 15 minutes at about 70°C. As the reaction mistreu cools, the product begins to precipitate out of solution. Add more 2-propanol (8780 mL, 6900 g, 8.5V) to the solution slowly over more then 60 minutes at about 70°C. The reactor vessel was cooled to about 20°C over more than 60 minutes and let sit for over 2 hrs. The product was filtered through an Aurora filter with a >25uM polypropylene filter cloth. Additional crops were obtained from the mother liquors by diluting with additional 2-propanol. The filter cakes were dried under a constant stream of nitrogen gas for at least 14 hrs to provide the desired product, N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)-l-phthalazinamine as an off-white solid. Yield: 2831 g (88.8%); purity 99.7%. MS m/z = 504 [M+H]+. Calc’d for C28 H21 N7 O S: 503.58.

The starting material 1 used/shown in Example 2 was prepared as follows:

and starting material 3, thienyl substituted phthalazinone, shown in Example 2 was prepared as follows:

Starting material 3

Synthesis of 4-(5-methylthiophen-2-yl)phthalazin-l(2//)-one

Step 1 : 2-(Dimethylamino)isoindoline-1.3-dione

A solution of isobenzofuran-l,3-dione (5.00 g, 34 mmol) and N,N-dimethylhydrazine (2.9 ml, 37 mmol) in toluene (75 ml, 34 mmol) was added p-TsOH H20 (0.32 g, 1.7 mmol). The Dean-Stark apparatus and a condenser were attached. The mixture was refluxed. After 4 hr, LCMS showed mainly product. The reaction was cooled to rt. Toluene was removed under reduced pressure the crude was dissolved in DCM, washed with sat NaHC03, water, and brine. The organic was dried over MgS04, filtered, and concentrated. Light yellow solid was obtained. !H NMR showed mainly product, 2-(dimethylamino)isoindoline-l,3-dione. MS Calcd for C10H10N2O2: [M]+ = 190. Found: [M+H]+ = 191.

Step 2 : 2-(Dimethylamino)-3 -hydroxy-3 -(5 -methylthiophen-2 -vDisoindolin- 1 -one

A solution of 2-bromo-5-methylthiophene (0.60 mL, 5.3 mmol) in THF (11 mL) was purged with nitrogen and cooled to -78 °C. «-Butyllithium (2.2 mL, 5.5 mmol; 2.5 M in THF) was added and the mixture was stirred under nitrogen for 30 min. This solution was cannulated into a flask containing a solution of 2-(dimethylamino)isoindoline-l,3-dione (1.5 g, 7.9 mmol) in THF (16 mL) at -78 °C under nitrogen. The reaction was allowed to warm to -30 °C over an hour, at which point LCMS showed complete conversion of 2-bromo-5-methylthiophene to product. The reaction was quenched by careful addition of saturated aqueous NH4C1. The reaction mixture was diluted with dichloromethane and water, and the layers were separated. The aqueous portion was extracted with additional dichloromethane, and the combined organic layers were dried with MgS04, filtered, concentrated, and purified by silica gel chromatography eluting with 0-2% MeOH in dichloromethane to provide intermediate A, as a light yellow solid, 2-(dimethylamino)-3-hydroxy-3-(5-methylthiophen-2-yl)isoindolin-l-one (1.2 g, 80% yield). !H NMR (400 MHz, DMSO-4) δ 7.68-7.65 (m, 1H). 7.63-7.59 (m, 1H), 7.57-7.51 (m, 1H), 7.37 (d, 1H, J=8), 7.09 (s, 1H), 6.69-6.66 (m, 1H), 6.65-6.62 (m, 1H), 2.81 (s, 6H), 2.40 (s, 3H). 13C NMR (400 MHz, DMSO-de) δ 165.0, 147.3, 141.6, 139.3, 132.7, 129.49, 129.46, 125.0, 124.7, 123.0, 122.1, 88.4, 44.7, 14.9. FT-IR (thin film, cm ) 3347, 3215, 1673. MS Calcd for Ci2H7ClN2S: [M]+ = 288. Found: [M+H]+= 289.

HRMS Calcd for Ci5H16N202S: [M+H]+= 288.1005, [M+Na]+ = 311.0825. Found:

[M+H]+ = 289.1022, [M+Na]+= 311.0838. mp = 138-140 °C.

Step 3: 4-(5-Methylthiophen-2-yl)phthalazin-l(2//)-one

2-(Dimethylamino)-3 -hydroxy-3 -(5 -methylthiophen-2-yl)isoindolin- 1 -one (1.1 g, 0.40 mmol), EtOH (4.0 mL), and hydrazine (0.19 mL, 59 mmol) were added into a RBF fitted with a reflux condenser. A nitrogen balloon was attached on top of the condenser. After refluxing overnight, the reaction was cooled to room temperature. An off-white solid precipitated. After cooling to 0 °C, water was added. The solid was filtered off with an aid of water and dried under vacuum to afford a white solid, 4-(5-methylthiophen-2-yl)phthalazin-l(2//)-one (0.82 g, 85% yield).

!H NMR (400 MHz, CDC13) δ 10.57 (s, 1H), 8.50-8.39 (m, 1H), 8.14-8.04 (m, 1H), 7.83- 7.69 (m, 2H), 7.20-7.17 (m, 1H), 6.82-6.71 (m, 1H), 2.47 (s, 3H). 13C NMR (400 MHz,

CDC13) 8 159.9, 142.5, 141.1, 134.3, 133.7, 131.7, 129.4, 128.8, 128.3, 127.1, 126.6,

125.8, 15.4. FT-IR (thin film, cm“1) 2891, 1660, 1334. MS Calcd for Ci3H10N2OS: [M]+

= 242. Found: [M+H]+= 243. HRMS Calcd for Ci3H10N2OS: [M+H]+= 243.0587. Found:

[M+H]+ = 243.0581. mp = 191-194 °C.

Alternatively, starting material 3 was prepared as follows:

The above scheme depicts the process by which intermediate-scale synthesis of thiophene-phthalazinone 5 (shown above) was prepared. Treatment of 50 grams of 3-methylthiophene with z-PrMgCl at 66 °C in the presence of catalytic TMP-H resulted in 98% conversion to the reactive species lb with a >40:1 regioisomeric ratio. After cooling to 20 °C, this mixture was added dropwise to a -20 °C slurry of phthalic anhydride in THF to provide keto acid 3 in 94% assay yield. While this intermediate could be crystallized from toluene/heptane, the crude reaction mixture was taken directly in a through -process conversion to the phthalazinone 5. To that end, removal of THF, MTBE, and residual 3-methylthiophene was accomplished through a distillative solvent switch into ethanol. The resulting solution of 3 was exposed to aqueous hydrazine at 80 °C. After 18 hours, the reaction was cooled and the precipitated product was filtered directly at 20 °C. This process provided 82.7 grams of 98.6 wt % thiophene-phthalazinone 5 in a weight-adjusted 85% yield over the two steps.

LCMS Method:

Samples were run on a Agilent model- 1100 LC-MSD system with an Agilent Technologies XDB-C8 (3.5 μ) reverse phase column (4.6 x 75 mm) at 30 °C. The flow rate was constant and ranged from about 0.75 mL/min to about 1.0 mL/min.

The mobile phase used a mixture of solvent A (H2O/0.1% HO Ac) and solvent B

(AcCN/O.1 HOAc) with a 9 min time period for a gradient from 10%> to 90%> solvent B. The gradient was followed by a 0.5 min period to return to 10% solvent B and a 2.5 min 10% solvent B re-equilibration (flush) of the column.

Other methods may also be used to synthesize AMG 900. Many synthetic chemistry transformations, as well as protecting group methodologies, useful in synthesizing AMG 900, are known in the art. Useful organic chemical transformation literature includes, for example, R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, 3rd edition, John Wiley and Sons (1999); L. Fieser and M. Fieser, Fieser and Fieser’s Reagents for Organic Synthesis, John Wiley and Sons (1994); A. Katritzky and

A. Pozharski, Handbook of Heterocyclic Chemistry, 2nd edition (2001); M. Bodanszky, A. Bodanszky, The Practice of Peptide Synthesis, Springer- Verlag, Berlin Heidelberg (1984); J. Seyden-Penne, Reductions by the Alumino- and Borohydrides in Organic Synthesis, 2nd edition, Wiley- VCH, (1997); and L. Paquette, editor, Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995).

AMG 900 was tested for its ability to reduce or inhibit tumor progression in various cell lines (in-vitro) and multiple solid tumor types (in-vivo), some of which have previously been exposed to and developed resistance to standard-of-care antimitotic agents, including taxanes and vinca alkaloids, as well as to other chemotherapeutic agents. The following Examples and resulting data will illustrate the ability of AMG 900 to treat cancer, including cancer resistant to the presently standard-of-care therapies, including antimitotic agents, such as paclitaxel, and other drugs used in conjunction with chemotherapy, such as doxorubicin. Unless otherwise indicated, the free base form of AMG 900 was used in the Examples described hereinbelow.

The following Examples describe the efforts of identifying and characterizing various crystalline solid state forms of various salts of AMG 900. Some attempts at forming a solid state crystalline form of a given salt failed, as shown in table 1 hereinbelow. To this end, synthesizing and/or forming &isolating a crystalline solid state form of AMG 900 was not, in any way, straightforward or routine. Rather, the ability to prepare and identify a crystalline solid state form of AMG 900 depended upon the particular salt of AMG 900 and/or the crystallization conditions employed.

PAPER

Journal of Medicinal Chemistry (2015), 58(13), 5189-5207

Discovery of N-(4-(3-(2-Aminopyrimidin-4-yl)pyridin-2-yloxy)phenyl)-4-(4-methylthiophen-2-yl)phthalazin-1-amine (AMG 900), A Highly Selective, Orally Bioavailable Inhibitor of Aurora Kinases with Activity against Multidrug-Resistant Cancer Cell Lines

Departments of Medicinal Chemistry, Pharmaceutical Research and Development, §Pharmacokinetics and Drug Metabolism, Molecular Structure, and Oncology Research, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States, and Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
J. Med. Chem., 2015, 58 (13), pp 5189–5207
DOI: 10.1021/acs.jmedchem.5b00183
*Phone: 617-444-5041. E-mail: MeyerS@amgen.com.

