AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO, Worlddrugtracker, Born in Mumbai in 1964 and graduated from Mumbai University, Completed his PhD from ICT ,1991, Mumbai, India, in Organic chemistry, The thesis topic was Synthesis of Novel Pyrethroid Analogues, Currently he is working with AFRICURE PHARMA as ADVISOR earlier GLENMARK LS Research centre as consultant,Principal Scientist, Process Research (bulk actives) at Mahape, Navi Mumbai, India. Prior to joining Glenmark, he worked with major multinationals like Hoechst Marion Roussel, now sSanofi, Searle India ltd, now Rpg lifesciences, etc. he is now helping millions, has million hits on google on all organic chemistry websites. His New Drug Approvals, Green Chemistry International, Eurekamoments in organic chemistry are some most read blogs He has hands on experience in initiation and developing novel routes for drug molecules and implementation them on commercial scale over a 32 year tenure, good knowledge of IPM, GMP, Regulatory aspects, he has several international drug patents published worldwide . He gas good proficiency in Technology transfer, Spectroscopy, Stereochemistry, Synthesis, polymorphism etc He suffered a paralytic stroke in dec 2007 and is bound to a wheelchair, this seems to have injected feul in him to help chemists around the world, he is more active than before and is pushing boundaries, he has one lakh connections on all networking sites, He makes himself available to all, contact him on +91 9323115463, amcrasto@gmail.com

Tariquidar

 phase 2, Uncategorized  Comments Off on Tariquidar
Dec 242013
 

 

 

Tariquidar

206873-63-4 CAS NO

XR 9576;XR9576;D06008.

Molecular Weight (MW) 646.73

Formula

C38H38N4O6

NMR

http://file.selleckchem.com/downloads/nmr/S802801-Tariquidar-HNMR-Selleck.pdf

http://www.medkoo.com/Product-Data/Tariquidar/Tariquidar-QC-BBC20130420-web.pdf

N-[2-[[4-[2-(6,7-Dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)ethyl]phenyl]carbamoyl]-4,5-dimethoxyphenyl]quinoline-3-carboxamide

 

Xenova (Originator), QLT PhotoTherapeutics (Licensee)
Modulators of the Therapeutic Activity of Antineoplastic Agents, Multidrug Resistance Modulators, ONCOLYTIC DRUGS, P-Glycoprotein (MDR-1) Inhibitors

Tariquidar (XR9576) is a potent and selective noncompetitive inhibitor of P-glycoprotein with Kd of 5.1 nM, reverses drug resistance in MDR cell Lines. Phase 3.

Tariquidar (INN/USAN) is a P-glycoprotein inhibitor[1] undergoing research as an adjuvant against multidrug resistance in cancer.

Tariquidar is a P-glycoprotein inhibitor undergoing research as an adjuvant against multidrug resistance in cancer. Tariquidar non-competitively binds to the p-glycoprotein transporter, thereby inhibiting transmembrane transport of anticancer drugs. Inhibition of transmembrane transport may result in increased intracellular concentrations of an anticancer drug, thereby augmenting its cytotoxicity

 

The resistance of tumours to treatment with certain cytotoxic agents is an obstacle to the successful chemotherapeutic treatment of cancer patients. A tumour may acquire resistance to a cytotoxic agent used in a previous treatment. A tumour may also manifest intrinsic resistance, or cross-resistance, to a cytotoxic agent to which it has not previously been exposed, that agent being unrelated by structure or mechanism of action to any agent used in previous treatments of the tumour.

Analogously, certain pathogens may acquire resistance to pharmaceutical agents used in previous treatments of the diseases or disorders to which those pathogens give rise. Pathogens may also manifest intrinsic resistance, or cross resistance, to pharmaceutical agents to which they have not previously been exposed. Examples of this effect include multi-drug resistant forms of malaria, tuberculosis, leishmaniasis and amoebic dysentery. These phenomena are referred to collectively as multi-drug resistance (MDR).

The most common form of MDR is caused by over-production in the cell membrane of P-gp, a protein which is able to reduce the accumulation of drugs in cells by pumping them out. This protein has been shown to be a major cause of multidrug resistance in tumour cells (Beck, W. T. Biochem. Pharmacol, 1987, 36,2879-2887).

In addition to cancer cells, p-glycoprotein has been found in many normal human tissues including the liver, small intestine, kidney, and blood-brain endothelium. P-gps are localised to the secretory domains of the cells in all these tissues. This localisation suggests that P-gp may play a role in limiting the absorption of foreign toxic substances across biological barriers.

Consequently, in addition to their ability to increase the sensitivity of cancer cells to cytotoxic agents, P-gp inhibitors are expected to increase the net oral absorption of certain drugs and improve the transport of drugs through the blood-brain barrier. Indeed, administration of cyclosporin, a P-gp inhibitor, has been shown to increase the intestinal absorption of acebutolol and vinblastine in rats by 2.6 and 2.2-fold respectively (Tereo, T. et al. J. Pharm. Pharmacol, 1996, 48, 1083-1089), while mice deficient in mdr la P-gp gene exhibit up to 100-fold increased senstivity to the centrally neurotoxic pesticide ivermectin (Schinkel, A. H. et al Cell 1994, 77, 491-502). Besides increased drug levels in the brain, the P-gp deficient mice were shown to have elevated drug levels in many tissues and decreased drug elimination.

Disadvantages of drugs which have so far been used to modulate MDR, termed resistance modifying agents or RMAs, are that they frequently possess a poor pharmacokinetic profile and/or are toxic at the concentrations required for MDR modulation.

It has now been found that a series of anthranilic acid derivatives have activity as inhibitors of P-gp and may therefore be used in overcoming the multi-drug resistance of tumours and pathogens. They also have potential utility in improving the absorption, distribution, metabolism and elimination characteristics of certain drugs.

  1.  Robey RW, Shukla S, Finley EM, Oldham RK, Barnett D, Ambudkar SV, Fojo T, Bates SE. Inhibition of P-glycoprotein (ABCB1)- and multidrug resistance-associated protein 1 (ABCC1)-mediated transport by the orally administered inhibitor, CBT-1((R)). Biochem Pharmacol 2008;3:1302-12. PMID 18234154.
  2. Contino M, Zinzi L, Cantore M, Perrone MG, Leopoldo M, Berardi F, Perrone R, Colabufo NA. Activity-lipophilicity relationship studies on P-gp ligands designed as simplified tariquidar bulky fragments. Bioorg Med Chem Lett. 2013 Jul 1;23(13):3728-31. doi: 10.1016/j.bmcl.2013.05.019. Epub 2013 May 16. PubMed PMID: 23726026.
  3. Matthew D. Hall, Kyle R. Brimacombe, Matthew S. Varonka, Kristen M. Pluchino, Julie K. Monda, Jiayang Li, Martin J. Walsh, Matthew B. Boxer, Timothy H. Warren§, Henry M. Fales, and Michael M. Gottesman.Synthesis and Structure–Activity Evaluation of Isatin-β-thiosemicarbazones with Improved Selective Activity toward Multidrug-Resistant Cells Expressing P-Glycoprotein, J. Med. Chem., 2011, 54 (16), pp 5878–5889.

EP 0934276; GB 2334521; JP 2001502683; US 6218393; WO 9817648

Bioorg Med Chem Lett1999,9,(4):595

 

 

 

4,5-Dimethoxy-2-nitrobenzoic acid (I) was converted to the corresponding acid chloride (II) upon treatment with SOCl2, and this was further coupled to aniline (III), producing amide (IV). Catalytic hydrogenation of the nitro group of (IV) afforded amine (V). Acid chloride (VII) –obtained by chlorination of 3-quinolinecarboxylic acid (VI) with SOCl2– was then condensed with amine (V) to furnish the title diamide.

 

………………………………..

https://www.google.co.in/patents/US6218393?pg=PA29&dq=US+6218393&hl=en&sa=X&ei=YAm5Up-IMs7_rAeg1oHoAg&ved=0CDcQ6AEwAA

 

 

Figure US06218393-20010417-C00318

 

Share
Dec 232013
 

Rigosertib

 

(E)-2-(2-Methoxy-5-(2-(2′,4′,6′-trimethoxyphenyl)vinylsulfonamido)phenylamino)acetic Acid

THERAPEUTIC CLAIM Antineoplastic
CHEMICAL NAMES
1. Glycine, N-[2-methoxy-5-[[[(1E)-2-(2,4,6-Trimethoxyphenyl)ethenyl]sulfonyl]
methyl]phenyl]-
2. N-[2-methoxy-5-({[(1E)-2-(2,4,6-trimethoxyphenyl)ethenyl]sulfonyl}methyl)
phenyl]glycine

MOLECULAR FORMULA C21H25NO8S
MOLECULAR WEIGHT 451.5

SPONSOR Onconova Therapeutics
CODE DESIGNATION –ON 01910
CAS REGISTRY NUMBER 592542-59-1

 

 

Chemical Formula: C21H24NNaO8S

Molecular Weight: 473.47

1225497-78-8

sodium (E)-2-((2-methoxy-5-(((2,4,6-trimethoxystyryl)sulfonyl)methyl)phenyl)amino)acetate

 

US Patent No.7,598,232, such as in Schemes 1-10

M.V. Reddy et al. J. Med. Chem. 2011, 54, 6254

Rigosertib (ON-01910 sodium salt)  is a synthetic benzyl styryl sulfone analogue with potential antineoplastic activity. Polo-like kinase 1 inhibitor ON 01910.Na inhibits polo-like kinase1 (Plk1), inducing selective G2/M arrest followed by apoptosis in a variety of tumor cells while causing reversible cell arrest at the G1 and G2 stage without apoptosis in normal cells. This agent may exhibit synergistic antitumor activity in combination with other chemotherapeutic agents. Plk1, named after the polo gene of Drosophila melanogaster, is a serine/threonine protein kinase involved in regulating mitotic spindle function in a non-ATP competitive manner.

Rigosertib is an inhibitor of two important cellular signaling pathways, PI3K and PLK, both of which are frequently over-active in cancer cells. PI3K signaling promotes the growth and survival of cells under stressful conditions, such as under low oxygen levels that are often found in tumors. If the PI3K pathway is over-active, apoptosis of cancer cells is diminished, leading to excessive cellular growth. By inhibiting the PI3K pathway, rigosertib promotes tumor cell apoptosis. Rigosertib also influences signals along the PI3K pathway, such as those leading to the production of cyclin D1.

The PLK pathway plays a critical role in maintaining proper organization and sorting of chromosomes during cell division. Too much PLK activity in cancer cells results in uncontrolled proliferation. By modulating PLK pathway activity in cancer cells, rigosertib inhibits cellular division, leading to chromosome disorganization and death in these cells.

Due to this dual effect on tumor cell survival and division pathways, we believe that rigosertib has potential to treat a variety of cancer types, including hematological diseases and solid tumors.  Ongoing clinical trials are evaluating the activity of rigosertib in:

  • Myelodysplastic Syndromes (MDS)
  • Pancreatic Cancer
  • Head & Neck Cancer
  • Other hematological diseases and solid tumors

Ongoing and completed Phase 1, Phase 2 and Phase 3 clinical trials have generated data in over 850 patients with advanced, heavily pre-treated solid tumors and hematological diseases and have demonstrated a desirable safety profile for rigosertib.

Rigosertib is a substituted styryl benzylsulfone that inhibits multiple kinases including phosphatidylinositol 3-kinase (PI3-K) and polo-like kinase 1 (PLK-1). Phase 1 and 2 studies have demonstrated its ability to delay transition of myelodysplasia syndrome (MDS) to acute myologenous leukemia (AML), which is a serious disease associated with high mortality. As a result, it is being studied in a Phase 3 trial in MDS patients who have failed previous chemotherapy with accepted agents

Polo-like kinases are enzymes that are involved in cell division and checkpoint regulation of mitosis; they also help maintain DNA integrity. They are overexpressed in a variety of human tumours but not in normal cells, making them a potential target for cancer chemotherapy. Rigosertib, a small molecule agent designed to target these kinases, is being developed by US biotech company Onconova.It remains active against numerous cancer cells that are resistant to other drugs, without affecting normal cells. Trials are furthest advanced in myelo-dysplastic syndrome (MDS). In a Phase I/II trial, patients with the MDS or acute myeloid leukaemia were given the drug by continuous intravenous infusion over a period of 72 to 144 hours every two weeks, for between five and 70 weeks.Three achieved a marrow complete response and two a haematological improvement. The five non-responders were the five patients with AML. It was well tolerated.

 

 

 

US7598232

Figure US07598232-20091006-C00077

 

………..

NMR

 

J. Med. Chem., 2013, 56 (13), pp 5562–5586

DOI: 10.1021/jm400575x

(E)-2-(2-Methoxy-5-(2-(2′,4′,6′-trimethoxyphenyl)vinylsulfonamido)phenylamino)acetic Acid (25a)

 pale-yellow solid, mp 110–112 °C. 1H NMR(DMSO-d6, 300 MHz): δ 3.71 (s, 2H, CH2), 3.82 (s, 3H, OCH3), 3.84 (s, 3H, OCH3), 3.87 (s, 6H, 2 × OCH3), 6.26 (s, 2H, Ar–H), 6.48 (d, J = 1.8 Hz, 1H, Ar–H), 6.65 (dd, J = 1.8, 8.1 Hz, 1H, Ar–H), 6.86 (d, J = 8.1 Hz, 1H, Ar–H), 6.94 (d, J = 15.6 Hz, 1H, ═CH), 7.60 (d, J = 15.6 Hz, 1H, CH═), 8.99 (br s, 1H, NH). HRMS found [M – H] (m/z): 451.1209. Calcd for C20H24N2O8m/z: 452.1253.

 

About Onconova Therapeutics, Inc.

Onconova Therapeutics is a clinical-stage biopharmaceutical company focused on discovering and developing novel products to treat cancer. Onconova’s clinical and pre-clinical stage drug development candidates are derived from its extensive chemical library and are designed to work against specific cellular pathways that are important in cancer cells, while causing minimal damage to normal cells. In addition to rigosertib, the Company’s most advanced product candidate, two other candidates are in clinical trials, and several candidates are in pre-clinical stages.  For more information, please visit http://www.onconova.com.

 

NEWTOWN, Pa., Nov. 7, 2013 (GLOBE NEWSWIRE) — Onconova Therapeutics, Inc. , a clinical-stage biopharmaceutical company focused on discovering and developing novel products to treat cancer, today announced two presentations relating to clinical trials of its most advance product candidate, rigosertib, at the 55th American Society of Hematology (ASH) Annual Meeting in New Orleans, Louisiana, December 7-10, 2013. The presentations will include data on efficacy, tolerability, and dosing regimen from the Phase 2 study (ONTARGET) of oral rigosertib in transfusion-dependent, lower risk MDS patients and response, overall survival, and longer-term follow-up data from a Phase 1/2 trial of IV rigosertib in higher risk post-hypomethylating agent treated MDS and AML patients.

Share
Dec 222013
 

ICOTINIB

 

N-(3-Ethynylphenyl)-7,8,10,11,13,14-hexahydro[1,4,7,10]tetraoxacyclododecino[2,3-g]quinazolin-4-amine

[1,4,7,10]Tetraoxacyclododecino[2,3-g]quinazolin-4-amine, N-(3-ethynylphenyl)-7,8,10,11,13,14-hexahydro-

 

BPI 2009H

610798-31-7  CAS BASE

 

Compound Structure

Icotinib Hydrochloride, 1204313-51-8, CS-0918, HY-15164, Conmana Zhejiang Beta Pharma Ltd.

 

Icotinib is a potent and specific EGFR inhibitor with IC50 of 5 nM, including the EGFR, EGFR(L858R), EGFR(L861Q), EGFR(T790M) and EGFR(T790M, L858R). Phase 4.Icotinib hydrochloride is the epidermal growth factor receptor kinase targeting a new generation of targeted anti-cancer drugs, completely independent from the original tumor clinical practitioners and experts of science, through eight years of the development, its first adaptation disease is advanced non-small cell lung cancer. Icotinib is an orally available quinazoline-based inhibitor of epidermal growth factor receptor (EGFR), with potential antineoplastic activity. Icotinib selectively inhibits the wild-type and several mutated forms of EGFR tyrosine kinase. This may lead to an inhibition of EGFR-mediated signal transduction and may inhibit cancer cell proliferation. EGFR, a receptor tyrosine kinase, is upregulated in a variety of cancer cell types. Icotinib was approved in China in 2011

Icotinib has been found to be noninferior to gefitinib in patients with non-small-cell lung cancer (NSCLC), according to reports from the phase III Chinese double-blind ICOGEN study.

“[I]cotinib is a valid therapeutic option for patients with non-small-cell lung cancer as a second-line or third-line treatment, although patients might find taking icotinib three times a day an inconvenience,” write Yan Sun (Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China) and colleagues.

Icotinib is an oral epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) that has exhibited good antitumor activity in phase II studies. However, it has a shorter half-life than gefitinib, another TKI, which means that it needs to be taken more often.


Design and discovery of 4-anilinoquinazoline ureas as multikinase inhibitors targeting BRAF, VEGFR-2 and EGFR. Qingwen Zhang, Yuanyuan Diao, Fei Wang, Ying Fu, Fei Tang, Qidong You, Houyuan Zhou, Med. Chem. Commun., 2013, 4, 979

  • Tyrosine kinase receptors are trans-membrane proteins that, in response to an extracellular stimulus, propagate a signaling cascade to control cell proliferation, angiogenesis, apoptosis and other important features of cell growth. One class of such receptors, epidermal growth factor receptor (EGFR) tyrosine kinases, are over-expressed in many human cancers, including brain, lung, liver, bladder, breast, head and neck, esophagus, gastrointestinal, breast, ovary, cervix or thyroid cancer.
  • EGFR is expressed in many types of tumor cells. Binding of cognate ligands (including EGF, TGFα (i.e., Transforming Growth Factor-α) and neuregulins) to the extracellular domain causes homo- or heterodimerization between family members; the juxtaposition of cytoplasmic tyrosine kinase domains results in transphosphorylation of specific tyrosine, serine and threonine residues within each cytoplasmic domain. The formed phosphotyrosines act as docking sites for various adaptor molecules and subsequent activation of signal transduction cascades (Ras/mitogen-activated, PI3K/Akt and Jak/STAT) that trigger proliferative cellular responses.
  • Various molecular and cellular biology and clinical studies have demonstrated that EGFR tyrosine kinase inhibitors can block cancer cell proliferation, metastasis and other EGFR-related signal transduction responses to achieve clinical anti-tumor therapeutic effects. Two oral EGFR kinase inhibitors with similar chemical structures are Gefitinib (Iressa; AstraZeneca), approved by the U.S. FDA for advanced non-small cell lung cancer in 2003 (and later withdrawn), and Erlotinib Hydrochloride (Tarceva; Roche and OSI), approved by the U.S. FDA for advanced non-small cell lung cancer and pancreatic cancer treatment in 2004.
  • Chinese Patent Publication No. CN1305860C discloses the structure of 4-[(3-ethynyl-phenyl)amino]-6,7-benzo-12-crown-quinoline (free base) on page 29, Example 15, Compound 23.

Icotinib was launched in China in August 2011, after approval by the State Food and Drug Administration. It is a targeted EGFR tyrosine kinase inhibitor that, like erlotinib (Tarceva) and gefitinib (Iressa), shows benefit in patients with EGFR m+ NSCLC.

…………………………………….. http://www.google.com/patents/EP2392576A1

    •  Formula I (Icotinib hydrochloride):

Figure imgb0011

Method 1:

    • Figure imgb0002

Method 2:

    • Figure imgb0003

Method 3:

  • Figure imgb0004
  • BPI-02 is obtained by recrystallization.

http://www.google.com/patents/EP2392576A1 Example 1Step 1

    • Figure imgb0005
    • Preparation: 16 kg (400 mol) of sodium hydroxide was dissolved in 80 L of water in a 400 L reactor, and then 18.8 L (140 mol) of triethylene glycol, 32 L of THF were added into the reactor. After cooling below 5 °C, a solution of 47.84 kg (260 mol) of tosyl chloride and 50 L of THF was added dropwise. Following the addition, the reaction mixture was kept at this temperature for 2 hours, and it was then poured into 240 L of ice water. The precipitate was formed and filtered, washed with a small amount of water, and dried. 58.64 kg of BPI-01 as a white crystalline powder was yielded at 91.4%. mp: 77-80 °C, HPLC: 97%. TLC (petroleum ether: ethyl acetate = 1:1) Rf = 0.87.
    • NMR data: 1H-NMR (CDCl3): δ ppm: 7.78 (d, 4H, J = 10.4 Hz, benzene protons by sulfonyl group); 7.34 (d, 4H, J = 11.6 Hz, benzene protons by methyl group); 4.129 (dd, 4H, J = 5.6 Hz, ethylene protons by the sulfonyl group); 3.64 (dd, 4H, J = 5.6 Hz, ethylene protons away from the sulfonyl group); 3.517 (s, 4H, ethylene protons in the middle); 2.438 (s, 6H, methyl protons on the benzene).

Step 2

    • Figure imgb0006
    • Preparation: A solution containing 3.64 kg (20 mol) of ethyl 3,4-dihydroxybenzoate and 12.4 kg (89.6 mol) of potassium carbonate in 300 L of N,N-dimethylformamide was stirred and heated to 85-90 °C for about 30 minutes. A solution of 9.17 kg (20 mol) of BPI-01 in 40 L of N,N-dimethylformamide was added dropwise over 1.5-2 hours. After the addition, the reaction was kept for 30 minutes; the reaction completion was confirmed by TLC (developing solvent: petroleum ether:ethyl acetate = 1:1, Rf = 0.58). The reaction mixture was removed from the reactor and filtered. Then, the filtrate was evaporated to remove N,N-dimethylformamide; 240 L of ethyl acetate was added to dissolve the residue. After filtration and vacuum evaporation, the residual solution was extracted with 300 L of petroleum ether. After evaporation of the petroleum ether, the residual solids were re-crystallized with isopropanol in a ratio of 1:2.5 (W/V); 1.68 kg of BPI-02 as a white powder was obtained in a yield of 28%. mp: 73-76 °C, HPLC: 96.4%. NMR data: 1H-NMR (CDCl3): δ ppm: 7.701 (d, 1H, J = 2.4 Hz, benzene proton at position 6); 7.68 (s, 1 H, benzene proton at position 2); 6.966 (d, 1H, J = 10.8 Hz, benzene proton at position 5); 4.374-3.81 (q, 2H, J = 9.6 Hz, methylene protons of the ethyl); 3.78-4.23 (dd, 12H, J = 4.8 Hz, crown ether protons); 1.394 (t, 3H, J = 9.6 Hz, methyl protons of the ethyl). MS: m/z 296.

Step 3

    • Figure imgb0007
    • Preparation: A solution of 592 g (2 mol) of BPI-02 and 600 mL of acetic acid in a 5 L reaction flask was cooled to 0°C; 1640 mL (25.4 mol) of concentrated nitric acid was slowly added. The internal temperature should not exceed 10 °C. While cooled below 0°C, 1 L of concentrated sulfuric acid was added dropwise. The internal temperature should not be higher than 5°C. After the addition, the reaction was kept at 0-5 °C for 1-2 hours. After completion of the reaction, the reaction solution was poured into 15 L of ice water in a plastic bucket. After mixing, filtration, and re-crystallization in ethanol, 449 g of BPI-03 as a light yellow to yellow crystalline powder was obtained in 65.7% yield. mp: 92-95 °C, HPLC: 98.2%. TLC (petroleum ether: ethyl acetate =1:1) Rf = 0.52. NMR data: 1H-NMR (CDCl3): δ ppm: 7.56 (s, 1H, benzene proton at position 5); 7.20 (s, 1H, benzene proton at position 2); 4.402 (q, 2H, J = 9.2 Hz, methylene protons of the ethyl); 4.294 (dd, 12H, J = 4.8 Hz, crown ether protons); 1.368 (t, 3H, J = 9.2 Hz, methyl protons of the ethyl).

Step 4

    • Figure imgb0008
    • Preparation: In a 3 L hydrogenation reactor, 2 L of methanol and 195 g (0.57 mol) of BPI-03 were added, and then 63 mL of acetyl chloride was slowly added. After a short stir, 33 g of Pd/C containing 40% water was added. The reaction was conducted under 4 ATM hydrogen until hydrogen absorption stopped, and then the reaction was kept for 1-2 hours. After completion of the reaction, the reaction mixture was transferred into a 5 L reactor. After filtration, crystallization, and filtration, the product was obtained. The mother liquor was concentrated under vacuum, and more product was obtained. The combined crops were 168 g of BPI-04 as a white to pink crystalline powder in a yield of 85%. mp: 198-201 °C, HPLC: 99.1 %. TLC (petroleum ether: ethyl acetate = 1:1) Rf = 0.33. NMR data: 1H-NMR (DMSO-d6): δ ppm: 8-9 (br., 3H, 2 protons of the amino group and a proton of the hydrochloric acid); 7.37 (s, 1H, benzene proton at position 5); 6.55 (s, 1H , benzene proton at position 2); 4.25 (q, 2H, J = 7.06 Hz, methylene protons of the ethyl); 4.05 (dd, 12H, J = 4.04 Hz, crown ether protons); 1.31 (t, 3H, J = 7.06 Hz, methyl protons of the ethyl).

Step 5

    • Figure imgb0009
    • Preparation: 1105 g (3.175 mol)of BPI-04, 4810 g (106.9 mol) of formamide, and 540 g (8.55 mol) of ammonium formate were added to a 10 L 3-neck bottle. The reaction mixture was heated to 165 °C under reflux for 4 hours. After cooling to room temperature, 3 L of water was added, and then the mixture was stirred for 10 minutes. After filtration, washing, and drying, 742 g of BPI-05 as a white crystalline powder was obtained in a yield of 80%. mp: 248-251 °C, HPLC: 99.78%. TLC (chloroform: methanol = 8:1) Rf = 0.55. NMR data: 1H-NMR (DMSO-d6): δ ppm: 12.06 (s, 1H, NH of the quinazoline); 8.0 (d, 1H, J = 3.28 Hz, proton of the quinazoline position 3); 7.62 (s, 1H, proton of the quinazoline position 6); 7.22 (s, 1H, proton of the quinazoline position 9); 4.25 (dd, 12H, J = 4.08 Hz, crown ether protons).

Step 6

    • Figure imgb0010
    • Preparation: 337 g (1.13 mol) of BPI-05, 7.1 L of chloroform, 1.83 L (19.58mol) of POCI3 and 132 ml of N,N-dimethylformamide were added to a 10 L 3-neck bottle. The reaction mixture was stirred at reflux temperature. After dissolution, reaction completion was checked by TLC (developing solvent: chloroform: methanol = 15:1, Rf = 0.56); the reaction took approximately 8 hours to complete. Then, the reaction solution was cooled and evaporated under vacuum to dryness. The residue was dissolved in 4 L of chloroform; 4 kg of crushed ice was poured into the solution and the mixture was stirred for 0.5 hours. After separation, the aqueous phase was extracted twice with 2 L of chloroform. The organic phases were combined, 4 L of ice water was added and the pH was adjusted with 6 N NaOH to pH 8-9 while the temperature was maintained below 30 °C. After separation, the organic phase was washed with saturated NaCl, dried over anhydrous sodium sulfate and the solvents removed by vacuum evaporation. The residual solids were washed with acetone and filtered; 268 g of BPI-06 as a white crystalline powder was obtained in a yield of 77% with mp: 164-167°C and HPLC purity of 99%. NMR data: 1H-NMR (CDCl3): δ ppm: 8.89 (s, 1H, proton of the quinazoline position 2); 7.68 (s, 1H, proton of the quinazoline position 9); 7.42 (s, 1H, proton of the quinazoline position 6); 4.38-3.81 (dd, 12H, J = 3.88 Hz, crown ether protons).

Step 7

  • Figure imgb0011
  • Preparation of the compound of the present invention: To a suspension of 20.8 g of BPI-06 in 500 mL of ethanol was added 25 mL of N,N-dimethylformamide and a solution of 8.98 g m-acetylene aniline in 200 mL of isopropanol. The reaction mixture was stirred at room temperature for 5 minutes until dissolved completely, and then the reaction solution was heated at reflux for 3 hours. After concentration and drying, the residual solids were dissolved in ethyl acetate, washed with water, and dried over anhydrous sodium sulfate. Thus, 27.1 g of the compound of Formula I was obtained as a white crystalline powder. NMR data: 1H-NMR (Bruker APX-400, solvent: DMSO-d6, TMS as internal standard): δ ppm: 3.58 (dd, 2H, two protons of the crown position 12); 3.60 (dd, 2H, two protons of the crown position 13); 3.73 (dd, 2H, two protons of the crown position 10); 3.80 (dd, 2H, two protons of the crown position 15); 4.30 (s, 1H, proton of the alkynyl); 4.34 (dd, 2H, two protons of the crown position 16); 4.40 (dd, 2H, two protons of the crown position 9); 7.39 (d, 1H, benzene proton at position 25); 7.46 (dd, 1H, benzene proton at position 26); 7.49 (s, 1H, proton of the quinazoline position 6); 7.82 (d, 1H, benzene proton at position 27); 7.94 (t due dd, 1H, proton of the quinazoline position 19); 8.85 (s, 1H, benzene proton at the position 23); 8.87 (s, 1H, proton of the quinazoline position 2); 11.70 (s, 1H, proton of the aromatic amine as salt); 14-16 (bs, 1H, hydrochloride), see Figure 5. NMR data: 13C-NMR (DMSO-d6), see Figure 6. Mass spectrometry (MS): Instrument: ZAB-HS, testing conditions: EI, 200°C, 700ev, MS measured molecular weight: m/z 427.

…………………………………..

https://www.google.co.in/patents/WO2013064128A1?cl=en&dq=icotinib&hl=en&sa=X&ei=1oi2UsP9LYa4rgfUzoF4&ved=0CDcQ6AEwAA

 

Figure imgf000003_0002

Synthesis of compound 1 A

1 Synthesis of Compound 2

Figure imgf000008_0003

2

79.5g 3,4 – dihydroxybenzene nitrile, 272g of potassium carbonate, acetonitrile (6L) was added to a 10L three-necked reaction flask, and dissolved with stirring, heated to reflux and reflux was added dropwise an acetonitrile solution of the compound 1 (compound 1, 200 g; acetonitrile , 2L), and completion of the dropping, the HPLC monitoring of the completion of the reaction, the mixture was cooled to room temperature, filtered, and the solvent was removed, and the resulting solid was washed with ethyl acetate was dissolved, filtered, and the filtrate was concentrated, the resulting residue was dissolved in petroleum ether by rotary evaporation, the resulting solid was purified to give 18.9g of the compound 2.

1 LAI MR (CDC1 3-Sppm): 7.30 ~ 7.33 (m, 1H); 7.25 (s, 1H); 6.97-6.99 (d, 1H); 4.19 – 4.23 (m, 4H); 3.83 ~ 3.91 (m, 4H); 3.77 (s, 4H). MS: (M + H) +250 2 Synthesis of compound A

Figure imgf000009_0001

2 A

41.6g of compound 2 was dissolved in 580ml of acetic acid, dropwise addition of 83ml of fuming nitric acid at 30 ° C under completion of the dropping, the dropwise addition of 42ml of concentrated sulfuric acid at 30 ° C under the reaction at room temperature overnight, TLC monitoring completion of the reaction, the reaction solution was poured into ice water 4L , the precipitated solid was filtered, washed with cold water (500 mL X 2), vacuum 35 ° C and dried crude A compound 46g, isopropanol recrystallization was purified to give 33g of compound A.

1 LAI MR (CDC1 3-Sppm): 7.90 (s, 1H); 7.36 (s, 1H); 4.33 ~ 4.36 (m, 4H); 3.87 ~ 3.89 (m, 4H); 3.737 (s, 4H). Embodiment of Example 2 Synthesis of Compound B

Figure imgf000009_0002

AB

32g of compound A, 30.5g of iron powder, 5% acetic acid solution in methanol 1070ml 2L reaction flask was heated to reflux

TLC monitoring of the end of the reaction cooled and concentrated, dissolved in ethyl acetate, filtered, dried over anhydrous NaS0 4 23g of compound B. The solvent was removed.

1HNMR (d 6-DMSO-Sppm): 7.07 (s, 1H); 6.36 (s, 1H); 5.73 (s, 2H); 3.95 ~ 4.22 (m, 4H); 3.77-3.78 (m, 2H); 3.34 3.62 (m, 6H).Embodiment of Example 3 Synthesis of Compound CI

Figure imgf000009_0003

B CI

500mL three-necked flask, the Add 5g compound B, 5g v, v-dimethyl formamide dimethyl acetal and 160ml of dioxane was heated to reflux the TLC monitoring progress of the reaction, the reaction time is about 12 hours, after the end of the reaction The reaction solution was cooled to room temperature, spin-dry to give 5.8g of compound Cl.

1 LAI MR (CDCl 3-Sppm): 7.56 (s, 1H); 7.15 (s, 1H); 6.51 (s, 1H); 4.12-4.18 (m, 4H); 3.89-3.91 (m, 2H); 3.78 -3.80 (m, 6H); 3.07 (s, 6H); Example 4 Icotinib Synthesis

 

Figure imgf000010_0001

5 g of the compound Cl, 2.2 g inter-aminophenyl acetylene, 230ml of acetic acid was added to a 500 ml reaction flask was heated to 100 ° c,

TLC monitoring of the reaction. The end of the reaction, the reaction system spin dry methanol was added, and shock dispersion, filtration, wash with methanol, 5g Icotinib.

^ M (d 6-DMSO-5ppm): 11.98 (s, IH); 9.50 (s, IH); 8.53 (s 1H); 8.14 (s, IH); 8.04-8.05 (m, IH); 7.90-7.92 (m, IH); 7.38-7.42 (m, IH); 7.31 (s IH); 7.20-7.22 (m, IH); 4.29-4.30 (m, 4H); 4.21 (s, IH); 3.74-3.81 ( m, 4H); 3.64 (s, 4H); 1.91 (s, 3H); Synthesis Example 5 Exe hydrochloride erlotinib

Figure imgf000010_0002

Exeter for Nick for; s

700mg Icotinib Add to a 100 ml reaction flask, add 40 ml of methanol, stirred pass into the hydrogen chloride gas or concentrated hydrochloric acid, and filtered to give crude hydrochloric acid Icotinib after, and purified by recrystallization from isopropanol to give 760mg hydrochloride Icotinib.

1HNMR (d 6-DMSO-Sppm): 11.37 (s, IH); 8.87 (s, IH); 8.63 (s, IH); 7.90 (s, IH); 7.78-7.80 (d, IH); 7.48-7.52 (m, IH); 7.40-7.41 (m, 2H); 4.36-4.38 (d, 4H); 4.30 (s, IH); 3.75-3.81 (d, 4H); 3.61 (s, 4H); Example 6 Synthesis of Compound B

Figure imgf000011_0001

AB

25g of compound A, 25 g of iron powder, 3% acetic acid in methanol solution 900ml with Example 2 are the same, to give 16.6g of compound B.

Embodiment of Example 7 Synthesis of Compound B

Figure imgf000011_0002

AB

40 g of compound A, 40 g of iron powder and 7% acetic acid in methanol solution was 1200ml, in Example 2, to give 28.4g of compound B.

Example 8 Compound B Synthesis

Figure imgf000011_0003

AB

25 g of compound A, 5 g of Pd / C in 3% acetic acid in methanol solution 900ml Add 2L reaction flask, of the hydrogen, TLC monitoring of the end of the reaction, filtered, and the solvent was removed to give 17g of compound B.

Example 9 Compound B Synthesis

Figure imgf000011_0004

AB

40g of compound A, 17 g of magnesium and 5% acetic acid in methanol solution 1200ml, in Example 2, to give 25.2g of compound B. Example 10 Compound B Synthesis

 

Figure imgf000012_0001

AB

25 g of compound A, 32.5g of zinc powder and 5% acetic acid in methanol solution 900ml with Example 2 are the same, to give 17.1g of compound B.

Example Synthesis of compound 11 B

 

Figure imgf000012_0002

AB

25g of compound A, 28 g of iron powder, 5% trifluoroacetic acid in methanol solution 700ml, in Example 2, 16g of compound B.

Embodiment Example 12 Synthesis of Compound C1

 

Figure imgf000012_0003

3g compound B, 3G v, v-dimethyl formamide dimethyl acetal and 140ml of dioxane, reflux the reaction time is 10-11 hours, the other in the same manner as in Example 3 to give 3.2g of the compound Cl.

Example 13 Synthesis of Compound C1

 

Figure imgf000012_0004

8g compound B, 8G N, v-dimethyl formamide dimethyl acetal and 180ml of dioxane under reflux for a reaction time of approximately 12-13 hours, with the same manner as in Example 3 to give 8.7g of compound C. Embodiment Example 14 Synthesis of Compound CI

Figure imgf000013_0001

3g compound B, 3 g of N, N-dimethyl formamide dimethyl acetal and 140ml of toluene, the reaction time is 13-15 hours under reflux, with the same manner as in Example 3 to give 2.9g of the compound Cl.

Example 15 Synthesis of Compound C1

Figure imgf000013_0002

The same as in Example 14, except that reaction time is 10 hours, to obtain 2.6g compound Cl t

Embodiment Example 16 Synthesis of Compound C1

 

Figure imgf000013_0003

500mL three-necked flask, add 3 g of compound B, 3.7 g v, v-dimethylformamide, diethyl acetal and 140ml of dioxane was heated to reflux, TLC monitoring the progress of the reaction, the reaction time of approximately 11-12 hours, After completion of the reaction, the mixture was cooled to room temperature, spin-dry the reaction solution to give 2.5g of the compound Cl.

Example 17 Synthesis of Compound C1

 

Figure imgf000013_0004

G of compound B, 5.1 g of the N, N-dimethyl formamide di-t-butyl acetal was dissolved in 140ml dioxane was heated to reflux the TLC monitoring progress of the reaction, the reaction time of approximately 11-12 hours after the completion of the reaction, was cooled to room temperature, the reaction solution was spin-dry to give 2.6g of the compound Cl.

Embodiment Example 18 Synthesis of Compound CI

 

Figure imgf000014_0001

3g compound B, 4.4g N, N-dimethyl formamide diisopropyl acetal was dissolved in 140ml dioxane was heated to reflux, tlc monitoring the progress of the reaction, the reaction time of approximately 11-12 hours after the completion of the reaction, was cooled to room temperature, the reaction solution was spin-dry to give 2.4g of the compound Cl.

The implementation of the synthesis of Example 19 Icotinib

 

Figure imgf000014_0002

3g compound Cl, 1.3 g inter-aminophenyl acetylene, 130 ml of acetic acid was added 250 ml reaction flask and heated to 70-80

V, TLC monitoring of the reaction. Spin dry the reaction system, methanol was added, and shock dispersion, filtered, and the methanol wash was 2.8g Icotinib. Implementation of Example 20 Icotinib synthesis

 

Figure imgf000014_0003

C1 Icotinib

. Example 25 Icotinib Hydrochloride synthesis

 

Figure imgf000016_0001

Icotinib Hydrochloride

The 500mg Icotinib Add to a 100 ml reaction flask, add 30ml of ethanol was stirred under hydrogen chloride gas was passed into the after, filtered crude hydrochloride Icotinib recrystallized from isopropanol to give 515mg hydrochlorideIcotinib. Example 26 Icotinib Hydrochloride Synthesis

500mg Icotinib Add 100 ml reaction flask, add 40 ml of tetrahydrofuran was stirred under hydrogen chloride gas was passed into the after, filtered crude hydrochloride Icotinib recrystallized from isopropanol to give 500mg hydrochlorideIcotinib. EXAMPLE 27 Icotinib Hydrochloride Synthesis

 

Figure imgf000016_0002

 

500mg Icotinib Add 100 ml reaction flask, add 50 ml of isopropanol and stirred under hydrogen chloride gas was passed into the after, filtered crude hydrochloride Icotinib recrystallized from isopropanol to give 500mg hydrochloride Icotinib.

………………………………………………………………….

 

 

http://www.google.com/patents/EP2392576A1 NMR data: 1H-NMR (Bruker APX-400, solvent: DMSO-d6, TMS as internal standard): δ ppm: 3.58 (dd, 2H, two protons of the crown position 12); 3.60 (dd, 2H, two protons of the crown position 13); 3.73 (dd, 2H, two protons of the crown position 10); 3.80 (dd, 2H, two protons of the crown position 15); 4.30 (s, 1H, proton of the alkynyl); 4.34 (dd, 2H, two protons of the crown position 16); 4.40 (dd, 2H, two protons of the crown position 9); 7.39 (d, 1H, benzene proton at position 25); 7.46 (dd, 1H, benzene proton at position 26); 7.49 (s, 1H, proton of the quinazoline position 6); 7.82 (d, 1H, benzene proton at position 27); 7.94 (t due dd, 1H, proton of the quinazoline position 19); 8.85 (s, 1H, benzene proton at the position 23); 8.87 (s, 1H, proton of the quinazoline position 2); 11.70 (s, 1H, proton of the aromatic amine as salt); 14-16 (bs, 1H, hydrochloride), see Figure 5. NMR data: 13C-NMR (DMSO-d6), see Figure 6. Mass spectrometry (MS): Instrument: ZAB-HS, testing conditions: EI, 200°C, 700ev, MS measured molecular weight: m/z 427.

Share

ERTUGLIFLOZIN

 diabetes  Comments Off on ERTUGLIFLOZIN
Dec 202013
 

ERTUGLIFLOZIN, PFIZER

THERAPEUTIC CLAIM Treatment of type 2 diabetes
CHEMICAL NAMES
1. β-L-Idopyranose, 1,6-anhydro-1-C-[4-chloro-3-[(4-ethoxyphenyl)methyl]phenyl]-5-C-(hydroxymethyl)-
2. (1S,2S,3S,4R,5S)-5-{4-chloro-3-[(4-ethoxyphenyl)methyl]phenyl}-1-(hydroxymethyl)-6,8-dioxabicyclo[3.2.1]octane-2,3,4-triol

PF-04971729, MK 8835

M. Wt: 436.88
Formula: C22H25ClO7
CAS No:. 1210344-57-2

Diabetes looms as a threat to human health worldwide. As a result, considerable research efforts are devoted to identify new and efficacious anti-diabetic agents lacking the side effects associated with some of the current drugs (hypoglycemia, weight gain).Inhibition of sodium-dependent glucose cotransporter 2 (SGLT2), a transporter located in the kidney, is a mechanism that promotes glucosuria and therefore, reduction of plasma glucose concentration. Since the mechanism operates in a glucose-dependent and insulin-independent manner, and is associated with weight loss, it has emerged as a very promising approach to the pathophysiologic treatment of type 2 diabetes. Ertugliflozin (PF-04971729), an anti-diabetic agent currently in development (Phase 3 clinical trials) and belonging to a new class of SGLT2 inhibitors bearing a dioxa-bicyclo[3.2.1]octane bridged ketal motif.

 

 

http://www.google.it/patents/WO2010023594A1?cl=en

Scheme 1 outlines the general procedures one could use to provide compounds of the present invention.

Figure imgf000012_0001

Scheme 1 AIIyI 2,3,4-tιϊ-O-benzyl-D-glucopyranoside (La, where Pg1 is a benzyl group) can be prepared by procedures described by Shinya Hanashima, et al., in Bioorganic & Medicinal Chemistry, 9, 367 (2001 ); Patricia A. Gent et al. in Journal of the Chemical Society, Perkin 1, 1835 (1974); Hans Peter Wessel in the Journal of Carbohydrate Chemistry, 7, 263, (1988); or Yoko Yuasa, et al., in Organic Process Research & Development, 8, 405-407

(2004). In step 1 of Scheme 1 , the hydroxymethylene group can be introduced onto the glycoside by means of a Swern oxidation followed by treatment with formaldehyde in the presence of an alkali metal hydroxide (e.g., sodium hydroxide). This is referred to as an aldol-Cannizzaro reaction. The Swern oxidation is described by Kanji Omura and Daniel Swern in Tetrahedron, 34, 1651 (1978). Modifications of this process known to those of skill in the art may also be used. For example, other oxidants, like stabilized 2- iodoxybenzoic acid described by Ozanne, A. et al. in Organic Letters, 5, 2903 (2003), as well as other oxidants known by those skilled in the art can also be used. The aldol Cannizzaro sequence has been described by Robert Schaffer in the Journal of The American Chemical Society, 81 , 5452 (1959) and Amigues, E.J., et al., in Tetrahedron, 63,

10042 (2007).

In step 2 of Scheme 1 , protecting groups (Pg2) can be added by treating intermediate (MD) with the appropriate reagents and procedures for the particular protecting group desired. For example, p-methoxybenzyl (PMB) groups may be introduced by treatment of intermediate (MD) with p-methoxybenzyl bromide or p-methoxybenzyl chloride in the presence of sodium hydride, potassium hydride, potassium te/t-butoxide in a solvent like tetrahydrofuran, 1 ,2-dimethoxyethane or Λ/,Λ/-dimethylformamide (DMF). Conditions involving para-methoxybenzyltrichloroacetimidate in presence of a catalytic amount of acid (e.g., trifluoromethanesulfonic acid, methanesulfonic acid, or camphorsulfonic acid) in a solvent such as dichloromethane, heptane or hexanes can also be used. Benzyl (Bn) groups may be introduced by treatment of intermediate (MD) with benzyl bromide or benzyl chloride in the presence of sodium hydride, potassium hydride, potassium te/t-butoxide in a solvent like tetrahydrofuran, 1 ,2-dimethoxyethane or Λ/,Λ/-dimethylformamide. Conditions involving benzylthchloroacetimidate in presence of a catalytic amount of acid (e.g., trifluoromethanesulfonic acid, methanesulfonic acid, or camphorsulfonic acid) in a solvent such as dichloromethane, heptane or hexanes can also be used. In step 3 of Scheme 1 , the allyl protection group is removed (e.g., by treatment with palladium chloride in methanol; cosolvent like dichloromethane may also be used; other conditions known by those skilled in the art could also be used, see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991 ) to form the lactol (Ld).

In step 4 of Scheme 1 , oxidation of the unprotected hydroxyl group to an oxo group (e.g., Swern oxidation) then forms the lactone (l-e).

In step 5 of Scheme 1 , the lactone (Le) is reacted with Λ/,O-dimethyl hydroxylamine hydrochloride to form the corresponding Weinreb amide which may exist in equilibrium in a closed/opened form, (l-f/l-g). The “Weinreb amide” (LgJ can be made using procedures well known to those of skill in the art. See, Nahm, S., and S. M. Weinreb, Tetrahedron Letters. 22 (39), 3815-1818 (1981 ). For example, intermediate (l-f/l-α) can be prepared from the commercially available Λ/,O-dimethylhydroxylamine hydrochloride and an activating agent (e.g., trimethylaluminum). In step 6 of Scheme 1 , the arylbenzyl group (Ar) is introduced using the desired organometallic reagent (e.g., organo lithium compound (ArLi) or organomagnesium compound (ArMgX)) in tetrahydrofuran (THF) at a temperature ranging from about -780C to about 2O0C followed by hydrolysis (upon standing in protic conditions) to the corresponding lactol (N) which may be in equilibrium with the corresponding ketone (Ni). The bridged ketal motif found in (A) and (B) can be prepared by removing the protecting groups (Pg2) using the appropriate reagents for the protecting groups employed. For example, the PMB protecting groups may be removed by treatment with trifluoroacetic acid in the presence of anisole and dichloromethane (DCM) at about O0C to about 230C (room temperature). The remaining protecting groups (Pg1) may then be removed using the appropriate chemistry for the particular protecting groups. For example, benzyl protecting groups may be removed by treating with formic acid in the presence of palladium (Pd black) in a protic solvent (e.g., ethanol/THF) at about room temperature to produce the final products (A) and (B). When R1 is CN, the use of a Lewis acid like boron trichloride at a temperature ranging from about -780C to about room temperature in a solvent like dichloromethane or 1 ,2-dichloroethane may also be used to remove benzyl protective and/or para- methoxybenzyl protective groups. When R1 is CN and R2 is (Ci-C4)alkoxy in intermdediate (l-i) or in products (A) or (B), upon treatment with a Lewis acid such as boron trichloride or boron tribomide, partial to complete de-alkylation to the corresponding phenol may occur to lead to the corresponding compound (A) or (B) where R1 is CN and R2 is OH. If this occurs, the (d- C4)alkoxy group may be re-introduced via selective alkylation using a (CrC4) alkyl iodide under mildly basic conditions, for example, potassium carbonate in acetone at a temperature ranging from about room temperature to about 56 degrees Celsius.

When R1 and/or R2 is (CrC4)alkyl-SO2– it is understood by one skilled in the art that the organometallic addition step 6 (Scheme 1 ) will be carried out on the corresponding (d- C4)alkyl-S- containing organometallic reagent. The thio-alkyl is then oxidized at a later stage to the corresponding sulfone using conventional methods known by those skilled in the art.

The compounds of the present invention may be prepared as co-crystals using any suitable method. A representative scheme for preparing such co-crystals is described in Scheme 2.

 

Figure imgf000016_0001

Scheme 2

In Scheme 2, wherein Me is methyl and Et is ethyl, in step 1 , 1-(5-bromo-2- chlorobenzyl)-4-ethoxybenzene is dissolved in 3:1 , toluene: tetrahydrofuran followed by cooling the resulting solution to <-70°C. To this solution is added hexyllithium while maintaining the reaction at <-65°C followed by stirring for 1 hour. (3R,4S,5R,6R)-3,4,5- ths(thmethylsilyloxy)-6-((trimethylsilyloxy)methyl)-tetrahydropyran-2-one (ll-a) is dissolved in toluene and the resulting solution is cooled to -150C. This solution is then added to the – 7O0C aryllithium solution followed by stirring for 1 hour. A solution of methanesulfonic acid in methanol is then added followed by warming to room temperature and stirring for 16 to 24 hours. The reaction is deemed complete when the α-anomer level is < 3%. The reaction is then basified by the addition of 5 M aqueous sodium hydroxide solution. The resulting salts are filtered off followed by concentration of the crude product solution. 2- methyltetrahydrofuran is added as a co-solvent and the organic phase is extracted twice with water. The organic phase is then concentrated to 4 volumes in toluene. This concentrate is then added to a 5:1 , heptane: toluene solution causing precipitate to form. The solids are collected and dried under vacuum to afford a solid.

In step 2 of Scheme 2, to (ll-b) in methylene chloride is added imidazole followed by cooling to O0C and then addition of trimethylsilylchlohde to give the persilylated product.

The reaction is warmed to room temperature and quenched by the addition of water, and the organic phase is washed with water. This crude methylene chloride solution of (ll-c) is dried over sodium sulfate and then taken on crude into the next step.

In step 3 of Scheme 2, the crude solution of (ll-c) in methylene chloride is concentrated to low volume and then the solvent is exchanged to methanol. The methanol solution of (ll-c) is cooled to O0C, then 1 mol% of potassium carbonate is added as a solution in methanol followed by stirring for 5 hours. The reaction is then quenched by addition of 1 mol% acetic acid in methanol, followed by warming to room temperature, solvent exchange to ethyl acetate, and then filtration of the minor amount of inorganic solids. The crude ethyl acetate solution of (ll-d) is taken directly into the next step.

In step 4 of Scheme 2, the crude solution of (ll-d) is concentrated to low volume, then diluted with methylene chloride and dimethylsulfoxide. Triethylamine is added followed by cooling to 1O0C and then sulfur trioxide pyridine complex is added in 3 portions as a solid at 10 minute intervals. The reaction is stirred an additional 3 hours at 1O0C before quenching with water and warming to room temperature. The phases are separated followed by washing the methylene chloride layer with aqueous ammonium chloride. The crude methylene chloride solution of (ll-e) is taken directly into the next step.

In step 5 of Scheme 2, the crude solution of (ll-e) is concentrated to low volume and then the solvent is exchanged to ethanol. Thirty equivalents of aqueous formaldehyde is added followed by warming to 550C. An aqueous solution of 2 equivalents of potassium phosphate, tribasic is added followed by stirring for 24 hours at 550C. The reaction temperature is then raised to 7O0C for an additional 12 hours. The reaction is cooled to room temperature, diluted with te/t-butyl methyl ether and brine. The phases are separated followed by solvent exchange of the organic phase to ethyl acetate. The ethyl acetate phase is washed with brine and concentrated to low volume. The crude concentrate is then purified by silica gel flash chromatography eluting with 5% methanol, 95% toluene. Product containing fractions are combined and concentrated to low volume.

Methanol is added followed by stirring until precipitation occurs. The suspension is cooled and the solids are collected and rinsed with heptane followed by drying. Product (ll-f) is isolated as a solid.

In step 6 of Scheme 2, compound (ll-f) is dissolved in 5 volumes of methylene chloride followed by the addition of 1 mol% SiliaBonc/® tosic acid and stirring for 18 hours at room temperature. The acid catalyst is filtered off and the methylene chloride solution of (ll-g) is taken directly into the next step co-crystallization procedure.

In step 7 of Scheme 2, the methylene chloride solution of (ll-g) is concentrated and then the solvent is exchanged to 2-propanol. Water is added followed by warming to 550C. An aqueous solution of L-pyroglutamic acid is added followed by cooling the resulting solution to room temperature. The solution is then seeded and granulated for 18 hours. After cooling, the solids are collected and rinsed with heptane followed by drying. Product (ll-h) is isolated as a solid.

An alternative synthesis route for compounds (A) of the present invention is depicted in Scheme 3 and described below.

 

Figure imgf000019_0001

Scheme 3

The synthesis of (lll-a), where R3 is an alkyl or fluoro substituted alkyl (except for the carbon adjacent to the oxygen atom) can be prepared in a similar way as described in step 1 of Scheme 2. In step 1 of Scheme 3, the primary hydroxyl group is selectively protected by an appropriate protective group. For example, a trityl group (Pg3 = Tr) can be introduced by treatment of intermediate (lll-a) with chlorotriphenylmethane in presence of a base like pyridine in a solvent like toluene, tetrahydrofuran or dichloromethane at a temperature ranging from about 0 degrees Celsius to about room temperature. Additional examples of such protective groups and experimental conditions are known by those skilled in the art and can be found in T. W. Greene, Protective Groups in Organic Synthesis. John Wiley & Sons, New York, 1991.

In step 2 of Scheme 3, the secondary hydroxyl groups can be protected by the appropriate protecting groups. For example, benzyl groups (Pg4 is Bn) can be introduced by treatment of intermediate (lll-b) with benzyl bromide or benzyl chloride in the presence of sodium hydride, potassium hydride, potassium te/t-butoxide in a solvent like tetrahydrofuran, 1 ,2-dimethoxyethane or Λ/,Λ/-dimethylformamide at a temperature ranging from about 0 degrees Celsius to about 80 degrees Celsius. Acetyl or benzoyl groups (Pg4 = Ac or Bz) may be introduced by treatment of intermediate (lll-b) with acetyl chloride, acetyl bromide or acetic anhydride or benzoyl chloride or benzoic anhydride in the presence of a base like triethylamine, Λ/,Λ/-diisopropylethylamine or 4-

(dimethylamino)pyridine in a solvent like tetrahydrofuran, 1 ,2-dimethoxyethane or dichloromethane at a temperature ranging from about 0 degrees Celsius to about 80 degrees Celsius.

In step 3 of Scheme 3, the primary hydroxyl group is deprotected to lead to intermediate (lll-d). When Pg3 is Tr, intermediate (lll-c) is treated in the presence of an acid like para-toluenesulfonic acid in a alcoholic solvent like methanol at a temperature ranging from about -20 degrees Celsius to about room temperature to provide intermediate (lll-d). Cosolvents like chloroform may be used.

In step 4 of Scheme 3, a hydroxymethylene group is introduced through a process similar to the one already described in Scheme 1 (step 1 ) and Scheme 2 (steps 4 and 5).

Other sources of formaldehyde, like paraformaldehyde in a solvent like ethanol at a temperature ranging from about room temperature to about 70 degrees Celsius in the presence of an alkali metal alkoxide can also be used in this step. When Pg4is Bn, this step provides intermediate (lll-e) and when Pg4 is Ac or Bz, this step provides intermediate (lll-f).

In step 5 of Scheme 3, intermediate (lll-e) is treated with an acid like trifluoroacetic acid or an acidic resin in a solvent like dichloromethane at a temperature ranging from about -10 degrees Celsius to about room temperature to produce intermediate (lll-g).

In step 6 of Scheme 3, the remaining protecting groups (Pg4) may then be removed using the appropriate chemistry for the particular protecting groups. For example, benzyl protecting groups may be removed by treating with formic acid in the presence of palladium (Pd black) in a protic solvent (e.g., ethanol/THF) at about room temperature to produce the final product (A).

In step 7 of Scheme 3, intermediate (lll-f) is treated with an acid like trifluoroacetic acid or an acidic resin in a solvent like dichloromethane at a temperature ranging from about -10 degrees Celsius to about room temperature to produce the final product (A). Another alternative scheme for synthesizing product (A) is depicted in Scheme 4 and described below.

 

Figure imgf000021_0001

Scheme 4 In step 1 of Scheme 4, intermediate (lll-a) is treated with the appropriate arylsulfonyl chloride R4SO2CI or arylsulfonic anhydride R4S(O)2OS(O)2R4 (wherein R4 is an optionally substituted aryl group, such as found in the arylsulfonyl chlorides 4-methyl-benzenesulfonyl chloride, 4-nitro-benzenesulfonyl chloride, 4-fluoro-benzenesulfonyl chloride, 2,6-dichloro- benzenesulfonyl chloride, 4-fluoro-2-methyl-benzenesulfonyl chloride, and 2,4,6-trichloro- benzenesulfonyl chloride, and in the arylsulfonic anhydride, p-toluenesulfonic anhydride) in presence of a base like pyridine, triethylamine, Λ/,Λ/-diisopropylethylamine in a solvent like tetrahydrofuran, 2-methyltetrahydrofuran at a temperature ranging from about -20 degrees Celsius to about room temperature. Some Lewis acids like zinc(ll) bromide may be used as additives. In step 2 of Scheme 4, intermediate (IV-a) is submitted to a Kornblum-type oxidation

(see, Kornblum, N., et al., Journal of The American Chemical Society, 81 , 4113 (1959)) to produce the corresponding aldehyde which may exist in equilibrium with the corresponding hydrate and/or hemiacetal form. For example intermediate (IV-a) is treated in the presence of a base like pyridine, 2,6-lutidine, 2,4,6-collidine, Λ/,Λ/-diisopropylethylamine, A- (dimethylamino)pyridine in a solvent like dimethyl sulfoxide at a temperature ranging from about room temperature to about 150 degrees Celsius. The aldehyde intermediate produced is then submitted to the aldol/Cannizzaro conditions described for step 1 (Scheme 1 ) and step 5 (Scheme 2) to produce intermediate (IV-b). In step 3 of Scheme 4, intermediate (IV-b) is treated with an acid like thfluoroacetic acid or an acidic resin in a solvent like dichloromethane at a temperature ranging from about -10 degrees Celsius to about room temperature to produce the final product (A).

When R2 is (C2-C4)alkynyl the process may be performed using Scheme 5, wherein R6 is H or (CrC2)alkyl.

 

Figure imgf000022_0001

Scheme 5

In step 1 of Scheme 5, which provides intermediate (V-i), the organometallic addition step is carried out in a similar way to the one described in Schemel , step 6, using the organometallic reagent derived from (V-a), where Pg5 is a suitable protective group for the hydroxyl group. For instance Pgs can be a te/t-butyldimethylsilyl group (TBS) (see

US2007/0054867 for preparation of for instance {4-[(5-bromo-2-chloro-phenyl)-methyl]- phenoxy}-te/t-butyl-dimethyl-silane).

In step 2 of Scheme 5, when Pg2 = PMB, intermediate (V-i) is treated with an acid like trifluoroacetic acid, methanesulfonic acid or an acidic resin in presence of anisole in a solvent like dichloromethane at a temperature ranging from about -10 degrees Celsius to about room temperature to produce intermediate (V-j).

In step 3 of Scheme 5, protecting groups (Pg5) and (Pg1) can be removed to provide (V-k). Typically (Pg5) is TBS and Pg1 is Bn. In this circumstance, the protecting groups are removed by sequential treatment of (V-j) with 1 ) tetrabutylammonium fluoride in a solvent like tetrahydrofuran or 2-methyltetrahydrofuran at a temperature ranging from 0 degrees

Celsius to about 40 degrees Celsius and 2) treatment with formic acid in the presence of palladium (Pd black) in a protic solvent (e.g., ethanol/THF) at about room temperature. In this sequence, the order of the 2 reactions is interchangeable.

In step 4 of Scheme 5, intermediate (V-k) is treated with N,N-bis- (trifluoromethanesulfonyl)-aniline in presence of a base like triethylamine or 4- dimethyaminopyridine in a solvent like dichloromethane or 1 ,2-dichloroethane at a temperature ranging from 0 degrees Celsius to about 40 degrees Celsius to produce intermediate (V-I).

In step 5 of Scheme 5, intermediate (V-I) is subjected to a Sonogashira-type reaction (see, Sonogashira, K. Coupling Reactions Between sp2 and sp Carbon Centers. In

Comprehensive Organic Synthesis (eds. Trost, B. M., Fleming, I.), 3, 521-549, (Pergamon, Oxford, 1991 )).

Figure imgf000006_0001

IS ERTUGLIFLOZIN

Example 4

(1 S.2S.3S.4R.5S)-5-[4-chloro-3-(4-ethoxy-benzyl)-Dhen yll- 1 -h vdroxymeth yl-6.8-dioxa- bicvclo[3.2.1loctane-2,3Λ-triol (4A) and (1S,2S,3SΛS,5S)-5-[4-chloro-3-(4-ethoxy- benzvD-phen yll- 1 -h vdroxymeth yl-6, 8-dioxa-bicvclo[3.2.1 loctane-2, 3, 4-triol (4B):

Figure imgf000067_0001

To a solution of {(2S,3S)-2,3,4-tris-benzyloxy-5-[4-chloro-3-(4-ethoxy-benzyl)-phenyl]-6,8- dioxa-bicyclo[3.2.1]oct-1-yl}-methanol (l-4k: 335 mg) in ethanol/tetrahydrofuran (10 ml_, 4/1 volume) was added successively formic acid (420 microL, 22 equivalents) and palladium black (208 mg, 4 equivalents) and the resulting mixture was stirred at room temperature. After 1 hour, additional formic acid (420 microL, 22 equivalents) and palladium black (208 mg, 4 equivalents) were added and the mixture was allowed to stir for an additional hour at room temperature. The palladium was filtered and the crude mixture obtained after evaporation of solvent was purified by HPLC preparative.

HPLC preparative: reverse phase C18 Gemini column 5 micrometer 30 x 100 mm, 40 mL/minute, gradient of acetonitrile/0.1 % formic acid : water/0.1 % formic acid; 25 to 50% of acetonitrile/0.1 % formic acid over 18 minutes; UV detection: 220 nm. The HPLC indicated a ratio of diastereomers of 1.1 :1 (4A:4B). 4A: (60 mg, 29% yield); Rt = 12.4 minutes; the fractions containing the product were concentrated under reduced pressure. The crude material was precipitated from ethyl acetate and heptane. The resulting white solid was washed with heptane 2 times and dried under reduced pressure.

MS (LCMS) 437.3 (M+H+; positive mode); 481.3 (M+HCO2 ~; negative mode). 1H NMR (400 MHz, methanol-d4) delta 7.43 (d, 1 H, J = 1.9 Hz), 7.36 (dd, 1 H, J = 8.3 and 2

Hz), 7.32 (d, 1 H, J = 8.3 Hz), 7.08-7.04 (m, 2H), 6.79-6.75 (m, 2H), 4.12 (d, 1 H, J = 7.5 Hz), 4.00 (s, 2H), 3.96 (q, 2H, J = 7.0 Hz), 3.81 (d, 1 H, J = 12.5 Hz), 3.75 (dd, 1 H, J = 8.3 and 1.3 Hz), 3.65 (d, 1 H, J = 12.5 Hz), 3.63 (t, 1 H, J = 8.2 Hz), 3.57 (dd, 1 H, J = 7.5 and 1.3 Hz), 3.52 (d, 1 H, J = 8.0 Hz), 1.33 (t, 3H, J = 6.9 Hz). HRMS calculated for C22H26O7CI (M+H+) 437.1361 , found 437.1360.

4B: (30 mg, 15% yield); Rt = 13.2 minutes; the fractions containing the product were concentrated under reduced pressure. The crude material was precipitated from ethyl acetate and heptane. The resulting white solid was washed with heptane 2 times and dried under reduced pressure.

MS (LCMS) 437.3 (M+H+; positive mode) 481.3 (M+HCO2 , negative mode). 1H NMR (400 MHz, methanol-d4) delta 7.48 (d, 1 H, J = 1.9 Hz) 7.40 (dd, 1 H, J = 8.1 and 1.9 Hz), 7.32 (d, 1 H, J = 8.3 Hz), 7.08-7.03 (m, 2H), 6.80-6.74 (m, 2H), 4.04-3.99 (m, 3H), 3.95 (q, 2H, J = 7 Hz), 3.89-3.81 (m, 4H), 3.73 (d, 1 H, J = 12.5 Hz), 3.49 (d, 1 H, J = 7.3 Hz), 1.32 (t, 3H, J = 7 Hz). HRMS calculated for C22H26O7CI (M+H+) 437.1361 , found 437.1358.
Merck & Co., Inc. and Pfizer Enter Worldwide Collaboration Agreement to Develop and Commercialize Ertugliflozin, an Investigational Medicine for Type 2 Diabetes

Monday, April 29, 2013 9:23 am EDT

Merck & Co., Inc. (NYSE: MRK), known as MSD outside the United States and Canada (“Merck”), and Pfizer Inc. (NYSE:PFE) today announced that they have entered into a worldwide (except Japan) collaboration agreement for the development and commercialization of Pfizer’s ertugliflozin (PF-04971729), an investigational oral sodium glucose cotransporter (SGLT2) inhibitor being evaluated for the treatment of type 2 diabetes. Ertugliflozin is Phase III ready, with trials expected to begin later in 2013.

“We are pleased to join forces with Merck in the battle against type 2 diabetes and the burden that it poses on global health,” said John Young, president and general manager, Pfizer Primary Care. “Through this collaboration, we believe we can build on Merck’s leadership position in diabetes care with the introduction of ertugliflozin, an innovative SGLT2 inhibitor discovered by Pfizer scientists.”

Under the terms of the agreement, Merck, through a subsidiary, and Pfizer will collaborate on the clinical development and commercialization of ertugliflozin and ertugliflozin-containing fixed-dose combinations with metformin and JANUVIA® (sitagliptin) tablets. Merck will continue to retain the rights to its existing portfolio of sitagliptin-containing products. Pfizer has received an upfront payment and milestones of $60 million and will be eligible for additional payments associated with the achievement of pre-specified future clinical, regulatory and commercial milestones. Merck and Pfizer will share potential revenues and certain costs on a 60/40 percent basis.

“Merck continues to build upon our leadership position in the oral treatment of type 2 diabetes through our own research and business development,” said Nancy Thornberry, senior vice president and Diabetes and Endocrinology franchise head, Merck Research Laboratories. “We believe ertugliflozin has the potential to complement our strong portfolio of investigational and marketed products, and we look forward to collaborating with Pfizer on its development.”

……………….

Development of an Early-Phase Bulk Enabling Route to Sodium-Dependent Glucose Cotransporter 2 Inhibitor Ertugliflozin

David Bernhardson, Thomas A. Brandt, Catherine A. Hulford, Richard S. Lehner, Brian R. Preston, Kristin Price, John F. Sagal, Michael J. St. Pierre, Peter H. Thompson, and Benjamin Thuma
pp 57–65
Publication Date (Web): January 3, 2014 (Article)
DOI: 10.1021/op400289z

 

Abstract Image

The development and optimization of a scalable synthesis of sodium-dependent glucose cotransporter 2 inhibitor, ertugliflozin, for the treatment of type-2 diabetes is described. Highlights of the chemistry are a concise, four-step synthesis of a structurally complex API from known intermediate 4 via persilylation–selective monodesilylation, primary alcohol oxidation, aldol-crossed-Cannizzaro reaction, and solid-phase acid-catalyzed bicyclic ketal formation. The final API was isolated as the l-pyroglutamic acid cocrystal.

Inline image 1

1= ertugliflozin

Inline image 2

Inline image 3

 

PF-04971729, a potent and selective inhibitor of the sodium-dependent glucose cotransporter 2, is currently in phase 2 trials for the treatment of diabetes mellitus. Inhibitory effects against the organic cation transporter 2-mediated uptake of [14C] metformin by PF- 04971729 also were very weak (IC50 900μM). The disposition of PF-04971729, an orally active selective inhibitor of the sodium-dependent glucose cotransporter 2, was studied after a single 25-mg oral dose of [14C]-PF-04971729 to healthy human subjects. The absorption of PF-04971729 in humans was rapid with a Tmax at ~ 1.0 h. Of the total radioactivity excreted in feces and urine, unchanged PF-04971729 collectively accounted for ~ 35.3% of the dose, suggestive of moderate metabolic elimination in humans.
 
 
References on PF-04971729:
[1]. 1. Amit S. Kalgutkar, Meera Tugnait, Tong Zhu, et al.Preclinical Species and Human Disposition of PF-04971729, a Selective Inhibitor of the Sodium-Dependent Glucose cotransporter 2 and Clinical Candidate for the Treatment of Type 2 . Diabetes Mellitus Drug Metabolism and Diposition, 2011, 39 (9):. 1609-1619
Abstract
(1S, 2S, 3S, 4R, 5S) -5 – [4-Chloro-3-(4-ethoxybenzyl) phenyl] -1 -hydroxymethyl-6 ,8-dioxabicyclo [3.2.1] octane-2 ,3,4-triol (PF-04971729), a potent and selective inhibitor of the sodium-dependent glucose cotransporter 2, is currently in phase 2 trials for the treatment of diabetes mellitus. This article describes the preclinical species and in vitro human disposition characteristics of PF-04971729 that were used in experiments performed to support the first-in-human study. Plasma clearance was low in rats (4.04 ml · min? 1 · kg? 1) and dogs (1.64 ml · min? 1 · kg? 1), resulting in half-lives of 4.10 and 7.63 h, respectively. Moderate to good bioavailability in rats (69%) and dogs (94%) was . observed after oral dosing The in vitro biotransformation profile of PF-04971729 in liver microsomes and cryopreserved hepatocytes from rat, dog, and human was qualitatively similar;. prominent metabolic pathways included monohydroxylation, O-deethylation, and glucuronidation No human-specific metabolites of PF-04971729 were detected in in vitro studies. Reaction phenotyping studies using recombinant enzymes indicated a role of CYP3A4/3A5, CYP2D6, and UGT1A9/2B7 in the metabolism of PF-04971729. No competitive or time-dependent inhibition of the major human cytochrome P450 enzymes was discerned with PF-04971729. Inhibitory effects against the organic cation transporter 2-mediated uptake of [14C] metformin by PF-04971729 also were very weak (IC50 =? 900 μM). Single-species allometric scaling of rat pharmacokinetics of PF-04971729 was used to predict human clearance, distribution volume, and oral bioavailability. Human pharmacokinetic predictions were consistent with the potential for a low daily dose. First-in-human studies after oral administration indicated that the human pharmacokinetics / dose predictions for PF -04971729 were in the range that is likely to yield a favorable pharmacodynamic response.
. [2] … Timothy Colin Hardman, Simon William Dubrey Development and potential role of type-2 sodium-glucose transporter Inhibitors for Management of type 2 Diabetes Diabetes Ther 2011 September; 2 (3):. 133-145
Abstract
There is a recognized need for new treatment options for type 2 diabetes mellitus (T2DM). Recovery of glucose from the glomerular filtrate represents an important mechanism in maintaining glucose homeostasis and represents a novel target for the management of T2DM. Recovery of glucose from the glomerular filtrate is executed principally by the type 2 sodium-glucose cotransporter (SGLT2). Inhibition of SGLT2 promotes glucose excretion and normalizes glycemia in animal models. First reports of specifically designed SGLT2 inhibitors began to appear in the second half of the 1990s. Several candidate SGLT2 inhibitors are currently under development, with four in the later stages of clinical testing. The safety profile of SGLT2 inhibitors is expected to be good, as their target is a highly specific membrane transporter expressed almost exclusively within the renal tubules. One safety concern is that of glycosuria , which could predispose patients to increased urinary tract infections. So far the reported safety profile of SGLT2 inhibitors in clinical studies appears to confirm that the class is well tolerated. Where SGLT2 inhibitors will fit in the current cascade of treatments for T2DM has yet to be established. The expected favorable safety profile and insulin-independent mechanism of action appear to support their use in combination with other antidiabetic drugs. Promotion of glucose excretion introduces the opportunity to clear calories (80-90 g [300-400 calories] of glucose per day) in patients that are generally overweight, and is expected to work synergistically with weight reduction programs. Experience will most likely lead to better understanding of which patients are likely to respond best to SGLT2 inhibitors, and under what circumstances.
[3]. Zhuang Miao, Gianluca Nucci, Neeta Amin. Pharmacokinetics, Metabolism and Excretion of the Anti-Diabetic Agent Ertugliflozin (PF-04971729) in Healthy Male the Subjects. Drug Metabolism and Diposition.
Abstract
The Disposition of ertugliflozin (PF-04971729) , an orally active selective inhibitor of the sodium-dependent glucose cotransporter 2, was studied after a single 25-mg oral dose of [14C]-PF-04971729 to healthy human subjects. Mass balance was achieved with approximately 91% of the administered dose recovered in urine and feces. The total administered radioactivity excreted in feces and urine was 40.9% and 50.2%, respectively. The absorption of PF-04971729 in humans was rapid with a Tmax at ~ 1.0 h. Of the total radioactivity excreted in feces and urine, unchanged PF-04971729 collectively accounted for ~ 35.3% of the dose, suggestive of moderate metabolic elimination in humans. The principal biotransformation pathway involved glucuronidation of the glycoside hydroxyl groups to yield three regioisomeric metabolites M4a, M4b and M4c (~ 39.3% of the dose in urine) of which M4c was the major regioisomer (~ 31.7% of the dose). The structure of M4a and M4c were confirmed to be PF-04971729-4-O-β-and-3-O-β-glucuronide , respectively, via comparison of the HPLC retention time and mass spectra with authentic standards. A minor metabolic fate involved oxidation by cytochrome P450 to yield monohydroxylated metabolites M1 and M3 and des-ethyl PF-04971729 (M2), which accounted for ~ 5.2% of the dose in excreta. In plasma, unchanged PF-04971729 and the corresponding 4-O-β-(M4a) and 3-O-β-(M4c) glucuronides were the principal components, which accounted for 49.9, 12.2 and 24.1% of the circulating radioactivity. Overall, these data suggest that PF-04971729 is well absorbed in humans, and eliminated largely via glucuronidation.
. [4] .. Tristan S. Maurer, Avijit Ghosh, Nahor Haddish-Berhane pharmacodynamic Model of Sodium-Glucose Transporter 2 (SGLT2) Inhibition: Implications for Quantitative Translational Pharmacology AAPS J. 2011; 13 (4): 576-584
Abstract
Sodium-glucose co-transporter-2 (SGLT2) inhibitors are an emerging class of agents for use in the treatment of type 2 diabetes mellitus (T2DM). Inhibition of SGLT2 leads to improved glycemic control through increased urinary glucose excretion (UGE). In this study, a biologically based pharmacokinetic / pharmacodynamic (PK / PD) model of SGLT2 inhibitor-mediated UGE was developed. The derived model was used to characterize the acute PK / PD relationship of the SGLT2 inhibitor, dapagliflozin, in rats. The quantitative translational pharmacology of dapagliflozin was examined through both prospective simulation and direct modeling of mean literature data obtained for dapagliflozin in healthy subjects. Prospective simulations provided time courses of UGE that were of consistent shape to clinical observations, but were modestly biased toward under prediction. Direct modeling provided an improved characterization of the data and precise parameter estimates which were reasonably consistent with those predicted from preclinical data. Overall, these results indicate that the acute clinical pharmacology of SGLT2 inhibitors in healthy subjects can be reasonably well predicted from preclinical data through rational accounting of species differences in pharmacokinetics, physiology, and SGLT2 pharmacology. Because these data can be generated at the earliest stages of drug discovery, the proposed model is useful in the design and development of novel SGLT2 inhibitors. In addition, this model is expected to serve as a useful foundation for future efforts to understand and predict the effects of SGLT2 inhibition under chronic administration and in other patient populations.
[5]. Yoojin Kim, Ambika R Babu Clinical potential of sodium-glucose cotransporter 2 Inhibitors in the Management of type 2 Diabetes Diabetes Obes Metab Syndr 2012; 5:…. 313-327
Abstract
Background The Kidney plays an Important role in glucose metabolism, and has been considered a target for therapeutic intervention. The sodium-glucose cotransporter type 2 (SGLT2) mediates most of the glucose reabsorption from the proximal renal tubule. Inhibition of SGLT2 leads to glucosuria and provides a unique mechanism to lower elevated blood glucose levels in diabetes. The purpose of this review is to explore the physiology of SGLT2 and discuss several SGLT2 inhibitors which have clinical data in patients with type 2 diabetes. Methods We performed a PubMed search using the terms “SGLT2” and “SGLT2 inhibitor” through April 10, 2012. Published articles, press releases, and abstracts presented at national and international meetings were considered. Results SGLT2 inhibitors correct a novel pathophysiological defect, have an insulin-independent action, are efficacious with glycosylated hemoglobin reduction ranging from 0.5% to 1.5%, promote weight loss, have a low incidence of hypoglycemia, complement the action of other antidiabetic agents, and can be used at any stage of diabetes. They are generally well tolerated. However, due to side effects, such as repeated urinary tract and genital infections, increased hematocrit, and decreased blood pressure, appropriate patient selection for drug initiation and close monitoring after initiation will be important. Results of ongoing clinical studies of the effect of SGLT2 inhibitors on diabetic complications and cardiovascular safety are crucial to determine the risk -benefit ratio. A recent decision by the Committee for Medicinal Products for Human Use of the European Medicines Agency has recommended approval of dapagliflozin for the treatment of type 2 diabetes as an adjunct to diet and exercise, in combination with other glucose-lowering medicinal products , including insulin, and as a monotherapy for metformin-intolerant patients. Clinical research also remains to be carried out on the long-term effects of glucosuria and other potential effects of SGLT2 inhibitors, especially in view of the observed increase in the incidence of bladder and breast cancer SGLT2 inhibitors represent a promising approach for the treatment of diabetes, and could potentially be an addition to existing therapies Keywords:.. sodium-glucose cotransporter type 2, SGLT2, inhibitors, kidney, glucosuria, oral diabetes agent, weight loss.
[6]. Clinical Trials with PF-04971729

……….

 

Papua New Guinea

Papua New Guinea – Wikipedia, the free encyclopedia

en.wikipedia.org/wiki/Papua_New_Guinea

Papua New Guinea (PNG; /ˈpæpə njuː ˈɡɪniː/ PAP-pə-new-GHIN-ee; Tok Pisin: Papua Niugini; Hiri Motu: Papua Niu Gini), officially the Independent State …

 

 

 

 

 

 

 

 

 

//////////

Share

A Drug Against Obesity?

 Uncategorized  Comments Off on A Drug Against Obesity?
Dec 202013
 

Small synthetic molecule identified that ameliorates obesity-related disorders in animal models

Read more

 

Share
Dec 192013
 

 

LUSEOGLIFLOZIN, CAS 898537-18-3
An antidiabetic agent that inhibits sodium-dependent glucose cotransporter 2 (SGLT2).

Taisho (Originator), PHASE 3

TS-071

better version

http://newdrugapprovals.org/2014/07/01/luseogliflozin-ts-071-strongly-inhibited-sglt2-activity/

WO 2010119990

WO2006073197

TS-071, an SGLT-2 inhibitor, is in phase III clinical development at Taisho for the oral treatment of type 1 and type 2 diabetes

In 2012, the product was licensed to Novartis and Taisho Toyama Pharmaceutical by Taisho in Japan for comarketing for the treatment of type 2 diabetes.

Diabetes is a metabolic disorder which is rapidly emerging as a global health care problem that threatens to reach pandemic levels. The number of people with diabetes worldwide is expected to rise from 285 million in 2010 to 438 million by 2030. Diabetes results from deficiency in insulin because of impaired pancreatic β-cell function or from resistance to insulin in body, thus leading to abnormally high levels of blood glucose.

Diabetes which results from complete deficiency in insulin secretion is Type 1 diabetes and the diabetes due to resistance to insulin activity together with an inadequate insulin secretion is Type 2 diabetes. Type 2 diabetes (Non insulin dependent diabetes) accounts for 90-95 % of all diabetes. An early defect in Type 2 diabetes mellitus is insulin resistance which is a state of reduced responsiveness to circulating concentrations of insulin and is often present years before clinical diagnosis of diabetes. A key component of the pathophysiology of Type 2 diabetes mellitus involves an impaired pancreatic β-cell function which eventually contributes to decreased insulin secretion in response to elevated plasma glucose. The β-cell compensates for insulin resistance by increasing the insulin secretion, eventually resulting in reduced β-cell mass. Consequently, blood glucose levels stay at abnormally high levels (hyperglycemia).

Hyperglycemia is central to both the vascular consequences of diabetes and the progressive nature of the disease itself. Chronic hyperglycemia leads to decrease in insulin secretion and further to decrease in insulin sensitivity. As a result, the blood glucose concentration is increased, leading to diabetes, which is self-exacerbated. Chronic hyperglycemia has been shown to result in higher protein glycation, cell apoptosis and increased oxidative stress; leading to complications such as cardiovascular disease, stroke, nephropathy, retinopathy (leading to visual impairment or blindness), neuropathy, hypertension, dyslipidemia, premature atherosclerosis, diabetic foot ulcer and obesity. So, when a person suffers from diabetes, it becomes important to control the blood glucose level. Normalization of plasma glucose in Type 2 diabetes patients improves insulin action and may offset the development of beta cell failure and diabetic complications in the advanced stages of the disease.

Diabetes is basically treated by diet and exercise therapies. However, when sufficient relief is not obtained by these therapies, medicament is prescribed alongwith. Various antidiabetic agents being currently used include biguanides (decrease glucose production in the liver and increase sensitivity to insulin), sulfonylureas and meglitinides (stimulate insulin production), a-glucosidase inhibitors (slow down starch absorption and glucose production) and thiazolidinediones (increase insulin sensitivity). These therapies have various side effects: biguanides cause lactic acidosis, sulfonylurea compounds cause significant hypoglycemia, a-glucosidase inhibitors cause abdominal bloating and diarrhea, and thiazolidinediones cause edema and weight gain. Recently introduced line of therapy includes inhibitors of dipeptidyl peptidase-IV (DPP-IV) enzyme, which may be useful in the treatment of diabetes, particularly in Type 2 diabetes. DPP-IV inhibitors lead to decrease in inactivation of incretins glucagon like peptide- 1 (GLP-1) and gastric inhibitory peptide (GIP), thus leading to increased production of insulin by the pancreas in a glucose dependent manner. All of these therapies discussed, have an insulin dependent mechanism.

Another mechanism which offers insulin independent means of reducing glycemic levels, is the inhibition of sodium glucose co-transporters (SGLTs). In healthy individuals, almost 99% of the plasma glucose filtered in the kidneys is reabsorbed, thus leading to only less than 1% of the total filtered glucose being excreted in urine. Two types of SGLTs, SGLT-1 and SGLT-2, enable the kidneys to recover filtered glucose. SGLT-1 is a low capacity, high-affinity transporter expressed in the gut (small intestine epithelium), heart, and kidney (S3 segment of the renal proximal tubule), whereas SGLT-2 (a 672 amino acid protein containing 14 membrane-spanning segments), is a low affinity, high capacity glucose ” transporter, located mainly in the S 1 segment of the proximal tubule of the kidney. SGLT-2 facilitates approximately 90% of glucose reabsorption and the rate of glucose filtration increases proportionally as the glycemic level increases. The inhibition of SGLT-2 should be highly selective, because non-selective inhibition leads to complications such as severe, sometimes fatal diarrhea, dehydration, peripheral insulin resistance, hypoglycemia in CNS and an impaired glucose uptake in the intestine.

Humans lacking a functional SGLT-2 gene appear to live normal lives, other than exhibiting copious glucose excretion with no adverse effects on carbohydrate metabolism. However, humans with SGLT-1 gene mutations are unable to transport glucose or galactose normally across the intestinal wall, resulting in condition known as glucose-galactose malabsorption syndrome.

Hence, competitive inhibition of SGLT-2, leading to renal excretion of glucose represents an attractive approach to normalize the high blood glucose associated with diabetes. Lower blood glucose levels would, in turn, lead to reduced rates of protein glycation, improved insulin sensitivity in liver and peripheral tissues, and improved cell function. As a consequence of progressive reduction in hepatic insulin resistance, the elevated hepatic glucose output which is characteristic of Type 2 diabetes would be expected to gradually diminish to normal values. In addition, excretion of glucose may reduce overall caloric load and lead to weight loss. Risk of hypoglycemia associated with SGLT-2 inhibition mechanism is low, because there is no interference with the normal counter regulatory mechanisms for glucose.

The first known non-selective SGLT-2 inhibitor was the natural product phlorizin

(glucose, 1 -[2-P-D-glucopyranosyloxy)-4,6-dihydroxyphenyl]-3-(4-hydroxyphenyl)- 1 – propanone). Subsequently, several other synthetic analogues were derived based on the structure of phlorizin. Optimisation of the scaffolds to achieve selective SGLT-2 inhibitors led to the discovery of several considerably different scaffolds.

C-glycoside derivatives have been disclosed, for example, in PCT publications

W.O20040131 18, WO2005085265, WO2006008038, WO2006034489, WO2006037537, WO2006010557, WO2006089872, WO2006002912, WO2006054629, WO2006064033, WO2007136116, WO2007000445, WO2007093610, WO2008069327, WO2008020011, WO2008013321, WO2008013277, WO2008042688, WO2008122014, WO2008116195, WO2008042688, WO2009026537, WO2010147430, WO2010095768, WO2010023594, WO2010022313, WO2011051864, WO201 1048148 and WO2012019496 US patents US65151 17B2, US6936590B2 and US7202350B2 and Japanese patent application JP2004359630. The compounds shown below are the SGLT-2 inhibitors which have reached advanced stages of human clinical trials: Bristol-Myers Squibb’s “Dapagliflozin” with Formula A, Mitsubishi Tanabe and Johnson & Johnson’s “Canagliflozin” with Formula B, Lexicon’s “Lx-421 1” with Formula C, Boehringer Ingelheim and Eli Lilly’s “Empagliflozin” with Formula D, Roche and Chugai’s “Tofogliflozin” with Formula E, Taisho’s “Luseogliflozin” with Formula F, Pfizer’ s “Ertugliflozin” with Formula G and Astellas and Kotobuki’s “Ipragliflozin” with Formula H.

 

Figure imgf000005_0001

Formula G                                                                                                                  Formula H

In spite of all these molecules in advanced stages of human clinical trials, there is still no drug available in the market as SGLT-2 inhibitor. Out of the potential candidates entering the clinical stages, many have been discontinued, emphasizing the unmet need. Thus there is an ongoing requirement to screen more scaffolds useful as SGLT-2 inhibitors that can have advantageous potency, stability, selectivity, better half-life, and/ or better pharmacodynamic properties. In this regard, a novel class of SGLT-2 inhibitors is provided herein

better version

http://newdrugapprovals.org/2014/07/01/luseogliflozin-ts-071-strongly-inhibited-sglt2-activity/

SYNTHESIS

EP1845095A1

 

      Example 5

    • Figure imgb0035

Synthesis of 2,3,4,6-tetra-O-benzyl-1-C-[2-methoxy-4-methyl-(4-ethoxybenzyl)phenyl]-5-thio-D-glucopyranose

    • Five drops of 1,2-dibromoethane were added to a mixture of magnesium (41 mg, 1.67 mmol), 1-bromo-3-(4-ethoxybenzyl)-6-methoxy-4-methylbenzene (0.51 g, 1.51 mmol) and tetrahydrofuran (2 mL). After heated to reflux for one hour, this mixture was allowed to stand still to room temperature to prepare a Grignard reagent. A tetrahydrofuran solution (1.40 mL) of 1.0 M i-propyl magnesium chloride and the prepared Grignard reagent were added dropwise sequentially to a tetrahydrofuran (5 mL) solution of 2,3,4,6-tetra-O-benzyl-5-thio-D-glucono-1,5-lactone (0.76 g, 1.38 mmol) while cooled on ice and the mixture was stirred for 30 minutes. After the reaction mixture was added with a saturated ammonium chloride aqueous solution and extracted with ethyl acetate, the organic phase was washed with brine and dried with anhydrous magnesium sulfate. After the desiccant was filtered off, the residue obtained by evaporating the solvent under reduced pressure was purified by silica gel column chromatography (hexane:ethyl acetate =4:1) to obtain (0.76 g, 68%) a yellow oily title compound.
      1H NMR (300 MHz, CHLOROFORM-d) δ ppm 1.37 (t, J=6.92 Hz, 3 H) 2.21 (s, 3 H) 3.51 – 4.20 (m, 12 H) 3.85 – 3.89 (m, 3 H) 4.51 (s, 2 H) 4.65 (d, J=10.72 Hz, 1 H) 4.71 (d, J=5.75 Hz, 1 H) 4.78 – 4.99 (m, 3 H) 6.59 – 7.43 (m, 26 H)

Example 6

    • [0315]
      Figure imgb0036

Synthesis of (1S)-1,5-anhydro-2,3,4,6-tetra-O-benzyl-1-[2-methoxy-4-methyl-5-(4-ethoxybenzyl)phenyl]-1-thio-D-glucitol

    • An acetonitrile (18 mL) solution of 2,3,4,6-tetra-O-benzyl-1-C-[2-methoxy-4-methyl-5-(4-ethoxybenzyl)phenyl]-5-thio-D-glucopyranose (840 mg, 1.04 mmol) was added sequentially with Et3SiH (0.415 mL, 2.60 mmol) and BF3·Et2O (0.198 mL, 1.56 mmol) at -18°C and stirred for an hour. After the reaction mixture was added with a saturated sodium bicarbonate aqueous solution and extracted with ethyl acetate, the organic phase was washed with brine and then dried with anhydrous magnesium sulfate. After the desiccant was filtered off, the residue obtained by evaporating the solvent under reduced pressure was purified by silica gel column chromatography (hexane:ethyl acetate=4:1) to obtain the title compound (640 mg, 77%).
      1H NMR (600 MHz, CHLOROFORM-d) δ ppm 1.35 (t, J=6.88 Hz, 3 H) 2.21 (s, 3 H) 3.02 – 3.21 (m, 1 H) 3.55 (t,J=9.40 Hz, 1 H) 3.71 (s, 1 H) 3.74 – 3.97 (m, 10 H) 4.01 (s, 1 H) 4.45 – 4.56 (m, 3 H) 4.60 (d, J=10.55 Hz, 2 H) 4.86 (s, 2 H) 4.90 (d, J=10.55 Hz, 1H) 6.58 – 6.76 (m, 5 H) 6.90 (d, J=7.34 Hz, 1 H) 7.09 – 7.19 (m, 5 H) 7.23 – 7.35 (m, 15 H).
      ESI m/z = 812 (M+NH4).

Example 7

    • Figure imgb0037

Synthesis of (1S)-1,5-anhydro-1-[3-(4-ethoxybenzyl)-6-methoxy-4-methylphenyl]-1-thio-D-glucitol

  • A mixture of (1S)-1,5-anhydro-2,3,4,6-tetra-O-benzyl-1-[2-methoxy-4-methyl-5-(4-ethoxybenzyl)phenyl]-1-thio-D-glucitol (630 mg, 0.792 mmol), 20% palladium hydroxide on activated carbon (650 mg) and ethyl acetate (10 mL) – ethanol (10 mL) was stirred under hydrogen atmosphere at room temperature for 66 hours. The insolubles in the reaction mixture were filtered off with celite and the filtrate was concentrated. The obtained residue was purified by silica gel column chromatography (chloroform:methanol =10:1) to obtain a colorless powdery title compound (280 mg, 81%) as 0.5 hydrate. 1H NMR (600 MHz, METHANOL- d4) δ ppm 1.35 (t, J=6.9 Hz, 3 H) 2.17 (s, 3 H) 2.92 – 3.01 (m, 1 H) 3.24 (t, J=8.71 Hz, 1 H) 3.54 – 3.60 (m, 1 H) 3.72 (dd, J=11.5, 6.4 Hz, 1 H) 3.81 (s, 3 H) 3.83 (s, 2 H) 3.94 (dd, J=11.5, 3.7 Hz, 1 H) 3.97 (q, J=6.9 Hz, 2 H) 4.33 (s, 1 H) 6.77 (d, J=8.3 Hz, 2 H) 6.76 (s, 1 H) 6.99 (d, J=8.3 Hz, 2 H) 7.10 (s, 1 H). ESI m/z = 452 (M+NH4+), 493 (M+CH3CO2-). mp 155.0-157.0°C. Anal. Calcd for C23H30O6S·0.5H2O: C, 62.28; H, 7.06. Found: C, 62.39; H, 7.10.

better version

http://newdrugapprovals.org/2014/07/01/luseogliflozin-ts-071-strongly-inhibited-sglt2-activity/

 

Share
Dec 192013
 

ChemSpider 2D Image | remogliflozin etabonate | C26H38N2O9

REMOGLIFLOZIN ETABONATE

5-methyl-4-[4-(1-methylethoxy)benzyl]-1-(1-methylethyl)-1H-pyrazol-3-yl 6-O-(ethoxycarbonyl)-β-D-glucopyranoside,

ethyl [(2R,3S,4S,5R,6S)-3,4,5-trihydroxy-6-[5-methyl-1-propan-2-yl-4-[(4-propan-2-yloxyphenyl)methyl]pyrazol-3-yl]oxy-oxan-2-yl]methyl carbonate
UNII:TR0QT6QSUL

CAS 442201-24-3

189075  BHV-091009  GSK-189075  GSK-189075A  KGT-1681 

BHV Pharma  Kissei (Originator)  , GlaxoSmithKline

Remogliflozin etabonate

Phase II

A SGLT-2 antagonist potentially for the treatment of type 2 diabetes, non-alcoholic steatohepatitis (NASH), obesity.

GSK-189075; GSK-189075A

  • Molecular FormulaC26H38N2O9
  • Average mass522.588

Remogliflozin etabonate (INN/USAN)[1] is a proposed drug of the gliflozin class for the treatment of non-alcoholic steatohepatitis (“NASH”) and type 2 diabetes. Remogliflozin is being developed by Avolynt, Inc.[2]

Remogliflozin etabonate, also known as GSK 189075A or GSK 189075, is a SGLT2 inhibitor under development for the treatment of type 2 diabetes. Remogliflozin etabonate is a pro-drug of remogliflozin. Remogliflozin inhibits the sodium-glucose transport proteins (SGLT), which are responsible for glucose reabsorption in the kidney. Blocking this transporter causes blood glucose to be eliminated through the urine. Remogliflozin is selective for SGLT2.

Remogliflozin etabonate also known as 5-methyl-4-[4-(1-methylethoxy)benzyl]-1-(1- methylethyl)-1H-pyrazol-3-yl 6-0-(ethoxycar onyl)-β-D-glucopyranoside of the following formula

(«):

Figure imgf000009_0001

(I)

Another nomenclature convention provides this molecule as 3-(6-0-ethoxycarbonyl-.beta.-D- glucopyranosyloxy)-4-[(4-isopropoxyphenyl)methyl]-1-isopropyl-5-methylpyrazole. Remogliflozin etabonate is also known as GSK 189075 or KGT-1681. Salts of compounds of formula (i) are useful as the active ingredient in the pharmaceutical presentation of the invention. Such salts may be as described in US Patent 7,084,123 issued August 1, 2006, herein incorporated by reference. Examples of such salts include acid addition salts with mineral acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, nitric acid, phosphoric acid and the like, acid addition salts with organic acids such as formic acid, acetic acid,

methanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, propionic acid, citric acid, succinic acid, tartaric acid, fumaric acid, butyric acid, oxalic acid, malonic acid, maieic acid, lactic acid, malic acid, carbonic acid, glutamic acid, aspartic acid, adipic acid, oleic acid, stearic acid and the like, and salts with inorganic bases such as a sodium salt, a potassium salt, a calcium salt, a magnesium salt and the like.

The compounds represented by the above formula (I) include their solvates with pharmaceutically acceptable solvents such as ethanol and water.

Remogliflozin etabonate may be prepared as described in US Patents 7,084,123 and 7,375,087, in particular Example 1 of US Patent 7,084,123, each herein incorporated by reference.

Remogliflozin etabonate is the pro-drug of remogliflozin (also known as GSK189074 or KGT-1650).

Remogliflozin etabonate has the potential to be used as monotherapy for the treatment of T2DM. To date, studies have assessed the efficacy, safety and tolerability up to 12 weeks, with varying efficacy so there is a need to characterize the profile of a number of selected formulated doses over a 12-week period. The study is designed with a placebo treatment arm to enable the profile of the drug to be further characterized and for maximal glycemic effect to be achieved. However to minimize the time which subjects may have sub-optimal glycemic control, the double blind study medication has been limited to 12 weeks duration. In addition, criteria have been included to allow the introduction of rescue therapy after 6 weeks for those subjects who have a high FP.

Diabetes mellitus is a diagnostic term for a group of disorders characterized by abnormal glucose homeostasis resulting in elevated blood sugar. There are many types of diabetes, but the two most common are type 1 (also referred to as insulin-dependent diabetes mellitus or IDDM) and type 2 (also referred to as non-insulin-dependent diabetes mellitus or NIDDM).

The etiology of the different types of diabetes is not the same; however, everyone with diabetes has two things in common: overproduction of glucose by the liver and little or no ability to move glucose out of the blood into the cells where it becomes the body’s primary fuel.

People who do not have diabetes rely on insulin, a hormone made in the pancreas, to move glucose from the blood into the cells of the body. However, people who have diabetes either do not produce insulin or can not efficiently use the insulin they produce; therefore, they can not move glucose into their cells. Glucose accumulates in the blood creating a condition called

hyperglycemia, and over time, can cause serious health problems. Diabetes is a syndrome with interrelated metabolic, vascular, and neuropathic components.

The metabolic syndrome, generally characterized by hyperglycemia, comprises alterations in carbohydrate, fat and protein metabolism caused by absent or markedly reduced insulin secretion and/or ineffective insulin action. The vascular syndrome consists of abnormalities in the blood vessels leading to cardiovascular, retinal and renal complications. Abnormalities in the peripheral and autonomic nervous systems are also part of the diabetic syndrome.

About 5% to 10% of the people who have diabetes have IDDM. These individuals do not produce insulin and therefore must inject insulin to keep their blood glucose levels normal. IDDM is characterized by low or undetectable levels of endogenous insulin production caused by destruction of the insulin-producing beta cells of the pancreas, the characteristic that most readily distinguishes IDDM from NIDDM. IDDM, once termed juvenile-onset diabetes, strikes young and older adults alike.

Approximately 90% to 95% of people with diabetes have NIDDM (type 2). NIDDM subjects produce insulin, but the cells in their bodies are insulin resistant: the cells do not respond properly to the hormone, so glucose accumulates in their blood. NIDDM is characterized by a relative disparity between endogenous insulin production and insulin requirements, leading to elevated blood glucose levels. In contrast to IDDM, there is always some endogenous insulin production in NIDDM; many NIDDM patients have normal or even elevated blood insulin levels, while other NIDDM patients have inadequate insulin production (Rotwein, R. et al. N. Engl. J. Med. 308, 65-71 (1983)). Most people diagnosed with NIDDM are age 30 or older, and half of all new cases are age 55 and older. Compared with whites and Asians, NIDDM is more common among Native Americans, African- Americans, Latinos, and Hispanics. In addition, the onset can be insidious or even clinically inapparent, making diagnosis difficult.

The primary pathogenic lesion on NIDDM has remained elusive. Many have suggested that primary insulin resistance of the peripheral tissues is the initial event. Genetic epidemiological studies have supported this view. Similarly, insulin secretion abnormalities have been argued as the primary defect in NIDDM. It is likely that both phenomena are important contributors to the disease process (Rimoin, D. L., et. al. Emery and Rimoin’s Principles and Practice of Medical Genetics 3ri Ed. 1: 1401-1402 (1996)).

Many people with NIDDM have sedentary lifestyles and are obese: they weigh approximately 20% more than the recommended weight for their height and build. Furthermore, obesity is characterized by hyperinsulinemia and insulin resistance, a feature shared with NIDDM, hypertension and atherosclerosis.

The patient with diabetes faces a 30% reduced lifespan. After age 45, people with diabetes are about three times more likely than people without diabetes to have significant heart disease and up to five times more likely to have a stroke. These findings emphasize the inter-relations between risks factors for NIDDM and coronary heart disease and the potential value of an integrated approach to the prevention of these conditions (Perry, I. J., et al., BMJ 310, 560-564 (1995)).

Diabetes has also been implicated in the development of kidney disease, eye diseases and nervous-system problems. Kidney disease, also called nephropathy, occurs when the kidney’s “filter mechanism” is damaged and protein leaks into urine in excessive amounts and eventually the kidney fails. Diabetes is also a leading cause of damage to the retina at the back of the eye and increases risk of cataracts and glaucoma. Finally, diabetes is associated with nerve damage, especially in the legs and feet, which interferes with the ability to sense pain and contributes to serious infections. Taken together, diabetes complications are one of the nation’s leading causes of death.

B. Obesity

Obesity and diabetes are among the most common human health problems in industrialized societies. In industrialized countries a third of the population is at least 20% overweight. In the United States, the percentage of obese people has increased from 25% at the end of the 1970’s, to 33% at the beginning the 1990’s. Obesity is one of the most important risk factors for NIDDM. Definitions of obesity differ, but in general, a subject weighing at least 20% more than the recommended weight for his/her height and build is considered obese. The risk of developing NIDDM is tripled in subjects 30% overweight, and three-quarters with NIDDM are overweight.

Obesity, which is the result of an imbalance between caloric intake and energy expenditure, is highly correlated with insulin resistance and diabetes in experimental animals and human.

However, the molecular mechanisms that are involved in obesity-diabetes syndromes are not clear. During early development of obesity, increased insulin secretion balances insulin resistance and protects patients from hyperglycemia (Le Stunff, et al. Diabetes 43, 696-702 (1989)). However, after several decades, β cell function deteriorates and non-insulin-dependent diabetes develops in about 20% of the obese population (Pederson, P. Diab. Metab. Rev. 5, 505-509 (1989)) and (Brancati, F. L., et al., Arch. Intern. Med. 159, 957-963 (1999)). Given its high prevalence in modern societies, obesity has thus become the leading risk factor for NIDDM (Hill, J. O., et al., Science 280, 1371-1374 (1998)). However, the factors which predispose a fraction of patients to alteration of insulin secretion in response to fat accumulation remain unknown.

Whether someone is classified as overweight or obese can be determined by a number of different methods, such as, on the basis of their body mass index (BMI) which is calculated by dividing body weight (kg) by height squared (m2). Thus, the units of BMI are kg/m2 and it is possible to calculate the BMI range associated with minimum mortality in each decade of life. Overweight is defined as a BMI in the range 25-30 kg/m2, and obesity as a BMI greater than 30 kg/m2(see table below). There are problems with this definition, such as, it does not take into account the proportion of body mass that is muscle in relation to fat (adipose tissue). To account for this, alternatively, obesity can be defined on the basis of body fat content: greater than 25% and 30% in males and females, respectively.

CLASSIFICATION OF WEIGHT BY BODY MASS INDEX (BMI)

Figure imgf000005_0001

As the BMI increases there is an increased risk of death from a variety of causes that is independent of other risk factors. The most common diseases associated with obesity are cardiovascular disease (particularly hypertension), diabetes (obesity aggravates the development of diabetes), gall bladder disease (particularly cancer) and diseases of reproduction. Research has shown that even a modest reduction in body weight can correspond to a significant reduction in the risk of developing coronary heart disease.

Obesity considerably increases the risk of developing cardiovascular diseases as well. Coronary insufficiency, atheromatous disease, and cardiac insufficiency are at the forefront of the cardiovascular complication induced by obesity. It is estimated that if the entire population had an ideal weight, the risk of coronary insufficiency would decrease by 25% and the risk of cardiac insufficiency and of cerebral vascular accidents by 35%. The incidence of coronary diseases is doubled in subjects less than 50 years of age who are 30% overweight.

C. Atherosclerosis

Atherosclerosis is a complex disease characterized by inflammation, lipid accumulation, cell death and fibrosis. Atherosclerosis is characterized by cholesterol deposition and monocyte infiltration into the subendothelial space, resulting in foam cell formation. Thrombosis subsequent to atherosclerosis leads to myocardial infarction and stroke. Atherosclerosis is the leading cause of mortality in many countries, including the United States. (See, e.g., Ruggeri, Nat Med (2002) 8: 1227-1234; Arehart et al, Circ Res, Circ. Res. (2008) 102:986-993.)

D. Osteoporosis

Osteoporosis is a disabling disease characterized by the loss of bone mass and microarchitectural deterioration of skeletal structure leading to compromised bone strength, which predisposes a patient to increased risk of fragility fractures. Osteoporosis affects more than 75 million people in Europe, Japan and the United States, and causes more than 2.3 million fractures in Europe and the United States alone. In the United States, osteoporosis affects at least 25% of all post-menopausal white women, and the proportion rises to 70% in women older than 80 years. One in three women older than 50 years will have an osteoporotic fracture that causes a considerable social and financial burden on society. The disease is not limited to women; older men also can be affected. By 2050, the worldwide incidence of hip fracture projected to increase by 310% in men and 240% in women. The combined lifetime risk for hip, forearm, and vertebral fractures presenting clinically is around 40%, equivalent to the risk for cardiovascular disease. Osteoporotic fractures therefore cause substantial mortality, morbidity, and economic cost. With an ageing population, the number of osteoporotic fractures and their costs will at least double in the next 50 years unless effective preventive strategies are developed. (See, e.g., Atik et al, Clin. Orthop. Relat. Res. (2006) 443: 19-24; Raisz, J. Clin. Invest. (2005) 115:3318-3325; and World Health Organization Technical Report Series 921 (2003), Prevention and

Management of Osteoporosis).

E. Inflammatory Bowel Disease (IBD)

Inflammatory bowel disease (IBD) is the general name for diseases that cause inflammation in the intestines and includes, e.g. Crohn’s disease, ulcerative colitis, and ulcerative proctitis. U.S. medical costs of inflammatory bowel disease for 1990 have been estimated to be $1.4 to $1.8 billion. Lost productivity has been estimated to have added an additional $0.4 to $0.8 billion, making the estimated cost of inflammatory bowel disease $1.8 to $2.6 billion. (See, e.g. , Pearson, Nursing Times (2004) 100:86-90; Hay et al., J. Clin.

Gastroenterol. (1992) 14:309-317; Keighley et al, Ailment Pharmacol. Ther. (2003) 18:66-70).

Enteritis refers to inflammation of the intestine, especially the small intestine, a general condition that can have any of numerous different causes. Enterocolitis refers to inflammation of the small intestine and colon.

Crohn’s disease (CD) is an inflammatory process that can affect any portion of the digestive tract, but is most commonly seen in the last part of the small intestine otherwise called the (terminal) ileum and cecum. Altogether this area is also known as the ileocecal region. Other cases may affect one or more of: the colon only, the small bowel only (duodenum, jejunum and/or ileum), the anus, stomach or esophagus. In contrast with ulcerative colitis, CD usually does not affect the rectum, but frequently affects the anus instead. The inflammation extends deep into the lining of the affected organ. The inflammation can cause pain and can make the intestines empty frequently, resulting in diarrhea. CD may also be called enteritis.

Granulomatous colitis is another name for CD that affects the colon. Ileitis is CD of the ileum which is the third part of the small intestine. Crohn’s colitis is CD affecting all or part of the colon.

Ulcerative colitis (UC) is an inflammatory disease of the large intestine, commonly called the colon. UC causes inflammation and ulceration of the inner lining of the colon and rectum. The inflammation of UC is usually most severe in the rectal area with severity diminishing (at a rate that varies from patient to patient) toward the cecum, where the large and small intestines join together. Inflammation of the rectum is called proctitis. Inflammation of the sigmoid colon (located just above the rectum) is called sigmoiditis. Inflammation involving the entire colon is termed pancolitis. The inflammation causes the colon to empty frequently resulting in diarrhea. As the lining of the colon is destroyed ulcers form releasing mucus, pus and blood. Ulcerative proctitis is a form of UC that affects only the rectum.

F. GPR119

GPR119 is a G protein-coupled receptor (GPR119; e.g. , human GPR119, GenBank® Accession No. AAP72125 and alleles thereof; e.g. , mouse GPR119, GenBank® Accession No. AY288423 and alleles thereof) and is selectively expressed on pancreatic beta cells. GPR119 activation leads to elevation of a level of intracellular cAMP, consistent with GPR119 being coupled to Gs. Agonists to GPR119 stimulate glucose-dependent insulin secretion in vitro and lower an elevated blood glucose level in vivo; see, e.g. , International Applications WO

04/065380 and WO 04/076413, and EP 1338651. In the literature, GPR119 has also been referred to as RUP3 (see, International Application WO 00/31258) and as Glucose-Dependent Insulinotropic Receptor GDIR (see, Jones, et. al. Expert Opin. Ther. Patents (2009), 19(10): 1339- 1359).

GPR119 agonists also stimulate the release of Glucose-dependent Insulinotropic Polypeptide (GIP), Glucagon-Like Peptide- 1 (GLP-1), and at least one other L-cell peptide, Peptide YY (PYY) (Jones, et. al. Expert Opin. Ther. Patents (2009), 19(10): 1339-1359); for specific references related to GPR119 agonists and the release of:

GIP, see Shah, Current Opinion in Drug Discovery & Development, (2009) 12:519-532; Jones, et al, Ann. Rep. Med. Chem. , (2009) 44: 149-170; WO 2007/120689; and WO 2007/120702;

GLP-1, see Shah, Current Opinion in Drug Discovery & Development, (2009) 12:519-532; Jones, et al, Ann. Rep. Med. Chem. , (2009) 44:149-170; Schwartz et. al., Cell Metabolism, 2010, 11 :445-447; and WO 2006/076231 ; and

PYY, see Schwartz et. al, Cell Metabolism, 2010, 11:445-447; and WO 2009/126245. As mentioned above, GPR119 agonists enhance incretin release and therefore can be used in treatment of disorders related to the incretins, such as, GIP, GLP-1, and PYY. However, a number of the incretins, such as, GIP and GLP-1, are substrates for the enzyme dipeptidyl peptidase-4 (DPP-IV). Jones and co-workers (Jones, et al, Ann. Rep. Med. Chem. , (2009) 44: 149- 170) have demonstrated that a combined administration of a GPR119 agonist, (2-Fluoro-4- methanesulfonyl-phenyl) – { 6-[4-(3 -isopropyl- [1,2,4] oxadiazol-5 -yl) -piperidin- 1 -yl] -5 -nitro- pyrimidin-4-yl} -amine (see, Compound B i l l in WO 2004/065380). and a DPP-IV inhibitor acutely increased plasma GLP-1 levels and improved glucose tolerance to a significantly greater degree than either agent alone.

G. Glucose-dependent Insulinotropic Polypeptide (GIP) Glucose-dependent insulinotropic polypeptide (GIP, also known as gastric inhibitory polypeptide) is a peptide incretin hormone of 42 amino acids that is released from duodenal endocrine K cells after meal ingestion. The amount of GIP released is largely dependent on the amount of glucose consumed. GIP has been shown to stimulate glucose-dependent insulin secretion in pancreatic beta cells. GIP mediates its actions through a specific G protein-coupled receptor, namely GIPR.

As GIP contains an alanine at position 2, it is an excellent substrate for DPP-IV, an enzyme regulating the degradation of GIP. Full-length GIP(l-42) is rapidly converted to bioinactive GIP(3-42) within minutes of secretion from the endocrine K cell. Inhibition of DPP- IV has been shown to augment GIP bioactivity. (See, e.g. , Drucker, Cell Metab (2006) 3: 153- 165; Mcintosh et al, Regul Pept (2005) 128: 159-165; Deacon, Regul Pept (2005) 128: 117-124; and Ahren et al., Endocrinology (2005) 146:2055-2059.). Analysis of full length bioactive GIP, for example in blood, can be carried out using N-terminal-specific assays (see, e.g. , Deacon et al., J Clin Endocrinol Metab (2000) 85:3575-3581).

Recently, GIP has been shown to promote bone formation. GIP has been shown to activate osteoblastic receptors, resulting in increases in collagen type I synthesis and alkaline phosphatase activity, both associated with bone formation. GIP has been shown to inhibit osteoclast activity and differentiation in vitro. GIP administration has been shown to prevent the bone loss due to ovariectomy. GIP receptor (GIPR) knockout mice evidence a decreased bone size, lower bone mass, altered bone microarchitecture and biochemical properties, and altered parameters for bone turnover, especially in bone formation. (See, e.g., Zhong et al., Am J Physiol Endocrinol Metab (2007) 292:E543-E548; Bollag et al., Endocrinology (2000) 141 : 1228-1235; Bollag et al., Mol Cell Endocrinol (2001) 177:35-41 ; Xie et al., Bone (2005) 37:759-769; and Tsukiyama et al., Mol Endocrinol (2006) 20: 1644-1651.)

The usefulness of GIP for maintaining or increasing bone density or formation has been acknowledged by the United States Patent and Trademark Office by issuance of United States Patent No. 6,410,508 for the treatment of reduced bone mineralization by administration of GIP peptide. However, current GIP peptide agonists suffer from a lack of oral bioavailability, negatively impacting patient compliance. An attractive alternative approach is to develop an orally active composition for increasing an endogenous level of GIP activity.

GPR119 agonists have been shown to stimulate the release of GIP; see Shah, Current Opinion in Drug Discovery & Development, (2009) 12:519-532; Jones, et al., Ann. Rep. Med. Chem., (2009) 44:149-170; WO 2007/120689; and WO 2007/120702.

H. Glucagon-Like Peptide-1 (GLP-1)

Glucagon-like peptide-1 (GLP-1) is an incretin hormone derived from the

posttranslational modification of proglucagon and secreted by gut endocrine cells. GLP-1 mediates its actions through a specific G protein-coupled receptor (GPCR), namely GLP-1R. GLP-1 is best characterized as a hormone that regulates glucose homeostasis. GLP-1 has been shown to stimulate glucose-dependent insulin secretion and to increase pancreatic beta cell mass. GLP-1 has also been shown to reduce the rate of gastric emptying and to promote satiety. The efficacy of GLP-1 peptide agonists in controlling blood glucose in type 2 diabetics has been demonstrated in several clinical studies [see, e.g. , Nauck et al., Drug News Perspect (2003) 16:413-422], as has its efficacy in reducing body mass [Zander et al., Lancet (2002) 359:824- 830].

GLP-1 receptor agonists are additionally useful in protecting against myocardial infarction and against cognitive and neurodegenerative disorders. GLP-1 has been shown to be cardioprotective in a rat model of myocardial infarction [Bose et al., Diabetes (2005) 54: 146- 151], and GLP-1 R has been shown in rodent models to be involved in learning andneuroprotection [During et al., Nat. Med. (2003) 9: 1173-1179; and Greig et al., Ann N Y Acad Sri (2004) 1035:290-315].

Certain disorders such as type 2 diabetes are characterized by a deficiency in GLP-1 [see, e.g., Nauck et al, Diabetes (2004) 53 Suppl 3:S190-196].

Current GLP-1 peptide agonists suffer from a lack of oral bioavailability, negatively impacting efficacy. Efforts to develop orally bioavailable non-peptidergic, small-molecule agonists of GLP-1 R have so far been unsuccessful (Mentlein, Expert Opin Investig Drugs (2005) 14:57-64). An attractive alternative approach is to develop an orally active composition for increasing an endogenous level of GLP-1 in the blood.

GPR119 agonists have been shown to stimulate the release of GLP-1, see Shah, Current Opinion in Drug Discovery & Development, (2009) 12:519-532; Jones, et al., Ann. Rep. Med. Chem., (2009) 44:149-170; Schwartz et. al., Cell Metabolism, 2010, 11:445-447; and WO2006/076231.

Clinical trials

Remogliflozin etabonate (RE) was shown to enhance urinary glucose excretion in rodents and humans. Early studies in diabetics improved plasma glucose levels.[3][4] Remogliflozin etabonate has been studied at doses up to 1000 mg.[5] A pair of 12-week phase 2b randomized clinical trials of diabetics published in 2015, found reductions in glycated hemoglobin and that it was generally well tolerated.[6]

Method of action

Remogliflozin etabonate is a pro-drug of remogliflozin. Remogliflozin inhibits the sodium-glucose transport proteins (SGLT), which are responsible for glucose reabsorption in the kidney. Blocking this transporter causes blood glucose to be eliminated through the urine.[7] Remogliflozin is selective for SGLT2.


Figure imgf000004_0002

Remogliflozin inhibits the sodium-glucose transport proteins, which are responsible for glucose reabsorption in the kidney. Blocking this transporter causes blood glucose to be eliminated through the urine.[3]

DPP IV inhibitors represent a novel class of agents that are being developed for the treatment or improvement in glycemic control in patients with type 2 diabetes. For example, DPP IV inhibitors and their uses are disclosed in WO 2002/068420, WO 2004/018467, WO 2004/018468, WO 2004/018469, WO 2004/041820, WO 2004/046148, WO 2005/051950, WO 2005/082906, WO 2005/063750, WO 2005/085246, WO 2006/027204, WO 2006/029769, WO2007/014886; WO 2004/050658, WO 2004/1 1 1051 , WO 2005/058901 , WO 2005/097798; WO 2006/068163, WO 2007/071738, WO 2008/017670; WO 2007/054201 or WO 2007/128761.

 

 

Chemical structures of remogliflozin etabonate (A), remogliflozin (B), sergliflozin (C), phlorizin (D), and T-1095 (E). Remogliflozin etabonate is metabolized to remogliflozin, its active form.

SYNTHESIS

JP 2011201871

CLIP, USE THE REF SHOWN BELOW

https://jstagebeta.jst.go.jp/article/cpb/64/7/64_c15-00982/_html

O-Glycosylation of 4-(Substituted benzyl)-1,2-dihydro-3H-pyrazol-3-one Derivatives with 2,3,4,6-Tetra-O-acyl-α-D-glucopyranosyl Bromide via N1-Acetylation of the Pyrazole Ring
Masahiro Kobayashi, Hidetoshi Isawa, Junichi Sonehara, Minoru Kubota, Tetsuji Ozawa
JOURNALS

Volume 64 (2016) Issue 7 Pages 1009-1018,

Some glucopyranosyloxypyrazole derivatives such as 1ad (Fig. 1) have been demonstrated to inhibit the low-affinity Na+-dependent glucose co-transporter SGLT2.13) Two types of SGLT are known, SGLT1 and SGLT2, both of which act as transmembrane glucose transporters. Although SGLT1 (high-affinity Na+-dependent glucose co-transporter) is expressed to some extent in the kidney and contributes to glucose reabsorption, it is mainly expressed in the small intestine, where it plays an important role in glucose absorption.4,5) SGLT2 is specifically expressed in the kidney and plays an important role in renal glucose reabsorption in the proximal tubule.6)Remogliflozin (1a), discovered at Kissei Pharmaceutical Co., Ltd., exhibits an inhibitory activity that is highly selective for SGLT2.7,8) Remogliflozin etabonate (1b), a prodrug of 1a, is metabolized to its active form 1a in the body, and may therefore be useful as a preventive or therapeutic agent for diseases attributable to hyperglycemia such as diabetes, complications related to diabetes, and obesity.9,10)

Remogliflozin etabonate (1b), a prodrug of 1a, is metabolized to its active form 1a in the body, and may therefore be useful as a preventive or therapeutic agent for diseases attributable to hyperglycemia such as diabetes, complications related to diabetes, and obesity.9,10)

Fig. 1. Glucopyranosyloxypyrazole Derivatives 1 Having an SGLT2 Inhibitory Activity

The synthetic strategy for 1b, given in Chart 1, shows that the 4-[(4-isopropoxyphenyl)methyl]-5-methyl-3-(2,3,4,6-tetra-O-acyl-β-D-glucopyranosyloxy)-1H-pyrazole derivative 2 is an important intermediate. O-Glycosylation of 4-[(4-isopropoxyphenyl)methyl]-5-methyl-1,2-dihydro-3H-pyrazol-3-one (3a) with a glycosyl donor (4 or 5) is therefore a key step in the production of 1b. Various O-glycosylation methods of 1,2-dihydro-3H-pyrazol-3-one derivatives 3have been reported, including the Koenigs–Knorr reaction, which employs a phase-transfer catalyst, and the Mitsunobu reaction.1,2,11–13) Although we also evaluated these methods for the preparation of 2, no successful results were obtained. Therefore, developing an efficient O-glycosylation method is strongly desired to establish scalable synthesis of 1b. We report here a convenient and practical method for the O-glycosylation of 3 with 2,3,4,6-tetra-O-acyl-α-D-glucopyranosyl bromide 5 via N1-acetylation of the pyrazole ring.

Chart 1. Synthetic Strategy for 1b

Remogliflozin etabonate (1b) was prepared by a four-step sequence starting from 2b, as shown in Chart 8. Introduction of an isopropyl group to 2b with 2-iodopropane in the presence of NaH in 1,3-dimethyl-2-imidazolidinone (DMI) provided 4-[(4-isopropoxyphenyl)methyl]-1-isopropyl-5-methyl-3-(2,3,4,6-tetra-O-acyl-β-D-glucopyranosyloxy)-1H-pyrazole derivative (15) in an 86% yield. The depivaloylation of 15 in the presence of sodium methoxide (MeONa) in MeOH provided Remogliflozin (1a) in a 99% yield. The reaction of 1a with ethyl chloroformate in the presence of 2,6-lutidine and pyridine in MeCN provided 16 as an ethanol solvate of 1b in a 72% yield. 16 was crystallized from a mixed solvent of methyl t-butyl ether (MTBE) and n-heptane to provide 1b in a 98% yield.

Chart 8. Preparation for 1b from 2bReagents: (a) 2-Iodopropane, NaH, DMI; (b) MeONa, MeOH; (c) Ethyl chloroformate, 2,3-lutidine, pyridine, MeCN; (d) MTBE, n-heptane.

In conclusion, an efficient and practical method for the synthesis of 2b, an important intermediate in the synthesis of 1b, was established. These results suggest that this O-glycosylation method could be applied in syntheses of additional glucopyranosyloxypyrazole derivatives exhibiting SGLT2 inhibitory activity such as 1c, d.

3-(β-D-Glucopyranosyloxy)-4-[(4-isopropoxyphenyl)methyl]-1-isopropyl-5-methyl-1H-pyrazole (1a)

A methanolic solution of 28% MeONa (1.93 g, 10 mmol) was added to a suspension of 15 (7.87 g, 10 mmol) in MeOH (75 mL) at room temperature. The mixture was then heated to 55°C and stirred for 3 h at this temperature. After cooling to 40°C, acetic acid (0.601 g, 10 mmol) was added dropwise to the reaction mixture. The reaction mixture was concentrated under reduced pressure to evaporate the methyl pivalate contained in the mixture. The residue was purified by silica gel chromatography (eluent dichloromethane–MeOH, 10 : 1) to provide 1a (4.45 g, 99% yield) as a pale yellowish oil.

[α]D20 −8.1 (c=1.0, DMSO).

IR (KBr) cm−1: 3407, 2975, 2931, 1506, 1466, 1384.

1H-NMR (CD3OD) δ: 1.26 (6H, d, J=6.0 Hz), 1.36 (6H, dd, J=3.8, 6.8 Hz), 2.09 (3H, s), 3.21–3.26 (1H, m), 3.33–3.43 (3H, m), 3.62–3.72 (3H, m), 3.77 (1H, dd, J=2.5, 12.1 Hz), 4.36–4.46 (1H, m), 4.46–4.55 (1H, m), 5.00–5.05 (1H, m), 6.76 (2H, d, J=8.7 Hz), 7.07 (2H, d, J=8.7 Hz).

13C-NMR (CD3OD) δ: 8.93 (q), 21.61 (q×2), 21.62 (q), 21.65 (q), 26.79 (t), 49.77 (d), 61.85 (t), 70.25 (d), 70.48 (d), 74.32 (d), 77.24 (d), 77.49 (d), 102.41 (d), 104.50 (s), 116.18 (d×2), 129.39 (d×2), 134.00 (s), 137.53 (s), 156.55 (s), 159.47 (s).

HR-MS (ESI) m/z: 451.2444 [M+H]+ (Calcd for C23H35N2O7: 451.2439).

5-Methyl-4-[4-(1-methylethoxy)benzyl]-1-(1-methylethyl)-1H-pyrazol-3-yl-6-O-(ethoxycarbonyl)-β-D-glucopyranoside (1b)

16 (1.50 g, 2.64 mmol) was dissolved in MTBE (10 mL) at 45°C The solution was concentrated under reduced pressure to evaporate EtOH. MTBE was added to the residue, and the weight was adjusted to 9.0 g. H2O (0.015 mL) and n-heptane (3.6 g) were added to the solution at 40°C and the solution was cooled to 25°C. The solution was seeded with 1a and stirred at 25°C for 3 h. The resulting slurry was warmed to 40°C, and then a mixed solvent of MTBE (0.44 g) and n-heptane (2.4 g) was added dropwise to the slurry while maintaining the temperature between 37 and 43°C. The slurry was stirred at 40°C for 1 h and for an additional 3 h at 10°C. The slurry was filtered and the wet cake washed successively with a mixed solvent of MTBE (1.5 g) and n-heptane (1.5 g) followed by n-heptane (3.0 g). The product was dried in vacuo at room temperature to give 1.35 g (98% yield) of

1a ( ERROR SHORLD BE 1b )as a white solid. mp 80–83°C. [α]D20 −19.3 (c=1.0, DMSO).

IR (KBr) cm−1: 3414, 2979, 1747, 1506, 1477, 1474, 1466, 1458, 1449, 1382, 1370, 1317.

1H-NMR (CD3OD) δ: 1.23 (3H, t, J=7.2 Hz), 1.26 (6H, d, J=6.1 Hz), 1.37 (6H, dd, J=2.3, 6.7 Hz), 2.07 (3H, s), 3.34–3.42 (4H, m), 3.61–3.69 (2H, m), 4.12 (2H, q, J=7.2 Hz), 4.21 (1H, dd, J=5.4, 11.5 Hz), 4.35 (1H, dd, J=1.7, 11.6 Hz), 4.35–4.45 (1H, m), 4.45–4.54 (1H, m), 5.04–5.06 (1H, m), 6.75 (2H, d, J=8.6 Hz), 7.06 (2H, d, J=8.6 Hz).

13C-NMR (CD3OD) δ: 9.70, 14.60, 22.43, 22.49, 22.54, 27.63, 50.53, 65.07, 67.67, 71.07, 71.21, 75.02, 75.56, 77.84, 103.25, 105.62, 116.98, 130.21, 134.81, 138.21, 156.65, 157.33, 159.99.

HR-MS (ESI) m/z: 523.2651 [M+H]+ (Calcd for C26H39N2O9: 523.2650).

5-Methyl-4-[4-(1-methylethoxy)benzyl]-1-(1-methylethyl)-1H-pyrazol-3-yl-6-O-(ethoxycarbonyl)-β-D-glucopyranoside Ethanolate (16)

A solution of ethyl chloroformate (522 mg, 4.81 mmol) in MeCN (1 mL) was added dropwise to a mixture of 1a(1.89 g, 4.19 mmol), 2,6-lutidine (672 mg, 6.28 mmol) and pyridine (13 mg, 0.17 mmol) in MeCN (10 mL) while maintaining the temperature between −3 and 3°C. The reaction mixture was stirred at 0°C for 2 h. After addition of glacial acetic acid (113 mg, 1.88 mmol), the reaction mixture was allowed to warm to room temperature. The reaction mixture was diluted with MTBE (10 mL) and 10% brine (5 mL), and then the layers were separated. The organic layer was washed twice with brine (5 mL), dried over anhydrous MgSO4 (2 g) and concentrated under reduced pressure. The residue was dissolved in EtOH (17 mL) and concentrated again under reduced pressure. EtOH was added to the residue, and the weight was adjusted to 9.3 g. To the EtOH solution, n-heptane (6 mL) was added and heated to 60°C to dissolve solids. The mixture was cooled to 45°C and stirred for 1 h at this temperature for an additional 1 h at 0–5°C. The slurry was filtered and the wet cake washed successively with a mixed solvent of EtOH (1.2 mL) and n-heptane (2.8 mL), which was cooled to 0°C, and then n-heptane (2.8 mL). The precipitate was dried in vacuo at room temperature to give 1.72 g (72% yield) of 16 as a white solid. mp 70–74°C. [α]D20 −17.7 (c=1.0, DMSO). IR (KBr) cm−1: 3353, 2980, 2926, 1753, 1731, 1508, 1477, 1467, 1449, 1386, 1371. 1H-NMR (CDCl3) δ: 1.23 (3H, t, J=7.0 Hz), 1.28 (3H, t, J=7.0 Hz), 1.30 (6H, d, J=6.0 Hz), 1.38 (6H, dd, J=2.3, 6.6 Hz), 2.06 (3H, s), 3.47–3.63 (6H, m), 3.71 (2H, q, J=7.0 Hz), 4.17 (2H, q, J=7.0 Hz), 4.24–4.31 (1H, m), 4.32–4.39 (2H, m), 4.43–4.52 (1H, m), 4.98 (1H, d, J=7.6 Hz), 6.77 (2H, d, J=8.6 Hz), 7.05 (2H, d, J=8.6 Hz). 13C-NMR (CDCl3) δ: 9.72, 14.21, 18.35, 22.09, 22.21, 22.25, 26.87, 49.44, 58.35, 64.23, 66.48, 69.49, 69.86, 73.65, 74.24, 76.44, 102.32, 104.67, 115.78, 129.10, 133.15, 136.55, 155.46, 155.96, 158.07. HR-MS (ESI) m/z: 523.2648 [M+H]+ (Calcd for C26H39N2O9: 523.2650).

1) Fujikura H., Fushimi N., Nishimura T., Nakabayashi T., Isaji M., PCT, WO 02/053573 (2002).

2) Ohsumi K., Umemura T., Matsueda H., Hatanaka T., Onuki A., Mae-zono K., Kageyama Y., Kondo N., PCT, WO 02/36602 (2002).

3) Kraemer G., Martin H., Adelgoss G., Dugi K., Duran A., Eickelmann P., Maier S., Pinnetti S., Ritter R., Schilcher G., Streicher R., Thomas L., PCT, WO 2007/080170 (2007).

4) Pajor A. M., Wright E. M., J. Biol. Chem., 267, 3557–3560 (1992).

5) Wright E. M., Am. J. Physiol. Renal Physiol., 280, F10–F18 (2001).

6) Kanai Y., Lee W. S., You G., Brown D., Hediger M. A., J. Clin. Invest., 93, 397–404 (1994).

7) Fujimori Y., Katsuno K., Nakashima I., Ishikawa-Takemura Y., Fujikura H., Isaji M., J. Pharmacol. Exp. Ther., 327, 268–276 (2008).

8) Mikhail N., Expert Opin. Investig. Drugs, 24, 1381–1387 (2015).

9) Isaji M., Curr. Opin. Investig. Drugs, 8, 285–292 (2007).

10) Katsuno K., Fujimori Y., Takemura Y., Hiratochi M., Itoh F., Komatsu Y., Fujikura H., Isaji M., J. Pharmacol. Exp. Ther., 320, 323–330 (2007).

11) Fujikura H., Nishimura T., Katsuno K., Hiratochi M., Iyobe A., Fujioka M., Isaji M., PCT, WO 01/16147 (2001).

12) Nishimura T., Fushimi N., Fujikura H., Katsuno K., Komatsu Y., Isaji M., PCT, WO 02/068439 (2002).

13) Washburn W. N., PCT, WO 03/020737 (2003).

14) Shng-Eun Y., Kyu Y. Y., Bull. Korean Chem. Soc., 10, 112 (1989).

15) Kees K. L., Fitzgerald J. J., Steiner K. E., Mattes J. F., Mihan B., Tosi T., Mondoro D., McCaleb M. L., J. Med. Chem., 39, 3920–3928 (1996).

16) Lemaire S., Houpis N. I., Xiao T., Li J., Digard E., Gozlan C., Liu R., Gavryushin A., Diène C., Wang Y., Farina V., Knochel P., Org. Lett., 14, 1480–1483 (2012).

17) Tokuoka Y., Kosobe T., Kawashima N., Kaji H., Nishino T., Ishizuka M., Kokai, Tokkyo Koho, JP2010053079 (2010).

PATENT

http://www.google.co.in/patents/WO2012040279A1?cl=en

In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from ethyl ((2R,35,45,5R,65)-3,4,5-trihydroxy-6-(4-(4-isopropoxybenzyl)- 1 -isopropyl-5-methyl- lH-pyrazol-3-yloxy)tetrahydro-2H-pyran-2-yl)methyl carbonate

(chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:

Figure imgf000076_0002

CLIP

Highly Selective Primary Alkoxycarboxylation and Esterification of Unprotected Pyranose Derivatives Mediated by Scandium(III) Triflate Catalysis
European Journal of Organic Chemistry (2012), 2012, (19), 3561-3565

http://onlinelibrary.wiley.com/doi/10.1002/ejoc.201200261/abstract

STR1

5-Methyl-1-(1-methylethyl)-4-({4-[(1-methylethyl)oxy]phenyl}methyl)-1H-pyrazol-3-yl 6-O- [(ethyloxy)carbonyl]--D-glucopyranoside (2): To a solution of 1 (1.5 Kg, 1.0 eq, 3.2 mol) in toluene (9.6 L) and ethanol (2.4 L) is added scandium triflate (2.4 g, 0.0015 eq) and diethylpyrocarbonate (597 g, 1.15 eq). The solution is heated to 45-55 ˚C for 1-6 hours before quenching with dilute acetic acid (4.5 L, 2.5 vol%). The mixture is cooled to 20 ˚C and the layers are allowed to separate. The bottom layer (aqueous) is discarded. The organic layer is washed again with dilute aq. acetic acid (4.5 L) and the aqueous layer discarded. The final organic layer is then concentrated under reduced pressure to about 2.25 volumes. MIBK (4.13 L), water (47 mL), and heptanes (12.8 L) are added and the desired compound is isolated by crystallization to afford a white solid. The cake is washed with 25% MIBK in heptanes and then dried under reduced pressure (30 ˚C) to afford the title compound 2 as a white solid (1.55 kg, 92% yield).

FTIR (ATR) Vmax cm-1: 3225, 2978, 1744, 1610, 1504, 1382, 1295, 1079, 1052; 1H NMR (500 MHz, DMSO-d6)  1.17 (t, J = 7.1 Hz, 3H), 1.22 (d, J = 6.1 Hz, 6H), 1.27 (dd, J1 = 6.7 Hz, J2 = 8.3 Hz, 6H), 2.06 (s, 3H), 3.12-3.29 (m, 3H), 3.38 (ddd, J1 = 1.8 Hz, J2 =6.1 Hz, J3 = 10.0 Hz, 1H), 3.51 (s, 2H), 4.08 (q, J = 7.1 Hz, 2H), 4.10 (dd, J1 = 6.1 Hz, J2 = 11.7 Hz, 1H), 4.29 (dd, J1 = 1.8 Hz, J2 = 11.7 Hz, 1H), 4.34 (sp, J = 6.4 Hz, 1H), 4.50 (sp, J = 6.0 Hz, 1H), 5.12 (d, J = 7.9 Hz, 1H), 5.14 (d, J = 5.3 Hz, 1H), 5.25 (d, J = 5.8 Hz, 1H), 5.32 (d, J = 5.4 Hz, 1H), 6.75 (d, J = 8.6 Hz, 2H), 7.08 (d, J = 8.6 Hz, 2H);

13C NMR (125 MHz, DMSO-d6)  9.1, 13.9, 21.8, 21.9, 22.2, 26.2, 48.3, 63.4, 66.6, 68.9, 69.5, 73.2, 73.8, 76.3, 100.6, 102.8, 115.3, 129.0, 133.2, 135.5, 154.4, 155.3, 157.8 ppm;

PATENT

US 7,084,123

http://www.google.co.in/patents/US7084123

EXAMPLE 1

3-(6-O-Ethoxycarbonyl-β-D-glucopyranosyloxy)-4-[(4-isopropoxyphenyl)methyl]-1-isopropyl-5-methylpyrazole

To a solution of 3-(β-D-glucopyranosyloxy)-4-[(4-isopropoxyphenyl)methyl]-1-isopropyl-5-methylpyrazole (0.10 g) in 2,4,6-trimethylpyridine (1 mL) was added ethyl chloroformate (0.072 g), and the mixture was stirred at room temperature overnight. To the reaction mixture were added citric acid monohydrate (3.3 g) and water, and the resulting mixture was purified by ODS solid phase extraction (washing solvent: distilled water, eluent: methanol). Further purification by column chromatography on silica gel (eluent: dichloromethane/methanol=1011) and recrystalization (recrystalization solvent: ethyl acetate/hexane=1/3) afforded 3-(6-O-ethoxycarbonyl-β-D-glucopyranosyloxy)-4-[(4-isopropoxyphenyl)methyl]-1-isopropyl-5-methylpyrazole (0.084 g).

1H-NMR (CD3OD) δ ppm: 1.23 (3H, t, J=7.0 Hz), 1.26 (6H, d, J=5.8 Hz), 1.30–1.40 (6H, m), 2.07 (3H, s), 3.25–3.45 (4H, m), 3.60–3.70 (2H, m), 4.12 (2H, q, J=7.0 Hz), 4.21 (1H, dd, J=5.4, 11.6 Hz), 4.34 (1H, dd, J=1.7, 11.6 Hz), 4.35–4.45 (1H, m), 4.45–4.55 (1H, m), 5.00–5.10 (1H, m), 6.70–6.80 (2H, m), 7.00–7.10 (2H, m)

CLIP

http://www.omicsonline.org/assessment-of-remogliflozin-etabonate-a-sodiumdependent-glucose-cotransporter-inhibitor-2155-6156.1000200.php?aid=7077

Image result for REMOGLIFLOZIN SYNTHESIS

References

  1.  Statement on a nonproprietory name adopted by the USAN council
  2. Yahoo Finance http://finance.yahoo.com/news/avolynt-announces-completion-phase-2b-143600142.html. Missing or empty |title=(help)
  3.  “Remogliflozin etabonate, a selective inhibitor of the sodium-glucose transporter 2, improves serum glucose profiles in type 1 diabetes.”. 35. Nov 2012: 2198–200. doi:10.2337/dc12-0508.PMID 23011728.
  4.  “Remogliflozin etabonate, a selective inhibitor of the sodium-dependent transporter 2 reduces serum glucose in type 2 diabetes mellitus patients.”. 14. Jan 2012: 15–22. doi:10.1111/j.1463-1326.2011.01462.x. PMID 21733056.
  5.  “Randomized trial showing efficacy and safety of twice-daily remogliflozin etabonate for the treatment of type 2 diabetes.”. Diabetes Obes Metab. 17: 94–7. Jan 2015.doi:10.1111/dom.12391. PMID 25223369.
  6.  “Randomized efficacy and safety trial of once-daily remogliflozin etabonate for the treatment of type 2 diabetes.”. 17. Jan 2015: 98–101. doi:10.1111/dom.12393. PMID 25238025.
  7.  Prous Science: Molecule of the Month November 2007 Archived January 6, 2008, at the Wayback Machine.

REFERENCES

1: Nakano S, Katsuno K, Isaji M, Nagasawa T, Buehrer B, Walker S, Wilkison WO, Cheatham B. Remogliflozin Etabonate Improves Fatty Liver Disease in Diet-Induced Obese Male Mice. J Clin Exp Hepatol. 2015 Sep;5(3):190-8. doi: 10.1016/j.jceh.2015.02.005. Epub 2015 Apr 28. PubMed PMID: 26628836; PubMed Central PMCID: PMC4632078.

2: Mikhail N. Remogliflozin etabonate: a novel SGLT2 inhibitor for treatment of diabetes mellitus. Expert Opin Investig Drugs. 2015;24(10):1381-7. doi: 10.1517/13543784.2015.1061501. Epub 2015 Aug 14. PubMed PMID: 26288025.

3: Mikhail N. Remogliflozin etabonate : a novel SGLT2 inhibitor for treatment of diabetes mellitus. Expert Opin Investig Drugs. 2015 Aug 14:1-7. [Epub ahead of print] PubMed PMID: 26271274.

4: O’Connor-Semmes R, Walker S, Kapur A, Hussey EK, Ye J, Wang-Smith L, Tao W, Dobbins RL, Cheatham B, Wilkison WO. Pharmacokinetics and Pharmacodynamics of the SGLT2 Inhibitor Remogliflozin Etabonate in Subjects with Mild and Moderate Renal Impairment. Drug Metab Dispos. 2015 Jul;43(7):1077-83. doi: 10.1124/dmd.114.062828. Epub 2015 May 1. PubMed PMID: 25934577.

5: Sykes AP, Kemp GL, Dobbins R, O’Connor-Semmes R, Almond SR, Wilkison WO, Walker S, Kler L. Randomized efficacy and safety trial of once-daily remogliflozin etabonate for the treatment of type 2 diabetes. Diabetes Obes Metab. 2015 Jan;17(1):98-101. doi: 10.1111/dom.12393. Epub 2014 Nov 3. PubMed PMID: 25238025.

6: Sykes AP, O’Connor-Semmes R, Dobbins R, Dorey DJ, Lorimer JD, Walker S, Wilkison WO, Kler L. Randomized trial showing efficacy and safety of twice-daily remogliflozin etabonate for the treatment of type 2 diabetes. Diabetes Obes Metab. 2015 Jan;17(1):94-7. doi: 10.1111/dom.12391. Epub 2014 Nov 3. PubMed PMID: 25223369.

7: Jackson VM, Price DA, Carpino PA. Investigational drugs in Phase II clinical trials for the treatment of obesity: implications for future development of novel therapies. Expert Opin Investig Drugs. 2014 Aug;23(8):1055-66. doi: 10.1517/13543784.2014.918952. Epub 2014 Jul 7. Review. PubMed PMID: 25000213.

8: Kapur A, O’Connor-Semmes R, Hussey EK, Dobbins RL, Tao W, Hompesch M, Smith GA, Polli JW, James CD Jr, Mikoshiba I, Nunez DJ. First human dose-escalation study with remogliflozin etabonate, a selective inhibitor of the sodium-glucose transporter 2 (SGLT2), in healthy subjects and in subjects with type 2 diabetes mellitus. BMC Pharmacol Toxicol. 2013 May 13;14:26. doi: 10.1186/2050-6511-14-26. PubMed PMID: 23668634; PubMed Central PMCID: PMC3700763.

9: Hussey EK, Kapur A, O’Connor-Semmes R, Tao W, Rafferty B, Polli JW, James CD Jr, Dobbins RL. Safety, pharmacokinetics and pharmacodynamics of remogliflozin etabonate, a novel SGLT2 inhibitor, and metformin when co-administered in subjects with type 2 diabetes mellitus. BMC Pharmacol Toxicol. 2013 Apr 30;14:25. doi: 10.1186/2050-6511-14-25. PubMed PMID: 23631443; PubMed Central PMCID: PMC3682882.

10: O’Connor-Semmes RL, Sandefer EP, Hussey EK, Tao W, Doll WJ, Page RC, Dobbins R. Regional gastrointestinal delivery of remogliflozin etabonate in humans. Biopharm Drug Dispos. 2013 Mar;34(2):79-86. doi: 10.1002/bdd.1824. Epub 2013 Jan 7. PubMed PMID: 23111980.

11: Mudaliar S, Armstrong DA, Mavian AA, O’Connor-Semmes R, Mydlow PK, Ye J, Hussey EK, Nunez DJ, Henry RR, Dobbins RL. Remogliflozin etabonate, a selective inhibitor of the sodium-glucose transporter 2, improves serum glucose profiles in type 1 diabetes. Diabetes Care. 2012 Nov;35(11):2198-200. doi: 10.2337/dc12-0508. Epub 2012 Sep 25. PubMed PMID: 23011728; PubMed Central PMCID: PMC3476920.

12: Sigafoos JF, Bowers GD, Castellino S, Culp AG, Wagner DS, Reese MJ, Humphreys JE, Hussey EK, O’Connor Semmes RL, Kapur A, Tao W, Dobbins RL, Polli JW. Assessment of the drug interaction risk for remogliflozin etabonate, a sodium-dependent glucose cotransporter-2 inhibitor: evidence from in vitro, human mass balance, and ketoconazole interaction studies. Drug Metab Dispos. 2012 Nov;40(11):2090-101. doi: 10.1124/dmd.112.047258. Epub 2012 Jul 30. PubMed PMID: 22851617.

13: Papazafiropoulou AK, Kardara MS, Pappas SI. Challenges for the treatment of diabetes mellitus. Recent Pat Endocr Metab Immune Drug Discov. 2011 Sep;5(3):203-9. Review. PubMed PMID: 21913881.

14: Dobbins RL, O’Connor-Semmes R, Kapur A, Kapitza C, Golor G, Mikoshiba I, Tao W, Hussey EK. Remogliflozin etabonate, a selective inhibitor of the sodium-dependent transporter 2 reduces serum glucose in type 2 diabetes mellitus patients. Diabetes Obes Metab. 2012 Jan;14(1):15-22. doi: 10.1111/j.1463-1326.2011.01462.x. Epub 2011 Oct 30. PubMed PMID: 21733056.

15: Fujimori Y, Katsuno K, Nakashima I, Ishikawa-Takemura Y, Fujikura H, Isaji M. Remogliflozin etabonate, in a novel category of selective low-affinity sodium glucose cotransporter (SGLT2) inhibitors, exhibits antidiabetic efficacy in rodent models. J Pharmacol Exp Ther. 2008 Oct;327(1):268-76. doi: 10.1124/jpet.108.140210. Epub 2008 Jun 26. PubMed PMID: 18583547.

Remogliflozin etabonate
Remogliflozin etabonate structure.svg
Systematic (IUPAC) name
5-methyl-4-[4-(1-methylethoxy)benzyl]-1-(1-methylethyl)-1H-pyrazol-3-yl 6-O-(ethoxycarbonyl)-β-D-glucopyranoside
Clinical data
Routes of
administration
Oral
Identifiers
CAS Number 442201-24-3 Yes
ATC code none
ChemSpider 8047110
UNII TR0QT6QSUL 
KEGG D10055 
ChEMBL CHEMBL494323 
Chemical data
Formula C26H38N2O9
Molar mass 522.586 g/mol

 

REMOGLIFLOZIN AND REMOGLIFLOZIN ETABONATE

Figure US20130096076A1-20130418-C00002
The compound is described for example in EP 1354888 A1.

 

Image result for REMOGLIFLOZIN NMR

 

Chemical Information

M.Wt Formula CAS No. Synonyms
450.53 C23H34N2O7 329045-45-6 Remogliflozin A; (2R,3S,4S,5R,6S)-2-(hydroxymethyl)-6-((4-(4-isopropoxybenzyl)-1-isopropyl-5-methyl-1H-pyrazol-3-yl)oxy)tetrahydro-2H-pyran-3,4,5-triolBMS-790052; EBP 883; BMS 790052

Structure Information of Remogliflozin

Smiles O[C@H]([C@H]([C@@H]([C@@H](CO)O1)O)O)[C@@H]1OC2=NN(C(C)C)C(C)=C2CC3=CC=C(OC(C)C)C=C3

 

///////////GSK-189075; GSK-189075A,  GSK189075A, GSK-189075A, GSK 189075,  GSK18907,  GSK-18907,  GSK 18907, Remogliflozin,  O-glycosylation , 1,2-dihydro-3H-pyrazol-3-ones derivative , N1-acetylation , glucopyranosyloxypyrazole derivative , Remogliflozin etabonate,  442201-24-3189075, BHV-091009, GSK-189075,  GSK-189075A,  KGT-1681, BHV Pharma  Kissei , GlaxoSmithKline,  SGLT-2 antagonist, type 2 diabetes, non-alcoholic steatohepatitis,  (NASH), obesity,

OC1C(COC(=O)OCC)OC(C(O)C1O)Oc(nn(C(C)C)c2C)c2Cc3ccc(cc3)OC(C)C

CC(C)Oc1ccc(cc1)Cc2c(nn(C(C)C)c2C)O[C@@H]3O[C@H](COC(=O)OCC)[C@@H](O)[C@H](O)[C@H]3O

Share
Dec 182013
 

TOFOGLIFLOZIN

托格列净

CSG-452, R-7201, RG-7201

CAS..1201913-82-7 monohydrate

903565-83-3 (anhydrous)

(1S,3′R,4′S,5′S,6′R)-6-(4-Ethylbenzyl)-6′-(hydroxymethyl)-3′,4′,5′,6′-tetrahydro-3H-spiro[2-benzofuran-1,2′-pyran]-3′,4′,5′-triol hydrate (1:1)

PMDA Pharmaceuticals and Medical Devices Agency, Japan Approved mar24, 2014

 

THERAPEUTIC CLAIM Treatment of diabetes mellitus
CHEMICAL NAMES
1. Spiro[isobenzofuran-1(3H),2′-[2H]pyran]-3′,4′,5′-triol, 6-[(4-ethylphenyl)methyl]-3′,4′,5′,6′-tetrahydro-6′-(hydroxymethyl)-, hydrate (1:1), (1S,3’R,4’S,5’S,6’R)-
2. (1S,3’R,4’S,5’S,6’R)-6-[(4-ethylphenyl)methyl]-6′-(hydroxymethyl)-3′,4′,5′,6′-tetrahydro-3H-spiro[2-benzofuran-1,2′-pyran]-3′,4′,5′-triol monohydrate
3. (1S,3’R,4’S,5’S,6’R)-6-[(4-ethylphenyl)methyl]-3′,4′,5′,6′-tetrahydro-6′-(hydroxymethyl)-
spiro[isobenzofuran-1(3H),2′-[2H]pyran]-3′,4′,5′-triol monohydrate

(3S,3’R,4’S,5’S,6’R)-5-[(4-ethylphenyl)methyl]-6′-(hydroxymethyl)spiro[1H-2-benzofuran-3,2′-oxane]-3′,4′,5′-triol;hydrate

MW404.5, MF C22H26O6

INNOVATOR  Chugai Pharmaceuticals

Sanofi, kowa

Deberza®………..KOWA/Apleway®……………SANOFI

CODE DESIGNATION CSG 452

Tofogliflozin (USAN, codenamed CSG452) is an experimental drug for the treatment of diabetes mellitus and is being developed byChugai Pharma in collaboration with Kowa and Sanofi.[1] It is an inhibitor of subtype 2 sodium-glucose transport protein (SGLT2), which is responsible for at least 90% of the glucose reabsorption in the kidney. As of September 2012, the drug is in Phase III clinical trials.[2][3]

Tofogliflozin is an SGLT-2 inhibitor first launched in 2014 in Japan by Sanofi and Kowa for the oral treatment of type II diabetes.

The product was discovered by Chugai and was licensed to Roche in 2007. In 2011, this license agreement was terminated. In 2012, the product was licensed to Kowa and Sanofi by Chugai Pharmaceutical in Japan for the treatment of diabetes type 2. In 2015, the license between Kowa and Chugai was expanded for developments and marketing of the agent in the U.S. and the E.U.

Chemistry

The active moiety or anhydrous form (ChemSpider ID: 28530778, CHEMBL2110731) has the chemical formula C22H26O6 and amolecular mass of 386.44 g/mol.

The United States Adopted Name tofogliflozin applies to the monohydrate, which is the form used as a drug.[4] The International Nonproprietary Name tofogliflozin applies to the anhydrous compound[5] and the drug form is referred to as tofogliflozin hydrate.

Several drugs are available for the treatment of type 2 diabetes mellitus (T2DM), but few patients achieve and maintain glycaemic control without weight gain and hypoglycaemias. Sodium glucose co-transporter 2 (SGLT-2) inhibitors are an emerging class of drugs with an original mechanism of action involving inhibition of renal glucose reabsorption. Two agents of this class, dapagliflozin and canagliflozin, have already been approved, although we need more data on cardiovascular outcomes along with bladder and breast cancer. Tofogliflozin is a further SGLT-2 inhibitor, which exhibits the highest selectivity for SGLT-2, the most potent antidiabetic action and a reduced risk of hypoglycaemia. Recently, a 52-week, multicentre, open-label, randomised controlled trial in Japanese T2DM patients has shown that tofogliflozin exhibits adequate safety and efficacy as monotherapy or as add-on treatment in patients suboptimally controlled with oral agents. Despite the very promising characteristics of this new drug, important questions remain to be answered, mainly additional data on safety outcomes and potential beneficial effects of tofogliflozin, for instance in prediabetes and diabetic nephropathy. Moreover, it would be welcome to examine the utility of its therapeutic use in combination with insulin and metformin.

Tofogliflozin has recently demonstrated safety and efficacy as monotherapy or add-on treatment . This is very important, granted our expectations of SGLT-2 inhibitors as useful alternative oral hypoglycaemic agents. Although important questions remain to be answered, the results of the new trial add to the importance of SGLT-2 inhibitors as a useful new class of oral hypoglycaemic agents.

 

CLIP

There are two scalable synthetic routes reported to prepare tofogliflozin.2 An efficient production synthesis of tofogliflozin hydrate from alcohol 2 was first described by Murakata et al. (Scheme 1, route 1).2a In 2016, Ohtake et al. reported an improved synthetic route, which achieved in just 7 linear steps (Scheme 1, route 2).2b They selected the optimal protecting groups for the purpose of chemoselective activation and crystalline purification, and obtained the pure tofogliflozin in a good overall yield. However, these methods suffer from several drawbacks. Firstly, some reagents, such as BH3 (Scheme 1, route 2) and 2-Methoxyproene (3, Scheme 1), are toxic or highly volatile. Meanwhile, the use of Palladium reagents may lead to an excess of residual heavy metal in the final product. Secondly, manufacturing costs in these methods are high due to the application of expensive raw materials and reagents. Last but not least, the key tactical stages that involve Br/Li exchange of aryl bromide followed by addition to gluconolactone 5 need the cryogenic conditions (< -60 oC), and this method is not suitable for industrial production. Herein, we report a newly developed synthetic method for tofogliflozin hydrate starting from readily available raw materials and affording good overall yield.

SCHEME 2 FOR

 

2. (a) Murakata, M.; Ikeda, T.; Kimura, N.; Kawase, A.; Nagase, M.; Yamamoto, K.; Takata, N.; Yoshizaki, S.; Takano, K. Crystal of spiroketal derivative, and process for production thereof. European Appl. EP 2308886 A1, April 13, 2011. (b) Ohtake, Y.; Emura, T.; Nishimoto, M.; Takano, K.; Yamamoto, K.; Tsuchiya, S.; Yeu, S.; Kito, Y.; Kimura, N.; Takeda, S.; Tsukazaki, M.; Murakata, M.; Sato, T. J. Org. Chem. 2016, 81, 2148.

 

 

Antidiabetic mechanism of SGLT-2 inhibitors.

CLIP

Ohtake, Y.; Sato, T.; Kobayashi, T.; Nishimoto, M.; Taka, N.; Takano, K.; Yamamoto, K.; Ohmori, M.; Yamaguchi, M.; Takami, K.; Yeu, S.-H.; Ahn, K.-H.; Matsuoka, H.; Morikawa, K.; Suzuki, M.; Hagita, H.; Ozawa, K.; Yamaguchi, K.; Kato, M.; Ikeda, S. J. Med. Chem. 2012, 55, 7828−7840

DOI: 10.1021/acs.joc.5b02734 J. Org. Chem. 2016, 81, 2148−2153

STR1

 

STR1

(1S,3′R,4′S,5′S,6′R)-6-[(4-Ethylphenyl)methyl]-6′-(hydroxymethyl)-3′,4′,5′,6′-tetrahydro-3H-spiro[2-benzofuran-1,2′- pyran]-3′,4′,5′-triol (1, tofogliflozin).

To a solution of 17b (89.9 g, 145 mmol) in DME (653 mL) and MeOH (73.0 mL), 2 N NaOH aq. solution (726 mL, 1.45 mol) was added dropwise for 1 h at waterbath temperature. After stirring at rt for 1 h, 2 N H2SO4 aq. solution (436 mL) was added slowly to the mixture. Water (700 mL) was added to the mixture, and the resultant mixture was extracted with AcOEt (500 mL × 2). The resultant organic layer was washed with brine (1.00 L) and then dried over anhydrous Na2SO4 (250 g). The mixture was concentrated in vacuo to obtain 1 (57.3 g, quant) as a colorless amorphous solid;

[α]D 26 +24.2° (c 1.02, MeOH);

1 H NMR (400 MHz, CD3OD) δ: 1.19 (3H, t, J = 7.6 Hz), 2.58 (2H, q, J = 7.6 Hz), 3.42−3.47 (1H, m), 3.63−3.67 (1H, m), 3.75−3.88 (4H, m), 3.95 (2H, s), 5.06 (1H, d, J = 12.5 Hz), 5.12 (1H, d, J = 12.5 Hz), 7.07−7.14 (4H, m), 7.17−7.23 (3H, m);

13C NMR (100 MHz, CD3OD) δ: 16.3, 29.4, 42.3, 62.8, 71.9, 73.4, 74.9, 76.2, 76.4, 111.6, 121.8, 123.6, 128.9, 129.9, 131.1, 139.7, 139.9, 140.2, 142.6, 143.2;

MS (ESI) m/z: 387 [M + H]+ ; HRMS (ESI) calcd for C22H27O6 [M + H]+ 387.1802, found 387.1801

DOI: 10.1021/acs.joc.5b02734 J. Org. Chem. 2016, 81, 2148−2153

Ohtake, Y.; Sato, T.; Kobayashi, T.; Nishimoto, M.; Taka, N.; Takano, K.; Yamamoto, K.; Ohmori, M.; Yamaguchi, M.; Takami, K.; Yeu, S.-H.; Ahn, K.-H.; Matsuoka, H.; Morikawa, K.; Suzuki, M.; Hagita, H.; Ozawa, K.; Yamaguchi, K.; Kato, M.; Ikeda, S. J. Med. Chem. 2012, 55, 7828−7840

 

 

str1

 

 

 

SGLT2 inhibitors inhibitors represent a novel class of agents that are being developed for the treatment or improvement in glycemic control in patients with type 2 diabetes. Glucopyranosyl-substituted benzene derivative are described in the prior art as SGLT2 inhibitors, for example in

WO 01/27128, WO 03/099836, WO 2005/092877, WO 2006/034489,

WO 2006/064033, WO 2006/117359, WO 2006/117360,

WO 2007/025943, WO 2007/028814, WO 2007/031548,

WO 2007/093610, WO 2007/128749, WO 2008/049923, WO 2008/055870, WO 2008/055940.

 

PATENTS

WO 2006080421

WO2009154276A1

WO 2011074675

WO 2012115249

 

Papers

Chinese Chemical Letters, 2013 ,  vol. 24,  2  pg. 131 – 133

Journal of Medicinal Chemistry, 2012 ,  vol. 55,  17  pg. 7828 – 7840

NMR

STR1

STR1
WO 2011074675

Figure JPOXMLDOC01-appb-C000048

1 H-NMR (CD 3 OD) δ: 1.19 (3H, t, J = 7.5Hz), 2.59 (2H, q, J = 7.5Hz) ,3.42-3 .46 (1H , m), 3.65 (1H, dd, J = 5.5,12.0 Hz) ,3.74-3 .82 (4H, m), 3.96 (2H, s), 5.07 (1H , d, J = 12.8Hz), 5.13 (1H, d, J = 12.8Hz) ,7.08-7 .12 (4H, m) ,7.18-7 .23 (3H, m) .
MS (ESI +): 387 [M +1] +.

 

 

Second set

http://pubs.acs.org/doi/full/10.1021/jm300884k

J. Med. Chem., 2012, 55 (17), pp 7828–7840

DOI: 10.1021/jm300884k

1H NMR (400 MHz, CD3OD) δ: 1.20 (3H, t, J = 7.6 Hz), 2.58 (2H, q, J = 7.6 Hz), 3.42–3.47 (1H, m), 3.63–3.67 (1H, m), 3.75–3.88 (4H, m), 3.95 (2H, s), 5.06 (1H, d, J = 12.3 Hz), 5.12 (1H, d, J = 12.5 Hz), 7.07–7.14 (4H, m), 7.17–7.23 (3H, m).

13C NMR (100 MHz, CD3OD) δ: 16.3, 29.4, 42.3, 62.8, 71.9, 73.4, 74.9, 76.2, 76.4, 111.6, 121.8, 123.6, 128.9, 129.9, 131.1, 139.7, 139.9, 140.2, 142.6, 143.2.

MS (ESI): 387 [M + H]+. HRMS (ESI), m/z calcd for C22H27O6 [M + H]+ 387.1802, found 387.1801.

THIRD SET

(1S,3′R,4′S,5′S,6′R)-6-[(4-Ethylphenyl)methyl]-6′-(hydroxymethyl)-3′,4′,5′,6′-tetrahydro-3H-spiro[2-benzofuran-1,2′- pyran]-3′,4′,5′-triol (1, tofogliflozin).

To a solution of 17b (89.9 g, 145 mmol) in DME (653 mL) and MeOH (73.0 mL), 2 N NaOH aq. solution (726 mL, 1.45 mol) was added dropwise for 1 h at waterbath temperature. After stirring at rt for 1 h, 2 N H2SO4 aq. solution (436 mL) was added slowly to the mixture. Water (700 mL) was added to the mixture, and the resultant mixture was extracted with AcOEt (500 mL × 2). The resultant organic layer was washed with brine (1.00 L) and then dried over anhydrous Na2SO4 (250 g). The mixture was concentrated in vacuo to obtain 1 (57.3 g, quant) as a colorless amorphous solid;

[α]D 26 +24.2° (c 1.02, MeOH);

1 H NMR (400 MHz, CD3OD) δ: 1.19 (3H, t, J = 7.6 Hz), 2.58 (2H, q, J = 7.6 Hz), 3.42−3.47 (1H, m), 3.63−3.67 (1H, m), 3.75−3.88 (4H, m), 3.95 (2H, s), 5.06 (1H, d, J = 12.5 Hz), 5.12 (1H, d, J = 12.5 Hz), 7.07−7.14 (4H, m), 7.17−7.23 (3H, m);

13C NMR (100 MHz, CD3OD) δ: 16.3, 29.4, 42.3, 62.8, 71.9, 73.4, 74.9, 76.2, 76.4, 111.6, 121.8, 123.6, 128.9, 129.9, 131.1, 139.7, 139.9, 140.2, 142.6, 143.2;

MS (ESI) m/z: 387 [M + H]+ ; HRMS (ESI) calcd for C22H27O6 [M + H]+ 387.1802, found 387.1801

DOI: 10.1021/acs.joc.5b02734 J. Org. Chem. 2016, 81, 2148−2153

Ohtake, Y.; Sato, T.; Kobayashi, T.; Nishimoto, M.; Taka, N.; Takano, K.; Yamamoto, K.; Ohmori, M.; Yamaguchi, M.; Takami, K.; Yeu, S.-H.; Ahn, K.-H.; Matsuoka, H.; Morikawa, K.; Suzuki, M.; Hagita, H.; Ozawa, K.; Yamaguchi, K.; Kato, M.; Ikeda, S. J. Med. Chem. 2012, 55, 7828−7840

 

PATENT

Prepn

WO 2011074675

[Example 1] (1S, 3’R, 4’S, 5’S, 6’R) -6 – [(4 – ethyl-phenyl) methyl] -3 ‘, 4’, 5 ‘, 6′-tetrahydro- -6′-(hydroxymethyl) – spiro [isobenzofuran -1 (3H), 2’-[2H] pyran] -3 ‘, 4′, one of the preparation step [compound of formula (IX)] 5’-triol Preparation of methanol (2 – hydroxymethyl-phenyl – bromo-4)

 

Figure JPOXMLDOC01-appb-C000042

 

To the mixing solution (1mol / L, 78.9kg, 88.4mol) of borane-tetrahydrofuran complex in tetrahydrofuran (6.34kg, 61.0mol) and, trimethoxyborane, two tetrahydrofuran (33.1kg) in – bromoterephthalic was added at below 30 ℃ solution (7.5kg, 30.6mol) of the acid, and the mixture was stirred for 1 hour at 25 ℃. Then cooled to 19 ℃ The reaction mixture was stirred for 30 minutes and added a mixed solution of tetrahydrofuran and methanol (3.0kg) of (5.6kg). In addition to methanol (15.0kg) in the mixture was kept for a while.

Again, to the mixing solution (1mol / L, 78.9kg, 88.4mol) of borane-tetrahydrofuran complex in tetrahydrofuran (6.34kg, 61.0mol) and, trimethoxyborane, two tetrahydrofuran (33.0kg) in – was added at below 30 ℃ solution (7.5kg, 30.6mol) of bromo terephthalic acid, and the reaction was carried out for 1 hour at 25 ℃. Then cooled to 18 ℃ The reaction mixture was stirred for 30 minutes and added a mixed solution of tetrahydrofuran and methanol (3.0kg) of (5.6kg). After addition of methanol (15.0kg) in the mixture is combined with the reaction mixture obtained in the previous reaction, and then the solvent was distilled off under reduced pressure. After addition of methanol (36kg) residue was obtained, and the solvent was evaporated under reduced pressure. Furthermore, (54 ℃ dissolved upon confirmation) which was dissolved by warming was added to methanol (36kg) to the residue. After cooling to room temperature the solution was stirred for 30 minutes added water (60kg). After addition of water (165kg) In addition to this mixture was cooled to 0 ℃, and the mixture was stirred for one hour. Centrifuge the obtained crystals were washed twice with water (45kg), and dried for 2 hours under reduced pressure to give (11.8kg, 54.4mol, 89% yield) of the title compound.

1 H-NMR (DMSO-d 6) δ: 4.49 (4H, t, J = 5.8Hz), 5.27 (1H, t, J = 5.8Hz), 5.38 (1H, t, J = 5.8Hz), 7.31 (1H, d, J = 7.5Hz), 7.47 (1H, d, J = 7.5Hz), 7.50 (1H, s).

Preparation of benzene (ethoxy methyl – methyl – – methoxy-1 1) – bromo-1 ,4 – 2:2 process bis

 

Figure JPOXMLDOC01-appb-C000043

 

(- Bromo-4 – 2-hydroxyethyl methyl phenyl) in tetrahydrofuran (57kg) in the solution (8.0kg, 36.9mol) of methanol, I added (185.12g, 0.74mol) of pyridinium p-toluenesulfonate. After cooling to -15 ℃ below the mixture, 2 – was added at -15 ℃ or less (7.70kg, 106.8mol) methoxy propene, and the mixture was stirred 1 h at -15 ~ 0 ℃. Was added aqueous potassium carbonate (25 wt%, 40kg) and the reaction mixture was warmed to room temperature and separate the organic layer was added toluene (35kg). After washing with water (40kg) The organic layer was evaporated under reduced pressure. Was dissolved in toluene (28kg) and the residue obtained was obtained as a toluene solution of the title compound.

1 H-NMR (CDCl 3) δ: 1.42 (6H, s), 1.45 (6H, s), 3.24 (3H, s), 3.25 (3H, s), 4.45 ( 2H, s), 4.53 (2H, s), 7.28 (1H, dd, J = 1.5,8.0 Hz), 7.50 (1H, d, J = 8.0Hz), 7. 54 (1H, d, J = 1.5Hz).
MS (ESI +): 362 [M +2] +.

Preparation of on – (3R, 4S, 5R, 6R) -3,4,5 – tris (trimethylsilyloxy)-6 – trimethylsilyloxy methyl – tetrahydropyran-2: Step 3

 

Figure JPOXMLDOC01-appb-C000044

 

Glucono -1,5 – – D-(+) in tetrahydrofuran (70kg) in the solution (35.8kg, 353.9mol) of N-methylmorpholine (7.88kg, 44.23mol) and lactone, chlorotrimethylsilane ( was added at 40 ℃ less 29.1kg, and 267.9mol), and the mixture was stirred for 2 hours at 30 ~ 40 ℃ resulting mixture. Was cooled to 0 ℃ the reaction mixture was added toluene (34kg) water (39kg), and the organic layer was separated. Twice sodium dihydrogen phosphate aqueous solution (5 wt%, 39.56kg) in, washed once with water (39kg) the organic layer the solvent was evaporated under reduced pressure. Was dissolved in toluene (34.6kg) and the residue obtained was obtained as a toluene solution of the title compound.

1 H-NMR (CDCl 3) δ: 0.13 (9H, s), 0.17 (9H, s), 0.18 (9H, s), 0.20 (9H, s), 3.74- 3.83 (3H, m), 3.90 (1H, t, J = 8.0Hz), 3.99 (1H, d, J = 8.0Hz), 4.17 (1H, dt, J = 2 .5,8.0 Hz).

Step 4: (1S, 3’R, 4’S, 5’S, 6’R) -3 ‘, 4’, 5 ‘, 6′-tetrahydro -6,6′ – bis (hydroxymethyl) – spiro [ (3H), 2’-[2H] pyran] -3 ‘, 4′, 5’-Preparation of triol isobenzofuran-1

 

Figure JPOXMLDOC01-appb-C000045

 

(Methyl – – – methoxy 1-ethoxy-methyl) – bromo-1 ,4 – 2 prepared in step 2 bis cooled to below -10 ℃ toluene solution of benzene, hexane solution to (15 wt% n-butyl lithium , was added at below 0 ℃ 18.2kg, and 42.61mol), and the mixture was stirred 1.5 h at 5 ℃ resulting mixture. (10.5kg, 40.7mol), was added tetrahydrofuran (33.4kg) then magnesium bromide diethyl ether complex in the mixture, and the mixture was stirred for 1 hour at 25 ℃. Was added at below -10 ℃ toluene solution of the on – tris (trimethylsilyloxy) -6 – – 3,4,5 cooled to -15 ℃ below the mixture prepared in step 3 trimethylsilyloxy methyl – tetrahydropyran-2 was. After stirring 0.5 h at -15 ℃ or less, poured into 20% aqueous ammonium chloride solution to (80kg) of this solution, and the organic layer was separated. After washing with water (80kg) and the organic layer obtained, and the solvent was evaporated under reduced pressure. I was dissolved in methanol (43kg) residue was obtained. Was stirred for 1 hour at 20 ℃ was added (1.4kg, 7.4mol) and p-toluenesulfonic acid monohydrate in the mixture. Thereafter, it was stirred for another hour and cooled to 0 ℃, centrifuged crystals obtained was washed with methanol (25kg), and dried for 8 hours at reduced pressure under 40 ℃, (5.47kg, yield the title compound I got 50%) rate.

1 H-NMR (DMSO-d 6) δ :3.20-3 .25 (1H, m) ,3.41-3 .45 (1H, m) ,3.51-3 .62 (4H, m) , 4.39 (1H, t, J = 6.0Hz) ,4.52-4 .54 (3H, m), 4.86 (1H, d, J = 4.5Hz), 4.93 (1H, d, J = 5.5Hz), 4.99 (1H, d, J = 12.5Hz), 5.03 (1H, d, J = 12.5Hz), 5.23 (1H, t, J = 5 .8 Hz) ,7.24-7 .25 (2H, m), 7.29 (1H, dd, J = 1.5,8.0 Hz).

Step 5: (1S, 3’R, 4’S, 5’S, 6’R) -6 – [(methoxycarbonyl) methyl] -3 ‘, 4’, 5 ‘, 6′-tetrahydro-3’ , 4 ‘, 5′-tris (methoxycarbonyl) oxy-6′-[(methoxycarbonyl) methyl] – Preparation of [(3H), 2’-[2H] pyran isobenzofuran] spiro

 

Figure JPOXMLDOC01-appb-C000046

 

(1S, 3’R, 4’S, 5’S, 6’R) – tetrahydro -6,6 ‘- bis (hydroxymethyl) – spiro [isobenzofuran -1 (3H), 2’-[2H] pyran ] -3 ‘, 4′, 5’-triol 4 (5.3kg, 17.8mol) and – dissolved in acetonitrile (35kg) (13.7kg, 112.1mol) a chloroformate, in the solution of dimethylaminopyridine I was added at 12 ℃ or less (10.01kg, 105.9mol) methyl. Heated to 20 ℃, After stirring for 1 h, was added ethyl acetate (40kg) and water (45kg), and the organic layer was separated and the mixture. Once (45.4kg) aqueous solution consisting of (9.01kg) sodium chloride and potassium hydrogen sulfate (1.35kg), sodium chloride aqueous solution (weight 10%, 44.5kg), sodium chloride aqueous solution (the organic layer was washed successively 20% by weight, in 45.0kg), and the solvent was evaporated under reduced pressure. Was dissolved in ethylene glycol dimethyl ether (18kg) and the residue obtained was then evaporated under reduced pressure. Was dissolved in ethylene glycol dimethyl ether (13.2kg) again and the residue obtained was obtained as ethylene glycol dimethyl ether solution of the title compound. I was used as it was in the six step.

1 H-NMR (CDCl 3) δ: 3.54 (3H, s), 3.77 (6H, s), 3.811 (3H, s), 3.812 (3H, s), 4.23 ( 1H, dd, J = 2.8,11.9 Hz), 4.32 (1H, dd, J = 4.0,11.9 Hz) ,4.36-4 .40 (1H, m), 5.11 -5.24 (5H, m), 5.41 (1H, d, J = 9.8Hz), 5.51 (1H, t, J = 9.8Hz), 7.25 (1H, d, J = 7.5Hz), 7.42 (1H, d, J = 7.5Hz), 7.44 (1H, s).
MS (ESI +): 589 [M +1] +, 606 [M +18] +.

Step 6: (1S, 3’R, 4’S, 5’S, 6’R) -6 – [(4 – ethyl-phenyl) methyl] -3 ‘, 4’, 5 ‘, 6’-tetrahydro-3 ‘4’, 5′-tris (methoxycarbonyl) oxy-6′-[(methoxycarbonyl) methyl] – Preparation of [(3H), 2′-[2H] pyran isobenzofuran] spiro

 

Figure JPOXMLDOC01-appb-C000047

 

[(Methoxycarbonyl) methyl] -3 ‘, 4’, 5 ‘, 6’-tetrahydro – (1S, 3’R, 4’S, 5’S, 6’R) -6 which had been prepared in Step 5 – 3 ‘, 4′, 5′-tris (methoxycarbonyl) oxy-6′-[(methoxycarbonyl) methyl] – spiro [isobenzofuran -1 (3H), 2’-[2H] pyran] Ethylene glycol dimethyl ether in solution, 2 – (2.46kg, 17.8mol), 4 butanol (25kg), anhydrous potassium carbonate – – methyl-2 were sequentially added (3.73kg, 24.9mol) ethyl phenyl boronic acid, in the reaction vessel was replaced with argon atmosphere, was bubbled with argon mixture. To the mixture – after the addition (0.72kg, 0.88mol) and palladium (II) chloride dichloromethane adduct [1,1 ‘-bis (diphenylphosphino) ferrocene], it was replaced with argon again inside of the vessel, one at 80 ℃ I was stirring time. After cooling, I added sequentially (0.859kg, 5.3mol) of ethylene glycol dimethyl ether (9.85kg), ethyl acetate (19kg), N-acetyl-L-cysteine ​​in the mixture. After stirring for 2.5 h the mixture was filtered and added Celite (5.22kg), and washed with ethyl acetate (78kg) and the filter residue. The combined washings and filtrate, and the solvent is evaporated off under reduced pressure, and in addition (0.58kg, 3.6mol) and ethanol (74kg), N-acetyl-L-cysteine ​​residue was obtained, which is heated to 70 ℃ or I was dissolved residue is then. After addition of water (9.4kg) in the solution, cooled to 60 ℃, and the mixture was stirred for 1 h. After confirming solid precipitated, cooled to 0 ℃ from 60 ℃ over 2.5 hours or more The mixture was stirred for 1 hour or more at 5 ℃ less. Centrifuge the resulting solid was washed twice with a mixture of water (35kg) and ethanol (55kg). Was dissolved at 70 ℃ ethanol (77kg) again, wet powder was obtained (10.21kg), cooled to 60 ℃ added water (9.7kg), and the mixture was stirred for 1 h. After confirming solid precipitated, cooled to 0 ℃ from 60 ℃ over 2.5 hours or more, and the mixture was stirred for 1 hour or more at 5 ℃ less. (9.45kg, dry powder rate 8.47kg, 13.7mol which was centrifuged obtained crystals were washed with a mixture of water (32kg) and ethanol (51kg), was obtained as a moist powder the title compound, 77% overall yield from the previous step).

1 H-NMR (CDCl 3) δ: 1.20 (3H, t, J = 7.5Hz), 2.60 (2H, q, J = 7.5Hz), 3.50 (3H, s), 3 .76 (3H, s), 3.77 (3H, s), 3.81 (3H, s), 3.96 (2H, s), 4.23 (1H, dd, J = 2.8,11 .9 Hz), 4.33 (1H, dd, J = 4.5,11.9 Hz) ,4.36-4 .40 (1H, m) ,5.11-5 .20 (3H, m), 5 .41 (1H, d, J = 10.0Hz), 5.51 (1H, t, J = 10.0Hz) ,7.07-7 .11 (4H, m), 7.14 (1H, d, J = 7.8Hz), 7.19 (1H, dd, J = 1.5,7.8 Hz), 7.31 (1H, d, J = 1.5Hz).
MS (ESI +): 619 [M +1] +, 636 [M +18] +.

Step 7: (1S, 3’R, 4’S, 5’S, 6’R) -6 – [(4 – ethyl-phenyl) methyl] -3 ‘, 4’, 5 ‘, 6’-tetrahydro-6 , 4 ‘, 5′-Preparation of triol’ – -3 [(3H), 2′-[2H] pyran isobenzofuran] spiro – (hydroxymethyl) ‘

 

Figure JPOXMLDOC01-appb-C000048

 

(1S, 3’R, 4’S, 5’S, 6’R) -6 – [(4 – ethyl-phenyl) methyl] -3 ‘, 4’, 5 ‘, 6′-tetrahydro-3’, 4 ‘, 5′-tris (methoxycarbonyl) oxy-6′-[(methoxycarbonyl) methyl] – wet powder spiro [(3H), 2’-[2H] pyran isobenzofuran -1] (8.92kg, In addition at 20 ℃ (4mol / L, 30.02kg, the 104.2mol) aqueous solution of sodium hydroxide, 1 hour the reaction mixture to a solution of (28kg) ethylene glycol dimethyl ether dry end conversion 8.00kg, of 12.9mol) the mixture was stirred. And the organic layer was separated by addition of water (8.0kg) in the mixture. The ethyl acetate aqueous sodium chloride solution (25 wt%, 40kg) and a (36kg) in the organic layer and the aqueous layer was removed after washing. The washed again aqueous sodium chloride solution (25 wt%, 40kg) in the organic layer was evaporated under reduced pressure. Were added and acetone (32.0kg) water (0.8kg) residue was obtained. After the solvent was evaporated under reduced pressure, dissolved in acetone (11.7kg) in water (15.8kg) and the residue obtained was cooled to below 5 ℃. Was added below 10 ℃ water (64kg) to the mixture, and the mixture was stirred for 1 hour at below 10 ℃. Centrifuge the resulting crystals were washed with a mixture of water (8.0kg) and (1.3kg) acetone. For 8 hours through-flow drying 13 ~ 16 ℃ temperature ventilation, under the conditions of 24-33% relative humidity the wet powder, the monohydrate crystal (3.94kg, 9.7mol, 75% yield) of the title compound I was obtained as: (4.502 wt% water content).

Method of measuring the amount of water:
Analysis: coulometric KF titration analyzer: trace moisture measurement device manufactured by Mitsubishi Chemical Corporation Model KF-100
Anolyte: Aqua micron AX (manufactured by Mitsubishi Chemical Corporation)
Catholyte: Aqua micron CXU (manufactured by Mitsubishi Chemical Corporation)

1 H-NMR (CD 3 OD) δ: 1.19 (3H, t, J = 7.5Hz), 2.59 (2H, q, J = 7.5Hz) ,3.42-3 .46 (1H , m), 3.65 (1H, dd, J = 5.5,12.0 Hz) ,3.74-3 .82 (4H, m), 3.96 (2H, s), 5.07 (1H , d, J = 12.8Hz), 5.13 (1H, d, J = 12.8Hz) ,7.08-7 .12 (4H, m) ,7.18-7 .23 (3H, m) .
MS (ESI +): 387 [M +1] +.

 

PATENT

US20110306778

Example 1 Synthesis of 1,1-anhydro-1-C-[5-(4-ethylphenyl)methyl-2-(hydroxymethyl)phenyl]-β-D-glucopyranose Step 1: Synthesis of 3,4,5-tris(trimethylsilyloxy)-6-trimethylsilyloxymethyl-tetrahydropyran-2-one

 

Figure US20110306778A1-20111215-C00017

 

To a solution of D-(+)-glucono-1,5-lactone (7.88 kg) and N-methylmorpholine (35.8 kg) in tetrahydrofuran (70 kg) was added trimethylsilyl chloride (29.1 kg) at 40° C. or below, and then the mixture was stirred at a temperature from 30° C. to 40° C. for 2 hours. After the mixture was cooled to 0° C., toluene (34 kg) and water (39 kg) were added thereto. The organic layer was separated and washed with an aqueous solution of 5% sodium dihydrogen phosphate (39.56 kg×2) and water (39 kg×1). The solvent was evaporated under reduced pressure to give the titled compound as an oil. The product was used in the next step without further purification.

1H-NMR (CDCl3) δ: 0.13 (9H, s), 0.17 (9H, s), 0.18 (9H, s), 0.20 (9H, s), 3.74-3.83 (3H, m), 3.90 (1H, t, J=8.0 Hz), 3.99 (1H, d, J=8.0 Hz), 4.17 (1H, dt, J=2.5, 8.0 Hz).

Step 2: Synthesis of 2,4-dibromo-1-(1-methoxy-1-methylethoxymethyl)benzene

 

Figure US20110306778A1-20111215-C00018

 

Under a nitrogen atmosphere, to a solution of 2,4-dibromobenzyl alcohol (40 g, 0.15 mol) in tetrahydrofuran (300 ml) was added 2-methoxypropene (144 ml, 1.5 mol) at room temperature, and then the mixture was cooled to 0° C. At the same temperature, pyridinium p-toluenesulfonic acid (75 mg, 0.30 mmol) was added and the mixture was stirred for 1 hour. The reaction mixture was poured into a saturated aqueous solution of sodium hydrogen carbonate cooled to 0° C., and extracted with toluene. The organic layer was washed with a saturated aqueous solution of sodium chloride, dried over anhydrous sodium sulfate, and the solvent was evaporated under reduced pressure to give the titled compound as an oil in quantitative yield. The product was used in the next step without further purification.

1H-NMR (CDCl3) δ: 1.44 (6H, s), 3.22 (3H, 4.48 (2H, s), 7.42 (1H, d, J=8.0 Hz), 7.44 (1H, dd, J=1.5, 8.0 Hz), 7.68 (1H, d, J=1.5 Hz).

Step 3: Synthesis of 2,3,4,5-tetrakis(trimethylsilyloxy)-6-trimethylsilyloxymethyl-2-(5-(4-ethylphenyl)hydroxymethyl-2-(1-methoxy-1-methylethoxymethyl)phenyl)tetrahydropyran

 

Figure US20110306778A1-20111215-C00019

 

Under a nitrogen atmosphere, 2,4-dibromo-1-(1-methoxy-1-methylethoxymethyl)benzene (70 g, 207 mmol), which was obtained in the previous step, was dissolved in toluene (700 mL) and t-butylmethyl ether (70 ml), and n-butyllithium in hexane (1.65 M, 138 ml, 227 mmol) was added dropwise at 0° C. over 30 minutes. After the mixture was stirred for 1.5 hours at 0° C., the mixture was added dropwise to a solution of 3,4,5-tris(trimethylsilyloxy)-6-trimethylsilyloxymethyl-tetrahydropyran-2-one (Example 1, 108 g, 217 mol) in tetrahydrofuran (507 ml) at −78° C., and the reaction mixture was stirred for 2 hours at the same temperature. Triethylamine (5.8 ml, 41 mmol) and trimethylsilyl chloride (29.6 ml, 232 mmol) were added thereto, and the mixture was warmed to 0° C. and stirred for 1 hour to give a solution containing 2,3,4,5-tetrakis(trimethylsilyloxy)-6-trimethylsilyloxymethyl-2-(5-bromo-2-(1-methoxy-1-methylethoxymethyl)phenyl)tetrahydropyran.

The resulting solution was cooled to −78° C., and n-butyllithium in hexane (1.65 M, 263 ml, 434 mmol) was added dropwise thereto at the same temperature. After the mixture was stirred at −78° C. for 30 minutes, 4-ethylbenzaldehyde (62 ml, 455 mmol) was added dropwise at −78° C., and the mixture was stirred at the same temperature for 2 hours. A saturated aqueous solution of ammonium chloride was added to the reaction mixture, and the organic layer was separated, and washed with water. The solvent was evaporated under reduced pressure to give a product containing the titled compound as an oil (238 g). The product was used in the next step without further purification.

A portion of the oil was purified by HPLC (column: Inertsil ODS-3, 20 mm I.D.×250 mm; acetonitrile, 30 mL/min) to give four diastereomers of the titled compound (two mixtures each containing two diastereomers).

Mixture of Diastereomers 1 and 2:

1H-NMR (500 MHz, CDCl3) δ: −0.47 (4.8H, s), −0.40 (4.2H, s), −0.003-0.004 (5H, m), 0.07-0.08 (1314, m), 0.15-0.17 (18H, m), 1.200 and 1.202 (3H, each t, J=8.0 Hz), 1.393 and 1.399 (3H, each s), 1.44 (3H, s), 2.61 (2H, q, J=8.0 Hz), 3.221 and 3.223 (3H, each s), 3.43 (1H, t, J=8.5 Hz), 3.54 (1H, dd, J=8.5, 3.0 Hz), 3.61-3.66 (1H, m), 3.80-3.85 (3H, m), 4.56 and 4.58 (1H, each d, J=12.4 Hz), 4.92 and 4.93 (1H, each d, J=12.4 Hz), 5.80 and 5.82 (1H, each d, J=3.0 Hz), 7.14 (2H, d, J=8.0 Hz), 7.28-7.35 (3H, m), 7.50-7.57 (2H, m).

MS (ESI+): 875 [M+Na]+.

Mixture of Diastereomers 3 and 4:

1H-NMR (500 MHz, toluene-d8, 80° C.) δ: −0.25 (4H, s), −0.22 (5H, s), 0.13 (5H, s), 0.16 (4H, s), 0.211 and 0.214 (9H, each s), 0.25 (9H, s), 0.29 (9H, s), 1.21 (3H, t, J=7.5 Hz), 1.43 (3H, s), 1.45 (3H, s), 2.49 (2H, q, J=7.5 Hz), 3.192 and 3.194 (3H, each s), 3.91-4.04 (4H, m), 4.33-4.39 (2H, m), 4.93 (1H, d, J=14.5 Hz), 5.10-5.17 (1H, m), 5.64 and 5.66 (1H, each s), 7.03 (2H, d, J=8.0 Hz), 7.28-7.35 (3H, m), 7.59-7.64 (1H, m), 7.87-7.89 (1H, m).

MS (ESI+): 875 [M+Na]+.

Step 4: Synthesis of 1,1-anhydro-1-C-[5-(4-ethylphenyl)hydroxymethyl-2-(hydroxymethyl)phenyl]-β-D-glucopyranose

 

Figure US20110306778A1-20111215-C00020

 

Under a nitrogen atmosphere, the oil containing 2,3,4,5-tetrakis(trimethylsilyloxy)-6-trimethylsilyloxymethyl-2-(5-(4-ethylphenyl)hydroxymethyl-2-(1-methoxy-1-methylethoxymethyl)phenyl)tetrahydropyran (238 g), which was obtained in the previous step, was dissolved in acetonitrile (693 ml). Water (37 ml) and 1N HCl aq (2.0 ml) were added and the mixture was stirred at room temperature for 5.5 hours. Water (693 ml) and n-heptane (693 ml) were added to the reaction mixture and the aqueous layer was separated. The aqueous layer was washed with n-heptane (693 ml×2), and water was evaporated under reduced pressure to give a product containing water and the titled compound (a diastereomer mixture) as an oil (187 g). The product was used in the next step without further purification.

1H-NMR (500 MHz, CD3OD) δ: 1.200 (3H, t, J=7.7 Hz), 1.201 (3H, t, J=7.7 Hz), 2.61 (2H, q, J=7.7 Hz), 3.44-3.48 (1H, m), 3.63-3.68 (111, m), 3.76-3.84 (4H, m), 5.09 (1H, d, J=12.8 Hz), 5.15 (1H, d, J=12.8 Hz), 5.79 (1H, s), 7.15 (2H, d, J=7.7 Hz), 7.24 and 7.25 (1H, each d, J=8.4 Hz), 7.28 (2H, d, J=7.7 Hz), 7.36 (1H, dd, J=8.4, 1.5 Hz), 7.40-7.42 (114, m).

MS (ESI+): 425 [M+Na]+.

Step 5: Synthesis of 1,1-anhydro-1-C-[5-(4-ethylphenyl)methyl-2-(hydroxymethyl)phenyl]-β-D-glucopyranose (crude product)

 

Figure US20110306778A1-20111215-C00021

 

To a solution of the oil containing 1,1-anhydro-1-C-[5-(4-ethylphenyl)hydroxymethyl-2-(hydroxymethyl)phenyl]-β-D-glucopyranose (187 g), which was obtained in the previous step, in 1,2-dimethoxyethane (693 ml) was added 5% Pd/C (26 g, 6.2 mmol, water content ratio: 53%), and the mixture was stirred in the atmosphere of hydrogen gas at room temperature for 4 hours. After filtration, the filtrate was evaporated under reduced pressure to give an oil containing the titled compound (59 g). The purity of the resulting product was 85.7%, which was calculated based on the area ratio measured by HPLC. The product was used in the next step without further purification.

1H-NMR (CD3OD) δ: 1.19 (3H, t, J=7.5 Hz), 2.59 (2H, q, J=7.5 Hz), 3.42-3.46 (1H, m), 3.65 (1H, dd, J=5.5, 12.0 Hz), 3.74-3.82 (4H, m), 3.96 (2H, s), 5.07 (1H, d, J=12.8 Hz), 5.13 (1H, d, J=12.8 Hz), 7.08-7.12 (4H, m), 7.18-7.23 (3H, m).

MS (ESI+): 387 [M+1]+.

Measurement Condition of HPLC:

Column: Cadenza CD-C18 50 mm P/NCD032

Mobile phase: Eluent A: H2O, Eluent B: MeCN

Gradient operation: Eluent B: 5% to 100% (6 min), 100% (2 min)

Flow rate: 1.0 mL/min

Temperature: 35.0° C.

Detection wavelength: 210 nm

Step 6: Synthesis of 1,1-anhydro-1-C-[5-(4-ethylphenyl)methyl-2-(hydroxymethyl)phenyl]-2,3,4,6-tetra-O-methoxycarbonyl-β-D-glucopyranose

 

Figure US20110306778A1-20111215-C00022

 

Under a nitrogen atmosphere, to a solution of the oil containing 1,1-anhydro-1-C-[5-(4-ethylphenyl)methyl-2-(hydroxymethyl)phenyl]-β-D-glucopyranose (59 g) and 4-(dimethylamino)pyridine (175 g, 1436 mmol) in acetonitrile (1040 ml) was added dropwose methyl chloroformate (95 ml, 1231 mmol) at 0° C. The mixture was allowed to warm to room temperature while stirred for 3 hours. After addition of water, the mixture was extracted with isopropyl acetate. The organic layer was washed with an aqueous solution of 3% potassium hydrogensulfate and 20% sodium chloride (three times) and an aqueous solution of 20% sodium chloride, dried over anhydrous sodium sulfate, and the solvent was evaporated under reduced pressure. To the resulting residue was added ethanol (943 mL) and the mixture was heated to 75° C. to dissolve the residue. The mixture was cooled to 60° C. and a seed crystal of the titled compound was added thereto. The mixture was cooled to room temperature and stirred for 1 hour. After precipitation of solid was observed, water (472 ml) was added thereto, and the mixture was stirred at room temperature for 2 hours. The resulting crystal was collected by filtration, washed with a mixture of water and ethanol (1:1), and dried under reduced pressure to give the titled compound (94 g). To the product (91 g) was added ethanol (1092 ml), and the product was dissolved by heating to 75° C. The solution was cooled to 60° C. and a seed crystal of the titled compound was added thereto. The mixture was cooled to room temperature and stirred for 1 hour. After precipitation of solid was observed, water (360 ml) was added thereto, and the mixture was stirred at room temperature for 2 hours. The resulting crystal was collected by filtration, washed with a mixture of water and ethanol (1:1), and dried under reduced pressure to give the titled compound [83 g, total yield from 2,4-dibromo-1-(1-methoxy-1-methylethoxymethyl)benzene used in Step 3: 68%].

1H-NMR (CDCl3) δ: 1.20 (3H, t, J=7.5 Hz), 2.60 (2H, q, J=7.5 Hz), 3.50 (3H, s), 3.76 (3H, s), 3.77 (3H, s), 3.81 (3H, s), 3.96 (2H, s), 4.23 (1H, dd, J=2.5, 11.8 Hz), 4.33 (1H, dd, J=4.5, 12.0 Hz), 4.36-4.40 (1H, m), 5.11-5.20 (3H, m), 5.41 (1H, d, J=10.0 Hz), 5.51 (1H, t, J=10.0 Hz), 7.07-7.11 (4H, m), 7.14 (1H, d, J=7.5 Hz), 7.19 (1H, dd, J=1.5, 7.8 Hz), 7.31 (1H, d, J=1.5 Hz).

MS (ESI+): 619 [M+1]+, 636 [M+18]+.

Another preparation was carried out in the same manner as Step 6, except that a seed crystal was not used, to give the titled compound as a crystal.

Step 7: Synthesis of 1,1-anhydro-1-C-[5-(4-ethylphenyl)methyl-2-(hydroxymethyl)phenyl]-β-D-glucopyranose

 

Figure US20110306778A1-20111215-C00023

 

To a solution of 1,1-anhydro-1-C-[5-(4-ethylphenyl)methyl-2-(hydroxymethyl)phenyl]-2,3,4,6-tetra-O-methoxycarbonyl-β-D-glucopyranose (8.92 kg as wet powder, corresponding to 8.00 kg of dry powder) in 1,2-dimethoxyethane (28 kg) was added a solution of sodium hydroxide (4 mol/L, 30.02 kg) at 20° C., and the mixture was stirred for 1 hour. Water (8.0 kg) was added to the mixture and the layers were separated. To the organic layer were added an aqueous solution of 25% sodium chloride (40 kg) and ethyl acetate (36 kg). The organic layer was separated, washed with an aqueous solution of 25% sodium chloride (40 kg), and the solvent was evaporated under reduced pressure. The purity of the resulting residue was 98.7%, which was calculated based on the area ratio measured by HPLC. To the resulting residue were added acetone (32.0 kg) and water (0.8 kg), and the solvent was evaporated under reduced pressure. To the resulting residue were added acetone (11.7 kg) and water (15.8 kg), and the solution was cooled to 5° C. or below. Water (64 kg) was added to the solution at 10° C. or below, and the mixture was stirred at the same temperature for 1 hour. The resulting crystal was collected by centrifugation, and washed with a mixture of acetone (1.3 kg) and water (8.0 kg). The resulting wet powder was dried by ventilation drying under a condition at air temperature of 13 to 16° C. and relative humidity of 24% to 33% for 8 hours, to give a monohydrate crystal (water content: 4.502%) of the titled compound (3.94 kg). The purity of the resulting compound was 99.1%, which was calculated based on the area ratio measured by HPLC.

1H-NMR (CD3OD) δ: 1.19 (3H, t, J=7.5 Hz), 2.59 (2H, q, J=7.5 Hz), 3.42-3.46 (1H, m), 3.65 (1H, dd, J=5.5, 12.0 Hz), 3.74-3.82 (4H, m), 3.96 (2H, s), 5.07 (1H, d, J=12.8 Hz), 5.13 (1H, d, J=12.8 Hz), 7.08-7.12 (4H, m), 7.18-7.23 (311, m).

MS (ESI+): 387 [M+1]+.

Measurement Condition of HPLC:

Column: Capcell pack ODS UG-120 (4.6 mm I.D.×150 mm, 3 μm, manufactured by Shiseido Co., Ltd.)

Mobile phase: Eluent A: H2O, Eluent B: MeCN

Mobile phase sending: Concentration gradient was controlled by mixing Eluent A and Eluent B as indicated in the following table.

 

TABLE 1
Time from
injection (min) Eluent A (%) Eluent B (%)
0 to 15 90→10 10→90
15 to 17.5 10 90
17.5 to 25 90 10

 

Flow rate: 1.0 mL/min

Temperature: 25.0° C.

Detection wavelength: 220 nm

Method for Measurement of Water Content:

Analysis method: coulometric titration method

KF analysis apparatus: Type KF-100 (trace moisture measuring apparatus manufactured by Mitsubishi Chemical Corporation)

Anode solution: Aquamicron AX (manufactured by Mitsubishi Chemical Corporation)

Cathode solution: Aquamicron CXU (manufactured by Mitsubishi Chemical Corporation)

 

 

PATENT

US20090030006

The compound of the present invention can be synthesized as shown in Scheme 1:

 

Figure US20090030006A1-20090129-C00005
Figure US20090030006A1-20090129-C00006

 

wherein R11 and R12 have the same meaning as defined above for substituents on Ar1, A is as defined above, and P represents a protecting group for a hydroxyl group.

CLIP

Tofogliflozin hydrate (Deberza)
Tofogliflozin hydrate, which is a sodium-glucose co-transporter 2 inhibitor, was approved in Japan for the treatment of type 2 diabetes
at the same time as luseogliflozin hydrate (XIX). The drug was discovered by Chugai Pharmaceutical and jointly developed
with Sanofi-Aventis and Kowa.263

Tofogliflozin hydrate reduces glucose levels by inhibiting the reuptake of glucose by selectively
inhibiting SGLT2, and plays a key role in the reuptake of glucose in the proximal tubule of the kidneys.264–266 The synthetic
approach described in Scheme 48 represents the largest scale reported to date in a patent application.263,266–268

Reduction of commercially available 2-bromoterephtalic acid (268, Scheme 48) through the use of trimethoxyborane and borane-THF proceeded in 89% yield to afford diol 269.

Subjection of this compound to 2-methoxypropene (270) under acidic conditions generated bis-acetonide 271. This bromide then underwent lithium–halogen exchange followed by exposure to magnesium bromide and treatment with lactone 272 (which was prepared by persilylation of commercially available (3R,4S,5S,6R)-3,4,5-trihydroxy-6-hydroxymethyl)tetrahydro-2Hpyran-2-one (277, Scheme 49).

This mixture was worked up with aqueous ammonium chloride and upon treatment with p-TsOH in methanol resulted in spiroacetal 273. Next, global protection of all alcohol functionalities within 273 was affected by reaction with methylchloroformate and DMAP in acetonitrile.

The benzyl carbonate within 274 was selectively exchanged via Suzuki coupling with 4-ethylphenylboronic acid (275) to afford methylene dibenzyl system 276. Subsequent treatment with aqueous sodium hydroxide in methanol followed by crystallization from 1:6 acetone and water furnished the desired product tofogliflozin hydrate (XXXIV) in 75% yield.

STR1

STR1

263 Takamitsu, K.; Tsutomu, S.; Masahiro, N. WO Patent 2006080421A1, 2006.
264. http://www.info.pmda.go.jp/shinyaku/P201400036/index.html.
265. Pafili, K.; Papanas, N. Expert Opin. Pharmacother. 2014, 15, 1197.

266. Ohtake, Y.; Sato, T.; Kobayashi, T.; Nishimoto, M.; Taka, N.; Takano, K.;Yamamoto, K.; Ohmori, M.; Yamaguchi, M.; Takami, K.; Yeu, S. Y.; Ahn, K. H.;Matsuoka, H.; Morikawa, K.; Suzuki, M.; Hagita, H.; Ozawa, K.; Yamaguchi, K.;Kato, M.; Ikeda, S. J. Med. Chem. 2012, 55, 7828.
267. Murakata, M.; Ikeda, T.; Kawase, A.; Nagase, M.; Kimura, N.; Takeda, S.;Yamamoto, K.; Takano, K.; Nishimoto, M.; Ohtake, Y.; Emura, T.; Kito, Y. WOPatent 2011074675A1, 2011.
268. Murakata, M.; Takuma, I.; Nobuaki, K.; Masahiro, N.; Kawase, A.; Nagase, M.;Yamamoto, K.; Takata, N.; Yoshizaki, S. WO Patent 2009154276A1, 2009.

 

Paper

A Scalable Synthesis of Tofogliflozin Hydrate

Pharmaceutical Research Center, Disha Pharmaceutical Group Co., Ltd., Weihai 264205, China
Org. Process Res. Dev., Article ASAP
Abstract Image

A newly process for the synthesis of tofogliflozin hydrate, a sodium-glucose cotransporter type 2 (SGLT2) inhibitor, was described. Three improvements were achieved, including the development of a regioselective Friedel–Crafts reaction, a high-yield reduction, and a mild metal–halogen exchange. These improvements ultimately resulted in the isolation of tofogliflozin hydrate as a white solid in >99% purity (HPLC area) and 23% overall yield after 12 steps without column chromatography.

 

 Tofogliflozin hydrate white solid with 99.56% purity by HPLC. Water content: 4.47%.

Mp: 71−80 oC. [α]20 D =  +23.9 (c = 1.0, CH3OH).

1H NMR (400 MHz, CD3OD) δ 7.23-7.18 (m, 3H), 7.12-7.08(m, 4H), 5.13 (d, J = 12.4 Hz, 1H), 5.07 (d, J = 12.4 Hz, 1H), 3.96 (s, 2H), 3.83-3.73 (m, 4H), 3.65 (dd, J = 11.9, 5.5 Hz, 1H), 3.41-3.47 (m, 1H), 2.59 (q, J = 7.6 Hz, 2H), 1.19 (t, J = 7.6 Hz, 3H).

13C NMR (100 MHz, CD3OD) δ 143.2, 142.6, 140.2, 139.9, 139.7, 131.2, 129.9, 128.9, 123.6, 121.8, 111.6, 76.4, 76.2, 74.9, 73.4, 71.9, 62.8, 42.3, 29.5, 16.3.

HRMS (ESI) m/z: [M+H]+ Calcd for C22H27O6 387.1802; Found 387.1805.

IR (KBr, cm-1) ν: 3362, 2962, 2927, 1637, 1513, 1429, 1095, 1034, 808, 770. Spectroscopic data were identical with those reported.1b, 2

1. (a) Suzuki, M.; Honda, K.; Fukazawa, M.; Ozawa, K.; Hagita, H.; Kawai, T.; Takeda, M.; Yata, T.; Kawai, M.; Fukuzawa, T.; Kobayashi, T.; Sato, T.; Kawabe, Y.; Ikeda, S. J. Pharmacol. Exp. Ther. 2012, 341, 692.

(b) Ohtake, Y.; Sato, T.; Kobayashi, T.; Nishimoto, M.; Taka, N.; Takano, K.; Yamamoto, K.; Ohmori, M.; Yamaguchi, M.; Takami, K.; Yeu, S.-H.; Ahn. K.-H.; Matsuoka, H.; Morikawa, K.; Suzuki, M.; Hagita, H.; Ozawa, K.; Yamaguchi, K.; Kato, M.; Ikeda, S. J. Med. Chem. 2012, 55, 7828.

(c) Ikeda, S.; Takano, Y.; Cynshi, O.; Tanaka, R.; Christ, A. D.; Boerlin, V.; Beyer, U.; Beck, A.; Ciorciaro, C.; Meyer, M.; Kadowaki, T. Diabetes, Obesity and Metabolism 2015, 17, 984.

2. (a) Murakata, M.; Ikeda, T.; Kimura, N.; Kawase, A.; Nagase, M.; Yamamoto, K.; Takata, N.; Yoshizaki, S.; Takano, K. Crystal of spiroketal derivative, and process for production thereof. European Appl. EP 2308886 A1, April 13, 2011.

(b) Ohtake, Y.; Emura, T.; Nishimoto, M.; Takano, K.; Yamamoto, K.; Tsuchiya, S.; Yeu, S.; Kito, Y.; Kimura, N.; Takeda, S.; Tsukazaki, M.; Murakata, M.; Sato, T. J. Org. Chem. 2016, 81, 2148.

References

  1.  Chugai Pharmaceutical: Development Pipeline
  2.  Nagata, T.; Fukazawa, M.; Honda, K.; Yata, T.; Kawai, M.; Yamane, M.; Murao, N.; Yamaguchi, K.; Kato, M.; Mitsui, T.; Suzuki, Y.; Ikeda, S.; Kawabe, Y. (2012). “Selective SGLT2 inhibition by tofogliflozin reduces renal glucose reabsorption under hyperglycemic but not under hypo- or euglycemic conditions in rats”. AJP: Endocrinology and Metabolism 304 (4): E414–E423. doi:10.1152/ajpendo.00545.2012.PMID 23249697.
  3.  Ohtake, Y.; Sato, T.; Kobayashi, T.; Nishimoto, M.; Taka, N.; Takano, K.; Yamamoto, K.; Ohmori, M.; Yamaguchi, M.; Takami, K.; Yeu, S. Y.; Ahn, K. H.; Matsuoka, H.; Morikawa, K.; Suzuki, M.; Hagita, H.; Ozawa, K.; Yamaguchi, K.; Kato, M.; Ikeda, S. (2012). “Discovery of Tofogliflozin, a NovelC-Arylglucoside with anO-Spiroketal Ring System, as a Highly Selective Sodium Glucose Cotransporter 2 (SGLT2) Inhibitor for the Treatment of Type 2 Diabetes”. Journal of Medicinal Chemistry 55 (17): 7828–7840. doi:10.1021/jm300884k.PMID 22889351.
  4.  Statement on a nonproprietary name adopted by the USAN council: Tofogliflozin.
  5.  http://www.who.int/entity/medicines/publications/druginformation/innlists/RL65.pdf
Tofogliflozin monohydrate
Tofogliflozin monohydrate skeletal 3D.svg
Systematic (IUPAC) name
(1S,3′R,4′S,5′S,6′R)-6-(4-Ethylbenzyl)-6′-(hydroxymethyl)-3′,4′,5′,6′-tetrahydro-3H-spiro[2-benzofuran-1,2′-pyran]-3′,4′,5′-triol hydrate (1:1)
Legal status
Legal status
  • Investigational
Identifiers
CAS Number 1201913-82-7
903565-83-3 (anhydrous)
ATC code None
PubChem CID 46908928
ChemSpider 28527871
KEGG D09978
ChEMBL CHEMBL2105711
Synonyms CSG452
Chemical data
Formula C22H28O7
Molar mass 404.45 g/mol

//////////TOFOGLIFLOZIN, 托格列净 , CSG-452, R-7201, RG-7201, 1201913-82-7  , 903565-83-3, oral hypoglycaemic agentsSGLT-2 inhibitorstype 2 diabetes mellitus, Deberza

CCc1ccc(cc1)Cc2ccc3c(c2)[C@]4([C@@H]([C@H]([C@@H]([C@H](O4)CO)O)O)O)OC3.O

The glucopyranosyl-substituted benzene derivatives are proposed as inducers of urinary sugar excretion and as medicaments in the treatment of diabetes.

The term “canagliflozin” as employed herein refers to canagliflozin, including hydrates and solvates thereof, and crystalline forms thereof and has the following structure:

Figure US20130035281A1-20130207-C00013

The compound and methods of its synthesis are described in WO 2005/012326 and WO 2009/035969 for example. Preferred hydrates, solvates and crystalline forms are described in the patent applications WO 2008/069327 for example.

atigliflozin, including hydrates and solvates thereof, and crystalline forms thereof and has the following structure:

Figure US20130035281A1-20130207-C00014

The compound and methods of its synthesis are described in WO 2004/007517 for example.

ipragliflozin, including hydrates and solvates thereof, and crystalline forms thereof and has the following structure:

Figure US20130035281A1-20130207-C00015

The compound and methods of its synthesis are described in WO 2004/080990, WO 2005/012326 and WO 2007/114475 for example.

tofogliflozin, including hydrates and solvates thereof, and crystalline forms thereof and has the following structure:

Figure US20130035281A1-20130207-C00016

The compound and methods of its synthesis are described in WO 2007/140191 and WO 2008/013280 for example.

remogliflozin and prodrugs of remogliflozin, in particular remogliflozin etabonate, including hydrates and solvates thereof, and crystalline forms thereof. Methods of its synthesis are described in the patent applications EP 1213296 and EP 1354888 for example.

sergliflozin and prodrugs of sergliflozin, in particular sergliflozin etabonate, including hydrates and solvates thereof, and crystalline forms thereof. Methods for its manufacture are described in the patent applications EP 1344780 and EP 1489089 for example.

luseoghflozin, including hydrates and solvates thereof, and crystalline forms thereof and has the following structure:

Figure imgf000031_0002

ertugliflozin, including hydrates and solvates thereof, and crystalline forms thereof and has the following structure:

Figure imgf000031_0003

and is described for example in WO 2010/023594.

The compound of the formula

Figure imgf000032_0001

is described for example in WO 2008/042688 or WO 2009/014970.

Dapagliflozin

Figure US20130096076A1-20130418-C00001

The compound is described for example in WO 03/099836. Crystalline forms are described for example in WO 2008/002824.

Remogliflozin and Remogliflozin Etabonate

Figure US20130096076A1-20130418-C00002

The compound is described for example in EP 1354888 A1.

Sergliflozin and Sergliflozin Etabonate

Figure US20130096076A1-20130418-C00003

The compounds are described in EP 1 329 456 A1 and a crystalline form ofSergliflozin etabonate is described in EP 1 489 089 A1.

1-Chloro-4-(β-D-glucopyranos-1-yl)-2-(4-ethyl-benzyl)-benzene

Figure US20130096076A1-20130418-C00004

The compound is described in WO 2006/034489.

(1S)-1,5-anhydro-1-[5-(azulen-2-ylmethyl)-2-hydroxyphenyl]-D-glucitol

Figure US20130096076A1-20130418-C00005

The compound (4-(Azulen-2-ylmethyl)-2-(β-D-glucopyranos-1-yl)-1-hydroxy-benzene) is described in WO 2004/013118 and WO 2006/006496. The crystalline choline salt thereof is described in WO 2007/007628.

(1S)-1,5-anhydro-1-[3-(1-benzothien-2-ylmethyl)-4-fluorophenyl]-D-glucitol

Figure US20130096076A1-20130418-C00006

The compound is described in WO 2004/080990 and WO 2005/012326. A cocrystal with L-proline is described in WO 2007/114475.

Thiophen Derivatives of the Formula (7-1)

Figure US20130096076A1-20130418-C00007

wherein R denotes methoxy or trifluoromethoxy. Such compounds and their method of production are described in WO 2004/007517, DE 102004063099 and WO 2006/072334.

1-(β-D-glucopyranosyl)-4-methyl-3-[5-(4-fluorophenyl)-2-thienylmethyl]benzene

Figure US20130096076A1-20130418-C00008

The compound is described in WO 2005/012326. A crystalline hemihydrate is described in WO 2008/069327.

Spiroketal Derivatives of the Formula (9-1)

Figure US20130096076A1-20130418-C00009

wherein R denotes methoxy, trifluoromethoxy, ethoxy, ethyl, isopropyl or tert. butyl. Such compounds are described in WO 2007/140191 and WO 2008/013280.

Share
Dec 172013
 

ALISKIREN

(2S,4S,5S,7S)-5-amino-N-(2-carbamoyl-2,2-dimethylethyl)-4-hydroxy-7-{[4-methoxy-3-(3-methoxypropoxy)phenyl]methyl}-8-methyl-2-(propan-2-yl)nonanamide,  CAS 173334-57-1, base

CAS 173334-58-2,aliskiren hemifumarate

Aliskiren is a renin inhibitor. It was approved by the U.S. Food and Drug Administration in 2007 for the treatment of hypertension.

2-C30-H53-N3-O6.C4-H4-O4
1219.599
Novartis (Originator), Speedel (Licensee)
CARDIOVASCULAR DRUGS, Heart Failure Therapy, Hypertension, Treatment of, Renal Failure, Agents for, RENAL-UROLOGIC DRUGS, Treatment of Renal Diseases, Renin Inhibitors

Tekturna contains aliskiren hemifumarate, a renin inhibitor, that is provided as tablets for oral administration. Aliskiren hemifumarate is chemically described as (2S,4S,5S,7S)-N-(2-carbamoyl-2-methylpropyl)-5-amino-4-hydroxy-2,7diisopropyl-8-[4-methoxy-3-(3-methoxypropoxy)phenyl]-octanamide hemifumarate and its structural formula is

Tekturna® (aliskiren) Structural Formula Illustration

Molecular formula: C30H53N3O6 • 0.5 C4H4O4

Aliskiren hemifumarate is a white to slightly yellowish crystalline powder with a molecular weight of 609.8 (free base- 551.8). It is soluble in phosphate buffer, n-octanol, and highly soluble in water.

Country
Patent Number
Approved
Expires (estimated)
Canada 2147056 2005-10-25 2015-04-13
United States 5559111 1998-07-21 2018-07-21

Aliskiren (INN) (trade names Tekturna, US; Rasilez, UK and elsewhere) is the first in a class of drugs called direct renin inhibitors. Its current licensed indication is essential (primary) hypertension.

Aliskiren was co-developed by the Swiss pharmaceutical companies Novartis andSpeedel.[1][2] It was approved by the US Food and Drug Administration in 2007 for the treatment of primary hypertension.[3]

In December 2011, Novartis had to halt a clinical trial of the drug after discovering increased incidence of nonfatal stroke, renal complications, hyperkalemia, and hypotension in patients with diabetes and renal impairment (ALTITUDE Trial ).[4] [5]

As a result, in April 20, 2012:

A new contraindication was added to the product label concerning the use of aliskiren with angiotensin receptor blockers (ARBs) or angiotensin-converting enzyme inhibitors (ACEIs) in patients with diabetes because of the risk of renal impairment, hypotension, and hyperkalemia.

A warning to avoid use of aliskiren with ARBs or ACEIs was also added for patients with moderate to severe renal impairment (i.e., where glomerular filtration rate is less than 60 ml/min).

Renin, the first enzyme in the renin-angiotensin-aldosterone system, plays a role in blood pressure control. It cleaves angiotensinogen to angiotensin I, which is in turn converted byangiotensin-converting enzyme (ACE) to angiotensin II. Angiotensin II has both direct and indirect effects on blood pressure. It directly causes arterial smooth muscle to contract, leading to vasoconstriction and increased blood pressure. Angiotensin II also stimulates the production of aldosterone from the adrenal cortex, which causes the tubules of the kidneys to increase reabsorption of sodium, with water following, thereby increasing plasma volume, and thus blood pressure. Aliskiren binds to the S3bp binding site of renin, essential for its activity.[6] Binding to this pocket prevents the conversion of angiotensinogen to angiotensin I. Aliskiren is also available as combination therapy withhydrochlorothiazide.[7]

Many drugs control blood pressure by interfering with angiotensin or aldosterone. However, when these drugs are used chronically, the body increases renin production, which drives blood pressure up again. Therefore, doctors have been looking for a drug to inhibit renin directly. Aliskiren is the first drug to do so.[8][9]

Aliskiren may have renoprotective effects independent of its blood pressure−lowering effect in patients with hypertension, type 2 diabetes, and nephropathy, who are receiving the recommended renoprotective treatment. According to the AVOID study, researchers found that treatment with 300 mg of aliskiren daily, as compared with placebo, reduced the mean urinary albumin-to-creatinine ratio by 20%, with a reduction of 50% or more in 24.7% of the patients who received aliskiren as compared with 12.5% of those who received placebo. Furthermore, the AVOID trial showed treatment with 300 mg of aliskiren daily reduces albuminuria in patients with hypertension, type 2 diabetes, and proteinuria, who are receiving the recommended maximal renoprotective treatment with losartan and optimal antihypertensive therapy. Therefore, direct renin inhibition will have a critical role in strategic renoprotective pharmacotherapy, in conjunction with dual blockade of the renin−angiotensin−aldosterone system with the use of ACE inhibitors and angiotensin II–receptor blockers, very high doses of angiotensin II−receptor blockers, and aldosterone blockade.[10]

Aliskiren is a minor substrate of CYP3A4 and, more important, P-glycoprotein:

  • It reduces furosemide blood concentration.
  • Atorvastatin may increase blood concentration, but no dose adjustment is needed.
  • Due to possible interaction with ciclosporin, the concomitant use of ciclosporin and aliskiren is contraindicated.
  • Caution should be exercised when aliskiren is administered with ketoconazole or other moderate P-gp inhibitors (itraconazole, clarithromycin, telithromycin, erythromycin, or amiodarone).
  • Doctors should stop prescribing aliskiren-containing medicines to patients with diabetes (type 1 or type 2) or with moderate to severe kidney impairment who are also taking an ACE inhibitor or ARB, and should consider alternative antihypertensive treatment as necessary.[13]
  • Aliskiren (I) is a second generation renin inhibitor with renin-angiotensin system (RAS) as its target. It’s used clinically in the form of Aliskiren hemifumarate (Rasilez®) and was approved by FDA in May, 2007.
  •  Aliskiren has the chemical name: (2S, 4S, 5S, 7S)-5-amino-N-(2-carbamoyl-2-methylpropyl)-4-hydroxy-2-isopropyl-7-[4-methoxy-3-(3-methoxypropoxy)benzyl]-8-methyloctanamide (CAS No.: 173334-57-1). Its chemical structure is illustrated with Formula I given below:
    Figure imgb0001
  •  The method of preparation for Aliskiren and its intermediates has been reported in US7132569 , WO0208172 , US5559111 (equivalent patent toCN1266118 ), US5606078 CN101016253 WO2007/045421 ,EP2062874 , Helvetica ChimicaActa (2005, 3263-3273).
  • In US7132569 , WO0208172 et al., the preparation of Aliskiren (I) comprises the following steps as described in reaction scheme 1: coupling 2-(3-methoxypropoxy)-4-((R)-2-(bromomethyl)-3-methylbutyl)-1-methoxybenzene (II) with (2S, 4E)-5-chloro-2-isopropyl-4-pentenoic acid derivative (III) to obtain the compound of formula IV; halolactonization of the compound of formula IV to obtain the compound of formula V; then substituting the compound of formula V with azide to obtain the compound of formula VI; ring-opening the compound of formula VI with 3-amino-2,2-dimethylpropionamide (VII) in the presence of 2-hydroxypyridine and triethylamine to obtain the compound of formula VIII and a final catalytic hydrogenation of the compound of formula VIII to obtain Aliskiren (I). This preparation process is illustrated in Reaction Scheme 1.

    Figure imgb0002
  • In the patented preparation described above, chiral starting materials with the compounds of formula II and III are utilized to obtain the compound of formula IV. However, the reactions followed after the preparation of the compound of formula IV, such as the halolactonization and especially the substitutive reaction between the compound of formula V and azide, have problems of low yields and numerous by-products, which is not conducive to industrial scale production.
  •  US5559111 (equivalent patent CN1266118 ) and US5606078 et al. report the preparation of the compound of formula XI via Grignard reaction with 4-bromo-1-methoxy-2-(3-methoxypropoxy)benzene (IX) and the compound of formula X as starting materials as illustated in Reaction Scheme 2:
    Figure imgb0003
  • In the patented preparation described above, there are multiple reaction steps in the preparation of the compound of formula X from the compound of formula XII. The key steps, as described in Reaction Scheme 3, involve selective reduction agents such as sodium tri-tert-butoxyaluminum hydride and diisobutylaluminium hydride to prepare aldehyde and the reaction conditions need to be very well-controlled.

    Figure imgb0004
    Figure imgb0005
  • [0009]
    The compound of formula XI prepared by reaction scheme 2 could then be converted into Aliskiren (I) after multiple catalytic hydrogenation, protection and de-protection. In this method of preparation, a stepwise catalytic hydrogenation, azido reduction and dehydroxylation were implemented to reduce by-products during the catalytic hydrogenation. In addition, it is necessary to protect and de-protect the free hydroxyl group during the preparation. This synthetic scheme has disadvantage of multiple synthetic steps, tedious operation, lengthy overall reaction duration, low yield and particularly high production cost for the starting compound of formula X.
  • WO2007/045421 has reported an improved preparation method in which the starting material 4-bromo-1-methoxy-2-(3-methoxypropoxy)benzene (IX) firstly reacts with the compound of formula XIII via Grignard reaction to obtain the compound of formula XIV, and then followed by catalytic hydrogenation and ketone reduction to yield the compound of formula XV-A, as illustrated in Reaction Scheme 4:

    Figure imgb0006
    Figure imgb0007
  •  In the above preparation, expensive reagents, such as sodium tri-tert-butoxyaluminum hydride and diisobutylaluminium hydride were eliminated, but additional synthetic steps were introduced. In addition, the preparation of the compound of formula XV-A prepared from the compound of formula XIV via ketone reduction required extended reaction time, great amount of catalyst with multiple small addition and good operation skills.
  •  EP2062874A1 provides a method in preparing the compound of formula XVI. In this method, the compound of formula XVII is obtained from the compound of formula XVI via halogenation. A corresponding Grignard reagent is firstly prepared from the compound of formula IX or XVII reacting with magnesium, which is then couples with another chemical in the presence of the metal catalyst iron(III) acetylacetonate (Fe(acac)3) to obtain the compound of formula XVIII as described in Reaction Scheme 5:
    Figure imgb0008
    Figure imgb0009
  • In EP2062874A1 , the compound of formula XVIII reacts with 3-amino-2,2-dimethylpropionamide (VII). The resulted product is then through reduction of the azio group to obtain Aliskiren (I). In this patent, detailed experimental protocol was not provided although N-methylpyrrolidone was mentioned as solvent. We found: 1) it is difficult to prepare the Grgnard reagent from the compound of formula IX; 2) the compounds of formula XVII and XVIII are not quite stable in the presence of iron(III) acetylacetonate. In addition, the yield in preparing the compound of formula XVIII was extremely low.

the spiro aldehyde (XLVII) is treated with N-benzylhydroxylamine in dichloromethane to give nitrone (LII), which is submitted to a Grignard reaction with the magnesium derivative of intermediate (XXX) in THF to afford the adduct (LIII) as a mixture of epimers at the amino group. Simultaneous N-dehydroxylation and cleavage of the spiro function of (LIII) by means of Zn, Cu (OAc) 2 in AcOH / water gives lactone (LIV), which is condensed with 3-amino- 2,2-dimethylpropionamide (XIX) by means of TEA and 2-hydroxypyridine giving the adduct (LV). Finally, the benzylamino group of (LV) is removed with H2 over Pd / C in methanol to yield a mixture of two epimers at the amino group, from which aliskiren is separated.
Tetrahedron Lett2001, 42, (29): 4819

NMR

ALISKIREN BASE

Figure imgb0023

EP2546243A1

MS m/z: 552.6 (M+H)+; 1H-NMR (400 MHz, CDCl3) δ 6.88-6.75 (m, 3H), 4.08-4.04 (t, J = 6.3Hz, 2H), 3.79 (s, 3H), 3.60-3.55 (t, J = 6.3Hz, 2H), 3.30 (s, 3H), 3.30-3.25 (m, 3H), 2.69 (m, 2H), 2.49 (m, 1H), 2.27 (m, 1H), 2.04 (m, 2H), 1.78-1.35 (m, 7H), 1.10 (m, 6H), 0.90 (m, 12H) ppm.

Paper

Abstract Image

A novel synthesis of the renin inhibitor aliskiren based on an unprecedented disconnection between C5 and C6 was developed, in which the C5 carbon acts as a nucleophile and the amino group is introduced by a Curtius rearrangement, which follows a simultaneous stereocontrolled generation of the C4 and C5 stereogenic centers by an asymmetric hydrogenation. Operational simplicity, step economy, and a good overall yield makes this synthesis amenable to manufacture on scale.

Convergent Synthesis of the Renin Inhibitor Aliskiren Based on C5–C6 Disconnection and CO2H–NH2 Equivalence

Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
Chemessentia SRL, Via Bovio 6, 28100 Novara, Italy
§ Dipartimento di Chimica e Chimica Industriale, Università degli Studi di Genova, Via Dodecaneso 31, 16146 Genova, Italy
Dipartimento di Chimica, Materiali e Ingegneria Chimica “G. Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy
Johnson Matthey Catalysis and Chiral Technologies, 28 Cambridge Science Park, Milton Road, Cambridge CB4 0FP, United Kingdom
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.5b00396
Publication Date (Web): January 5, 2016
Copyright © 2016 American Chemical Society
PAPER
 
PAPER
EP 0678500; EP 0678503; JP 1996053434; JP 1996081430; US 5559111; US 5627182; US 5646143
Alkylation of 3-hydroxy-4-methoxybenzyl alcohol (I) with 1-bromo-3-methoxypropane (II) gives ether (III). Subsequent conversion of benzyl alcohol (III) into bromide (IV) is carried out using bromotrimetylsilane. The chiral isovaleryloxazolidinone (V) is alkylated with bromide (IV) by means of LiHMDS to afford (VI), which is hydrolyzed to the (S)-2-aryl-2-isopropylpropionic acid (VII) by means of lithium peroxide. The reduction of acid (VII) to the corresponding alcohol with NaBH4/I2 reagent, followed by treatment with PPh3 and NBS, provides bromide (VIII). Alkylation of the chiral dimethoxydihydropyrazin (IX) with bromide (VIII) produces (X). Further hydrolysis of the pyrazine ring of (X) with HCl, followed by Boc protection of the resulting (S,S)-amino ester, yields compound (XI). Reduction of the ester group of (XI) with DIBAL gives aldehyde (XII). This compound is condensed with the Grignard reagent (XIII) to afford the diastereomeric mixture of amino alcohols (XIV). Treatment of mixture (XIV) with 2,2-dimethoxypropane (XV) and TsOH produces a mixture of oxazolidines, from which the required (S,S,S)-isomer (XVI) is isolated by flash chromatography. Hydrogenolitic deprotection of the benzyl ether of (XVI) gives alcohol (XVII).
This alcohol is oxidized to aldehyde with NMMO and tetrapropylammonium perruthenate (TPAP), and further oxidized to carboxylic acid (XVIII) with KMnO4 and tetrabutylammonium bromide (TBAB). Coupling of (XVIII) with aminoamide (XIX) by means of diethyl cyanophosphonate and TEA gives (XX). Finally, acid hydrolysis of the oxazolidine ring and Boc protecting groups of (XX) furnishes the corresponding amino alcohol, which is finally converted to the hemifumarate salt.
WO 0109079; WO 0109083
 Alternatively, the chiral azido intermediate (XXXIV) can also be synthesized as follows: Alkylation of oxazolidinone (V) with 1-chloro-3-iodopropene (XLVIII) by means of LiHMDS in THF gives compound (XLIX), which is condensed with the magnesium derivative of the phenylpropyl chloride (XXX) to yield, after working up, amide (L). Bromination of (L) with NBS and phosphoric acid affords the bromolactone (LI), which by treatment with NaN3 in tripropylene glycol/water provides the azido derivative (XXXIV).
WO 0202500
The condensation of benzaldehyde (I) with ethyl isovalerate (II) by means of hexyl lithium and DIA in THF gives the hydroxyester (III), which is acylated with Ac2O and DMAP in THF to yield the acetoxy derivate (IV). The elimination reaction in (IV) by means of t-BuOK in THF affords the unsaturated ester (V), which is hydrolyzed with KOH in ethanol to provide the unsaturated free acid (VI). Finally, this compound is enantioselectively reduced with H2 over several chiral Rh catalysts {[Rh(NBD)2BF4, [Rh(NBD)(OCOCF3)2], [Rh(NBD)Cl2], etc} to give the target intermediate 2(R)-isopropyl-3-[4-methoxy-3-(3-methoxypropoxy)phenyl]propionic acid (VII). (see scheme 26758001a, intermediate (VII)).
WO 0208172
The condensation of ethyl isovalerate (I) with 1,3-dichloropropene (II) by means of BuLi and DIA in THF gives 5-chloro-2-isopropyl-4-pentenoic acid ethyl ester (III), which is hydrolyzed with NaOH in ethanol to yield the corresponding racemic acid (IV). The optical resolution of (IV) is carried out by means of cinchonidine and TEA in THF to afford 5-chloro-2(S)-isopropyl-4-pentenoic acid (V), which can also be obtained by asymmetric synthesis as follows: Condensation of 4(S)-benzyl-3-(3-methylbutyryl)oxazolidin-2-one (VI) with 3-iodo-1-propenyl chloride (VII) by means of LiHMDS in THF gives 4(S)-benzyl-3-(2(S)-isopropyl-3-methylbutyryl)oxazolidin-2-one (VIII), which is hydrolyzed with LiOH in THF/water to afford the chiral pentanoic acid (V). The reaction of (V) with oxalyl chloride in toluene gives the corresponding acyl chloride (IX), which is treated with dimethylamine and pyridine in dichloromethane to yield the dimethylamide (X). The condensation of (X) with the chiral chloro derivative (XI) (obtained by reaction of the corresponding alcohol (XII) with CCl4 and trioctylphosphine) by means of Mg and 1,2-dibromoethane in THF affords the octenamide (XIII). The cyclization of (XIII) by means of phosphoric acid and simultaneous bromination with NBS in THF provides the chiral bromolactone (XIV), which is opened by means of dimethylamine and Et2AlCl in dichloromethane to give the chiral 5-bromo-4-hydroxy-2,7-diisopropyloctanamide (XV). The reaction of (XV) with acetic anhydride and pyridine in dichloromethane yields the acetoxy derivative (XVI), which is treated with LiN3 to afford the 5(S)-azido derivative (XVII).
The cyclization of (XVII) by means of TsOH in refluxing methanol gives the chiral lactone (XVIII), which is condensed with 3-amino-2,2-dimethylpropionamide (XIX) by means of TEA and 2-hydroxypyridine at 90 C to yield the corresponding amide (XX). Finally, the azido group of (XX) is reduced with H2 over Pd/C in tert-butyl methyl ether to afford the target Aliskiren.
WO 0202508
The condensation of the chiral chloro derivative (I) with 5-chloro-[2(S)-isopropyl]-4-pentanoic acid methyl ester (II) by means of Mg and dibromoethane in THF gives the chiral octenoic ester (III) which is converted to the corresponding acid (IV) by means of LiOH in THF/methanol/water. The reaction of (IV) with NBS in dichloromethane yields the bromolactone (V), which is treated with LiOH in isopropanol to yield the epoxide (VI). This compound, without isolation, is treated with HCl in the same solvent to afford the chiral hydroxylactone (VII). The reaction of the OH group of (VII) with MsCl and pyridine in toluene provides the mesylate (VIII), which is treated with NaN3 in hot 1,3-dimethylperhydropyrimidin-2-one to give the azido derivative (IX). The condensation of (IX) with 3-amino-2,2-dimethylpropionamide (X) by means of 2-hydroxypyridine in hot TEA yields the carboxamide (XI). Finally, the azido group of (XI) is reduced with H2 over Pd/C in tert-butyl methyl ether to provide the target Aliskiren.
Tetrahedron Lett 2000,41(51),10085
The intermediate gamma-butyrolactone (XXVIII) has been obtained as follows: Allylation of the imidazolidinone intermediate (V) with allyl bromide (XXI) and LiHMDS in THF gives the chiral intermediate (XXII), which by dihydroxylation and cleavage of the chiral auxiliary with OsO4 and NMMO in tert-butanol/acetone/water yields the lactone alcohol (XXIII). Oxidation of (XXIII) with NaIO4 and RuCl3 in CCl4/acetonitrile/water affords the carboxylic acid (XXIV), which by treatment with (COCl)2 in toluene provides the acyl chloride (XXV). Esterification of (XXV) with benzyl alcohol gives the corresponding benzyl ester as a diastereomeric mixture, from which the desired isomer (XXVI) is separated by flash chromatography. Hydrogenolysis of the benzyl ester (XXVI) with H2 over Pd/C in ethyl acetate yields the carboxylic acid (XXVII), which is treated with oxalyl chloride in toluene to afford the desired gamma-butyrolactone intermediate (XXVIII).
  1. Gradman A, Schmieder R, Lins R, Nussberger J, Chiang Y, Bedigian M (2005). “Aliskiren, a novel orally effective renin inhibitor, provides dose-dependent antihypertensive efficacy and placebo-like tolerability in hypertensive patients”. Circulation 111 (8): 1012–8.doi:10.1161/01.CIR.0000156466.02908.EDPMID 15723979.
  2.  Straessen JA, Li Y, and Richart T (2006). “Oral Renin Inhibitors”Lancet 368 (9545): 1449–56. doi:10.1016/S0140-6736(06)69442-7PMID 17055947.
  3. “First Hypertension Drug to Inhibit Kidney Enzyme Approved”CBC. 2007-03-06. Retrieved 2007-03-14.[dead link]
  4. Healthzone.ca: Blood-pressure drug reviewed amid dangerous side effects
  5.  Parving, Hans-Henrik; Barry M. Brenner, M.D., Ph.D., John J.V. McMurray, M.D., Dick de Zeeuw, M.D., Ph.D., Steven M. Haffner, M.D., Scott D. Solomon, M.D., Nish Chaturvedi, M.D., Frederik Persson, M.D., Akshay S. Desai, M.D., M.P.H., Maria Nicolai
  6. Alkylation of 3-hydroxy-4-methoxybenzyl alcohol (I) with 1-bromo-3-methoxypropane (II) gives ether (III). Subsequent conversion of benzyl alcohol (III) into bromide (IV) is carried out using bromotrimetylsilane. The chiral isovaleryloxazolidinone (V) is alkylated with bromide (IV) by means of LiHMDS to afford (VI), which is hydrolyzed to the (S)-2-aryl-2-isopropylpropionic acid (VII) by means of lithium peroxide. The reduction of acid (VII) to the corresponding alcohol with NaBH4/I2 reagent, followed by treatment with PPh3 and NBS, provides bromide (VIII). Alkylation of the chiral dimethoxydihydropyrazin (IX) with bromide (VIII) produces (X). Further hydrolysis of the pyrazine ring of (X) with HCl, followed by Boc protection of the resulting (S,S)-amino ester, yields compound (XI). Reduction of the ester group of (XI) with DIBAL gives aldehyde (XII). This compound is condensed with the Grignard reagent (XIII) to afford the diastereomeric mixture of amino alcohols (XIV). Treatment of mixture (XIV) with 2,2-dimethoxypropane (XV) and TsOH produces a mixture of oxazolidines, from which the required (S,S,S)-isomer (XVI) is isolated by flash chromatography. Hydrogenolitic deprotection of the benzyl ether of (XVI) gives alcohol (XVII).des, M.D., Alexia Richard, M.Sc., Zhihua Xiang, Ph.D., Patrick Brunel, M.D., and Marc A. Pfeffer, M.D., Ph.D. for the ALTITUDE Investigators (2012). “Cardiorenal End Points in a Trial of Aliskiren for Type 2 Diabetes”NEJM 367 (23): 2204–13. doi:10.1056/NEJMoa1208799PMID 23121378.
  7. J “Chemistry & Biology : Structure-based drug design: the discovery of novel nonpeptide orally active inhibitors of human renin”. ScienceDirect. Retrieved 2010-01-20.
  8.  Baldwin CM, Plosker GL.[1]doi:10.2165/00003495-200969070-00004. Drugs 2009; 69(7):833-841.
  9.  Ingelfinger JR (June 2008). “Aliskiren and dual therapy in type 2 diabetes mellitus”N. Engl. J. Med. 358 (23): 2503–5.doi:10.1056/NEJMe0803375PMID 18525047.
  10.  PharmaXChange: Direct Renin Inhibitors as Antihypertensive Drugs
  11.  Parving HH, Persson F, Lewis JB, Lewis EJ, Hollenberg NK. “Aliskiren Combined with Losartan in Type 2 Diabetes and Nephropathy,” N Engl J Med 2008;358:2433-46.
  12.  Drugs.com: Tekturna
  13.  Cardiorenal end points in a trial of aliskiren for type 2 diabetes, N Engl J MED. 2012;367(23):2204-2213
  14. European Medicines Agency recommends new contraindications and warnings for aliskiren-containing medicines.

Drugs Fut2001, 26, (12): 1139

Tetrahedron Lett 2001, 42: 4819-23.

Tetrahedron Lett2000, 41, (51): 10085

EP 0678500; EP 0678503; JP 1996053434; JP 1996081430; US 5559111; US ​​5627182; US 5646143, WO 0109079; WO 0109083

Aliskiren
Aliskiren Structural Formulae V.1.svg
Systematic (IUPAC) name
(2S,4S,5S,7S)-5-amino-N-(2-carbamoyl-2,2-dimethylethyl)-4-hydroxy-7-{[4-methoxy-3-(3-methoxypropoxy)phenyl]methyl}-8-methyl-2-(propan-2-yl)nonanamide
Clinical data
AHFS/Drugs.com monograph
MedlinePlus a607039
Licence data EMA:Link, US FDA:link
Pregnancy
category
  • C in first trimester
    D in second and third trimesters
Legal status
Routes of
administration
PO (oral)
Pharmacokinetic data
Bioavailability Low (approximately 2.5%)
Metabolism Hepatic, CYP3A4-mediated
Biological half-life 24 hours
Excretion Renal
Identifiers
CAS Number 173334-57-1 Yes
ATC code C09XA02
C09XA52 (with HCT)
PubChem CID: 5493444
IUPHAR/BPS 4812
DrugBank DB01258 Yes
ChemSpider 4591452 
UNII 502FWN4Q32 Yes
KEGG D03208 Yes
ChEBI CHEBI:601027 
ChEMBL CHEMBL1639 
Chemical data
Formula C30H53N3O6
Molecular mass 551.758 g/mol

STR1

 

SEE……..http://newdrugapprovals.org/2016/01/18/aliskiren/

////

O=C(N)C(C)(C)CNC(=O)[C@H](C(C)C)C[C@H](O)[C@@H](N)C[C@@H](C(C)C)Cc1cc(OCCCOC)c(OC)cc1

Share

PITAVASTATIN

 GENERIC, Uncategorized  Comments Off on PITAVASTATIN
Dec 162013
 

 

PITAVASTATIN, LIVALO, Itavastatin calcium, Nisvastatin, NKS-104, NK-104,

 

(3R, 5S) -7 – [2-Cyclopropyl-4-(4-fluorophenyl) quinolin-3-yl] -3,5-dihydroxy-6 (E)-heptenoic acid calcium salt (2:1)

CAS REGISTRY NUMBER

147526-32-7  CA SALT, 147511-69-1 (free acid), 141750-63-2 (lactone), 192565-91-6 (monoK salt)

rotation is +

alpha(D20) +6.8° (c 1.74, CHCl3)

ALSO

Bioorganic and Medicinal Chemistry Letters, 1999 ,  VOL 9,  20  pg. 2977 – 2982…….alpha(D20) +23.1° (c 1.0, acn/water(1:))

Helvetica Chimica Acta, 2007 ,  vol. 90, 6  pg. 1069 – 1081…alpha(D20) +22.9° (c 1.0, acn/water)

(3R,5S,6E)-7-[2-cyclopropyl-4-(4-fluorophenyl)quinolin-3-yl]-3,5-dihydroxyhept-6-enoic acid

Pitavastatin a lipid-lowering agent that belongs to the statin class of medications for treatment of dyslipidemia. It is also used for primary and secondary prevention of cardiovascular disease. FDA approved in Aug 3, 2009.

2-C25-H23-FN-O4.Ca, 881.01

 
Nissan Chemical (Originator), Kowa (Licensee), Novartis (Licensee), Recordati (Licensee), Sankyo (Licensee)
 
Lipoprotein Disorders, Treatment of, METABOLIC DRUGS, APOA1 Expression Enhancers, HMG-CoA Reductase Inhibitors, SPP1 (Osteopontin) Expression Inhibitors
 
Launched-2003

Statin drugs are currently the most therapeutically effective drugs available for reducing the level of Low density lipoprotein (LDL) in the blood stream of a patient at risk for cardiovascular disease. A high level of LDL in the
bloodstream has been linked to the formationof coronary lesions which obstruct the flow of blood and can rupture and promote thrombosis. It is well known that inhibitors against HMG CoA reductase which is rate limiting enzyme for cholesterol biosynthesis  have been clinically proved to be potentially useful anti-hyperlipoproteinemic agents
and they are considered very effective curative and preventive for coronary artery sclerosis or atherosclerosis .
Pitavastatin calcium was  discovered by Nissan Chemical Industries Limited  Japan and developedfurther by Kowa Pharmaceuticals Tokyo Japan is a novel member of the medication class of statins.

LIVALO (pitavastatin) is an inhibitor of HMG-CoA reductase. It is a synthetic lipid-lowering agent for oral administration.

The chemical name for pitavastatin is (+)monocalcium bis{(3R, 5S, 6E)-7-[2-cyclopropyl-4-(4-fluorophenyl)-3-quinolyl]-3,5dihydroxy-6-heptenoate}. The structural formula is:

 

 

LIVALO (pitavastatin) Structural Formula Illustration

 

The empirical formula for pitavastatin is C50H46CaF2N2O8 and the molecular weight is 880.98. Pitavastatin is odorless and occurs as white to pale-yellow powder. It is freely soluble in pyridine, chloroform, dilute hydrochloric acid, and tetrahydrofuran, soluble in ethylene glycol, sparingly soluble in octanol, slightly soluble in methanol, very slightly soluble in water or ethanol, and practically insoluble in acetonitrile or diethyl ether. Pitavastatin is hygroscopic and slightly unstable in light.

Each film-coated tablet of LIVALO contains 1.045 mg, 2.09 mg, or 4.18 mg of pitavastatin calcium, which is equivalent to 1 mg, 2 mg, or 4 mg, respectively of free base and the following inactive ingredients: lactose monohydrate, low substituted hydroxypropylcellulose, hypromellose, magnesium aluminometasilicate, magnesium stearate, and film coating containing the following inactive ingredients: hypromellose, titanium dioxide, triethyl citrate, and colloidal anhydrous silica.

 

Pitavastatin (usually as a calcium salt) is a member of the blood cholesterol loweringmedication class of statins,[1] marketed in the United States under the trade nameLivalo. Like other statins, it is an inhibitor of HMG-CoA reductase, the enzyme that catalyses the first step of cholesterol synthesis. It has been available in Japan since 2003, and is being marketed under licence in South Korea and in India.[2] It is likely that pitavastatin will be approved for use in hypercholesterolaemia (elevated levels of cholesterol in the blood) and for the prevention of cardiovascular disease outside South and Southeast Asia as well.[3] In the US, it received FDA approval in 2009.[4]

Pitavastatin is used to lower serum levels of total cholesterol, LDL-C, apolipoprotein B, and triglycerides, and raise levels of HDL-C for the treatment of dyslipidemia.

Like the other statins, pitavastatin is indicated for hypercholesterolaemia (elevated cholesterol) and for the prevention of cardiovascular disease. A 2009 study showed that pitavastatin increased HDL cholesterol (24.6%), especially in patients with HDL lower than 40 mg/dl, in addition to greatly reducing LDL cholesterol (–31.3%).[5] As a consequence, pitavastatin is most likely to be appropriate for patients with metabolic syndrome with high LDL, low HDL and diabetes mellitus.

Common statin-related side effects (headaches, stomach upset, abnormal liver function tests and muscle cramps) were similar to other statins. However, pitavastatin seems to lead to fewer muscle side effects than certain statins that are lipid-soluble, as a result of the fact that pitavastatin is water-soluble (as is pravastatin, for example).[6] One study found that coenzyme Q10 was not reduced as much as with certain other statins (though this is unlikely given the inherent chemistry of the HMG-CoA reductase pathway that all statin drugs inhibit).[3][7]

Hyperuricemia or increased levels of serum uric acid have been reported with pitavastatin.[8]

Most statins are metabolised in part by one or more hepatic cytochrome P450enzymes, leading to an increased potential for drug interactions and problems with certain foods (such as grapefruit juice). Pitavastatin appears to be a substrate ofCYP2C9, and not CYP3A4 (which is a common source of interactions in other statins). As a result, pitavastatin is less likely to interact with drugs that are metabolized via CYP3A4, which might be important for elderly patients who need to take multiple medicines.[3]

Pitavastatin (previously known as itavastatin, itabavastin, nisvastatin, NK-104 or NKS-104) was discovered in Japan by Nissan Chemical Industries and developed further byKowa PharmaceuticalsTokyo.[3] Pitavastatin was approved for use in the United States by the FDA on 08/03/2009 under the trade name Livalo. Pitavastatin has been also approved by the Medicines and Healthcare products Regulatory Agency (MHRA) in UK on 17 August 2010.

  1.  Kajinami, K; Takekoshi, N; Saito, Y (2003). “Pitavastatin: efficacy and safety profiles of a novel synthetic HMG-CoA reductase inhibitor”.Cardiovascular drug reviews 21 (3): 199–215. PMID 12931254edit
  2.  Zydus Cadila launches pitavastatin in India
  3. Mukhtar, R. Y. A.; Reid, J.; Reckless, J. P. D. (2005). “Pitavastatin”. International Journal of Clinical Practice 59 (2): 239–252.doi:10.1111/j.1742-1241.2005.00461.xPMID 15854203edit
  4.  The Seventh Statin; Pitavastatin
  5.  http://www.ncbi.nlm.nih.gov/pubmed/19907105
  6.  ScienceDaily (11 May 2013). “Alternative Cholesterol-Lowering Drug for Patients Who Can’t Tolerate Statins”ScienceDaily.
  7.  Clin Pharmacol Ther. 2008 May;83(5):731-9. Epub 2007 Oct 24. Comparison of effects of pitavastatin and atorvastatin on plasma coenzyme Q10 in heterozygous familial hypercholesterolemia: results from a crossover study. Kawashiri MA, Nohara A, Tada H, Mori M, Tsuchida M, Katsuda S, Inazu A, Kobayashi J, Koizumi J, Mabuchi H, Yamagishi M.
  8.  Ogata, N.; Fujimori, S.; Oka, Y.; Kaneko, K. (2010). “Effects of Three Strong Statins (Atorvastatin, Pitavastatin, and Rosuvastatin) on Serum Uric Acid Levels in Dyslipidemic Patients”. Nucleosides, Nucleotides and Nucleic Acids 29 (4–6): 321.doi:10.1080/15257771003741323edit

Thumb

Country
Patent Number
Approved
Expires (estimated)
United States 7,022,713 2009-08-03 2024-02-19
United States 6,465,477 2009-08-03 2016-12-20
United States 5,856,336 2009-08-03 2016-01-05
United States 5,854,259 2009-08-03 2015-12-29
United States 5,753,675 2009-08-03 2015-05-19

JP 1993310700, JP 1994025092

Tetrahedron Lett 1993, 34, 51, 8263-6.

Bioorg Med Chem2001, 9, (10): 2727

Drugs Fut1998, 23, (8) :847-859

Bull Chem Soc Jpn1995, 68, (1) :364-72

Tetrahedron Asymmetry1993, 4, (2) :201-4

Tetrahedron Lett1993, 34, (51) :8267-70

A Endo; J. Med. Chem. 1985, 28, 401.
AM Gotta; LC Smith; IXth International Symp. Drugs Affecting Lipid Metabolism, 1986, 30- 31
Y Fujikawa, Nissan Chemical Industries Ltd, EP304063 (A3), 1989.
S Ahmed; CS Madsen; PD Stein; J. Med. Chem. 2008, 51, 2722-2733.
K Turner; Org. Process. Res. Dev, 2004, 8, 823-833.
Z Casar; M Steinbocher; J Kosmrlj; J. Org. Chem. 2010, 75(19), 6681-6684.
KL Baumann; Tetrahedron Letters, 1992, 33, 2283-2284.
N Miyachi; Y Yanagawa; H Iwasaki; Tetrahedron Lett. 1993, 34, 8267-8270.
T Minami; K Takahashi; T Hiyama; Tetrahedron Lett. 1993, 34, 3, 513-516.
DA Evans; AH Hoveyda; J. Org. Chem. 1990, 55, 5190-5192.
J Castorer; LA Sorbera; PA Leeson; Drugs Fut. 23(8), 1998, 847-859.
T Hiyama; K Takahashi; T Minami; Bull. Chem. Soc. Jpn. 1995, 68, 364-372.
MS Reddy; M Bairy; K Reddy; Oriental Journal of Chemistry. 2007, 23, 559-564.
RN Moore; G Bigam; JK Chan; AM Hogg; JC Vederas; J. Am. Chem. Soc. 1985, 107 3694-3701.
DS Johnson; JJ Li; Art of Drug Synthesis, John Wiley & Sons, New Jersey, 2007, 177-181.
MT Stone; Organic Lett. 2011, 13, 2326-2329.
SR Manne, SR Maramreddy, WO2007132482 (A2), 2007.
SD Dwivedi, DJ Patel, AP Shah, Cadila Healthcare Ltd, US0022102 (A1), 2012.

 

……………………………………………………..

 

……………………………

The reaction of 1 (R) ,7,7-trimethylbicyclo [2.2.1] heptan-2-one (I) with 1 -naphthylmagnesium bromide (II) gives the tertiary alcohol (III), which by reaction with SOCl2 and then with NaHCO3 yields 2 – (1-naphthyl) -1 (R) ,7,7-trimethylbicyclo [2.2.1] heptene (IV ). Hydroboration of (IV) with BH3 followed by oxidation with H2O2 affords 4 (S) ,7,7-trimethyl-3exo-(1-naphthyl) bicyclo [2.2.1] heptan-2exo-ol (V), which is submitted to transesterification with methyl acetoacetate (VI) and dimethyl-aminopyridine (DMAP) to give the corresponding ester (VII). The condensation of (VII) with N-methoxy-N-methyl-3-[2-cyclopropyl-4-( 4-fluorophenyl) quinolin-3-yl] -2 (E)-propenamide (VIII) by means of NaH yields the corresponding chiral 3,5-dioxoheptenoic acid ester (IX), which is selectively reduced first with diisobutylaluminum hy-dride acid (DIBAL) and then with diethylmethoxyborane and sodium borohydride affording the 3 (R), 5 (S)-dihydroxyheptenoic ester (X). Finally, this compound is saponified with NaOH and treated with acetic acid / sodium acetate. The intermediate amide (VIII ) is obtained by condensation of 2-cyclopropyl-4-(4-fluorophenyl) quinoline-3-carbaldehyde (XI) with N-methoxy-N-methylacetamide (XII) by means of butyllithium to the hydroxy propionamide (XIII), which is then dehydrated with methanesulfonyl chloride and triethylamine in the usual way).

…………………

 

 

A systematic chiral synthesis of NK-104 and its enantiomer (X) has been reported: The oxidation of the already known 2-cyclopropyl-4-(4-fluorophenyl)quinoline-3-methanol (I) with DMSO, P2O5 and triethylamine gives the corresponding aldehyde (II), which is condensed with diethyl cyanomethylphosphonate by means of NaOH in toluene yielding the propenenitrile (III). The reduction of (III) with DIBAL affords the unsaturated aldehyde (IV), which is condensed with ethyl acetoacetate by means of NaH and n-BuLi to provide the 3-oxo-5-hydroxy-6-heptenoic acid ethyl ester derivative (V). The highly syn stereoselective reduction of (V) by means of diethylmethoxyborane and NaBH4 yields the desired syn racemic mixture of erythro-beta,delta-dihydroxyesters (VII), which is submitted to optical resolution with chiral (+)-alpha-methylbenzylamine [(+)-MBA] to obtain NK-104 free acid (VIII), which is finally treated with NaOH and CaCl2. The enantiomer of NK-104 has been obtained by optical resolution of the racemic mixture (VII) with (-)-alpha-methylbenzylamine to obtain the enantiomeric free acid (IX), which is treated with NaOH and CaCl2 as before.

 

Fujikawa, Y.; Suzuki, M.; Iwasaki, H.; Kitahara, M.; Sakashita, M.; Sakoda, R.;. Synthesis and biological evaluations of quinolone-based HMG-CoA reductase inhibitors Bioorg Med Chem 2001, 9 , 10, 2727

 

………

 

 

 

ADDITIONAL UPDATED INFO

Pitavastatin calcium is a novel member of the medication class of statins. Marketed in the United States under the trade name Livalo, it is like other statin drugs an inhibitor of HMG-CoA reductase, the enzyme that catalyses the first step of cholesterol synthesis. It is likely that pitavastatin will be approved for use in hypercholesterolaemia (elevated levels of cholesterol in the blood) and for the prevention of cardiovascular disease outside South and Southeast Asia as well.

Pitavastatin calcium is chemically known as (3R,5S)-7-[2-cyclopropyl-4-(4-fluorophenyl)quinolin-3-yl]-3,5-dihydroxy-6(E)-heptenoic acid calcium salt having the formula IA is known in the literature.

 

Figure US20120022102A1-20120126-C00001

 

Pitavastatin is a synthetic lipid-lowering agent that acts as an inhibitor of 3-hydroxy-3-methylglutaryl-coenzyme a (HMG-CoA) reductase (HMG-CoA Reductase inhibitor). This enzyme catalyzes the conversions of HMG-CoA to mevalonate, inhibitors are commonly referred to as “statins”. Statins are therapeutically effective drugs used for reducing low density lipoprotein (LDL) particle concentration in the blood stream of patients at risk for cardiovascular disease. Pitavastatin is used in the treatment of hyperchloesterolemia and mixed dyslipidemia.

Pitavastatin calcium has recently been developed as a new chemically synthesized and powerful statin by Kowa Company Ltd, Japan. On the basis of reported data, the potency of Pitavastatin is dose-dependent and appears to be equivalent to that of Atorvastatin. This new statin is safe and well tolerated in the treatment of patients with hypercholesterolaemia. Significant interactions with a number of other commonly used drugs can be considered to be extremely low.

Pitavastatin was disclosed for the first time in US patents US 4,761,419, US 5,01 1 ,930 and US 5,753,675. The process disclosed in these patents for the preparation of Pitavastatin is as shown below:

 

Figure imgf000003_0001

wherein R is hydrogen or protecting group.

US 5,284,953 discloses a process for the preparation of Pitavastatin calcium, which employs optically active a-methylbenzylamine as a resoluting agent.

The above processes are economically not viable, as resolution is carried out in final stage.

US 6,835,838 B2 discloses a process for the preparation of Pitavastatin calcium, which is as shown below:

 

Figure imgf000003_0002
Figure imgf000004_0001

However, it has been observed that the above process of lactonization results in ~10- 15% of unreacted Pitavastatin ethyl ester and therefore results in low yield. Further, -10% of Pitavastatin acid results during the above lactonization process and therefore does not produce a single product which is required to keep adequate control for an intermediate through specifications to have consistently better quality of the finished product.

Processes for the preparation of Pitavastatin are described in EP-A-0304063 and EP-A-1099694 and in the publications by N. Miyachi et al. in Tetrahedron Letters (1993) vol. 34, pages 8267-8270 and by K. Takahashi et al. in Bull. Chem. Soc. Japan (1995) Vol. 68, 2649-2656. These publications describe the synthesis of Pitavastatin in great detail but do not describe the hemi-calcium salt of Pitavastatin. The publications by L A. Sorbera et al. in Drugs of the Future (1998) vol. 23, pages 847-859 and by M. Suzuki at al. in Bioorganic & Medicinal Chemistry Letters (1999) vol. 9, pages 2977-2982 describe Pitavastatin calcium, however, a precise procedure for its preparation is not given. A full synthetic procedure for the preparation of Pitavastatin calcium is described in EP-A-0520406. In the process described in this patent Pitavastatin calcium is obtained by precipitation from an aqueous solution as a white crystalline material with a melting point of 190-192° C.

US20090182008 A1 discloses polymorphic form A, B, C, D, E, and F, and the amorphous form of Pitavastatin Calcium salt (2:1). In particular, crystalline Form A having water content from about. 5% to about 15% and process for its preparation are disclosed.

US20090176987 A1 also discloses polymorphic form crystal form A of Pitavastatin Calcium which contains from 5 to 15% of water and which shows, in its X-ray powder diffraction as measured by using CuKa radiation, a peak having a relative intensity of more than 25% at a diffraction angle (20) of 30.16°.

WO2007/132482 A1 discloses a novel process for the preparation of Pitavastatin Calcium by condensing bromide salt of formula-3 with aldehyde compound of formula-4 to obtain olefinic compound of formula-5 and converting olefinic compound to Pitavastatin Calcium via organic amine salt for purification.

Pitavastatin and its process were disclosed in U.S. Pat. No. 5,753,675.

Pitavastatin calcium and its process were disclosed in U.S. Pat. No. 5,856,336. PCT publication no. WO 2004/072040 (herein after referred to ‘040 patent) disclosed crystalline polymorph A, polymorph B, polymorph C, polymorph D, polymorph E, polymorph F and amorphous form of pitavastatin calcium

  • Synthesis of pitavastatin via cross-coupling reaction is disclosed inTetrahedron Lett. 1993, 34, 8263-8266, and in Tetrahedron Lett. 1993, 34, 8267-8270.
  • A method for the preparation of pitavastatin via epichlorohydrin is described in Tetrahedron: Asymmetry 1993, 4, 201-204.
  • Synthesis of pitavastatin heterocycle and pitavastatin molecule assembly via aldol condensation reaction is disclosed in Bioorg. Med. Chem. Lett. 1999, 9, 2977-2982, and Bioorg. Med. Chem. 2001, 9, 2727-2743:

    Figure imgb0010
    Figure imgb0011
  • PCT application WO 2003/064382 describes a method for preparation of pitavastatin by asymmetric aldol reaction, in which titanium complex is used as a catalyst.
  • HWE route to pitavastatin by utilization of 3-formyl substituted pitavastatin heterocycle is disclosed in Helv. Chim. Acta 2007, 90, 1069-1081:

  • Methods for preparation of pitavastatin heterocycle derivatives are described in Bull. Chem. Soc. Jpn. 1995, 68, 364-372, Heterocycles 1999, 50, 479-483, Lett. Org. Chem. 2006, 3, 289-291, and in Org. Biomol. Chem. 2006, 4, 104-110, as well as in the international patent applications WO 95/11898 and WO 2004/041787 
  • WO 95/11898 and Bull. Chem. Soc. Jpn. 1995, 68, 364-372 disclose synthesis of PTVBR from PTVOH with PBr3:

    Figure imgb0013

 

 

WO 1995/1 1898 Al discloses a process for the preparation of Pitavastatin, which is as shown below:

 

Figure imgf000005_0001

wherein Y represents P+RnRi2Ri3Hal or P(W)Ri4R15; R9a, R% and R]0 are protecting groups each of Rn, Rj2> R^, Ri4 and R15 which are independent of one another, is optionally substituted alkyl or optionally substituted aryl group; R14 and Rj5 together form a 5- or 6-membered ring; Hal is chlorine, bromine or iodine; and W is O or S.

The above process results in 2-5% of Cis isomer of Pitavastatin which requires further purification and therefore results poor yield.

US 6,875,867 B2 discloses a process for the preparation of Pitavastatin arginine salt, which is as shown below:

 

Figure imgf000005_0002

Saponification / Base

 

Figure imgf000006_0001

During the above process Trifluoroacetic acid or hydrochloric acid is used to break the acetonide and the Pitavastatin ester formed is converted in situ to its corresponding alkali salt by treating with base, such as sodium hydroxide.

US20090182008 A1 discloses polymorphic form A, B, C, D, E, and F, and the amorphous form of Pitavastatin Calcium salt (2:1). In particular, crystalline Form A having water content from about. 5% to about 15% and process for its preparation are disclosed.

 

……………………………………..

nmr

http://scholarsresearchlibrary.com/dpl-vol4-iss5/DPL-2012-4-5-1553-1557.pdf

calcium bis-(E)-3,5-dihydroxy-7-[4’-(4’’-flurophenyl)-2’-
cyclopropyl-quinoline-3-yl]-hept-6-enoate , pitavastatin calcium
Melting Point: 207 degC;

IR υmax (KBr) cm-1: 3366 (OH), 2911, 1603 (C=O), 1567 (C=N), 1513 (C=C),

1488 (C-H), 1416 (C-H), 1313, 1275, 1221 (C-O-C), 1158, 1065 (C-H), 972, 843, 763.

1H-NMR (500MHz, DMSO-d6):

δ 1.01 (m, 2H), 1.09 (m, 1H), 1.19 (m, 2H), 1.41 (m, 1H),

1.98 (dd, 1H, J1 =8.5,
J2 =15.5Hz), 2.11(d, 1H, J1 =3.0, J2 =15.5Hz), 2.50 (m, 2H),

3.66 (m, 1H), 4.13 (m, 1H), 4.95 (s, 1H), 5.58 (dd, 1H,
J1 =5.5, J2 =10.5Hz), 6.49 (d, 1H, J = 16.0Hz),

7.35 (m, 6H), 7.59 (m, 1H, J = 7.0Hz), 7.83 (d, 1H, J =8.5Hz).

 

13CNMR & DEPT (125.76MHz, DMSO-d6):

δ 11.12(CH2, C-17), 11.23(CH2,C-18), 15.80(CH2, C-16), 44.29(CH2,
C-22), 44.61(CH2, C-24), 66.61(C-O, C-23),

69.34(C-O,C-21),115.53(C=C, C-20), 15.62(CH), 115.79(CH),
123.59(CH), 126.07(C=C, C-19),

128.79(CH),129.20(CH),130.07(CH), 32.30(CH),

132.56(CH), 133.51(C),
142.60(C), 144.09(C), 146.37(C),

161.02(C), 163.00(C), 179.13(C=O, C-25).

ESI-MS: m/z (%) 318 (100), 274 (23), 423 (13), 422 (M+, 70); EI calcd for C25H24FNO4, 421.461; found, 422.220
(M+).

…………………

………………

 

 

Share
Follow

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

Join other followers: