Quantcast
Channel: DR ANTHONY MELVIN CRASTO Ph.D – New Drug Approvals
Viewing all 1640 articles
Browse latest View live

Elpamotide

$
0
0

STR1

STR1

Elpamotide str drawn bt worlddrugtracker

Elpamotide

L-Arginyl-L-phenylalanyl-L-valyl-L-prolyl-L-alpha-aspartylglycyl-L-asparaginyl-L-arginyl-L-isoleucine human soluble (Vascular Endothelial Growth Factor Receptor) VEGFR2-(169-177)-peptide

MF C47 H76 N16 O13
Molecular Weight, 1073.2164
L-Isoleucine, L-arginyl-L-phenylalanyl-L-valyl-L-prolyl-L-α-aspartylglycyl-L-asparaginyl-L-arginyl-
  • 10: PN: WO2008099908 SEQID: 10 claimed protein
  • 14: PN: WO2009028150 SEQID: 1 claimed protein
  • 18: PN: JP2013176368 SEQID: 18 claimed protein
  • 1: PN: WO2009028150 SEQID: 1 claimed protein
  • 2: PN: WO2010027107 TABLE: 1 claimed sequence
  • 6: PN: WO2013133405 SEQID: 6 claimed protein
  • 8: PN: US8574586 SEQID: 8 unclaimed protein
  • 8: PN: WO2004024766 SEQID: 8 claimed sequence
  • 8: PN: WO2010143435 SEQID: 8 claimed protein

Phase III

A neoangiogenesis antagonist potentially for the treatment of pancreatic cancer and biliary cancer.

OTS-102

CAS No.673478-49-4, UNII: S68632MB2G

Company OncoTherapy Science Inc.
Description Angiogenesis inhibitor that incorporates the KDR169 epitope of vascular endothelial growth factor (VEGF) receptor 2 (KDR/Flk-1; VEGFR-2)
Molecular Target Vascular endothelial growth factor (VEGF) receptor 2 (VEGFR-2) (KDR/Flk-1)
Mechanism of Action Angiogenesis inhibitor; Vaccine
Therapeutic Modality Preventive vaccine: Peptide vaccine
  • Originator OncoTherapy Science
  • Class Cancer vaccines; Peptide vaccines
  • Mechanism of Action Cytotoxic T lymphocyte stimulants
  • 16 Jun 2015 No recent reports on development identified – Phase-II/III for Pancreatic cancer (Combination therapy) and Phase-II for Biliary cancer in Japan (SC)
  • 09 Jan 2015 Otsuka Pharmaceutical announces termination of its license agreement with Fuso Pharmaceutical for elpamotide in Japan
  • 01 Feb 2013 OncoTherapy Science and Fuso Pharmaceutical Industries complete a Phase-II trial in unresectable advanced Biliary cancer and recurrent Biliary cancer (combination therapy) in Japan (UMIN000002500)

STR1

Elpamotide str drawn bt worlddrugtracker

Elpamotide , credit kegg

Elpamotide is a neoangiogenesis inhibitor in phase II clinical trials at OncoTherapy Science for the treatment of inoperable advanced or recurrent biliary cancer. Phase III clinical trials was also ongoing at the company for the treatment of pancreas cancer, but recent progress report for this indication are not available at present.

Consisting of VEGF-R2 protein, elpamotide is a neovascular inhibitor with a totally novel mechanism of action. Its antitumor effect is thought to work by inducing strong immunoreaction against new blood vessels which provide blood flow to tumors. The drug candidate only act against blood vessels involved in tumor growth and is associated with few adverse effects.

Gemcitabine is a key drug for the treatment of pancreatic cancer; however, with its limitation in clinical benefits, the development of another potent therapeutic is necessary. Vascular endothelial growth factor receptor 2 is an essential target for tumor angiogenesis, and we have conducted a phase I clinical trial using gemcitabine and vascular endothelial growth factor receptor 2 peptide (elpamotide). Based on the promising results of this phase I trial, a multicenter, randomized, placebo-controlled, double-blind phase II/III clinical trial has been carried out for pancreatic cancer. The eligibility criteria included locally advanced or metastatic pancreatic cancer. Patients were assigned to either the Active group (elpamotide + gemcitabine) or Placebo group (placebo + gemcitabine) in a 2:1 ratio by the dynamic allocation method. The primary endpoint was overall survival. The Harrington-Fleming test was applied to the statistical analysis in this study to evaluate the time-lagged effect of immunotherapy appropriately. A total of 153 patients (Active group, n = 100; Placebo group, n = 53) were included in the analysis. No statistically significant differences were found between the two groups in the prolongation of overall survival (Harrington-Fleming P-value, 0.918; log-rank P-value, 0.897; hazard ratio, 0.87, 95% confidence interval [CI], 0.486-1.557). Median survival time was 8.36 months (95% CI, 7.46-10.18) for the Active group and 8.54 months (95% CI, 7.33-10.84) for the Placebo group. The toxicity observed in both groups was manageable. Combination therapy of elpamotide with gemcitabine was well tolerated. Despite the lack of benefit in overall survival, subgroup analysis suggested that the patients who experienced severe injection site reaction, such as ulceration and erosion, might have better survival

The vaccine candidate was originally developed by OncoTherapy Science. In January 2010, Fuso Pharmaceutical, which was granted the exclusive rights to manufacture and commercialize elpamotide in Japan from OncoTherapy Science, sublicensed the manufacturing and commercialization rights to Otsuka Pharmaceutical. In 2015, the license agreement between Fuso Pharmaceutical and OncoTherapy Science, and the license agreement between Fuso Pharmaceutical and Otsuka Pharmaceutical terminated.

WO 2010143435

US 8574586

WO 2012044577

WO 2010027107

WO 2013133405

WO 2009028150

WO 2008099908

WO 2004024766

PATENT

WO2013133405

The injectable formulation containing peptides, because peptides are unstable to heat, it is impossible to carry out terminal sterilization by autoclaving. Therefore, in order to achieve sterilization, sterile filtration step is essential. Sterile filtration step is carried out by passing through the 0.22 .mu.m following membrane filter typically absolute bore is guaranteed. Therefore, in the stage of pre-filtration, it is necessary to prepare a peptide solution in which the peptide is completely dissolved. However, peptides, since the solubility characteristics by its amino acid sequence differs, it is necessary to select an appropriate solvent depending on the solubility characteristics of the peptide. In particular, it is difficult to completely dissolve the highly hydrophobic peptide in a polar solvent, it requires a great deal of effort on the choice of solvent. It is also possible to increase the solubility by changing the pH, or depart from the proper pH range as an injectable formulation, in many cases the peptide may become unstable.
 In recent years, not only one type of peptide, the peptide vaccine formulation containing multiple kinds of peptides as an active ingredient has been noted. Such a peptide vaccine formulation is especially considered to be advantageous for the treatment of cancer.
 The peptide vaccine formulation for the treatment of cancer, to induce a specific immune response to the cancer cells, containing the T cell epitope peptides of the tumor-specific antigen as an active ingredient (e.g., Patent Document 1). Tumor-specific antigens these T-cell epitope peptide is derived, by exhaustive expression analysis using clinical samples of cancer patients, for each type of cancer, specifically overexpressed in cancer cells, only rarely expressed in normal cells It never is one which has been identified as an antigen (e.g., Patent Document 2). However, even in tumor-specific antigens identified in this way, by a variety of having the cancer cells, in all patients and all cancer cells, not necessarily the same as being highly expressed. That is, there may be a case in which the cancer in different patients can be an antigen that is highly expressed cancer in a patient not so expressed. Further, even in the same patient, in the cellular level, cancer cells are known to be a heterogeneous population of cells (non-patent document 1), another even antigens expressed in certain cancer cells in cancer cells may be the case that do not express. Therefore, in one type of T-cell epitope peptide vaccine formulations containing only, there is a possibility that the patient can not be obtained a sufficient antitumor effect is present. Further, even in patients obtained an anti-tumor effect, the cancer cells can not kill may be present. On the other hand, if the vaccine preparation comprising a plurality of T-cell epitope peptide, it is likely that the cancer cells express any antigen. Therefore, it is possible to obtain an anti-tumor effect in a wider patient, the lower the possibility that cancer cells can not kill exists.
 The effect of the vaccine formulation containing multiple types of T-cell epitope peptide as described above, the higher the more kinds of T-cell epitope peptides formulated. However, if try to include an effective amount of a plurality of types of T cell peptide, because the peptide content of the per unit amount is increased, to completely dissolve the entire peptide becomes more difficult. Further, because it would plurality of peptides having different properties coexist, it becomes more difficult to maintain all of the peptide stability.
 For example, in European Patent Publication No. 2111867 (Patent Document 3), freeze-dried preparation of the vaccine formulation for the treatment of cancer comprising a plurality of T-cell epitope peptides have been disclosed. This freeze-dried preparation, in the preparation of peptide solution before freeze drying, each peptide depending on its solubility properties, are dissolved in a suitable solvent for each peptide. Furthermore, when mixing the peptide solution prepared in order to prevent the precipitation of the peptide, it is described that mixing the peptide solution in determined order. Thus, to select a suitable solvent for each peptide, possible to consider the order of mixing each peptide solution is laborious as the type of peptide increases.

In order to avoid difficulties in the formulation preparation, as described above, a vaccine formulation comprising one type of T-cell epitope peptides, methods for multiple types administered to the same patient is also contemplated. However, when administering plural kinds of vaccine preparation, it is necessary to vaccination of a plurality of locations of the body, burden on a patient is increased. Also peptide vaccine formulation, the DTH (Delayed Type Hypersensitivity) skin reactions are often caused called reaction after inoculation. Occurrence of skin reactions at a plurality of positions of the body, increases the discomfort of the patient. Therefore, in order to reduce the burden of patients in vaccination is preferably a vaccine formulation comprising a plurality of T-cell epitope peptide. Further, even when the plurality of kinds administering the vaccine formulation comprising a single type of epitope peptides, when manufacturing each peptide formulation is required the task of selecting an appropriate solvent for each peptide.

Patent Document 1: International Publication No. WO 2008/102557
Patent Document 2: International Publication No. 2004/031413 Patent
Patent Document 3: The European Patent Publication No. 2111867
PATENT
PATENT

///////////Elpamotide, Phase III,  A neoangiogenesis antagonist, pancreatic cancer and biliary cancer, OTS-102, OncoTherapy Science Inc, peptide

CC[C@H](C)[C@@H](C(=O)O)NC(=O)[C@H](CCCNC(=N)N)NC(=O)[C@H](CC(=O)N)NC(=O)CNC(=O)[C@H](CC(=O)O)NC(=O)[C@@H]1CCCN1C(=O)[C@H](C(C)C)NC(=O)[C@H](Cc2ccccc2)NC(=O)[C@H](CCCNC(=N)N)N


Filed under: Peptide drugs, Phase3 drugs Tagged: A neoangiogenesis antagonist, Elpamotide, OncoTherapy Science Inc, OTS-102, pancreatic cancer and biliary cancer, peptide, Phase III

Oliceridine

$
0
0

TRV130.svg

Oliceridine.png

Oliceridine

N-[(3-methoxythiophen-2-yl)methyl]-2-[(9R)-9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl]ethan-1-amine

[(3-Methoxythiophen-2-yl)methyl]({2-[(9R)-9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9- yl]ethyl})amine

Phase III

A mu-opioid receptor ligand potentially for treatment of acute postoperative pain.

TRV-130; TRV-130A

CAS No.1401028-24-7

Molecular Formula: C22H30N2O2S
Molecular Weight: 386.5508 g/mol
  • Originator Trevena

Trevena, Inc.

  • Class Analgesics; Small molecules
  • Mechanism of Action Beta arrestin inhibitors; Opioid mu receptor agonists
  • Orphan Drug Status No
  • On Fast track Postoperative pain
    • Phase III Postoperative pain
    • Phase II Pain

    Most Recent Events

    • 09 Mar 2016Trevena intends to submit NDA to US FDA in 2017
    • 22 Feb 2016Oliceridine receives Breakthrough Therapy status for Pain in USA
    • 19 Jan 2016Phase-III clinical trials in Postoperative pain in USA (IV) (NCT02656875)

Oliceridine (TRV130) is an opioid drug that is under evaluation in human clinical trials for the treatment of acute severe pain. It is afunctionally selective μ-opioid receptor agonist developed by Trevena Inc. Oliceridine elicits robust G protein signaling, with potencyand efficacy similar to morphine, but with far less β-arrestin 2 recruitment and receptor internalization, it displays less adverse effectsthan morphine.[1][2][3]

In 2015, the product was granted fast track designation in the U.S. for the treatment of moderate to severe acute pain. In 2016, the compound was granted FDA breakthrough therapy designation for the management of moderate to severe acute pain.

Oliceridine (TRV130) is an intravenous G protein biased ligand that targets the mu opioid receptor. Trevena is developing TRV130 for the treatment of moderate to severe acute pain where intravenous therapy is preferred, with a clinical development focus in acute postoperative pain

TRV 130 HCl is a novel μ-opioid receptor (MOR) G protein-biased ligand; elicits robust G protein signaling(pEC50=8.1), with potency and efficacy similar to morphine, but with far less beta-arrestin recruitment and receptor internalization.

NMR

STR1

Oliceridine (TRV130) – Mu Opioid Biased Ligand for Acute Pain

Target Indication Lead
Optimization
Preclinical
Development
Phase
1
Phase
2
Phase
3
Ownership
Oliceridine (TRV130) Mu-receptor Moderate to
Severe Pain
intravenous Trevena Logo

Oliceridine (TRV130) is an intravenous G protein biased ligand that targets the mu opioid receptor. Trevena is developing TRV130 for the treatment of moderate to severe acute pain where intravenous therapy is preferred, with a clinical development focus in acute postoperative pain.

Recent TRV130 News

Opioid receptors (ORs) mediate the actions of morphine and morphine-like opioids, including most clinical analgesics. Three molecularly and pharmacologically distinct opioid receptor types have been described: δ, κ and μ. Furthermore, each type is believed to have sub-types. All three of these opioid receptor types appear to share the same functional mechanisms at a cellular level. For example, activation of the opioid receptors causes inhibition of adenylate cyclase, and recruits β-arrestin.

When therapeutic doses of morphine are given to patients with pain, the patients report that the pain is less intense, less discomforting, or entirely gone. In addition to experiencing relief of distress, some patients experience euphoria. However, when morphine in a selected pain-relieving dose is given to a pain-free individual, the experience is not always pleasant; nausea is common, and vomiting may also occur. Drowsiness, inability to concentrate, difficulty in mentation, apathy, lessened physical activity, reduced visual acuity, and lethargy may ensue.

There is a continuing need for new OR modulators to be used as analgesics. There is a further need for OR agonists as analgesics having reduced side effects. There is a further need for OR agonists as analgesics having reduced side effects for the treatment of pain, immune dysfunction, inflammation, esophageal reflux, neurological and psychiatric conditions, urological and reproductive conditions, medicaments for drug and alcohol abuse, agents for treating gastritis and diarrhea, cardiovascular agents and/or agents for the treatment of respiratory diseases and cough.

 PAPER

Structure activity relationships and discovery of a g protein biased mu opioid receptor ligand, ((3-Methoxythiophen-2-yl)methyl)a2((9R)-9-(pyridin-2-y1)-6-oxaspiro-(4.5)clecan-9-yl)ethylpamine (TRV130), for the treatment of acute severe pain
J Med Chem 2013, 56(20): 8019

Structure–Activity Relationships and Discovery of a G Protein Biased μ Opioid Receptor Ligand, [(3-Methoxythiophen-2-yl)methyl]({2-[(9R)-9-(pyridin-2-yl)-6-oxaspiro-[4.5]decan-9-yl]ethyl})amine (TRV130), for the Treatment of Acute Severe Pain

Trevena, Inc., 1018 West 8th Avenue, Suite A, King of Prussia, Pennsylvania 19406, United States
J. Med. Chem., 2013, 56 (20), pp 8019–8031
DOI: 10.1021/jm4010829
Publication Date (Web): September 24, 2013
Copyright © 2013 American Chemical Society
*Phone: 610-354-8840. Fax: 610-354-8850. E-mail: dchen@trevenainc.com.

Abstract

Abstract Image

The concept of “ligand bias” at G protein coupled receptors has been introduced to describe ligands which preferentially stimulate one intracellular signaling pathway over another. There is growing interest in developing biased G protein coupled receptor ligands to yield safer, better tolerated, and more efficacious drugs. The classical μ opioid morphine elicited increased efficacy and duration of analgesic response with reduced side effects in β-arrestin-2 knockout mice compared to wild-type mice, suggesting that G protein biased μ opioid receptor agonists would be more efficacious with reduced adverse events. Here we describe our efforts to identify a potent, selective, and G protein biased μ opioid receptor agonist, TRV130 ((R)-30). This novel molecule demonstrated an improved therapeutic index (analgesia vs adverse effects) in rodent models and characteristics appropriate for clinical development. It is currently being evaluated in human clinical trials for the treatment of acute severe pain.

http://pubs.acs.org/doi/abs/10.1021/jm4010829

[(3-Methoxythiophen-2-yl)methyl]({2-[(9R)-9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl] ethyl})amine ((R)-30)

Using a procedure described in method A, (R)-39e was converted to (R)-30 as a TFA salt. 1H NMR (400 MHz, CDCl3) δ 11.70 (brs, 1H), 9.14 (d, J = 66.6, 2H), 8.72 (d, J = 4.3, 1H), 8.19 (td,J = 8.0, 1.4, 1H), 7.70 (d, J = 8.1, 1H), 7.63 (dd, J = 7.0, 5.8, 1H), 7.22 (d, J = 5.5, 1H), 6.78 (d,J = 5.6, 1H), 4.08 (m, 2H), 3.80 (m, 4H), 3.69 (dd, J = 11.2, 8.7, 1H), 2.99 (d, J = 4.8, 1H), 2.51 (t, J = 9.9, 1H), 2.35 (m, 3H), 2.18 (td, J = 13.5, 5.4, 1H), 1.99 (d, J = 14.2, 1H), 1.82 (m, 2H), 1.65 (m, 1H), 1.47 (m, 4H), 1.14 (m, 1H), 0.73 (dt, J = 13.2, 8.9, 1H). LC-MS (API-ES) m/z = 387.0 (M + H).

Patent

WO 2012129495

http://www.google.com/patents/WO2012129495A1?cl=en

Scheme 1: Synthesis of Spirocyclic Nitrile

NCCH2C02CH3 AcOH, NH4OAc

Figure imgf000050_0001
Figure imgf000050_0002

1-5 1-6 1-7

Chiral HPLC separation n=1-2

R= phenyl, substituted phenyl, aryl,

Figure imgf000050_0003

s

Scheme 2: Converting the nitrile to the opioid receptor ligand (Approach 1)

Figure imgf000051_0001

2-4

Scheme 3: Converting the nitrile to the opioid receptor ligand (Approach 2)

Figure imgf000051_0002

1-8B 3-1 3-2 n=1-2

In some embodiments, the same scheme is applied to 1 -7 and 1 -8A. Scheme 4: Synthesis of Non-Spirocyclic Nitrile

Figure imgf000052_0001

4-1 4-2 4-3

KOH, ethylene glycol R= phenyl, substituted phenyl, aryl,

substituted aryl, pyridyl, substituted pyridyl, heat heteroaryl, substituted heteroaryl,

Figure imgf000052_0002

carbocycle, heterocycle and etc.

In some embodiments, 4-1 is selected from the group consisting of

Figure imgf000052_0003

4-1 A 4-1 B 4-1 C 4-1 D 4-1 E

Scheme 5: Synthesis of Other Spirocyclic Derived Opioid Ligands

Figure imgf000053_0001

5-1 5-2 5-3

Scheme 6: Allyltrimethylsilane Approach to Access the Quaternary Carbon Center

RMgX, or RLi

Figure imgf000053_0002

Scheme 7: N-linked Pyrrazole Opioid Receptor Ligand

Figure imgf000054_0001
Figure imgf000055_0001

[(3-Methoxythiophen-2-yl)methyl]({2-[(9R)-9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9- yl]ethyl})amine

Figure imgf000144_0001

Into a vial were added 2-[(9R)-9-(Pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl]ethan-l -amine (500 mg, 1.92 mmole), 18 mL CH2C12 and sodium sulfate (1.3 g, 9.6 mmole). The 3- methoxythiophene-2-carboxaldehyde (354 mg, 2.4 mmole) was then added, and the misture was stirred overnight. NaBH4 (94 mg, 2.4 mmole) was added to the reaction mixture, stirred for 10 minutes, and then MeOH (6.0 mL) was added, stirred l h, and finally quenched with water. The organics were separated off and evaporated. The crude residue was purified by a Gilson prep HPLC. The desired fractions collected and concentrated and lyophilized. After lyophilization, residue was partitioned between CH2C12 and 2N NaOH, and the organic layers were collected. After solvent was concentrated to half of the volume, 1.0 eq of IN HC1 in Et20 was added,and majority of solvent evaporated under reduced pressure. The solid obtained was washed several times with Et20 and dried to provide [(3-methoxythiophen-2-yl)methyl]({2-[(9R)-9-(pyridin-2- yl)-6-oxaspiro[4.5]decan-9-yl]ethyl})amine monohydrochloride (336 mg, 41% yield, m/z 387.0 [M + H]+ observed) as a white solid. The NMR for Compound 140 is described herein.

Example 15: Synthesis of [(3-methoxythiophen-2-yl)methyl]({2-[(9R)-9- (pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl]ethyl})amine (Compound 140).

Methyl 2-cyano-2-[6-oxaspiro[4.5]decan-9-ylidene]acetate (mixture of E and Z isomers)

Figure imgf000141_0001

A mixture of 6-oxaspiro[4.5]decan-9-one (13.74 g, 89.1 mmol), methylcyanoacetate (9.4 ml, 106.9 mmol), ammonium acetate (1.79 g, 26.17.mmol) and acetic acid (1.02 ml, 17.8 mmol) in benzene (75 ml) was heated at reflux in a 250 ml round bottom flask equipped with a Dean-Stark and a reflux condenser. After 3h, TLC (25%EtOAc in hexane, PMA stain) showed the reaction was completed. After cooling, benzene (50 ml) was added and the layer was separated, the organic was washed by water (120 ml) and the aqueous layer was extracted by CH2CI2 (3 x 120 ml). The combined organic was washed with sat’d NaHCCb, brine, dried and concentrated and the residual was purified by flash chromatography (340 g silica gel column, eluted by EtOAc in hexane: 5% EtOAc, 2CV; 5-25%, 14CV; 25-40%,8 CV) gave a mixture of E and Z isomers: methyl 2-cyano-2-[6- oxaspiro[4.5]decan-9-ylidene]acetate ( 18.37 g, 87.8 % yield, m/z 236.0 [M + H]+ observed) as a clear oil. -cyano-2-[9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl]acetate

Figure imgf000141_0002

A solution of 2-bromopyridine (14.4 ml, 150 mmo) in THF (75 ml) was added dropwise to a solution of isopropylmagnesium chloride (75 ml, 2M in THF) at 0°C under N2, the mixture was then stirred at rt for 3h, copper Iodide(2.59 g, 13.6 mmol) was added and allowed to stir at rt for another 30 min before a solution of a mixture of E and Z isomers of methyl 2-cyano-2-[6-oxaspiro[4.5]decan-9-ylidene]acetate (16 g, 150 mmol) in THF (60 ml) was added in 30 min. The mixture was then stirred at rt for 18h. The reaction mixture was poured into a 200 g ice/2 N HC1 (100 ml) mixture. The product was extracted with Et20 (3×300 ml), washed with brine (200 ml), dried (Na2S04) and concentrated. The residual was purified by flash chromatography (100 g silica gel column, eluted by EtOAc in hexane: 3% 2CV; 3-25%, 12 CV; 25-40% 6CV gave methyl 2-cyano-2-[9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl]acetate (15.44 g, 72% yield, m/z 315.0 [M + H]+ observed) as an amber oil .

-[9-(Pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl]acetonitrile

Figure imgf000142_0001

Ethylene glycol (300 ml) was added to methyl 2-cyano-2-[9-(pyridin-2-yl)-6- oxaspiro[4.5]decan-9-yl]acetate( 15.43 g, 49 mmol) followed by potassium hydroxide (5.5 g , 98 mmol), the resulting mix was heated to 120oC, after 3 h, the reaction mix was cooled and water (300 ml) was added, the product was extracted by Et20(3 x 400 ml), washed with water(200 ml), dried (Na2S04) and concentrated, the residual was purified by flash chromatography (340 g silica gel column, eluted by EtOAc in hexane: 3% 2CV; 3-25%, 12 CV; 25-40% 6CV to give 2-[9-(Pyridin-2-yl)-6-oxaspiro[4.5]decan-9- yl]acetonitrile (10.37 g, 82% yield, m/z 257.0 [M + H]+ observed).

-yl)-6-oxaspiro[4.5]decan-9-yl]acetonitrile

Figure imgf000142_0002

racemic 2-[9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl]acetonitrile was separated by chiral HPLC column under the following preparative-SFC conditions: Instrument: SFC-80 (Thar, Waters); Column: Chiralpak AD-H (Daicel); column temperature: 40 °C; Mobile phase: Methanol /CO2=40/60; Flow: 70 g/min; Back pressure: 120 Bar; Cycle time of stack injection: 6.0min; Load per injection: 225 mg; Under these conditions, 2-[9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl]acetonitrile (4.0 g) was separated to provide the desired isomer, 2-[(9R)-9-(Pyridin-2-yI)-6- oxaspiro[4.5]decan-9-yl]acetonitrile (2.0 g, >99.5% enantiomeric excess) as a slow- moving fraction. The absolute (R) configuration of the desired isomer was later determined by an X-ray crystal structure analysis of Compound 140. [0240] -[(9R)-9-(Pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl]ethan-l-amine

Figure imgf000143_0001

LAH (1M in Et20, 20ml, 20 mmol) was added to a solution of 2-[(9R)-9-(pyridin-2-yl)- 6-oxaspiro[4.5]decan-9-yl]acetonitrile (2.56 g, 10 mmol) in Et20 (100 ml, 0.1M ) at OoC under N2. The resulting mix was stirred and allowed to warm to room temperature. After 2 h, LCMS showed the reaction had completed. The reaction was cooled at OoC and quenched with water ( 1.12 ml), NaOH (10%, 2.24 ml) and another 3.36 ml of water. Solid was filtered and filter pad was washed with ether (3 x 20 ml). The combined organic was dried and concentrated to give 2-[(9R)-9-(Pyridin-2-yl)-6- oxaspiro[4.5]decan-9-yl]ethan-l -amine (2.44 g, 94% yield, m/z 260.6 [M + H]+ observed) as a light amber oil.

Alternatively, 2-[(9R)-9-(Pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl]ethan-l -amine was prepared by Raney-Nickel catalyzed hydrogenation.

An autoclave vessel was charged with 2-[(9R)-9-(pyridin-2-yl)-6-oxaspiro[4,5]decan-9- yl] acetonitrile and ammonia (7N solution in methanol). The resulting solution was stirred at ambient conditions for 15 minutes and treated with Raney 2800 Nickel, slurried in water. The vessel was pressurized to 30 psi with nitrogen and agitated briefly. The autoclave was vented and the nitrogen purge repeated additional two times. The vessel was pressurized to 30 psi with hydrogen and agitated briefly. The vessel was vented and purged with hydrogen two additional times. The vessel was pressurized to 85-90 psi with hydrogen and the mixture was warmed to 25-35 °C. The internal temperature was increased to 45-50 °C over 30-60 minutes. The reaction mixture was stirred at 45-50 °C for 3 days. The reaction was monitored by HPLC. Once reaction was deemed complete, it was cooled to ambient temperature and filtered through celite. The filter cake was washed with methanol (2 x). The combined filtrates were concentrated under reduced pressure at 40-45 °C. The resulting residue was co-evaporated with EtOH (3 x) and dried to a thick syrupy of 2-[(9R)-9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl]ethan-l -amine.

References

  1.  Chen XT, Pitis P, Liu G, Yuan C, Gotchev D, Cowan CL, Rominger DH, Koblish M, Dewire SM, Crombie AL, Violin JD, Yamashita DS (October 2013). “Structure-Activity Relationships and Discovery of a G Protein Biased μ Opioid Receptor Ligand, [(3-Methoxythiophen-2-yl)methyl]({2-[(9R)-9-(pyridin-2-yl)-6-oxaspiro-[4.5]decan-9-yl]ethyl})amine (TRV130), for the Treatment of Acute Severe Pain”. J. Med. Chem. 56 (20): 8019–31.doi:10.1021/jm4010829. PMID 24063433.
  2.  DeWire SM, Yamashita DS, Rominger DH, Liu G, Cowan CL, Graczyk TM, Chen XT, Pitis PM, Gotchev D, Yuan C, Koblish M, Lark MW, Violin JD (March 2013). “A G protein-biased ligand at the μ-opioid receptor is potently analgesic with reduced gastrointestinal and respiratory dysfunction compared with morphine”. J. Pharmacol. Exp. Ther. 344 (3): 708–17.doi:10.1124/jpet.112.201616. PMID 23300227.
  3.  Soergel DG, Subach RA, Sadler B, Connell J, Marion AS, Cowan C, Violin JD, Lark MW (October 2013). “First clinical experience with TRV130: Pharmacokinetics and pharmacodynamics in healthy volunteers”. J Clin Pharmacol 54(3): 351–7. doi:10.1002/jcph.207. PMID 24122908.

External links

Patent ID Date Patent Title
US2015246904 2015-09-03 Opioid Receptor Ligands And Methods Of Using And Making Same
US8835488 2014-09-16 Opioid receptor ligands and methods of using and making same
US2013331408 2013-12-12 Opioid Receptor Ligands and Methods of Using and Making Same
Oliceridine
TRV130.svg
Systematic (IUPAC) name
N-[(3-methoxythiophen-2-yl)methyl]-2-[(9R)-9-pyridin-2-yl-6-oxaspiro[4.5]decan-9-yl]ethanamine
Clinical data
Routes of
administration
IV
Legal status
Legal status
Identifiers
CAS Number 1401028-24-7
ATC code none
PubChem CID 66553195
ChemSpider 30841043
UNII MCN858TCP0
ChEMBL CHEMBL2443262
Synonyms TRV130
Chemical data
Formula C22H30N2O2S
Molar mass 386.55 g·mol−1

////////TRV-130; TRV-130A, Oliceridine, Phase III, Postoperative pain, trevena, mu-opioid receptor ligand, fast track designation, breakthrough therapy designation

COc1ccsc1CNCC[C@]2(CCOC3(CCCC3)C2)c4ccccn4


Filed under: Breakthrough Therapy Designation, FAST TRACK FDA, Phase3 drugs Tagged: Breakthrough Therapy Designation, Fast Track Designation, mu-opioid receptor ligand, Oliceridine, Phase III, Postoperative pain, trevena, TRV-130; TRV-130A

Galunisertib

$
0
0

Galunisertib

Phase III

A TGF-beta receptor type-1 inhibitor potentially for the treatment of myelodysplastic syndrome (MDS) and solid tumours.

LY-2157299

CAS No.700874-72-2

4-[2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl]quinoline-6-carboxamide
6-Quinolinecarboxamide, 4-[5,6-dihydro-2-(6-methyl-2-pyridinyl)-4H-pyrrolo[1,2-b]pyrazol-3-yl]-
700874-72-2
  • Molecular FormulaC22H19N5O
  • Average mass369.419 Da

Eli Lilly and Company

4-(2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)quinoline-6-carboxamide

4-(2-(6-Methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)quinolin-6-carboxamide monohydrate 

Anal. Calcd for C22H19N5O·H2O: C, 68.20; H, 5.46; N, 18.08. Found: C, 68.18; H, 5.34; N, 17.90.

1H NMR (DMSO-d6: δ) 1.74 (s, 3H), 2.63 (m, 2H), 2.82 (br s, 2H), 4.30 (t, J = 7.2 Hz, 2H), 6.93 (m, 1H), 7.37 (s, 1H), 7.41 (d, J = 4.4 Hz, 1H), 7.56 (m, 1H), 7.58 (m, 1H), 8.04, (s, 1H), 8.04 (d, J = 4.4 Hz, 1H), 8.12 (dd, J = 8.8, 1.6 Hz, 1H), 8.25 (d, J = 2.0 Hz, 1H), 8.87 (d, J = 4.4 Hz, 1H).

13C NMR (DMSO-d6: δ) 22.56, 23.24, 25.58, 48.01, 109.36, 117.74, 121.26, 122.95, 126.73, 127.16 (2C), 129.01, 131.10, 136.68, 142.98, 147.20, 148.99, 151.08, 151.58, 152.13, 156.37, 167.47.

IR (KBr): 3349, 3162, 3067, 2988, 2851, 1679, 1323, 864, 825 cm–1.

HRMS (m/z M + 1): Calcd for C22H19N5O: 370.1653. Found: 370.1662.

GalunisertibAn orally available, small molecule antagonist of the tyrosine kinase transforming growth factor-beta (TGF-b) receptor type 1 (TGFBR1), with potential antineoplastic activity. Upon administration, galunisertib specifically targets and binds to the kinase domain of TGFBR1, thereby preventing the activation of TGF-b-mediated signaling pathways. This may inhibit the proliferation of TGF-b-overexpressing tumor cells. Dysregulation of the TGF-b signaling pathway is seen in a number of cancers and is associated with increased cancer cell proliferation, migration, invasion and tumor progression.

.

  • OriginatorEli Lilly
  • DeveloperEli Lilly; National Cancer Institute (USA); Vanderbilt-Ingram Cancer Center; Weill Cornell Medical College
  • ClassAntineoplastics; Pyrazoles; Pyridines; Pyrroles; Quinolines; Small molecules
  • Mechanism of ActionPhosphotransferase inhibitors; Transforming growth factor beta1 inhibitors
    • Phase II/IIIMyelodysplastic syndromes
    • Phase IIBreast cancer; Glioblastoma; Hepatocellular carcinoma
    • Phase I/IIGlioma; Non-small cell lung cancer; Pancreatic cancer
    • Phase ICancer; Solid tumours

    Most Recent Events

    • 26 Apr 2016Eli Lilly plans a pharmacokinetics phase I trial in Healthy volunteers in United Kingdom (PO) (NCT02752919)
    • 16 Apr 2016Pharmacodynamics data from a preclinical study in Cancer presented at the 107th Annual Meeting of the American Association for Cancer Research (AACR-2016)
    • 06 Apr 2016Eli Lilly and AstraZeneca plan a phase Ib trial for Pancreatic cancer (Second-line therapy or greater, Metastatic disease, Recurrent, Combination therapy) in USA, France, Italy, South Korea and Spain (PO) (NCT02734160)

Transforming growth factor-beta (TGF-β) signaling regulates a wide range of biological processes. TGF-β plays an important role in tumorigenesis and contributes to the hallmarks of cancer, including tumor proliferation, invasion and metastasis, inflammation, angiogenesis, and escape of immune surveillance. There are several pharmacological approaches to block TGF-β signaling, such as monoclonal antibodies, vaccines, antisense oligonucleotides, and small molecule inhibitors. Galunisertib (LY2157299 monohydrate) is an oral small molecule inhibitor of the TGF-β receptor I kinase that specifically downregulates the phosphorylation of SMAD2, abrogating activation of the canonical pathway. Furthermore, galunisertib has antitumor activity in tumor-bearing animal models such as breast, colon, lung cancers, and hepatocellular carcinoma. Continuous long-term exposure to galunisertib caused cardiac toxicities in animals requiring adoption of a pharmacokinetic/pharmacodynamic-based dosing strategy to allow further development. The use of such a pharmacokinetic/pharmacodynamic model defined a therapeutic window with an appropriate safety profile that enabled the clinical investigation of galunisertib. These efforts resulted in an intermittent dosing regimen (14 days on/14 days off, on a 28-day cycle) of galunisertib for all ongoing trials. Galunisertib is being investigated either as monotherapy or in combination with standard antitumor regimens (including nivolumab) in patients with cancer with high unmet medical needs such as glioblastoma, pancreatic cancer, and hepatocellular carcinoma. The present review summarizes the past and current experiences with different pharmacological treatments that enabled galunisertib to be investigated in patients.

Company Eli Lilly and Co.
Description Transforming growth factor (TGF) beta receptor 1 (TGFBR1; ALK5) inhibitor
Molecular Target Transforming growth factor (TGF) beta receptor 1 (TGFBR1) (ALK5)
Mechanism of Action Transforming growth factor (TGF) beta 1 inhibitor
Therapeutic Modality Small molecule

Bristol-Myers Squibb and Lilly Enter Clinical Collaboration Agreement to Evaluate Opdivo (nivolumab) in Combination with Galunisertib in Advanced Solid Tumors

Bristol-Myers Squibb and Lilly

NEW YORK & INDIANAPOLIS–(BUSINESS WIRE)– Bristol-Myers Squibb Company (NYSE:BMY) and Eli Lilly and Company (NYSE:LLY) announced today a clinical trial collaboration to evaluate the safety, tolerability and preliminary efficacy of Bristol-Myers Squibb’s immunotherapy Opdivo (nivolumab) in combination with Lilly’s galunisertib (LY2157299). The Phase 1/2 trial will evaluate the investigational combination of Opdivo and galunisertib as a potential treatment option for patients with advanced (metastatic and/or unresectable) glioblastoma, hepatocellular carcinoma and non-small cell lung cancer.

Opdivo is a human programmed death receptor-1 (PD-1) blocking antibody that binds to the PD-1 receptor expressed on activated T-cells. Galunisertib (pronounced gal ue” ni ser’tib) is a TGF beta R1 kinase inhibitor that in vitro selectively blocks TGF beta signaling. TGF beta promotes tumor growth, suppresses the immune system and increases the ability of tumors to spread in the body. This collaboration will address the hypothesis that co-inhibition of PD-1 and TGF beta negative signals may lead to enhanced anti-tumor immune responses than inhibition of either pathway alone.

“Advanced solid tumors represent a serious unmet medical need among patients with cancer,” said Michael Giordano, senior vice president, Head of Development, Oncology, Bristol-Myers Squibb. “Our clinical collaboration with Lilly underscores Bristol-Myers Squibb’s continued commitment to explore combination regimens from our immuno-oncology portfolio with other mechanisms of action that may accelerate the development of new treatment options for patients.”

“Combination therapies will be key to addressing tumor heterogeneity and the inevitable resistance that is likely to develop to even the most promising new tailored therapies,” said Richard Gaynor, M.D., senior vice president, Product Development and Medical Affairs, Lilly Oncology. “To that end, having multiple cancer pathways and technology platforms will be critical in an era of combinations to ensure sustainability beyond any single asset.”

The study will be conducted by Lilly. Additional details of the collaboration were not disclosed.

About Galunisertib

Galunisertib (pronounced gal ue” ni ser’tib) is Lilly’s TGF beta R1 kinase inhibitor that in vitro selectively blocks TGF beta signaling. TGF beta promotes tumors growth, suppresses the immune system, and increases the ability of tumors to spread in the body.

Immune function is suppressed in cancer patients, and TGF beta worsens immunosuppression by enhancing the activity of immune cells called T regulatory cells. TGF beta also reduces immune proteins, further decreasing immune activity in patients

Galunisertib is currently under investigation as an oral treatment for advanced/metastatic malignancies, including Phase 2 evaluation in hepatocellular carcinoma, myelodysplastic syndromes (MDS), glioblastoma, and pancreatic cancer.

PATENT

WO 2004048382

The disclosed invention also relates to the select compound of Formula II:

Figure imgf000005_0001

Formula II

2-(6-methyl-pyridin-2-yI)-3-[6-amido-quinolin-4-yl)-5,6-dihydro-4H-pyrrolo[l,2- bjpyrazole and the phannaceutically acceptable salts thereof.

The compound above is genetically disclosed and claimed in PCT patent application PCT/US02/11884, filed 13 May 2002, which claims priority from U.S. patent application U. S . S .N. 60/293 ,464, filed 24 May 2001 , and incorporated herein by reference. The above compound has been selected for having a surprisingly superior toxicology profile over the compounds specifically disclosed in application cited above.

The following scheme illustrates the preparation of the compound of Formula II.

Scheme II

Figure imgf000007_0001

Cs2C03

Figure imgf000007_0002

The following examples further illustrate the preparation of the compounds of this invention as shown schematically in Schemes I and II. Example 1

Preparation of 7-(2-morpholin-4-yI-ethoxy)-4-(2-pyridin-2-yl-5,6-dihydro-4H- pyrroIo[l,2-b]pyrazol-3-yl)-q inoline

A. Preparation of 4-(2-pyridin-2-yl-5,6-dihydro-4H-pyrrolo[l,2-b]pyrazol-3-yl)- 7-[2-(tetrahydropyran-2-yIoxy)ethoxy]quinoIine

Heat 4-(2-pyridm-2-yl-5,6-dihydro-4H-pyrrolo[l,2-b]pyrazol-3-yl)-quinolin-7-ol (376 mg, 1.146 mmol), cesium carbonate (826 mg, 2.54 mmol), and 2-(2- bromoethoxy)tetrahydro-2H-pyran (380 μL, 2.52 mmol) in DMF (5 mL) at 120 °C for 4 hours. Quench the reaction with saturated sodium chloride and then extract with chloroform. Dry the organic layer over sodium sulfate and concentrate in vacuo. Purify the reaction mixture on a silica gel column eluting with dichloromethane to 10% methanol in dichloromethane to give the desired subtitled intermediate as a yellow oil (424 mg, 81%). MS ES+m/e 457.0 (M+l).

EXAMPLE 2

Preparation of 2-(6-methyl-pyridin-2-yl)-3-[6-amido-quinolin-4-yl)-5,6-dihydro-4H-pyrrolo[l,2- b]pyrazole

A. Preparation of 6-bromo-4-methyI-quinoline

Stir a solution of 4-bromo-phenylamine (1 eq), in 1,4-dioxane and cool to approximately 12 °C. Slowly add sulfuric acid (2 eq) and heat at reflux. Add methyl vinyl ketone (1.5 eq) drop wise into the refluxing solution. Heat the solution for 1 hour after addition is complete. Evaporate the reaction solution to dryness and dissolve in methylene chloride. Adjust the solution to pH 8 with 1 M sodium carbonate and extract three times with water. Chromatograph the residue on SiO (70/30 hexane/ethyl acetate) to obtain the desired subtitled inteπnediate. MS ES+ m e = 158.2 (M+l). B. Preparation of 6-methyl-pyridine-2-carboxylic acid methyl ester

Suspend 6-methyl-pyridine-2-carboxylic acid (10 g, 72.9 mmol) in methylene chloride (200 mL). Cool to 0 °C. Add methanol (10 mL), 4-dimethylaminopyridine (11.6 g, 94.8 mmol), and l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC)

(18.2 g, 94.8 mmol). Stir the mixture at room temperature for 6 hours, wash with water and brine, and dry over sodium sulfate. Filter the mixture and concentrate in vacuo.

Chromatograph the residue on SiO2 (50% ethyl acetate/hexanes) to obtain the desired subtitled intermediate, 9.66 g (92%), as a colorless liquid. 1H NMR (CDC13) 6 7.93-7.88 (m, IH), 7.75-7.7 (m, IH), 7.35-7.3 (m, IH), 4.00 (s, 3H), 2.60 (s, 3H).

C. Preparation of 2-(6-bromo-quinoIin-4-yl)-l-(6-methyl-pyridin-2-yl)-ethanone Dissolve 6-bromo-4-methyl-quinoline (38.5 g, 153 mmol) in 600 mL dry THF.

Cool to -70° C and treat with the dropwise addition of 0.5 M potassium hexamethyldisilazane (KN(SiMe )2 (400 mL, 200 mmol) over 2 hours while keeping the temperature below -65 °C. Stir the resultant solution at -70°C for 1 hour and add a solution of 6-methylpyridine-2-carboxylic acid methyl ester (27.2, 180 mmol) in 100 mL dry THF dropwise over 15 minutes. During the addition, the mixture will turn from dark red to pea-green and form a precipitate. Stir the mixture at -70°C over 2 hours then allow it to warm to ambient temperature with stirring for 5 hours. Cool the mixture then quench with 12 N HC1 to pH=l . Raise the pH to 9 with solid potassium carbonate. Decant the solution from the solids and extract twice with 200 mL ethyl acetate. Combine the organic extracts, wash with water and dry over potassium carbonate. Stir the solids in 200 mL water and 200 mL ethyl acetate and treat with additional potassium carbonate. Separate the organic portion and dry with the previous ethyl acetate extracts. Concentrate the solution in vacuo to a dark oil. Pass the oil through a 300 mL silica plug with methylene chloride then ethyl acetate. Combine the appropriate fractions and concentrate in vacuo to yield an amber oil. Rinse the oil down the sides of the flask with methylene chloride then dilute with hexane while swirling the flask to yield 38.5 g (73.8 %) of the desired subtitled intermediate as a yellow solid. MS ES+ = 341 (M+l)v D. Preparation of l-[2-(6-bromo-quinolin-4-yI)-l-(6-methyl-pyridin-2-yl)- ethylideneamino]-pyrrolidin-2-one

Stir a mixture of 2-(6-bromo-quinolin-4-yl)-l-(6-methyl-pyridin-2-yl)-ethanone (38.5 g, 113 mmol) and 1-aminopyrrolidinone hydrochloride (20 g, 147 mmol) in 115 mL pyridine at ambient temperature for 10 hours. Add about 50 g 4 A unactivated sieves. Continue stirring an additional 13 h and add 10-15 g silica and filter the mixture through a 50 g silica plug. Elute the silica plug with 3 L ethyl acetate. Combine the filtrates and concentrate in vacuo. Collect the hydrazone precipitate by filtration and suction dry to yield 33.3 g (69.7%) of the desired subtitled intermediate as an off-white solid. MS ES+ = 423 (M+l).

E. Preparation of 6-bromo-4-[2-(6-methyl-pyridin-2-yι)-5,6-dihydro-4H- pyrrolo[l,2-b]pyrazol-3-yl]-quinoline

To a mixture of (1.2 eq.) cesium carbonate and l-[2-(6-bromo-qumolin-4-yl)-l- (6-methyl-pyridin-2-yl)-ethylideneamino]-pyrrolidin-2-one (33.3 g, 78.7 mmol) add 300 mL dry N,N-dimethylformamide. Stir the mixture 20 hours at 100°C. The mixture may turn dark during the reaction. Remove the N,N-dimethylformamide in vacuo. Partition the residue between water and methylene chloride. Extract the aqueous portion with additional methylene chloride. Filter the organic solutions through a 300 mL silica plug, eluting with 1.5 L methylene chloride, 1.5 L ethyl acetate and 1.5 L acetone. Combine the appropriate fractions and concentrate in vacuo. Collect the resulting precipitate by filtration to yield 22.7 g (71.2%) of the desired subtitled intermediate as an off-white solid. MS ES+ = 405 (M+l).

F. Preparation of 4-[2-(6-methyl-pyridin-2-yl)-5,6-dihydro-4H-pyrrolo[l,2- b]pyrazol-3-yl]-quinoline-6-carboxylic acid methyl ester

Add 6-bromo-4-[2-(6-methyl-pyridin-2-yl)-5,6-dihydro-4H-pyrrolo[l,2- b]pyrazol-3-yl]-quinoline (22.7 g, 45 mmol) to a mixture of sodium acetate (19 g, 230 mmol) and the palladium catalyst [1,1 ‘- bis(diphenylphosphino)ferrocene]dichloropalladium(II), complex with dichloromethane (1:1) (850 mg, 1.04 mmol) in 130 mL methanol. Place the mixture under 50 psi carbon monoxide atmosphere and stir while warming to 90° C over 1 hour and with constant charging with additional carbon monoxide. Allow the mixture to cool over 8 hours, recharge again with carbon monoxide and heat to 90 °C. The pressure may rise to about 75 PSI. The reaction is complete in about an hour when the pressure is stable and tic (1 : 1 toluene/acetone) shows no remaining bromide. Partition the mixture between methylene chloride (600 mL) and water (1 L). Extract the aqueous portion with an additional portion of methylene chloride (400 mL.) Filter the organic solution through a 300 mL silica plug and wash with 500 mL methylene chloride, 1200 mL ethyl acetate and 1500 mL acetone. Discard the acetone portion. Combine appropriate fractions and concentrate to yield 18.8 g (87.4%) of the desired subtitled intermediate as a pink powder. MS ES+ = 385 (M+l).

G. Preparation of 2-(6-methyl-pyridin-2-yl)-3-[6-amido-quinolin-4-yι)-5,6- dihydro-4H-pyrrolo[l,2-b]pyrazole

Figure imgf000012_0001

Warm a mixture of 4-[2-(6-methyl-pyridin-2-yl)-5,6-dihydro-4H-pyrrolo[l,2- b]pyrazol-3-yl]-quinolme-6-carboxylic acid methyl ester in 60 mL 7 N ammonia in methanol to 90 °C in a stainless steel pressure vessel for 66 hours. The pressure will rise to about 80 PSI. Maintain the pressure for the duration of the reaction. Cool the vessel and concentrate the brown mixture in vacuo. Purify the residual solid on two 12 g Redi- Pak cartridges coupled in series eluting with acetone. Combine appropriate fractions and concentrate in vacuo. Suspend the resulting nearly white solid in methylene chloride, dilute with hexane, and filter. The collected off-white solid yields 1.104 g (63.8%) of the desired title product. MS ES+ = 370 (M+l).

PAPER

http://pubs.acs.org/doi/abs/10.1021/op4003054

Application of Kinetic Modeling and Competitive Solvent Hydrolysis in the Development of a Highly Selective Hydrolysis of a Nitrile to an Amide

Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285, United States
Org. Process Res. Dev., 2014, 18 (3), pp 410–416
DOI: 10.1021/op4003054
Publication Date (Web): February 11, 2014
Copyright © 2014 American Chemical Society
*Telephone: (317) 276-2066. E-mail: niemeier_jeffry_k@lilly.com (J.K.N.)., *Telephone: (317) 433-3769. E-mail: rrothhaar@lilly.com(R.R.R.).

Abstract

Abstract Image

A combination of mechanism-guided experimentation and kinetic modeling was used to develop a mild, selective, and robust hydroxide-promoted process for conversion of a nitrile to an amide using a substoichiometric amount of aqueous sodium hydroxide in a mixed water and N-methyl-2-pyrrolidone solvent system. The new process eliminated a major reaction impurity, minimized overhydrolysis of the product amide by selection of a solvent that would be sacrificially hydrolyzed, eliminated genotoxic impurities, and improved the intrinsic safety of the process by eliminating the use of hydrogen peroxide. The process was demonstrated in duplicate on a 90 kg scale, with 89% isolated yield and greater than 99.8% purity.

Patent ID Date Patent Title
US2015289795 2015-10-15 METHODS AND KITS FOR THE PROGNOSIS OF COLORECTAL CANCER
US2014348889 2014-11-27 Compositions and Methods for Treating and Preventing Neointimal Stenosis
US2014328860 2014-11-06 METHODS FOR STIMULATING HEMATOPOIETIC RECOVERY BY INHIBITING TGF BETA SIGNALING
US2014127228 2014-05-08 INHIBITION OF TGFBETA SIGNALING TO IMPROVE MUSCLE FUNCTION IN CANCER
US2014128349 2014-05-08 ADMINISTERING INHIBITORS OF TGFBETA SIGNALING IN COMBINATION WITH BENZOTHIAZEPINE DERIVATIVES TO IMPROVE MUSCLE FUNCTION IN CANCER PATIENTS
US2013071931 2013-03-21 PROCESS FOR HEPATIC DIFFERENTIATION FROM INDUCED HEPATIC STEM CELLS, AND INDUCED HEPATIC PROGENITOR CELLS DIFFERENTIATED THEREBY
US7872020 2011-01-18 TGF-[beta] inhibitors
US7834029 2010-11-16 QUINOLINYL-PYRROLOPYRAZOLES
US7265225 2007-09-04 Quinolinyl-pyrrolopyrazoles

REFERENCES

1: Rodón J, Carducci M, Sepulveda-Sánchez JM, Azaro A, Calvo E, Seoane J, Braña I, Sicart E, Gueorguieva I, Cleverly A, Pillay NS, Desaiah D, Estrem ST, Paz-Ares L, Holdhoff M, Blakeley J, Lahn MM, Baselga J. Pharmacokinetic, pharmacodynamic and biomarker evaluation of transforming growth factor-β receptor I kinase inhibitor, galunisertib, in phase 1 study in patients with advanced cancer. Invest New Drugs. 2014 Dec 23. [Epub ahead of print] PubMed PMID: 25529192.

2: Kovacs RJ, Maldonado G, Azaro A, Fernández MS, Romero FL, Sepulveda-Sánchez JM, Corretti M, Carducci M, Dolan M, Gueorguieva I, Cleverly AL, Pillay NS, Baselga J, Lahn MM. Cardiac Safety of TGF-β Receptor I Kinase Inhibitor LY2157299 Monohydrate in Cancer Patients in a First-in-Human Dose Study. Cardiovasc Toxicol. 2014 Dec 9. [Epub ahead of print] PubMed PMID: 25488804.

3: Rodon J, Carducci MA, Sepulveda-Sanchez JM, Azaro A, Calvo E, Seoane J, Brana I, Sicart E, Gueorguieva I, Cleverly AL, Sokalingum Pillay N, Desaiah D, Estrem ST, Paz-Ares L, Holdoff M, Blakeley J, Lahn MM, Baselga J. First-in-Human Dose Study of the Novel Transforming Growth Factor-β Receptor I Kinase Inhibitor LY2157299 Monohydrate in Patients with Advanced Cancer and Glioma. Clin Cancer Res. 2014 Nov 25. pii: clincanres.1380.2014. [Epub ahead of print] PubMed PMID: 25424852.

4: Huang C, Wang H, Pan J, Zhou D, Chen W, Li W, Chen Y, Liu Z. Benzalkonium Chloride Induces Subconjunctival Fibrosis Through the COX-2-Modulated Activation of a TGF-β1/Smad3 Signaling Pathway. Invest Ophthalmol Vis Sci. 2014 Nov 18;55(12):8111-22. doi: 10.1167/iovs.14-14504. PubMed PMID: 25406285.

5: Cong L, Xia ZK, Yang RY. Targeting the TGF-β receptor with kinase inhibitors for scleroderma therapy. Arch Pharm (Weinheim). 2014 Sep;347(9):609-15. doi: 10.1002/ardp.201400116. Epub 2014 Jun 11. PubMed PMID: 24917246.

6: Gueorguieva I, Cleverly AL, Stauber A, Sada Pillay N, Rodon JA, Miles CP, Yingling JM, Lahn MM. Defining a therapeutic window for the novel TGF-β inhibitor LY2157299 monohydrate based on a pharmacokinetic/pharmacodynamic model. Br J Clin Pharmacol. 2014 May;77(5):796-807. PubMed PMID: 24868575; PubMed Central PMCID: PMC4004400.

7: Oyanagi J, Kojima N, Sato H, Higashi S, Kikuchi K, Sakai K, Matsumoto K, Miyazaki K. Inhibition of transforming growth factor-β signaling potentiates tumor cell invasion into collagen matrix induced by fibroblast-derived hepatocyte growth factor. Exp Cell Res. 2014 Aug 15;326(2):267-79. doi: 10.1016/j.yexcr.2014.04.009. Epub 2014 Apr 26. PubMed PMID: 24780821.

8: Giannelli G, Villa E, Lahn M. Transforming growth factor-β as a therapeutic target in hepatocellular carcinoma. Cancer Res. 2014 Apr 1;74(7):1890-4. doi: 10.1158/0008-5472.CAN-14-0243. Epub 2014 Mar 17. Review. PubMed PMID: 24638984.

9: Dituri F, Mazzocca A, Peidrò FJ, Papappicco P, Fabregat I, De Santis F, Paradiso A, Sabbà C, Giannelli G. Differential Inhibition of the TGF-β Signaling Pathway in HCC Cells Using the Small Molecule Inhibitor LY2157299 and the D10 Monoclonal Antibody against TGF-β Receptor Type II. PLoS One. 2013 Jun 27;8(6):e67109. Print 2013. PubMed PMID: 23826206; PubMed Central PMCID: PMC3694933.

10: Bhola NE, Balko JM, Dugger TC, Kuba MG, Sánchez V, Sanders M, Stanford J, Cook RS, Arteaga CL. TGF-β inhibition enhances chemotherapy action against triple-negative breast cancer. J Clin Invest. 2013 Mar 1;123(3):1348-58. doi: 10.1172/JCI65416. Epub 2013 Feb 8. PubMed PMID: 23391723; PubMed Central PMCID: PMC3582135.

11: Bhattachar SN, Perkins EJ, Tan JS, Burns LJ. Effect of gastric pH on the pharmacokinetics of a BCS class II compound in dogs: utilization of an artificial stomach and duodenum dissolution model and GastroPlus,™ simulations to predict absorption. J Pharm Sci. 2011 Nov;100(11):4756-65. doi: 10.1002/jps.22669. Epub 2011 Jun 16. PubMed PMID: 21681753.

12: Bueno L, de Alwis DP, Pitou C, Yingling J, Lahn M, Glatt S, Trocóniz IF. Semi-mechanistic modelling of the tumour growth inhibitory effects of LY2157299, a new type I receptor TGF-beta kinase antagonist, in mice. Eur J Cancer. 2008 Jan;44(1):142-50. Epub 2007 Nov 26. PubMed PMID: 18039567.

References

http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4539082/

http://www.ncbi.nlm.nih.gov/pubmed/26057634

https://clinicaltrials.gov/ct2/show/NCT0242334

Bhattachar, Shobha N.; Journal of Pharmaceutical Sciences 2011, 100(11), 4756-4765 

Investigational new drugs (2015), 33(2), 357-70.

//////////TGF-β, TGF-βRI kinase inhibitor, ALK5, galunisertib, LY2157299, cancer, clinical trials, PHASE 3

CC1=CC=CC(=N1)C2=NN3CCCC3=C2C4=C5C=C(C=CC5=NC=C4)C(=O)N

Filed under: Phase3 drugs Tagged: ALK5, CANCER, clinical trials, galunisertib, LY2157299, PHASE 3, TGF-β, TGF-βRI kinase inhibitor

Quality Documentation of API mix in the Marketing Authorisation Procedure

$
0
0

DRUG REGULATORY AFFAIRS INTERNATIONAL

For different reasons, the manufacture of APIs may sometimes require adding excipients. In the context of an authorisation procedure, this practice reveals to be problematic. Read more here about the data required for the quality documentation of a API mix in an ASMF or a CEP.

http://www.gmp-compliance.org/enews_05334_Quality-Documentation-of-API-mix-in-the-Marketing-Authorisation-Procedure_15339,15332,S-WKS_n.html

The manufacture of APIs sometimes requires adding of one or several excipients like for example an antioxidant or an inert matrix for stabilisation purposes. Occasionally, corresponding mixtures can be manufactured to optimize workability for further processing or filling (e.g. improvement of flowability). Yet, within a marketing authorisation procedure, such an API mix can possibly be accepted differently than the pure API.

To clarify the questions around this topic, EMA’s QWP has published a document entitled “Quality Working Party questions and answers on API mix“. Please find hereinafter a summary of the questions addressed in the document:

What is an API mix?

View original post 476 more words


Filed under: Uncategorized

EMA publishes finalised Process Validation Guideline for Biotech Products

$
0
0

DRUG REGULATORY AFFAIRS INTERNATIONAL

Approximately two years ago the EMA published a draft guideline on process validation for the manufacture of biotech products. Now the final guideline has been published under the title “Guideline on process validation for the manufacture of biotechnology-derived active substances and data to be provided in the regulatory submission“.

READ

http://www.gmp-compliance.org/enews_05342_EMA-publishes-finalised-Process-Validation-Guideline-for-Biotech-Prodcts_15435,15373,15298,15250,Z-VM_n.html

Approximately two years ago the EMA published a draft guideline on process validation for the manufacture of biotech products. Now the final guideline has been published under the title “Guideline on process validation for the manufacture of biotechnology-derived active substances and data to be provided in the regulatory submission”.

The scope of the guideline is to provide guidance on the data to be included in a regulatory submission to demonstrate that the active substance manufacturing process is in a validated state. The guideline focuses on recombinant proteins and polypeptides, their derivates, and products of which they are components (e.g…

View original post 300 more words


Filed under: Uncategorized

Sun Pharma and Merck & Co. Inc. Enter into Licensing Agreement for Tildrakizumab, MK 3222

$
0
0

Tildrakizumab (MK-3222)

Company Merck & Co. Inc.
Description Anti-IL-23 antibody
Molecular Target Interleukin-23 (IL-23)
Mechanism of Action Antibody
Therapeutic Modality Biologic: Antibody
Latest Stage of Development Phase III
Standard Indication Psoriasis
Indication Details Treat moderate to severe chronic plaque psoriasis
Regulatory Designation
Partner Sun Pharmaceutical Industries Ltd.

Tildrakizumab is a monoclonal antibody designed for the treatment of immunologically mediated inflammatory disorders.[1]

Tildrakizumab was designed to block interleukin-23, a cytokine that plays an important role in managing the immune system and autoimmune disease. Originally developed by Schering-Plough, this drug is now part of Merck‘s clinical program, following that company’s acquisition of Schering-Plough.

Sun Pharmaceutical acquired worldwide rights to tildrakizumab for use in all human indications from Merck in exchange for an upfront payment of U.S. $80 million. Upon product approval, Sun Pharmaceutical will be responsible for regulatory activities, including subsequent submissions, pharmacovigilance, post approval studies, manufacturing and commercialization of the approved product. [2]

As of March 2014, the drug was in phase III clinical trials for plaque psoriasis. The two trials will enroll a total of nearly 2000 patients, and preliminary results are expected in June, 2015. [3][4]

References

http://clinicaltrials.gov/ct2/show/NCT01722331?term=SCH-900222&phase=2&fund=2&rank=2

Sun Pharma and Merck & Co. Inc. Enter into Licensing Agreement for Tildrakizumab, MK 3222

WHITEHOUSE STATION, N.J., and MUMBAI, India, Wednesday, September 17, 2014 (BUSINESS WIRE) – Merck & Co., Inc., (NYSE:MRK), known as MSD outside the United States and Canada, and Sun Pharmaceutical Industries Ltd. (Reuters: SUN.BO, Bloomberg: SUNP IN, NSE: SUNPHARMA, BSE: 524715) through their respective subsidiaries, today announced an exclusive worldwide licensing agreement for Merck’s investigational therapeutic antibody candidate, tildrakizumab, (MK-3222), which is currently being evaluated in Phase 3 registration trials for the treatment of chronic plaque psoriasis, a skin ailment.

Under terms of the agreement, Sun Pharma will acquire worldwide rights to tildrakizumab for use in all human indications from Merck in exchange for an upfront payment of U.S. $80 million. Merck will continue all clinical development and regulatory activities, which will be funded by Sun Pharma. Upon product approval, Sun Pharma will be responsible for regulatory activities, including subsequent submissions, pharmacovigilance, post approval studies, manufacturing and commercialization of the approved product. Merck is eligible to receive undisclosed payments associated with regulatory (including product approval) and sales milestones, as well as tiered royalties ranging from mid-single digit through teen percentage rates on sales.

“Consistent with our previously announced global initiative to sharpen our commercial and R&D focus, including prioritizing our late stage pipeline candidates, we are pleased to enter into this agreement with Sun Pharma to help realize the potential of tildrakizumab for patients with chronic plaque psoriasis,” said Iain D. Dukes, Ph.D., senior vice president, Business Development and Licensing, Merck Research Laboratories.

“Sun Pharma is very pleased to enter into this collaboration with Merck, a recognized leader in the field of inflammatory/immunology therapies, for this late-stage candidate for chronic plaque psoriasis,” said Kirti Ganorkar, senior vice president, Business Development, Sun Pharma. “This collaboration is a part of our strategy towards building our pipeline of innovative dermatology products in a market with strong growth potential.”

The transaction is subject to customary closing conditions, including the requirements under the Hart Scott-Rodino Antitrust Improvements Act.

About Tildrakizumab

Tildrakizumab is an investigational humanized, anti-IL-23p19 monoclonal antibody that binds specifically to IL-23p19 and is therefore designed to selectively block the cytokine IL-23. Human genetics suggest that inhibiting IL-23 is effective for treating inflammatory conditions. In clinical studies for the treatment of chronic plaque psoriasis, tildrakizumab demonstrates efficacy in blocking inflammation by blocking IL-23. Other potential indications, which may be evaluated in future, include psoriatic arthritis and Crohn’s Disease.

Further details of the Phase 3 clinical trials can be found at: http://clinicaltrials.gov

About Merck

Today’s Merck is a global healthcare leader working to help the world be well. Merck is known as MSD outside the United States and Canada. Through our prescription medicines, vaccines, biologic therapies, and consumer care and animal health products, we work with customers and operate in more than 140 countries to deliver innovative health solutions. We also demonstrate our commitment to increasing access to healthcare through far-reaching policies, programs and partnerships. For more information, visit www.merck.com and connect with us on Twitter, Facebook and YouTube.

About Sun Pharma

Established in 1983, listed since 1994 and headquartered in India, Sun Pharmaceutical Industries Ltd. (Reuters: SUN.BO, Bloomberg: SUNP IN, NSE: SUNPHARMA, BSE: 524715) is an international specialty pharmaceutical company with over 75% sales from global markets. It manufactures and markets a large basket of pharmaceutical formulations as branded generics as well as generics in US, India and several other markets across the world. For the year ending March 2014, overall revenues were at US$2.7 billion, of which US contributed US$1.6 billion. In India, the company is a leader in niche therapy areas of psychiatry, neurology, cardiology, nephrology, gastroenterology, orthopedics and ophthalmology. The company has strong skills in product development, process chemistry, and manufacturing of complex dosage forms. More information about the company can be found at www.sunpharma.com.

Tildrakizumab
Monoclonal antibody
Type ?
Source Humanized (from mouse)
Target IL23
Identifiers
CAS Number 1326244-10-3
ATC code none
ChemSpider none
Chemical data
Formula C6426H9918N1698O2000S46
Molar mass 144.4 kg/mol

///////Sun Pharma, Merck & Co. Inc, Licensing Agreement, Tildrakizumab, mk 3222


Filed under: Biosimilar drugs, Monoclonal antibody Tagged: Licensing Agreement, Merck & Co. Inc, mk 3222, sun pharma, Tildrakizumab

Processes for Constructing Homogeneous Antibody Drug Conjugates

$
0
0
Abstract Image

Antibody drug conjugates (ADCs) are synthesized by conjugating a cytotoxic drug or “payload” to a monoclonal antibody. The payloads are conjugated using amino or sulfhydryl specific linkers that react with lysines or cysteines on the antibody surface. A typical antibody contains over 60 lysines and up to 12 cysteines as potential conjugation sites. The desired DAR (drugs/antibody ratio) depends on a number of different factors and ranges from two to eight drugs/antibody. The discrepancy between the number of potential conjugation sites and the desired DAR, combined with use of conventional conjugation methods that are not site-specific, results in heterogeneous ADCs that vary in both DAR and conjugation sites. Heterogeneous ADCs contain significant fractions with suboptimal DARs that are known to possess undesired pharmacological properties. As a result, new methods for synthesizing homogeneous ADCs have been developed in order to increase their potential as therapeutic agents. This article will review recently reported processes for preparing ADCs with improved homogeneity. The advantages and potential limitations of each process are discussed, with emphasis on efficiency, quality, and in vivo efficacy relative to similar heterogeneous ADCs.

Table 1. Examples of Heterogeneous ADCs Currently in Clinical Trials for Cancer Indicationsa
ADC Sponsor Indications Status Payload Linked to Target
Adcetris Seattle Genetics HL and ALCL approved MMAE cysteine CD30
Kadcyla Genentech/Roche breast cancer approved DM1 lysine Her2
inotuzumab ozogamicin Pfizer NHL and ALL Phase III calicheamicin lysine CD22
lorvotuzumab mertansine Immunogen SCLC Phase II DM1 lysine CD56
glembatumumab vedotin Celldex BC, melanoma Phase II MMAE cysteine GPNMB
PSMA-ADC Progenics prostate Phase II MMAE cysteine FOLH1
SAR-3419 Sanofi DLBCL, ALL Phase II DM4 lysine CD19
ABT-414 Abbvie glioblastoma Phase II MMAE cysteine EGFR
BT-062 Biotest mult. myeloma Phase II DM4 lysine CD138
HLL1-Dox Immunomedics CLL, MM, NHL Phase II doxorubicin cysteine CD74
Immu-130 Immunomedics CRC Phase II SN-38 cysteine CEACAM5
Immu-132 Immunomedics solid tumors Phase II SN-38 cysteine EGP1
SYD985 Synthon breast cancer Phase II duocarmycin cysteine Her2
SAR-3419 Sanofi DLBCL, ALL Phase II DM4 lysine CD19
IMGN853 ImmunoGen solid tumors Phase I DM4 lysine FOLR1
IMGN529 ImmunoGen BCL,CLL, NHL Phase I DM1 lysine CD37
ASG-22M6E Astellas solid tumors Phase I MMAE cysteine nectin-4
AGS-16M8F Astellas RCC Phase I MMAF cysteine AGS16
AMG 172 Amgen RCC Phase I DM1 lysine CD27L
AMG 595 Amgen glioblastoma Phase I DM1 lysine EGFR8
BAY94-9343 Bayer solid tumors Phase I DM4 lysine mesothelin

Processes for Constructing Homogeneous Antibody Drug Conjugates

Igenica Biotherapeutics, 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.6b00067
Publication Date (Web): April 14, 2016
Copyright © 2016 American Chemical Society
*Igenica Biotherapeutics 863A Mitten Road, Suite 100B Burlingame, CA 94010, USA. E-mail: dyjackson@comcast.net. Cell: 650-339-3948.
ACS Editors’ Choice – This is an open access article published under an ACS AuthorChoice License, which permits copying and redistribution of the article or any adaptations for non-commercial purposes.

//////Processes, Constructing,  Homogeneous,  Antibody Drug Conjugates


Filed under: Uncategorized Tagged: Antibody Drug Conjugates, Constructing, Homogeneous, Processes

A New Antibiotic (E)-3-(3-Carboxyphenyl)-2-(4-cyanostyryl)quinazolin-4(3H)-one, from University Of Notre Dame

$
0
0

SCHEMBL16024086.png

STR1

(E)-3-(3-Carboxyphenyl)-2-(4-ethynylstyryl)quinazolin-4(3H)-one

(E)-3-(2-(4-Cyanostyryl)-4-Oxoquinazolin-3(4h)-Yl)benzoic Acid;

1624273-22-8  CAS

NA SALT 1624273-21-7 CAS

INNOVATORS

University Of Notre Dame

Mayland Chang, Shahriar Mobashery, Renee BOULEY INVENTORS

C24H15N3O3
Molecular Weight: 393.3942 g/mol
 1H NMR (500 MHz, DMSO-d6) δ 4.32 (s, 1H), 6.34 (d, J = 15.55 Hz, 1H), 7.35 (d, J = 8.37 Hz, 2H), 7.44 (d, J = 8.37 Hz, 2H), 7.49 (d, J = 7.58 Hz, 1H), 7.55 (t, J = 7.98 Hz, 1H), 7.58 (t, J = 7.78 Hz, 1H), 7.78 (d, J = 8.17 Hz, 1H), 7.87 (m, 3H), 8.05 (d, J = 7.78 Hz, 1H), 8.13 (d, J = 7.98 Hz, 1H).
13C NMR (126 MHz, DMSO-d6) δ 82.70, 83.24, 120.66, 121.04, 122.84, 126.51, 126.81, 127.31, 127.83, 129.98, 130.12, 132.33, 132.39, 133.49, 134.90, 135.21, 137.21, 137.99, 147.36, 151.04, 161.37, 166.58.
HRMS (m/z): [M + H]+, calcd for C25H17N2O3, 393.1234; found, 393.1250. HRMS (m/z): [M + Na]+, calcd for C25H16N2NaO3, 415.1053; found, 415.1054.
The emergence of resistance to antibiotics over the past few decades has created a state of crisis in the treatment of bacterial infections.Over the years, β-lactams were the antibiotics of choice for treatment of S. aureus infections. However, these agents faced obsolescence with the emergence of methicillin-resistant S. aureus (MRSA). Presently, vancomycin, daptomycin, linezolid, or ceftaroline are used for treatment of MRSA infections, although only linezolid can be dosed orally. Resistance to all four has emerged. Thus, new anti-MRSA antibiotics are sought, especially agents that are orally bioavailable.  a new antibiotic (E)-3-(3-carboxyphenyl)-2-(4-cyanostyryl)quinazolin-4(3H)one, with potent activity against S. aureus, including MRSA. We document that quinazolinones of our design are inhibitors of cell-wall biosynthesis in S. aureus and do so by binding to dd-transpeptidases involved in cross-linking of the cell wall.  quinazolinones possess activity in vivo and are orally bioavailable. This antibiotic holds promise in treating difficult infections by MRSA.
STR1

PAPER

Journal of the American Chemical Society (2015), 137(5), 1738-1741.

http://pubs.acs.org/doi/abs/10.1021/jacs.5b00056

Discovery of Antibiotic (E)-3-(3-Carboxyphenyl)-2-(4-cyanostyryl)quinazolin-4(3H)-one

Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
Department of Crystallography and Structural Biology, Instituto de Química-Física “Rocasolano”, Consejo Superior de Investigaciones Científicas, Madrid, Spain
§ Freimann Life Sciences Center and Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556, United States
J. Am. Chem. Soc., 2015, 137 (5), pp 1738–1741
DOI: 10.1021/jacs.5b00056
Publication Date (Web): January 28, 2015
Copyright © 2015 American Chemical Society

Abstract

Abstract Image

In the face of the clinical challenge posed by resistant bacteria, the present needs for novel classes of antibiotics are genuine. In silico docking and screening, followed by chemical synthesis of a library of quinazolinones, led to the discovery of (E)-3-(3-carboxyphenyl)-2-(4-cyanostyryl)quinazolin-4(3H)one (compound 2) as an antibiotic effective in vivo against methicillin-resistant Staphylococcus aureus (MRSA). This antibiotic impairs cell-wall biosynthesis as documented by functional assays, showing binding of 2 to penicillin-binding protein (PBP) 2a. We document that the antibiotic also inhibits PBP1 of S. aureus, indicating a broad targeting of structurally similar PBPs by this antibiotic. This class of antibiotics holds promise in fighting MRSA infections.

PATENT

WO 2014138302

http://www.google.com/patents/WO2014138302A1?cl=en

Staphylococcus aureus is a common bacterium found in moist areas of the body and skin. S. aureus can also grow as a biofilm, representing the leading cause of infection after implantation of medical devices. Approximately 29% (78.9 million) of the US population is colonized in the nose with S. aureus, of which 1.5% (4.1 million) is methicillin-resistant S. aureus (MRSA). In 2005, 478,000 people in the US were hospitalized with a S. aureus infection, of these 278,000 were MRSA infections, resulting in 19,000 deaths. MRSA infections have been increasing from 2% of S. aureus infections in intensive care units in 1974 to 64% in 2004, although more recent data report stabilization. Approximately 14 million outpatient visits occur every year in the US for suspected S. aureus skin and soft tissue infections. About 76% of these infections are caused by S. aureus, of which 78% are due to MRSA, for an overall rate of 59%. Spread of MRSA is not limited to nosocomial (hospital-acquired) infections, as they are also found in community-acquired infections. Over the years, β-lactams were antibiotics of choice in treatment of S. aureus infections. However, these agents faced obsolescence with the emergence of

MRSA. Presently, vancomycin, daptomycin or linezolid are agents for treatment of MRSA infections, although only linezolid can be dosed orally. Resistance to all three has emerged. Thus, new anti-MRSA therapeutic strategies are needed, especially agents that are orally bioavailable.

Clinical resistance to β-lactam antibiotics by MRSA has its basis predominantly in acquisition of the mecA gene, which encodes penicillin-binding protein 2a (PBP2a). PBP2a, a cell-wall DD- transpeptidase, is refractory to inhibition by essentially all commercially available β-lactams (ceftaroline is an exception), antibiotics that irreversibly acylate the active-site serine of typical PBPs. PBPs catalyze biosynthesis of the bacterial cell wall, which is essential for the survival of the bacterium. Accordingly, new ηοη-β-lactam antibiotics that inhibit PBP2a are needed to combat drug-resistant strains of bacteria. SUMMARY

Staphylococcus aureus is responsible for a number of human diseases, including skin and soft tissue infections. Annually, 292,000 hospitalizations in the US are due to S. aureus infections, of which 126,000 are related to methicillin-resistant Staphylococcus aureus (MRSA), resulting in 19,000 deaths. A novel structural class of antibiotics has been discovered and is described herein. A lead compound in this class shows high in vitro potency against Gram-positive bacteria comparable to those of linezolid and superior to vancomycin (both considered gold standards) and shows excellent in vivo activity in mouse models of MRSA infection.

The invention thus provides a novel class of ηοη-β-lactam antibiotics, the quinazolinones, which inhibit PBP2a by an unprecedented mechanism of targeting both its allosteric and active sites. This inhibition leads to the impairment of the formation of cell wall in living bacteria. The quinazolinones described herein are effective as anti-MRSA agents both in vitro and in vivo. Furthermore, they exhibit activity against other Gram-positive bacteria. The quinazolinones have anti-MRSA activity by themselves. However, these compounds synergize with β-lactam antibiotics. The use of a combination of a quinazolinone with a β-lactam antibiotic can revive the clinical use of β-lactam antibacterial therapy in treatment of MRSA infections. The invention provides a new class of quinazolinone antibiotics, optionally in combination with other antibacterial agents, for the therapeutic treatment of methicillin- resistant Staphylococcus aureus and other bacteria.

The quinazolinone compounds described herein can be prepared using standard synthetic techniques known to those of skill in the art. Examples of such techniques are described by Khajavi et al. (J. Chem. Res. (S), 1997, 286-287) and Mosley et al. (J. Med. Chem. 2010, 53, 5476-5490). A general preparatory scheme for preparing the compounds described herein, for example, compounds of Formula

Figure imgf000030_0001
Figure imgf000031_0001

wherein each of the variables are as defined for one or more of the formulas described herein, such as Formula (A).

EXAMPLES

Example 1. Compound Preparation

Chemistry. Organic reagents and solvents were purchased from Sigma- Aldrich. lH and 13C NMR spectra were recorded on a Varian INOVA-500. High-resolution mass spectra were obtained using a Bruker micrOTOF/Q2 mass spectrometer.

Figure imgf000034_0001

2-Methyl-4H-benzo[</| [l,3]oxazin-4-one (3). Anthranilic acid (20 g, 146 mmol) was dissolved in triethyl orthoacetate (45 mL, 245 mmol) and refluxed for 2 h. The reaction mixture was cooled on ice for 4 h to crystallize the intermediate. The resulting crystals were filtered and washed with hexanes to give 3 (17 g, 72% yield). lH NMR (500 MHz, CDC13) δ 2.47 (s, 3H), 7.50 (t, J= 7.38 Hz, 1H), 7.54 (d, J = 7.98 Hz, 1H), 7.80 (t, J= 7.18 Hz, 1H), 8.18 (d, J= 7.78 Hz, 1H). 13C NMR (126 MHz, CDCI3) δ 21.59, 1 16.84, 126.59, 128.42, 128.66, 136.77, 146.61, 159.89, 160.45. HRMS (m/z): [M + H]+, calcd for C9H8NO2, 162.0550; found , 162.0555.

2-Methyl-3-(3-carboxyphenyl)-quinazolin-4(3//)-one (4). Compound 3 (2 g, 12.4 mmol) and 3- aminophenol (1.7 g, 12.4 mmol) were suspended in glacial acetic acid (8 mL, 140 mmol), and dissolved upon heating. The reaction was refluxed for 5 h, at which point 5 mL water was added to the cooled reaction mixture. The resulting precipitate was filtered and washed with water, followed by cold ethanol and hexane to give 4 (3.19 g, 92% yield). lH (500 MHz, DMSO-d6) δ 2.87 (s, 3H), 7.52 (t, J= 7.38 Hz, 1H), 7.66-7.73 (m, 3H), 7.84 (t, J= 7.38 Hz, 1H), 8.01 (s, 1H), 8.09 (t, J= 7.58 Hz, 2H). 13C NMR (126 MHz, DMSO-de) δ 24.13, 120.48, 126.32, 126.47, 126.72, 129.52, 129.83, 130.01, 132.40, 133.07, 134.67, 138.18, 147.37, 154.13, 161.44, 166.58. HRMS (m/z): [M + H]+, calcd for C16H13N2O3 ,

281.0921 ; found, 281.0917.

Sodium (£)-3-(3-carboxyphenyl)-2-(4-cyanostyryl)quinazolin-4(3H)-one (2). Compound 4 (1.0 g, 3.6 mmol) and 4-formylbenzonitrile (0.56 g, 4.3 mmol) were suspended in glacial acetic acid (5 mL, 87 mmol), a suspension that dissolved upon heating. The reaction was refluxed for 18 h and 5 mL water was added to the cooled reaction mixture. The resulting precipitate was filtered and washed with water, followed by cold ethanol and hexanes to afford the carboxylic acid (0.77g, 75% yield). HRMS (m/z): [M + H]+, calcd for C24H16N3O3, 394.1 186; found 394.1214. The carboxylic acid (0.45 g, 1.1 mmol) was dissolved in hot ethanol, to which sodium 2-ethylhexanoate (0.28 g, 1.7 mmol) was added. The reaction mixture was stirred on ice for 2 h. The precipitate was filtered and washed with cold ethanol. The product was obtained by dissolving the precipitate in about 5 mL of water and subsequent lyophilization of the solution to give 2 as the sodium salt (0.4 g, 85% yield).

¾ NMR (500 MHz, DMSO- de) δ 6.47 (d, J= 15.55 Hz, 1H), 7.59 (m, 3H), 7.74 (d, J= 5.38 Hz, 2H), 7.79 (m, 3H), 7.91 (m, 2H), 8.05 (s, 1H), 8.14 (d, J= 7.78 Hz, 2H).

13C NMR (126 MHz, DMSO-de) δ 11 1.56, 1 18.61, 120.76, 123.42, 126.50, 127.01, 127.35, 128.26, 129.99, 130.06, 130.12, 132.33, 132.83, 133.46, 134.89, 136.95, 137.03, 139.25, 147.21, 150.74, 161.25, 166.52.

HRMS (m/z): [M + H]+, calcd for C24Hi5N3NaO3, 416.1006; found, 416.0987.

PAPER

http://pubs.acs.org/doi/full/10.1021/acs.jmedchem.6b00372

Structure–Activity Relationship for the 4(3H)-Quinazolinone Antibacterials

Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
Freimann Life Sciences Center and Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556, United States
J. Med. Chem., Article ASAP
DOI: 10.1021/acs.jmedchem.6b00372
Publication Date (Web): April 18, 2016
Copyright © 2016 American Chemical Society
*S.M.: e-mail, mobashery@nd.edu; phone, 574-631-2933., *M.C.: e-mail, mchang@nd.edu; phone, 574-631-2965.
ACS Editors’ Choice – This is an open access article published under an ACS AuthorChoice License, which permits copying and redistribution of the article or any adaptations for non-commercial purposes.
Abstract Image

We recently reported on the discovery of a novel antibacterial (2) with a 4(3H)-quinazolinone core. This discovery was made by in silico screening of 1.2 million compounds for binding to a penicillin-binding protein and the subsequent demonstration of antibacterial activity againstStaphylococcus aureus. The first structure–activity relationship for this antibacterial scaffold is explored in this report with evaluation of 77 variants of the structural class. Eleven promising compounds were further evaluated for in vitro toxicity, pharmacokinetics, and efficacy in a mouse peritonitis model of infection, which led to the discovery of compound 27. This new quinazolinone has potent activity against methicillin-resistant (MRSA) strains, low clearance, oral bioavailability and shows efficacy in a mouse neutropenic thigh infection model.

NMR

STR1

STR1

INVENTORS

Renee Bouley

Renee Bouley selected to receive prestigious ACS Predoctoral Fellowship

Published: July 02, 2013

Renee Bouley

Renee Bouley, a third year graduate student in the Department of Chemistry and Biochemistry, has been selected to receive a prestigious American Chemical Society (ACS) Division of Medicinal Chemistry Predoctoral Fellowship.  Bouley is one of only four recipients chosen for the 2013-2014 cycle.

This award supports doctoral candidates working in the area of medicinal chemistry who have demonstrated superior achievements as graduate students and who show potential for future work as independent investigators. These fellowships have been awarded annually since 1991 and include one year stipend support and an invitation to present the fellow’s research results at a special awards session at the ACS National Meeting.

Bouley’s work, conducted under the advisement of Shahriar Mobashery, Navari Family Professor in Life Sciences, and Mayland Chang, Research Professor and Director of the Chemistry-Biochemistry-Biology Interface (CBBI) Program, centers around the discovery of a new class of antibiotics that are selective against staphylococcal species of bacteria, including hard-to-treat methicillin-resistant Staphylococcus aureus (MRSA).  She has already identified a class of compounds that has in vitro activity against bacteria and demonstrated efficacy in mice. Bouley spent three months in 2012 in the laboratory of Prof. Juan Hermoso at Consejo Superior de Investigaciones Cientificas in Madrid, Spain, where she solved the crystal structure of the lead compound in complex with its target protein. Her studies have shown an unprecedented mechanism of action that opens opportunities for clinical resurrection of β-lactam antibiotics in combination with the new antibiotics. Bouley’s work during her fellowship tenure will explore structural analogs of these compounds with the goal of optimizing their potency in vivo and improving their drug-like properties.

Bouley is already the recipient of a National Institutes of Health Ruth L. Kirschstein National Research Service Award – CBBI (Chemistry-Biochemistry-Biology Interface) Program, a CBBI Research Internship Award, and an American Heart Association Predoctoral Fellowship (declined)………..https://www.linkedin.com/in/renee-bouley-43243215

University of Notre Dame

MAYLAND CHANG

http://chemistry.nd.edu/people/mayland-chang/

MAYLAND CHANG

  • Research Professor; Director, Chemistry-Biochemistry-Biology Interface (CBBI) Program
  • Office: 247 NSH
  • Phone: (574) 631-2965

Dr. Chang obtained B.S. degrees in biological sciences and chemistry from the University of Southern California, and a Ph.D. in chemistry from the University of Chicago.  Subsequently, she conducted postdoctoral research at Columbia University as a National Institutes of Health postdoctoral fellow.  She joined the faculty of the University of Notre Dame in 2003.  Previously, Dr. Chang was Chief Operating Officer of University Research Network, Inc., Senior Scientist with Pharmacia Corporation, and Senior Chemist at Dow Chemical Company.  She has characterized the ADME properties of numerous drugs, as well as prepared NDAs, INDs, Investigator’s Brochures, product development plans, and candidate drug evaluations.

Shahriar Mobashery

Shahriar Mobashery

Shahriar Mobashery

Navari Professor at University of Notre Dame

The Mobashery research program integrates computation, biochemistry, molecular biology, and the organic synthesis of medically important molecules. Bringing together these different disciplines is required to produce both scientific and medical advances for very difficult, but critically important clinical problems.

http://chemistry.nd.edu/people/shahriar-mobashery/

https://www.linkedin.com/in/shahriar-mobashery-71b67b4b

/////// 1624273-22-8, Antibiotic,  (E)-3-(3-Carboxyphenyl)-2-(4-cyanostyryl)quinazolin-4(3H)-one, methicillin-resistant S. aureus, MRSA, 1624273-21-7, PRECLINICAL

O=C(O)c1cc(ccc1)N3C(=Nc2ccccc2C3=O)/C=C/c4ccc(C#N)cc4


Filed under: Preclinical drugs Tagged: (E)-3-(3-Carboxyphenyl)-2-(4-cyanostyryl)quinazolin-4(3H)-one, 1624273-21-7, 1624273-22-8, antibiotic, methicillin-resistant S. aureus, MRSA, preclinical

Nolatrexed Dihydrochloride

$
0
0

Nolatrexed.png

Nolatrexed

NDA Filed in china

A thymidylate synthase inhibitor potentially for the treatment of hepatocellular carcinoma and nasopharyngeal cancer.

AG-337

CAS No. 147149-76-6 (free)

free form data

(eluents: CH3CN−H2O = 10−90, pH 4.94; Rt = 11.8 min); Rf = 0.31 [ethyl acetate/(0.63 M NH3 in ethanol) = 6/4]; Mp 300−302 °C (lit.:(J. Med. Chem. 1993, 36, 733746) a tan solid; Mp 301−302 °C); MS (ESI+) m/z: 285.1 [M + 1]+; the major impurity: 3.0% (Rt = 13.0 min); Mp 73−77 °C; 1H NMR (DMSO-d6): δ 7.95 (d, J = 6.4 Hz, 4 H), 8.81 (d, J = 6.4 Hz, 4 H);

MS (ESI+) m/z: 219.2 [M − 1]+;

Nolatrexed dihydrochloride.png

152946-68-4(Nolatrexed Dihydrochloride)

2-amino-6-methyl-5-pyridin-4-ylsulfanyl-1H-quinazolin-4-one;dihydrochloride

Nolatrexed dihydrochloride; Thymitaq; 152946-68-4; Nolatrexeddihydrochloride; AG 337; AG-337; 
Molecular Formula: C14H14Cl2N4OS
Molecular Weight: 357.25816 g/mol

diHCl data

IR (KBr cm−1): 3401, 3058, 2929, 1701, 1621, 1471, 799;

1H NMR (DMSO-d6): δ 2.43 (s, 3H, −CH3), 7.53 (d,J = 6.9 Hz, 2H, Pyr-H), 7.67 (d, J = 8.5 Hz, 1H, Ar−H), 7.92 (d, J = 8.5 Hz, 1 Hz, Ar−H), 8.30 (br s, 3H, NH3), 8.52 (d, J = 6.9 Hz, 2H, Pyr-H); MS (ESI+) m/z: 285 [M − 1−2Cl]+; (ESI+) m/z: 283 [M − 1− 2HCl]+.

Pfizer (Originator) , Gilead,LG Life Sciences,北京康辰药业

Nolatrexed is a thymidylate synthase inhibitor.[1][2]

Phase I studies of p.o. administered nolatrexed dihydrochloride (AG337, THYMITAQ), a nonclassical thymidylate synthase inhibitor, were performed to establish the maximum tolerated dose and a recommended dose for Phase II studies. The bioavailability and pharmacokinetic and pharmacodynamic properties of oral nolatrexed were also studied. Forty-five patients were treated with oral nolatrexed every 6 h for 5 days at doses of 288-1000 mg/m2/day. The bioavailability of the oral preparation was determined, and the effect of a standard meal on nolatrexed absorption was investigated at a dose of 800 mg/m2/day. Nolatrexed plasma concentrations were analyzed by high-performance liquid chromatography. Nolatrexed was rapidly absorbed with a median bioavailability of 89% (range 33-116%), with 88% of patients above 70%. The dose-limiting toxicities were gastrointestinal, and the recommended Phase II oral dose was 800 mg/m2/day. After a standard meal, the peak plasma nolatrexed concentration achieved was lower (median, 8.3 microg/ml versus 15.0 microg/ml; P = 0.001), and the time taken to reach the peak was longer (median, 180 min versus 45 min; P = 0.00003), but the trough concentration was higher (median, 3.6 microg/ml versus 2.1 microg/ml; P = 0.004) when compared with the fasted state. The area under the nolatrexed plasma concentration versus time curve was not affected by food. Average trough nolatrexed concentration, but not dose, was significantly related to the % decrease in both thrombocytes (r2 = 0.58; C50 = 6.0 microg/ml, where C50 is the plasma concentration associated with a 50% decrease in thrombocytes) and neutrophils (r2 = 0.63; C50 = 0.6 microg/ml). Nolatrexed can be safely administered as an oral preparation at a dose of 800 mg/m2/day for 5 days. Bioavailability was close to 100% and, because inhibition of thymidylate synthase by nolatrexed is rapidly reversible, the slower absorption after a standard meal may result in a shorter duration of noninhibitory concentrations between doses.

Catalytic hydrogenation of 2-bromo-4 -nitrotoluene (I) over Raney-Ni provided aniline (II). Reaction of (II) with chloral hydrate and hydroxylamine gave rise to the isonitrosoacetanilide (III), which was subsequently cyclized to the isatin (IV) by heating in concentrated H2SO4. Oxidative cleavage of isatin (IV) produced the anthranilic acid (V). This was converted to the benzoxazinone (VI) upon refluxing with acetic anhydride. Ring opening of benzoxazinone (VI) with MeOH, followed by acidic hydrolysis of the acetamide function, yielded the anthranilate ester (VII). The quinazoline derivative (VIII) was then obtained by treatment of anthranilate (VII) with chloroformamidine hydrochloride in refluxing diglyme. Finally, displacement of the bromide group of (VIII) with the sodium thiolate of 4-mercaptopyridine (IX) under Ullmann conditions afforded the title pyridyl sulfide.

Dissertation title [BT] A New Method for Synthesis of Nolatrexed Dihydrochloride
Hangul title Nolatrexed dihydrochloride Synthesis Process Development
Author Xueqing Zhao, Fei Li, Weiping Zhuang, Xiaowen Xue, Yuanyang Lian, Jianhui Fan and Dongsheng Fang
Japjimyeong ORG PROCESS RES DEV Issue year 2010
Gwonho details 14 (2) The surface 346-350
ABSTRACT
A new synthetic method for nolatrexed dihydrochloride (thymitaq) has been developed. The synthesis was accomplished in three steps featuring the direct conversion of the starting 4-bromo-5-methylisatin into the methyl anthranilate by potassium peroxydisulfate / sodium methoxide. In the final Ullmann reaction potassium carbonate was employed in place of sodium hydride, and the amount of copper catalysts was significantly reduced. Moreover, sodium sulfide solution was utilized to efficiently remove copper under approximately neutral conditions instead of hydrogen sulfide / methanol under strongly acidic conditions. By means of these modifications, nolatrexed dihydrochloride was ensured to be prepared in good yield and high purity.
Contents

Nolatrexed dihydrochloride (2-Amino-6-methyl-5-(4-pyridylthio) -3 H-quinazolin-4-one dihydrochloride, thymitag, 1) is the HCC cancer therapeutic agent to the TS (thymidylate synthase) folate binding site on the TS inhibitor as DNA replication inhibition, DNA damage, S-phase cell cycle arrest, and caspase-dependent apoptosis induction and clinical 2 on theresults look HCC patients, the survival benefit of showing the current phase III study is in progress in it. under scheme 1 is conducted in a number of synthesis team Nolatrexedillustrates the development process

Scheme 1. Synthetic routes A-F from 4-bromo-5-methylisatin (2) to nolatrexed dihydrochloride (1)

The scheme 1 When the complex first synthesis process but is A : 23457 · HCl1 or in part, 6 pass through a B step ( 2365 ) to obtain the desired compound with, but However, these processes are of the desired product quality control had a disadvantage unfulfilled this . after C, D, E process was developed during the E step is a step wherein compound 8 from the first to the one-pot is the most superior process consists in the process also drug of the compound for use as a quality control has difficulty in . more recentlyWennerberg is a new process F compounds were reported for 3 compound directly from the 7fully in the process I scored quality control could be the place . in the process, each reactionstep partially changed by the use of a reagent zoom impurity to minimize the formation of .However, this process also work-up, and purification there have difficulties to process the authors reported a new efficient way .
Scheme 2. Synthetic route G from 4-bromo-5-methylisatin (2) to nolatrexed dihydrochloride (1)
Scheme 2 The process reported to also have specifically not a new process only takes the best features from several processes previously reported , significant differences that the author is proud director teen two direct compound from 5 will get the , also reported in other processes already advanced mercaptopyridine introducing    Ullmann reaction in the processimpurity , to reduce the formation of NaH , instead of K2CO3 were used the copper catalyst in order to minimize the amount of copper scavenge used to H2S instead of Na2S was used . the compound obtained in the process 1 of the purity is 96.6% and 3% with impurities of the 4,4′-dithiodipyridine this was confirmed copper impurity is 20 ppm was below . last Nolatrexed dihydrochloride in the process to obtain a 99.7% purity I scored the desired product , 0.3% ofunidentified impurity, and 10 ppm less than copper because it contains should think very advanced process compared to the previous number of ways . Fortunately Ullmann key contained in the reaction impurity in 4,4′-dithiodipyridine was automatically removed from the crystallization process of the last reaction.

Korea Research Institute of Chemical Technology provides incurable disease treatment and research center, Dr. jaedu
View original http://pubs.acs.org/doi/full/10.1021/op9002517
Route 1

Reference:1. J. Med. Chem. 1993, 36, 733-746.

2. WO9320055A1.

Route 2

Reference:1. Org. Process Res. Dev. 2008, 12, 1195-1200.

Route 3

Reference:1. Org. Process Res. Dev. 2010, 14, 346-350.

2. CN1335307A.

Route 4
Ref Chemical Reagents 2011, 33, 1131-1134..

References

  1. Hughes AN, Rafi I, Griffin MJ, et al. (January 1999). “Phase I studies with the nonclassical antifolate nolatrexed dihydrochloride (AG337, THYMITAQ) administered orally for 5 days”. Clin. Cancer Res. 5 (1): 111–8. PMID 9918208.
  2. “Nolatrexed”. PubChem.gov. Pub Chem. Retrieved 12 August 2014.
Nolatrexed
Nolatrexed.png
Names
IUPAC name
2-Amino-6-methyl-5-(4-pyridylthio)-1H-quinazolin-4-one
Identifiers
147149-76-6 Yes
ChemSpider 97268 
Jmol 3D model Interactive image
PubChem 108189
UNII K75ZUN743Q Yes
Properties
C14H12N4OS
Molar mass 284.34 g/mol
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).
///////Nolatrexed,  thymidylate synthase inhibitor, AG337, THYMITAQ,
CC1=C(C2=C(C=C1)NC(=NC2=O)N)SC3=CC=NC=C3.Cl.Cl

Filed under: NDA Tagged: AG337, Nolatrexed, thymidylate synthase inhibitor, THYMITAQ

CADROFLOXACIN

$
0
0

Cadrofloxacin StructureCadrofloxacin.png

Cadrofloxacin , CS 940

3-Quinolinecarboxylic acid, 1-cyclopropyl-8-(difluoromethoxy)-6-fluoro-1,4-dihydro-7-[(3S)-3-methyl-1-piperazinyl]-4-oxo-, hydrochloride (1:1)

UNII-1YOQ7J9ACY; 153808-85-6; CADROFLOXACIN HYDROCHLORIDE; 1-cyclopropyl-8-(difluoromethoxy)-6-fluoro-7-[(3s)-3-methylpiperazin-1-yl]-4-oxo-1,4-dihydroquinoline-3-carboxylic acid;

1-cyclopropyl-8-(difluoromethoxy)-6-fluoro-7-[(3S)-3-methylpiperazin-1-yl]-4-oxoquinoline-3-carboxylic acid

NDA Filed in china

Molecular Formula: C19H20F3N3O4
Molecular Weight: 411.37501 g/mol

Company:HengRui (Originator), Daiichi Sankyo (Originator), UBE (Originator)

A quinolone antibiotic potentially for the treatment of bacterial infections.

Research Code CS-940

CAS No. 153808-85-6(FREE)

Cas 128427-55-4(Cadrofloxacin HCl)

HYDROCHLORIDE

Molecular Weight 447.84
Formula C19H20F3N3O4 • HCl
  • OriginatorSankyo; Ube Industries
  • DeveloperSankyo
  • ClassAntibacterials; Quinolones; Small molecules
  • Mechanism of ActionType II DNA topoisomerase inhibitors
    • 20 Jun 1996An animal study has been added to the Bacterial infections pharmacodynamics section
    • 24 Mar 1995Phase-II clinical trials for Bacterial infections in Japan (PO)

Cadrofloxacin hydrochloride was studied for the treatment of bacterial infections.The compound was originally developed by UBE and Daiichi Sankyo. However, this study was discontinued. The compound currently was developed by Hengrui.

SYNTHESIS

Decarboxylation of 3,5,6-trifluoro-4- hydroxyphthalic acid (I) upon heating at 140 C in an autoclave furnished 2,4,5-trifluoro-3-hydroxybenzoic acid (II). This was converted to ethyl ester (III) by refluxing in EtOH in the presence of H2SO4. Condensation of (III) with chlorodifluoromethane and NaH in hot DMF produced the corresponding difluoromethyl ether, and subsequent basic hydrolysis of the ethyl ester yielded 3- (difluoromethoxy) -2, 4,5-trifluorobenzoic acid (IV). Alternatively, acid (II) was converted to acid chloride with SOCl2 and subsequently condensed with ammonia to give amide (V). After formation of the difluoromethyl ether (VI) under similar conditions as above, acid (IV) was obtained by diazotization of the amide function of (VI) in hot sulfuric acid. The difluoromethoxy acid (IV) was also prepared by direct alkylation of hydroxy acid (II) with chlorodifluoromethane in the presence of NaOH in hot DMF. acid (IV) was activated as the corresponding acid chloride (VII) with SOCl2. Condensation of acid chloride (VII) with the magnesium salt of diethyl malonate gave rise to the benzoylmalonate (VIII). Further decarbethoxylation of (VIII) by heating in the presence of p-toluenesulfonic acid yielded keto ester (IX). This was condensed with triethyl orthoformate in the presence of Ac2O to give the ethoxyacrylate (X), which was converted to enamine (XII) by treatment with cyclopropylamine (XI). The target quinolone system (XIII) was then obtained by intramolecular cyclization of (XII) in the presence of NaH. Then, ethyl ester (XII) cleavage using boron trifluoride etherate provided the key quinolonecarboxylic acid boron chelate (XIV)

Route 
US5073556A / US5348961A.
1 to 8 of 8
Patent ID Date Patent Title
US2011159049 2011-06-30 PHARMACEUTICAL COMPOSITION
US2010330165 2010-12-30 USE OF CHEMOTHERAPEUTIC AGENTS
US2007196504 2007-08-23 PHARMACEUTICAL COMPOSITION
US2007197501 2007-08-23 Use Of Chemotherapeutic Agents
US2007148235 2007-06-28 PHARMACEUTICAL COMPOSITION
US2005152975 2005-07-14 Pharmaceutical composition
US2004022848 2004-02-05 Medicinal composition
US2003045544 2003-03-06 Use of chemotherapeutic agents

//////CS 940, Quinolone antibiotic , CADROFLOXACIN, NDA

CC1CN(CCN1)C2=C(C=C3C(=C2OC(F)F)N(C=C(C3=O)C(=O)O)C4CC4)F


Filed under: NDA Tagged: CADROFLOXACIN, CS 940, NDA, Quinolone antibiotic

Arbaclofen

$
0
0

Arbaclofen placarbil.svg

Arbaclofen placarbil

(3R)-3-(4-chlorophenyl)-4-[[(1S)-2-methyl-1-(2-methylpropanoyloxy)propoxy]carbonylamino]butanoic acid

NDA filed

A GABA (B) receptor agonist potentially for the treatment of muscle spasticity.

AGI-006; STX-209; OS-440

CAS No. 69308-37-8 free

847353-30-4 placarbil

Arbaclofen placarbil (ar-bac-loe-fen pla-kar-bil, also known as XP19986) is a prodrug of Rbaclofen. Arbaclofen placarbil possesses more favorable pharmacokinetic profile than baclofen, with less fluctuations in plasma drug levels. It was being developed as a potential treatment for patients with GERD and spasticity due to multiple sclerosis; however, in May 2013 XenoPort announced the termination of development because of unsuccessful results in phase III clinical trials.[1]

Arbaclofen Placerbil is a prodrug of Arbaclofen, which is a selective gamma-amino-butyric acid type B receptor agonist and the R-enantiomer of baclofen. It was discovered, and has been patented by XenoPort as a new chemical entity with an improved pharmacokinetic profile compared to baclofen, which allows for sustained release properties. ArbaclofenPlacerbil was believed to have therapeutic potential in treating gastroesophogeal reflux disease (GERD) and plasticity; however due to discouraging clinical trial results, the drug was abandoned by XenoPort in 2011 for the treatment of GERD. On May 20th, 2013, XenoPort announced plans to terminate the development of Arbaclofen Placerbil for the treatment of multiple sclerosis.

Autism spectrum disorder (ASD) is a behaviorally defined disorder which has increased in prevalence over the last two decades. Despite decades of research, no effective treatment is currently available. Animal models, as well as other lines of evidence, point to abnormalities in the balance of cortical excitation to inhibition in individuals with ASD, with this imbalance resulting in an overall increase in cortical excitation. To reduce cortical excitatory glutamate pathways, arbaclofen, a selective agonist of the gamma aminobutyric acid receptor type B, has been developed. This article reviews the evidence for this treatment for ASD using a systematic review methodology. Overall, a systematic search of the literature revealed 148 relevant references with the majority of these being review papers or news items that mentioned the potential promise of arbaclofen. Five original studies were identified, four of which used STX209, a form of arbaclofen developed by Seaside Therapeutics, Inc., and one which used R-baclofen. In an animal model, treatment of Fragile X, a genetic disease with ASD features, demonstrated a reversal of behavioral, neurological, and neuropathological features associated with the disease. One double-blind, placebo-controlled study treated children and adults with Fragile X. Results from this study were promising, with signs of improvement in social function, especially in the most severely socially impaired. Two studies, one open-label and one double-blind, placebo-controlled, were conducted in children, adolescents, and young adults with ASD. These studies suggested some improvements in socialization, although the effects were limited and may have been driven by individuals with ASD that were higher-functioning. These studies and others that have used arbaclofen for the treatment of gastroesophageal reflux suggest that arbaclofen is safe and well-tolerated. Clearly, further clinical studies are needed in order to refine the symptoms and characteristics of children with ASD that are best treated with arbaclofen.

Arbaclofen placarbil.png

 Fig. 1.

Fig. 1.

The Structures of R-baclofen (1), arbaclofen placarbil (2), R-baclofen lactam (3), and the potential γ-hydroxy metabolite of R-baclofen (4).

Route 2

Reference:1. Chem. Pharm. Bull. 1995, 43, 1302-1306.

Route 3
Route 4

Reference:1. J. Am. Chem. Soc. 2005, 127, 119-125.

2. WO2007066828A1 / US2009137819A1.

Route 5

Reference:1. US2012029230A1

Route 1

Reference:1. Tetrahedron-Asymmetr. 1992, 3, 1213-1221.

2. Tetrahedron Lett. 1991, 32, 6949-6952.

.

References

Arbaclofen placarbil
Arbaclofen placarbil.svg
Systematic (IUPAC) name
(3R)-3-(4-chlorophenyl)-4-[[[(1S)-2-methyl-1-[(2-methylpropanoyl)oxy]propoxy]carbonyl]amino]butanoic acid
Clinical data
Pregnancy
category
  • N/A
Legal status
Legal status
  • Development terminated
Identifiers
CAS Number 847353-30-4
ATC code none
PubChem CID 11281011
ChemSpider 9456008
KEGG D08861 Yes
ChEMBL CHEMBL2107312 Yes
Chemical data
Formula C19H26ClNO6
Molar mass 399.86 g/mol

///////AGI-006,  STX-209,  OS-440, Arbaclofen, autism spectrum disorder, Fragile X, gamma-aminobutyric acid, arbaclofen, R-baclofen, STX209

CC(C)[C@@H](OC(=O)C(C)C)OC(=O)NC[C@H](CC(=O)O)C1=CC=C(C=C1)Cl

DISCLAIMER

I , Dr A.M.Crasto is writing this blog to share the knowledge/views, after reading Scientific Journals/Articles/News Articles/Wikipedia. My views/comments are based on the results /conclusions by the authors(researchers). I do mention either the link or reference of the article(s) in my blog and hope those interested can read for details. I am briefly summarising the remarks or conclusions of the authors (researchers). If one believe that their intellectual property right /copyright is infringed by any content on this blog, please contact or leave message at below email address amcrasto@gmail.com. It will be removed ASAP

Filed under: NDA Tagged: AGI-006, Arbaclofen, autism spectrum disorder, Fragile X, gamma-aminobutyric acid, OS-440, R-baclofen, STX-209, STX209

FDA issues rule for data collection of antimicrobial sales and distribution by animal species

$
0
0

05/10/2016 09:28 AM EDT
Additional data help further target efforts to ensure judicious use of medically important antimicrobials
The U.S. Food and Drug Administration (FDA) finalized a rule today that revises its annual reporting requirements for drug sponsors of all antimicrobials sold or distributed for use in animals intended for human consumption or food-producing animals. Companies are now required to provide estimates of sales broken down by major food-producing species (cattle, swine, chickens and turkeys) in addition to the overall estimates they already submit on the amount of antimicrobial drugs they sell or distribute for use in food-producing animals.

May 10, 2016

Release

The U.S. Food and Drug Administration finalized a rule today that revises its annual reporting requirements for drug sponsors of all antimicrobials sold or distributed for use in animals intended for human consumption or food-producing animals. Companies are now required to provide estimates of sales broken down by major food-producing species (cattle, swine, chickens and turkeys) in addition to the overall estimates they already submit on the amount of antimicrobial drugs they sell or distribute for use in food-producing animals.

The new sales data will improve the agency’s understanding of how antimicrobials are sold and distributed for use in major food-producing species and help further target efforts to ensure judicious use of medically important antimicrobials.

Section 105 of the Animal Drug User Fee Amendments of 2008 (ADUFA 105) requires antimicrobial drug sponsors to annually report to the FDA the amount of all antimicrobial drugs they sell and distribute for use in food-producing animals, including those antibiotics that are not used in human medicine. ADUFA 105 also requires the FDA to prepare summary reports of sales and distribution information received from drug sponsors each year, by antimicrobial class for classes with three or more distinct sponsors, and to provide those summaries to the public. Prior to finalizing this rule, animal drug sponsors were not required to submit sales or distribution data by particular species.

Adding the requirement for sponsors to report species-specific sales estimates will also complement the data collection plan the FDA is developing, as part of the National Strategy for Combating Antibiotic-Resistant Bacteria (CARB), with the U.S. Department of Agriculture and the Centers for Disease Control and Prevention, to obtain additional on-farm use and resistance data. The collection of data from multiple sources, including enhanced sales data from antimicrobial animal drug sponsors, is important for providing a comprehensive and science-based picture of antimicrobial drug use and resistance in animal agriculture.

“This information will further enhance FDA’s ongoing activities related to slowing the development of antimicrobial resistance to help ensure that safe and effective antimicrobial new animal drugs will remain available for use in human and animal medicine,” said Dr. William T. Flynn, D.V.M., M.S., deputy director for science policy in the FDA’s Center for Veterinary Medicine.

The final rule also includes a provision to improve the timeliness of annual reports by requiring the FDA to publish its summary report of the antimicrobial sales and distribution information it collects for each calendar year by Dec. 31 of the following year.

The rule was proposed in May 2015, and takes into consideration hundreds of public comments from the veterinary community, animal feed manufacturing and livestock production associations, drug manufacturers, consumer groups and other stakeholders. Drug sponsors are required to comply with the reporting requirements in the final rule when submitting their reports covering the period of calendar year 2016.

///////FDA ,  data collection, antimicrobial sales, distribution, animal species


Filed under: FDA 2016 Tagged: animal species, antimicrobial sales, data collection, distribution, fda

Buthionine Sulphoximine

$
0
0

Skeletal formula of buthionine sulfoximine

Buthionine Sulphoximine

NDA Filed in china

A gamma-glutamylcysteine synthetase inhibitor potentially for the treatment of solid tumors.

 NSC-326231; BSO

CAS No. 5072-26-4

BUTHIONINE SULFOXIMINE; DL-Buthionine-[S,R]-sulfoximine; 5072-26-4; Buthionine sulfoxamine; Buthionine-S,R-sulfoximine; Buthione sulfoximine;

Molecular Formula: C8H18N2O3S
Molecular Weight: 222.30512 g/mol

Buthionine sulfoximine (BSO) is a sulfoximine which reduces levels of glutathione and is being investigated as an adjunct withchemotherapy in the treatment of cancer.[1] The compound inhibits gamma-glutamylcysteine synthetase, the enzyme required in the first step of glutathione synthesis. Buthionine sulfoximine may also be used to increase the sensitivity of parasites to oxidativeantiparasitic drugs.[2]

Buthionine sulphoximine is an oncolytic agent in early clinical development at the National Cancer Institute (NCI) for the treatment of neuroblastoma in pediatric patients in combination with melphalan and bone marrow or peripheral stem cell transplantation.

DATA

STR1

STR1

1H NMR

STR1

13C NMR

Synthesis

Methionine and buthionine sulfoximines: Syntheses under mild and safe imidation/oxidation conditions
Advanced Synthesis&Catalysis (2014), 356, (10), 2209-2213

Abstract

Thumbnail image of graphical abstract

Methionine and buthionine sulfoximines (MSO and BSO) are non-natural amino acids known to inhibit the biosynthesis of glutathione (GSH). The current syntheses of these biologically active molecules involve harsh reaction conditions and the use of hazardous reagents for the sulfur imidation. Here, improved syntheses of MSO and BSO are presented including safe and mild one-pot imidation/oxidation sequences and single-step deprotections of three different functionalities.

Methionine and Buthionine Sulfoximines: Syntheses under Mild and Safe Imidation/Oxidation Conditions

  1. Laura Buglioni,
  2. Vincent Bizet and
  3. Carsten Bolm*

DOI: 10.1002/adsc.201400354

http://onlinelibrary.wiley.com/doi/10.1002/adsc.201400354/abstract

References

  1.  Defty, CL; Marsden, JR (2012). “Melphalan in regional chemotherapy for locally recurrent metastatic melanoma.”. Current topics in medicinal chemistry 12 (1): 53–60. PMID 22196271.
  2.  “Definition of buthionine sulfoximine – National Cancer Institute Drug Dictionary”.

BUTHIONINE SULFOXIMINE.png

Buthionine sulfoximine
Skeletal formula of buthionine sulfoximine
Ball-and-stick model of buthionine sulfoximine as a zwitterion
Names
IUPAC name
2-amino-4-(butylsulfonimidoyl)butanoic acid
Other names
BSO
Identifiers
5072-26-4 
ChEBI CHEBI:28714 Yes
ChemSpider 19896 Yes
Jmol 3D model Interactive image
MeSH Buthionine+sulfoximine
PubChem 21157
Properties
C8H18N2O3S
Molar mass 222.305 g/mol
Density 1.29 g/mL
Melting point 215 °C (419 °F; 488 K)
Boiling point 382.3 °C (720.1 °F; 655.5 K)
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).

////NSC-326231,  BSO, 5072-26-4, Butionine sulfoximine, Neuroblastoma

CCCCS(=N)(=O)CCC(C(=O)O)N


Filed under: Uncategorized Tagged: 5072-26-4, BSO, Butionine sulfoximine, Neuroblastoma, NSC-326231

EMA’s new Draft Guideline on the Sterilisation of Medicinal Products, APIs, Excipients and Primary Containers

$
0
0

DRUG REGULATORY AFFAIRS INTERNATIONAL

For medicinal products administrated in sterile form, the process to reduce the microbial level is a critical manufacturing step with regard to quality. The EMA has recently published the draft of a guideline on that topic which contains a range of clarifications. Read more about the coming requirements on sterilisation of medicinal products, APIs, excipients and final containers

see

http://www.gmp-compliance.org/enews_05350_EMA-s-new-Draft-Guideline-on-the-Sterilisation-of-Medicinal-Products–APIs–Excipients-and-Primary-Containers_15435,S-WKS_n.html

As referred to in the European Pharmacopoeia, the procedure for terminal sterilisation of a medicinal product, an API, or an excipient is generally the method of choice. Yet, this might be difficult in many cases for product stability reasons. That’s why other microbial reduction processes can be used like sterilising filtration or aseptic processing. So far, there has been some uncertainty about these methods and their acceptance in a marketing authorisation procedure or a variation application, and about which data have to be submitted.

EMA’s new draft guideline entitled “Guideline…

View original post 781 more words


Filed under: Uncategorized

USP publishes draft of a new general chapter for plastic components used in manufacturing

$
0
0

DRUG REGULATORY AFFAIRS INTERNATIONAL

In the Pharmacopoeial Forum (PF)  42(3) (May-June 2016) the USP General Chapters – Packaging and Distribution Expert Committee proposes a new general chapter  <661.3> Plastic Components and Systems Used in Pharmaceutical Manufacturing and a revised version of general chapter <1661> Evaluation of Plastic Packaging and Manufacturing Systems and Their Materials of construction with Respect to Their User Safety Impact. Read more about USPs Proposal on Plastic Components and Systems Used in Pharmaceutical Manufacturing.

<1661> Evaluation of Plastic Packaging and Manufacturing Systems and Their Materials of construction with Respect to Their User Safety Impact. Read more about USPs Proposal on Plastic Components and Systems Used in Pharmaceutical Manufacturing.

see

http://www.gmp-compliance.org/enews_05341_USP-publishes-draft-of-a-new-general-chapter–661.3–for-plastic-components-used-in-manufacturing_15303,15493,Z-PKM_n.html

In the Pharmacopoeial Forum (PF)  42(3) (May-June 2016) the USP General Chapters – Packaging and Distribution Expert Committee proposes a new chapter to address the qualification of plastic components used in the manufacture of APIs (pharmaceutical and…

View original post 754 more words


Filed under: Uncategorized

APIs from Legitimate and Reliable Sources

$
0
0

DRUG REGULATORY AFFAIRS INTERNATIONAL

APIs from Legitimate and Reliable Sources

1. Introduction

Counterfeit and sub-standard APIs are increasingly present. Not only are they a fact of non-compliance but also they form a serious and increasing risk for patient safety. Various initiatives have been taken such as the founding of the FDA Counterfeit Drug Task Force, the European Commission’s current “Public consultation in preparation of a legal proposal to combat counterfeit medicines for human use” and the WHO Program “IMPACT” (International Medical Products Anti-Counterfeiting Taskforce).

API =Active pharmaceutical ingredient (synonym: drug substance)

Counterfeit API =Active pharmaceutical ingredient for which source and/or quality are falsely represented on the label, on the certificate of analysis or otherwise

Rogue API =API that is counterfeit or severely, deliberately non-compliant.

This writeup focuses on the interaction between the API manufacturer and the medicinal product manufacturer and provides possible measures that may be taken by both partners in order to ensure only…

View original post 2,586 more words


Filed under: Uncategorized

BMS-520, a Potent and Selective Isoxazole-Containing S1P1 Receptor Agonist

$
0
0

img

BMS-520
CAS: 1236188-38-7
MF: C23H17F3N4O4
MW: 470.1202

Synonym: BMS-520; BMS 520; BMS520.

INNOVATOR Bristol-Myers Squibb Company

INVENTORS

Scott Hunter Watterson, Alaric J. Dyckman,William J. Pitts, Steven H. Spergel

1-[4-[5-[3-Phenyl-4-(trifluoromethyl)isoxazol-5-yl]-1,2,4-oxadiazol-3-yl]benzyl]azetidine-3-carboxylic acid

 1-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)-1,2,4-oxadiazol-3-yl)benzyl)azetidine-3-carboxylic acid

US2011300165

1H NMR (500 MHz, DMSO-d6) δ: 3.20–3.46 (m, 5H), 3.66 (s, 2H), 7.53 (d, J = 8.25 Hz, 2H), 7.60–7. 70 (m, 5H), and 8.06 (d, J = 7. 70 Hz, 2H);

MS m/e 471(M+H+);

HPLC (XBridge 5 μ C18 4.6 × 50 mm, 4 mL/min, solvent A: 10% MeOH/water with 0.2% H3PO4, solvent B: 90% MeOH/water with 0.2% H3PO4, gradient with 0–100% B over 4 min): 3.14 min;

Anal. Calcd for C23H17N4O4F3•0.01 EtOH: C, 58.72; H, 3.65; N, 11.90. Found: C, 58.63; H, 3.41; N, 11.84.

BMS-520 is a potent and selective S1P1 agonist. BMS-520 demonstrated impressive efficacy when administered orally in a rat model of arthritis and in a mouse experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis. Agonism of S1P1, in particular, has been shown to play a significant role in lymphocyte trafficking from the thymus and secondary lymphoid organs, resulting in immunosuppression.

Sphingosine-1 -phosphate (SlP) has been demonstrated to induce many cellular effects, including those that result in platelet aggregation, cell proliferation, cell morphology, tumor cell invasion, endothelial cell and leukocyte chemotaxis, endothelial cell in vitro angiogenesis, and lymphocyte trafficking. SlP receptors are therefore good targets for a wide variety of therapeutic applications such as tumor

15 growth inhibition, vascular disease, and autoimmune diseases. SlP signals cells in part via a set of G protein-coupled receptors named SlPi or SlPl, SIP2 or S1P2, SIP3 or S1P3, SlP4 Or S1P4, and SlP5 or S1P5 (formerly called EDG-I, EDG-5, EDG-3, EDG-6, and EDG-8, respectively). [0003] SlP is important in the entire human body as it is also a major regulator of

20 the vascular and immune systems. In the vascular system, SlP regulates angiogenesis, vascular stability, and permeability. In the immune system, SlP is recognized as a major regulator of trafficking of T- and B-cells. SlP interaction with its receptor SlPi is needed for the egress of immune cells from the lymphoid organs (such as thymus and lymph nodes) into the lymphatic vessels. Therefore, modulation

25 of SlP receptors was shown to be critical for immunomodulation, and SlP receptor modulators are novel immunosuppressive agents.

The SlPi receptor is expressed in a number of tissues. It is the predominant family member expressed on lymphocytes and plays an important role in lymphocyte trafficking. Downregulation of the SlPi receptor disrupts lymphocyte

30 migration and homing to various tissues. This results in sequestration of the lymphocytes in lymph organs thereby decreasing the number of circulating lymphocytes that are capable of migration to the affected tissues. Thus, development of an SlPi receptor agent that suppresses lymphocyte migration to the target sites associated with autoimmune and aberrant inflammatory processes could be efficacious in a number of autoimmune and inflammatory disease states. [0005] Among the five SlP receptors, SlPi has a widespread distribution and is highly abundant on endothelial cells where it works in concert with S IP3 to regulate cell migration, differentiation, and barrier function. Inhibition of lymphocyte recirculation by non-selective SlP receptor modulation produces clinical immunosuppression preventing transplant rejection, but such modulation also results in transient bradycardia. Studies have shown that SlPi activity is significantly correlated with depletion of circulating lymphocytes. In contrast, SIP3 receptor agonism is not required for efficacy. Instead, SIP3 activity plays a significant role in the observed acute toxicity of nonselective SlP receptor agonists, resulting in the undesirable cardiovascular effects, such as bradycardia and hypertension. (See, e.g., Hale et al, Bioorg. Med. Chem. Lett., 14:3501 (2004); Sanna et al, J. Biol. Chem., 279: 13839 (2004); Anliker et al., J. Biol. Chem., 279:20555 (2004); Mandala et al., J. Pharmacol. Exp. Ther., 309:758 (2004).)

An example of an SlPi agonist is FTY720. This immunosuppressive compound FTY720 (JPI 1080026-A) has been shown to reduce circulating lymphocytes in animals and humans, and to have disease modulating activity in animal models of organ rejection and immune disorders. The use of FTY720 in humans has been effective in reducing the rate of organ rejection in human renal transplantation and increasing the remission rates in relapsing remitting multiple sclerosis (see Brinkman et al., J. Biol. Chem., 277:21453 (2002); Mandala et al., Science, 296:346 (2002); Fujino et al., J. Pharmacol, and Exp. Ther., 305:45658 (2003); Brinkman et al., Am. J. Transplant, 4: 1019 (2004); Webb et al., J.

Neuroimmunol, 153: 108 (2004); Morris et al., Eur. J. Immunol, 35:3570 (2005); Chiba, Pharmacology & Therapeutics, 108:308 (2005); Kahan et al., Transplantation, 76: 1079 (2003); and Kappos et al., N. Engl. J. Med, 335: 1124 (2006)). Subsequent to its discovery, it has been established that FTY720 is a prodrug, which is phosphorylated in vivo by sphingosine kinases to a more biologically active agent that has agonist activity at the SlPi, SIP3, SlP4, and SIP5 receptors. It is this activity on the SlP family of receptors that is largely responsible for the pharmacological effects of FTY720 in animals and humans.

Clinical studies have demonstrated that treatment with FTY720 results in bradycardia in the first 24 hours of treatment (Kappos et al., N. Engl. J. Med., 335: 1124 (2006)). The observed bradycardia is commonly thought to be due to agonism at the SIP3 receptor. This conclusion is based on a number of cell based and animal experiments. These include the use of SIP3 knockout animals which, unlike wild type mice, do not demonstrate bradycardia following FTY720 administration and the use of SlPi selective compounds. (Hale et al., Bioorg. Med. Chem. Lett., 14:3501 (2004); Sanna et al., J. Biol. Chem., 279: 13839 (2004); and Koyrakh et al., Am. J. Transplant., 5:529 (2005)).

The following applications have described compounds as SlPi agonists: WO 03/061567 (U.S. Publication No. 2005/0070506), WO 03/062248 (U.S. Patent No. 7,351,725), WO 03/062252 (U.S. Publication No. 2005/0033055), WO 03/073986 (U.S. Patent No. 7,309,721), WO 03/105771, WO 05/058848, WO

06/047195, WO 06/100633, WO 06/115188, WO 06/131336, WO 2007/024922, WO 07/116866, WO 08/023783 (U.S. Publication No. 2008/0200535), and WO 08/074820. Also see Hale et al., J. Med. Chem., 47:6662 (2004). [0009] There still remains a need for compounds useful as SlPi agonists and yet having selectivity over Sl P3.

SCHEMBL2572635.png

BMS 520

Paper

Journal of Medicinal Chemistry (2016), 59(6), 2820-2840

Potent and Selective Agonists of Sphingosine 1-Phosphate 1 (S1P1): Discovery and SAR of a Novel Isoxazole Based Series

Bristol-Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
J. Med. Chem., 2016, 59 (6), pp 2820–2840
DOI: 10.1021/acs.jmedchem.6b00089
Publication Date (Web): February 28, 2016
Copyright © 2016 American Chemical Society
*Phone: 609-252-6778. E-mail: scott.watterson@bms.com.

Abstract

Abstract Image

Sphingosine 1-phosphate (S1P) is the endogenous ligand for the sphingosine 1-phosphate receptors (S1P1–5) and evokes a variety of cellular responses through their stimulation. The interaction of S1P with the S1P receptors plays a fundamental physiological role in a number of processes including vascular development and stabilization, lymphocyte migration, and proliferation. Agonism of S1P1, in particular, has been shown to play a significant role in lymphocyte trafficking from the thymus and secondary lymphoid organs, resulting in immunosuppression. This article will detail the discovery and SAR of a potent and selective series of isoxazole based full agonists of S1P1. Isoxazole 6d demonstrated impressive efficacy when administered orally in a rat model of arthritis and in a mouse experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis.

SEE…..http://pubs.acs.org/doi/abs/10.1021/acs.jmedchem.6b00089

PAPER

Abstract Image

This article reports an efficient scale-up synthesis of 1-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)-1,2,4-oxadiazol-3-yl)benzyl)azetidine-3-carboxylic acid (BMS-520), a potent and selective isoxazole-containing S1P1 receptor agonist. This process features a highly regioselective cycloaddition leading to a key intermediate, ethyl 3-phenyl-4-(trifluoromethyl)isoxazole-5-carboxylate, a chemo-selective hydrolysis of its regioisomers, as well as an improved method for 1,2,4-oxadiazole formation, relative to the original synthesis. The improved process was applied to the preparation of multiple batches of BMS-520 for preclinical toxicological studies.

An Efficient Scale-Up Synthesis of BMS-520, a Potent and Selective Isoxazole-Containing S1P1 Receptor Agonist

Discovery Chemistry, Bristol-Myers Squibb Research and Development, Route 206 and Provinceline Road, Princeton, New Jersey 08543, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.6b00112
Publication Date (Web): May 05, 2016
Copyright © 2016 American Chemical Society
.HPLC purity 99.8%; tR= 7.62 min (method A); 99.9%; tR = 8.45 min (method B);
LCMS (ESI) m/z calcd for C23H17F3N4O4 [M + H]+ 445.2. Found: 471.3.
1H NMR (500 MHz, DMSO-d6) δ ppm 3.20–3.46 (m, 5H), 3.66 (s, 2H), 7.53 (d, J = 8.25 Hz, 2H), 7.60–7.70 (m, 5H), and 8.06 (d, J = 7.70 Hz, 2H).
Anal. Calcd for C23H17N4O4F3, 0.44% water: C, 58.42; H, 3.70; N, 11.83. Found: C, 58.52; H, 3.43; N, 11.86.

PATENT

WO 2010085581

Scheme 1

Figure imgf000037_0001

Scheme 2

Figure imgf000038_0001

Scheme 3

Figure imgf000039_0001

Scheme 4

Figure imgf000040_0001
Figure imgf000040_0002

Scheme 5

Figure imgf000041_0001

Scheme 6

Figure imgf000042_0001

Example 1

l-(4-(5-(3-Phenyl-4-(trifluoromethyl)isoxazol-5-yl)-l,2,4-oxadiazol-3- yl)benzyl)azetidine-3-carboxylic acid

Figure imgf000049_0001

1-A. 4,4,4-Trifluorobut-2-yn-l-ol

Figure imgf000049_0002

To a solution of diisopropylamine (24.7 mL, 176 mmol) in ether (100 mL) at -78 0C was added a 1OM solution of butyllithium in ether (17.6 mL, 176 mmol) over 5 min. After 10 min. at -78°C, 2-bromo-3,3,3-trifluoroprop-l-ene (14.0 g, 80 mmol) was added to the pale yellow solution. After an additional 10 min., paraformaldehyde (2.40 g, 80 mmol) was added, the dry-ice bath was removed, and the reaction mixture was stirred at room temperature overnight. As the reaction mixture approached room temperature, it became dark in color. The reaction was quenched with a IN aqueous solution of hydrochloric acid (100 mL), diluted with ether (500 mL), washed with a IN aqueous solution of hydrochloric acid (2 x 100 mL), washed with brine 100 mL, and dried over anhydrous sodium sulfate. Concentration under reduced pressure afforded a dark liquid which was distilled under low-vacuum (-50 Torr, ~50 0C) to give 4,4,4-trifluorobut-2-yn-l-ol (7.1 g, 57.2 mmol, 72 % yield) as a pale yellow liquid. 1H NMR (500 MHz, CDCl3) δ ppm 2.31 (br. s., IH) and 4.38 – 4.42 (m, 2H).An Alternative Preparation of 1 -A: 4,4,4-Trifluorobut-2-yn- 1 -ol

HO

-CF, (1-A) [00117] To an ether (pre-dried over magnesium sulfate) solution of phenanthroline (2.16 mg, 0.012 mmol) (indicator) at -78°C under nitrogen was added a 2M solution of n-butyl lithium in pentane. An orange color immediately appeared. Trifluoromethylacetylene gas was bubbled through the solution at -78°C. After ~4 min. of gas introduction, the orange color almost completely disappeared, the reaction solution became cloudy (due to some precipitation), and a pale light orange color persisted. Paraformaldehyde was added, and the dry ice/isopropanol bath was removed after 5 min. and replaced with a 00C ice-bath. Stirring was continued for 45 min., the ice bath was removed, and stirring was continued for an additional 1.25 h. The reaction flask was immersed in a 00C ice bath, and a saturated aqueous solution of ammonium chloride (20.0 mL) was added. The layers were separated, and the organic layer was washed with water (2x), washed with brine, and dried over anhydrous sodium sulfate. Concentration under low-vacuum (~50 Torr) without heat afforded a dark brown liquid which was purified by vacuum distillation (~50 Torr, -50 0C) to give 4,4,4-trifluorobut-2-yn-l-ol (7.1 g, 57.2 mmol, 72 % yield) as a colorless liquid.

1-B. N-Hydroxybenzimidoyl chloride

Figure imgf000050_0001

This compound was prepared according to the method of Liu, K.C. et al, J. Org. Chem., 45:3916-1918 (1980).To a colorless, homogeneous solution of (E)-benzaldehyde oxime (24.4 g, 201 mmol) in N,N-dimethylformamide (60 mL) at room temperature was added N- chlorosuccinimide (26.9 g, 201 mmol) portion-wise over 30 min. During each addition, the reaction mixture would turn yellow and then gradually return to near colorlessness. Additionally, an exotherm was noted with each portion added to ensure that the reaction initiated after the addition of N-chlorosuccinimide. An ice bath was available, if required, to cool the exotherm. After the addition was complete, the homogeneous reaction mixture was stirred overnight at room temperature. The reaction mixture was diluted with 250 mL of water and extracted with ether (3 x 100 mL). The organic layers were combined, washed with water (2 x 100 mL), washed with a 10% aqueous solution of lithium chloride (2 x 100 mL), and washed with brine (100 mL). The aqueous layers were back extracted with ether (100 mL), and the combined organic layers (400 mL) were dried over anhydrous sodium sulfate. Concentration under reduced pressure afforded (Z)-N-hydroxybenzimidoyl chloride (30.84 g, 198 mmol, 98 % yield) as a fluffy, pale yellow solid. The product had an HPLC ret. time = 1.57 min. – Column: CHROMOLITH® SpeedROD 4.6 x 50 mm (4 min.); Solvent A = 10% MeOH, 90% H2O, 0.1% TFA; Solvent B = 90% MeOH, 10% H2O, 0.1% TFA. LC/MS M+1 = 155.8. 1H NMR (500 MHz, DMSO-d6) δ ppm 7.30 – 7.64 (m, 3H), 7.73 – 7.87 (m, 2H), and 12.42 (s, IH).

l-C. 3-Phenyl-4-(trifluoromethyl)isoxazol-5-yl)methanol

Figure imgf000051_0001

To a pale yellow, homogeneous mixture of N-hydroxybenzimidoyl chloride (5.50 g, 35.4 mmol) and 4,4,4-trifluorobut-2-yn-l-ol (5.46 g, 39.6 mmol) in dichloroethane (85 mL) in a 250 mL round bottom flask at 700C was added triethylamine (9.85 mL, 70.7 mmol) in 22 mL of dichloroethane over 2.5 h via an addition funnel (the first -50% over 2 h and the remaining 50% over 0.5 h). After the addition was complete, the reaction mixture was complete by HPLC (total time at 700C was 3 h). The reaction mixture was stirred at room temperature overnight. [00121] The reaction mixture was diluted with dichloromethane (100 mL), washed with water (100 mL), and the organic layer was collected. The aqueous layer was extracted with dichloromethane (2 x 50 mL), and the combined organic layers were dried over anhydrous sodium sulfate. The solvent was removed under reduced pressure. Analysis indicated that the product mixture was composed of a 86: 14 mixture of the desired regioisomer (1-C), (3-phenyl-4-(trifluoromethyl)isoxazol-5- yl)methanol, and the undesired regioisomer, (3-phenyl-5-(trifluoromethyl)isoxazol-4- yl)methanol. The mixture was purified by silica gel chromatography using a mixture of ethyl acetate and hexane (1% to pack and load – 5% – 9% – 12%) to afford (3- phenyl-4-(trifluoromethyl)isoxazol-5-yl)methanol (5.34 g, 21.96 mmol, 62.1 % yield) as a pale yellow oil. The compound had an HPLC ret. time = 1.91 min. – Column: CHROMOLITH® SpeedROD 4.6 x 50 mm (4 min.); Solvent A = 10% MeOH, 90% H2O, 0.1% TFA; Solvent B = 90% MeOH, 10% H2O, 0.1% TFA. LC/MS M+1 =244.2. 1H NMR (500 MHz, CDCl3) δ ppm 2.21 (br. s., IH), 4.97 (s, 2H), 7.47 – 7.56 (m, 3H), and 7.65 (d, J=6.60 Hz, 2H).

1-D. 3-Phenyl-4-(trifluoromethyl)isoxazole-5-carboxylic acid

Figure imgf000052_0001

Preparation of Jones’ Reagent

To an orange, homogeneous solution of chromium trioxide (12.4 g, 0.123 mol) in water (88.4 mL) at 00C was added sulfuric acid (10.8 mL) dropwise via addition funnel over 30 min. with stirring. The addition funnel was rinsed with water

(1 mL) to give 1.23 M solution of Jones’ Reagent (0.123 mol of reagent in 100 mL of solvent).

To a solution of (3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)methanol

(5.24 g, 21.6 mmol) in acetone (75 mL) at room temperature (immersed in a water bath) was added Jones’ Reagent (43.8 mL, 53.9 mmol) via addition funnel slowly over 1.5 h. The dark reaction mixture was stirred at room temperature overnight. By HPLC, the reaction was 93% complete. An additional 0.5 equivalents (9 mL) of the Jones’ Reagent was added. After 1 h, the reaction was 95% complete. After an additional 3h, the reaction was 96% complete. An additional 0.5 equivalents (9 mL) of the Jones’ Reagent was added. The reaction mixture was stirred for an additional 2.5 h. By HPLC, the reaction was 97% complete. Isopropyl alcohol (6 mL) was added, and the mixture was stirred for 90 min, resulting in a dark green precipitate. The mixture was diluted with ether (600 mL), washed with a 2% aqueous solution of sodium hydrogen sulfite (5 x 100 mL), and the organic layer was collected. The aqueous layer was back-extracted with ether (2 x 100 mL). By HPLC, there was no additional product in the aqueous layer. The combined organic layers were washed with water (100 mL), washed with a saturated aqueous solution of brine (100 mL), and dried over anhydrous sodium sulfate. The aqueous layer was back-extracted with ether (100 mL), and the organic layer was added to the previous organic layers. The solution was concentration under reduced pressure to give 3-phenyl-4-

(trifluoromethyl)isoxazole-5-carboxylic acid as an off-white solid. The solid was diluted with dichloromethane (200 mL), washed with a 2% aqueous solution of sodium hydrogen sulfite, washed with brine, and dried over anhydrous sodium sulfate. Concentration under reduced pressure afforded 3-phenyl-4- (trifluoromethyl)isoxazole-5-carboxylic acid (3.84 g, 14.93 mmol, 69.3 % yield) as a pale yellow solid. The product was 96% pure by HPLC with a ret. time = 1.60 min. – Column: CHROMOLITH® SpeedROD 4.6 x 50 mm (4 min.); Solvent A = 10% MeOH, 90% H2O, 0.1% TFA; Solvent B = 90% MeOH, 10% H2O, 0.1% TFA. LC/MS M+1 = 258.2. [00124] The sodium hydrogen sulfite aqueous layer still contained a significant amount of product. The brine layer contained no additional product and was discarded. The aqueous layer was saturated with sodium chloride, the pH was adjusted to -3.5, and the solution was extracted with ether (3 x 100 mL). The organic layer was dried over anhydrous sodium sulfate and concentrated to afford additional 3-phenyl-4-(trifluoromethyl)isoxazole-5-carboxylic acid (1.12 g, 4.36 mmol, 20.21 % yield) as a white solid. The product was >99% pure by HPLC with a ret. time = 1.60 min. – Column: CHROMOLITH® SpeedROD 4.6 x 50 mm (4 min.); Solvent A = 10% MeOH, 90% H2O, 0.1% TFA; Solvent B = 90% MeOH, 10% H2O, 0.1% TFA. LC/MS M+1 = 258.1. 1H NMR (500 MHz, DMSO-(I6) δ ppm 7.55 – 7.63 (m, 5H).  The products were combined to give 4.96 g (90% yield) of 3-phenyl-4- (trifluoromethyl)isoxazole-5-carboxylic acid.

An Alternative Preparation of 1-D: 3 -Phenyl -4-(trifluoromethyl)isoxazole-5- carboxylic acid starting with (3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)methanol

Figure imgf000054_0001

A mixture of (3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)methanol (2.1 g, 8.64 mmol), TEMPO (0.094 g, 0.604 mmol), and a sodium phosphate buffer (0.67M) (32.2 mL, 21.59 mmol) was heated to 35°C. A solution of sodium phosphate buffer (40 mL, pH -6.5) consisting of a 1: 1 solution OfNaH2PO4 (20 mL, 0.67M) and Na2HPO4 (20 mL, 0.67M) was prepared in acetonitrile (30 mL) was prepared prior to use. Solutions of sodium chlorite (3.91 g, 34.5 mmol) in water (4.5 mL) and bleach (4.3 mL, 6% wt.) were added simultaneously over 40min. The reaction was monitored by HPLC, and after 2 h, -30% of the starting material remained. After 6 h, 10% remained. Additional bleach (100 μL) was added, and the reaction mixture was left at room temperature overnight. [00127] Additional bleach (100 μL) was added. The resulting mixture was allowed to stir at 35°C for additional 2 h. HPLC indicated complete conversion. The reaction was quenched by the slow addition of a solution of sodium sulfite (2.07 mL, 43.2 mmol) in water (90 mL) at 00C, resulting in the disappearance of the brown reaction color. The solvent was removed under reduced pressure, and the remaining aqueous residue was extracted with ethyl acetate (3 x 40 mL). The organic layers were combined, washed with water (8 mL), washed with brine (8 mL), and dried over anhydrous sodium sulfate. Concentration under reduced pressure afforded 3 -phenyl – 4-(trifluoromethyl)isoxazole-5-carboxylic acid (2.2 g, 8.55 mmol, 99 % yield) as a pale yellow solid. An alternative procedure for the for the preparation of 3-phenyl-4-(trifluoromethyl) isoxazole-5-carboxylic acid starting with 4,4,4-trifluorobut-2ynoate (1-D)

Figure imgf000055_0001

Alt.1 -D- 1. Ethyl 3 -phenyl-4-(trifluoromethyl)isoxazole-5-carboxylate

Figure imgf000055_0002

To a pale yellow mixture of (Z)-N-hydroxybenzimidoyl chloride (1.04 g, 6.68 mmol) and ethyl 4,4,4-trifluorobut-2-ynoate (1.238 g, 7.45 mmol) in diethyl ether (20 mL) at room temperature was added triethylamine (1.86 mL, 13.4 mmol) over 15 min., resulting in a precipitant. After the addition was complete, the pale yellow slurry was stirred at room temperature over a weekend. The heterogeneous reaction mixture was filtered under reduced pressure to remove the triethylamine hydrochloride salt, and the filtrate was concentrated to give the product mixture as a dark yellow, viscous oil (2.03 g). By HPLC, the reaction mixture was composed of a mixture of the desired regioisomer, ethyl 3-phenyl-4-(trifluoromethyl)isoxazole-5- carboxylate, and the undesired regioisomer, ethyl 3-phenyl-5- (trifluoromethyl)isoxazole-4-carboxylate, in an approximately 15:85 ratio. The compound mixture was dissolved in hexane and sonicated for 5 min. The hexane was decanted off, and the dark red, oily residue was found to have only trace product by HPLC. The hexane was removed under reduced pressure, and the residue (1.89 g) was purified by preparative HPLC. The desired fractions containing ethyl 3-phenyl- 4-(trifluoromethyl)isoxazole-5-carboxylate were concentrated, and the residue was diluted with dichloromethane, washed with a saturated aqueous solution of sodium bicarbonate, and dried over anhydrous sodium sulfate. Concentration under reduced pressure afforded ethyl 3-phenyl-4-(trifluoromethyl) isoxazole-5-carboxylate (0.087 g, 0.305 mmol, 4.6 % yield) as a pale yellow solid. The compound had an HPLC ret. time = 2.88 min. – Column: CHROMOLITH® SpeedROD 4.6 x 50 mm (4 min.); Solvent A = 10% MeOH, 90% H2O, 0.1% TFA; Solvent B = 90% MeOH, 10% H2O, 0.1% TFA. 1H NMR (400 MHz, CDCl3) δ ppm 1.46 (t, J=7.15 Hz, 3H), 4.53 (q, J=7.03 Hz, 2H), 7.48 – 7.55 (m, 3H), and 7.58 (d, J=7.53 Hz, 2H).

An Alternative Preparation of 1-D-l : Ethyl 3-phenyl-4-(trifluoromethyl)isoxazole-5- carboxylic acid starting with ethyl 4,4,4-trifluorobut-2-enoate

1-D-l. Ethyl 2,3-dibromo-4,4,4-trifluorobutanoate

Br L /COOEt

Br (1-D-l) [00129] Bromine (18.4 mL, 357 mmol) was added dropwise over 30 minutes to a solution of (E)-ethyl 4,4,4-trifluorobut-2-enoate (50 g, 297 mmol) in carbon tetrachloride (50 mL) at room temperature under nitrogen. The resulting dark red solution was refluxed for 4 hours. Additional bromine (2ml) was added and heating was continued until the HPLC analysis showed that the starting material had been consumed. The reaction mixture was concentrated under reduced pressure to give light brown oil which used in the next step without purification. HPLC (XBridge 5μ Cl 8 4.6×50 mm, 4 mL/min, Solvent A: 10 % MeOH/water with 0.2 % H3PO4, Solvent B: 90 % MeOH/water with 0.2 % H3PO4, gradient with 0-100 % B over 4 minutes): 2.96 and 3.19 minutes.

l-D-2. (Z/E)-Ethyl 2-bromo-4,4,4-trifluorobut-2-enoate

,COOEt

F3C

Br (l-D-2)

To a solution of ethyl 2,3-dibromo-4,4,4-trifluorobutanoate (1-B-l) in hexane (200 mL) cooled to 00C was added triethylamine (49.7 ml, 357mmol) drop- wise over 35 minutes, during which time a white precipitate formed. The reaction mixture was stirred for an additional 2 hours until LC indicated complete conversion. The solid was filtered and rinsed with hexane (3 x 5OmL), and the filtrate was concentrated and passed through a short silica gel pad eluting with 10% ethyl acetate/hexane to give (Z/E)-ethyl 2-bromo-4,4,4-trifluorobut-2-enoate (65.5 g, 265mmol, 89 % yield for two steps) as a colorless oil. Alternatively, the crude product can be purified by distillation (85 0C / -60 mmHg). 1H NMR (CDCl3, 400 MHz) 5 7.41 (q, IH, J= 7.28 Hz), 4.35 (q, 2H, J= 7.11 Hz), 1.38 (t, 3H, J= 7.15 Hz); HPLC (XBridge 5μ Cl 8 4.6×50 mm, 4 mL/min, Solvent A: 10 % MeOH/water with 0.2 % H3PO4, Solvent B: 90 % MeOH/water with 0.2 % H3PO4, gradient with 0- 100 % B over 4 minutes): 3.09 minutes.

1-D-l. Ethyl 3 -phenyl -4-(trifluoromethyl)isoxazole-5-carboxylate

Figure imgf000057_0001

(Z/E)-Ethyl 2-bromo-4,4,4-trifluorobut-2-enoate, l-D-3, (39.7 g, 161 mmol) and N-hydroxybenzimidoyl chloride (30 g, 193mmol) were dissolved in ethyl acetate (15OmL). Indium (III) chloride (8.89 g, 40.2mmol) was added and the resulting mixture stirred for 60 minutes at RT under N2. Potassium hydrogen carbonate (32.2 g, 321mmol) was added to the reaction mixture which was allowed to stir overnight for 14 hours at RT. The solvent was removed in vacuo. The residue was re-suspended in 30OmL hexane and stirred for lOmiutes then filtered. The filter cake was washed with hexane (3X3 OmL) and the combined filtrate was concentrated in vacuo to give crude product, which was further purified with flash chromatography to generate 33g product (72%) as light yellowish oil as a mixture of the desired isomer 1-D-l and undesired isomer 1-D-la in a ratio of -30/1. MS m/e 286.06(M+H+); 1H NMR (CDCl3, 400 MHz) δ 7.56 (m, 5H), 4.53 (q, 2H, J= 7.3 Hz), 1.46 (t, 3H, J= 7.2 Hz); HPLC (XBridge 5μ C18 4.6×50 mm, 4 mL/min, Solvent A: 10 % MeOH/water with 0.2 % H3PO4, Solvent B: 90 % MeOH/water with 0.2 % H3PO4, gradient with 0-100 % B over 4 minutes): 3.57 minutes.

Alt.1-D. 3-Phenyl-4-(trifluoromethyl)isoxazole-5-carboxylic acid, lithium salt

Figure imgf000057_0002

A mixture of ethyl 3-phenyl-4-(trifluoromethyl)isoxazole-5-carboxylate, 1-D-l, (0.085 g, 0.298 mmol) and lithium hydroxide hydrate (0.013 g, 0.298 mmol) in methanol (2.0 mL) and water (1.0 mL) was stirred at room temperature overnight. The reaction mixture was concentrated to dryness to give 3-phenyl-4- (trifluoromethyl)isoxazole-5-carboxylic acid, lithium salt (0.079 g, 0.299 mmol, 100 % yield) as a pale yellow solid. The compound had an HPLC ret. time = 1.72 min. – Column: CHROMOLITH® SpeedROD 4.6 x 50 mm (4 min.); Solvent A = 10% MeOH, 90% H2O, 0.1% TFA; Solvent B = 90% MeOH, 10% H2O, 0.1% TFA. LC/MS M+1 = 258.0. 1H NMR (400 MHz, CDCl3) δ ppm 7.49 – 7.57 (m, 3H) and 7.58 – 7.62 (m, 2H).1-E. 3-Phenyl-4-(trifluoromethyl)isoxazole-5-carbonyl fluoride

Figure imgf000058_0001

To a mixture of 3-phenyl-4-(trifluoromethyl)isoxazole-5-carboxylic acid (3.00 g, 11.7 mmol) and pyridine (1.132 mL, 14.0 mmol) in dichloromethane (100 mL) at room temperature was added 2,4,6-trifluoro-l,3,5-triazine (cyanuric fluoride) (1.18 mL, 14.0 mmol). The reaction mixture was stirred at room temperature overnight, diluted with dichloromethane (300 mL), washed with an ice-cold solution of 0.5N aqueous hydrochloric acid (2 x 100 mL), and the organic layer was collected. The aqueous layer was back-extracted with dichloromethane (200 mL), and the combined organic layers were dried anhydrous sodium sulfate and concentrated to afford 3-phenyl-4-(trifluoromethyl)isoxazole-5-carbonyl fluoride (2.91 g, 11.2 mmol, 96 % yield) as a yellow, viscous oil. The product was found to react readily with methanol and on analysis was characterized as the methyl ester, which had an HPLC ret. time = 2.56 min. – Column: CHROMOLITH® SpeedROD 4.6 x 50 mm (4 min.); Solvent A = 10% MeOH, 90% H2O, 0.1% TFA; Solvent B = 90% MeOH, 10% H2O, 0.1% TFA. LC/MS M+1 = 272.3 (methyl ester).1-F. tert-Butyl l-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)-l,2,4-oxadiazol- 3-yl)-benzyl)azetidine-3-carboxylate

Figure imgf000059_0001

A suspension of 3-phenyl-4-(trifluoromethyl)isoxazole-5-carbonyl fluoride (2.91 g, 11.2 mmol), (Z)-tert-butyl 1-(4-(N’- hydroxycarbamimidoyl)benzyl)azetidine-3-carboxylate (Int. l, 3.43 g, 11.2 mmol), and Hunig’s Base (2.55 mL, 14.6 mmol) in acetonitrile (20 mL) was stirred at room temperature over the weekend. The reaction mixture had completely solidified (pinkish-tan in color), but was judged complete by HPLC and LCMS. The reaction mixture was partitioned between a saturated aqueous of sodium bicarbonate (150 mL) and dichloromethane (150 mL). The aqueous layer was extracted with dichloromethane (2 x 100 mL), and the combined organic layers were dried over anhydrous sodium sulfate. Concentration under reduced pressure afforded a tan solid which was purified by flash silica gel chromatography using a mixture of ethyl acetate in hexane (0-50%) to afford tert-butyl l-(4-(5-(3-phenyl-4-(trifluoromethyl) isoxazol-5-yl)-l,2,4-oxadiazol-3-yl)benzyl)azetidine-3-carboxylate (4.60 g; 78%) as a white, crystalline solid. The material was suspended in methanol (-75 mL) and was sonicated for 5 minutes. The MeOH was removed under reduce pressure, and the residue was re-suspended in methanol (-50 mL) with sonication. Vacuum filtration and drying afforded tert-butyl l-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)- l,2,4-oxadiazol-3-yl)benzyl)azetidine-3-carboxylate (4.04 g, 7.67 mmol, 68 % yield) as a white, crystalline solid. The methanol filtrate was concentrated to afford additional tert-butyl l-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)- 1,2,4- oxadiazol-3-yl)benzyl)azetidine-3-carboxylate (570 mg; 10%) as a slightly off- white solid. The compound had an HPLC retention time = 3.12 min. – Column: CHROMOLITH® SpeedROD 4.6 x 50 mm (4 min.); Solvent A = 10% MeOH, 90% H2O, 0.1% TFA; Solvent B = 90% MeOH, 10% H2O, 0.1% TFA. LC/MS M+1 =527.1. 1H NMR (500 MHz, CDCl3) δ ppm 1.47 (s, 9H) 3.28 – 3.37 (m, 3H), 3.60 (br. s., 2H), 3.74 (br. s., 2H), 7.49 (d, J=7.70 Hz, 2H), 7.53 – 7.62 (m, 3H), 7.69 (d, J=7.15 Hz, 2H), and 8.16 (d, J=7.70 Hz, 2H). 1. Preparation of l-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)-l,2,4- oxadiazol-3-yl)benzyl)azetidine-3-carboxylic acid

A mixture of tert-butyl l-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5- yl)-l,2,4-oxadiazol-3-yl)benzyl)azetidine-3-carboxylate (6.12 g, 11.6 mmol) and trifluoroacetic acid (50.1 mL, 651 mmol) was stirred at room temperature for 1.5 h. By HPLC, the deprotection appeared to be complete after 1 h. The TFA was removed under reduced pressure, and the oily residue was diluted with water (100 mL) and sonicated for 5 min. The resulting suspension was stirred for an additional 10 min until a consistent white suspension was observed. A IN aqueous solution of sodium hydroxide was added portion-wise until the pH was ~4.5 (42 mL of IN NaOH). Over time, the pH drifted back down to 3-4, and additional IN aqueous sodium hydroxide had to be added. The suspension was stirred overnight at room temperature. Several drops of IN aqueous sodium hydroxide were added to re-adjust the pH to 4.5, and after 60 min., the pH appeared to be stable. The solid was collected by vacuum filtration, washed with water several times, and dried under reduced pressure for 5 h. The solid was then suspended in methanol (110 mL) in a 150 mL round bottom flask and sonicated for 15 min. During the sonication, the solution became very thick. An additional 25 mL of methanol was added, and the suspension was stirred overnight. The product was collected by vacuum filtration, washed with methanol (-50 mL), and dried under reduced pressure. The solid was transferred to a 250 mL round bottom flask, re-suspended in methanol (115 mL), sonicated for 5 min., and stirred for 60 min. The solid was collected by vacuum filtration, washed with methanol (~50 mL), and dried over well under reduced pressure to give l-(4-(5-(3- phenyl-4-(trifluoromethyl)isoxazol-5-yl)-l,2,4-oxadiazol-3-yl)benzyl)azetidine-3- carboxylic acid (5.06 g, 10.7 mmol, 92 % yield) as a crystalline, white solid. The product had an HPLC ret. time = 2.79 min. – Column: CHROMOLITH® SpeedROD 4.6 x 50 mm (4 min.); Solvent A = 10% MeOH, 90% H2O, 0.1% TFA; Solvent B = 90% MeOH, 10% H2O, 0.1% TFA. LC/MS M+1 = 471.3. 1H NMR (500 MHz, DMSO-d6) δ ppm 3.20 – 3.46 (m, 5H), 3.66 (s, 2H), 7.53 (d, J=8.25 Hz, 2H), 7.60 – 7.70 (m, 5H), and 8.06 (d, J=7.70 Hz, 2H).

HPLC purity 100/99.8%, ret. time = 7.62 min. (A linear gradient using 5% acetonitrile, 95% water, and 0.05% TFA (Solvent A) and 95% acetonitrile, 5% water, and 0.05% TFA (Solvent B); t = 0 min., 10% B, t = l2 min., 100% B (15 min.) was employed on a SunFire C18 3.5u 4.6 x 150 mm column. Flow rate was 2 ml/min and UV detection was set to 220/254 nm.).

HPLC purity 100/99.9%, ret. time = 8.45 min. (A linear gradient using 5% acetonitrile, 95% water, and 0.05% TFA (Solvent A) and 95% acetonitrile, 5% water, and 0.05% TFA (Solvent B); t = 0 min., 10% B, t = l2 min., 100% B (15 min.) was employed on a XBridge Ph 3.5u 4.6 x 150 mm column. Flow rate was 2 ml/min and UV detection was set to 220/254 nm.).

CONSTRUCTION

Figure imgf000049_0002

Figure imgf000050_0001

Figure imgf000051_0001

Figure imgf000055_0001

Alt.1 -D- 1. Ethyl 3 -phenyl-4-(trifluoromethyl)isoxazole-5-carboxylate

Figure imgf000055_0002

Figure imgf000057_0001

Figure imgf000057_0002

Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000049_0001

ADDITIONAL INFORMATION

Sphingosine 1-phosphate (S1P) is the endogenous ligand for the sphingosine 1-phosphate receptors (S1P1–5) and evokes a variety of cellular responses through their stimulation. The interaction of S1P with the S1P receptors plays a fundamental physiological role in a number of processes including vascular development and stabilization, lymphocyte migration, and proliferation

REFERENCES

Watterson, S. H.; Guo, J.; Spergel, S. H.; Langevine, C. L.; Moquin, R. V.; Shen, D.
R.; Yarde, M.; Cvijic, M. E.; Banas, D.; Liu, R.; Suchard, S. J.; Gillooly, K.; Taylor,
T.; Rex-Rabe, S.; Shuster, D. J.; McIntyre, K. W.; Cornelius, G.; Darienzo, C.;
Marino, A.; Balimane, P.; Warrack, B.; Saltercid, L.; McKinnon, M.; Barrish, J. C.;
Carter, P. C.; Pitts, W. J.; Xie, J.; Dyckman, D. J. J. Med. Chem. 2016, 59, 2820.

Watterson, S.H.; Guo, J.; Spergel, S.H.; et al.
Potent and selective agonists of Sphingosine-1-Phosphate 1 (S1P1): The discovery and SAR of a novel isoxazole based series
241st Am Chem Soc (ACS) Natl Meet (March 27-30, Anaheim) 2011, Abst MEDI 96

/////Potent and Selective Isoxazole-Containing S1P1 Receptor Agonist, BMS 520, Sphingosine-1-Phosphate 1 (S1P1)

O=C(C1CN(CC2=CC=C(C3=NOC(C4=C(C(F)(F)F)C(C5=CC=CC=C5)=NO4)=N3)C=C2)C1)O


Filed under: Preclinical drugs Tagged: BMS 520, Potent and Selective Isoxazole-Containing S1P1 Receptor Agonist, Sphingosine-1-Phosphate 1 (S1P1)

7th Annual Clinical Trials Summit 2016, 24th May 2016, The Lalit Hotel, Mumbai, India

$
0
0

7th Annual Clinical Trials Summit 2016
                                                              

 

            
                    “A critical guide for
successfully conducting clinical trials”
24th May 2016, The Lalit Hotel, Mumbai, India
Greetings from Virtue
Insight,
 
I am happy to invite you
and your colleagues to be a sponsor/ delegate for our upcoming “7th
Annual Clinical Trials Summit 2016” The conference will Be held on 14th
May 2016, The Lalit Hotel, Mumbai, India. 
CONFIRMED SPEAKERS  FROM :- Takeda Pharmaceuticals (UK), Clinical Research &
Development, Cadila, Sanofi Aventis, Johnson & Johnson, GNH India, Clintech
India, Boehringer Ingelheim, Reliance Life Sciences, Abbott, Glenmark
Pharmaceuticals, Sanofi, Nishith Desai Associates, Novartis, Tata Consultancy
Services, Janssen India (Pharmaceutical companies of Johnson & Johnson),
SIRO Clinpharm, and few more.. 
CONFERENCE BOOKING DETAILS:-
·
Standard
Price:- 1 or 2 Delegates – (INR 7,000 + Tax (14.5%) per delegate)
·
Group
Discounts – 3 or 4 Delegates – (INR 6,500 + Tax (14.5%) per delegate) 
·
Group
Discounts – 5 and above Delegates – (INR 5,500 + Tax (14.5%) per delegate)
·
Conference
Sponsor & Exhibition Stall – Should you wish to Sponsor, or purchase a
Exhibition Stall (Booth) or a paid Speaker Slot, you can simply email your
interest and queries to TEL:
+ 91
9171350244

or
deepak@virtueinsight.co.in, deepakrajvirtueinsight@gmail.com  
Thank you for your time and
consideration. I look forward to hearing from you.  
PS: – Please refer your
friends or colleagues by forwarding this email to anyone you think may benefit
from it.  
 
Best Regards,
Deepak Raj
Delegate and Sponsorship Sales
Virtue Insight
Gsm
–   + 91
9171350244
Tel –      + 91 44 65515693
Skype –  edeepakraj143
clinical
trials”
“A critical guide for successfully conducting clinical trials”

Filed under: Uncategorized

CDRI 830

$
0
0

 

CDRI 830

CDRI S006-830

N-[2-[4-[(4-methoxyphenyl)-thiophen-2-ylmethyl]phenoxy]ethyl]-N-propan-2-ylpropan-2-amine

Molecular Formula: C26H33NO2S
Molecular Weight: 423.61072 g/mol

CHEMBL3417617.png

CDRI-830 of thiophene containing trisubstituted methane (TRSM) class was identified as an anti-tubercular lead with MIC value of 1.33 mg/L against Mycobacterium tuberculosis H37Rv strain, non-toxicity against Vero C-1008 cell line (selectivity index >10), ex vivo efficacy (in mouse and human macrophages) equivalent to first line TB drugs, lung CFU count (2.2×107) comparable to pyrazinamide (1.9×107) and ethambutol (1.27×107). CDRI-830 has exhibited potent bactericidal activity against single and multi-drug resistant clinical isolates of M. tuberculosis. Furthermore, CDRI-830 has demonstrated good pharmacokinetic properties with fast intestinal absorption, peak plasma concentration one hour post oral dose, optimum elimination half-life (9-13 h), plasma protein binding (~60%), favorable bioavailability (45-50%) and mean residence time (18-20 h).

CDRI S006-830 is a potent triethylamine containing thiophene antitubercular compound of the Central Drug Research Institute, India. The present study aimed to conduct comprehensive metabolic investigations of CDRI S006-830 to corroborate its preclinical investigations. Preliminary metabolic investigations were performed to assess the metabolic stability, enzyme kinetics, reaction phenotyping, and metabolite identification of CDRI S006-830 in rat, rabbit, dog, and human liver microsomes using liquid chromatography with mass spectrometry. The observed in vitro t1/2 and Clint values were 9.9 ± 1.29, 4.5 ± 0.52, 4.5 ± 0.86, 17 ± 5.21 min and 69.60 ± 8.37, 152.0 ± 17.26, 152.34 ± 27.63, 33.62 ± 21.04 μL/min/mg in rat, rabbit, dog and human liver microsomes respectively. These observations suggested that CDRI S006-830 rapidly metabolized in the presence of NADPH in liver microsomes of rat, rabbit and dog while moderately metabolized in human liver microsomes. It was observed that CDRI S006-830 exhibited monophasic Michaelis–Menten kinetics. The metabolism of CDRI S006-830 was primarily mediated by CYP3A4 and was deduced by CYP reaction phenotyping with known potent inhibitors. CYP3A4 involvement was also confirmed by cDNA-expressed recombinant human isozyme activity with different CYPs. Four major phase-I metabolites of S006-830, (M-1 to M-4) were detected in rat, rabbit, dog (except M4) and human liver microsomes……..http://onlinelibrary.wiley.com/doi/10.1002/dta.1802/abstract?systemMessage=Wiley+Online+Library+will+be+unavailable+on+Saturday+14th+May+11%3A00-14%3A00+BST+%2F+06%3A00-09%3A00+EDT+%2F+18%3A00-21%3A00+SGT+for+essential+maintenance.Apologies+for+the+inconvenience.

 

NMR

str1

 

13C NMR

str1

Abstract

The triarylmethane antituberculosis drug CDRI-830 is synthesized. The triarylmethane derivative 4 is prepared from ether 6 by a rearrangement process. The total synthesis of the drug CDRI-830 is achieved in a good overall yield of 35% from a simple thiophene derivative 8.

Synthetic Communications: An International Journal for Rapid Communication of Synthetic Organic Chemistry

Volume 44, Issue 23, 2014

Total Synthesis of an Experimental Antitubercular Drug CDRI-830

 

Total Synthesis of an Experimental Antitubercular DrugDOI:

10.1080/00397911.2014.942745

http://www.tandfonline.com/doi/abs/10.1080/00397911.2014.942745

 

REFERENCES

http://www.ingentaconnect.com/content/ben/cpa/2015/00000011/00000001/art00008?crawler=true

S006-830 against H37RV, single, multi-drug resistant M. tuberculosis; CFU in the lungs with S006-830, EMB, PZA (European Journal of Medicinal Chemistry 2015, 95, 357-368, J Antimicrob Chemother. 2012; 67(5):1188-97, Bioorg Med Chem Lett, 2008, 18, 289-292)

Panda, G., Parai, M.K., Das, S.K., Shagufta, Sinha,M., Chaturvedi, V., Srivastava, A.K., Manju,
Y.S., Gaikwad, A.N., and Sinha, S.: Effect of substituents on diarylmethanes for antitubercular activity.
European Journal of Medicinal Chemistry;2007,42, 410-419
1.  DiaryloxyMethanoPhenanthrenes: A New Class of Antituberculosis Agents, G. Panda,Shagufta, Jitendra Kumar Mishra, Vinita Chaturvedi, Anil K. Srivastava, Manju, RanjanaSrivastava and Brahm S. Srivastava,        1178DEL2004 Filing date 24/06/04
2. Thiophene containing Trisubstituted Methanes (TRSMs) as antitubercular agents, Gautam Panda, Maloy Kumar Parai, Priyanka Singh, Sudhir Sinha, Vinita Chaturvedi, Anil Gaikwad, PCT in process (685/DEL/2010) dt 20-2-2010

/////////

c1c(ccc(c1)OC)C(c2ccc(cc2)OCCN(C(C)C)C(C)C)c3sccc3


Filed under: Uncategorized Tagged: CDRI 830

Letermovir, AIC 246

$
0
0

Letermovir skeletal.svg

Letermovir, MK 8828, AIC 246

2-[(4S)-8-fluoro-2-[4-(3-methoxyphenyl)piperazin-1-yl]-3-[2-methoxy-5-(trifluoromethyl)phenyl]-4H-quinazolin-4-yl]acetic acid

 CAS 917389-32-3

Letermovir; UNII-1H09Y5WO1F; AIC-246; 2-((4S)-8-Fluoro-2-(4-(3-methoxyphenyl)piperazin-1-yl)-3-(2-methoxy-5-(trifluoromethyl)phenyl)-4H-quinazolin-4-yl)acetic acid; 2-[(4S)-8-fluoro-2-[4-(3-methoxyphenyl)piperazin-1-yl]-3-[2-methoxy-5-(trifluoromethyl)phenyl]-4H-quinazolin-4-yl]acetic acid; Letermovir [INN]

Molecular Formula: C29H28F4N4O4
Molecular Weight: 572.550633 g/mol

Letermovir (INN) is an antiviral drug that is being developed for the treatment of cytomegalovirus (CVM) infections. It has been tested in CMV infected patients with allogeneic stem cell transplants and may also be useful for other patients with a compromised immune system such as those with organ transplants or HIV infections.[1]

The drug has been granted fast track status by the US Food and Drug Administration (FDA) and orphan drug status by the European Medicines Agency.[1]

The drug candidate is under development by Merck & Co., Inc as investigative compound MK-8828.[2]

AIC246, also known as letermovir, is a novel anti-CMV compound with IC50 value of 5.1 ± 1.2 nM. It targets the pUL56 (amino acid 230-370) subunit of the viral terminase complex [1].
The subunit pUL56 is a component of the terminase complex which is responsible for packaging unit length DNA into assembling virions.
AIC246 has a novel mode of action targets the enzyme UL56 terminase and keep active to other drug-resistant virus. The anti-HCMV activity of AIC246 was evaluated in vitro by using different HCMV laboratory strains, GCV-resistant viruses. The result showed that the inhibitory potentcy of AIC246 surpasses the current gold standard GCV by more than 400-fold with respect to EC50s (mean, ∼4.5 nM versus ∼2 μM) and by more than 2,000-fold with respect to EC90 values (mean, ∼6.1 nM versus ∼14.5 μM).  In the CPE-RA strains, the EC50 values of AIC 246 ranged from 1.8 nM to 6.1 nM [2].
In mouse model with HCMV subcutaneous xenograft, oral administration of AIC246 caused significant a dose-dependent reduction of the HCMV titer. 30 mg/kg/d AIC246 for 9 days induced PFU reduction with maximum efficiency, compared with the gold standard GCV at the ED50 and ED90 level [2].
References:
[1].Verghese PS, Schleiss MR. Letermovir Treatment of Human Cytomegalovirus Infection Anti-infective Agent. Drugs Future. 2013, 38(5):291-298.
[2]. Lischka P1, Hewlett G, Wunberg T, et al.In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.Antimicrob Agents Chemother. 2010, 54(3):1290-1297.

NMR

STR1

STR1

Human cytomegalovirus (HCMV) remains the leading viral cause of birth defects and life-threatening disease in transplant recipients. All approved antiviral drugs target the viral DNA polymerase and are associated with severe toxicity issues and the emergence of drug resistance. Attempts to discover improved anti-HCMV drugs led to the identification of the small-molecular-weight compound AIC246 (Letermovir). AIC246 exhibits outstanding anti-HCMV activity in vitro and in vivo and currently is undergoing a clinical phase IIb trial. The initial mode-of-action studies suggested that the drug acts late in the HCMV replication cycle via a mechanism distinct from that of polymerase inhibitors. Here, we extend our mode-of-action analyses and report that AIC246 blocks viral replication without inhibiting the synthesis of progeny HCMV DNA or viral proteins. The genotyping of mutant viruses that escaped AIC246 inhibition uncovered distinct point mutations in the UL56 subunit of the viral terminase complex. Marker transfer analyses confirmed that these mutations were sufficient to mediate AIC246 resistance. The mapping of drug resistance to open reading frame UL56 suggests that viral DNA processing and/or packaging is targeted by AIC246. In line with this, we demonstrate that AIC246 affects the formation of proper unit-length genomes from viral DNA concatemers and interferes with virion maturation. However, since AIC246-resistant viruses do not exhibit cross-resistance to previously published terminase inhibitors, our data suggest that AIC246 interferes with HCMV DNA cleavage/packaging via a molecular mechanism that is distinct from that of other compound classes known to target the viral terminase.

PATENT

WO 2006133822


Scheme 2:

Chromatography
on a chiral phase

Scheme 4:

Scheme 5:

Synthesis of {8-fluoro-2- [4- (3-methoxyphenyl) piperazin-l -yl] -3- [2-methoxy-5- (trifluoromethyl) phenyl] -3,4-dihydroquinazolin-4-yl }acetic acid

xample 1

N- (2-bromo-6-fluoφhenyl) -N ‘- [2-methoxy-5- (trifluoromethyl) phenyl] urea

2-methoxy-5-trifluoromethylphenyl isocyanate (274.3 g) are dissolved in acetonitrile (1 L), then 2-bromo-6-fluoroaniline (200 g) was added with acetonitrile (50 mL) flushed. The resulting clear solution is at 38 h reflux (ca. 85 0 stirred C), then under vacuum at 40 0 concentrated C a dogged mush. This is filtered off, with acetonitrile (260 mL, to 0-5 0 C cooled) washed and incubated overnight at 45 0 dried C in the VDO using entraining nitrogen. Thus, a total of 424.3 g of N- (2-bromo-6-fluorophenyl) -N ‘- get [2-methoxy-5- (trifluoromethyl) phenylJ-urea as a solid, corresponding to 99.2% of theory.

1 H NMR (300 MHz, d 6 -DMSO): δ = 8.93 (s, IH), 8.84 (s, IH), 8.52 (d, V = 2.3, 2H), 7, 55 (d, 2 = Vr = 7.7, IH), 7.38 to 7.26 (m, 3H), 7.22 (d, 2 J = 8.5, IH), 4.00 (s, 3H) ppm;

– – MS (API-ES-pos.): M / z = 409 [(M + H) + , 100%];

HPLC (Method 1): R τ = 22.4 and 30.6 min.

example 2

N- (2-bromo-6-fluorophenyl) -N ‘- [2-methoxy-5- (trifluoromethyl) phenyl] urea (Alterhativsynthese)

2-methoxy-5-trifluoromethylphenyl isocyanate (1.19 kg) are at about 35 0 dissolved melted and C in acetonitrile (4.2 L), then 2-bromo-6-fluoroaniline (870 g) was added and with acetonitrile ( 380 mL) rinsed. The resulting clear solution is at 74-88 45 h 0 stirred C, then under vacuum (200 mbar) at 50 0 C to a dogged mush concentrated (amount of distillate 4.4 L). This is at room temperature with diisopropylether (1.5 L), washed aspirated, with diisopropylether (1.15 L) washed and at 45 0 C in the VDO using entraining nitrogen to constant weight (24 h) dried. Thus, a total of 1, 63 kg Η- (2-bromo-6-fluoro-phenyl) -W- – obtained [2-methoxy-5 (trifluoromethyl) phenyl] urea as a solid, corresponding to 87.5% of theory.

HPLC (Method 1): R τ = 22.6 and 30.8 min.

example 3

{8-Fluor-3-[2-methoxy-5-(trifluormethyl)phenyl]-2-oxo-l,2,3,4-tetrahydrochinazolin-4-yl}essigsäuremethylester

N- (2-bromo-6-fluorophenyl) -N- [2-methoxy-5- (trifluoromethyl) phenyl] urea (300 g) under a nitrogen atmosphere in isobutyronitrile (1.2 L) was suspended, then triethylamine

(21O mL), bis (acetonitrile) dichloropalladium (7.5 g), tris- (o-tolyl) phosphine (18.0 g) and

Methyl acrylate (210 mL) were added in this order. The resulting suspension is for 16 hours at reflux (ca. 102 0 stirred C) and then cooled to room temperature. Water (1.2 L) is added and the mixture 1 at room temperature stirred, then aspirated and washed with water / methanol h: washed and acetonitrile (10O mL) (1 1 30O mL). The residue is treated overnight at 45 0 dried C in the VDO using entraining nitrogen. Thus, a total of 208 g as a solid, corresponding to 68.5% of theory.

1 H NMR (300 MHz, d 6 -DMSO): δ = 9.73 (s, IH), 7.72 (d, 2 J = 7.3, IH), 7.71 (s, IH), 7 , 33 (d, 2 J = 9.3, IH), 7.15 (dd, 2 J = 9.6, 2 J = 8.6, IH), 7.01 (d, 2 J = 7.3 , IH), 6.99 to 6.94 (m, IH), 5.16 (t, 2 , J = 5.9, IH), 3.84 (s, 3H), 3.41 (s, 3H) , 2.81 (dd, 2 J = 15.4, 2 J = 5.8, IH), 2.62 (dd, 2 J = 15.4, 2 J = 6.3, IH) ppm;

MS (API-ES-pos.): M / z = 413 [(M + H) + , 100%], 825 [(2M + H) + , 14%];

HPLC (Method 1): R τ = 19.3 min; Pd (ICP): 16,000 ppm.

example 4

{8-Fluor-3-[2-methoxy-5-(trifluormethyl)phenyl]-2-oxo-l,2,3,4-tetrahydrochinazolin-4-yl}essigsäuremethylester (Alternative synthesis)

N- (2-bromo-6-fluorophenyl) -N ‘- [2-methoxy-5- (trifluoromethyl) phenyl] urea (2.5 kg) is suspended under a nitrogen atmosphere in isobutyronitrile (9 L), then triethylamine (1.31 kg), bis (acetonitrile) dichloropalladium (64.9 g), tris (o-tolyl) phosphine (149 g) and methyl acrylate (1.59 kg) were added in this order. The resulting suspension is 22 hours at 90-100 0 stirred C, then cooled to room temperature. Water (9 L) is added and stirred, then aspirated and washed with water / methanol (1: 1, 2.5 L) at room temperature, the mixture for 1 hour and acetonitrile (850 mL). The residue is treated overnight at 45 0 dried C in the VDO using entraining nitrogen to constant weight (21 h). Thus, a total of 1.90 kg as a solid, corresponding to 74.9% of theory.

HPLC (Method 1): R τ = 19.4 min.

example 5

{2-Chlor-8-fluor-3-[2-methoxy-5-(trifluormethyl)phenyl]-3,4-dihydrochinazolin-4-yl}essigsäure-methylester / chlorination

A solution of 2.84 kg {8-fluoro-3- [2-methoxy-5- (trifluoromethyl) phenyl] -2-oxo-l, 2,3,4-tetrahydroquinazolin-4-yl} acetic acid methyl ester in 14.8 l of chlorobenzene is heated to reflux and the solvent is distilled off until water no longer separates. It is to 12O 0 cooled C. Within 10 min phosphorus oxychloride are metered in 3.17 kg, and then is added within a further 10 min 2.10 kg DBU. It is heated to reflux for 9 hours.

For working up the mixture is cooled to 40 0 C., stirred overnight and dosed the reactor contents to 11.4 L of water, previously estimated at 40 0 was tempered C. For dosing an internal temperature of 40-45 to 0 C, are satisfied. The mixture is allowed to cool to room temperature, 11.4 L of dichloromethane, filtered through a Seitz filter plate and the phases are separated. The organic phase is washed with 11.4 L of water, 11.4 L of an aqueous saturated sodium bicarbonate solution and again with 11.4 L of water. The organic phase is concentrated on a rotary evaporator in vacuo and the remaining residue (2.90 kg) is used without further treatment in the next step.

1 H NMR (300 MHz, d 6 -DMSO): δ = 7.93 to 7.82 (m, 2H), 7.38 (d, 2 J = 8.9, IH), 7.17 (m, 2H), 6.97 to 6.91 (m, IH), 5.45 and 5.29 (m and t, 2 , J = 5.4, IH), 3.91 and 3.84 (2s, 3H) , 3.48 (s, 3H), 3.0 to 2.6 (m, 2H) ppm;

MS (CI, NH 3 ): m / z = 431 [(M + H) + , 100%];

HPLC (Method 1): R τ = 23.5 min; typical Pd value (ICP): 170 ppm.

example 6

{8-Fluor-2-[4-(3-methoxyphenyl)piperazin-l-yl]-3-[2-methoxy-5-(trifluormethyl)phenyl]-3,4-dihydrochinazolin-4-yl}essigsäuremethylester / Amination – –

(52.5 g) is dissolved in 1,4-dioxane (10O mL), then (25.8 g) and DBU (20.4 g) was added at room temperature 3-methoxyphenylpiperazine, whereupon the temperature rises. The mixture is stirred at reflux for 22 h, then cooled to room temperature, with ethyl acetate (500 mL) and water (200 mL) and the phases separated. The organic phase (200 mL) washed with 0.2N hydrochloric acid (three times 100 mL) and water, dried over sodium sulfate and evaporated. Thus, a total of 62.5 g obtained as a solidified foam, which is reacted as the crude product without further purification.

HPLC (Method 1): R τ = 16.6 min.

example 7

{8-Fluor-2-[4-(3-methoxyphenyl)piperazin-l-yl]-3-[2-methoxy-5-(trifluormethyl)phenyl]-3,4-dihydrochinazolin-4-yl}essigsäuremethylester / Pot chlorination + amination

(50.0 g) is introduced in chlorobenzene (300 mL), then chlorobenzene is partially distilled (5O mL). The mixture is heated to 120 0 cooled C., DBU (36.9 g) is added, then at 120-128 is 0 C phosphorous oxychloride (33.4 mL) over 10 min. metered. The mixture (approximately 130 at reflux for 9 hours 0 C) stirred. Subsequently, at 40 0cooled C, slowly at 40-45 0 C with water (200 mL), cooled to room temperature and diluted with dichloromethane (200 mL), stirred and then the phases separated. The organic phase is washed with water (200 mL), saturated aqueous sodium bicarbonate solution (200 mL) and again water (200 mL), dried over sodium sulfate, concentrated by rotary evaporation and then under high vacuum at 50 0 dried C. The residue (48.1 g) is dissolved in chlorobenzene (20 mL), then with 1,4-dioxane (80 mL) at room temperature and 3-methoxyphenylpiperazine (23.6 g) and DBU (18.7 g) was added, whereupon the temperature rises. The mixture is stirred at reflux for 22 h, then cooled to room temperature, with ethyl acetate (500 mL) and water (200 mL) and the phases separated. The organic phase (200 mL) washed with 0.2N hydrochloric acid (three times 100 mL) and water, dried over sodium sulfate and evaporated. Thus, a total of 55.6 g obtained as a solidified foam, which is reacted as the crude product without further purification.

HPLC (Method 1): R τ = 16.2 min.

example 8

(^)-{8-Fluor-2-[4-(3-methoxyphenyl)piperazin-l-yl]-3-(2-methoxy-5-trifluormethylphenyl)-3,4-dihydrochinazolin-4-yl} acetate / saponification racemate

(64 g) is dissolved in 1,4-dioxane (45O mL) and IN sodium hydroxide solution (325 mL) and stirred for 2 h at room temperature, then dried in vacuo at 30 0 , a part of the solvent C is distilled off (400 mL). Toluene is added (300 mL) and the phases separated. The aqueous phase is washed with toluene (15O mL twice), then the combined organic phases again with IN sodium hydroxide solution (50 mL) are extracted. The pH of the combined aqueous phases with 2N hydrochloric acid (about 150 mL) to 7.5, then MIBK (15O mL) is added. The phases are separated, the aqueous phase extracted again with MIBK (15O mL), then dried the combined MIBK phases over sodium sulfate and at 45 0 concentrated C. Thus, a total of 64 g as an amorphous solid in quantitative yield.

HPLC (Method 1): R τ = 14.9 min.

Scheme 6:

Separation of enantiomers of {8-fluoro-2- [4- (3-methoxyphenyl) piperazin-l -yl] -3- [2-methoxy-5- (tri-fluoromethyl) phenyl] -3,4-dihydroquinazolin-4-yl } acetate

x (2S, 3S) -2,3-bis [(4-methylbenzoyl) – oxyjbemsteinsäure
x EtOAc

example 9

(2S, 3 £) -2,3-bis [(4-methylbenzoyl) oxy] succinic acid (1: 1 salt) / crystallization

(62.5 g, crude product) is dissolved and filtered in ethyl acetate (495 mL). To the filtrate is (35 25 ‘,) added 2,3-bis [(4-methylbenzoyl) oxy] succinic acid (42.0 g), the mixture for 30 minutes. stirred at room temperature, then with (35 25 “) -2,3-bis [(4-methylbenzoyl) oxy] -succinic acid – (l: l salt) (165 mg) was inoculated and stirred for 3 days at room temperature, then to 0-3 0 cooled C and stirred for a further 3 h, the suspension is suction filtered and washed with cold ethyl acetate (0-10. 0 C, 35 mL ) washed. the crystals are at 40 h 18 0 C in the VDO using entraining nitrogen dried. Thus 37.1 g of the salt are obtained as a solid, corresponding to 30.4% of theory over three stages (chlorination, amination and crystallization) on the racemate, or 60.8% based on the resulting S enantiomer.

– – 1 H NMR (300 MHz, d 6 -DMSO): δ = 7.90 (d, 2 J = 7.8, 4H), 7.56 (d, 2 J = 8.3, IH), 7 , 40 (d, 2 J = 7.8, 4H), 7.28 to 7.05 (m, 4H), 6.91 to 6.86 (m, 2H), 6.45 (d, 2 J = 8.3, IH), 6.39 to 6.36 (m, 2H), 5.82 (s, 2H), 4.94 (m, IH), 4.03 (q, 2 J = 7.1 , 2H), 3.83 (brs, 3H), 3.69 (s, 3H), 3.64 (s, 3H), 3.47 to 3.36 (m, 8H and water, 2H), 2, 98 to 2.81 (m, 5H), 2.58 to 2.52 (m, IH), 2.41 (s, 6H), 1.99 (s, 3H), 1.18 (t, 2 J = 7.2, 3H) ppm;

HPLC (Method 1): R τ = 16.6 and 18.5 min.

example 10

(25,3iS) -2,3-bis [(4-methylbenzoyl) oxy] succinic acid (1: 1 salt) / recrystallization

(2S, 3S) -2,3-bis [(4-methy lbenzoyl) oxy] succinic acid – { (l: l salt) (36.8 g) is suspended in ethyl acetate (37o mL) and (77 by heating to reflux 0 C) dissolved. The mixture is slowly cooled to room temperature. Here there is a spontaneous crystallization. The suspension is stirred at RT for 16 h, then 0-5 0 cooled C and stirred for another 3 h. The suspension is suction filtered and washed with cold ethyl acetate (0-10 0 C, twice 15 ml). The crystals are at 45 h 18 0 C in the VDO using entraining nitrogen dried. Thus 33.6 g of the salt are obtained as a solid, corresponding to 91.3% of theory.

HPLC (Method 1): R τ = 16.9 and 18.8 min .;

HPLC (Method 3): 99.9% ee

example 11

(5)-{8-Fluor-2-[4-(3-methoxyphenyl)piperazin-l-yl]-3-(2-methoxy-5-trifluormethylphenyl)-3,4-dihydrochinazolin-4-yl}essigsäure

(2IS I , 3S) -2,3-bis [(4-methylbenzoyl) oxy] succinic acid (l: l salt) (10.1 g, containing 14 ppm of Pd) are suspended in ethyl acetate (100 mL) and shaken with saturated aqueous sodium bicarbonate solution (10O mL) shaken until both phases are clear. The phases are separated, the organic phase is evaporated. The residue is dissolved in 1,4-dioxane (100 mL) and IN sodium hydroxide solution (31.2 mL) and stirred for 3 h at room temperature. Subsequently, the pH is adjusted with IN hydrochloric acid (about 17 mL) is set to 7.5, MIBK (8O mL) was added, then the pH is adjusted with IN hydrochloric acid (about 2 mL) adjusted to 7.0. The phases are separated, the organic phase dried over sodium sulfate and concentrated. The residue is dissolved in ethanol and concentrated (40 mL), then again in ethanol (40 mL) and concentrated under high vacuum at 50 0 C dried. The solidified foam is at 45 h 18 0 C in the VDO using entraining nitrogen dried. Thus, a total of 5.05 g as an amorphous solid, corresponding to 85.0% of theory.

1 H NMR (300 MHz, d 6 -DMSO): δ = 7.53 (d, 2 J = 8.4, IH), 7.41 (brs, IH), 7.22 (d, 2 J = 8 , 5, IH), 7.09 to 7.01 (m, 2H), 6.86 (m, 2H), 6.45 (dd, V = 8.2, 3 J = 1.8, IH) 6.39 to 6.34 (m, 2H), 4.87 (t, 2 J = 7.3, IH), 3.79 (brs, 3H), 3.68 (s, 3H), 3.50 -3.38 (m, 4H), 2.96 to 2.75 (m, 5H), 2.45 to 2.40 (m, IH) ppm;

MS (API-ES-neg.): M / z = 571 [(MH), 100%];

HPLC (Method 1): R τ = 15.1 min;

HPLC (Method 2): 99.8% ee; Pd (ICP): <1 ppm.

example 12

(2 / ?, 3Λ) -2,3-bis [(4-methylbenzoyl) oxy] succinic acid (1: 1 salt) / crystallization R-isomer from the mother liquor

The mother liquor from a crystallization of (2IS ‘, 3S) -2,3-bis [(4-methylbenzoyl) oxy] -succinic acid – {8-fluoro-2- [4- (3-methoxyphenyl) piperazin-l -yl] -3- [2-methoxy-5- (trifluoromethyl) phenyl] -3,4-dihydroquinazolin-4-yl} acetic acid methyl ester (l: l-salt) in 279 g scale is washed with saturated aqueous sodium bicarbonate solution (1.5 L ) shaken, the phases are separated and the organic phase is shaken with semi-saturated aqueous sodium bicarbonate solution (1.5 L). The phases are separated, the organic phase dried over sodium sulfate and evaporated. The residue (188.4 g) is dissolved in ethyl acetate (1.57 L), then (2R, 3R) -2,3-bis [(4-methylbenzoyl) oxy] succinic acid (121.7 g) was added and the mixture 10 min. stirred at room temperature. Is then treated with (2R, 3R) -2,3-bis [(4-methyl-benzoyl) oxy] succinic acid – (l: l salt) (0.38 g) was inoculated and stirred for 18 h at room temperature, then to 0-3 0 cooled C and stirred for another 3 h. The suspension is suction filtered and washed with cold ethyl acetate (0-10 0 C, 50O ml). The crystals are at 40 h 18 0 C in the VDO using entraining nitrogen dried. So a total of 160 g of the salt are obtained as a solid.

HPLC (Method 1): R τ = 16.6 and 18.5 min .;

HPLC (Method 3): -99.0% ee

example 13

(i?)-{8-Fluor-2-[4-(3-methoxyphenyl)piperazin-l-yl]-3-(2-methoxy-5-trifluormethylphenyl)-3,4-dihydrochinazolin-4-yl} acetate / production R-isomer

(2Λ, 3 /?) – 2,3-bis [(4-methylbenzoyl) oxy] succinic acid – {8-fluoro-2- [4- (3-methoxy-phenyl) pipera-tine 1-yl] -3- [ 2-methoxy-5- (trifluormethy l) pheny l] -3, 4-dihydroquinazolin-4-y 1} -acetic acid methyl ester (1: 1 salt) (170 g) are suspended in ethyl acetate (85O mL) and as long as with saturated aqueous sodium bicarbonate (850 mL) shaken until both phases are clear (about 5 min.). The phases are separated, the solvent of the organic phase under normal pressure with 1, 4-dioxane to a final temperature of 99 0 exchanged C (portions distilled total 2.55 L solvent, and 2.55 L of 1,4-dioxane used). The mixture is cooled to room temperature and 18 at room temperature IN sodium hydroxide solution (525 mL) stirred. Subsequently, the pH value with concentrated hydrochloric acid (about 35 mL) is set to 7.5, MIBK (85O mL) was added, then the pH with concentrated hydrochloric acid (ca. 1O mL) adjusted to 7.0. The phases are separated, the organic phase dried over sodium sulfate and concentrated. The residue is dissolved in ethanol and concentrated (350 mL), then again in ethanol (350 mL) at 50 and 0 concentrated C. Thus, a total of 91.6 g as an amorphous solid, corresponding to 91.6% of theory.

HPLC (method 1): R 7 = 14.8 min.

– – Example 14

{8-Fluor-2-[4-(3-methoxyphenyl)piperazin-l-yl]-3-(2-methoxy-5-trifluormethylphenyl)-3,4-dihydrochinazolin-4-yl} acetate / racemization R-enantiomer

acetic acid (50 g) is dissolved in acetonitrile (500 mL) and treated with sodium methoxide (30% in methanol, 32.4 mL) and then stirred at reflux for 60 h. After cooling to room temperature the mixture is concentrated in vacuo to half, then with hydrochloric acid (20% strength, ca. 20 ml) adjusted to pH 7.5, MIBK (200 mL) was added and hydrochloric acid (20%) on pH 7 adjusted. The phases are separated, the organic phase dried over sodium sulfate and evaporated to the hard foam. The residue is dissolved in ethanol and concentrated (15O mL), then again in ethanol (15O mL) and concentrated. Thus, 54.2 g as an amorphous solid in quantitative yield.

HPLC (Method 1): R τ = 14.9 min .;

HPLC (method 4): 80.8 wt.%.

example 15

{8-Fluor-2-[4-(3-methoxyphenyl)piperazin-l-yl]-3-[2-methoxy-5-(trifluormethyl)phenyl]-3,4-dihydrochinazolin-4-yl}essigsäuremethylester / Esterification racemate

acetic acid (54 g) (540 g) was dissolved in methanol, then concentrated sulfuric acid (7.85 mL) is added. The mixture is stirred at reflux for 26 h, then cooled and concentrated in vacuo to about one third of the original volume. Water (15O mL) and dichloromethane (15O mL) are added, then the phases are separated. The organic phase is washed with saturated sodium bicarbonate solution (two times 140 mL), dried over sodium sulfate and concentrated to a foamy residue. This is twice in succession in ethanol (150 mL) and concentrated, dried in vacuo using entraining nitrogen then 18 h. Thus, a total of 41.6 g as an amorphous solid, corresponding to 75.2% of theory.

HPLC (Method 1): R τ = 16.8 min .;

HPLC (method 4): 85.3 wt.%;

HPLC (Method 3): -8.5% ee

example 16

(25 1 , 3S) -2,3-bis [(4-methylbenzoyl) oxy] succinic acid – { (1: 1 salt) / crystallization of esterified racemate

(41.0 g) is suspended in ethyl acetate (287 mL), then (2S, 3IS) -2,3-bis [(4-methylbenzoyl) oxy] succinic acid (27.5 g) was added. The mixture is 30 minutes. stirred at room temperature, then with (2 <S ‘, 3S) -2,3-bis [(4-methylbenzoyl) oxy] succinic acid – (1: 1 salt) (0.08 g) was inoculated. The suspension is stirred at RT for 16 h, then 0-5 0 cooled C and stirred for another 3 h, then filtered off with suction and washed with cold ethyl acetate (0-10 0 C, four times 16 ml). The crystals are at 45 h 18 0 C in the VDO using entraining nitrogen dried. So a total of 25.4 g of the salt are obtained as a solid, corresponding to 37.4% of theory.

HPLC (Method 1): R τ = 16.9 and 18.8 min .;

HPLC (method 4): 99.5 wt.%;

HPLC (Method 3): 99.3% ee

example 17

(iS)-{8-Fluor-2-[4-(3-methoxyphenyl)piperazin-l-yl]-3-(2-methoxy-5-trifluormethylphenyl)-3,4-dihydrochinazolin-4-yl} acetate / saponification crystals

(25,3S) -2,3-bis [(4-methylbenzoyl) oxy] succinic acid – (l rl salt) (25.1 g) is suspended in ethyl acetate (25O mL) and shaken with saturated aqueous sodium bicarbonate solution (250 mL) shaken until both phases are clear. The phases are separated, the organic phase is evaporated. Dissolve the residue in 1, 4-dioxane (25O mL) and IN sodium hydroxide solution (77.4 mL) and stirred for 18 h at room temperature. Subsequently, the pH is adjusted with IN hydrochloric acid (about 50 mL) is set to 7.5, was added MIBK (240 mL), then the pH is adjusted with IN hydrochloric acid (about 15 mL) adjusted to 7.0. The phases are separated, the organic phase dried over sodium sulfate and concentrated. The residue is dissolved in ethanol and concentrated (90 mL), then again in ethanol (90 mL) and concentrated. The solidified foam is at 45 h 180 C in the VDO using entraining nitrogen dried. Thus, a total of 12 g as an amorphous solid, corresponding to 81.2% of the theory.

HPLC (Method 1): R τ = 15.1 min;

HPLC (Method 2): 97.5% ee; Pd (ICP): <20 ppm.

Alternative method for the racemization:

example 18

(i)-{8-Fluor-2-[4-(3-methoxyphenyl)piperazin-l-yl]-3-(2-methoxy-5-trifluormethylphenyl)-3,4-dihydrochinazolin-4-yl} acetic acid / saponification enriched R isomer from the mother liquor after crystallization

The mother liquor from a crystallization of (2 J S ‘, 35) -2,3-bis [(4-methylbenzoyl) oxy] -succinic acid – (l: l-salt) in 207 g scale is shaken with saturated aqueous sodium bicarbonate (500 mL), the phases are separated and the organic phase is shaken with semi-saturated aqueous sodium bicarbonate solution (500 mL). The phases are separated, the organic phase dried over sodium sulfate and evaporated. The residue is dissolved in ethanol (500 mL) and rotary evaporated to a hard foam. This is in 1,4-dioxane (1.6 L) and IN sodium hydroxide solution (1.04 L) and stirred at room temperature for 18 h, then toluene is added (1.5 L) and the phases separated. The aqueous phase is adjusted with hydrochloric acid (20% strength, ca. 155 ml) of pH 14 to pH 8, then is added MIBK (1.25 L) and hydrochloric acid (20% strength, ca. 25 mL) to pH 7 readjusted. The phases are separated, the organic phase dried over sodium sulfate and evaporated to the hard foam. This is at 45 h 18 0 C in the VDO using entraining nitrogen dried. Thus, a total of 150 g obtained as (R / S) mixture as an amorphous solid.

HPLC (Method 2): 14.6% ee

– – Example 19

(i)-{8-Fluor-2-[4-(3-methoxyphenyl)piperazin-l-yl]-3-(2-methoxy-5-trifluormethylphenyl)-3,4-dihydrochinazolin-4-yl} acetate / racemization

(150 g, R / S mixture with -14.6% ee) is dissolved in acetonitrile (1.5 L) and treated with sodium methoxide (30% in methanol, 97.2 mL) was added, then stirred at reflux for 77 h , After cooling to room temperature the mixture is concentrated in vacuo to half, then with hydrochloric acid (20% strength, ca. 80 mL) made of pH 13 to pH 7.5, was added MIBK (0.6 L) and treated with hydrochloric acid ( 20% strength, ca. 3 mL) adjusted to pH. 7 The phases are separated, the organic phase dried over sodium sulfate and evaporated to the hard foam. The residue is dissolved in ethanol and concentrated (500 mL), then again in ethanol (500 mL) and concentrated, then 18 h at 450 dried C in the VDO using entraining nitrogen. Thus, a total of 148 g as an amorphous solid, corresponding to 98.7% of theory.

HPLC (Method 2): 1.5% ee

example 20

{8-Fluor-2-[4-(3-methoxyphenyl)piperazin-l-yl]-3-[2-methoxy-5-(trifluormethyl)phenyl]-3,4-dihydrochinazolin-4-yl}essigsäuremethylester (Esterification)

(±) – {8-fluoro-2- [4- (3-methoxyphenyl l) piperazin-1 -yl] -3- (2-methoxy-5-trifluormethy lphenyl) -3, 4-dihydroquinazolin-4-yl} acetic acid (148 g) (1480 g) was dissolved in methanol, then concentrated sulfuric acid (21.5 mL) is added. The mixture is stirred at reflux for 6 h, then cooled and concentrated in vacuo to about one third of the original volume. Water (400 mL) and dichloromethane (400 mL) are added, then the phases are separated. The organic phase (diluted twice 375 mL, 300 mL water) with saturated sodium bicarbonate solution, dried over sodium sulfate and concentrated to a foamy residue. This is twice in succession in ethanol (each 400 mL) and concentrated, dried in vacuo using entraining nitrogen then 18 h. Thus, a total of 124 g as an amorphous solid, corresponding to 81.9% of theory.

HPLC (Method 1): R τ = 16.9 min .;

example 21

(25.35) -2,3-bis [(4-methylbenzoyl) oxy] succinic acid – (1: 1 salt) / crystallization of esterified racemate

(2S, 3S) -2,3-bis [(4-methylbenzoyl) oxy] succinic acid – (1: 1 salt) (123 g, 14.4% ee) is suspended in ethyl acetate (861 mL) and filtered, then (2IS ‘, 3IS) -2,3-bis [(4-methylbenzoyl) oxy ] succinic acid (82.5 g). The mixture 30 min. stirred at room temperature, then with (2 £, 3 <S) -2,3-bis [(4-methylbenzoyl) oxy] succinic acid – (1: 1 salt) (0.24 g) was inoculated. The suspension is stirred for 4 days at RT, then concentrated to approximately 600 mL and again with (25 ‘, 3 1 -2,3-bis [(4-methylbenzoyl) oxy] succinic acid S) – (l: l salt) (0.24 g) was inoculated. The suspension is stirred for 1 week at RT, to 0-5 0 cooled C and further stirred for 3 hours, then filtered off with suction and washed with cold ethyl acetate (0-10 0 C, 4 x 40 ml). The crystals are at 45 h 18 0 C in the VDO using entraining nitrogen dried. So a total of 1 1.8 g of salt are obtained as a solid, corresponding to 5.8% of theory.

Scheme 7:

example 22

N- (2-Fluoφhenyl) -N ‘- [2-methoxy-5- (trifluoromethyl) phenyl] urea

2-methoxy-5-trifluoromethylphenyl isocyanate (1057.8 g) is dissolved in acetonitrile (4240 mL), then 2-fluoro aniline (540.8 g) was added with acetonitrile (50 mL) flushed.The resulting clear solution is stirred for 4 h at reflux (about 82 ° C), then seeded at about 78 ° C and about 15 min. touched. The suspension is on 0 0 cooled C, aspirated and the product with acetonitrile (950 mL, to 0-5 0 cooled C) washed. The product is dried overnight at 45 ° C in a vacuum drying oven using entraining nitrogen. Thus, a total of 1380.8 g of N- (2-fluorophenyl) -N ‘- [2-methoxy-5- (trifluoromethyl) phenyl] -harnstqff obtained as a solid, corresponding to 86.4% of theory.

1 H NMR (500 MHz, d 6 -DMSO): δ = 9.36 (s, IH), 9.04 (s, IH), 8.55 (d, 1.7 Hz, IH), 8.17 ( t, 8.2 Hz, IH), 7.33 (d, 8.5 Hz, IH), 7.20 to 7.26 (m, 2H), 7.14 (t, 7.6 Hz, IH), 7, 02 (m, IH), 3.97 (s, 3H) ppm;

MS (API-ES-pos.): M / z = 329 [(M + H) + , 100%];

HPLC: R τ = 48.7 min.

Instrument: HP 1100 Multiple Wavelength detection; Column: Phenomenex-Prodigy ODS (3) 100A, 150 mm x 3 mm, 3 microns; Eluent A: (1.36 g KH 2 PO 4 +0.7 mL H 3PO 4 ) / L water, eluent B:

acetonitrile; Gradient: 0 min 20% B, 40 min 45% B, 50 min 80% B, 65 min 80% B; Flow: 0.5 mL / min; Temp .: 55 0 C; UV detection: 210 nm.

example 23

Methyl (2E) -3- {3-fluoro-2 – [({[2-methoxy-5 – (trifluormethy l) pheny 1] amino} carbonylation l) amino] pheny 1} acrylate

N- (2-fluorophenyl) -N ‘- [2-methoxy-5- (trifluoromethyl) phenyl] urea (0.225 kg) is dissolved in acetic acid (6.75 L) and (30.3 g) was added with palladium acetate. Then 65% oleum is (247.5 g) is added and then methyl acrylate (90 g). The solution is stirred overnight at room temperature. Then, at about 30 0 C and about 30 mbar acetic acid (3740 g) were distilled off. The suspension is treated with water (2.25 L) and stirred for about 1 hour. The product is drained, washed twice with water (0.5 L) and incubated overnight at 50 0 dried C in a vacuum drying oven using entraining nitrogen. Thus, a total of 210.3 g of methyl (2E) -3- {3-fluoro-be 2 – [({[2-methoxy-5- (trifluoromethyl) phenyl] amino} carbonyl) amino] phenyl} acrylate obtained as a solid, corresponding to 72.2% of theory.

1 H NMR (300 MHz, d 6 -DMSO): δ = 9.16 (s, IH), 8.84 (s, IH), 8.45 (d, 1.7 Hz, IH), 7.73 ( m, 2H), 7.33 (m, 3H), 7.22 (d, 8.6 Hz, IH), 6.70 (d, 16Hz, IH), 3.99 (s, 3H), 3.71 (s, 3H) ppm;

MS (API-ES-pos.): M / z = 429.9 [(M + NH,) + ]; 412.9 [(M + H) + ]

HPLC: R τ = 46.4 min.

Instrument: HP 1100 Multiple Wavelength detection; Column: Phenomenex-Prodigy ODS (3) 100A, 150 mm x 3 mm, 3 microns; Eluent A: (1.36 g KH 2 PO 4 +0.7 mL H 3PO 4 ) / L water, eluent B: acetonitrile; Gradient: 0 min 20% B, 40 min 45% B, 50 min 80% B, 65 min 80% B; Flow: 0.5 mL / min; Temp .: 55 0 C; UV detection: 210 nm.

example 24

{8-FluorO-[2-methoxy-5-(trifluormethyl)phenyl]-2-oxo-l,2,3,4-tetrahydrochinazolin-4-yl}essigsäuremethylester

Methyl (2E) -3- {3-fluoro-2 – [({[2-methoxy-5- (trifluoromethyl) phenyl] amino} carbonyl) amino] phenyl} acrylate (50 g) is dissolved in acetone (1.2 L) was suspended and 3.7 g) was added l, 8-diazabicyclo [5.4.0] undec-7-ene (. The suspension is heated to reflux (ca..56 ° C) and stirred for 4 h. The resulting clear solution is hot through diatomaceous earth (5 g) was filtered. The diatomaceous earth is rinsed with warm acetone (100 ml). Subsequently, acetone (550 g) was distilled off. The resulting suspension is in 3 h at O 0 cooled and stirred C. The product is drained, washed twice with cold acetone (50 ml) and incubated overnight at 45 0 dried C in a vacuum drying oven using entraining nitrogen. Thus, a total of 44.5 g of {8-fluoro-3- [2-methoxy-5- (trifluoromethyl) phenyl] -2-oxo-1, 2, 3, 4-tetrahydrochinazo-lin-4-yl} acetic acid methyl ester as a solid, corresponding to 89% of theory.

1 H NMR (300 MHz, d 6 -DMSO): δ = 9.73 (s, IH), 7.72 (d, 2 J = 7.3, IH), 7.71 (s, IH), 7 , 33 (d, 2 J = 9.3, IH), 7.15 (dd, 2 J = 9.6, 2 J = 8.6, IH), 7.01 (d, 2 J = 7.3 , IH), 6.99 to 6.94 (m, IH), 5.16 (t, 2 J =

5.9, IH), 3.84 (s, 3H), 3.41 (s, 3H), 2.81 (dd, 1 J = 15.4, V = 5.8, IH), 2.62 (dd, 2 Vr = = 15.4, V = 6.3, IH) ppm;

MS (API-ES-pos.): M / z = 413 [(M + H) + , 100%], 825 [(2M + H) + , 14%];

HPLC: R τ = 37.1 min.

Instrument: HP 1100 Multiple Wavelength detection; Column: Phenomenex-Prodigy ODS (3) 100A, 150 mm x 3 mm, 3 microns; Eluent A: (1.36 g KH 2 PO 4 +0.7 mL H 3PO 4 ) / L water, eluent B: acetonitrile; Gradient: 0 min 20% B, 40 min 45% B, 50 min 80% B, 65 min 80% B; Flow: 0.5 mL / min; Temp .: 55 0 C; UV detection: 210 nm.

PATENT

WO 2015088931

Human cytomegalovirus (HCMV) is ubiquitously distributed in the human population. In immunocompetent adults infections are mainly asymptomatic, but in

immunocompromised patients, such as transplant recipients or AIDS patients, life threatening infections occur at a high rate. HCMV is also the leading cause of birth defects among congenitally transmitted viral infections.

Various substituted heterocyclic compounds are inhibitors of the HCMV terminase enzyme. Included in these heterocycles are quinazolines related to Compound A, as defined and described below. These compounds and pharmaceutically acceptable salts thereof are useful in the treatment or prophylaxis of infection by HCMV and in the treatment, prophylaxis, or delay in the onset or progression of HCMV infection. Representative quinazoline compounds that are useful for treating HCMV infection are described, for example, in US Patent Patent No. 7, 196,086. Among the compounds disclosed in US7, 196,086, is (S)-2-(8-fluoro-3-(2-methoxy-5-(trifluoromethyl)phenyl)-2-(4-(3-methoxyphenyl)piperazin-l-yl)-3,4-dihydroquinazolin-4-yl)acetic acid, hereinafter referred to as Compound A. Compound A is a known inhibitor of HCMV terminase. The structure of Compound A is as follows:

Compound A

US Patent Nos. 7,196,086 and 8,084,604 disclose methodology that can be employed to prepare Compound A and related quinazoline-based HCMV terminase inhibitors. These methods are practical routes for the preparation of Compound A and related heterocyclic compounds.

EXAMPLE 6

Preparation of Compound A

To a slurry of compound 7 (20g, 18.9 mmol) in MTBE (40.0 mL) at room temperature was added a solution of sodium phosphate dibasic dihydrate (8.42 g, 47.3 mmol) in water (80 mL) and the resulting slurry was allowed to stir at room temperature for 40 minutes. The reaction mixture was transferred to a separatory funnel and the organic phase was collected and washed with a solution of sodium phosphate dibasic dihydrate (3.37 g, 18.91 mmol) in water (40.0 mL). A solution of KOH (4.99 g, 76 mmol) in water (80 mL) and methanol (10.00 mL) was then added to the organic phase and the resulting mixture was heated to 50 °C and allowed to stir at this temperature for 6 hours. MTBE (20 mL) and water (40 mL) were then added to the

reaction mixture and the resulting solution was transferred to a separatory funnel and the aqueous layer was collected and washed with MTBE (20 mL). Additional MTBE (40 mL) was added to the aqueous layer and the resulting solution was adjusted to pH 4-5 via slow addition of concentrated HCl. The resulting acidified solution was transferred to a separatory funnel and the organic phase was collected, concentrated in vacuo and solvent switched with acetone, maintaining a 30 mL volume. The resulting acetone solution was added dropwise to water and the precipitate formed was filtered to provide compound A as a white solid (10 g, 92%). XH NMR (500 MHz, d6-DMSO): δΗ 12.6 (1H, s), 7.52 (1H, dd, J= 8.6, 1.3 Hz), 7.41 (1H, brs), 7.22 (1H, d, J= 7.2 Hz), 7.08-7.02 (2H, m), 6.87-6.84 (2H, m), 6.44 (1H, dd, J= 8.3, 1.8 Hz), 6.39 (1H, t, J= 2.1 Hz), 6.35 (1H, dd, J= 8.1, 2.0 Hz), 4.89 (1H, t, J= 7.3 Hz), 3.79 (3H, br s), 3.68 (3H, s), 3.47 (2H, br s), 3.39 (2H, br s), 2.96-2.93 (2H, m), 2.82-2.77 (3H, m), 2.44 (1H, dd, J = 14.8, 7.4 Hz).

XAMPLE 1

Preparation of Intermediate Compound 2


N,N-dicyclohexylmethylamine

IPAC, 80°C

To a degassed solution of 2-bromo-6-fluoroaniline (1, 99.5 g, 0.524 mol), methyl acrylate (95.0 mL, 1.05 mol), Chloro[(tri-tert-butylphosphine)-2-(2-aminobiphenyl)] palladium(II) (0.537 g, 1.05 mmol) in isopropyl acetate (796 mL), was added degassed N,N-dicyclohexylmethylamine (135 mL, 0.628 mol). The resulting reaction was heated to 80 °C and allowed to stir at this temperature for 5 hours. The resulting slurry was cooled to 20 °C and filtered. The filtrate was washed with 1 M citric acid to provide a solution that contained compound 2 (99.3 g, 97% assay yield) in isopropyl acrylate, which was used without further purification. ‘H NMR (500 MHz, d-CHCl3): δΗ 7.79 ppm (1H, d, J= 15.9 Hz), 7.17 ppm (1H, d, J= 8.2 Hz), 7.00 ppm (1H, ddd, J= 10.7, 8.2, 1.2 Hz), 6.69 ppm (1H, td, J = 8.2, 5.1 Hz), 6.38 ppm (1H, d, J= 15.9 Hz), 4.06 ppm (2H, br s), 3.81 ppm (3H, s).

EXAMPLE 2

Preparation of Intermediate Compound 3

To a solution of compound 2 (48.8 g, 0.250 mol) in 683 mL of isopropyl acetate was added 244 mL of water, followed by di-sodium hydrogen phosphate (53.2 g, 0.375 mol). To the resulting solution was added phenyl chloroformate (39.2 mL, 0.313 mol) dropwise over 30 minutes. The resulting reaction was heated to 30 °C and allowed to stir at this temperature for 5 hours for 4 hours and then was heated to 60 °C and allowed to stir at this temperature for 5 hours for an additional 2 hours to remove excess phenyl chloroformate. An additional 293 mL of isopropyl acetate was then added and the reaction mixture was allowed to stir at room temperature until the solids completely dissolved into solution. The resulting reaction mixture was transferred to a separatory funnel and the organic phase was washed with 98 mL of water and collected to provide a solution of compound 3 in isopropyl acetate, which was used without further purification. XH NMR (500 MHz, d-acetonitrile): δΗ 7.91 ppm (1H, d, J= 15.9 Hz), 7.85 ppm (1H, br s), 7.63 ppm (1H, d, J= 7.9 Hz), 7.45-7.39 ppm (3H, m), 7.33-7.27 ppm (2H, m), 7.21 ppm (2H, br), 6.60 ppm (1H, d, J= 16.0 Hz).

EXAMPLE 3

Preparation of Intermediate Compound 4

A solution of compound 3 (79.0 g, 0.250 mol), 2-methoxy-5-(trifluoromethyl)aniline (52.7 g, 0.276 mol), and 4-dimethylaminopyridine (0.92 g, 0.0075 mol) in isopropyl acetate (780 mL) was heated to reflux and allowed to stir at this temperature for 5 hours. The resulting slurry was cooled to 20 °C, then allowed to stir at this temperature for for two hours at this temperature, then filtered. The collected filter cake was dried in vacuo to provide compound 5 (95.0 g, 0.230 mol) as a white solid, which was used without further purification. ¾ NMR (500 MHz, d-TFA): δΗ 7.98 ppm (1H, d, J= 16.1 Hz), 7.87 ppm (1H, s), 7.47 ppm (1H, d, J = 7.9 Hz), 7.41 ppm (1H, d, J= 8.5 Hz), 7.35 ppm (1H, q, J= 8.5 Hz), 7.19 ppm (1H, t, J= 8.6 Hz), 6.98 ppm (1H, d, J= 8.6 Hz), 6.56 ppm (1H, d, J= 16.0 Hz), 3.85 ppm (6H, br s).

EXAMPLE 4

Preparation of Intermediate Compound 6

To a stirred suspension of compound 4 (14.0 g, 34.0 mmol) in toluene (140 mL) at room temperature was added 2-picoline (10.1 mL, 102 mmol) followed by PCI5 (8.19 g, 37.3 mmol). The resulting reaction was heated to 40 °C and allowed to stir at this temperature for 4 hours, then was cooled to 0 °C and cautiously (internal temperature kept <15 °C) quenched with KOH (2 M, 102 mL). The resulting solution was allowed to warm to room temperature, allowed to stir for 30 minutes, then was filtered and the filtrate transferred to a separatory funnel. The organic phase was washed sequentially with H3PO4 (1M, 50 mL) and H20 (50 mL) to provide a solution of compound 5 in toluene, which was used without further purification. XH NMR (500 MHz, d6-DMSO): δΗ 7.96 (1H, d, J= 16.2 Hz), 7.74 (1H, d, J= 7.9 Hz), 7.61 (1H, dd, J= 6.7, 1.6 Hz), 7.50 (1H, d, J= 1.9 Hz), 7.43 (1H, t, J= 9.2 Hz), 7.30 (1H, d, J= 8.4 Hz), 7.28 (1H, m), 6.79 (1H, d, J= 16.2 Hz), 3.91 (3H, s), 3.74 (3H, s).

To the solution of compound 5 at room temperature was added an aqueous solution of piperazine hydrochloride (0.40 M, 93.3 mL, 37.3 mmol) followed by Na2HP04 (14.5 g, 102 mmol). The resulting reaction was allowed to stir for 1 hour at room temperature, then transferred to a separatory funnel. The organic phase was washed sequentially with aH2P04 (50 mL) and H20 (50 mL). Salicylic acid (5.16 g, 37.3 mmol) was then added to the organic phase, and the resulting solution was cooled to 0 °C and allowed to stir at this temperature for 1 hour to provide a slurry which was filtered and washed with cold toluene (50 mL). The filter cake was dried under air to provide compound 6 (23.0 g, 31.7 mmol, 93 %) as a white crystalline solid: XH NMR (500 MHz, d6-DMSO): δΗ 12.9 (1H, br s), 7.75 (1H, dd, J= 7.8, 1.8 Hz), 7.72 (1H, d, J= 16.1 Hz), 7.40 (1H, td, J= 7.2, 1.7 Hz), 7.27 (1H, d, J= 7.8 Hz), 7.17 (1H, m), 7.16 (1H, t, J= 8.2 Hz), 7.02 (1H, br s), 6.95 (1H, t, J= 8.6 Hz), 6.88-6.81 (3H, m), 6.78 (1H, br s), 6.60 (1H, dd, J= 8.2, 2.0 Hz), 6.54 (1H, m), 6.48 (1H, d, J= 16.1 Hz), 6.43 (1H, dd, J= 8.0, 2.1 Hz), 3.73 (3H, s), 3.71 (3H, s), 3.69 (4H, br s), 3.68 (3H, s).

Free Base: XH NMR (500 MHz, CD3CN): δΗ 7.91 (1H, d, J= 16.1 Hz), 7.29 (1H, d, J= 8.0 Hz), 7.24 (1H, d, J= 1.4 Hz), 7.20 (1H, t, J= 8.1 Hz), 7.15 (1H, dd, J= 8.6, 1.4 Hz), 6.94 (1H, m), 6.92 (1H, t, J= 8.1 Hz), 6.80 (1H, td, J= 8.1, 5.4 Hz), 6.60 (1H, dd, J= 8.3, 2.2 Hz), 6.54 (1H, t, J= 2.2 Hz), 6.50 (1H, d, J= 16.1 Hz), 6.47 (2H, m), 3.80 (3H, s), 3.79 (3H, s), 3.72 (3H, s), 3.63 (4H, t, J= 5.1 Hz), 3.25 (4H, t, J= 5.0 Hz).

2: 1 NDSA Salt: ‘H NMR (500 MHz, d6-DMSO): δΗ 10.2 (2H, br s), 8.86 (1H, d, J= 8.6 Hz), 7.92 (1H, d, J= 7.0 Hz), 7.47-7.37 (4H, m), 7.27-7.14 (4H, m), 6.96 (1H, d, J= 8.6 Hz), 6.65 (1H, d, J= 8.3 Hz), 6.59 (1H, s), 6.54 (1H, d, J= 15.9 Hz), 6.47 (1H, d, J= 8.3 Hz), 3.91 (4H, m), 3.77 (3H, s), 3.76 (3H, s), 3.74 (3H, s), 3.43 (4H, m). 1,5 -naphthalene disulfonic acid

EXAMPLE 5

Preparation of Intermediate Compound 7

To a suspension of compound 6 (12.5 g, 16.6 mmol) in 125 mL of toluene was added 50 mL of 0.43M aqueous K3P04. The resulting reaction was allowed to stir for 1 hour at room temperature and the reaction mixture was transferred to a separatory funnel. The organic phase was collected, washed once with 30 mL 0.43M aqueous K3P04then cooled to 0 °C and aqueous K3P04 (60 mL, 0.43 M, 25.7 mmol) was added. To the resulting solution was added a room temperature solution of ((lS,2S,4S,5R)-l-(3,5-bis(trifluoromethyl)benzyl)-2-((R)-

hydroxy( 1 -(3 -(trifluoromethyl)benzyl)quinolin- 1 -ium-4-yl)methyl)-5-vinylquinuclidin- 1 -ium bromide) (0.704 g, 0.838 mmol) in 1.45 mL of DMF. The resulting reaction was allowed to stir at 0 °C until the reaction was complete (monitored by HPLC), then the reaction mixture was transferred to a separatory funnel and the organic phase was collected and washed sequentially with 1M glycolic acid (25 mL) and water (25 mL). The organic phase was filtered through solka flok and concentrated in vacuo to a total volume of 60 mL. Ethyl acetate (20 mL) was added to the resulting solution, followed by (S,S)-Di-P-Toluoyl-D-tartaric acid (5.61 g, 14.1 mmol). Penultimate seed (0.2 g) was added the resulting solution was allowed to stir at room

temperature for 12 hours. The solution was then filtered and the collected solid was washed twice with ethyl acetate, then dried in vacuo to provide compound 7 as its DTTA salt ethyl acetate solvate (13.8 g, 78%) . ‘H NMR (500 MHz, d6-DMSO): δΗ 13.95 (2H, br s), 7.90 (4H, d, J= 8.1 Hz), 7.55 (1H, dd, J= 8.6, 1.3 Hz), 7.38 (4H, d, J= 8.1 Hz), 7.26 (1H, d, J= 7.8 Hz), 7.09-7.05 (3H, m), 6.91-6.86 (2H, m), 6.44 (1H, dd, J= 8.2, 1.7 Hz), 6.39 (1H, t, J= 2.0 Hz), 6.36 (1H, dd, J= 8.2, 2.0 Hz), 5.82 (2H, s), 4.94 (1H, t, J= 7.1 Hz), 4.02 (2H, q, J= 7.1 Hz), 3.83 (3H, br s), 3.68 (3H, s), 3.64 (3H, s), 3.47 (2H, br s), 3.37 (2H, br s), 2.95 (2H, br s), 2.87- 2.80 (3H, m), 2.56 (1H, dd, J= 14.3, 7.0 Hz), 2.39 (6H, s), 1.98 (3H, s), 1.17 (3H, t, J= 7.1 Hz).

PAPER

Asymmetric Synthesis of Letermovir Using a Novel Phase-Transfer-Catalyzed Aza-Michael Reaction

Department of Process Chemistry, Merck and Co., Inc., P.O. Box 2000, Rahway, New Jersey 07065, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.6b00076
Publication Date (Web): May 13, 2016
Copyright © 2016 American Chemical Society

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

Abstract

Abstract Image

The development of a concise asymmetric synthesis of the antiviral development candidate letermovir is reported, proceeding in >60% yield over a total of seven steps from commercially available materials. Key to the effectiveness of this process is a novel cinchonidine-based PTC-catalyzed aza-Michael reaction to configure the single stereocenter.

http://pubs.acs.org/doi/full/10.1021/acs.oprd.6b00076

(S)-2-(8-Fluoro-3-(2-methoxy-5-(trifluoromethyl)phenyl)-2-(4-(3-methoxyphenyl)piperazin-1-yl)-3,4-dihydroquinazolin-4-yl)acetic Acid (Letermovir, 1)

 letermovir (1, 20.2 g, 35.3 mmol, 100 wt %, 94%) as an amorphous white powder. 1H NMR (DMSO-d6, 600 MHz) δH 7.52 (dd, J = 8.7, 1.7 Hz, 1H), 7.40 (brs, 1H), 7.21 (m, 1H), 7.07 (t, J = 8.2 Hz, 1H), 7.04 (m, 1H), 6.87 (m, 2H), 6.44 (dd, J = 8.2, 1.9 Hz, 1H), 6.40 (t, J = 2.3 Hz, 1H), 6.36 (dd, J = 8.0, 2.0 Hz, 1H), 4.89 (t, J = 7.2 Hz, 1H), 3.80 (brs, 3H), 3.68 (s, 3H), 3.39–3.48 (m, 4H), 2.82–2.95 (m, 4H), 2.80 (dd, J = 14.8, 7.4 Hz, 1H), 2.46 (dd, J = 14.9, 7.4 Hz, 1H); 13C NMR (DMSO-d6, 150 MHz) δC 171.8, 160.2, 156.5, 154.6 (d, JCF = 246.3 Hz), 153.2, 152.2, 134.2, 132.3 (d, JCF = 11.2 Hz), 129.6, 124.1 (q, JCF = 271.3 Hz), 123.8 (q, JCF = 3.7 Hz), 122.4, 122.1 (q, JCF = 7.1 Hz), 121.4 (q, JCF = 29.2 Hz), 120.8, 114.5 (d, JCF = 19.5 Hz), 113.3, 108.3, 104.6, 101.9, 59.0, 56.3, 54.8, 47.9, 45.6, 40.0; HR-MS calcd for C29H29F4N4O4+ [M + H]+ 573.2119, found 573.2117 (Δ = 0.2 mmu).

References

Masangkay, Estel Grace (July 29, 2014). “Merck Kicks Off Phase 3 Study Of CMV Drug Letermovir”. Retrieved 8 Oct 2014.

Patent ID Date Patent Title
US8084604 2011-12-27 Process for the Preparation of Dihydroquinazolines
US2007191387 2007-08-16 Substituted dihydroquinazolines
Patent ID Date Patent Title
US2015133461 2015-05-14 PHARMACEUTICAL COMPOSITION CONTAINING AN ANTIVIRALLY ACTIVE DIHYDROQUINAZOLINE DERIVATIVE
US2015050241 2015-02-19 METHOD OF TREATING VIRAL INFECTIONS
US2015045371 2015-02-12 Salts of a dihydroquinazoline derivative
US2015038514 2015-02-05 SODIUM AND CALCIUM SALTS OF DIHYDROQUINAZOLINE DERIVATIVE AND USE THEREOF AS ANTIVIRAL AGENTS
US2015038728 2015-02-05 NOVEL ARYLATED CAMPHENES, PROCESSES FOR THEIR PREPARATION AND USES THEREOF
US8816075 2014-08-26 Process for the preparation of dihydroquinazolines
US2014193802 2014-07-10 IDENTIFICATION OF AN ALTERED THERAPEUTIC SUSCEPTIBILITY TO ANTI-HCMV COMPOUNDS AND OF A RESISTANCE AGAINST ANTI-HCMV COMPOUNDS
US2014178432 2014-06-26 PRODUCTION OF DENSE BODIES (DB) FROM HCMV-INFECTED CELLS
US8372972 2013-02-12 Process for the preparation of dihydroquinazolines
US8084604 2011-12-27 Process for the Preparation of Dihydroquinazolines
Letermovir
Letermovir skeletal.svg
Systematic (IUPAC) name
{(4S)-8-Fluoro-2-[4-(3-methoxyphenyl)-1-piperazinyl]-3-[2-methoxy-5-(trifluoromethyl)phenyl]-3,4-dihydro-4-quinazolinyl}acetic acid
Clinical data
Routes of
administration
Oral
Legal status
Legal status
  • Investigational
Identifiers
ATC code None
PubChem CID 45138674
ChemSpider 26352849
UNII 1H09Y5WO1F Yes
ChEMBL CHEMBL1241951
Synonyms AIC246
Chemical data
Formula C29H28F4N4O4
Molar mass 572.55 g/mol

/////Letermovir, MK 8828, AIC 246, fast track status, US Food and Drug Administrationorphan drug status ,  European Medicines Agency

COC1=C(C=C(C=C1)C(F)(F)F)N2[C@H](C3=C(C(=CC=C3)F)N=C2N4CCN(CC4)C5=CC(=CC=C5)OC)CC(=O)O


Filed under: Preclinical drugs Tagged: AIC 246, European Medicines Agency, Fast Track status, Letermovir, MK 8828, Orphan Drug Status, US Food and Drug Administration
Viewing all 1640 articles
Browse latest View live