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.

Abstract

Abstract Image

Efforts to improve upon the physical properties and metabolic stability of Aurora kinase inhibitor14a revealed that potency against multidrug-resistant cell lines was compromised by increased polarity. Despite its high in vitro metabolic intrinsic clearance, 23r (AMG 900) showed acceptable pharmacokinetic properties and robust pharmacodynamic activity. Projecting from in vitro data to in vivo target coverage was not practical due to disjunctions between enzyme and cell data, complex and apparently contradictory indicators of binding kinetics, and unmeasurable free fraction in plasma. In contrast, it was straightforward to relate pharmacokinetics to pharmacodynamics and efficacy by following the time above a threshold concentration. On the basis of its oral route of administration, a selectivity profile that favors Aurora-driven pharmacology and its activity against multidrug-resistant cell lines, 23r was identified as a potential best-in-class Aurora kinase inhibitor. In phase 1 dose expansion studies with G-CSF support, 23r has shown promising single agent activity.

N-(4-(3-(2-Aminopyrimidin-4-yl)pyridin-2-yloxy)phenyl)-4-(4-methylthiophen-2-yl)phthalazin-1-amine (23r)

Applying similar SNAr conditions as for 23b, reaction of 22r and 20a in 2-butanol provided the title compound (2.08 g, 49%) as an off-white solid; mp (DSC) 216 °C.
1H NMR (400 MHz, DMSO-d6) δ ppm 9.36 (s, 1 H) 8.64–8.69 (m, 1 H) 8.41–8.44 (m, 1 H) 8.36–8.40 (m, 1 H) 8.35 (d, J = 5.2 Hz, 1 H) 8.23 (dd, J = 4.8, 2.0 Hz, 1 H) 8.00–8.10 (m, 2 H) 7.91–7.97 (m, 2 H) 7.52 (d, J = 1.0 Hz, 1 H) 7.26–7.33 (m, 3 H) 7.16–7.22 (m, 2 H) 6.74 (br s, 2 H) 2.34 (br s, 3 H).
13C NMR (150 MHz, DMSO-d6) δ 163.81, 160.72, 160.67, 158.68, 151.64, 148.50, 148.36, 147.14, 139.86, 139.24, 137.72, 137.10, 132.61, 131.74, 130.24, 125.27, 124.89, 122.92, 122.83, 122.44, 121.56, 121.52, 119.11, 118.23, 109.93, 15.6
HRMS m/z [M + H]+ Calcd for C28H21N7OS: 504.1601. Found: 504.1607.
Table 1. Aurora Kinase Inhibitors with Known Structures That Have Entered Clinical Trials

ID, compd name AKa AK cell assayb (nM) most potently inhibited non-AKs (nM)c [total kinases in panel] admin route
1 (MK-0457/VX-680/tozasertib)(7) A/B MDA-MB-231 p-HH3(12) 43 FLT3 (6), PLK4 (9), ABL (13), MLCK (15), RET (28); [317](16) IV
2 (PHA-739358/danusertib)(8) A/B MDA-MB-231 p-HH3(12) 49 ABL (25), RET (31), TrkA (31), FGFR1 (47); [35](17) IV
3a(AZD1152/barasertib)d,(9) B MDA-MB-231 p-HH3(12) 16 FLT3 (8), cKIT (17), PDGFRA (38), PDGFRB (41), RET (80); [317](16) IV
4 (AT9283)(18) A/B HCT-116 DNA ploidy ∼30 JAK2 (1), JAK3 (1) Abl (T315I) (4), 9 others ≤10 nM; [230] IV
5 (SNS-314)(19) A/B HCT-116 DNA ploidy(20) 9 TrkB (5), TrkA (12), FLT4 (14), Fms (15), DDR2 (82), Axl (84); [219] IV
6 (GSK1070916)(21) B HCT-116 p-HH3(22) 20 FLT1 (42), TIE2 (59), SIK (70), FLT4 (74), FGFR (78); [328](22) IV
7 (ENMD-2076)(23) A HCT-116 p-AurA 130 FLT3 (2), RET (10), FLT4 (16), SRC (20), TrkA (24), Fms (25); [100] PO
8 (CYC116)(24) A/B A549 p-HH3 480 VEGFR2 (44), FLT3 (44), CDK2 (390); [23] PO
9 (ABT-348)(25) A/B HCT-116 p-HH3 21 VEGFR1 (1), FLT3 (1), VEGFR2 (2), CSF-1R (3), PDGFR-α (11); [128] PO
10 (AS703569/R763)(26) A/B A549 p-HH3 14 cell-based assays: VEGFR2 (11), FLT3 (27), AMPK (201); [10] PO
11 (PF-03814735)(27) A/B MDA-MB-231 p-HH3 ∼50 FLT1 (10), FAK (22), TrkA (30), 17 others ≥90% inh@100 nM; [220] PO
12 (MK-5108)(28) A HeLa S3 ↑p-HH3+ cells <1000 TrkA (2), ABL (8), FLT4 (12), TrkB (13), VEGFR2 (30); [233] PO
13a (MLN8054)(29) A HCT-116 p-AurA 34 DRAK2 (8), BLK (68), DRAK1 (190), FGR (220); [317](16) PO
13b (MLN8237/alisertib)(30) A HeLa p-AurA 7 %inh@1 μM: EphA2 (111), FGR (97), CAMK2A (95), EphA4 (94); [220] PO

a

AK = Aurora kinase family member(s) inhibited (AurA and/or AurB; AurC potency not listed).

b

Cell line; substrate or phenotype detected.

c

Kinase activities of greatest potency listed in published literature.

d

Listed enzyme and cellular potency data is for 3b, the parent of prodrug 3a.

References on AMG 900

Patent ID Date Patent Title
US2016008316 2016-01-14 USE OF DIANHYDROGALACTITOL AND ANALOGS OR DERIVATIVES THEREOF IN COMBINATION WITH PLATINUM-CONTAINING ANTINEOPLASTIC AGENTS TO TREAT NON-SMALL-CELL CARCINOMA OF THE LUNG AND BRAIN METASTASES
US2016009785 2016-01-14 NOVEL FUSION MOLECULES AND USES THEREOF
US2015266868 2015-09-24 PHARMACEUTICALLY ACTIVE COMPOUNDS
US2015079022 2015-03-19 USE OF AMG 900 FOR THE TREATMENT OF CANCER
US2015072988 2015-03-12 USE OF N-(4-((3-(2-AMINO-4-PYRIMIDINYL)-2-PYRIDINYL)OXY)PHENYL)-4-(4-METHYL-2-THIENYL)-1-PHTHALAZINAMINE IN COMBINATION WITH HISTONE DEACETYLASE INHIBITORS FOR TREATMENT OF CANCER
US8921367 2014-12-30 Use of AMG 900 for the treatment of cancer
US2014163052 2014-06-12 FUSED TRICYCLIC DUAL INHIBITORS OF CDK 4/6 AND FLT3
US2014127271 2014-05-08 BLOCK COPOLYMERS FOR STABLE MICELLES
US2014113879 2014-04-24 BLOCK COPOLYMERS FOR STABLE MICELLES
US2014114051 2014-04-24 BLOCK COPOLYMERS FOR STABLE MICELLES
Patent ID Date Patent Title
US2014114051 2014-04-24 BLOCK COPOLYMERS FOR STABLE MICELLES
US2014066430 2014-03-06 AURORA KINASE MODULATORS AND METHOD OF USE
US8623885 2014-01-07 Fused tricyclic dual inhibitors of CDK 4/6 and FLT3
US2012028917 2012-02-02 Use Of N-(4-((3-(2-Amino-4-Pyrimidinyl)-2-Pyridinyl)Oxy)Phenyl)-4-(4-Methyl-2-Thienyl)-1-Phthalazinamine In The Treatment Of Antimitotic Agent Resistant Cancer
US2011263530 2011-10-27 Aurora Kinase Modulators and Method of Use
US8022221 2011-09-20 Aurora kinase modulators and method of use
US7560551 2009-07-14 Aurora kinase modulators and method of use
WO2003055491A1 20 Dec 2002 10 Jul 2003 Astrazeneca Ab Substituted quinazoline derivatives as inhibitors of aurora kinases
WO2004000833A1 19 Jun 2003 31 Dec 2003 Vertex Pharmaceuticals Incorporated Processes for preparing substituted pyrimidines and pyrimidine derivatives as inhibitors of protein kinase
WO2004016612A2 13 Aug 2003 26 Feb 2004 Cyclacel Limited New purine derivatives
WO2004037814A1 27 Oct 2003 6 May 2004 Vertex Pharmaceuticals Incorporated Indazolinone compositions useful as kinase inhibitors
WO2004039774A2 19 May 2003 13 May 2004 Merck & Co., Inc. Mitotic kinesin inhibitors
WO2004092607A1 30 Mar 2004 28 Oct 2004 Carbone Lorraine Composants Ventilated disc brake pads
WO2005113494A2 9 May 2005 1 Dec 2005 Amgen Inc. Nitrogenated heterocyclic derivatives as protein kinase modulators and use for the treatment of angiogenesis and cancer
EP1702919A1 28 Dec 2004 20 Sep 2006 Banyu Pharmaceutical Co., Ltd. Novel 2-heteroaryl-substituted benzimidazole derivative
US6919338 21 Jun 2001 19 Jul 2005 Astrazeneca Ab Substituted quinazoline derivatives and their use as inhibitors of aurora-2 kinase
Citing Patent Filing date Publication date Applicant Title
WO2008124083A3 * 3 Apr 2008 15 Jan 2009 Amgen Inc Aurora kinase modulators and method of use
WO2009117157A1 * 19 Mar 2009 24 Sep 2009 Amgen Inc. Aurora kinase modulators and method of use
WO2010017240A2 * 4 Aug 2009 11 Feb 2010 Amgen Inc. Aurora kinase modulators and methods of use
WO2010017240A3 * 4 Aug 2009 1 Apr 2010 Amgen Inc. Aurora kinase modulators and methods of use
WO2011031842A1 9 Sep 2010 17 Mar 2011 Amgen Inc. N-4 ( – ( ( 3- ( 2 -amino-4 pyrimidinyl) -2 -pyridinyl) oxy) phenyl) -4- (4-methyl-2-thienyl) -1-phthalazinamine for use in the treatment of antimitotic agent resistant cancer
WO2012129344A1 21 Mar 2012 27 Sep 2012 Amgen Inc. Fused tricyclic dual inhibitors of cdk 4/6 and flt3
WO2015084649A1 25 Nov 2014 11 Jun 2015 Amgen Inc. Crystalline forms of n-(4-((3-(2-amino-4-pyrimidinyl) – 2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)-1 -phthalazinamine pharmaceutically acceptable salts and uses thereof
EP2818170A1 9 Sep 2010 31 Dec 2014 Amgen, Inc N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridin yl)oxy)phenyl)-4-(4-methyl-2-thienyl)-1-phthalazinamine for use in the treatment of antimitotic agent resistant cancer
EP2937349A1 21 Mar 2012 28 Oct 2015 Amgen Inc. Fused tricyclic dual inhibitors of cdk 4/6 and flt3
US7994185 4 May 2009 9 Aug 2011 Glaxo Smith Kline LLC Benzene sulfonamide thiazole and oxazole compounds
US8362241 29 Jan 2013 Amgen Inc. Inhibitors of PI3 kinase and/or mTOR
US8404694 19 Mar 2009 26 Mar 2013 Amgen Inc. Aurora kinase modulators and method of use
US8415345 4 May 2009 9 Apr 2013 Glaxo SmithKline LLC Benzene sulfonamide thiazole and oxazole compounds
US8586751 10 Jun 2010 19 Nov 2013 Bristol-Myers Squibb Company Nicotinamide compounds useful as kinase modulators
US8637500 16 Dec 2009 28 Jan 2014 Amgen Inc. Aminopyridine and carboxypyridine compounds as phosphodiesterase 10 inhibitors
US8642759 31 Jan 2013 4 Feb 2014 Glaxosmithkline Llc Benzene sulfonamide thiazole and oxazole compounds
US8772480 19 Nov 2012 8 Jul 2014 Amgen Inc. Inhibitors of PI3 kinase and/or mTOR
US9126935 7 Aug 2009 8 Sep 2015 Amgen Inc. Aurora kinase modulators and methods of use
US9233956 25 Nov 2013 12 Jan 2016 Novartis Ag Benzene sulfonamide thiazole and oxazole compounds

///////////945595-80-2, AMG 900,  aurora kinase (ARK) inhibitor,  treatment of leukemia and solid tumours, AMGEN, 2014, orphan drug designation,  U.S. for the treatment of ovarian cancer

CC1=CSC(=C1)C2=NN=C(C3=CC=CC=C32)NC4=CC=C(C=C4)OC5=C(C=CC=N5)C6=NC(=NC=C6)N

Share

Maralixibat Chloride, ماراليكسيبات كلوريد , 氯马昔巴特 , Мараликсибата хлорид

 breakthrough designation  Comments Off on Maralixibat Chloride, ماراليكسيبات كلوريد , 氯马昔巴特 , Мараликсибата хлорид
Jun 152016
 

STR1

 

2D chemical structure of 228113-66-4

Maralixibat chloride

Maralixibat Chloride,  ماراليكسيبات كلوريد ,  氯马昔巴特 , Мараликсибата хлорид

SHP625, Maralixibat chloride, Molecular Formula C40-H56-N3-O4-S.Cl, Molecular Weight, 710.4184

4-Aza-1-azoniabicyclo(2.2.2)octane, 1-((4-((4-((4R,5R)-3,3-dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro-4-hydroxy-1,1-dioxido-1-benzothiepin-5-yl)phenoxy)methyl)phenyl)methyl)-, chloride (1:1)

1-[4-({4-[(4R,5R)-3,3-Dibutyl-7-(dimethylamino)-4-hydroxy-1,1-dioxido-2,3,4,5-tetrahydro-1-benzothiepin-5-yl]phenoxy}methyl)benzyl]-4-aza-1-azoniabicyclo[2.2.2]octane chloride

4-Aza-1-azoniabicyclo[2.2.2]octane, 1-[[4-[[4-[(4R,5R)-3,3-dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro-4-hydroxy-1,1-dioxido-1-benzothiepin-5-yl]phenoxy]methyl]phenyl]methyl]-, chloride

(4R.5R)-1- r.4- r _4- .3.3 -Dibutyl-7- (dimethylamino) -2.3 ,4.5- tetrahydro-4-hydroxy-1, l-dioxido-l-benzothiepin-5- yl] henoxy] ethyl] phenyl1methyl] -4-aza-l- azoniabicyclo [2.2.2] octane

(4Rcis)-1-[[4-[[4-[3,3-Dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro-4-hydroxy-1,1-dioxido-1-benzothiepin-5-yl]phenoxy]methyl]phenyl]methyl]-4-aza-1-azoniabicyclo[2.2.2]octane Chloride Salt

(4R,5R)- 1 -((4-(4-(3,3-dibutyl-7-(dimemylamino)-2,3,4,5-tetrahydro-4- hydroxy- 1 , 1 -diυxido- 1 -benzithiepin-5-yl)pheπoxy)methyl)phenyl)methyl-4-aza- 1 – azoniabicyclo[2.2.2]octane chloride

Cas: 228113-66-4, Free form 716313-53-0
UNII: V78M04F0XC, LUM 001, Lopixibat chloride, Treatment of Cholestatic Liver Diseases

Inventors James Li, Ching-Cheng Wang, David B. Reitz, Victor Snieckus, Horng-Chih Huang,Andrew J. Carpenter, Less «
Applicant G.D. Searle & Co.

Several drawings of Maralixibat chloride

STR1

 

 

ChemSpider 2D Image | maralixibat chloride | C40H56ClN3O4S

STR1Figure imgf000053_0001

It is well established that agents which inhibit the 20 transport of bile acids across the ileum can also cause a decrease in the level of cholesterol in blood serum. Stedronski, in “Interaction of bile acids and cholesterol with nonsystemic agents having hypocholesterolemic properties,” Biochimica et Biophysica Acta, 1210 (1994) 255- 25287, discusses biochemistry, physiology, and known active agents affecting bile acids and cholesterol.

A class of ileal bile acid transport-inhibiting compounds which was recently discovered to be useful for influencing the level of blood serum cholesterol is 30 tetrahydrobenzothiepine-l,l-dioxides (THBDO compounds). (U.S. Patent Application No. 08/816,065)

Some classes of compounds show enhanced potency as pharmaceutical therapeutics after they have been enantiomerically-enriched (see, for example, Richard B. Silverman, The Organic Chemistry of Drug Design and Drug Action, Academic Press, 1992, pp. 76-82) . Therefore, THBDO compounds that have been enantiomerically-enriched are of particular interest.

A class of chemistry useful as intermediates in the preparation of racemic THBDO compounds is tetrahydrobenzothiepine-1-oxides (THBO compounds) . THBDO compounds and THBO compounds possess chemical structures in which a phenyl ring is fused to a seven-member ring. A method of preparing enantiomerically-enriched samples of another phenyl/seven-member fused ring system, the benzothiazepines, is described by Higashikawa (JP 59144777) , where racemic benzothiazepine derivatives are optically resolved on a chromatographic column containing chiral crown ethers as a stationary phase. Although optical resolution is achieved, the Higashikawa method is limited to producing only small quantities of the enantiomerically-enriched benzothiazepine derivatives. Giordano (CA 2068231) reports the cyclization of (2S, 3S) -aminophenylthiopropionates in the presence of a phosphonic acid to produce (2S, 3S) -benzothiazepin-4-ones . However, that preparation is constrained by the need to use enantiomerically-enriched starting materials rather than racemic starting materials. In addition, the Giordano method controls the stereochemistry of the seven-member ring of the benzothiazepin-4-one only at the 2- and 3 -positions. The 4- and 5-positions of the seven-member ring of the benzothiazepin-4-one are not asymmetric centers, and the stereochemistry at these sites therefore cannot be controlled by the Giordano method. A method by which enantiomerically-enriched 1,5- benzothiazepin-3-hydroxy-4 (5H) -one compounds have been produced is through the asymmetric reduction of 1,5- benzothiazepin-3,4 (2H, 5H) -dione compounds, reported by Yamada, et al . (J. Org. Chem. 1996, 61 (24), 8586-8590). The product is obtained by treating the racemic 1,5- benzothiazepin-3,4 (2H, 5H) -dione with the reaction product of an optically active alpha-amino acid and a reducing agent, for example sodium borohydride. Although a product with high optical purity was achieved, the method is limited by the use of a relatively expensive chemical reduction step.

The microbial reduction of racemic 1, 5-benzothiazepin- 3 , 4 (2H, 5H) -dione compounds to produce enantiomerically- enriched 1, 5-benzothiazepin-3-hydroxy-4 (5H) -one compounds is reported by Patel et al . , U.S. Patent 5,559,017. This method is limited by the inherent problems of maintaining a viable and pure bacterial culture of the appropriate species and variety. In addition, that method is limited in scale, producing only microgram quantities of the desired product. Until now, there have been no reported processes for preparing enantiomerically-enriched THBDO compounds or enantiomerically-enriched THBO compounds. Furthermore, there have been no reported processes for controlling the stereochemistry at the 4- and 5-positions of the seven- member rings of THBDO compounds or THBO compounds

FDA Grants Breakthrough Designation to Shire’s Rare GI Therapies

Tue, 06/14/2016

Shire announced that the U.S. Food and Drug Administration (FDA) has granted Breakthrough Therapy Designation for two investigational products for rare diseases: SHP621 (budesonide oral suspension, or BOS) for eosinophilic esophagitis (EoE), and SHP625 (maralixibat) for progressive familial intrahepatic cholestasis type 2 (PFIC2).

“Receiving Breakthrough Therapy Designation on two pipeline products this past week reflects the potential of our strong and innovative pipeline of more than 60 programs,” said Flemming Ornskov, M.D., MPH, and CEO, Shire. “Shire is committed to bringing innovation to the rare and specialty areas we focus on. We persevere to see compounds through the many stages of development through their challenges and successes, and always keep patients with unmet needs top of mind.”

EoE is a serious, chronic and rare disease that stems from an elevated number of eosinophils, a type of white blood cell, that infiltrate the walls of the esophagus. EoE is characterized by an inflammation of the esophagus that may lead to difficulty swallowing (dysphagia). The diagnosed prevalence of EoE ranges from approximately 15-55 cases per 100,000 persons, with high-end estimates reported by studies in Western regions.

PFIC refers to a group of autosomal-recessive liver disorders of childhood that disrupt bile formation and present with cholestasis. The symptoms of PFIC include severe itching of the skin (pruritus), and jaundice. PFIC is estimated to affect 1 in 50,000 to 1 in 100,000 births. PFIC2 is the most common type of PFIC, accounting for around half of cases.

According to the FDA, Breakthrough Therapy Designation is granted to a therapy that is intended to treat a serious or life-threatening disease or condition and preliminary clinical evidence indicates that the drug may demonstrate substantial improvement on one or more clinically significant endpoints over current standard of care. Under the designation, the FDA provides intensive guidance, organizational commitment involving senior managers, and eligibility for rolling and priority review of the application; this process helps ensure patients have access to therapies as soon as possible, pending approval. Breakthrough Therapy Designation does not guarantee that FDA will ultimately approve BOS for EoE or maralixibat for PFIC2, and the timing of any such approval is uncertain.

“On behalf of patients in the United States with EoE and PFIC2, we are so pleased that the FDA has granted Breakthrough Therapy Designation to BOS and maralixibat,” said Philip J. Vickers, Ph.D., Head of R&D, Shire. “We look forward to working with the agency to continue their development and, pending FDA approval, deliver these therapeutic options to the patients who need them most.”

Source: Shire

Patent

WO 2003022804

It is well established that agents which inhibit the transport of bile acids across the tissue of the ileum can also cause a decrease in the levels of cholesterol in blood serum. Stedronski, in “Interaction of bile acids and cholesterol with nonsystemic agents having hypocholesterolemic properties,” Biochimica et Biophysica Acta, 1210 (1994) 255-287 discusses biochemistry, physiology, and known active agents surrounding bile acids and cholesterol. Bile acids are actively transported across the tissue of the ileum by an apical sodium co-dependent bile acid transporter (ASBT), alternatively known as an ileal bile acid transporter (IBAT).
A class of ASBT-inhibiting compounds that was recently discovered to be useful for influencing the level of blood serum cholesterol comprises tetrahydrobenzothiepine oxides (THBO compounds, PCT Patent Application No. WO 96/08484). Further THBO compounds useful as ASBT inhibitors are described in PCT Patent Application No. WO 97/33882.
Additional THBO compounds useful as ASBT inhibitors are described in U.S. Patent No. 5,994,391. Still further THBO compounds useful as ASBT inhibitors are described in PCT Patent Application No. WO 99/64409. Included in the THBO class are tetrahydrobenzo-thiepine-l -oxides and tetrahydrobenzothiepine- 1,1 -dioxides. THBO compounds possess chemical structures in which a phenyl ring is fused to a seven-member ring.

Published methods for the preparation of THBO compounds include the synthesis through an aromatic sulfone aldehyde intermediate. For example l-(2,2-dibutyl-3-oxopropylsulfonyl)-2-((4-methoxyphenyl)methyl)benzene (29) was cyclized with potassium t-butoxide to form tetrahydrobenzothiepine- 1,1 -dioxide (svn-24) as shown in Eq. 1.

Compound 29 was prepared by reacting 2-chloro-5-nitrobenzoic acid chloride with anisole in the presence of aluminum trichloride to produce a chlorobenzophenone compound; the chlorobenzophenone compound was reduced in the presence of trifluoromethanesulfonic acid and triethylsilane to produce a chlorodiphenylmethane compound; the
chlorodiphenylmethane compound was treated with lithium sulfide and 2,2-dibutyl-3-(methanesulfonato)propanal to produce l-(2,2-dibutyl-3-oxopropylthio)-2-((4-methoxyphenyl)methyl)-4-dimethylaminobenzene (40); and 40 was oxidized with m-chloroperbenzoic acid to produce 29. The first step of that method of preparing compound 29 requires the use of a corrosive and reactive carboxylic acid chloride that was prepared by the reaction of the corresponding carboxylic acid with phosphorus pentachloride.
Phosphorus pentachloride readily hydrolyzes to produce volatile and hazardous hydrogen chloride. The reaction of 2,2-dibutyl-3-(methanesulfonato)propanal with the lithium sulfide and the chlorodiphenylmethane compound required the intermediacy of a cyclic tin compound to make the of 2,2-dibutyl-3-(methanesulfonato)propanal. The tin compound is expensive and creates a toxic waste stream.
In WO 97/33882 compound syn-24 was dealkylated using boron tribromide to produce the phenol compound 28. Boron tribromide is a corrosive and hazardous material that generates hydrogen bromide gas and requires special handling. Upon hydrolysis, boron tribromide also produces borate salts that are costly and time-consuming to separate and dispose of.

An alternative method of preparing THBO compounds was described in WO
97/33882, wherein a 1,3-propanediol was reacted with thionyl chloride to form a cyclic sulfite compound. The cyclic sulfite compound was oxidized to produce a cyclic sulfate compound. The cyclic sulfate was condensed with a 2-methylthiophenol that had been deprotonated with sodium hydride. The product of the condensation was a (2-methylphenyl) (3′-hydroxypropyl)thioether compound. The thioether compound was oxidized to form an thioether aldehyde compound. The thioether aldehyde compound was further oxidized to form an aldehyde sulfone compound which in turn was cyclized in the presence of potassium t-butoxide to form a 4-hydroxytetrahydrobenzothiepine 1,1 -dioxide compound. This cyclic sulfate route to THBO compounds requires an expensive catalyst. Additionally it requires the use of SOCI2, which in turn requires special equipment to handle.
PCT Patent Application No. WO 97/33882 describes a method by which the phenol compound 28 was reacted at its phenol hydroxyl group to attach a variety of functional groups to the molecule, such as a quaternary ammonium group. For example, (4R,5R)-28 was reacted with l,4-bis(chloromethyl)benzene (?,??’-dichloro-p-xylene) to produce the chloromethyl benzyl- ether (4R,5R)-27. Compound (4R,5R)-27 was treated with diazabicyclo[2.2.2]octane (DABCO) to produce (4R,5R)-l-((4-(4-(3,3-dibutyl-7-(dimemylamino)-2,3,4,5-tetrahydro-4-hydroxy-l , 1 -dioxido-1 -benzothiepin-5-yl)phenoxy)methyl)phenyl)methyl-4-aza-l-azomabicyclo[2.2.2]octane chloride (41). This method suffers from low yields because of a propensity for two molecules of compound (4R,5R)-28 to react with one molecule of l,4-bis(chloromethyl)benzene to form a bis(benzothiepine) adduct. Once the bis-adduct forms, the reactive chloromethyl group of compound (4R,5R)-27 is not available to react with an amine to form the quaternary ammonium product.

A method of preparing enantiomerically enriched tetrahydrobenzothiepine oxides is described in PCT Patent Application No. WO 99/32478. In that method, an aryl-3- hydroxypropylsulfide compound was oxidized with an asymmetric oxidizing agent, for example (lR (->(8,9-dichloro-10-camphorsulfonyl)oxaziridine, to yield a chiral aryl-3-hydroxypropylsulfoxide. Reaction of the aryl-3-hydroxypropylsulfoxide with an oxidizing agent such as sulfur trioxide pyridine complex yielded an aryl-3-propanalsulfoxide. The aryl- 3-propanalsulfoxide was cyclized with a base such as potassium t-butoxide to
enantioselectively produce a tetrahydrobenzothiepine- 1 -oxide. The tetrahydrobenzothiepine- 1 -oxide was further oxidized to produce a tetrahydrobenzothiepine- 1 , 1 -dioxide. Although this method could produce tetrahydrobenzothiepine- 1,1 -dioxide compounds of high enantiomeric purity, it requires the use of an expensive asymmetric oxidizing agent.
Some 5-amidobenzothiepine compounds and methods to make them are described in

PCT Patent Application Number WO 92/18462.
In Svnlett. 9, 943-944(1995) 2-bromophenyl 3-benzoyloxy-l-buten-4-yl sulfone was treated with tributyl tin hydride and AIBN to produce 3-benzoyloxytetrahydrobenzothiepine-1,1 -dioxide.
In addition to forming the desired ASBT inhibitors, it is also desirable to form such

ASBT inhibitors of higher purity and having lower levels of residual solvent impurities. This is especially so with respect to ASBT inhibitors having a positively charged substituent, for example, the compounds designated as 41 (supra) and 60 (infra).
It is further desirable to provide methods for making such high purity ASBT inhibitors.

Example 11.

Preparation of (4R,5R)- 1 -((4-(4-(3,3-dibutyl-7-(dimemylamino)-2,3,4,5-tetrahydro-4- hydroxy- 1 , 1 -diυxido- 1 -benzithiepin-5-yl)pheπoxy)methyl)phenyl)methyl-4-aza- 1 – azoniabicyclo[2.2.2]octane chloride,
41


41

Ste l. Preparation of (4R.5R1-26.


( 4R, 5R) -26
A 1000 mL 4 neck jacketed Ace reactor flask was fitted with a mechanical stirrer, a nitrogen inlet, an addition funnel or condenser or distilling head with receiver, a
thermocouple, four internal baffles and a 28 mm Teflon turbine agitator. The flask was purged with nitrogen gas and charged with 25.0 grams of (4R,5R)-28 and 125 mL of N,N-dimethylacetamide (DMAC). To this was added 4.2 grams of 50% sodium hydroxide. The mixture was heated to 50°C and stiπed for 15 minutes. To the flask was added 8.3 grams of 55 dissolved in 10 mL of DMAC, all at once. The temperature was held at 50°C for 24 hrs. To the flask was added 250 mL of toluene followed by 125 mL of dilution water. The mixture was stiπed for 15 minutes and the layers were then allowed to separate at 50°C. The flask was then charged with 125 mL of saturated sodium chloride solution and stiπed 15 minutes. Layers separated cleanly in 30 seconds at 50°C. Approximately half of the solvent was distilled off under vacuum at 50°C. The residual reaction mixture contained (4R,5R)-26.

Step 2. Preparation of (4R.5RV27.


( 4R, 5R) -27
Toluene was charged back to the reaction mixture of Step 1 and the mixture was cooled to 35°C. To the mixture was then added 7.0 grams of thionyl chloride over 5 minutes. The reaction was exothermic and reached 39°C. The reaction turned cloudy on first addition of thionyl chloride, partially cleared then finally remained cloudy. The mixture was stirred for 0.5 hr and was then washed with 0.25N NaOH. The mixture appeared to form a small amount of solids that diminished on stirring, and the layers cleanly separated. The solvent was distilled to a minimum stir volume under vacuum at 50°C. The residual reaction mixture contained (4R,5R)-27.

Step 3. Preparation of 41.
To the reaction mixture of Step 2 was charged with 350 mL of methyl ethyl ketone (MEK) followed by 10.5 mL water and 6.4 grams of diazabicyclo[2.2.2]octane (DABCO) dissolved in 10 mL of MEK. The mixture was heated to reflux, and HPLC showed <0.5% of (4R,5R)-27. The reaction remained homogenous initially then crystallized at the completion of the reaction. An additional 5.3 mL of water was charged to the flask to redissolve product. Approximately 160 mL of solvent was then distilled off at atmospheric pressure. The mixture started to form crystals after 70 mL of solvent was distilled. Water separated out of distillate indicating a ternary azeotrope between toluene, water and methyl ethyl ketone (MEK). The mixture was then cooled to 25°C. The solids were filtered and washed with 150 mL MEK, and let dry under vacuum at 60°C. Isolated 29.8.0 g of off-white crystalline 4 Example 11a.
Alternate Preparation of (4R,5R)-l-((4-(4-(3,3-dibutyl-7-(dimemylamino)-2,3,4,5-tetrahydro- 4-hydroxy- 1 , 1 -dioxido- 1 -benzithiepin-5-yl)phenoxy)methyl)phenyl)methyl-4-aza- 1 – azoniabicyclo[2.2.2]octane chloride, Form II of 41

A 1000 mL 4 neck jacketed Ace reactor flask is fitted with a mechanical stiπer, a nitrogen inlet, an addition funnel or condenser or distilling head with receiver, a
thermocouple, four internal baffles and a 28 mm Teflon turbine agitator. The flask is purged with nitrogen gas and charged with 25.0 grams of (4R,5R)-28 and 100 mL of N,N-dimethylacetamide (DMAC). The mixture is heated to 50°C and to it is added 4.02 grams of 50% sodium hydroxide. The mixture is stiπed for 30 minutes. To the flask is added 8.7 grams of 55 dissolved in 12.5 mL of DMAC, all at once. The charge vessel is washed with 12.5 mL DMAC and the wash is added to the reactor. The reactor is stiπed for 3 hours. To the reactor is added 0.19 mL of 49.4% aq. NaOH and the mixture is stirred for 2 hours. To the mixture is added 0.9 g DABCO dissolved in 12.5 mL DMAC. The mixture is stiπed 30 to 60 minutes at 50°C. To the flask is added 225 mL of toluene followed by 125 mL of dilution water. The mixture is stiπed for 15 minutes and the layers are then allowed to separate at 50°C. The bottom aqueous layer is removed but any rag layer is retained. The flask is then charged with 175 mL of 5% hydrochloric acid solution and stiπed 15 minutes. Layers are separated at 50°C to remove the bottom aqueous layer, discarding any rag layer with the aqueous layer. Approximately half of the solvent is distilled off under vacuum at a maximum pot temperature of 80°C. The residual reaction mixture contains (4R,5R)-26.

Step 2. Preparation of (4R.5RV27.

Toluene (225 mL) is charged back to the reaction mixture of Step 1 and the mixture is cooled to 30°C. To the mixture is then added 6.7 grams of thionyl chloride over 30 to 45 minutes. The temperature is maintained below 35°C. The reaction turns cloudy on first addition of thionyl chloride, then at about 30 minutes the layers go back together and form a clear mixture. The mixture is stiπed for 0.5 hr and is then charged with 156.6 mL of 4% NaOH wash over a 30 minute period. The addition of the wash is stopped when the pH of the mixture reaches’ 8.0 to 10.0. The bottom aqueous layer is removed at 30°C and any rag layer is retained with the organic layer. To the mixture is charged 175 mL of saturated NaCl wash with agitation. The layers are separated at 30°C and the bottom aqueous layer is removed, discarding any rag layer with the aqueous layer. The solvent is distilled to a minimum stir volume under vacuum at 80°C. The residual reaction mixture contains (4R,5R)-27.

Step 3. Preparation of 41.
To the reaction mixture of Step 2 is charged 325 mL of methyl ethyl ketone (MEK) and 13 mL water. Next, the reactor is charged 6.2 grams of diazabicyclo[2.2.2]octane (DABCO) dissolved in 25 mL of MEK. The mixture is heated to reflux and held for 30 minutes. Approximately 10% of solvent volume is then distilled off. The mixture starts to form crystals during distillation. The mixture is then cooled to 20°C for 1 hour. The off-white crystalline 41 (Form U) is filtered and washed with 50 mL MEK, and let dry under vacuum at 100°C.

Example lib.
Alternate Preparation of (4R,5R)-1 -((4-(4-(3,3-dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro- 4-hydroxy- 1 , 1 -dioxido- 1 -benzithiepin-5-yl)phenoxy)methyl)phenyl)methyl-4-aza- 1 – azoniabicyclo[2.2.2]octane chloride, Form II of 41

A 1000 mL 4 neck jacketed Ace reactor flask is fitted with a mechanical stiπer, a nitrogen inlet, an addition funnel or condenser or distilling head with receiver, a
thermocouple, four internal baffles and a Teflon turbine agitator. The flask is purged with nitrogen gas and charged with 25.0 grams of (4R,5R)-28 and 125 mL of N,N-dimethylacetamide (DMAC). The mixture is heated to 50°C and to it is added 7.11 grams of 30% sodium hydroxide over a period of 15 to 30 minutes with agitation. The mixture is stiπed for 30 minutes. To the flask is added 9.5 grams of solid 55. The reactor is stiπed for 3 hours. To the mixture is added 1.2 g of solid DABCO. The mixture is stiπed 30 to 60 minutes at 50°C. To the flask is added 225 mL of toluene followed by 125 mL of water. The mixture is stirred for 15 minutes and the layers are then allowed to separate at 50°C. The bottom aqueous layer is removed but any rag layer is retained with the organic layer. The flask is then charged with 175 mL of 5% hydrochloric acid solution and stirred 15 minutes. Layers are separated at 50°C to remove the bottom aqueous layer, discarding any rag layer with the aqueous layer. The flask is then charged with 225 mL of water and stirred 15 minutes. The layers are allowed to separate at 50°C. The bottom aqueous layer is removed, discarding any rag layer with the aqueous layer. Approximately half of the solvent is distilled off under vacuum at a maximum pot temperature of 80°C. The residual reaction mixture contains (4R,5R)-26.

Step 2. Preparation of (4R.5RV27.

Toluene (112.5 mL) is charged back to the reaction mixture of Step 1 and the mixture is cooled to 25°C. To the mixture is then added 7.3 grams of thionyl chloride over 15 to 45 minutes. The temperature of the mixture is maintained above 20°C and below 40°C. The reaction turns cloudy on first addition of thionyl chloride, then at about 30 minutes the layers go back together and form a clear mixture. The mixture is then charged with 179.5 mL of 4% NaOH wash over a 30 minute period. The mixture is maintained above 20°C and below 40°C during this time. The addition of the wash is stopped when the pH of the mixture reaches 8.0 to 10.0. The mixture is then allowed to separate at 40°C for at least one hour.

The bottom aqueous layer is removed and any rag layer is retained with the organic layer. To the mixture is charged 200 mL of dilution water. The mixture is stiπed for 15 minutes and then allowed to separate at 40°C for at least one hour. The bottom aqueous layer is removed, discarding any rag layer with the aqueous layer. The solvent is distilled to a minimum stir volume under vacuum at 80°C. The residual reaction mixture contains (4R,5R)-2 .

Step 3. Preparation of 41.
To the reaction mixture of Step 2 is charged 350 mL of methyl ethyl ketone (MEK) and 7 mL water. The mixture is stiπed for 15 minutes and the temperature of the mixture is adjusted to 25°C. Next, the reactor is charged with 6.7 grams of solid
diazabicyclo[2.2.2]octane (DABCO). The mixture is maintained at 25°C for three to four hours. It is then heated to 65°C and maintained at that temperature for 30 minutes. The mixture is then cooled to 25°C for 1 hour. The off-white crystalline 41 (Form II) is filtered and washed with 50 mL MEK, and let dry under vacuum at 100°C.

Example 12.
Alternate preparation of (4R,5R)-1 -((4-(4-(3,3-dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro- 4-hydroxy- 1 , 1 -dioxido- 1 -benzithiepin-5-yl)phenoxy)methyl)phenyl)methyl-4-aza- 1 – azoniabicyclo[2.2.2]octane chloride, Form I of 41

(4R,5R)-27 (2.82 kg dry basis, 4.7 mol) was dissolved in MTBE (9.4 L). The solution of (4R,5R)-22 was passed through a 0.2 mm filter cartridge into the feeding vessel. The flask and was rinsed with MTBE (2 x 2.5 L). The obtained solution as passed through the cartridge filter and added to the solution of (4R,5R)-2 in the feeding vessel. DABCO
(diazabicyclo[2.2.2]octane, 0.784 kg, 7.0 mol) was dissolved in MeOH (14.2 L). The DABCO solution was passed through the filter cartridge into the 100 L nitrogen-flushed reactor. The Pyrex bottle and the cartridge filter were rinsed with MeOH (7.5 L) and the solution was added to the reactor. The (4R,5R)-22 solution was added from the feeding vessel into the reactor at 37°C over a period of 10 min, while stirring. Methanol (6.5 L) was added to the Pyrex bottle and via the cartridge filter added to the feeding vessel to rinse the remaining (4R,5R)-2 into the reactor. The reaction mixture was brought to 50-60°C over 10-20 min and stiπed at that temperature for about 1 h. The mixture was cooled to 20-25°C over a period of 1 h. To the reaction mixture, methyl t-butyl ether (MTBE) (42 L) was added over a period of 1 h and stiπed for a minimum of 1 h at 20 – 25°C. The suspension was filtered through a Buchner funnel. The reactor and the filter cake were washed with MTBE (2 x 14 L). The solids were dried on a rotary evaporator in a 20 L flask at 400 – 12 mbar, 40°C, for 22 h. A white crystalline solid was obtained. The yield of 4 . (Form I) was 3.08 kg (2.97 kg dry, 93.8 %) and the purity 99.7 area % (HPLC; Kromasil C 4, 250 x 4.6 mm column; 0.05% TFA in H2O/0.05% TFA in ACN gradient, UV detection at 215 nm).

Example 12a.
Conversion of Form I of Compound 41 into Form II of Compound 41.

To 10.0 grams of Form I of 4 . in a 400 mL jacketed reactor is added 140 mL of MEK. The reactor is stirred (358 φm) for 10 minutes at 23 °C for 10 minutes and the stirring rate is then changed to 178 φm. The suspension is heated to reflux over 1 hour using a programmed temperature ramp (0.95°C/minute) using batch temperature control (cascade mode). The delta Tmaχ is set to 5°C. The mixture is held at reflux for 1 hour. The mixture is cooled to

25°C. After 3 hours at 25°C, a sample of the mixture is collected by filtration. Filtration is rapid (seconds) and the filtrate is clear and colorless. The white solid is dried in a vacuum oven (80°C, 25 in. Hg) to give a white solid. The remainder of the suspension is stirred at 25°C for 18 hours. The mixture is filtered and the cake starts to shrink as the mother liquor reaches the top of the cake. The filtration is stopped and the reactor is rinsed with 14 mL of MEK. The reactor stirrer speed is increased from 100 to 300 φm to rinse the reactor. The rinse is added to the filter and the solid is dried with a rapid air flow for 5 minutes. The solid is dried in a vacuum oven at 25 in. Hg for 84 hours to give Form II of 4

PATENT

WO 2014144650

Scheme 3:

PAPER

Journal of Medicinal Chemistry (2005), 48(18), 5853-5868

Discovery of Potent, Nonsystemic Apical Sodium-Codependent Bile Acid Transporter Inhibitors (Part 2)

Department of Discovery Chemistry and Department of Cardiovascular Disease, Pharmacia, 700 Chesterfield Parkway W, Chesterfield, Missouri 63017, Office of Science and Technology, Chemical Science Division, Pharmacia, 800 Lindbergh Boulevard, Creve Coeur, Missouri 63167, Department of Pharmaceutical Sciences, Pharmacia, Skokie, Illinois, and Department of Chemistry, University of Missouri, St. Louis, Missouri
J. Med. Chem., 2005, 48 (18), pp 5853–5868
DOI: 10.1021/jm0402162

http://pubs.acs.org/doi/abs/10.1021/jm0402162

Abstract

Abstract Image

In the preceding paper several compounds were reported as potent apical sodium-codependent bile acid transporter (ASBT) inhibitors. Since the primary site for active bile acid reabsorption is via ASBT, which is localized on the luminal surface of the distal ileum, we reasoned that a nonsystemic inhibitor would be desirable to minimize or eliminate potential systemic side effects of an absorbed drug. To ensure bioequivalency and product stability, it was also essential that we identify a nonhygroscopic inhibitor in its most stable crystalline form. A series of benzothiepines were prepared to refine the structure−activity relationship of the substituted phenyl ring at the 5-position of benzothiepine ring and to identify potent, crystalline, nonhygroscopic, and efficacious ASBT inhibitors with low systemic exposure.

compd R IC50 (nM)b hygroscp I wt gain (%)c hygroscp II % wt gain (%)d crystallinitye
74 OCH2C6H4(p)CH2(N+)DB 0.28 1.59 2.1 yes

(4Rcis)-1-[[4-[[4-[3,3-Dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro-4-hydroxy-1,1-dioxido-1-benzothiepin-5-yl]phenoxy]methyl]phenyl]methyl]-4-aza-1-azoniabicyclo[2.2.2]octane Chloride Salt (74). Following a similar procedure as in General Method B, the title compound 74 was prepared from the corresponding chloromethyl benzyl ether and DABCO as a white solid, mp 223−230 °C (dec); 1H NMR (CDCl3) δ 0.89 (m, 6H), 1.27−1.52 (br m, 10H), 1.63 (m, 1H), 2.20 (m, 1H), 2.81 (s, 6H), 3.06 (ABq, JAB = 15.1 Hz, J = 43.3 Hz, 2H), 3.16 (s, 6H), 3.76 (s, 6H), 4.11 (d, J = 7.7 Hz, 1H), 5.09 (s, 2H), 5.14 (s, 2H), 5.48 (s, 1H), 5.96 (s, 1H), 6.49 (d, J = 8.9 Hz, 1H), 6.99 (d, J = 8.0 Hz, 2H), 7.26 (m, 1H), 7.44 (d, J = 8.0 Hz, 2H), 7.52 (d, J = 7.4 Hz, 2H), 7.68 (d, J = 7.4 Hz, 2H), 7.87 (d, J = 8.9 Hz, 1H). HRMS calcd for C40H56N3O4S:  674.3992; found, 674.4005. Anal. Calcd for C40H56N3O4S:  ‘ C, 67.62; H, 7.95; N, 5.92; S, 4.51. Found:  C, 67.48; H, 8.32; N, 5.85; S, 4.60.

a All compounds were prepared using method B in Scheme 3.b Taurocholate is transported across the baby hamster kidney cell membrane.c % weight gain in a 25 °C, 57% humidity chamber for 2 weeks.d % weight gain in a 40 °C, 80% humidity chamber for 2 weeks.e Crystallinity as determined by X-ray powder diffraction analysis.f (N+)DB is a DABCO terminal group with the quaternary ammonium attached to the linke

ANY ERROR EMAIL amcrasto@gmail.com, +919323115463

PATENT

https://www.google.com/patents/WO1999032478A1?cl=en

Inventors James Li, Ching-Cheng Wang, David B. Reitz, Victor Snieckus, Horng-Chih Huang,Andrew J. Carpenter,
Applicant G.D. Searle & Co.

Example 10. Preparation of enantiomerically-enriched (4R.5R)-1- r.4- r _4- .3.3 -Dibutyl-7- (dimethylamino) -2.3 ,4.5- tetrahydro-4-hydroxy-1, l-dioxido-l-benzothiepin-5- yl] henoxy] ethyl] phenyl1methyl] -4-aza-l- azoniabicyclo [2.2.2] octane chloride ( (4R,5R) -XXVII) ♦

Figure imgf000053_0001

( (4R,5R) -XXVII) * = chiral center

Step 1. Preparation of 4-flUoro-2- ( (4- methoxyphenyl) methyl) -phenol To a stirred solution of 23.66 g of 95% sodium hydride (0.94 mol) in 600 mL of dry toluene was added 100.0 g of 4- fluorophenol (0.89 mol) at 0°C. The mixture was stirred at 90°C for 1 hour until gas evolution stopped. The mixture was cooled down to room temperature and a solution of 139.71 g of 3 -methoxybenzyl chloride (0.89 mol) in 400 mL of dry toluene was added. After refluxing for 24 hours, the mixture was cooled to room temperature and quenched with 500 mL of water. The organic layer was separated, dried over MgS04, and concentrated under high vacuum. The remaining starting materials were removed by distillation. The crude dark red oil was filtered through a layer of 1 L of silica gel with neat hexane to yield 53.00 g (25.6%) of the product as a pink solid: *H NMR (CDC13) d 3.79 (s, 3H) , 3.90 (s, 2H) , 4.58 (s, IH) , 6.70-6.74 (m, IH) , 6.79-6.88 (m, 4H) , 7.11-7.16 (m, 2H) .

Step 2. Preparation of 4-fluoro-2- ( (4- methoxyphenyl) methyl) -thiophenol

Step 2a. Preparation of thiocarbamate To a stirred solution of 50.00 g (215.30 mmol) of 4- fluoro-2- ( ( -methoxyphenyl) methyl) -phenol in 500 mL of dry DMF was added 11.20 g of 60% sodium hydride dispersion in mineral oil (279.90 mmol) at 2°C. The mixture was allowed to warm to room temperature and 26.61 g of dimethylthiocarbamoyl chloride (215.30 mmol) was added. The reaction mixture was stirred at room temperature overnight. The mixture was quenched with 100 mL of water in an ice bath. The solution was extracted with 500 mL of diethyl ether. The ether solution was washed with 500 mL of water and 500 mL of brine. The ether solution was dried over MgS04 and stripped to dryness. The crude product was filtered through a plug of 500 mL silica gel using 5% ethyl acetate/hexane to yield 48.00 g (69.8%) of the product as a pale white solid: XH NMR (CDC13) d 3.21 (s, 3H) , 3.46 (s, 3H) , 3.80 (s, 3H) , 3.82 (s, 2H) , 6.78-6.86 (m, 3H) , 6.90- 7.00 (m, 2H) , 7.09 (d, J = 8.7 Hz, 2H) .

Step 2b. Rearrangement and hydrolysis of thiocarbamate to 4-fluoro-2- ( (4 -methoxyphenyl) methyl) -thiophenol A stirred solution of 48.00 g (150.29 mmol) of thiocarbamate (obtained from Step 2a) in 200 mL of diphenyl ether was refluxed at 270°C overnight. The solution was cooled down to room temperature and filtered through 1 L of silica gel with 2 L of hexane to remove phenyl ether. The rearrangement product was washed with 5% ethyl acetate/hexane to give 46.00 g (95.8%) of the product as a pale yellow solid: XH NMR (CDC13) d 3.02 (s, 3H) , 3.10 (s, 3H) , 3.80 (s, 3H) , 4.07 (s, 2H) , 6.82-6.86 (m, 3H) , 6.93 (dt, J = 8.4 Hz, 2.7 Hz, IH) , 7.08 (d, J = 8.7 Hz, 2H) , 7.49 (dd, J = 6.0 Hz, 8.7 Hz, IH) . To a solution of 46.00 g (144.02 mmol) of the rearrangement product (above) in 200 mL of methanol and 200 mL of THF was added 17.28 g of NaOH (432.06 mmol) . The mixture was refluxed under nitrogen overnight . The solvents were evaporated off and 200 mL of water was added. The aqueous solution was washed with 200 mL of diethyl ether twice and placed in an ice bath. The aqueous mixture was acidified to pH 6 with concentrated HCl solution. The solution was extracted with 300 mL of diethyl ether twice. The ether layers were combined, dried over MgS04 and stripped to dryness to afford 27.00 g (75.5%) of the product as a brown oil: XH NMR (CDC13) d 3.24 (s, IH) , 3.80 (s, 3H) , 3.99 (s, 2H) , 6.81-6.87 (m, 4H) , 7.09 (d, J = 8.7 Hz, 2H) , 7.27- 7.33 (m, IH) .

Step 3. Preparation of dibutyl cyclic sulfate

Step 3a. Preparation of 2 , 2-dibutyl-l, 3-propanediol . To a stirred solution of di-butyl-diethylmalonate (Aldrich) (150g, 0.55 mol in dry THF (700ml) in an acetone/dry ice bath was added LAH (1 M THF) 662 ml (1.2 eq. , 0.66 mol) dropwise maintaining the temperature between -20 to 0°C. The reaction was stirred at RT overnight. The reaction was cooled to -20°C and 40 ml of water, and 80 mL of 10% NaOH and 80 ml of water were added dropwise. The resulting suspension was filtered. The filtrate was dried over sodium sulphate and concentrated in vacuo to give diol 598.4 g (yield 95%) as an oil. MS spectra and proton and carbon NMR spectra were consistent with the product.

Step 3b. Preparation of dibutyl cyclic sulfite

A solution of 2 , 2-dibutyl-l, 3-propanediol (103 g, 0.548 0 mol, obtained from Step 3a) and triethylamine (221 g, 2.19 mol) in anhydrous methylene chloride (500 ml) was stirred at 0°C under nitrogen. To the mixture, thionyl chloride (97.8* g, 0.82 mol) was added dropwise and within 5 min the solution turned yellow and then black when the addition was 5 completed within half an hour. The reaction mixture was stirred for 3 hrs. at 0°C. GC showed that there was no starting material left. The mixture was washed with ice water twice then with brine twice . The organic phase was dried over magnesium sulfate and concentrated under vacuum 0 to give 128 g (100%) of the dibutyl cyclic sulfite as a black oil. Mass spectrum (MS) was consistent with the product .

Step 3c. Oxidation of dibutyl cyclic sulfite to 5 dibutyl cyclic sulfate

To a solution of the dibutyl cyclic sulfite (127.5 g , 0.54 mol, obtained from Step 3b) in 600 ml acetonitrile and 500 ml of water cooled in an ice bath under nitrogen was added ruthenium (III) chloride (1 g) and sodium periodate 0 (233 g, 1.08 mol) . The reaction was stirred overnight and the color of the solution turned black. GC showed that there was no starting material left. The mixture was extracted with 300 ml of ether and the ether extract was washed three times with brine. The organic phase was dried over magnesium sulfate and passed through celite. The filtrate was 5 concentrated under vacuum and to give 133 g (97.8%) of the dibutyl cyclic sulfate as an oil. Proton and carbon NMR and MS were consistent with the product.

Step 4. Preparation of aryl-3-hydroxypropylsulfide

10 To a stirred solution of 27.00 g (108.73 mmol) of 4- fluoro-2- ( (4-methoxyphenyl) methyl) thiophenol (obtained from Step 2) in 270 mL of diglyme was added 4.35 g of 60% sodium-, hydride dispersion in mineral oil (108.73 mmol) at 0°C. After gas evolution ceased, 29.94 g (119.60 mmol) of the

15 dibutyl cyclic sulfate (obtained from Step 3c) was added at 0°C and stirred for 10 minutes. The mixture was allowed to warm up to room temperature and stirred overnight. The solvent was evaporated and 200 mL of water was added. The solution was washed with 200 mL of diethyl ether and added

2025 mL of concentrated sulfuric acid to make a 2.0 M solution that was refluxed overnight. The solution was extracted with ethyl acetate and the organic solution was dried over MgS04 and concentrated in vacuo. The crude aryl-3 – hydroxypropylsulfide was purified by silica gel

25 chromatography (Waters Prep 500) using 8% ethyl acetate/hexane to yield 33.00 g (72.5%) of the product as a light brown oil: E NMR (CDC13) d 0.90 (t, J = 7.1 Hz, 6H) , 1.14-1.34 (m, 12H) , 2.82 (s, 2H) , 3.48 (s, 2H) , 3.79 (s, 3H) , 4.10 (s, 2H) , 6.77-6.92 (m, 4H) , 7.09 (d, J = 8.7 Hz,

302H) , 7.41 (dd, J = 8.7 Hz, 5.7 Hz, IH) . Step 5. Preparation of enantiomerically-enriched aryl-3 – hydroxypropylsulfoxide

To a stirred solution of 20.00 g (47.78 mmol) of aryl- 53 -hydroxypropylsulfide (obtained from Step 4) in 1 L of methylene chloride was added 31.50 g of 96% (12?) – ( -) – (8 , 8- dichloro-10-camphor-sulfonyl) oxaziridine (100.34 mmol, Aldrich) at 2°C. After all the oxaziridine dissolved the mixture was placed into a -30 °C freezer for 72 hours. The

10 solvent was evaporated and the crude solid was washed with 1 L of hexane. The white solid was filtered off and the hexane solution was concentrated in vacuo. The crude oil was purified on a silica gel column (Waters Prep 500) using 15% ethyl acetate/hexane to afford 19.00 g (95%) of the

15 enantiomerically-enriched aryl-3 -hydroxypropylsulfoxide as a colorless oil: lH NMR (CDC13) d 0.82-0.98 (m, 6H) , 1.16-1.32 (m, 12H) , 2.29 (d, J – 13.8 Hz, IH) , 2.77 (d, J = 13.5 Hz, IH) , 3.45 (d, J = 12.3 Hz, IH) , 3.69 (d, J = 12.3 Hz, IH) , 3.79 (s, 3H) , 4.02 (q, J = 15.6 Hz, IH) , 6.83-6.93 (m, 3H) ,

207.00 (d, J = 8.1 Hz, 2H) , 7.18-7.23 (m, IH) , 7.99-8.04 (m, IH) . Enantiomeric excess was determined by chiral HPLC on a (2?,2?) -Whelk-0 column using 5% ethanol/hexane as the eluent. It showed to be 78% e.e. with the first eluting peak as the major product.

25

Step 6. Preparation of enantiomerically-enriched aryl-3- propanalsulfoxide

To a stirred solution of 13.27 g of triethylamine (131.16 mmol, Aldrich) in 200 mL dimethyl sulfoxide were

30 added 19.00 g (43.72 mmol) of enantiomerically-enriched aryl-3 -hydroxypropylsulfoxide (obtained from Step 5) and 20.96 g of sulfur trioxide-pyridine (131.16 mmol, Aldrich) at room temperature. After the mixture was stirred at room temperature for 48 hours, 500 mL of water was added to the mixture and stirred vigorously. The mixture was then 5 extracted with 500 mL of ethyl acetate twice. The ethyl acetate layer was separated, dried over MgS04, and concentrated in vacuo. The crude oil was filtered through 500 mL of silica gel using 15% ethyl acetate/hexane to give 17.30 g (91%) of the enantiomerically-enriched aryl-3-

10 propanalsulfoxide as a light orange oil: lE NMR (CDC13) d 0.85-0.95 (m, 6H) , 1.11-1.17 (m, 4H) , 1.21-1.39 (m, 4H) , 1.59-1.76 (m, 4H) , 1.89-1.99 (m, IH) , 2.57 (d, J = 14.1 Hz, IH) , 2.91 (d, J = 13.8 Hz, IH) , 3.79 (s, 3H) , 3.97 (d, J = 15.9 Hz, IH) , 4,12 (d, J = 15.9 Hz, IH) , 6.84-6.89 (m, 3H) ,

157.03 (d, J = 8.4 Hz, 2H) , 7.19 (dt, J = 8.4 Hz, 2.4 Hz, IH) , 8.02 (dd, J = 8.7 Hz, 5.7 Hz, IH) , 9.49 (s, IH) .

Step 7. Preparation of the enantiomerically-enriched tetrahydrobenzothiepine-1-oxide (4R, 5R)

20 To a stirred solution of 17.30 g (39.99 mmol) of enantiomerically-enriched aryl-3 -propanalsulfoxide (obtained from Step 6) in 300 mL of dry THF at -15°C was added 48 mL of 1.0 M potassium t-butoxide in THF (1.2 equivalents) under nitrogen. The solution was stirred at -15°C for 4 hours.

25 The solution was then quenched with 100 mL of water and neutralized with 4 mL of concentrated HCl solution at 0°C. The THF layer was separated, dried over MgS04, and concentrated in vacuo. The enantiomerically-enriched tetrahydrobenzothiepine-1-oxide (4R,5R) was purified by

30 silica gel chromatography (Waters Prep 500) using 15% ethyl acetate/hexane to give 13.44 g (77.7%) of the product as a white solid: ‘H NMR (CDC13) d 0.87-0.97 (m, 6H) , 1.16-1.32 (m, 4H) , 1.34-1.48 (m, 4H) , 1.50-1.69 (m, 4H) , 1.86-1.96 (m, IH) , 2.88 (d, J = 13.0 Hz, IH) , 3.00 (d, J = 13.0 Hz, IH) , 3.85 (s, 3H) , 4.00 (s, IH) , 4.48 (s, IH) , 6.52 (dd, J = 9.9 5Hz, 2.4 Hz, IH) , 6.94 (d, J = 9 Hz, 2H) , 7.13 (dt, J = 8.4 Hz, 2.4 Hz, IH) , 7.38 (d, J = 8.7 Hz, 2H) , 7.82 (dd, J = 8.7 Hz, 5.7 Hz, IH) .

Step 8. Preparation of enantiomerically-enriched

10 tetrahydrobenzothiepine-1, 1-dioxide (4R, 5R)

To a stirred solution of 13.44 g (31.07 mmol) of enantiomerically-enriched tetrahydrobenzothiepine-1-oxide (obtained from Step 7) in 150 mL of methylene chloride was added 9.46 g of 68% m-chloroperoxybenzoic acid (37.28 mmol,

15 Sigma) at 0 °C. After stirring at 0 °C for 2 hours, the mixture was allowed to warm up to room temperature and stirred for 4 hours. 50 mL of saturated Na2S03 was added into the mixture and stirred for 30 minutes. The solution was then neutralized with 50 mL of saturated NaHC03 solution.

20 The methylene chloride layer was separated, dried over MgS04, and concentrated in vacuo to give 13.00 g (97.5%) of the enantiomerically-enriched tetrahydrobenzothiepine-1, 1- dioxide (4R,5R) as a light yellow solid: ‘H NMR (CDC13) d 0.89-0.95 (m, 6H) , 1.09-1.42 (m, 12H) , 2.16-2.26 (m, IH) ,

253.14 (q, J = 15.6 Hz, IH) , 3.87 (s, 3H) , 4.18 (s, IH) , 5.48 (s, IH) , 6.54 (dd, J = 10.2 Hz, 2.4 Hz, IH) , 6.96-7.07 (m, 3H) , 7.40 (d, J = 8.1 Hz, 2H) , 8.11 (dd, J = 8.6 Hz, 5.9 Hz, IH) .

30 Step 9. Preparation of enantiomerically-enriched 7-

(dimethylamino) tetrahydrobenzothiepine-1 , 1-dioxide (4R.5R) – To a solution of 13.00 g (28.98 mmol) of enantiomerically-enriched tetrahydrobenzothiepine-1, 1- dioxide (obtained from Step 8) in 73 mL of dimethylamine (2.0 M in THF, 146 mmol) in a Parr Reactor was added ca . 20 5 mL of neat dimethylamine . The mixture was sealed and stirred at 110 °C overnight, and cooled to ambient temperature. The excess dimethylamine was evaporated. The crude oil was dissolved in 200 mL of ethyl acetate and washed with 100 mL of water, dried over MgS04 and

10 concentrated in vacuo. Purification on a silica gel column (Waters Prep 500) using 20% ethyl acetate/hexane gave 12.43 g (90.5%) of the enantiomerically- enriched 7- (dimethylamino) tetrahydrobenzothiepine-1, 1-dioxide (4R, 5R) as a colorless solid: *H NMR (CDC13) d 0.87-0.93 (m, 6H) ,

151.10-1.68 (m, 12H) , 2.17-2.25 (m, IH) , 2.81 (s, 6H) , 2.99 (d, J = 15.3 Hz, IH) , 3.15 (d, J = 15.3 Hz, IH) , 3.84 (s, 3H) , 4.11 (d, J = 7.5 Hz, IH) , 5.49 (s, IH) , 5.99 (d, J = 2.4 Hz, IH) , 6.51 (dd, J = 8.7 Hz, 2.4 Hz, IH) , 6.94 (d, J = 8.7 Hz, 2H) , 7.42 (d, J = 8.4 Hz, 2H) , 7.90 (d, J = 8.7 Hz,

20 IH) . The product was determined to have 78% e.e. by chiral HPLC on a Chiralpak AD column using 5% ethanol/hexane as the eluent. Recrystallization of this solid from ethyl acetate/hexane gave 1.70 g of the racemic product. The remaining solution was concentrated and recrystallized to

25 give 9.8 g of colorless solid. Enantiomeric excess of this solid was determined by chiral HPLC on a Chiralpak AD column using 5% ethanol/hexane as the eluent. It showed to have 96% e.e with the first eluting peak as the major product.

30 Step 10: Demethylation of 5- (4 ‘ -methoxyphenyl) -7-

(dimethylamino) tetrahydrobenzothiepine-1.1-dioxide (4R, 5R) To a solution of 47 g (99 mmol) of enantiomeric- enriched (dimethylamino) tetrahydrobenzothiepine-1, 1-dioxide (obtained from Step 9) in 500 mL of methylene chloride at -10 °C was added dropwise a solution of boron tribromide (297 mL, 1M in methylene chloride, 297 mmol), and the resulting solution was stirred cold (-5 °C to 0 °C) for 1 hour or until the reaction was complete. The reaction was cooled in an acetone-dry ice bath at -10 °C, and slowly quenched with 300 mL of water. The mixture was warmed to 10 °C, and further diluted with 300 mL of saturated sodium bicarbonate solution to neutralize the mixture. The aqueous layer was separated and extracted with 300 mL of methylene chloride, and the combined extracts were washed with 200 mL of water, brine, dried over MgS04 and concentrated in vacuo. The residue was dissolved in 500 mL of ethyl acetate and stirred with 50 mL of glacial acetic acid for 30 minutes at ambient temperature. The mixture was washed twice with 200 mL of water, 200 mL of brine, dried over MgS04 and concentrated in vacuo to give the crude 4-hydroxyphenyl intermediate. The solid residue was recrystallized from methylene chloride to give 37.5 g (82%) of the desired (4R, 5R) -5- (4′ – hydoxyphenyl) -7- (dimethylamino) tetrahydrobenzothiepine-1, 1- dioxide as a white solid: *H NMR (CDC13) d 0.84-0.97 (m, 6H) , 1.1-1.5 (m, 10H) , 1.57-1.72 (m, IH) , 2.14-2.28 (m, IH) , 2.83 (s, 6H) , 3.00 (d, J = 15.3 Hz, IH) , 3.16 (d, J – 15.3 Hz, IH) , 4.11 (s, 2H) , 5.48 (s, IH) , 6.02 (d, J – 2.4 Hz, IH) , 6.55 (dd, J = 9, 2.4 Hz, IH) , 6.88 (d, 8 , 7 Hz , 2H) , 7.38 (d, J – 8.7 Hz, 2H) , 7.91 (d, J = 9 Hz, 2H) .

Step 11: Preparation of enantiomerically-enriched chlorobenzyl intermediate Treat a solution of enantiomerically-enriched (4R,5R)- 5- (4′ -hydoxypheny1) -7- (dimethylamino) tetrahydrobenzothiepine-1, 1-dioxide (5.0 g, 10.9 mmol, obtained from Step 10) in acetone (100 mL) at 25 °C under N2 with powdered 5 K2C03 (2.3 g, 16.3 mmol, 1.5 eq.) and a, a’ -dichloro-p-xylene (6.7 g, 38.1 mmol, 3.5 eq.) . Stir the resulting solution at 65 °C for about 48 hours. Cool the reaction mixture to 25 °C and concentrate to 1/5 of original volume. Dissolve the residue in EtOAc (150 mL) and wash with water (2 x 150 mL) .

10 Extract the aqueous layer with EtOAc (2 x 150 mL) and wash the combined organic extracts with saturated aqueous NaCI (2 x 150 mL. Dry the combined extracts with MgS04 and concentrate in vacuo to provide the crude product . Purification by flash chromatography (5.4 x 45 cm silica,

1525-40% EtOAc/hexane) will afford the enantiomerically- enriched chlorobenzyl intermediate .

Step 12: Preparation of enantiomerically-enriched (4R.5R)- 1- r [4- [ [4- [3 , 3-Dibutyl-7- (dimethylamino) -2,3 , 4 , 5-tetrahvdro-

204 -hydroxy-1.1-dioxido-1-benzothiepin-5- yl] phenoxy] methyll phenyl! methyl] -4-aza-l- azoniabicyclo f2.2.2] octane chloride (XXVII)

Treat a solution of the enantiomerically-enriched chlorobenzyl intermediate (4.6 g, 7.7 mmol, obtained from

25 above in Step 11) in acetonitrile (100 mL) at 25 °C under N2 with diazabicyclo [2.2.2] -octane (DABCO, 0.95 g, 8.5 mmol, 1.1 eq.) and stir at 35 °C for 2 hours. Collect the precipitated solid and wash with CH3CN. Recrystallization from CH3OH/Et20 will give the desired title compound (XXVII) .

ANY ERROR,  EMAIL amcrasto@gmail.com, +919323115463

 

///////////FDA, Breakthrough Designation,  Shire, Rare GI Therapies, SHP625, maralixibat, progressive familial intrahepatic , Maralixibat chloride, 228113-66-4, UNII: V78M04F0XC, LUM 001, Lopixibat chloride, cholestasis type 2 (PFIC2), Maralixibat Chloride,  ماراليكسيبات كلوريد ,  氯马昔巴特 , Мараликсибата хлорид

CCCCC1(CS(=O)(=O)c2ccc(cc2[C@H]([C@H]1O)c3ccc(cc3)OCc4ccc(cc4)C[N+]56CCN(CC5)CC6)N(C)C)CCCC.[Cl-]

Share
Follow

Get every new post on this blog delivered to your Inbox.

Join other followers: