Quantcast
Channel: New Drug Approvals
Viewing all 2871 articles
Browse latest View live

FDA approves treatment Poteligeo (mogamulizumab-kpkc) for two rare types of non-Hodgkin lymphoma

$
0
0

 

FDA approves treatment for two rare types of non-Hodgkin lymphoma

The U.S. Food and Drug Administration today approved Poteligeo (mogamulizumab-kpkc) injection for intravenous use for the treatment of adult patients with relapsed or refractory mycosis fungoides (MF) or Sézary syndrome (SS) after at least one prior systemic therapy. This approval provides a new treatment option for patients with MF and is the first FDA approval of a drug specifically for SS.

August 8, 2018

Release

The U.S. Food and Drug Administration today approved Poteligeo (mogamulizumab-kpkc) injection for intravenous use for the treatment of adult patients with relapsed or refractory mycosis fungoides (MF) or Sézary syndrome (SS) after at least one prior systemic therapy. This approval provides a new treatment option for patients with MF and is the first FDA approval of a drug specifically for SS.

“Mycosis fungoides and Sézary syndrome are rare, hard-to-treat types of non-Hodgkin lymphoma and this approval fills an unmet medical need for these patients,” said Richard Pazdur, M.D., director of the FDA’s Oncology Center of Excellence and acting director of the Office of Hematology and Oncology Products in the FDA’s Center for Drug Evaluation and Research. “We are committed to continuing to expedite the development and review of this type of targeted therapy that offers meaningful treatments for patients.”

Non-Hodgkin lymphoma is a cancer that starts in white blood cells called lymphocytes, which are part of the body’s immune system. MF and SS are types of non-Hodgkin lymphoma in which lymphocytes become cancerous and affect the skin. MF accounts for about half of all lymphomas arising from the skin. It causes itchy red rashes and skin lesions and can spread to other parts of the body. SS is a rare form of skin lymphoma that affects the blood and lymph nodes.

Poteligeo is a monoclonal antibody that binds to a protein (called CC chemokine receptor type 4 or CCR4) found on some cancer cells.

The approval was based on a clinical trial of 372 patients with relapsed MF or SS who received either Poteligeo or a type of chemotherapy called vorinostat. Progression-free survival (the amount of time a patient stays alive without the cancer growing) was longer for patients taking Poteligeo (median 7.6 months) compared to patients taking vorinostat (median 3.1 months).

The most common side effects of treatment with Poteligeo included rash, infusion-related reactions, fatigue, diarrhea, musculoskeletal pain and upper respiratory tract infection.

Serious warnings of treatment with Poteligeo include the risk of dermatologic toxicity, infusion reactions, infections, autoimmune problems (a condition where the immune cells in the body attack other cells or organs in the body), and complications of stem cell transplantation that uses donor stem cells (allogeneic) after treatment with the drug.

The FDA granted this application Priority Review and Breakthrough Therapydesignation. Poteligeo also received Orphan Drug designation, which provides incentives to assist and encourage the development of drugs for rare diseases.

The FDA granted this approval to Kyowa Kirin, Inc.

///////////////// Poteligeo, mogamulizumab-kpkc, fda 2018, Kyowa Kirin, Priority Review, Breakthrough Therapy designation,  Orphan Drug designation

Spiramycin, スピラマイシン

$
0
0

8025-81-8.pngSpiramycin I.svg

ChemSpider 2D Image | [(11E,13E)-6-({5-[(4,5-Dihydroxy-4,6-dimethyltetrahydro-2H-pyran-2-yl)oxy]-4-(dimethylamino)-3-hydroxy-6-methyltetrahydro-2H-pyran-2-yl}oxy)-10-{[5-(dimethylamino)-6-methyltetrahydro-2H-pyran-2-yl]oxy }-4-hydroxy-5-methoxy-9,16-dimethyl-2-oxooxacyclohexadeca-11,13-dien-7-yl]acetaldehyde | C43H74N2O14Spiramycin.png2D chemical structure of 8025-81-8

ThumbChemSpider 2D Image | 033ECH6IFG | C43H74N2O14

Spiramycin

スピラマイシン

CAS: 8025-81-8

Sanofi INNOVATOR

Molecular Formula: C43H74N2O14
Molecular Weight: 843.065 g/mol

[(11E,13E)-6-({5-[(4,5-Dihydroxy-4,6-dimethyltetrahydro-2H-pyran-2-yl)oxy]-4-(dimethylamino)-3-hydroxy-6-methyltetrahydro-2H-pyran-2-yl}oxy)-10-{[5-(dimethylamino)-6-methyltetrahydro-2H-pyran-2-yl]oxy }-4-hydroxy-5-methoxy-9,16-dimethyl-2-oxooxacyclohexadeca-11,13-dien-7-yl]acetaldehyde

2-[(11E,13E)-6-[5-(4,5-dihydroxy-4,6-dimethyloxan-2-yl)oxy-4-(dimethylamino)-3-hydroxy-6-methyloxan-2-yl]oxy-10-[5-(dimethylamino)-6-methyloxan-2-yl]oxy-4-hydroxy-5-methoxy-9,16-dimethyl-2-oxo-1-oxacyclohexadeca-11,13-dien-7-yl]acetaldehyde

Leucomycin V, 9-O-[(2R,5S,6R)-5-(dimethylamino)tetrahydro-6-methyl-2H-pyran-2-yl]-

033ECH6IFG
24916-50-5 [RN]
Spiramycin I
[(4R,5S,6S,7R,9R,10R,11E,13E,16R)-6-{[(2S,3R,4R,5S,6R)-5-{[(2S,4R,5S,6S)-4,5-dihydroxy-4,6-dimethyloxan-2-yl]oxy}-4-(dimethylamino)-3-hydroxy-6-methyloxan-2-yl]oxy}-10-{[(2R,5S,6S)-5-(dimethylamino)-6
  • Provamycin
  • Rovamycin
  • RP 5337
  • Sequamycin
  • IL 5902
  • NSC-64393
  • ATC:J01FA02
  • Use:macrolide antibiotic
  • EINECS:232-429-6
  • LD50:130 mg/kg (M, i.v.); 2900 mg/kg (M, p.o.);
    170 mg/kg (R, i.v.); 3550 mg/kg (R, p.o.);
    5200 mg/kg (dog, p.o.)

2018/7/2 japan approved, UNII: 71ODY0V87H

Solubility

Slightly soluble in water

O’Neil, M.J. (ed.). The Merck Index – An Encyclopedia of Chemicals, Drugs, and Biologicals. Cambridge, UK: Royal Society of Chemistry, 2013., p. 1621

Soluble in most organic solvents

O’Neil, M.J. (ed.). The Merck Index – An Encyclopedia of Chemicals, Drugs, and Biologicals. Cambridge, UK: Royal Society of Chemistry, 2013., p. 1621

Spectral Properties

UV max (ethanol): 231nm

O’Neil, M.J. (ed.). The Merck Index – An Encyclopedia of Chemicals, Drugs, and Biologicals. Cambridge, UK: Royal Society of Chemistry, 2013., p. 1621

Specific optical rotation: -80 deg at 20 deg C/D

O’Neil, M.J. (ed.). The Merck Index – An Encyclopedia of Chemicals, Drugs, and Biologicals. Cambridge, UK: Royal Society of Chemistry, 2013., p. 1621
スピラマイシン
Spiramycin

スピラマイシン酢酸エステル JP17
Spiramycin Acetate

A macrolide antibiotic produced by Streptomyces ambofaciens. The drug is effective against gram-positive aerobic pathogens, N. gonorrhoeae, and staphylococci. It is used to treat infections caused by bacteria and Toxoplasma gondii.

Spiramycin is a macrolide antimicrobial agent with activity against gram-positive organisms, including Streptococcus pyogenes (group A beta-hemolytic streptococci), S. viridans, Corynebacterium diphtheriae, and methicillin-sensitive Staphylococcus aureus. Spiramycin is a 16-membered ring macrolide. It was discovered in 1952 as a product of Streptomyces ambofaciens. As a preparation for oral administration it has been used since 1955, in 1987 also the parenteral form was introduced into practice. The antibacterial spectrum comprises Gram-positive cocci and rods, Gram-negative cocci and also Legionellae, mycoplasmas, chlamydiae, some types of spirochetes, Toxoplasma gondii and Cryptosporidium species. Enterobacteria, pseudomonads and pathogenic moulds are resistant. Its action is mainly bacteriostatic, on highly sensitive strains it exerts a bactericide action.

Spiramycin is a macrolide antibiotic and antiparasitic It is used to treat toxoplasmosis and various other infections of soft tissues. Although used in Europe, Canada and Mexico,[1] spiramycin is still considered an experimental drug in the United States, but can sometimes be obtained by special permission from the FDA for toxoplasmosis in the first trimester of pregnancy.[2]

Spiramycin has been used in Europe since the year 2000 under the trade name “Rovamycine”, produced by Rhone-Poulenc Rorer and Famar Lyon, France and Eczacıbaşı İlaç, Turkey. It also goes under the name Rovamycine in Canada (distributed by OdanLaboratories), where it is mostly marketed to dentists for mouth infections.

Spiramycin is a 16-membered ring macrolide. It was discovered in 1952 as a product of Streptomyces ambofaciens. As a preparation for oral administration it has been used since 1955, in 1987 also the parenteral form was introduced into practice. The antibiotic action involves inhibition of protein synthesis in the bacterial cell during translocation. Resistance to spiramycin can develop by several mechanisms and its prevalence is to a considerable extent proportional to the frequency of prescription in a given area. The antibacterial spectrum comprises Gram-positive cocci and rods, Gram-negative cocci and also Legionellae, mycoplasmas, chlamydiae, some types of spirochetes, Toxoplasma gondii and Cryptosporidium species. Enterobacteria, pseudomonads and pathogenic moulds are resistant. Its action is mainly bacteriostatic, on highly sensitive strains it exerts a bactericide action. As compared with erythromycin, it is in vitro weight for weight 5 to 20 less effective, an equipotential therapeutic dose is, however, only double. This difference between the effectiveness in vitro and in vivo is explained above all by the great affinity of spiramycin to tissues where it achieves concentrations many times higher than serum levels. An important part is played also by the slow release of the antibiotic from the tissue compartment, the marked action on microbes in sub-inhibition concentrations and the relatively long persisting post-antibiotic effect. Its great advantage is the exceptionally favourable tolerance-gastrointestinal and general. It is available for parenteral and oral administration

Synthesis Path

  • From culture of Streptomyces ambofaciens.

Trade Names

Country Trade Name Vendor Annotation
D Rovamycine Teofarma
Selectomycin Grünenthal
F Bi Missilor Pierre Fabre
Birodogyl Sanofi-Aventis
Missilor Pierre Fabre comb.
Rodogyl Pierre Fabre
Rovamycine Grünenthal
I Rovamicina Sanofi-Aventis
Rovamycina Teofarma
Spiromix Pulitzer

Spiramycin

Title: Spiramycin
CAS Registry Number: 8025-81-8
Manufacturers’ Codes: RP-5337
Trademarks: Selectomycin (Grñenthal); Rovamicina (RPR); Rovamycin (RPR)
Literature References: Antibiotic substance classified in the erythromycin-carbomycin group and produced by Streptomyces ambofaciens from soil of northern France: Cosar et al., C.R. Seances Soc. Biol. Ses Fil. 234, 1498 (1952); Pinnert-Sindico et al.,Antibiot. Annu. 1954-1955, 724; Ninet, Verrier, US 2943023 (1960 to Rhône-Poulenc), see also US 3000785 (1961 to Rhône-Poulenc). Antibacterial activity and toxicity: H. Sous et al., Arzneim.-Forsch. 8, 386 (1958). Separation into 3 components named spiramycin I, II and III: Preud’homme, Charpentier, US 2978380 and US 3011947 (1961 to Rhône-Poulenc). Structure: Kuehne, Benson, J. Am. Chem. Soc. 87, 4660 (1965). Revised structure: Omura et al., ibid. 91, 3401 (1969); Mitscher et al., J. Antibiot. 26,55 (1973). Revised configuration at C-9: Freiberg et al., J. Org. Chem. 39, 2474 (1974). Symposium on pharmacology, antibacterial spectrum, and clinical efficacy: J. Antimicrob. Chemother. 22, Suppl. B, 1-213 (1988).
Properties: Amorphous base, slightly sol in water. [a]D20 -80° (methanol). uv max (ethanol): 231 nm. Sol in most organic solvents. Active on gram-positive bacteria and rickettsiae. Cross resistance between microorganisms resistant to erythromycin and carbomycin. LD50 in rats (mg/kg): 9400 orally; 1000 s.c.; 170 i.v. (Sous).
Optical Rotation: [a]D20 -80° (methanol)
Absorption maximum: uv max (ethanol): 231 nm
Toxicity data: LD50 in rats (mg/kg): 9400 orally; 1000 s.c.; 170 i.v. (Sous)
Derivative Type: Embonate
Trademarks: Spira 200 (RMB)
Derivative Type: Hexanedioate
Additional Names: Spiramycin adipate
Trademarks: Stomamycin (Chassot); Suanovil (Biokema)
Derivative Type: Spiramycin I
CAS Registry Number: 24916-50-5
Additional Names: Foromacidin A
Molecular Formula: C43H74N2O14
Molecular Weight: 843.05
Percent Composition: C 61.26%, H 8.85%, N 3.32%, O 26.57%
Properties: Crystals, mp 134-137°. [a]D20 -96°.
Melting point: mp 134-137°
Optical Rotation: [a]D20 -96°
Derivative Type: Spiramycin I triacetate
Properties: Crystals, mp 140-142°. [a]D20 -92.5°.
Melting point: mp 140-142°
Optical Rotation: [a]D20 -92.5°
Derivative Type: Spiramycin II
CAS Registry Number: 24916-51-6
Additional Names: Foromacidin B
Molecular Formula: C45H76N2O15
Molecular Weight: 885.09
Percent Composition: C 61.07%, H 8.65%, N 3.17%, O 27.11%
Properties: Crystals, mp 130-133°. [a]D20 -86°.
Melting point: mp 130-133°
Optical Rotation: [a]D20 -86°
Derivative Type: Spiramycin II diacetate
Properties: Crystals from cyclohexane, mp 156-160°. [a]D20 -98.4°.
Melting point: mp 156-160°
Optical Rotation: [a]D20 -98.4°
Derivative Type: Spiramycin III
CAS Registry Number: 24916-52-7
Additional Names: Foromacidin C
Molecular Formula: C46H78N2O15
Molecular Weight: 899.12
Percent Composition: C 61.45%, H 8.74%, N 3.12%, O 26.69%
Properties: Crystals, mp 128-131°. [a]D20 -83°.
Melting point: mp 128-131°
Optical Rotation: [a]D20 -83°
Derivative Type: Spiramycin III diacetate
Properties: Crystals from cyclohexane, mp 140-142°. [a]D20 -90.4°.
Melting point: mp 140-142°
Optical Rotation: [a]D20 -90.4°
Therap-Cat: Antibacterial.
Therap-Cat-Vet: Antibacterial; growth promotant.
Keywords: Antibacterial (Antibiotics); Macrolides.
Spiramycin
Spiramycin I.svg
Clinical data
Synonyms 2-[(4R,5S,6S,7R,9R,10R,11E,13E,16R)-6-{[(2S,3R,4R,5S,6R)-5-{[(2S,5S,6S)-4,5-dihydroxy-4,6-dimethyloxan-2-yl]oxy}-4-(dimethylamino)-3-hydroxy-6-methyloxan-2-yl]oxy}-10-{[(2R,5S,6R)-5-(dimethylamino)-6-methyloxan-2-yl]oxy}-4-hydroxy-5-methoxy-9,16-dimethyl-2-oxo-1-oxacyclohexadeca-11,13-dien-7-yl]acetaldehyde
Routes of
administration
oral
ATC code
Legal status
Legal status
  • In general: ℞ (Prescription only)
Identifiers
CAS Number
PubChem CID
ChemSpider
UNII
KEGG
ChEBI
ChEMBL
NIAID ChemDB
E number E710 (antibiotics) Edit this at Wikidata
ECHA InfoCard 100.029.476 Edit this at Wikidata
Chemical and physical data
Formula C43H74N2O14
Molar mass 843.053 g/mol
3D model (JSmol)
Solubility in water Insoluble in water; Very soluble in acetonitrile and methanol; Almost completely(>99.5) in ethanol. mg/mL (20 °C)

References

  1. Jump up^ Spiramycin advanced consumer information | Drugs.com
  2. Jump up^ Toxoplasmosis at MayoClinic.com

References

    • Pinnert-Sindico, S. et al.: Antibiot. Annu. (ABANAE) 1954-1955, 724.
    • US 2 943 023 (Rhône-Poulenc; 28.6.1960; F-prior. 30.5.1956).
    • US 2 978 380 (Rhône-Poulenc; 4.4.1961; F-prior. 30.11.1955).
    • US 3 000 785 (Rhône-Poulenc; 19.9.1961; F-prior. 31.7.1953).
    • US 3 011 947 (Rhône-Poulenc; 5.12.1961; F-prior. 30.11.1955).
    • CN 107266512

      CN 107840864

DB06145.png

Spiramycin I)

///////////Spiramycin, スピラマイシン , japan 2018, Provamycin, Rovamycin, RP 5337, Sequamycin, IL 5902, NSC-64393, ATC:J01FA02, Use:macrolide antibiotic, EINECS:232-429-6

 

O=CCC4C(OC2OC(C(OC1OC(C)C(O)C(O)(C)C1)C(N(C)C)C2O)C)C(OC)C(O)CC(=O)OC(C)C\C=C\C=C\C(OC3OC(C)C(N(C)C)CC3)C(C)C4

FDA approves new treatment Galafold (migalastat) for a rare genetic disorder, Fabry disease

$
0
0

FDA approves new treatment for a rare genetic disorder, Fabry disease

The U.S. Food and Drug Administration today approved Galafold (migalastat), the first oral medication for the treatment of adults with Fabry disease. The drug is indicated for adults with Fabry disease who have a genetic mutation determined to be responsive (“amenable”) to treatment with Galafold based on laboratory data. Fabry disease is a rare and serious genetic disease that results from buildup of a type of fat called globotriaosylceramide (GL-3) in blood vessels, the kidneys, the heart, the nerves and other organs.

August 10, 2018

Release

The U.S. Food and Drug Administration today approved Galafold (migalastat), the first oral medication for the treatment of adults with Fabry disease. The drug is indicated for adults with Fabry disease who have a genetic mutation determined to be responsive (“amenable”) to treatment with Galafold based on laboratory data. Fabry disease is a rare and serious genetic disease that results from buildup of a type of fat called globotriaosylceramide (GL-3) in blood vessels, the kidneys, the heart, the nerves and other organs.

“Thus far, treatment of Fabry disease has involved replacing the missing enzyme that causes the particular type of fat buildup in this disease. Galafold differs from enzyme replacement in that it increases the activity of the body’s deficient enzyme,” said Julie Beitz, M.D., director of the Office of Drug Evaluation III in FDA’s Center for Drug Evaluation and Research.

Fabry disease is an inherited disorder caused by mutations (alterations) in the alpha-galactosidase A (GLA) gene located on the X-chromosome. Fabry disease is rare and affects both males and females. It is estimated that classic Fabry disease (the most severe type) affects approximately one in 40,000 males. The later-onset type is more frequent, and in some populations, may occur in one in 1,500 to 4,000 males. Patients with Fabry disease develop slowly progressive kidney disease, cardiac hypertrophy (enlargement of the heart), arrhythmias (abnormal heart rhythm), stroke and early death.

The efficacy of Galafold was demonstrated in a six-month, placebo-controlled clinical trial in 45 adults with Fabry disease. In this trial, patients treated with Galafold over six months had a greater reduction in globotriaosylceramide (GL-3) in blood vessels of the kidneys (as measured in kidney biopsy samples) as compared to patients on placebo.The safety of Galafold was studied in four clinical trials which included a total of 139 patients with Fabry disease.

The most common adverse drug reactions in patients taking Galafold in clinical trials were headache, nasal and throat irritation (nasopharyngitis), urinary tract infection, nausea, and fever (pyrexia).

Galafold was approved using the Accelerated Approval pathway, under which the FDA may approve drugs for serious conditions where there is an unmet medical need and where a drug is shown to have certain effects that are reasonably likely to predict a clinical benefit to patients. A further study is required to verify and describe the clinical benefits of Galafold, and the sponsor will be conducting a confirmatory clinical trial of Galafold in adults with Fabry disease.

Galafold  was granted Priority Review designation, under which the FDA’s goal is to take action on an application within six months of application filing where the agency determines that the drug, if approved, would provide a significant improvement in treating, diagnosing or preventing a serious condition over available therapies. Galafold also received Orphan Drug designation, which provides incentives to assist and encourage the development of drugs for rare diseases.
The FDA granted approval of Galafold to Amicus Therapeutics U.S., Inc.

///////////////fda 2018, Galafold, migalastat, Fabry disease, Amicus Therapeutics

FDA approves first-of-its kind targeted RNA-based therapy Onpattro (patisiran) to treat a rare disease

$
0
0

Image result for patisiran

FDA approves first-of-its kind targeted RNA-based therapy to treat a rare disease

First treatment for the polyneuropathy of hereditary transthyretin-mediated amyloidosis in adult patients

The U.S. Food and Drug Administration today approved Onpattro (patisiran) infusion for the treatment of peripheral nerve disease (polyneuropathy) caused by hereditary transthyretin-mediated amyloidosis (hATTR) in adult patients. This is the first FDA-approved treatment for patients with polyneuropathy caused by hATTR, a rare, debilitating and often fatal genetic disease characterized by the buildup of abnormal amyloid protein in peripheral nerves, the heart and other organs. It is also the first FDA approval of a new class of drugs called small interfering ribonucleic acid (siRNA) treatment

Continue reading…

https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/UCM616518.htm?utm_campaign=08102018_PR_FDA%20approves%20new%20drug%20for%20rare%20disease%2C%20hATTR&utm_medium=email&utm_source=Eloqua

August 10, 2018

Release

The U.S. Food and Drug Administration today approved Onpattro (patisiran) infusion for the treatment of peripheral nerve disease (polyneuropathy) caused by hereditary transthyretin-mediated amyloidosis (hATTR) in adult patients. This is the first FDA-approved treatment for patients with polyneuropathy caused by hATTR, a rare, debilitating and often fatal genetic disease characterized by the buildup of abnormal amyloid protein in peripheral nerves, the heart and other organs. It is also the first FDA approval of a new class of drugs called small interfering ribonucleic acid (siRNA) treatment.

“This approval is part of a broader wave of advances that allow us to treat disease by actually targeting the root cause, enabling us to arrest or reverse a condition, rather than only being able to slow its progression or treat its symptoms. In this case, the effects of the disease cause a degeneration of the nerves, which can manifest in pain, weakness and loss of mobility,” said FDA Commissioner Scott Gottlieb, M.D. “New technologies like RNA inhibitors, that alter the genetic drivers of a disease, have the potential to transform medicine, so we can better confront and even cure debilitating illnesses. We’re committed to advancing scientific principles that enable the efficient development and review of safe, effective and groundbreaking treatments that have the potential to change patients’ lives.”

RNA acts as a messenger within the body’s cells, carrying instructions from DNA for controlling the synthesis of proteins. RNA interference is a process that occurs naturally within our cells to block how certain genes are expressed. Since its discovery in 1998, scientists have used RNA interference as a tool to investigate gene function and its involvement in health and disease. Researchers at the National Institutes of Health, for example, have used robotic technologies to introduce siRNAs into human cells to individually turn off nearly 22,000 genes.

This new class of drugs, called siRNAs, work by silencing a portion of RNA involved in causing the disease. More specifically, Onpattro encases the siRNA into a lipid nanoparticle to deliver the drug directly into the liver, in an infusion treatment, to alter or halt the production of disease-causing proteins.

Affecting about 50,000 people worldwide, hATTR is a rare condition. It is characterized by the buildup of abnormal deposits of protein fibers called amyloid in the body’s organs and tissues, interfering with their normal functioning. These protein deposits most frequently occur in the peripheral nervous system, which can result in a loss of sensation, pain, or immobility in the arms, legs, hands and feet. Amyloid deposits can also affect the functioning of the heart, kidneys, eyes and gastrointestinal tract. Treatment options have generally focused on symptom management.

Onpattro is designed to interfere with RNA production of an abnormal form of the protein transthyretin (TTR). By preventing the production of TTR, the drug can help reduce the accumulation of amyloid deposits in peripheral nerves, improving symptoms and helping patients better manage the condition.

“There has been a long-standing need for a treatment for hereditary transthyretin-mediated amyloidosis polyneuropathy. This unique targeted therapy offers these patients an innovative treatment for their symptoms that directly affects the underlying basis of this disease,” said Billy Dunn, M.D., director of the Division of Neurology Products in the FDA’s Center for Drug Evaluation and Research.

The efficacy of Onpattro was shown in a clinical trial involving 225 patients, 148 of whom were randomly assigned to receive an Onpattro infusion once every three weeks for 18 months, and 77 of whom were randomly assigned to receive a placebo infusion at the same frequency. The patients who received Onpattro had better outcomes on measures of polyneuropathy including muscle strength, sensation (pain, temperature, numbness), reflexes and autonomic symptoms (blood pressure, heart rate, digestion) compared to those receiving the placebo infusions. Onpattro-treated patients also scored better on assessments of walking, nutritional status and the ability to perform activities of daily living.

The most common adverse reactions reported by patients treated with Onpattro are infusion-related reactions including flushing, back pain, nausea, abdominal pain, dyspnea (difficulty breathing) and headache. All patients who participated in the clinical trials received premedication with a corticosteroid, acetaminophen, and antihistamines (H1 and H2 blockers) to reduce the occurrence of infusion-related reactions. Patients may also experience vision problems including dry eyes, blurred vision and eye floaters (vitreous floaters). Onpattro leads to a decrease in serum vitamin A levels, so patients should take a daily Vitamin A supplement at the recommended daily allowance.

The FDA granted this application Fast TrackPriority Review and Breakthrough Therapy designations. Onpattro also received Orphan Drug designation, which provides incentives to assist and encourage the development of drugs for rare diseases.

Approval of Onpattro was granted to Alnylam Pharmaceuticals, Inc.

////////////// Onpattro, patisiran, fda 2018, Fast TrackPriority Review, Breakthrough Therapy,  Orphan Drug designation

FDA approves new vaginal ring for one year of birth control

$
0
0

FDA approves new vaginal ring for one year of birth control

The U.S. Food and Drug Administration today approved Annovera (segesterone acetate and ethinyl estradiol vaginal system), which is a combined hormonal contraceptive for women of reproductive age used to prevent pregnancy and is the first vaginal ring contraceptive that can be used for an entire year. Annovera is a reusable donut-shaped (ring), non-biodegradable, flexible vaginal system that is placed in the vagina for three weeks followed by one week out of the vagina, at which time women may experience a period (a withdrawal bleed). This schedule is repeated every four weeks for one year (thirteen 28-day menstrual cycles).

August 10, 2018

Release

The U.S. Food and Drug Administration today approved Annovera (segesterone acetate and ethinyl estradiol vaginal system), which is a combined hormonal contraceptive for women of reproductive age used to prevent pregnancy and is the first vaginal ring contraceptive that can be used for an entire year. Annovera is a reusable donut-shaped (ring), non-biodegradable, flexible vaginal system that is placed in the vagina for three weeks followed by one week out of the vagina, at which time women may experience a period (a withdrawal bleed). This schedule is repeated every four weeks for one year (thirteen 28-day menstrual cycles).

“The FDA is committed to supporting innovation in women’s health and today’s approval builds on available birth control options,” states Victor Crentsil, M.D., acting deputy director of the Office of Drug Evaluation III in FDA’s Center for Drug Evaluation and Research.

Annovera is washed and stored in a compact case for the seven days not in use. Annovera does not require refrigeration prior to dispensing and can withstand storage temperatures up to 30°C (86°F).

The efficacy and safety of Annovera were studied in three, open label clinical trials with healthy women ranging from 18 to 40 years of age. Based on the results, about two to four women out of 100 women may get pregnant during the first year they use Annovera.

All hormonal contraception carries serious risks. Annovera carries a boxed warning relating to cigarette smoking and serious cardiovascular events. Women over 35 who smoke should not use Annovera. Cigarette smoking increases the risk of serious cardiovascular events from combination hormonal contraceptive use.

Annovera is contraindicated and should not be used in women with:

  • A high risk of arterial or venous thrombotic diseases;
  • Current or history of breast cancer or other estrogen- or progestin-sensitive cancer;
  • Liver tumors, acute hepatitis, or severe (decompensated) cirrhosis;
  • Undiagnosed abnormal uterine bleeding;
  • Hypersensitivity to any of the components of Annovera; and
  • Use of Hepatitis C drug combinations containing ombitasvir/paritaprevir/ritonavir, with or without dasabuvir.

The most common side effects in women using Annovera are similar to those of other combined hormonal contraceptive products and include headache/migraine, nausea/vomiting, yeast infections, abdominal pain, dysmenorrhea (painful menstruation), breast tenderness, irregular bleeding, diarrhea and genital itching.

The FDA is requiring postmarketing studies to further evaluate the risks of venous thromboembolism, and the effects of CYP3A modulating drugs and tampon use on the pharmacokinetics of Annovera.

The FDA granted approval of Annovera to The Population Council, Inc.

/////////////fda 2018, Annovera, segesterone acetate, ethinyl estradiol, vaginal system

Patisiran

$
0
0

Patisiran

Sense strand:
GUAACCAAGAGUAUUCCAUdTdT
Anti-sense strand:
AUGGAAUACUCUUGGUUACdTdT
RNA, (A-U-G-G-A-A-Um-A-C-U-C-U-U-G-G-U-Um-A-C-dT-dT), complex with RNA (G-Um-A-A-Cm-Cm-A-A-G-A-G-Um-A-Um-Um-Cm-Cm-A-Um-dT-dT) (1:1),
ALN-18328, 6024128  , ALN-TTR02  , GENZ-438027  , SAR-438037  , 50FKX8CB2Y (UNII code)

 for RNA, (A-U-G-G-A-A-Um-A-C-U-C-U-U-G-G-U-Um-A-C-dT-dT), complex with RNA(G-Um-A-A-Cm-Cm-A-A-G-A-G-Um-A-Um-Um-Cm-Cm-A-Um-dT-dT) (1:1)

Nucleic Acid Sequence

Sequence Length: 42, 21, 2112 a 7 c 7 g 4 t 12 umultistranded (2); modified

CAS 1420706-45-1

Treatment of Amyloidosis,

SEE…..https://endpts.com/gung-ho-alnylam-lands-historic-fda-ok-on-patisiran-revving-up-the-first-global-rollout-for-an-rnai-breakthrough/

Lipid-nanoparticle-encapsulated double-stranded siRNA targeting a 3 untranslated region of mutant and wild-type transthyretin mRNA

Patisiran (trade name Onpattro®) is a medication for the treatment of polyneuropathy in people with hereditary transthyretin-mediated amyloidosis. It is the first small interfering RNA-based drug approved by the FDA. Through this mechanism, it is a gene silencing drug that interferes with the production of an abnormal form of transthyretin.

Chemical structure of Patisiran.

During its development, patisiran was granted orphan drug statusfast track designationpriority review and breakthrough therapy designation due to its novel mechanism and the rarity of the condition it is designed to treat.[1][2] It was approved by the FDA in August 2018 and is expected to cost around $345,000 to $450,000 per year.[3]

Patisiran was granted orphan drug designation in the U.S. and Japan for the treatment of familial amyloid polyneuropathy. Fast track designation was also granted in the U.S. for this indication. In the E.U., orphan drug designation was assigned to the compound for the treatment of transthyretin-mediated amyloidosis (initially for the treatment of familial amyloid polyneuropathy)

Hereditary transthyretin-mediated amyloidosis is a fatal rare disease that is estimated to affect 50,000 people worldwide. Patisiran is the first drug approved by the FDA to treat this condition.[4]

Patisiran is a second-generation siRNA therapy targeting mutant transthyretin (TTR) developed by Alnylam for the treatment of familial amyloid polyneuropathy. The product is delivered by means of Arbutus Biopharma’s (formerly Tekmira Pharmaceuticals) lipid nanoparticle technology

“A lot of peo­ple think it’s win­ter out there for RNAi. But I think it’s spring­time.” — Al­ny­lam CEO John Maraganore, NYT, Feb­ru­ary 7, 2011.

Patisiran — designed to silence messenger RNA and block the production of TTR protein before it is made — is number 6 on Clarivate’s list of blockbusters set to launch this year, with a 2022 sales forecast of $1.22 billion. Some of the peak sales estimates range significantly higher as analysts crunch the numbers on a disease that afflicts only about 30,000 people worldwide.

PATENT

WO 2016033326

https://patents.google.com/patent/WO2016033326A2

Transthyretin (TTR) is a tetrameric protein produced primarily in the liver.

Mutations in the TTR gene destabilize the protein tetramer, leading to misfolding of monomers and aggregation into TTR amyloid fibrils (ATTR). Tissue deposition results in systemic ATTR amyloidosis (Coutinho et al, Forty years of experience with type I amyloid neuropathy. Review of 483 cases. In: Glenner et al, Amyloid and Amyloidosis, Amsterdam: Excerpta Media, 1980 pg. 88-93; Hou et al., Transthyretin and familial amyloidotic polyneuropathy. Recent progress in understanding the molecular mechanism of

neurodegeneration. FEBS J 2007, 274: 1637-1650; Westermark et al, Fibril in senile systemic amyloidosis is derived from normal transthyretin. Proc Natl Acad Sci USA 1990, 87: 2843-2845). Over 100 reported TTR mutations exhibit a spectrum of disease symptoms.

[0004] TTR amyloidosis manifests in various forms. When the peripheral nervous system is affected more prominently, the disease is termed familial amyloidotic

polyneuropathy (FAP). When the heart is primarily involved but the nervous system is not, the disease is called familial amyloidotic cardiomyopathy (FAC). A third major type of TTR amyloidosis is called leptomeningeal/CNS (Central Nervous System) amyloidosis.

[0005] The most common mutations associated with familial amyloid polyneuropathy

(FAP) and ATTR-associated cardiomyopathy, respectively, are Val30Met (Coelho et al, Tafamidis for transthyretin familial amyloid polyneuropathy: a randomized, controlled trial. Neurology 2012, 79: 785-792) and Vall22Ile (Connors et al, Cardiac amyloidosis in African Americans: comparison of clinical and laboratory features of transthyretin VI 221 amyloidosis and immunoglobulin light chain amyloidosis. Am Heart J 2009, 158: 607-614). [0006] Current treatment options for FAP focus on stabilizing or decreasing the amount of circulating amyloidogenic protein. Orthotopic liver transplantation reduces mutant TTR levels (Holmgren et al, Biochemical effect of liver transplantation in two Swedish patients with familial amyloidotic polyneuropathy (FAP-met30). Clin Genet 1991, 40: 242-246), with improved survival reported in patients with early-stage FAP, although deposition of wild-type TTR may continue (Yazaki et al, Progressive wild-type transthyretin deposition after liver transplantation preferentially occurs into myocardium in FAP patients. Am J Transplant 2007, 7:235-242; Adams et al, Rapid progression of familial amyloid polyneuropathy: a multinational natural history study Neurology 2015 Aug 25; 85(8) 675-82; Yamashita et al, Long-term survival after liver transplantation in patients with familial amyloid polyneuropathy. Neurology 2012, 78: 637-643; Okamoto et al., Liver

transplantation for familial amyloidotic polyneuropathy: impact on Swedish patients’ survival. Liver Transpl 2009, 15: 1229-1235; Stangou et al, Progressive cardiac amyloidosis following liver transplantation for familial amyloid polyneuropathy: implications for amyloid fibrillogenesis. Transplantation 1998, 66:229-233; Fosby et al, Liver transplantation in the Nordic countries – An intention to treat and post-transplant analysis from The Nordic Liver Transplant Registry 1982-2013. Scand J Gastroenterol. 2015 Jun; 50(6):797-808.

Transplantation, in press).

[0007] Tafamidis and diflunisal stabilize circulating TTR tetramers, which can slow the rate of disease progression (Berk et al, Repurposing diflunisal for familial amyloid polyneuropathy: a randomized clinical trial. JAMA 2013, 310: 2658-2667; Coelho et al., 2012; Coelho et al, Long-term effects of tafamidis for the treatment of transthyretin familial amyloid polyneuropathy. J Neurol 2013, 260: 2802-2814; Lozeron et al, Effect on disability and safety of Tafamidis in late onset of Met30 transthyretin familial amyloid polyneuropathy. Eur J Neurol 2013, 20: 1539-1545). However, symptoms continue to worsen on treatment in a large proportion of patients, highlighting the need for new, disease-modifying treatment options for FAP.

[0008] Description of dsRNA targeting TTR can be found in, for example,

International patent application no. PCT/US2009/061381 (WO2010/048228) and

International patent application no. PCT/US2010/05531 1 (WO201 1/056883). Summary

[0009] Described herein are methods for reducing or arresting an increase in a

Neuropathy Impairment Score (NIS) or a modified NIS (mNIS+7) in a human subject by administering an effective amount of a transthyretin (TTR)-inhibiting composition, wherein the effective amount reduces a concentration of TTR protein in serum of the human subject to below 50 μg/ml or by at least 80%. Also described herein are methods for adjusting a dosage of a TTR- inhibiting composition for treatment of increasing NIS or Familial Amyloidotic Polyneuropathy (FAP) by administering the TTR- inhibiting composition to a subject having the increasing NIS or FAP, and determining a level of TTR protein in the subject having the increasing NIS or FAP. In some embodiments, the amount of the TTR- inhibiting composition subsequently administered to the subject is increased if the level of TTR protein is greater than 50 μg/ml, and the amount of the TTR- inhibiting composition subsequently administered to the subject is decreased if the level of TTR protein is below 50 μg/ml. Also described herein are formulated versions of a TTR inhibiting siRNA.

Image result for Alnylam

PATENT

WO 2016203402

PAPERS

Annals of Medicine (Abingdon, United Kingdom) (2015), 47(8), 625-638.

Pharmaceutical Research (2017), 34(7), 1339-1363

Annual Review of Pharmacology and Toxicology (2017), 57, 81-105

CLIP

Image result for Alnylam

Alnylam Announces First-Ever FDA Approval of an RNAi Therapeutic, ONPATTRO™ (patisiran) for the Treatment of the Polyneuropathy of Hereditary Transthyretin-Mediated Amyloidosis in Adults
Aug 10,2018

− First and Only FDA-approved Treatment Available in the United States for this Indication –

− ONPATTRO Shown to Improve Polyneuropathy Relative to Placebo, with Reversal of Neuropathy Impairment Compared to Baseline in Majority of Patients –

− Improvement in Specified Measures of Quality of Life and Disease Burden Demonstrated Across Diverse, Global Patient Population –

− Alnylam to Host Conference Call Today at 3:00 p.m. ET. −

CAMBRIDGE, Mass.–(BUSINESS WIRE)–Aug. 10, 2018– Alnylam Pharmaceuticals, Inc. (Nasdaq: ALNY), the leading RNAi therapeutics company, announced today that the United States Food and Drug Administration (FDA) approved ONPATTRO™ (patisiran) lipid complex injection, a first-of-its-kind RNA interference (RNAi) therapeutic, for the treatment of the polyneuropathy of hereditary transthyretin-mediated (hATTR) amyloidosis in adults. ONPATTRO is the first and onlyFDA-approved treatment for this indication. hATTR amyloidosis is a rare, inherited, rapidly progressive and life-threatening disease with a constellation of manifestations. In addition to polyneuropathy, hATTR amyloidosis can lead to other significant disabilities including decreased ambulation with the loss of the ability to walk unaided, a reduced quality of life, and a decline in cardiac functioning. In the largest controlled study of hATTR amyloidosis, ONPATTRO was shown to improve polyneuropathy – with reversal of neuropathy impairment in a majority of patients – and to improve a composite quality of life measure, reduce autonomic symptoms, and improve activities of daily living.

Image result for Alnylam

This press release features multimedia. View the full release here:https://www.businesswire.com/news/home/20180810005398/en/

ONPATTRO™ (patisiran) packaging and product vial (Photo: Business Wire)ONPATTRO™ (patisiran) packaging and product vial (Photo: Business Wire)

“Alnylam was founded on the vision of harnessing the potential of RNAi therapeutics to treat human disease, and this approval heralds the arrival of an entirely new class of medicines. We believe today draws us ever-closer to achieving our Alnylam 2020 goals of becoming a fully integrated, multi-product biopharmaceutical company with a sustainable pipeline,” said John Maraganore, Ph.D., Chief Executive Officer of Alnylam. “With the potential for the sequential launches of several new medicines in the coming years, we believe we have the opportunity to meaningfully impact the lives of people around the world in need of new approaches to address serious diseases with significant unmet medical needs.”

“Today’s historic approval marks the arrival of a first-of-its kind treatment option for a rare and devastating condition with limited treatment options,” said Akshay Vaishnaw, M.D., Ph.D., President of R&D at Alnylam. “We extend our deepest gratitude to the patients who participated in the ONPATTRO clinical trials and their families and caregivers who supported them. We are also grateful for the tireless efforts of the investigators and study staff, without whom this important milestone would not have been possible. We also look forward to working with the FDA to potentially expand the ONPATTRO indication in the future.”

The FDA approval of ONPATTRO was based on positive results from the randomized, double-blind, placebo-controlled, global Phase 3 APOLLO study, the largest-ever study in hATTR amyloidosis patients with polyneuropathy. Results from the APOLLO study were published in the July 5, 2018, issue of The New England Journal of Medicine.

In APOLLO, the safety and efficacy of ONPATTRO were evaluated in a diverse, global population of hATTR amyloidosis patients in 19 countries, with a total of 39 TTR mutations. Patients were randomized in a 2:1 ratio to receive intravenous ONPATTRO (0.3 mg per kg of body weight) or placebo once every 3 weeks for 18 months. The study showed that ONPATTRO improved measures of polyneuropathy, quality of life, activities of daily living, ambulation, nutritional status and autonomic symptoms relative to placebo in adult patients with hATTR amyloidosis with polyneuropathy. The primary endpoint of the APOLLO study was the modified Neuropathy Impairment Score +7 (mNIS+7), which assesses motor strength, reflexes, sensation, nerve conduction and postural blood pressure.

  • Patients treated with ONPATTRO had a mean 6.0-point decrease (improvement) in mNIS+7 score from baseline compared to a mean 28.0-point increase (worsening) for patients in the placebo group, resulting in a mean 34.0-point difference relative to placebo, after 18 months of treatment.
  • While nearly all ONPATTRO-treated patients experienced a treatment benefit relative to placebo, 56 percent of ONPATTRO-treated patients at 18 months of treatment experienced reversal of neuropathy impairment (as assessed by mNIS+7 score) relative to their own baseline, compared to four percent of patients who received placebo.
  • Patients treated with ONPATTRO had a mean 6.7-point decrease (improvement) in Norfolk Quality of Life Diabetic Neuropathy (QoL-DN) score from baseline compared to a mean 14.4-point increase (worsening) for patients in the placebo group, resulting in a mean 21.1-point difference relative to placebo, after 18 months of treatment.
  • As measured by Norfolk QoL-DN, 51 percent of patients treated with ONPATTRO experienced improvement in quality of life at 18 months relative to their own baseline, compared to 10 percent of the placebo-treated patients.
  • Over 18 months of treatment, patients treated with ONPATTRO experienced significant benefit vs. placebo for all other secondary efficacy endpoints, including measures of activities of daily living, walking ability, nutritional status, and autonomic symptoms.
  • The most common adverse events that occurred more frequently with ONPATTRO than with placebo were upper respiratory tract infections and infusion-related reactions. To reduce the risk of infusion-related reactions, patients received premedications prior to infusion.

“FDA approval of ONPATTRO represents an entirely new approach to treating patients with polyneuropathy in hATTR amyloidosis and shows promise as a new era in patient care,” said John Berk, M.D., Associate Professor of Medicine at Boston University School of Medicine and assistant director of the Amyloidosis Center at Boston University School of Medicine. “Given the strength of the APOLLO data, including data showing the possibility of halting or improving disease progression in many patients, ONPATTRO holds tremendous promise for people living with this disease.”

“For years I have witnessed the tragic impact of hATTR amyloidosis on generations of families. Today, we celebrate the FDA approval of ONPATTRO,” said Muriel Finkel, President of Amyloidosis Support Groups. “It’s extremely gratifying to see promising science translate into a treatment option that will allow patients to potentially experience an improvement in their disease and an improvement in their overall quality of life.”

“Today’s approval is significant in so many respects. It means the hATTR amyloidosis community of patients, families, caregivers and healthcare professionals in the United States now has a treatment option that offers renewed hope,” said Isabelle Lousada, Founder and Chief Executive Officer of the Amyloidosis Research Consortium. “With an FDA-approved treatment now available, I am more optimistic than ever that we can increase awareness of this rare disease and encourage more people to get tested and receive the proper diagnosis.”

ONPATTRO is expected to be available for shipment to healthcare providers in the U.S. within 48 hours.

Alnylam is committed to helping people access the medicines they are prescribed and will be offering comprehensive support services for people prescribed ONPATTRO through Alnylam Assist™. Visit AlnylamAssist.com for more information or call 1-833-256-2748.

ONPATTRO was reviewed by the FDA under Priority Review and had previously been granted Breakthrough Therapy and Orphan Drug Designations. On July 27, patisiran received a positive opinion from the Committee for Medicinal Products for Human Use (CHMP) for the treatment of hereditary transthyretin-mediated amyloidosis in adults with stage 1 or stage 2 polyneuropathy under accelerated assessment by the European Medicines Agency. The recommended Summary of Product Characteristics (SmPC) for the European Union (EU) includes data on secondary and exploratory endpoints. Expected in September, the European Commission will review the CHMP recommendation to make a final decision on marketing authorization, applicable to all 28 EU member states, plus Iceland, Liechtenstein and Norway. Regulatory filings in other markets, including Japan, are planned beginning in mid-2018.

Visit ONPATTRO.com for more information,

About ONPATTRO™ (patisiran) lipid complex injection
ONPATTRO was approved by the U.S. Food and Drug Administration (FDA) for the treatment of the polyneuropathy of hereditary transthyretin-mediated (hATTR) amyloidosis in adults. ONPATTRO is the first and only RNA interference (RNAi) therapeutic approved by the FDA for this indication. ONPATTRO utilizes a novel approach to target and reduce production of the TTR protein in the liver via the RNAi pathway. Reducing the TTR protein leads to a reduction in the amyloid deposits that accumulate in tissues. ONPATTRO is administered through intravenous (IV) infusion once every 3 weeks following required premedication and the dose is based on actual body weight. Home infusion may be an option for some patients after an evaluation and recommendation by the treating physician, and may not be covered by all insurance plans. Regardless of the setting, ONPATTRO infusions should be performed by a healthcare professional. For more information about ONPATTRO, visit ONPATTRO.com.

About hATTR Amyloidosis
Hereditary transthyretin (TTR)-mediated amyloidosis (hATTR) is an inherited, progressively debilitating, and often fatal disease caused by mutations in the TTR gene. TTR protein is primarily produced in the liver and is normally a carrier of vitamin A. Mutations in the TTR gene cause abnormal amyloid proteins to accumulate and damage body organs and tissue, such as the peripheral nerves and heart, resulting in intractable peripheral sensory neuropathy, autonomic neuropathy, and/or cardiomyopathy, as well as other disease manifestations. hATTR amyloidosis represents a major unmet medical need with significant morbidity and mortality. The median survival is 4.7 years following diagnosis. Until now, people living with hATTR amyloidosis in the U.S. had no FDA-approved treatment options.

Alnylam Assist™
As part of Alnylam’s commitment to making therapies available to those who may benefit from them, Alnylam Assist will offer a wide range of services to guide patients through treatment with ONPATTRO, including financial assistance options for eligible patients, benefit verification and claims support, and ordering assistance and facilitation of delivery via specialty distributor or specialty pharmacy. Patients will have access to dedicated Case Managers who can provide personalized support throughout the treatment process and Patient Education Liaisons to help patients gain a better understanding of the disease. Visit AlnylamAssist.com for more information.

About RNAi
RNAi (RNA interference) is a natural cellular process of gene silencing that represents one of the most promising and rapidly advancing frontiers in biology and drug development today. Its discovery has been heralded as “a major scientific breakthrough that happens once every decade or so,” and was recognized with the award of the 2006 Nobel Prize for Physiology or Medicine. RNAi therapeutics are a new class of medicines that harness the natural biological process of RNAi. Small interfering RNA (siRNA), the molecules that mediate RNAi and comprise Alnylam’s RNAi therapeutic platform, function upstream of today’s medicines by potently silencing messenger RNA (mRNA) – the genetic precursors – that encode for disease-causing proteins, thus preventing them from being made. This is a revolutionary approach in developing medicines to improve the care of patients with genetic and other diseases.

About Alnylam
Alnylam (Nasdaq: ALNY) is leading the translation of RNA interference (RNAi) into a whole new class of innovative medicines with the potential to improve the lives of people afflicted with rare genetic, cardio-metabolic, and hepatic infectious diseases. Based on Nobel Prize-winning science, RNAi therapeutics represent a powerful, clinically validated approach for the treatment of a wide range of severe and debilitating diseases. Founded in 2002, Alnylam is delivering on a bold vision to turn scientific possibility into reality, with a robust discovery platform. ONPATTRO, available in the U.S. for the treatment of the polyneuropathy of hereditary transthyretin-mediated (hATTR) amyloidosis in adults, is Alnylam’s first U.S. FDA-approved RNAi therapeutic. Alnylam has a deep pipeline of investigational medicines, including three product candidates that are in late-stage development. Looking forward, Alnylam will continue to execute on its “Alnylam 2020” strategy of building a multi-product, commercial-stage biopharmaceutical company with a sustainable pipeline of RNAi-based medicines to address the needs of patients who have limited or inadequate treatment options. Alnylam employs over 800 people worldwide and is headquartered in Cambridge, MA. For more information about our people, science and pipeline, please visit www.alnylam.com and engage with us on Twitter at @Alnylam or on LinkedIn.

Image result for patisiran

FDA approves first-of-its kind targeted RNA-based therapy to treat a rare disease

First treatment for the polyneuropathy of hereditary transthyretin-mediated amyloidosis in adult patients

The U.S. Food and Drug Administration today approved Onpattro (patisiran) infusion for the treatment of peripheral nerve disease (polyneuropathy) caused by hereditary transthyretin-mediated amyloidosis (hATTR) in adult patients. This is the first FDA-approved treatment for patients with polyneuropathy caused by hATTR, a rare, debilitating and often fatal genetic disease characterized by the buildup of abnormal amyloid protein in peripheral nerves, the heart and other organs. It is also the first FDA approval of a new class of drugs called small interfering ribonucleic acid (siRNA) treatment

Continue reading…

https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/UCM616518.htm?utm_campaign=08102018_PR_FDA%20approves%20new%20drug%20for%20rare%20disease%2C%20hATTR&utm_medium=email&utm_source=Eloqua

August 10, 2018

Release

The U.S. Food and Drug Administration today approved Onpattro (patisiran) infusion for the treatment of peripheral nerve disease (polyneuropathy) caused by hereditary transthyretin-mediated amyloidosis (hATTR) in adult patients. This is the first FDA-approved treatment for patients with polyneuropathy caused by hATTR, a rare, debilitating and often fatal genetic disease characterized by the buildup of abnormal amyloid protein in peripheral nerves, the heart and other organs. It is also the first FDA approval of a new class of drugs called small interfering ribonucleic acid (siRNA) treatment.

“This approval is part of a broader wave of advances that allow us to treat disease by actually targeting the root cause, enabling us to arrest or reverse a condition, rather than only being able to slow its progression or treat its symptoms. In this case, the effects of the disease cause a degeneration of the nerves, which can manifest in pain, weakness and loss of mobility,” said FDA Commissioner Scott Gottlieb, M.D. “New technologies like RNA inhibitors, that alter the genetic drivers of a disease, have the potential to transform medicine, so we can better confront and even cure debilitating illnesses. We’re committed to advancing scientific principles that enable the efficient development and review of safe, effective and groundbreaking treatments that have the potential to change patients’ lives.”

RNA acts as a messenger within the body’s cells, carrying instructions from DNA for controlling the synthesis of proteins. RNA interference is a process that occurs naturally within our cells to block how certain genes are expressed. Since its discovery in 1998, scientists have used RNA interference as a tool to investigate gene function and its involvement in health and disease. Researchers at the National Institutes of Health, for example, have used robotic technologies to introduce siRNAs into human cells to individually turn off nearly 22,000 genes.

This new class of drugs, called siRNAs, work by silencing a portion of RNA involved in causing the disease. More specifically, Onpattro encases the siRNA into a lipid nanoparticle to deliver the drug directly into the liver, in an infusion treatment, to alter or halt the production of disease-causing proteins.

Affecting about 50,000 people worldwide, hATTR is a rare condition. It is characterized by the buildup of abnormal deposits of protein fibers called amyloid in the body’s organs and tissues, interfering with their normal functioning. These protein deposits most frequently occur in the peripheral nervous system, which can result in a loss of sensation, pain, or immobility in the arms, legs, hands and feet. Amyloid deposits can also affect the functioning of the heart, kidneys, eyes and gastrointestinal tract. Treatment options have generally focused on symptom management.

Onpattro is designed to interfere with RNA production of an abnormal form of the protein transthyretin (TTR). By preventing the production of TTR, the drug can help reduce the accumulation of amyloid deposits in peripheral nerves, improving symptoms and helping patients better manage the condition.

“There has been a long-standing need for a treatment for hereditary transthyretin-mediated amyloidosis polyneuropathy. This unique targeted therapy offers these patients an innovative treatment for their symptoms that directly affects the underlying basis of this disease,” said Billy Dunn, M.D., director of the Division of Neurology Products in the FDA’s Center for Drug Evaluation and Research.

The efficacy of Onpattro was shown in a clinical trial involving 225 patients, 148 of whom were randomly assigned to receive an Onpattro infusion once every three weeks for 18 months, and 77 of whom were randomly assigned to receive a placebo infusion at the same frequency. The patients who received Onpattro had better outcomes on measures of polyneuropathy including muscle strength, sensation (pain, temperature, numbness), reflexes and autonomic symptoms (blood pressure, heart rate, digestion) compared to those receiving the placebo infusions. Onpattro-treated patients also scored better on assessments of walking, nutritional status and the ability to perform activities of daily living.

The most common adverse reactions reported by patients treated with Onpattro are infusion-related reactions including flushing, back pain, nausea, abdominal pain, dyspnea (difficulty breathing) and headache. All patients who participated in the clinical trials received premedication with a corticosteroid, acetaminophen, and antihistamines (H1 and H2 blockers) to reduce the occurrence of infusion-related reactions. Patients may also experience vision problems including dry eyes, blurred vision and eye floaters (vitreous floaters). Onpattro leads to a decrease in serum vitamin A levels, so patients should take a daily Vitamin A supplement at the recommended daily allowance.

The FDA granted this application Fast TrackPriority Review and Breakthrough Therapy designations. Onpattro also received Orphan Drug designation, which provides incentives to assist and encourage the development of drugs for rare diseases.

Approval of Onpattro was granted to Alnylam Pharmaceuticals, Inc.

References

  1. Jump up^ “FDA approves first-of-its kind targeted RNA-based therapy to treat a rare disease” (Press release). U.S. Food and Drug Administration. 10 August 2018. Retrieved 11 August 2018.
  2. Jump up^ Brooks, Megan (10 August 2018). “FDA OKs Patisiran (Onpattro) for Polyneuropathy in hAATR”Medscape. WebMD. Retrieved 10 August 2018.
  3. Jump up^ Lipschultz, Bailey; Cortez, Michelle (10 August 2018). “Rare-Disease Treatment From Alnylam to Cost $450,000 a Year”Bloomberg. Retrieved 11 August 2018.
  4. Jump up^ Loftus, Peter (10 August 2018). “New Kind of Drug, Silencing Genes, Gets FDA Approval”Wall Street Journal. Retrieved 10 August 2018.

////////////// Onpattro, patisiran, fda 2018, Fast TrackPriority Review, Breakthrough Therapy,  Orphan Drug designation, Alnylam Pharmaceuticals, ALN-18328,  6024128  , ALN-TTR02  , GENZ-438027  , SAR-438037  , 50FKX8CB2Y

CC1=CN(C2OC(COP(=O)(O)OC3C(O)C(OC3COP(=O)(O)OC4C(O)C(OC4COP(=O)(O)OC5C(O)C(OC5COP(=O)(O)OC6C(O)C(OC6COP(=O)(O)OC7C(O)C(OC7COP(=O)(O)OC8C(O)C(OC8COP(=O)(O)OC9C(O)C(OC9COP(=O)(O)OC%10C(O)C(OC%10COP(=O)(O)OC%11C(O)C(OC%11COP(=O)(O)OC%12C(O)C(OC%12COP(=O)(O)OC%13C(O)C(OC%13COP(=O)(O)OC%14C(O)C(OC%14COP(=O)(O)OC%15C(O)C(OC%15COP(=O)(O)OC%16C(O)C(OC%16COP(=O)(O)OC%17C(O)C(OC%17CO)n%18cnc%19C(=O)NC(=Nc%18%19)N)N%20C=C(C)C(=O)NC%20=O)n%21cnc%22c(N)ncnc%21%22)n%23cnc%24c(N)ncnc%23%24)N%25C=C(C)C(=NC%25=O)N)N%26C=C(C)C(=NC%26=O)N)n%27cnc%28c(N)ncnc%27%28)n%29cnc%30c(N)ncnc%29%30)n%31cnc%32C(=O)NC(=Nc%31%32)N)n%33cnc%34c(N)ncnc%33%34)n%35cnc%36C(=O)NC(=Nc%35%36)N)N%37C=C(C)C(=O)NC%37=O)n%38cnc%39c(N)ncnc%38%39)N%40C=C(C)C(=O)NC%40=O)N%41C=C(C)C(=O)NC%41=O)C(OP(=O)(O)OCC%42OC(C(O)C%42OP(=O)(O)OCC%43OC(C(O)C%43OP(=O)(O)OCC%44OC(C(O)C%44OP(=O)(O)OCC%45OC(CC%45OP(=O)(O)OCC%46OC(CC%46O)N%47C=C(C)C(=O)NC%47=O)N%48C=C(C)C(=O)NC%48=O)N%49C=C(C)C(=O)NC%49=O)n%50cnc%51c(N)ncnc%50%51)N%52C=C(C)C(=NC%52=O)N)C2O)C(=O)N=C1N.CC%53=CN(C%54CC(O)C(COP(=O)(O)OC%55CC(OC%55COP(=O)(O)OC%56C(O)C(OC%56COP(=O)(O)OC%57C(O)C(OC%57COP(=O)(O)OC%58C(O)C(OC%58COP(=O)(O)OC%59C(O)C(OC%59COP(=O)(O)OC%60C(O)C(OC%60COP(=O)(O)OC%61C(O)C(OC%61COP(=O)(O)OC%62C(O)C(OC%62COP(=O)(O)OC%63C(O)C(OC%63COP(=O)(O)OC%64C(O)C(OC%64COP(=O)(O)OC%65C(O)C(OC%65COP(=O)(O)OC%66C(O)C(OC%66COP(=O)(O)OC%67C(O)C(OC%67COP(=O)(O)OC%68C(O)C(OC%68COP(=O)(O)OC%69C(O)C(OC%69COP(=O)(O)OC%70C(O)C(OC%70COP(=O)(O)OC%71C(O)C(OC%71COP(=O)(O)OC%72C(O)C(OC%72COP(=O)(O)OC%73C(O)C(OC%73COP(=O)(O)OC%74C(O)C(OC%74CO)n%75cnc%76c(N)ncnc%75%76)N%77C=CC(=O)NC%77=O)n%78cnc%79C(=O)NC(=Nc%78%79)N)n%80cnc%81C(=O)NC(=Nc%80%81)N)n%82cnc%83c(N)ncnc%82%83)n%84cnc%85c(N)ncnc%84%85)N%86C=C(C)C(=O)NC%86=O)n%87cnc%88c(N)ncnc%87%88)N%89C=CC(=NC%89=O)N)N%90C=CC(=O)NC%90=O)N%91C=CC(=NC%91=O)N)N%92C=CC(=O)NC%92=O)N%93C=CC(=O)NC%93=O)n%94cnc%95C(=O)NC(=Nc%94%95)N)n%96cnc%97C(=O)NC(=Nc%96%97)N)N%98C=CC(=O)NC%98=O)N%99C=C(C)C(=O)NC%99=O)n1cnc2c(N)ncnc12)N3C=CC(=NC3=O)N)N4C=C(C)C(=O)NC4=O)O%54)C(=O)NC%53=O

Evocalcet, エボカルセト , Эвокальцет , إيفوكالسيت , 依伏卡塞 ,

$
0
0

Evocalcet.pngImage result for EvocalcetEvocalcet.svg

Evocalcet

C24H26N2O2,  374.484 

Evocalcet; UNII-E58MLH082P; E58MLH082P; 870964-67-3; Evocalcet [INN]; Orkedia (TN)
エボカルセト

Эвокальцет [Russian] [INN]

إيفوكالسيت [Arabic] [INN]
依伏卡塞 [Chinese] [INN]
2-[4-[(3S)-3-[[(1R)-1-naphthalen-1-ylethyl]amino]pyrrolidin-1-yl]phenyl]acetic acid
KHK-7580
MT-4580
UNII:E58MLH082P
{4-[(3S)-3-{[(1R)-1-(1-Naphthyl)ethyl]amino}-1-pyrrolidinyl]phenyl}acetic acid
10098
870964-67-3 [RN]
Benzeneacetic acid, 4-[(3S)-3-[[(1R)-1-(1-naphthalenyl)ethyl]amino]-1-pyrrolidinyl]-
E58MLH082P
KHK-7580 / KHK7580 / MT-4580

Image result for Evocalcet

エボカルセト
Evocalcet

C24H26N2O2 : 374.48
[870964-67-3]

WP_000286

KHK 7580 …..example

3.008
Figure US20140080770A1-20140320-C00373
Figure US20140080770A1-20140320-C00374
Figure US20140080770A1-20140320-C00375
2HCl MS · APCI: 375[M + H]+

Figure imgb0350

in EP1757582

4-(3S-(1R-(1-naphthyl)ethylamino)pyrrolidin-1- yl)phenylacetic acid

4-​[(3S)​-​3-​[[(1R)​-​1-​(1-​naphthalenyl)​ethyl]​amino]​-​1-​pyrrolidinyl]​-Benzeneacetic acid,

BASE ….870964-67-3

DI HCL SALT …….870856-31-8

MF C24 H26 N2 O2 BASE

MW 374.48 BASE

KHK-7580

KHK-7580; MT-4580

Mitsubishi Tanabe Pharma Corp… innovator

Kyowa Hakko Kirin Co Ltd.. licencee

4-(3S-(1R-(1-naphthyl)ethylamino)pyrrolidin-1-yl)phenylacetic acid,

Evocalcet (trade name Orkedia) is a drug for the treatment of hyperparathyroidism.[1] It acts as a calcium-sensing receptor agonist.[2]

In 2018, it was approved in Japan for treatment of secondary hyperparathyroidism in patients on dialysis.[3]

useful as calcium-sensitive receptor (CaSR) agonists for treating hyperparathyroidism.  a CaSR agonist, being developed by Kyowa Hakko Kirin, under license from Mitsubishi Tanabe, for treating secondary hyperparathyroidism (phase 2 clinical, as of March 2015).

WO2005115975,/EP1757582

http://www.google.co.in/patents/EP1757582A1?cl=en

Example no

3.008
Figure US20140080770A1-20140320-C00373
Figure US20140080770A1-20140320-C00374
Figure US20140080770A1-20140320-C00375
2HCl MS · APCI: 375[M + H]+

Figure imgb0350

WO 2015034031A1

http://worldwide.espacenet.com/publicationDetails/biblio?DB=worldwide.espacenet.com&II=0&ND=3&adjacent=true&locale=en_EP&FT=D&date=20150312&CC=WO&NR=2015034031A1&KC=A1

Mitsubishi Tanabe Pharma Corporation

The present invention provides a novel crystal form of an arylalkylamine
compound. Specifically, a novel crystal form of
4-(3S-(1R-(1-naphthyl)ethylamino)pyrrolidin-1- yl)phenylacetic acid has
excellent stability, and is therefore useful as an active ingredient for
a medicine. The present invention also provides an industrially
advantageous method for producing an arylalkylamine compound.

WP_000287

WO 2015034031A1

http://worldwide.espacenet.com/publicationDetails/biblio?DB=worldwide.espacenet.com&II=0&ND=3&adjacent=true&locale=en_EP&FT=D&date=20150312&CC=WO&NR=2015034031A1&KC=A1
Mitsubishi Tanabe Pharma Corporation

The present invention provides a novel crystal form of an arylalkylamine compound. Specifically, a novel crystal form of 4-(3S-(1R-(1-naphthyl)ethylamino)pyrrolidin-1- yl)phenylacetic acid has excellent stability, and is therefore useful as an active ingredient for a medicine. The present invention also provides an industrially advantageous method for producing an arylalkylamine compound.

 

PATENT

http://www.google.co.in/patents/US20140080770?cl=und

Reference Example 3.001

Figure US20140080770A1-20140320-C00042

(1) To a mixed solution containing 33.5 g of 3-hydroxypiperidine and 62.7 ml of triethylamine dissolved in 250 ml of methylene chloride was added dropwise a solution of 55.7 ml of benzyloxycarbonyl chloride in 150 ml of methylene chloride, and the mixture was stirred at room temperature for 16 hours. To the reaction mixture were added a saturated aqueous citric acid and chloroform, the mixture was stirred and the liquids were separated. The organic layer was dried, the solvent was evaporated, and the residue was purified by silica gel column chromatography (hexane:ethyl acetate=4:1→0:1) to obtain 75.5 g of benzyl 3-hydroxypiperidine-1-carboxylate.

MS•APCI (m/z): 236 [M+H]+

(2) 800 ml of a solution of 52.4 ml of oxalyl chloride in methylene chloride was cooled to −78° C., 53.2 ml of DMSO was added dropwise to the solution, and the mixture was stirred at −78° C. for 0.5 hour. A solution of 75.5 g of benzyl 3-hydroxypiperidine-1-carboxylate dissolved in 200 ml of methylene chloride was added dropwise to the mixture, and further 293 ml of triethylamine was added dropwise to the same, and the mixture was stirred for 16 hours while a temperature thereof was gradually raised to room temperature. To the reaction mixture were added a saturated aqueous sodium bicarbonate solution and chloroform, the mixture was stirred and the liquids were separated. The organic layer was dried and concentrated to obtain 83.7 g of 1-benzyloxycarbonyl-3-piperidone. MS•APCI (m/z): 234 [M+H]+
(3) To a solution of 83.7 g of 1-benzyloxycarbonyl-3-piperidone dissolved in 1.2 liters of methylene chloride was added 55.0 g of (R)-(+)-1-(1-naphthyl)ethylamine, and after the mixture was stirred at room temperature for 2 hours, 69 ml of acetic acid and 160 g of sodium triacetoxy borohydride were added to the mixture, and the mixture was stirred at room temperature for 15 hours. To the reaction mixture was added an aqueous sodium hydroxide to make the mixture basic, and then, chloroform was added to the mixture, the mixture was stirred and the liquids were separated. The organic layer was dried and concentrated, and the residue was purified by silica gel column chromatography (hexane:ethyl acetate=4:1→0:1) to obtain 98.7 g of benzyl 3-[(R)-1-(naphthalen-1-yl)ethylamino]piperidine-1-carboxylate. MS•APCI (m/z): 389 [M+H]+
(4) To a solution of 40.95 g of triphosgene dissolved in 800 ml of methylene chloride was added dropwise a mixed solution containing 80.6 g of benzyl 3-[(R)-1-(naphthalen-1-yl)ethylamino]piperidine-1-carboxylate and 86.6 ml of triethylamine dissolved in 200 ml of methylene chloride at 0° C., and the mixture was stirred at room temperature for 16 hours. To the reaction mixture was added water, the mixture was stirred and the liquids were separated. The organic layer was dried and concentrated, and the residue was washed with 200 ml of diethyl ether, and the crystal collected by filtration was recrystallized from chloroform and diethyl ether to obtain 48.9 g of benzyl (R)-3-[chlorocarbonyl-(R)-1-(naphthalen-1-yl)ethylamino]piperidine-1-carboxylate.

Further, the filtrate was purified by silica gel column chromatography (hexane:ethyl acetate=8:1→0:1) to obtain 5.82 g of benzyl (R)-3-[chlorocarbonyl-(R)-1-(naphthalen-1-yl)ethylamino]piperidine-1-carboxylate and 14.5 g of benzyl (S)-3-[chlorocarbonyl-(R)-1-(naphthalen-1-yl)ethylamino]piperidine-1-carboxylate.

(5) To a solution containing 54.6 g of benzyl (R)-3-[chlorocarbonyl-(R)-1-(naphthalen-1-yl)ethylamino]piperidine-1-carboxylate dissolved in 700 ml of tetrahydrofuran was added 350 ml of water, and the mixture was stirred under reflux for 15 hours. After tetrahydrofuran was evaporated, a saturated aqueous sodium bicarbonate solution and chloroform were added thereto, the mixture was stirred and the liquids were separated. The organic layer was dried and concentrated, and the residue was purified by silica gel column chromatography (hexane:ethyl acetate=4:1→0:1) to obtain 24.3 g of benzyl (R)-3-[(R)-1-(naphthalen-1-yl)ethylamino]piperidine-1-carboxylate. MS•APCI (m/z): 389 [M+H]+
(6) To a solution containing 24.2 g of benzyl (R)-3-[(R)-1-(naphthalen-1-yl)ethylamino]piperidine-1-carboxylate dissolved in 250 ml of methanol was added 2.5 g of palladium carbon (10% wet), and the mixture was shaked under hydrogen atmosphere at 3 atm at room temperature for 40 hours. Palladium carbon was removed, and the solvent was evaporated, the residue was washed with ethyl acetate-chloroform (10:1), and collected by filtration to obtain 15.3 g of (R)-3-[(R)-1-(naphthalen-1-yl)ethylamino]piperidine (the following Reference example Table, Reference example 3.001(a)). MS•APCI (m/z): 255 [M+H]+
(7) By using 14.5 g of benzyl (S)-3-[chlorocarbonyl-(R)-1-(naphthalen-1-yl)ethylamino]piperidine-1-carboxylate, the same treatment was carried out as in the above-mentioned (5) to obtain 4.74 g of benzyl (S)-3-[(R)-1-(naphthalen-1-yl)ethylamino]piperidine-1-carboxylate. MS•APCI (m/z): 389 [M+H]+

Moreover, by using 4.7 g of benzyl (S)-3-[(R)-1-(naphthalen-1-yl)ethylamino]piperidine-1-carboxylate, the same treatment was carried out as in the above-mentioned (6) to obtain 2.89 g of (S)-3-[(R)-1-(naphthalen-1-yl)ethylamino]piperidine. MS•APCI (m/z): 255 [M+H]+

(8) To a solution of 3.46 g of (S)-3-[(R)-1-(naphthalen-1-yl)ethylamino]piperidine dissolved in 15 ml of methanol was added dropwise 20 ml of a solution of 4M hydrochloric acid in ethyl acetate, and the mixture was stirred. The reaction mixture was concentrated under reduced pressure, diethyl ether was added to the residue, washed and dried to obtain 3.33 g of (S)-3-[(R)-1-(naphthalen-1-yl)ethylamino]piperidine dihydrochloride

3.008
Figure US20140080770A1-20140320-C00373
Figure US20140080770A1-20140320-C00374
Figure US20140080770A1-20140320-C00375
2HCl MS · APCI: 375[M + H]+
TABLE A3
Figure US20140080770A1-20140320-C00350
Example No. R1—X—
Figure US20140080770A1-20140320-C00351
—Ar Salt Physical properties, etc.

CLIP

see all at   http://drugpatentsint.blogspot.in/2015/03/wo-2015034031.html

see all at   http://drugpatentsint.blogspot.in/2015/03/wo-2015034031.html

see all at   http://drugpatentsint.blogspot.in/2015/03/wo-2015034031.html

see all at   http://drugpatentsint.blogspot.in/2015/03/wo-2015034031.html
see all at   http://drugpatentsint.blogspot.in/2015/03/wo-2015034031.html

do not miss out on above click

 http://www.kyowa-kirin.com/research_and_development/pipeline/

KHK7580 -Secondary Hyperparathyroidism

JP

Company Mitsubishi Tanabe Pharma Corp.
Description Calcium receptor agonist
Molecular Target
Mechanism of Action Calcium-sensing receptor (CaSR) agonist
Therapeutic Modality Small molecule
Latest Stage of Development Phase II
Standard Indication Thyroid disease
Indication Details Treat hyperparathyroidism in patients receiving hemodialysis; Treat secondary hyperparathyroidism (SHPT)
Regulatory Designation
Partner

Kyowa Hakko Kirin Co. Ltd.

August 29, 2014

Kyowa Hakko Kirin Announces Commencement of Phase 2b Clinical Study of KHK7580 in Patients with Secondary Hyperparathyroidism in Japan

Tokyo, Japan, August 29, 2014 — Kyowa Hakko Kirin Co., Ltd. (Tokyo: 4151, President and CEO: Nobuo Hanai, “Kyowa Hakko Kirin”) today announced the initiation of a phase 2b clinical study evaluating KHK7580 for secondary hyperparathyroidism patients receiving hemodialysis in Japan.

This randomized, placebo-controlled, double-blind, parallel-group, multi-center study is designed to evaluate efficacy and safety in cohorts comprising KHK7580, its placebo and cinacalcet and initial dose of KHK7580 for secondary hyperparathyroidism patients receiving hemodialysis.

KHK7580 is a small molecular compound produced by Mitsubishi Tanabe Pharma Corporation (President & Representative Director, CEO: Masayuki Mitsuka, “Mitsubishi Tanabe Pharma”). Kyowa Hakko Kirin signed a license agreement of KHK7580 with Mitsubishi Tanabe Pharma for the rights to cooperative research, develop, market and manufacture the product in Japan and some part of Asia on March 2008.

The Kyowa Hakko Kirin Group is contributing to the health and prosperity of the world’s people by pursuing advances in life sciences and technology and creating new value.

Outline of this study

CLINICALTRIALS.GOV IDENTIFIER New window opensNCT02216656
TARGET POPULATION Secondary hyperparathyroidism patients receiving hemodialysis
TRIAL DESIGN Randomized, placebo-controlled, double-blind (included open arm of cinacalcet), parallel-group, multi-center study
ADMINISTRATION GROUP KHK7580, Placebo, cinacalcet
TARGET NUMBER OF SUBJECTS 150
PRIMARY OBJECTIVE Efficacy
TRIAL LOCATION Japan
TRIAL DURATION Jul. 2014 to Jun. 2015

Contact:

Kyowa Hakko Kirin
Media Contact:
+81-3-3282-1903
or
Investors:
+81-3-3282-0009

Update on march 2016

New comment waiting approval on New Drug Approvals

M.F. Balandrin commented on KHK 7580 structure cracked

KHK 7580 …..example 3.008 2HCl MS · APCI: 375[M + H]+ in …

The calcimimetic agent, KHK-7580, currently entering Phase III clinical trials, has now been given the INN (WHO) generic name, evocalcet. Its chemical structure has also now been published and it is, in fact, correct as proposed by Dr. Crasto (Well Done!!):

http://www.drugspider.com/drug/evocalcet

https://tripod.nih.gov/ginas/app/substance/f580b9fd

http://www.medkoo.com/products/6729

(Etymologically, in classical Latin, “evolutio” refers to “the unrolling of a scroll” and “evocare” refers to a “call out”…).

http://www.medkoo.com/products/6729

img

Name: Evocalcet
CAS#: 870964-67-3
Chemical Formula: C24H26N2O2
Exact Mass: 374.19943

Evocalcet is a calcium-sensing receptor agonist. The calcium-sensing receptor (CaSR) is a Class C G-protein coupled receptor which senses extracellular levels of calcium ion. The calcium-sensing receptor controls calcium homeostasis by regulating the release of parathyroid hormone (PTH). CaSR is expressed in all of the organs of the digestive system. CaSR plays a key role in gastrointestinal physiological function and in the occurrence of digestive disease. High dietary Ca2+ may stimulate CaSR activation and could both inhibit tumor development and increase the chemotherapeutic sensitivity of cancer cells in colon cancer tissues. (Last update: 12/15/2015).

Synonym: MT-4580; MT 4580; MT4580; KHK-7580; KHK7580; KHK 7580; Evocalcet

IUPAC/Chemical Name: 2-(4-((S)-3-(((R)-1-(naphthalen-1-yl)ethyl)amino)pyrrolidin-1-yl)phenyl)acetic acid

2

https://tripod.nih.gov/ginas/app/substance/f580b9fd

Structure of EVOCALCET

http://www.drugspider.com/drug/evocalcet

INN NAME
Evocalcet
LAB CODE(S)
MT-4580
KHK-7580
CHEMICAL NAME
{4-[(3S)-3-{[(1R)-1-(Naphthalen-1-yl)ethyl]amino}pyrrolidin-1-yl]phenyl}acetic acid
CHEMICAL STRUCTURE
MOLECULAR FORMULA
C24H26N2O2
SMILES
O=C(O)CC1=CC=C(N2C[C@@H](N[C@@H](C3=C4C=CC=CC4=CC=C3)C)CC2)C=C1
CAS REGISTRY NUMBER
870964-67-3
ORPHAN DRUG STATUS
No
ON FAST TRACK
No
NEW MOLECULAR ENTITY
Yes
ORIGINATOR
DEVELOPER(S)
CLASS
MECHANISM OF ACTION
WHO ATC CODE(S)
EPHMRA CODE(S)
CLINICAL TRIAL(S)
CONDITIONS INTERVENTIONS PHASES RECRUITMENT SPONSOR/COLLABORATORS
Secondary Hyperparathyroidism Drug: KHK7580 Phase 3 Recruiting Kyowa Hakko Kirin Company, Limited
Secondary Hyperparathyroidism Drug: KHK7580 Phase 3 Recruiting Kyowa Hakko Kirin Company, Limited
Secondary Hyperparathyroidism Drug: KHK7580|Drug: KRN1493 Phase 2|Phase 3 Recruiting Kyowa Hakko Kirin Company, Limited
Secondary Hyperparathyroidism Drug: Placebo|Drug: KHK7580 low dose|Drug: KHK7580 middle dose|Drug: KHK7580 high dose|Drug: KRN1493 Phase 2 Completed Kyowa Hakko Kirin Company, Limited
Hyperparathyroidism Drug: KHK7580 Phase 1|Phase 2 Completed Kyowa Hakko Kirin Company, Limited
Secondary Hyperparathyroidism Drug: KHK7580 Phase 1 Completed Kyowa Hakko Kirin Company, Limited
UPDATED ON
11 Oct 2015

CLIP

https://www.sciencedirect.com/science/article/pii/S0960894X18303676

Image result for Evocalcet

Scheme 1. Synthesis of key intermediates S4S5S9, and S10. Reagents and conditions: (a) Tf2O, i-Pr2NEt, CH2Cl2, −20 °C. Then, (R)-(+)-1-(1-naphthyl)ethylamine, −20 °C to rt (S1 57%); (b) triphosgene, Et3N, CH2Cl2, −20 °C to 5 °C. Then, i-Pr2NEt, tert-butanol, 70 °C; (c) separation via silica gel chromatography (S2 31%, S3 33% in 2 steps); (d) HClchloroform1,4-dioxane, rt (S4 > 94%, S5 > 94%); (e) (R)-(+)-1-(1-naphthyl)ethylamine, NaBH(OAc)3acetic acid, CH2Cl2, rt (S6 79%); (f) triphosgene, Et3N, CH2Cl2, 0 °C to rt; (g) separation via filtration and silica gel chromatography (S7 58%, S8 16% in 2 steps); (h) water, tetrahydrofuranreflux; (i) H2, Pd/C, methanol, rt (S9 50% in 2 steps); (j) HCl, ethyl acetate, methanol, rt (S10 31% in 3steps).

Scheme 2. Synthesis of 215 and evocalcet (16). Reagents and conditions: (a) aryl iodide or aryl bromide, palladium acetate, (rac)-BINAP, sodium tert-butoxide, toluene, 80 °C or reflux; (b) HCl, ethyl acetate or 1,4-dioxane, rt; (c) tert-butyl 4-fluorobenzoate, potassium carbonate, DMSO, 130 °C; (d) HCl, 1,4-dioxane, 45 °C; (e) 2-aminoethanol, EDC hydrochlorideHOBt, Et3N, DMF, rt; (f) 5-(4-bromophenyl)-2-(triphenylmethyl)–2H-tetrazole, Pd2(dba)3, (2-biphenyl)di-tert-butylphosphine, sodium tert-butoxide, toluene, rt; (g) HCl, water, 1,4-dioxane, rt; (h) tert-butyl 4-bromobenzoate, palladium acetate, (rac)-BINAP, sodium tert-butoxide, toluene, reflux; (i) trifluoroacetic acid, rt. Then, HCl, ethyl acetate or 1,4-dioxane, rt (2 23%, 3 21%, 4 44%, 5 34%, 620%, 7 55%, 8 29%, 9 21%, 10 19%, 11 40%, 13 26%, 14 69%, 15 69% in 2 steps); (j) 3-(trifluoromethoxy)phenylboronic acid, copper acetate, Et3N, CH2Cl2molecular sieve 4A, rt (S117%); (k) (COCl)2, DMSO, Et3N, CH2Cl2, −60 °C to rt (S12 was used in the next step without purification); (l) (R)-(+)-1-(1-naphthyl)ethylamine, NaBH(OAc)3acetic acid, CH2Cl2, rt. Then, separation of isomers; (m) HCl, ethyl acetate, rt (12 10% in 2 steps); (n) ethyl 4-bromophenylacetate, Pd2(dba)3, (2-biphenyl)di-tert-butylphosphine, sodium tert-butoxide, toluene, rt (S13 63%); (o) aqueous sodium hydroxide solution, ethanol, rt (evocalcet (16) 73%).

evocalcet as a white crystal. MS-APCI (m/z): 375 [M+H]+ .

1H NMR (400 MHz, DMSO-d6) δ 8.25-–8.37 (m, 1H), 7.88–7.97 (m, 1H), 7.79 (d, J = 7.9 Hz, 1H), 7.74 (d, J = 6.9 Hz, 1H), 7.39–7.57 (m, 3H), 7.01 (d, J = 8.6 Hz, 2H), 6.38 (d, J = 8.6 Hz, 2H), 4.74 (q, J = 6.4 Hz, 1H), 3.37 (s, 2H), 3.18–3.34 (m, 3H), 3.03–3.15 (m, 1H), 2.89–3.02 (m, 1H), 1.95–2.11 (m, 1H), 1.80–1.94 (m, 1H), 1.40 (d, J = 6.4 Hz, 3H).

Anal. Calcd for C24H26N2O2: C 76.98; H 7.00; N 7.48. Found: C 76.83; H 7.06; N 7.46.

HPLC 99.6% (25.4 min, Inertsil ODS-3V [5 μm, 4.6 × 250 mm], 0.05% TFA in H2O/0.05% TFA in CH3CN [95:5 to 0:100/60 min]).

References

  1. Jump up^ Kawata, Takehisa; Tokunaga, Shin; Murai, Miki; Masuda, Nami; Haruyama, Waka; Shoukei, Youji; Hisada, Yutaka; Yanagida, Tetsuya; Miyazaki, Hiroshi; Wada, Michihito; Akizawa, Tadao; Fukagawa, Masafumi (2018). “A novel calcimimetic agent, evocalcet (MT-4580/KHK7580), suppresses the parathyroid cell function with little effect on the gastrointestinal tract or CYP isozymes in vivo and in vitro”. PLOS ONE13 (4): e0195316. doi:10.1371/journal.pone.0195316PMID 29614098.
  2. Jump up^ Miyazaki, Hiroshi; Ikeda, Yousuke; Sakurai, Osamu; Miyake, Tsutomu; Tsubota, Rie; Okabe, Jyunko; Kuroda, Masataka; Hisada, Yutaka; Yanagida, Tetsuya; Yoneda, Hikaru; Tsukumo, Yukihito; Tokunaga, Shin; Kawata, Takehisa; Ohashi, Rikiya; Fukuda, Hajime; Kojima, Koki; Kannami, Ayako; Kifuji, Takayuki; Sato, Naoya; Idei, Akiko; Iguchi, Taku; Sakairi, Tetsuya; Moritani, Yasunori (2018). “Discovery of evocalcet, a next-generation calcium-sensing receptor agonist for the treatment of hyperparathyroidism”. Bioorganic & Medicinal Chemistry Letters28 (11): 2055–2060. doi:10.1016/j.bmcl.2018.04.055.
  3. Jump up^ “Kyowa Hakko Kirin Launches ORKEDIA® TABLETS (Evocalcet) for the Treatment of Secondary Hyperparathyroidism in Patients on Maintenance Dialysis in Japan” (Press release). Kyowa Hakko Kirin. May 22, 2018.
Evocalcet
Evocalcet.svg
Clinical data
Trade names Orkedia
Identifiers
CAS Number
PubChem CID
DrugBank
UNII
Chemical and physical data
Formula C24H26N2O2
Molar mass 374.48 g·mol−1

///////////////Evocalcet,  エボカルセト , Эвокальцет ,  إيفوكالسيت , 依伏卡塞 , JAPAN 2018, KHK-7580, MT-4580, UNII:E58MLH082P, ORKEDIA

SMILES Code: O=C(O)CC1=CC=C(N2C[C@@H](N[C@@H](C3=C4C=CC=CC4=CC=C3)C)CC2)C=C1

 C[C@H](c1cccc2c1cccc2)N[C@H]3CCN(C3)c4ccc(cc4)CC(=O)O

Tesirine

$
0
0

Tesirine.png2D chemical structure of 1595275-62-9

Tesirine

Molecular Formula: C75H101N9O23
Molecular Weight: 1496.673 g/mol

UNII-8DVQ435K46;

CAS 1595275-62-9

(11S,11aS)-4-((2S,5S)-37-(2,5-Dioxo-2,5-dihydro-1H-pyrrol-1-yl)-5-isopropyl-2-methyl-4,7,35-trioxo-10,13,16,19,22,25,28,31-octaoxa-3,6,34-triazaheptatriacontanamido)benzyl 11-hydroxy-7-methoxy-8-((5-(((S)-7-methoxy-2-methyl-5-oxo-5,11a-dihydro-1H-benzo[e]pyrrolo[1,2-a][1,4]diazepin-8-yl)oxy)pentyl)oxy)-2-methyl-5-oxo-11,11a-dihydro-1H-benzo[e]pyrrolo[1,2-a][1,4]diazepine-10(5H)-carboxylate 

SG3249, Tesirine

[4-[[(2S)-2-[[(2S)-2-[3-[2-[2-[2-[2-[2-[2-[2-[2-[3-(2,5-dioxopyrrol-1-yl)propanoylamino]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]propanoylamino]-3-methylbutanoyl]amino]propanoyl]amino]phenyl]methyl (6S,6aS)-3-[5-[[(6aS)-2-methoxy-8-methyl-11-oxo-6a,7-dihydropyrrolo[2,1-c][1,4]benzodiazepin-3-yl]oxy]pentoxy]-6-hydroxy-2-methoxy-8-methyl-11-oxo-6a,7-dihydro-6H-pyrrolo[2,1-c][1,4]benzodiazepine-5-carboxylate

PATENT

WO 2014057074

In 2012, tesirine (SG3249) was developed by Spirogen, as a drug linker combining a set of desired properties: fast and straightforward conjugation to antibody cysteines by maleimide Michael addition, good solubility in aqueous/DMSO (90/10) systems, and a traceless cleavable linker system delivering the highly potent pyrrolobenzodiazepine (PBD) DNA cross-linker SG3199

Image result for tesirine

Image result for tesirine

Image result for tesirine

CLIP

Image result for tesirine

CLIP

Scale-up Synthesis of Tesirine

 SpirogenQMB Innovation Centre42 New Road, E1 2AX London, United Kingdom
§ PharmaronNo. 6, Taihe Road, BDA, Beijing, 100176, People’s Republic of China
 Lonza AGRottenstrasse 6, CH – 3930 Visp, Switzerland
# Novasep Ltd1 Rue Démocrite, 72000 Le Mans, France
 Early Chemical Development, Pharmaceutical SciencesIMED Biotech UnitAstraZeneca, Macclesfield, United Kingdom
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.8b00205
Abstract Image

This work describes the enabling synthesis of tesirine, a pyrrolobenzodiazepine antibody–drug conjugate drug-linker. Over the course of four synthetic campaigns, the discovery route was developed and scaled up to provide a robust manufacturing process. Early intermediates were produced on a kilogram scale and at high purity, without chromatography. Midstage reactions were optimized to minimize impurity formation. Late stage material was produced and purified using a small number of key high-pressure chromatography steps, ultimately resulting in a 169 g batch after 34 steps. At the time of writing, tesirine is the drug-linker component of eight antibody–drug conjugates in multiple clinical trials, four of them pivotal

 CLIP

Design and Synthesis of Tesirine, a Clinical Antibody–Drug Conjugate Pyrrolobenzodiazepine Dimer Payload

QMB Innovation Centre, Spirogen, 42 New Road, E1 2AX London, U.K.
ACS Med. Chem. Lett.20167 (11), pp 983–987
DOI: 10.1021/acsmedchemlett.6b00062
Publication Date (Web): May 24, 2016
Copyright © 2016 American Chemical Society
This article is part of the Antibody-Drug Conjugates and Bioconjugates special issue.
Abstract Image

Pyrrolobenzodiazepine dimers are an emerging class of warhead in the field of antibody–drug conjugates (ADCs). Tesirine (SG3249) was designed to combine potent antitumor activity with desirable physicochemical properties such as favorable hydrophobicity and improved conjugation characteristics. One of the reactive imines was capped with a cathepsin B-cleavable valine-alanine linker. A robust synthetic route was developed to allow the production of tesirine on clinical scale, employing a flexible, convergent strategy. Tesirine was evaluated in vitro both in stochastic and engineered ADC constructs and was confirmed as a potent and versatile payload. The conjugation of tesirine to anti-DLL3 rovalpituzumab has resulted in rovalpituzumab-tesirine (Rova-T), currently under evaluation for the treatment of small cell lung cancer.

https://cdn-pubs.acs.org/doi/suppl/10.1021/acsmedchemlett.6b00062/suppl_file/ml6b00062_si_001.pdf

SG3249 (tesirine) (860 mg, 73% over 2 steps). LC/MS, method 2, 2.65 min (ES+) m/z (relative intensity) 1496.78 ([M+H] +. , 20). [] 24 D = +262 (c = 0.056, CHCl3).

1H NMR (400 MHz, DMSO-d6) δ 9.95 (s, 1H), 8.20 (d, J = 7.0 Hz, 1H), 8.03 (t, J = 5.6 Hz, 1H), 7.97 – 7.84 (m, 2H), 7.55 (d, J = 8.1 Hz, 2H), 7.32 (s, 1H), 7.18 (d, J = 8.0 Hz, 2H), 7.10 – 6.96 (m, 3H), 6.84 (s, 1H), 6.79 – 6.57 (m, 4H), 5.59 (d, J = 9.4 Hz, 1H), 5.16 (d, J = 12.7 Hz, 1H), 4.81 (d, J = 12.4 Hz, 1H), 4.38 (t, J = 7.1 Hz, 1H), 4.32 – 4.17 (m, 2H), 4.17 – 4.07 (m, 1H), 4.07 – 3.87 (m, 3H), 3.80 (d, J = 14.2 Hz, 6H), 3.74 – 3.62 (m, 1H), 3.59 (t, J = 7.2 Hz, 4H), 3.55 – 3.42 (m, 28H), 3.35 (d, J = 5.2 Hz, 2H), 3.21 – 3.11 (m, 2H), 3.11 – 2.98 (m, 2H), 2.98 – 2.83 (m, 1H), 2.49 – 2.28 (m, 5H), 2.03 – 1.88 (m, 1H), 1.87 – 1.65 (m, 10H), 1.64 – 1.47 (m, 2H), 1.29 (t, J = 5.9 Hz, 3H), 0.85 (dd, J = 17.1, 6.7 Hz, 6H).

13C NMR (126 MHz, DMSO-d6) δ 171.55, 171.29, 171.16, 170.78, 169.91, 164.80, 162.52, 155.03, 150.25, 139.27, 134.99, 128.84, 123.13, 122.67, 121.76, 119.28, 111.93, 110.93, 86.05, 70.21, 70.16, 70.02, 69.95, 69.46, 68.93, 68.79, 67.39, 57.94, 56.16, 54.03, 49.49, 38.97, 38.89, 36.39, 34.53, 34.40, 31.04, 28.69, 28.65, 22.72, 19.60, 18.55, 18.37, 13.88, 13.82. HRMS (ESI) m/z Calc. C75H101N9O23 1495.70831 found 1495.70444.

FT-IR (ATR, cm‐1 ) 3311, 2911, 2871, 1706, 1643, 1623, 1601, 1512, 1435, 1411, 1243, 1213, 1094, 1075, 946, 827, 747, 695, 664.

Patent ID Title Submitted Date Granted Date
US2015297746 PYRROLOBENZODIAZEPINE-ANTIBODY CONJUGATES
2013-10-11
2015-10-22
US2017267778 HUMANIZED ANTI-TN-MUC1 ANTIBODIES AND THEIR CONJUGATES
2015-04-15
Patent ID Title Submitted Date Granted Date
US2015283258 PYRROLOBENZODIAZEPINE – ANTI-PSMA ANTIBODY CONJUGATES
2013-10-11
2015-10-08
US2015283262 PYRROLOBENZODIAZEPINE-ANTIBODY CONJUGATES
2013-10-11
2015-10-08
US2015283263 PYRROLOBENZODIAZEPINE-ANTIBODY CONJUGATES
2013-10-11
2015-10-08
US2016106861 AXL ANTIBODY-DRUG CONJUGATE AND ITS USE FOR THE TREATMENT OF CANCER
2014-04-28
2016-04-21
US2014127239 PYRROLOBENZODIAZEPINES AND CONJUGATES THEREOF
2013-10-11
2014-05-08
Patent ID Title Submitted Date Granted Date
US2017320960 NOVEL ANTI-MFI2 ANTIBODIES AND METHODS OF USE
2015-09-04
US2016015828 NOVEL ANTIBODY CONJUGATES AND USES THEREOF
2014-02-21
2016-01-21
US2015265722 PYRROLOBENZODIAZEPINE-ANTI-CD22 ANTIBODY CONJUGATES
2013-10-11
2015-09-24
US2015273077 PYRROLOBENZODIAZEPINE-ANTI-HER2 ANTIBODY CONJUGATES
2013-10-11
2015-10-01
US2015273078 PYRROLOBENZODIAZEPINE-ANTI-PSMA ANTIBODY CONJUGATES
2013-10-11
2015-10-01

.//////////Tesirine, SG3249, SG 3249

CC1=CN2C(C1)C=NC3=CC(=C(C=C3C2=O)OC)OCCCCCOC4=C(C=C5C(=C4)N(C(C6CC(=CN6C5=O)C)O)C(=O)OCC7=CC=C(C=C7)NC(=O)C(C)NC(=O)C(C(C)C)NC(=O)CCOCCOCCOCCOCCOCCOCCOCCOCCNC(=O)CCN8C(=O)C=CC8=O)OC


TEBIPENEM PIVOXIL, テビペネムピボキシル , тебипенем пивоксил , تيبيبينام بيفوكسيل ,

$
0
0

Tebipenem pivoxil.png

ChemSpider 2D Image | tebipenem pivoxil | C22H31N3O6S2Image result for TEBIPENEM PIVOXIL

TEBIPENEM PIVOXIL

テビペネムピボキシル

тебипенем пивоксил [Russian] [INN]
تيبيبينام بيفوكسيل [Arabic] [INN]

2,2-dimethylpropanoyloxymethyl (4R,5S,6S)-3-[1-(4,5-dihydro-1,3-thiazol-2-yl)azetidin-3-yl]sulfanyl-6-[(1R)-1-hydroxyethyl]-4-methyl-7-oxo-1-azabicyclo[3.2.0]hept-2-ene-2-carboxylate

Molecular Formula: C22H31N3O6S2
Molecular Weight: 497.625 g/mol

Tebipenem pivoxil; 161715-24-8; Orapenem; UNII-95AK1A52I8; TBPM-PI; Tebipenem pivoxil(L-084)

(+)-hydroxymethyl(4R,5S,6S )-6-[(1R )-1-hydroxyethyl]-4-methyl-7-oxo-3-{[1-(2-thiazolin-2-yl)-3-azetidinyl]thio}-1-azabicyclo[3.2.0]hept-2-ene-2-carboxylate,2-pivalate

(4R,5R,6S)-3-[[1-(4,5-Dihydro-2-thiazolyl)-3-azetidinyl]thio]-6-[(1R)-1-hydroxyethyl]-4-methyl-7-oxo-1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid (2,2-dimethyl-1-oxopropoxy) methyl ester
[(2,2-Dimethylpropanoyl)oxy]methyl (4R,5S,6S)-3-{[1-(4,5-dihydro-1,3-thiazol-2-yl)-3-azetidinyl]sulfanyl}-6-[(1R)-1-hydroxyethyl]-4-methyl-7-oxo-1-azabicyclo[3.2.0]hept-2-ene-2-carboxylate
161715-24-8 [RN]
1-Azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid, 3-[[1-(4,5-dihydro-2-thiazolyl)-3-azetidinyl]thio]-6-[(1R)-1-hydroxyethyl]-4-methyl-7-oxo-, (2,2-dimethyl-1-oxopropoxy)methyl ester, (4R,5S,6S)-
7924
95AK1A52I8
UNII:95AK1A52I8
2,2-dimethylpropanoyloxymethyl (4R,5S,6S)-3-[1-(4,5-dihydrothiazol-2-yl)azetidin-3-yl]sulfanyl-6-[(1R)-1-hydroxyethyl]-4-methyl-7-oxo-1-azabicyclo[3.2.0]hept-2-ene-2-carboxylate
L 084
L084
L-084;Orapenem;L084;L 084
ME1211
MFCD17215369
L-084; ME-1211, SPR-994
TBPM-PI
MOA:Carbapenem antibiotic
Indication:Otitis Media; Otorhinolaryngological infection; Bacterial Pneumonia
Company:WYETH, Meiji Seika (Originator)
2009-04-22 japan approved

Tebipenem (brand name: Orapenem) is a broad-spectrum orally-administered antibiotic, from the carbapenem subgroup of β-lactam antibiotics. It was developed as a replacement drug to combat bacteria that had acquired antibiotic resistance to commonly used antibiotics.[1][2] Tebipenem is formulated as the ester tebipenem pivoxil due to the better absorption and improved bioavailability of this form.[3] It has performed well in clinical trials for ear infection and looks likely to be further developed in future.[4] It is only marketed in Japan.[5] Tebipenem is the first carbapenem whose prodrug form, the pivalyl ester, is orally available.[6]

Tebipenem pivoxil, an oral carbapenem prodrug, was launched in Japan in 2009 by Meiji Seika Pharma for the treatment of bacterial infection in children. The drug candidate was originally developed at Wyeth Pharmaceuticals (now Pfizer) and was subsequently licensed to Meiji Seika.

Tebipenem pivoxil was approved by Pharmaceuticals and Medical Devices Agency of Japan (PMDA) on Apr 22, 2009. It was developed and marketed as Orapenem® by Meiji Seika in Japan.

In 2017, the product was licensed to Spero Therapeutics by Meiji Seika Pharma for worldwide development and commercialization, except in Japan and certain Asian countries, where Meiji will retain rights.

In 2017, the FDA granted the drug qualified infectious disease product designation for complicated urinary tract infections (cUTI), diabetic foot infections (DFI) and community acquired bacterial pneumonia (CABP)

Tebipenem pivoxil is a broad-spectrum orally-administered antibiotic, from the carbapenem subgroup of β-lactam antibiotics. Carbapenems are a class of beta-lactam antibiotics, which act by inhibiting the synthesis of the peptidoglycan layer of bacterial cell walls. It is used to treat otorhinolaryngological infection, otitis media and bacterial pneumonia.

Orapenem® is available as granules for oral use, containing 100 mg Tebipenem pivoxil/g granules. According to the weight of children, 4 mg/kg, and twice a day after dinner.

Image result for TEBIPENEM PIVOXIL

CLIP

Image result for TEBIPENEM PIVOXIL

Clip

https://www.pharmacodia.com/yaodu/html/v1/chemicals/8c465432a5c7eaa0f3fc7c03401ce607.html

PATENTS

WO 2015070394

US5659043

WO9721712

CN 103613526

CN 103012406

CN 102775410

CN 106083858

EP 1580191

PATENT

https://patents.google.com/patent/CN104341421A/en

 Alternatively Bipei Nan ester (Tebipenempivoxil) (I), chemical name: (+) – (4R, 5S, 6S) -6 – [(lR) -l- hydroxyethyl] -4-methyl-7 – oxo-3 [[1- (2-thiazol-2-yl) -3-azetidinyl] thio] -1-azabicyclo [3. 2.0] hept-2-ene 2-carboxylic acid methyl -2- pivaloyl), of the formula:

[0003]

Figure CN104341421AD00031

[0004] developed by the American company Pfizer, for Bipei Nan ester fine granules developed by the Japanese company Meiji, in February 2009 was approved in Japan, and listed in April 2009. Alternatively Bipei Nan prodrug esters are for Bipei Nan, the lower the water after oral administration of the parent drug esterase for Bipei Nan, penicillin-binding protein binding to bacteria (the PBP), inhibition of bacterial cell wall synthesis, and is the only can oral carbapenem antibiotics.

[0005] Alternatively esters Bipei Nan structural characteristics, is a C3-side chain is thiazolyl substituted azetidinyl group, while in the C2 position by matching volts carboxylic ester forming a prodrug, increased oral absorbability; its oral absorption is better than in most β_ lactam antibiotics already on the market now. Bipei Nan for a broad spectrum antibiotic; especially for the PRSP in recent years, mainly due to the infection caused by children (penicillin-resistant Streptococcus pneumoniae), MRSP (erythromycin-resistant Streptococcus pneumoniae) and Haemophilusinfluenzae (Haemophilus influenzae) showed a very strong antibacterial effect. Alternatively Bipei Nan as prodrugs compared to for Bipei Nan horses volts for Bipei Nan ester having a better absorption kinetics, has good stability.

[0006] TakeshiIsoda like literature SynthesesandPharmacokineticStudiesof ProdrugEstersfortheDevelopmentofOralCarbapenem, L-084 (TheJournalof Antibiotics (2006) 59, 241 – 247; doi:. 10 · 1038 / ja 2006. 34) discloses a method for the synthesis of ester Bipei Nan : the Bipei Nan Alternatively, benzyltriethylammonium chloride, and chloromethyl pivalate was dissolved in N, N- dimethylformamide was added a solution of N, N- diisopropylethylamine, in the reaction was stirred for 4h at 45 ° C, the reaction was cooled to complete 5 ° C, was added ethyl acetate and water, the mixture was adjusted with aqueous citric acid I.OM PH = 4, the organic phase was discarded, the aqueous phase was adjusted with potassium bicarbonate to PH = 7. 6, the mixture extracted with ethyl acetate, the organic phase was washed with water and aqueous sodium chloride, dried over anhydrous magnesium sulfate, and concentrated under reduced pressure, the residue was subjected to silica gel column to give a yellow solid which was slurried with ethyl acetate to give colorless crystals. The column chromatography method requires not only consume a large amount of organic solvent together IJ, and a long time, so this method is not suitable for industrial production. In addition, the resulting large solid slurried with ethyl acetate, a solid dispersion is not easy, affect the uniformity of the product.

[0007] Patent US5534510, EP0632039, JP10-195076 discloses a method for synthesizing Bipei Nan ester is: dissolved after lyophilization for Bipei Nan aqueous sodium bicarbonate, lyophilized solid was dissolved in N, N- two dimethylformamide, adding a specific acid, methyl iodide, LH stirred at room temperature, ethyl acetate was added the reaction was completed, the organic phase was washed with saturated aqueous sodium bicarbonate, washed with brine, dried over anhydrous magnesium sulfate, the solvent was removed, the residue was subjected Alternatively Bipei Nan silica gel column to give the ester.The Mr. Fang Fati Bipei Nan for Bipei Nan into the sodium salt of pivalic acid with methyl iodide, prior to lyophilization to remove water required for the reaction, or affect the subsequent reaction with methyl iodide pivalate, lyophilized difficult when enlarged and the operation will take longer, the method further column chromatography operations shall therefore not suitable for industrial production.

[0008] Chinese patent CN102633801A disclosed for the preparation of esters Bipei Nan, including the following steps of: for Bipei Nan I. 37g, N, N- 11 ml of dimethylformamide, potassium carbonate 0 · 5g, tetrabutylphosphonium bromide 0 · 03g, reaction at 0 · 5h -KTC, Stuttgart dropwise at this temperature, methyl iodide 〇.88g acid, ethyl acetate was added the reaction was completed Ilml insolubles were filtered off, the filtrate was washed with water 22ml, water Ilml phase was extracted once with ethyl acetate, the combined ethyl acetate, washed with water and ethyl acetate are added 11 ml of water, adjusted to 3.5 with aqueous citric acid, phases were separated, the aqueous phase washed with ethyl acetate Ilml with the aqueous phase is added acetic acid 22ml ethyl ester, was adjusted to 7.5 with aqueous sodium bicarbonate, phase separation, washed with water and 22ml ethyl acetate, the ethyl acetate phase over anhydrous sodium sulfate was added 0. 5g, dried decolorizing charcoal, the filtrate was concentrated to a volume, stirring crystallization, cooling to 〇~5 ° C followed by stirring crystallization, filtration, and dried to give a white solid 〇.54g. Pivalate methyl iodide using the method of low-temperature reaction, post-treatment operation complicated, solvent volume, increasing both cost and environmental pollution, and methyl iodide pivalate unstable, expensive, not suitable for industrial production.

Figure CN104341421AD00041

 Example 1

[0029] Alternatively the Bipei Nan 18g, N, N- dimethylformamide 162mL, potassium 6. 54g, tetrabutylammonium bromide (λ45g, the reaction 60min, was added dropwise at this temperature at 25 ° C for chloromethyl pivalate 8. 93g, after the completion of the reaction, water was added and stirred for 10 minutes, 320ml, 160ml ethyl acetate was added to extract the aqueous phase extracted with ethyl acetate and once with 160ml ethyl acetate were combined, washed with water, phase separation, ethyl acetate washed with water 640ml paint ester, the ethyl acetate phase over anhydrous sodium sulfate was added 60g, decolorizing charcoal and dried, and the filtrate was concentrated, was added dropwise 25 ° C under stirring for crystallization 162mL isopropyl ether, filtered and dried to give a white solid 17. 64g. purity by HPLC 99.87%, the yield was 89.7%.

[0030] TBPN1HNMR data (CDCl3):. 5 989-5 978ppm (1H, d, 5 5, H-13.), 5 858-5 847ppm (1H, d, 5 5, Η-13.)… , 4 · 436-4. 390ppm (2H, m, H-22e, H-24e), 4. 232-4. 217ppm (2H, m, H-2, H-9), 4. 173-4. 146ppm (1H, m, H-21), 4. 039-3. 960ppm (4H, m, H-22a, H-24a, H-28), 3. 402-3. 372ppm (2H, t, 7. 5 , H-27), 3. 243-3. 230ppm (1H, m, H-3), 3. 199-3. 167ppm (1H, m, H-8), I. 348-1. 337ppm (3H, d, 5. 5, H-1), I. 24ppm (12H, m, H-17, H-18, H-19, H-10) ·

[0031] TBPN13C bandit R data (CDCl3):.. 176 958ppm (1C, C-15), 172 425ppm (1C, C-4), 164.286ppm (lC, C-25), 159.397ppm (lC, C- ll), 150.363ppm (lC, C-7), 124.570ppm (1C, C-6), 79.826 (1C, C-13), 65. 824ppm (1C, C-2), 60. 905ppm (1C, C -28), 59. 990ppm (1C, C-24), 59. 850ppm (lC, C-3), 58. 164ppm (1C, C-22), 56. 257ppm (lC, C-9), 43. 939ppm (1C, C-8), 38. 767ppm (1C, C-16), 36. 339ppm (1C, C-27), 33. 170 (1C, C-21), 26. 887ppm (3C, C- 17, C-18, C-19), 21.962ppm (lC, Cl), 16.805ppm (1C, C-10)

Patent

Publication numberPriority datePublication dateAssigneeTitle
CN102276611A *2011-05-182011-12-14深圳万乐药业有限公司Recrystallization method for purifying esters for Bipei Nan
CN102532139A *2010-12-072012-07-04重庆医药工业研究院有限责任公司Method for preparing tebipenem
CN102633801A *2012-03-132012-08-15深圳科兴生物工程有限公司Method for preparing tebipenem ester
F

Publication numberPriority datePublication dateAssigneeTitle

CN106543186A *2016-11-072017-03-29山东大学Monocrystal A of tebipenem pivoxil and preparation method thereof
KR101774812B1 *2016-05-272017-09-06(주)하이텍팜Preparation method for tebipenem pivoxil

PAPER

Chem. Pharm. Bull. 2006, 54, 1408-1411.

https://www.jstage.jst.go.jp/article/cpb/54/10/54_10_1408/_pdf

PAPER

J. Antibiot. 200659, 241-247.

PRESENTATION

Abe, T.; Hayashi, K.; Mihira, A.; Satoh, C.; Tamai, S.; Yamamoto, S.; Hikida, M.; Kumagai, T.; Kitamura, M.
L-084, a new oral carbapenem: Synthesis and structure-activity relationships of C2-substituted 1beta-methylcarbapenems
38th Intersci Conf Antimicrob Agents Chemother (ICAAC) (September 24-27, San Diego) 1998, Abst F-64

CLIP

EP 0632039; EP 0717042; JP 1996053453; US 5534510; US 5659043; US 5783703

Halogenation of allylamine (I) with either bromine or sulfuryl chloride produced the corresponding (halomethyl)aziridines (II). Subsequent treatment of (II) with n-butyllithium at -78 C yielded 1-azabicyclobutane (III). Opening of the bicyclic system of (III) with formic acid followed by acid hydrolysis provided 3-hydroxyazetidine (IV). This was condensed with 2-(methylsulfanyl)thiazoline (V) to give thiazolinylazetidine (VI). Alternatively, 3-hydroxyazetidine (IV) was condensed with 2-chloroethyl isothiocyanate (VII) to give the intermediate thiourea (VIII), which cyclized to the thiazoline (VI). Conversion of the hydroxyl group of (VI) into the thioacetate (IX) was carried out by either coupling with thioacetic acid under Mitsunobu conditions or by conversion to mesylate (X) followed by displacement with potassium thioacetate. The required thiol (XI) was then obtained from (IX) by basic hydrolysis of the thioacetate ester.

1-azabicyclobutane (III) was opened with thioacetic acid with concomitant N-acetylation yielding (XII). Further acid hydrolysis of (XII) gave 3-mercaptoazetidine (XIII). Condensation of (XIII) with either 2-(methylthio)thiazoline (V) or 2-chloroethyl isothiocyanate (VII) then produced thiazolinylazetidine (XI).

A further procedure consisted in the opening of 1-azabicyclobutane (III) with 2-mercaptothiazoline (XIV) to give (XV). Subsequent rearrangement of (XV) in the presence of methanesulfonic acid produced thiazolinyl azetidine (XI).

Condensation of (phosphoryloxy)carbapenem (XVI) with 3-mercapto-1-(1,3-thiazolin-2-yl)azetidine (XI) gave thioether (XVII). The p-nitrobenzyl ester group of (XVII) was then deprotected with Zn powder to afford the target carboxylic acid.

References

  1. Jump up^ El-Gamal, M. I.; Oh, C. H. (2010). “Current status of carbapenem antibiotics”. Current Topics in Medicinal Chemistry10 (18): 1882–1897. doi:10.2174/156802610793176639PMID 20615191.
  2. Jump up^ Fujimoto, K.; Takemoto, K.; Hatano, K.; Nakai, T.; Terashita, S.; Matsumoto, M.; Eriguchi, Y.; Eguchi, K.; Shimizudani, T.; Sato, K.; Kanazawa, K.; Sunagawa, M.; Ueda, Y. (2012). “Novel Carbapenem Antibiotics for Parenteral and Oral Applications: In Vitro and in Vivo Activities of 2-Aryl Carbapenems and Their Pharmacokinetics in Laboratory Animals”Antimicrobial Agents and Chemotherapy57 (2): 697–707. doi:10.1128/AAC.01051-12PMC 3553697Freely accessiblePMID 23147735.
  3. Jump up^ Kato, K.; Shirasaka, Y.; Kuraoka, E.; Kikuchi, A.; Iguchi, M.; Suzuki, H.; Shibasaki, S.; Kurosawa, T.; Tamai, I. (2010). “Intestinal Absorption Mechanism of Tebipenem Pivoxil, a Novel Oral Carbapenem: Involvement of Human OATP Family in Apical Membrane Transport”. Molecular Pharmaceutics7 (5): 1747–1756. doi:10.1021/mp100130bPMID 20735088.
  4. Jump up^ Sugita, R. (2013). “Good transfer of tebipenem into middle ear effusion conduces to the favorable clinical outcomes of tebipenem pivoxil in pediatric patients with acute otitis media”. Journal of Infection and Chemotherapy19 (3): 465–471. doi:10.1007/s10156-012-0513-5PMID 23393013.
  5. Jump up^ Rossi, S, ed. (7 August 2014). “Tebipenem Pivoxil”Martindale: The Complete Drug Reference. London, UK: Pharmaceutical Press. Retrieved 6 April 2015.
  6. Jump up^ Hazra, S; Xu, H; Blanchard, J (June 2014). “Tebipenem, a New Carbapenem Antibiotic is a Slow Substrate that Inhibits the β-Lactamase from Mycobacterium tuberculosis” (PDF). Biochemistry53 (22): 3671–3678. doi:10.1021/bi500339jPMC 4053071Freely accessiblePMID 24846409.
Patent ID Title Submitted Date Granted Date
US2014296257 High-Loading Water-Soluable Carrier-Linked Prodrugs
2012-08-10
2014-10-02
US2012156259 Biodegradable Polyethylene Glycol Based Water-Insoluble Hydrogels
2010-07-30
2012-06-21
US2009238867 Nanoparticulate Anidulafungin Compositions and Methods for Making the Same
2009-09-24
US2008103169 COMPOSITIONS COMPRISING ACID LABILE PROTON PUMP INHIBITING AGENTS, AT LEAST ONE OTHER PHARMACEUTICALLY ACTIVE AGENT AND METHODS OF USING SAME
2008-05-01
US8093294 Metallo-[beta]-lactamase inhibitors
2008-04-17
2012-01-10
Patent ID Title Submitted Date Granted Date
US2017304388 NEW INDICATION OF CARDIOVASCULAR DRUGS FOR PREPARATION OF CANCER INHIBITION PHARMACEUTICAL COMPOSITION
2015-10-23
US9278091 COMPOUNDS FOR THE TREATMENT OF CLOSTRIDIUM DIFFICILE ASSOCIATED DISEASE
2015-02-12
2015-07-30
US2016082123 Hydrogel-Linked Prodrugs Releasing Tagged Drugs
2014-04-16
2016-03-24
US2015087688 PRODRUGS OF HYDROXYL-COMPRISING DRUGS
2013-04-24
2015-03-26
US2014243254 Polymeric Hyperbranched Carrier-Linked Prodrugs
2012-08-10
2014-08-28
Patent ID Title Submitted Date Granted Date
US8987308 COMPOUNDS FOR THE TREATMENT OF CLOSTRIDIUM DIFFICILE-ASSOCIATED DISEASE
2012-11-30
2013-04-18
US9115079 NDM INHIBITOR
2012-07-26
2014-08-07
US8975416 ANTIBACTERIAL COMPOUNDS
2012-01-26
US9260375 METALLO-BETA-LACTAMASE INHIBITORS
2011-11-29
2012-03-22
US2016354315 DOSAGE FORMS AND USE THEREOF
2016-06-03
Tebipenem
Tebipenem pivoxil2DCSD.svg
Tebipenem pivoxil
Clinical data
Trade names Orapenem
Routes of
administration
Oral
Legal status
Legal status
  • Prescription-only in Japan; investigational elsewhere
Identifiers
CAS Number
KEGG
PDB ligand
Chemical and physical data
Formula C22H31N3O6S2
Molar mass 497.63 g/mol
3D model (JSmol)

/////////////////TEBIPENEM PIVOXIL, orapenem, テビペネムピボキシル  ,тебипенем пивоксил , تيبيبينام بيفوكسيل , L-084,  ME-1211, JAPAN 2009, SPR-994 , , Qualified infectious disease product designation

CC1C2C(C(=O)N2C(=C1SC3CN(C3)C4=NCCS4)C(=O)OCOC(=O)C(C)(C)C)C(C)O

Ambrisentan, أمبريسنتان , 安立生坦 ,アンブリセンタン

$
0
0

Ambrisentan structure.svgChemSpider 2D Image | ambrisentan | C22H22N2O4Ambrisentan.png

Ambrisentan

BSF-208075; LU-208075

(+)-(2S)-2-[(4,6-dimethylpyrimidin-2-yl)oxy]-3-methoxy-3,3-diphenylpropanoic acid

  • Molecular FormulaC22H22N2O4
  • Average mass378.421 Da
(2S)-2-[(4,6-dimethylpyrimidin-2-yl)oxy]-3-methoxy-3,3-diphenylpropanoic acid
177036-94-1 [RN]
8128
HW6NV07QEC
أمبريسنتان [Arabic] [INN]
安立生坦 [Chinese] [INN]
QA-7701
UNII:HW6NV07QEC
BSF208075
Letairis
Letairis®
LU208075
Trade Name:Letairis® / Volibris®
MOA:Type A endothelin receptor (ETA) antagonist
Indication:Pulmonary arterial hypertension
Company:Abbott (Originator) , Gilead,GlaxoSmithKline
アンブリセンタン
Ambrisentan

C22H22N2O4 : 378.42
[177036-94-1

Ambrisentan (U.S. trade name Letairis; E.U. trade name Volibris; India trade name Pulmonext by MSN labs) is a drug indicated for use in the treatment of pulmonary hypertension.

The peptide endothelin constricts muscles in blood vessels, increasing blood pressure. Ambrisentan, which relaxes those muscles, is an endothelin receptor antagonist, and is selective for the type A endothelin receptor (ETA).[1] Ambrisentan significantly improved exercise capacity (6-minute walk distance) compared with placebo in two double-blind, multicenter trials (ARIES-1 and ARIES-2).[2]

Ambrisentan was approved by the U.S. Food and Drug Administration (FDA) and European Medicines Agency, and designated an orphan drug, for the treatment of pulmonary hypertension.[3][4][5][6][7]

Ambrisentan is an endothelin receptor antagonist used in the therapy of pulmonary arterial hypertension (PAH). Ambrisentan has been associated with a low rate of serum enzyme elevations during therapy, but has yet to be implicated in cases of clinically apparent acute liver injury.

Ambrisentan was first approved by the U.S. Food and Drug Administration (FDA) on Jun 15, 2007, then approved by the European Medicines Agency (EMA) on Apr 21, 2008 and approved by Pharmaceuticals and Medical Devices Agency of Japan (PMDA) on Jul 23, 2010. In 2000, Abbott, originator of ambrisentan, granted Myogen (acquired by Gilead in 2006) a license to the compound for the treatment of PAH. In 2006, GlaxoSmithKline obtained worldwide rights to market the compound for PAH worldwide, with the exception of the U.S. It is marketed as Letairis® by Gilead in US.

Ambrisentan is an endothelin receptor antagonist, and is selective for the type A endothelin receptor (ETA). It is indicated for the treatment of pulmonary arterial hypertension (PAH) (WHO Group 1) to improve exercise ability and delay clinical worsening. Studies establishing effectiveness included predominantly patients with WHO Functional Class II-III symptoms and etiologies of idiopathic or heritable PAH (64%) or PAH associated with connective tissue diseases (32%).

Letairis® is available as film-coated tablet for oral use, containing 5 or 10 mg of free Ambrisentan. The recommended starting dose is 5 mg once daily with or without food, and increase the dose to 10 mg once daily if 5 mg is tolerated.

Recent Developments and Publications

Last Updated 9/2/2015
8/15/2015Reprod. Toxicol. Endothelin receptor activation mediates strong pulmonary vasoconstriction and positive inotropic effect on the heart. These physiologic effects are vital for the development of the fetal cardiopulmonary system. As such, endothelin receptor antagonists such as Ambrisentan are teratogenic.[8]
8/27/2015NEJM Ambrisentan when used in combination therapy with Tadalafil was found to be more efficacious in treating treatment naive patients with WHO class II or III Pulmonary Arterial Hypertension than monotherapy using either drug.[9]
Approval Date Approval Type Trade Name Indication Dosage Form Strength Company Review Classification
2007-06-15 Marketing approval Letairis Pulmonary arterial hypertension Tablet, Film coated 5 mg/10 mg Gilead Priority; Orphan
Approval Date Approval Type Trade Name Indication Dosage Form Strength Company Review Classification
2008-04-21 Marketing approval Volibris Pulmonary arterial hypertension Tablet, Film coated 5 mg/10 mg GlaxoSmithKline Orphan
Approval Date Approval Type Trade Name Indication Dosage Form Strength Company Review Classification
2010-07-23 Marketing approval Volibris Pulmonary arterial hypertension Tablet, Film coated 2.5 mg GlaxoSmithKline
Approval Date Approval Type Trade Name Indication Dosage Form Strength Company Review Classification
2010-10-19 Marketing approval 凡瑞克/Volibris Pulmonary arterial hypertension Tablet 5 mg GlaxoSmithKline
2010-10-19 Marketing approval 凡瑞克/Volibris Pulmonary arterial hypertension Tablet 10 mg GlaxoSmithKline

Clinical uses

Ambrisentan is indicated for the treatment of pulmonary arterial hypertension (WHO Group 1) in patients with WHO class II or III symptoms to improve exercise capacity and delay clinical worsening.

Image result for ambrisentan

Birth defects

Endothelin receptor activation mediates strong pulmonary vasoconstriction and positive inotropic effect on the heart. These physiologic effects are vital for the development of the fetal cardiopulmonary system. In addition to this, endothelin receptors are also known to play a role in neural crest cell migration, growth, and differentiation. As such, endothelin receptor antagonists such as Ambrisentan are known to be teratogenic.

Ambrisentan has a high risk of liver damage, and of birth defects if a woman becomes pregnant while taking it. In the U.S., doctors who prescribe it, and patients who take it, must enroll in a special program, the LETAIRIS Education and Access Program (LEAP), to learn about those risks. Ambrisentan is available only through specialty pharmacies.

External links

PATENT

WO9611914A1 / US7109205B2.

WO2010070658A2 / US2011263854A1.

WO2011004402A2 / US2012184573A1.

WO2013030410A2 / US2014011992A1.

CN103709106A.

CN103420811A.

str1

PATENT

https://patents.google.com/patent/WO2012167406A1/en

Ambrisentan and darusentan first reported in U. Med. Chem. 1996, 39, 2123-2128), as a selective antagonist of endothelin receptor A, followed by their pharmacological properties have been studied further, published in J. Med. Chem. 1996, 39, 2123-2128), US patent US 5932730, WO 2009/017777 A2 in. The formula (I), when R is methyl, Chinese name is (+) – ambrisentan, Chinese chemical name is (+) – (2S) -2 – [(4, 6- dimethyl-pyrimidine 2-yl) – oxy] -3-methoxy-3,3-diphenyl-propionic acid; English name is (+) – ambrisentan, English name: (S) -2- (4,6-dimethylpyrimidin -2-yloxy) -3-methoxy-3,3-diphenylpropanoic acid; when R is methoxy, Chinese as (+) – darusentan, Chinese chemical name is (+) – (2S) -2- [ (4,6-dimethoxypyrimidin-2-yl) – oxy] -3-methoxy-3,3-diphenyl-propionic acid; English name is (+ darusentan, English name: (S) – . 2- (4,6-dimethoxypyrimidin-2-yloxy) -3-methoxy-3,3-diphenylpropanoic acid ambrisentan now been approved by the FDA in the United States, the trade name Letairis, for the oral treatment of pulmonary hypertension; up Lu bosentan new drugs may be resistant hypertension (Resistant hypertension) of.

Existing ambrisentan or darusentan synthetic techniques include benzophenone Darzens reaction of an epoxy compound and a racemic methyl chloroacetate, the racemic epoxide opening catalyst in a solution of boron trifluoride diethyl ether ring to give the chiral alcohol latent after substitution reaction and then after hydrolysis reaction ambrisentan or darusentan. Existing obtained optically pure (+) – ambrisentan or (+) – darusentan methods rely mainly on resolution techniques. For example, the split is by a latent chiral alcohol or R- L- proline methyl phenethylamine, see WO 2010/070658 A2, WO 2011/004402 A2. It is well known as chiral utilization of raw materials is not high, resulting in increased costs, limiting industrial-scale applications.

 Example 1, (+) – ambrisentan ((2S) -2 – [(4,6- dimethyl-pyrimidin-2-yl) – oxy] -3-methoxy-3,3-diphenyl propionic acid) of

Preparation of 3,3-diphenyl-2,3-epoxy-propionate (1) (2S)

Figure imgf000006_0001

As indicated above Formula Scheme, wherein, Ph is phenyl; Ac is acetyl;

To a 50 L reactor equipped with a mechanical stirrer was added 3.0 L of acetonitrile was dissolved in 3,3-diphenyl acrylate (0.536 mol, 135.0 g), was dissolved in 1.5 L of acetonitrile to give a concentration of 0.12 M 4 M ethylenediamine of formula (IV) shown fructose derived chiral ketones and tetra-n-butylammonium hydrogen sulfate (36 mmol, 12.2 g), was then added containing 3.0 L Ι χ ΙΟ “an aqueous solution of disodium ; cooling liquid into the reaction vessel dissection, the kettle temperature adjusted to -5 ° C- + 5 ° C; was added in batches with stirring pulverized with the pulverizer medicine through a 1.85 kg potassium hydrogen sulfate complex salt mixture (Oxone®), and 0.78 kg NaHCO 3 (9.29mol), and takes about 4.5 hours complete addition of the above mixture, after the addition the reaction mixture was continued stirring the reaction under this condition (in the system, 3,3- diphenyl acrylate, over a potassium bisulfate salts and complexes of formula molar ratio of fructose derived chiral ketone (IV) is shown in h 5: 0.34), and the timing detection reactions by gas chromatography; the end of the reaction after 5 hours , 5.0 L of water was added to dilute the reaction solution, and extracted with 5.0 L of ethyl acetate; the aqueous phase was added 2.5 L of acetic Extracted with ethyl; organic phases were combined and concentrated to remove the solvent to give homogeneous Qing 162.56 g (2S) -3,3- diphenyl-2,3-epoxy-propionate, crude yield greater than 99%, No purification processing the next reaction, nuclear magnetic conversion was 92%, measured by HPLC enantiomeric excess of 86.9%, Analytical conditions: column model Chiralcel OD-H, n has a volume ratio of the embankment and isopropanol 98: 2 analysis of wavelength 210 nm, the mobile phase flow rate of 1 mL / min, t! = 9.5 min, t 2 = 13.01 min, 86.9% ee.

IR (fi lm) 1760, 1731 cm- 1; ¾ NMR [400 MHz, CDC1 3] δ 7.46-7.44 (m, 2H), 7.36-7.31 (m, 8H), 3.99 (m, 3H), 0.96 (t , J = 7.2Hz, 3H); 13 C NMR [100 MHz, CDC1 3] δ 166.99, 138.98, 135.62, 128.67, 128.53, 128.36, 128.13, 127.04, 66.57, 62.16, 61.43, 13.96.

(2) (2S) -2-phenyl-3,3-hydroxy-3-methoxy propionate

Figure imgf000006_0002

The step (1) 162.56 g obtained in unpurified (2S) – 3,3-diphenyl acrylate epoxy crude compound was dissolved in 100 mL of methanol, 1 mL of boron trifluoride etherate ((2S ) – mole fraction of ethylene-3,3-diphenyl acrylate and boron trifluoride diethyl ether ratio of 1: 0.013) for the epoxy ring opening reaction; after controlling the reaction temperature is 20 ° C, reacted for 8 hours , the reaction solution was concentrated, ethyl acetate and aqueous extraction of the reaction solution after the ethyl acetate was concentrated to give 166.0 g of intermediate (2S) -2- hydroxy-3-methoxy-3,3-diphenyl acetic acid ester, crude yield of 92%, measured by high performance liquid enantiomeric excess of 86.9%, Analytical conditions: column model Chiralcel OD-H, n-and isopropyl alcohol embankment has a volume ratio of 98: 2, the wavelength analysis 210 nm, mobile phase flow rate of 1 mL / min,

Figure imgf000007_0001

min, t 2 = 14.51 min, 85.8% ee .;

IR (film) 1769, 1758 cm “1; 1H NMR [400 MHz, CDC1 3] δ 7.50-7.28 (m, 10H), 5.18 (s, 1H), 4.10 (t, 2H), 3.20 (s, J = 7.2Hz, 3H), 3.03 (s , 1H), 1.17 (t, J = 7.2Hz, 3H); 13 C NMR [100 MHz, CDC1 3] δ 172.48, 141.13, 140.32, 128.97, 128.73, 128.99, 127.81, 127.76, 127.62, 85.01, 77.42, 61.76, 52.62, 14.07.

-3-methoxy-3,3-diphenyl propionate (3) (2S) -2- [- oxo – dimethyl-pyrimidin-2-yl)]

Figure imgf000007_0002

Step (2) obtained in 166.0 g of intermediate (2S) -2- hydroxy-3-methoxy-3,3-diphenyl-propionate were added N, N- dimethylformamide 750 mL , potassium carbonate 45.54 g, was added 4,6-dimethyl after stirring for about half an hour 2-methanesulfonyl-pyrimidin nucleophilic substitution reaction at 80 ° C in an oil bath, the system, (2S) -2- hydroxy -3-methoxy-3,3-diphenyl-ethyl, 4,6-dimethyl-2 molar fraction ratio methylsulfonylpyrimidine and potassium carbonate is 1: 1.2: 0.6; nuclear magnetic after complete consumption of starting material was monitored after about 3 hours, water was added and the reaction solution was extracted with ethyl acetate, the ethyl acetate layer was concentrated to give 237.70 g of intermediate (2S) -2 – [(4,6- dimethyl-pyrimidin-2-yl ) – oxy] -3-methoxy-3,3-diphenyl propionate, crude yield greater than 99%, measured by HPLC enantiomeric excess of 85.9%, Analytical conditions: column Chiralcel OD model volume -H, isopropanol and n has embankment ratio of 98: 2, analysis wavelength was 210 nm, the mobile phase flow rate of 1 mL / min, t ^ lO.15 min, t 2 = 11.87 min, 85.9% ee .

IR (film) 1750cm “VH NMR [400 MHz, CDC1 3] δ 7.45 (d, J = 7.2 Hz, 2H), 7.39 (d, J = 7.2 Hz, 2H), 7.33-7.19 (m, 7H), 6.70 (s, 1H), 6.12 ( s, 1H), 4.01-3.85 (m, 2H), 3.50 (s, 3H) 2.38 (s, 6H), 0.93 (t, J = 6.8 Hz, 3H); 13 C NMR [100 MHz, CDC1 3] δ 169.51, 168.70, 163.86, 142.50, 141.29, 128.54, 128.03, 127.97, 127.94, 127.47, 127.40, 115.03, 83.76, 79.23, 77.43, 60.66, 53.92, 23.99, 13.93;. Anal Calcd For C 24 H 26 N 2 O 4 : C, 70.92; H, 6.45; N, 6.89 Found:. C, 70.72; H, 6.47; N, 6.83.

(4) (28) -2 – [(4,6-dimethyl-2-yl) – oxy] -3-methoxy-3,3-diphenyl-propionic acid ((+) – Abe Students Tanzania) preparation

Figure imgf000007_0003

To step (3) 237.7 g of the intermediate obtained (2S) -2 – [(4,6- dimethyl-pyrimidin-2-yl) – oxy] -3-methoxy-3,3-diphenyl propionate was dissolved in 1.2 L of organic solvent is 1,4-dioxane was added 600 mL of an aqueous solution containing 92.3 g of sodium hydroxide (wherein, (2S) -2 – [(4,6- dimethyl pyrimidin-2-yl) – oxy] -3-methoxy-3,3-diphenyl propionate and sodium hydroxide molar fraction ratio of 1: 4), the reaction temperature was 80 ° C, the reaction after 8 hours, the reaction solution was concentrated, using (1 L, 0.5 L, 0.5 L) and extracted with ether to remove organic impurities, the aqueous phase was extracted after addition of hydrochloric acid to adjust pH 3, large amount of solid appears; then the aqueous phase was added 1.0 L ethyl acetate, filtered to remove insolubles (insolubles which was found after analysis racemic ambrisentan, 23.37 g), the organic layer was concentrated, i.e., optically pure can be obtained 103.9 g (+) – ambrisentan, from 3 , 3-diphenyl acrylate departure, the optically pure (+) – ambrisentan, a yield of 52.3%. A small amount of the obtained reaction with ambrisentan diazo embankment derived (2S) -2 – [(4,6- dimethyl-pyrimidin-2-yl) – oxy] -3-methoxy-3,3 methyl diphenyl measured enantiomeric excess ambrisentan. (2S) -2 – [(4,6- dimethyl-pyrimidin-2-yl) – oxy] -3-methoxy-3,3-diphenyl-propionic acid methyl ester: HPLC measured enantiomer excess of 99.1%, Analytical conditions: column model Chiralcel OD-H, n has a volume ratio of isopropanol embankment 98:! 2, analysis wavelength was 210 nm, the mobile phase flow rate of 1 mL / min, t = 11.61 min, t 2 = 14.05 min , 99.1% ee.

[a] D 25 = + 174.2 (c = 0.5, MeOH); mp> 150 ° C turns yellow,> 180 ° C into a black, 182 ° C melt; 1H NMR [400 MHz, CDC1 3] δ 7.43 ( d, J = Hz, 2H) , 7.29-7.19 (m, 8H), 6.63 (s, 1H), 6.30 (s, 1H), 3.26 (s, 3H) 2.31 (s, 6H); 13 C NMR [100 MHz, CDC1 3] δ 178.98,170.54, 169.70, 163.48, 139.91, 138.91, 128.77, 128.67, 128.22, 128.08, 115.34, 84.67, 77.55, 53.49, 23.93; 1H NMR [400 MHz, DMSO] δ 12.53 (s, 1H ), 7.34-7.20 (m, 10H) , 6.95 (s, 1H), 6.14 (s, 1H), 3.37 (s, 3H) 2.34 (s, 6H); 13 C NMR [100 MHz, DMSO] δ 169.01, 163.14, 142.59, 141.41, 127.80, 127.68, 127.64, 127.19, 126.95, 114.72, 83.12, 77.55, 52.99, 23.30.

CLIP

SEE https://www.pharmacodia.com/yaodu/html/v1/chemicals/a01610228fe998f515a72dd730294d87.html

CLIP

http://www.orgsyn.org/demo.aspx?prep=v89p0350#ref68

Image result for ambrisentan

Shi and coworkers recently obtained 120 g of virtually enantiopure (+)-ambrisentan (97) without the need for column chromatography (Scheme 23).68 (+)-Ambrisentan, an endothelin-1 receptor antagonist, is currently used to treat hypertension. Ketone 2-catalyzed epoxidation afforded 96 in 90% conversion and 85% ee. Compound 97 was further enriched via precipitation and filtration of the racemate.

  1. Peng, X.; Li, P.; Shi, Y. J. Org. Chem201277, 701-703.

Clip

https://pubs.acs.org/doi/10.1021/acs.oprd.8b00184

Process Research for (+)-Ambrisentan, an Endothelin-A Receptor Antagonist

 Collaborative Innovation Center of Yangze River Delta Region Green PharmaceuticalsZhejiang University of Technology18 Chaowang Road, Hangzhou 310014, China
 Department of Pharmaceutial EngineeringChina Pharmaceutical University24 Tongjiaxiang, Nanjing 210009, China
§ Shanghai Institute of Pharmaceutical IndustryChina State Institute of Pharmacetical Industry285 Gebaini Road, Pudong, Shanghai 201203, China
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.8b00184
Publication Date (Web): August 6, 2018
Copyright © 2018 American Chemical Society
Abstract Image

An efficient and robust synthetic route to (+)-ambrisentan ((+)-AMB) was designed by recycling the unwanted isomer from the resolution mother liquors. The racemization of AMB in the absence of either acid or base in the given solvents was reported. The recovery process was developed to produce racemates with purities over 99.5%. The mechanism of the formation of the process-related impurities of (+)-AMB is also discussed in detail. (+)-AMB was obtained in 47% overall yield with >99.5% purity and 99.8% e.e. by chiral resolution with only one recycling of the mother liquors on a 100-g scale without column purification.

https://pubs.acs.org/doi/suppl/10.1021/acs.oprd.8b00184/suppl_file/op8b00184_si_001.pdf

PaPER

https://pdfs.semanticscholar.org/3801/d5a98a526a4386c431e25d3ac99a328bfae2.pdf

CHEMICAL ENGINEERING TRANSACTIONS VOL. 46, 2015 A publication of The Italian Association of Chemical Engineering Online at http://www.aidic.it/cet Guest Editors: Peiyu Ren, Yancang Li, Huiping Song Copyright © 2015, AIDIC Servizi S.r.l., ISBN 978-88-95608-37-2; ISSN 2283-9216

Improved Synthesis Process of Ambrisentan and Darusentan Jian Lia , Lei Tian*b, c a School of Environmental Science, Nanjing Xiaozhuang University, 3601 Hongjing Road, Nanjing, Jiangsu, 211171, China b School of Petroluem Engineer, Yangtze University, Wuhan, Hubei, 430100, P. R. China c Key Laboratory of Exploration Technologies for Oil and Gas Resources (Yangtze University), Ministry of Education tianlei4665@163.com

2-hydroxy-3-phenoxy-3, 3-diphenylpropinate (5) was prepared from benzophenone via Darzens, methanolysis and hydrolysis reaction. The compound (5) was salified with (S)-dehydroabietylamine (7) and diasterotropic resolution was carried out to provide the key intermediate (S)-2-hydroxy-3-methoxy-3, 3-diphenylpropionic acid (6). Compound (6) was condensed with 2-methylsulfonyl-4, 6-dimethylpyrimidine and 2-methoxysulfonyl4, 6-dimethylpyrimidine to afford ambrisentan (1) and darusentan (7), respectively. Two products were with excellent charity and chemical purity. The total yield of the synthesis was 30.1% and 29.6%, respectively.

str1

Synthesis of methyl 3, 3-diphenyloxirane-2-carboxylate (3) To a solution of sodium methanolate (4.3 g, 79.6 mmol) in dry THF (25 mL) was added the solution of benzophenone (7.2 g, 39.5 mmol) and methyl chloroacetate (6.6 g, 60.8 mmol) in dry THF (15 mL) and stirred at -10 °C for 2 h. The mixture was quenched with water (50 mL). The solution was extracted with diethyl ether (80 mL×3). The organic phases were combined and washed with saturated NaCl. The solution was dried over Na2SO4, filtered, and evaporated under reduced pressure to afford a light yellow oil. The residue (3) can apply in next step without further purification (8.24 g, 82.1%). 1H-NMR (CDCl3): δ 3.52 (s, 3H), 3.99 (s, 1H), 7.32-7.45 (m, 10H).

Synthesis of 2-hydroxy-3-methoxy-3, 3-diphenylpropanoic acid (5)

To a solution of compound (3) (8.2 g, 31.6 mmol) in methanol (40 mL) was added p-toluene sulfonic acid (0.5 g) and stirred at for 0.5 h to afford the solution containing compound (4). Aqueous solution of NaOH (10% wt.) (60 mL) was added to the solution of compound (4) and the mixture was stirred at refluxed for 1h (ester disappeared by TLC). The solution was evaporated in order to remove a lot of methanol. The residue was acidified to pH 2 by conc. HCl. The solution was stirred for overnight and white solid stayed at the aqueous layer. The precipitate was filtered and deeply dried under vacuum to afford (5). (7.34 g, 85.3%). 1H-NMR (CDCl3): δ 3.22 (s, 3H), 5.14 (br, 1H), 5.20 (d, 1H), 7.18-7.37 (m, 10H), 12.30 (1H, br). Synthesis of (S)-2-hydroxy-3-methoxy-3, 3-diphenylpropanoate (6) The solution of compound (5) (14 g, 51.4 mmol) in methyltertiarybutylether (140 mL) was stirred and refluxed for 0.5 h. Dehydroabietylamine (7) (14.7 g, 51.4 mmol) in methyltertiarybutylether (50 mL) was added dropwise in 10 min. After addition, the reaction mixture was stirred for 1 h under reflux temperature. The reaction mixture was cooled to 0 °C and continued to stir for 2 h. The solid ((R, S)-diastereoisomers) was precipitated from the solution, filtered, washed with acetonitrile. The filtrate was diluted with water (100 mL) and acidified to pH 2 by conc. HCl. The aqueous solution was extracted with methylteriarybutylether (50 mL×4). The organic phases were combined and washed with water (80 mL). The organic phase was separated, dried over anhydrous Na2SO4 and evaporated under reduced pressure to afford white residue. The residue was recystallized from toluene to afford (6) as a white solid. (5.53 g, 39.5%). 1H-NMR (CDCl3): δ 3.22 (s, 3H), 5.14 (br, 1H), 5.20 (d, 1H), 7.18-7.37 (m, 10H), 12.30 (1H, br). [α] 20 D =12.3°(c=1.8% in ethanol).

Synthesis of (+)-ambrisentan (1) To a solution of compound (6) (3.6 g, 13.1 mmol) and NaNH2 (1.0 g, 25.6 mmol) in DMF (20 mL) was added 4, 6-dimethyl-2-(methylsulfonyl) pyrimidine (3.63 g, 19.6 mmol) in DMF (10 mL) slowly. After addition, the reaction was stirred for 5 h at room temperature. The solution was quenched with water (20 mL) and acidified to pH 2 by 10% H2SO4 aqueous solution. The mixture was extracted with ethyl acetate (50 mL ×4). The combined organic layers were washed with water (30 mL) and saturated NaCl solution (30 mL). The organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure. The resulting residue was recrystallized from iso-propyl alcohol (30 mL) and water (40 mL), and precipitate formed was filtered off. The cake was deeply dried under vacuum to afford (1) as a white solid. (4.27 g, 86.1%). 1H-NMR (CDCl3): δ 2.39 (s, 6H), 3.32 (s, 3H), 6.43 (s, 1H), 6.70 (s, 1H), 7.28-7.40 (m, 8H), 7.53-7.56 (d, 2H). MS-EI (m/z): 377(M-H). HPLC (XDB-C18, CH3OH/10mmol/L NaH2PO4 + 0.1% H3PO4 = 70/30, 1.0 mL/min): tR 5.2 min (>99.0%); ee= 99.0%.

Patent EP2547663A1

File:Ambrisentan synthesis.svg

Image result for ambrisentan

Title: Ambrisentan
CAS Registry Number: 177036-94-1
CAS Name: (aS)-a-[(4,6-Dimethyl-2-pyrimidinyl)oxy]-b-methoxy-b-phenylbenzenepropanoic acid
Manufacturers’ Codes: BSF-208075; LU-208075
Molecular Formula: C22H22N2O4
Molecular Weight: 378.42
Percent Composition: C 69.83%, H 5.86%, N 7.40%, O 16.91%
Literature References: Nonpeptide endothelin ETA receptor antagonist. Prepn: H. Riechers et al., WO 9611914eidemUS5932730 (1996, 1998 both to BASF); H. Riechers et al., J. Med. Chem. 39, 2123 (1996). Pharmacology: H. Vatter et al., Clin. Neuropharmacol. 26, 73 (2003). Clinical evaluation in pulmonary arterial hypertension: N. Galié et al., J. Am. Coll. Cardiol. 46, 529 (2005). Review of development and therapeutic potential: G. E. Billman, Curr. Opin. Invest. Drugs 3, 1483-1486 (2002).
Derivative Type: (±)-Form
CAS Registry Number: 713516-99-5
Properties: Crystals from diethylether, mp 190-191°.
Melting point: mp 190-191°
Therap-Cat: Antihypertensive.
Keywords: Antihypertensive; Endothelin Receptor Antagonist.

References

  1. Jump up^ Vatter H, Seifert V (2006). “Ambrisentan, a non-peptide endothelin receptor antagonist”. Cardiovasc Drug Rev24 (1): 63–76. doi:10.1111/j.1527-3466.2006.00063.xPMID 16939634.
  2. Jump up^ Frampton JE (2011). “Ambrisentan”. American Journal of Cardiovascular Drugs11 (4): 215–26. doi:10.2165/11207340-000000000-00000PMID 21623643.
  3. Jump up^ Pollack, Andrew (2007-06-16). “Gilead’s Drug Is Approved to Treat a Rare Disease”The New York TimesArchived from the original on May 24, 2013. Retrieved 2007-05-25.
  4. Jump up^ “U.S. Food and Drug Administration Approves Gilead’s Letairis Treatment of Pulmonary Arterial Hypertension” (Press release). Gilead Sciences. 2007-06-15. Archived from the original on 2007-09-27. Retrieved 2007-06-16.
  5. Jump up^ “FDA Approves New Orphan Drug for Treatment of Pulmonary Arterial Hypertension” (Press release). Food and Drug Administration. 2007-06-15. Archived from the original on 23 June 2007. Retrieved 2007-06-22.
  6. Jump up^ “GlaxoSmithKline’s Volibris (ambrisentan) receives authorisation from the European Commission for the treatment of Functional Class II and III Pulmonary Arterial Hypertension” (Press release). GlaxoSmithKline. 2008-04-25. Archived from the original on 30 April 2008. Retrieved 2008-04-29.
  7. Jump up^ Waknine, Yael (2005-05-09). “International Approvals: Ambrisentan, Oral-lyn, Risperdal”Medscape. Retrieved 2007-06-16.
  8. Jump up^ de Raaf MA, Beekhuijzen M, Guignabert C, Vonk Noordegraaf A, Bogaard HJ (2015). “Endothelin-1 receptor antagonists in fetal development and pulmonary arterial hypertension”. Reproductive Toxicology56: 45–51. doi:10.1016/j.reprotox.2015.06.048PMID 26111581.
  9. Jump up^ Galiè, Nazzareno; Barberà, Joan A.; Frost, Adaani E.; Ghofrani, Hossein-Ardeschir; Hoeper, Marius M.; McLaughlin, Vallerie V.; Peacock, Andrew J.; Simonneau, Gérald; Vachiery, Jean-Luc; Grünig, Ekkehard; Oudiz, Ronald J.; Vonk-Noordegraaf, Anton; White, R. James; Blair, Christiana; Gillies, Hunter; Miller, Karen L.; Harris, Julia H.N.; Langley, Jonathan; Rubin, Lewis J. (2015). “Initial Use of Ambrisentan plus Tadalafil in Pulmonary Arterial Hypertension”. New England Journal of Medicine373 (9): 834–44. doi:10.1056/NEJMoa1413687.
  1. US5703017
  2. CA2201785
  3. US5840722
  4. US7601730
  5. US8377933
  6. US7109205
  7. USRE42462
  8. US8349843
  9. US9474752
  10. US9549926

Patent

Publication numberPriority datePublication dateAssigneeTitle
CN1160396A *1994-10-141997-09-24巴斯福股份公司Carboxylic acid derivs., their preparation and their use
WO2003066614A1 *2002-02-042003-08-14Colorado State University Research FoundationAsymmetric epoxidation of electron deficient olefins
WO2010070658A2 *2008-11-052010-06-24Msn Laboratories LimitedImproved process for the preparation of endothelin receptor antagonists
CN102276536A *2011-06-102011-12-14中国科学院化学研究所An optically pure (+) – ambrisentan and optically pure (+) – darusentan preparation
Family To Family Citations
US6030975A *1997-03-142000-02-29Basf AktiengesellschaftCarboxylic acid derivatives, their preparation and use in treating cancer
JP2010535210A2007-07-312010-11-18アボット ゲーエムベーハー ウント コンパニー カーゲーMetabolites and derivatives of ambrisentan
WO2010091877A3 *2009-02-132010-11-11Ratiopharm GmbhProcess for producing ambrisentan
WO2011004402A32009-07-102011-03-10Cadila Healthcare LimitedImproved process for the preparation of ambrisentan and novel intermediates thereof

Non-Patent Citation

Title
WANG, BIN ET AL.: ‘A Diacetate Ketone-Catalyzed Asymmetric Epoxidation of Olefins’ J. ORG. CHEM. vol. 74, 23 April 2009, pages 3986 – 3989 *

Publication numberPriority datePublication dateAssigneeTitle
Family To Family Citations
CN102276536B *2011-06-102015-04-29中国科学院化学研究所Preparation method of optically pure (+)-ambrisentan and optically pure (+)-darusentan
CN103420811B *2012-05-182015-04-15上海医药工业研究院Intermediate compound used for preparing Ambrisentan, preparation method thereof, and preparation of Ambrisentan
CN103524425A *2012-07-042014-01-22天津药物研究院Crystal form V of ambrisentan as well as preparation method and application thereof
CN102850300A *2012-09-182013-01-02中国科学院化学研究所Preparation method of alpha,beta-epoxy amide compounds
CN104592129A *2013-10-302015-05-06武汉启瑞药业有限公司Improved method used for preparing ambrisentan
CN103709106A *2013-12-062014-04-09石家庄博策生物科技有限公司Stereoselectivity preparation method for Letairis
CN103755569A *2013-12-262014-04-30上海皓骏医药科技有限公司Preparation method for ambrisentan intermediate compound
Ambrisentan
Ambrisentan structure.svg
Clinical data
AHFS/Drugs.com Monograph
License data
Pregnancy
category
  • AU: X (High risk)
  • US: X (Contraindicated)
Routes of
administration
Oral
ATC code
Legal status
Legal status
Pharmacokinetic data
Bioavailability Undetermined
Protein binding 99%
Elimination half-life 15 hours (terminal)
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
ChemSpider
UNII
ChEMBL
ECHA InfoCard 100.184.855 Edit this at Wikidata
Chemical and physical data
Formula C22H22N2O4
Molar mass 378.421 g/mol
3D model (JSmol)

/////////////Ambrisentan,  أمبريسنتان ,  安立生坦 , BSF-208075,  LU-208075, アンブリセンタン

CC1=CC(=NC(=N1)OC(C(=O)O)C(C2=CC=CC=C2)(C3=CC=CC=C3)OC)C

USFDA has released GUIDANCE for Quality Attributes of *CHEWABLE TABLETS

$
0
0

DRUG REGULATORY AFFAIRS INTERNATIONAL

Image result for Quality Attributes of *CHEWABLE TABLETS

*CQAs of CHEWABLE TABLETS (CT)*
USFDA has released GUIDANCE for Quality Attributes of *CHEWABLE TABLETS*
According to this latest guideline, FDA has recommended sponsor/applicant should also incorporate following CQAs:
*1. PATIENT ACCEPTABILITY*
Acceptable Taste, Mouthfeel & Aftertaste With-
*2. HARDNESS / BREAKING FORCE / CRUSHING STRENGTH*
Hardness of CTshould be kept  low  (i.e.  <12 kp).
A higher hardness  value  (e.g.,  >12 kp)  may  be  considered if  justified.  An example  of  such justification could be  demonstrating  significant disintegration and/or  reduction in hardness  of  such  tablets  following  brief  i.e.  30 seconds  in-vitro exposure to simulated saliva (1 mL) before chewing to ensure patient compliance without  GI  obstruction (choking in throat / blocking bowel movement) in the case if patient swallow tablet without chewing due to high hardness
*3. CHEWING DIFFICULTY INDEX*
CDI is a value derived from the relationship between two methods used for measuring tablet strength: diametral compression (diametrical tensile strength)…

View original post 109 more words

FDA approves first drug Oxervate (cenegermin) for neurotrophic keratitis, a rare eye disease

$
0
0
The U.S. Food and Drug Administration today approved the first drug, Oxervate (cenegermin), for the treatment of neurotrophic keratitis, a rare disease affecting the cornea (the clear layer that covers the colored portion of the front of the eye).
“While the prevalence of neurotrophic keratitis is low, the impact of this serious condition on an individual patient can be devastating,” said Wiley Chambers, M.D., an ophthalmologist in the FDA’s Center for Drug Evaluation and Research. “In the past, it has often been necessary to turn to surgical interventions; these treatments are usually only palliative in this disease. Today’s approval provides a novel topical treatment and a major advance that offers complete corneal healing for many of these patients.”

August 22, 2018

Release

The U.S. Food and Drug Administration today approved the first drug, Oxervate (cenegermin), for the treatment of neurotrophic keratitis, a rare disease affecting the cornea (the clear layer that covers the colored portion of the front of the eye).

“While the prevalence of neurotrophic keratitis is low, the impact of this serious condition on an individual patient can be devastating,” said Wiley Chambers, M.D., an ophthalmologist in the FDA’s Center for Drug Evaluation and Research. “In the past, it has often been necessary to turn to surgical interventions; these treatments are usually only palliative in this disease. Today’s approval provides a novel topical treatment and a major advance that offers complete corneal healing for many of these patients.”

Neurotrophic keratitis is a degenerative disease resulting from a loss of corneal sensation. The loss of corneal sensation impairs corneal health causing progressive damage to the top layer of the cornea, including corneal thinning, ulceration, and perforation in severe cases. The prevalence of neurotrophic keratitis has been estimated to be less than five in 10,000 individuals.

The safety and efficacy of Oxervate, a topical eye drop containing cenegermin, was studied in a total of 151 patients with neurotrophic keratitis in two, eight-week, randomized controlled multi-center, double-masked studies. In the first study, patients were randomized into three different groups. One group received Oxervate, a second group received an eye drop with a different concentration of cenegermin, and the third group received an eye drop without cenegermin. In the second study, patients were randomized into two groups. One group was treated with Oxervate eye drops and the other group was treated with an eye drop without cenegermin. All eye drops in both studies were given six times daily in the affected eye(s) for eight weeks. In the first study, only patients with the disease in one eye were enrolled, while in the second study, patients with the disease in both eyes were treated in both eyes (bilaterally). Across both studies, complete corneal healing in eight weeks was demonstrated in 70 percent of patients treated with Oxervate compared to 28 percent of patients treated without cenegermin (the active ingredient in Oxervate).

The most common adverse reactions in patients taking Oxervate are eye pain, ocular hyperemia (enlarged blood vessels in the white of the eyes), eye inflammation and increased lacrimation (watery eyes).

Oxervate was granted Priority Review designation, under which the FDA’s goal is to take action on an application within six months of application filing where the agency determines that the drug, if approved, would provide a significant improvement in the safety or effectiveness of the treatment, diagnosis or prevention of a serious condition. Oxervate also received Orphan Drug designation, which provides incentives to assist and encourage the development of drugs for rare diseases.

The FDA granted approval of Oxervate to Dompé farmaceutici SpA.

/////////////fda 2018, Oxervate, cenegermin, orphan drug, priority review

Cenegermin

$
0
0
Image result for cenegermin
Active Substance General information The active substance in Oxervate, cenegermin, is a recombinant human Nerve Growth factor (rhNGF) produced in E. coli strain HMS174. The molecule is identical to human Nerve Growth factor (NGF), a naturally occurring human protein. In humans, NGF is naturally produced as pre-pro-peptide, secreted into the endoplasmic reticulum and cleaved by furin protease. The pro-sequence is further cleaved during the production process by enzymatic hydrolysis. Therefore these two amino acid changes have no influence on the final active ingredient (rhNGF), which is identical to the naturally secreted human protein. The 3D structure of rhNGF is a non-covalent dimer with three intra-molecular disulphide bridges. Cenegermin contains 118 amino acids and has a relative molecular mass of 13,266 Daltons and the following molecular formula: C583H908N166O173S8. Figure 1 shows the protein sequence of recombinant human ProNGFrh ProNGF (Figure 1A), and a map of the disulphide bridges (Figure IB):
Cenegermin sequence:
SSSHPIFHRGEFSVCDSVSVWVGDKTTATDIKGKEVMVLGEVNIN
NSVFKQYFFETKCRDPNPVDSGCRGIDSKHWNSYCTTTHTFVKAL
TMDGKQAAWRFIRIDTACVCVLSRKAVR
CAS 1772578-74-1
rhNGF, Nerve growth factor – Anabasis/Dompe; Oxervate; Sentinel
  • OriginatorAnabasis Pharma
  • DeveloperDompe Farmaceutici; Ospedale San Raffaele
  • ClassEye disorder therapies; Nerve growth factors; Neuroprotectants; Proteins
  • Mechanism of ActionNerve growth factor receptor agonists; Neuron stimulants
  • Orphan Drug StatusYes – Keratitis; Retinitis pigmentosa
  • Highest Development Phases
  • RegisteredKeratitis
  • Phase II Dry eyes; Glaucoma; Retinitis pigmentosa
  • APPROVED FDA AUG  2018

Most Recent Events

  • 28 Jul 2018No recent reports of development identified for phase-I development in Glaucoma in Italy (Ophthalmic, Drops)
  • 29 May 2018Phase-II clinical trials in Glaucoma (Ophthalmic) (http://www.dompe.com/RnD-Pipeline/)
  • 01 May 2018Dompé Farmaceutici completes a phase I trial in Glaucoma in USA (Ophthalmic) (NCT02855450)
  • Image result for cenegermin
Cenegermin (planned brand names OxervateSentinel), also known as recombinant human nerve growth factor (rhNGF), is a recombinant form of human nerve growth factor (NGF). It was approved in the European Union as an eye drop formulation for the treatment of moderate or severe neurotrophic keratitis in adults on 6 July 2017.[2][3][1] As a recombinant form of NGF, cenegermin is a peripherally selective agonist of the TrkA and LNGFR (p75NTR) which must be administered parenterally.[3] In addition to neurotrophic keratitis, cenegermin is also under development for the treatment of dry eyesretinitis pigmentosa, and glaucoma.[3] It was developed by Anabasis Pharma, Dompé Farmaceutici, and Ospedale San Raffaele.[3]
Cenegermin is a human beta-nerve growth factor (beta-ngf)-(1-118)- peptide (non-covalent dimer) produced in escherichia coli. It received European Union Approval in July, 2017 for the treatment of moderate to severe neurotrophic keratitis.
In 2013, orphan drug designations in the E.U. and in the U.S. were assigned to the candidate for the treatment of retinitis pigmentosa. The product was granted additional orphan drug designation for the treatment of neurotrophic keratitis in the U.S. and the E.U. in 2014 and 2015, respectively.
Cenegermin, a recombinant human nerve growth factor developed by Dompé was first approved in July 2017 in the E.U. for the treatment of moderate to severe neurotrophic keratitis (NK) in adults
Clip
The U.S. Food and Drug Administration today approved the first drug, Oxervate (cenegermin), for the treatment of neurotrophic keratitis, a rare disease affecting the cornea (the clear layer that covers the colored portion of the front of the eye).
“While the prevalence of neurotrophic keratitis is low, the impact of this serious condition on an individual patient can be devastating,” said Wiley Chambers, M.D., an ophthalmologist in the FDA’s Center for Drug Evaluation and Research. “In the past, it has often been necessary to turn to surgical interventions; these treatments are usually only palliative in this disease. Today’s approval provides a novel topical treatment and a major advance that offers complete corneal healing for many of these patients.”

August 22, 2018

Release

The U.S. Food and Drug Administration today approved the first drug, Oxervate (cenegermin), for the treatment of neurotrophic keratitis, a rare disease affecting the cornea (the clear layer that covers the colored portion of the front of the eye).

“While the prevalence of neurotrophic keratitis is low, the impact of this serious condition on an individual patient can be devastating,” said Wiley Chambers, M.D., an ophthalmologist in the FDA’s Center for Drug Evaluation and Research. “In the past, it has often been necessary to turn to surgical interventions; these treatments are usually only palliative in this disease. Today’s approval provides a novel topical treatment and a major advance that offers complete corneal healing for many of these patients.”

Neurotrophic keratitis is a degenerative disease resulting from a loss of corneal sensation. The loss of corneal sensation impairs corneal health causing progressive damage to the top layer of the cornea, including corneal thinning, ulceration, and perforation in severe cases. The prevalence of neurotrophic keratitis has been estimated to be less than five in 10,000 individuals.

The safety and efficacy of Oxervate, a topical eye drop containing cenegermin, was studied in a total of 151 patients with neurotrophic keratitis in two, eight-week, randomized controlled multi-center, double-masked studies. In the first study, patients were randomized into three different groups. One group received Oxervate, a second group received an eye drop with a different concentration of cenegermin, and the third group received an eye drop without cenegermin. In the second study, patients were randomized into two groups. One group was treated with Oxervate eye drops and the other group was treated with an eye drop without cenegermin. All eye drops in both studies were given six times daily in the affected eye(s) for eight weeks. In the first study, only patients with the disease in one eye were enrolled, while in the second study, patients with the disease in both eyes were treated in both eyes (bilaterally). Across both studies, complete corneal healing in eight weeks was demonstrated in 70 percent of patients treated with Oxervate compared to 28 percent of patients treated without cenegermin (the active ingredient in Oxervate).

The most common adverse reactions in patients taking Oxervate are eye pain, ocular hyperemia (enlarged blood vessels in the white of the eyes), eye inflammation and increased lacrimation (watery eyes).

Oxervate was granted Priority Review designation, under which the FDA’s goal is to take action on an application within six months of application filing where the agency determines that the drug, if approved, would provide a significant improvement in the safety or effectiveness of the treatment, diagnosis or prevention of a serious condition. Oxervate also received Orphan Drug designation, which provides incentives to assist and encourage the development of drugs for rare diseases.

The FDA granted approval of Oxervate to Dompé farmaceutici SpA.

Cenegermin
Clinical data
Trade names Oxervate, Sentinel
Synonyms Recombinant human nerve growth factor; rhNGF; human beta-nerve growth factor (beta-NGF)-(1-118) peptide (non-covalent dimer) produced in Escherichia coli[1]
Routes of
administration
Eye drops
ATC code
Identifiers
CAS Number
DrugBank
ChemSpider
  • None
UNII
KEGG
Chemical and physical data
Formula C583H908N166O173S8
Molar mass 13266.94 g/mol

References

External links

////////////fda 2018, Oxervate, cenegermin, orphan drug, priority review, EU 2017, DOMPE, neurotrophic keratitis

BINIMETINIB, биниметиниб , بينيميتينيب , 美替尼 , ビニメチニブ

$
0
0

Figure imgf000024_0001ChemSpider 2D Image | Binimetinib | C17H15BrF2N4O3

Binimetinib.svgBinimetinib.png

Binimetinib

MEK-162
биниметиниб [Russian] [INN]
بينيميتينيب [Arabic] [INN]
贝美替尼 [Chinese] [INN]
ビニメチニブ
5-[(4-bromo-2-fluorophenyl)amino]-4-fluoro-N-(2-hydroxyethoxy)-1-methyl-1H-benzimidazole-6-carboxamide
5-(4-Bromo-2-fluorophenylamino)-4-fluoro-1-methyl-1H-benzimidazole-6-carbohydroxamic acid 2-hydroxyethyl ester
6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxyethyoxy)-amide
606143-89-9  CAS
C17H15BrF2N4O3, 441.227
UNII-181R97MR71
181R97MR71
1H-Benzimidazole-6-carboxamide, 5-[(4-bromo-2-fluorophenyl)amino]-4-fluoro-N-(2-hydroxyethoxy)-1-methyl-
tyrosine kinase inhibitor, antineoplastic

Array BioPharma Inc;PHASE 3 Cancer, ovary (serous)

Novartis PHASE 3 Melanoma

CAS 606143-89-9 [RN]
9764
ARRY-162
ARRY-438162, NVP-MEK162

MEK-1 protein kinase inhibitor; MEK-2 protein kinase inhibitor

Liver injury; Melanoma; Noonan syndrome; Ovary tumor; Solid tumor

On June 27, 2018, the Food and Drug Administration approved encorafenib and binimetinib in combination patients with unresectable or metastatic melanoma with a BRAF V600E or V600K mutation, as detected by an FDA-approved test

Binimetinib, also known as Mektovi and ARRY-162, is an anti-cancer small molecule that was developed by Array Biopharma to treat various cancers.[1] Binimetinib is a selective inhibitor of MEK, a central kinase in the tumor-promoting MAPK pathway.[2] Inappropriate activation of the pathway has been shown to occur in many cancers.[2] In June 2018 it was approved by the FDA in combination with encorafenib for the treatment of patients with unresectable or metastatic BRAF V600E or V600K mutation-positive melanoma.[3]

Binimetinib, also known as Mektovi, is a potent is a potent and selective oral mitogen-activated protein kinase 1/2 (MEK 1/2) inhibitor which is combined with Encorafenib [4],[8].

On June 27, 2018, the Food and Drug Administration approved the combination of Encorafeniband binimetinib (BRAFTOVI and MEKTOVI, from Array BioPharma Inc.) in combination for patients with unresectable or metastatic melanoma with the BRAF V600E or V600K mutations, as detected by an FDA-approved test [8].

Binimetinib was originally developed by Array BioPharma, then licensed to Novartis for worldwide development in 2010. But Array Biopharma regained full worldwide rights of the product in 2015. And in 2015, Pierre Fabre acquired exclusive rights to commercialize the product.

Mechanism of action

Binimetinib is an orally available inhibitor of mitogen-activated protein kinase kinase (MEK), or more specifically, a MAP2K inhibitor.[4]MEK is part of the RAS pathway, which is involved in cell proliferation and survival. MEK is upregulated in many forms of cancer.[5]Binimetinib, uncompetitive with ATP, binds to and inhibits the activity of MEK1/2 kinase, which has been shown to regulate several key cellular activities including proliferation, survival, and angiogenesis.[6] MEK1/2 are dual-specificity threonine/tyrosine kinases that play key roles in the activation of the RAS/RAF/MEK/ERK pathway and are often upregulated in a variety of tumor cell types.[7] Inhibition of MEK1/2 prevents the activation of MEK1/2 dependent effector proteins and transcription factors, which may result in the inhibition of growth factor-mediated cell signaling.[8] As demonstrated in preclinical studies, this may eventually lead to an inhibition of tumor cell proliferation and an inhibition in production of various inflammatory cytokines including interleukin-1, -6 and tumor necrosis factor.[8]

Development

In 2015, it was in phase III clinical trials for ovarian cancer,[9] BRAF mutant melanoma,[10] and NRAS Q61 mutant melanoma.[11]

In December 2015, the company announced that the mutant-NRAS melanoma trial was successful.[12] In the trial, those receiving binimetinib had a median progression-free survival of 2.8 months versus 1.5 months for those on the standard dacarbazinetreatment.[13] NDA submitted Jun 2016,[14] and the FDA should decide by 30 June 2017.[15]

In April 2016, it was reported that the phase III trial for low-grade ovarian cancer was terminated due to lack of efficacy.[16]

Binimetinib was studied for treatment of rheumatoid arthritis, but a phase II trial did not show benefit.

In 2017, the FDA informed Array Biopharma that the phase III trial data was not sufficient and the New Drug Application was withdrawn.[17]

In June 2018 it was approved for the treatment of certain melanomas by the FDA in combination with encorafenib.[3]

Growth factor-mediated proliferative signals are transmitted from the extracellular environment to the nucleus through several pathways, including the RAS/RAF/ MEK pathway. The RAS/RAF/MEK kinase signal transduction pathway is activated through initial extracellular binding and stimulation of tyrosine receptor kinases (RTKs) by their respective cognate ligands. Upon autophosphorylation of specific tyrosine residues in the cytosolic domain of RTKs, the Grb2-Sos complex translocates to the plasma membrane, and converts the inactive RAS’GDP to active RAS’GTP. The interaction between the Grb2 docking protein and the activated kinases or the phosphorylated receptor associated proteins is mediated by the Src Homology (SH2) domain of the signaling protein that recognizes specific phosphotyrosine sequences. RAS undergoes a conformational change upon guanosine 5 ‘-triphosphate (GTP) binding and causes the recruitment of RAF- 1 to the cytoplasmic membrane where it is phosphorylated by several kinases and simultaneous disphosphorylated at key residues by protein phosphatase-2B. Activated RAF phosphorylates the mitogen- activated protein kinase kinase (MEK) on two serine residues in the activation loop, which results in the activation of this protein kinase. MEK then phosphorylates and activates extracellular signal-regulated kinase (ERK), allowing its translocation to the nucleus where it phosphorylates transcriptional factors permitting the expression of a variety of genes.

The RAS/RAF/MEK signal transduction pathway is deregulated, often through mutations that result in ectopic protein activation, in roughly 1/3 of human cancers. This deregulation in turn results in a wide array of cellular changes that are integral to the etiology and maintenance of a cancerous phenotype including, but not limited to, the promotion of proliferation and evasion of apoptosis (Dhillon et al., Oncogene, 2007, 26: 3279-3290).

Accordingly, the development of small molecule inhibitors of key members of the RAS/ RAF/ MEK signal transduction pathway has been the subject of intense effort within the pharmaceutical industry and oncology community.

MEK is a major protein in the RAS/ RAF/ MEK pathway, which signals toward cell proliferation and survival, and frequently activated in tumors that have mutations in the RAS or RAF oncogenes or in growth receptor tyrosine kinases. MEK is a key player in the RAS/RAF/MEK pathway as it is downstream of RAS and RAF. Despite being only rarely mutated in cancer (Murugan et al., Cell Cycle, 2009, 8: 2122-2124; Sasaki et al., J. Thorac. Oncol., 2010, 5: 597-600), inhibitors of the MEK1 and MEK2 proteins have also been targeted for small molecule inhibition owing to their central position within the RAS/ RAF/ MEK signal transduction pathway signaling cascade (Fremin and Meloche, J. Hematol.

Oncol., 2010, 3:8). Recently a potent MEK inhibitor failed to demonstrate efficacy in clinical trials in patients with advanced non-small cell lung cancer (Haura et al., Clin. Cancer Res., 2010, 16: 2450-2457). The reason for failure in this trial is not clear.

6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxyethyoxy)-amide (hereinafter, “Compound A”) is a benzimidazole compound that is a known potent and selective inhibitor of the MEK1 and MEK2 proteins, and useful in the treatment of hyperproliferative diseases, particularly cancer, in mammals. For example, in a recently published Phase I study of 28 patients suffering from unresectable, locally advanced or metastatic biliary cancer and who had received < 1 prior systemic therapy, oral Compound A treatment (60 mg twice daily) resulted in 1 complete regression, 1 partial regression and 11 stable disease diagnoses after at least 6 weeks of treatment (Finn et al., J. Clin. Oncol. 30, 2012 (Supplement 4, 2012 Gastrointestinal Cancers Symposium, Abstract No. 220). Compound A has also been demonstrated to be effective in the treatment of patients with either BRAFV600 or NRAS-mutant melanoma (Ascierto et al., J. Clin. Oncol. 30, 2012 (Supplement, 2012 ASCO Annual Meeting, Abstract No. 8511).

The compound, as well as a process for its preparation, is disclosed in PCT Pub. No. WO 03/077914

MEK-162, a potent, orally active MEK1/2 inhibitor, is in phase III clinical trials at Array BioPharma and licensee Novartis for the treatment of metastatic or unresectable cutaneous melanoma with NRAS mutations and in combination with LGX-818 in adult patients with BRAF V600. Phase III studies are also under way at Array BioPharma for the treatment of low grade serous carcinomas of the ovary, fallopian tube or primary peritoneum following at least one prior platinum-based chemotherapy regimen and no more than three lines of prior chemotherapy regimens. Novartis and Array BioPharma are also conducting phase II clinical studies for the treatment of locally advanced and unresectable or metastatic malignant cutaneous melanoma, harboring BRAFV600E mutations; in BRAF mutated melanoma in combination with AMG-479 and for the treatment of Noonan’s syndrome, and in non-small cell lung cancer harboring KRAS or EGFR mutation and in combination with erlotinib. MEK-162 is being evaluated in phase I/II as first line treatment of advanced biliary tract carcinoma and for the treatment of adult patients with mutant or wild-type RAS metastatic colorectal cancer. The product is in early clinical trials at Array Biopharma for the treatment of biliary cancer.

According to Array, MEK-162 may also provide broad therapeutic benefits in the treatment of chronic degenerative diseases. However, a phase II trial for the treatment of stable rheumatoid arthritis (RA) did not meet its primary endpoint. Based on these data, the company focused development of MEK-162 solely in oncology.

In 2010, MEK-162 was licensed to Novartis by Array BioPharma for worldwide development. In 2013, orphan drug designation was assigned in Japan for the treatment of malignant melanoma with NRAS or BRAF V600 mutation.

WO-2014063024 DEALS WITH Preparation, crystalline forms, and formulations comprising binimetinib. Binimetinib is a MEK-1/2 inhibitor originally claimed in WO03077914, which Array and Novartis are developing for the treatment of cancer, including melanoma, low-grade serous ovarian cancer, and other solid tumors, as well as Noonan syndrome hypertrophic cardiomyopathy and hepatic impairment. See also WO2014018725 for the most recent filing on the agent

PATENT

WO 03/077914

http://www.google.com/patents/WO2003077914A1?cl=en

Schemes 1-4.

Scheme 1

Figure imgf000029_0001
Figure imgf000029_0002

Scheme la

Figure imgf000030_0001

Scheme 2

Figure imgf000031_0001

Scheme 3

Figure imgf000032_0001

17 18

Scheme 4

Figure imgf000033_0001

25

Scheme 5

Figure imgf000034_0001
Figure imgf000034_0002

General synthetic methods which may be referred to for preparing some of the compounds of the present invention are provided in PCT published application number WO 00/42022 (published July 20, 2000). The foregoing patent application is incorporated herein by reference in its entirety.

 similar ie chloro instead of fluoro

Example 52

Figure imgf000112_0001

6-(4-Bromo-2-chloro-phenylamino)-7-chloro-3-methyl-3H-benzoimidazole-5- carboxylic acid (2-hydroxy-ethoxy)-amide (lOcc) Step A: 3-Chloro-2,4-difluoro-5-nitro-benzoic acid 2a

3-Chloro-2,4-difluoro-benzoic acid la (3.00 g, 15.6 mmol) is added to a stirred solution of concentrated H2SO4 (16 mL) and fuming nitric acid (0.85 mL, 20.3 mmol). After 3 hours a precipitate forms. The yellow slurry is poured onto ice water (100 mL). The aqueous mixture is extracted with diethyl ether (3x). The organic extracts are dried (Na2SO4) and concentrated under reduced pressure to give 3.50 g (95%) of clean desired product as a pale yellow solid.

Step B: 4-Amino-3-chloro-2-fluoro-5-nitro-benzoic acid 3a

Ammonium hydroxide solution (6.88 g, -30% in water, 58.9 mmol) is added to a solution of 3-chloro-2,4-difluoro-5-nitro-benzoic acid 2a (3.5 g, 14.7 mmol) in water (16 mL) at 0 °C with stirring. Upon completion of the ammonium hydroxide addition the reaction mixture is warmed to room temperature. After 5 hours the reaction mixture is cooled to 0 °C and concentrated HCl is carefully added until the pH of the reaction mixture is near zero. The solid is collected by filtration and washed with water and diethyl ether. The solids are transferred to a round bottom flask as a solution in MeOH and EtOAc and concentrated under reduced pressure to give 2.96 g of a yellow solid. The filtrate is partitioned between diethyl ether and water and the organic layer is washed with brine. The combined organic extracts are dried (Na2SO ) and concentrated under reduced pressure to give 0.65 g of product. Recovered a total of 3.61 g (104%) of pure desired product, that is carried forward without further purification.

Step C: 4~Amino-3-chloro-2-fluoro-5-nitro-benzoic acid methyl ester 4a

To a stirred solution of 4-amino-3-chloro-2-fluoro-5-nitro-benzoic acid 3a (3.61 g, 15.4 mmol) in THF (30 mL) and MeOH (10 mL), TMS diazomethane (9.23 mL, 2.0 M solution in hexanes, 18.5 mmol) is added. After completion of reaction, the reaction mixture is concentrated via rotary evaporation with acetic acid in the trap. The recovered oily solid is triturated with diethyl ether to provide 1.51 g of a yellow solid. The filtrate is concentrated and triturated with diethyl ether to give an additional 0.69 g of yellow solid. A total of 2.20 g (57%) of pure desired product is recovered.

Step D: 4-Amino-3-chloro-5-nitro-2-phenylamino-benzoic acid methyl ester 5c

4-Amino-3-chloro-2-fluoro-5-nitro-benzoic acid methyl ester 4a (2.20 g, 8.84 mmol) is suspended in MeOH (9.4 mL) and aniline (3.22 mL, 35.4 mmol) is added. The reaction mixture is heated to reflux with stirring under a nitrogen atmosphere. After 19 hours, the reaction is complete. Distilled water (3.22 mL) is added to the reaction mixture and refluxing is continued for one hour. The reaction mixture is cooled to 0 °C in an ice bath for 20 minutes. The reaction mixture is filtered and washed with 3:10 distilled water/MeOH (65 mL total) and then with MeOH. The solid is dissolved with CH2C12 and concentrated under reduced pressure to give 2.40 g (84%) of pure desired product. MS APCI (-) m/z 320.3 (M-l) detected.

Step E: 4, 5-Diamino-3-chloro-2-phenylamino-benzoic acid methyl ester 6b

4-Amino-3-chloro-5-nitro-2-phenylamino-benzoic acid methyl ester 5c (0.50 g, 1.55 mmol) is dissolved into 2:1 EtOH/MeOH (15.5 mL). Saturated aqueous NH4C1 (15 mL), Zn powder (1.02 g, 15.6 mmol), and THF (10 mL) are added. After stirring for 20 hours, the reaction mixture is diluted with CH C12/THF and water. The organic layer is washed with water (3x). The combined organic extracts are dried (Na2SO4) and concentrated under reduced pressure. The solids are triturated with ether to give 0.32 g (70%) clean desired product. Step F: 7-Chloro-6-phenylamino-3H-benzoimidazole-5-carboxylic acid methyl ester 7c

4,5-Diamino-3-chloro-2-phenylamino-benzoic acid methyl ester 6b (0.32 g, 1.09 mmol) and formamidine acetate (72 mg, 1.64 mmol) in EtOH (36 mL) are heated, with stirring, to 80 °C. After 44 hours, the reaction mixture is cooled to room temperature and diluted with EtOAc and washed with water (3x), saturated NaHCO3, and brine. The combined organic extracts are dried (Na2SO4) and concentrated under reduced pressure to give 0.33 g (99%) clean desired product as a solid. MS APCI (+) m/z 302.3 (M+l) detected.

Step G: 6-(4-Bromo-phenylamino)-7-chloro-3H-benzoimidazole-5-carboxylic acid methyl ester 8g

7-Chloro-6-phenylamino-3H-benzoimidazole-5-carboxylic acid methyl ester 7c (0.327 g, 1.08 mmol) is dissolved into DMF (16 mL) and NBS (0.193 g, 1.08 mmol) is added. After one hour, the reaction mixture is quenched by the addition of saturated aqueous NaHSO3. The reaction mixture is then partitioned between EtOAc/THF and water. The organic layer is washed with water and brine. The combined organic extracts are dried (Na2SO ) and concentrated under reduced pressure. The recovered solid is triturated with ether to give 0.225 g (54%) pure desired product. MS ESI (+) m/z 382, 384 (M+, Br pattern) detected.

Step H: 6-(4-Bromo-2-chloro-phenylamino)- 7 -chloro-3H-benzoimidazole-5 -carboxylic acid methyl ester lOdd 6-(4-Bromo-phenylamino)-7-chloro-3H-benzoimidazole-5-carboxylic acid methyl ester 8g (0.225 g, 0.591 mmol) is dissolved in DMF (2 mL) and NCS (79 mg, 0.591 mmol) is added. After the NCS is in solution concentrated HCl (0.005 mL, 0.059 mmol) is added. After 2 hours, sodium bicarbonate, water and NaHSO3 are added to the reaction mixture. Solids are filtered and washed with water and ether to give 0.141 g (57%) of clean desired product as a tan solid. MS APCI (-) m/z 414, 416 (M-, Br pattern) detected.

Step I: 6-(4-Bromo-2-chloro-phenylamino)-7-chloro-3-methyl-3H-benzoimidazole-5- carboxylic acid methyl ester lOee

6-(4-Bromo-2-chloro-phenylamino)-7-chloro-3H-benzoimidazole-5-carboxylic acid methyl ester lOdd (0.141 g, 0.34 mmol), potassium carbonate (0.141 g, 1.02 mmol), and iodomethane (0.063 mL, 1.02 mmol) are dissolved in dimethylformamide (3 mL). After 20 hours, the reaction mixture is diluted with EtOAc and washed with water (3x), potassium carbonate, and brine. The organic layer is dried (Na2SO4) and concentrated to a brown oil. The N3 and Nl alkylated regioisomers are separated by flash chromatography (EtOAc). The recovery of the N3 alkylated regioisomer is 20.4 mg (28%). MS ESI (+) m/z 428, 430 (M+, Br pattern) detected.

Step J: 6-(4-Bromo-2-chloro-phenylamino)-7-chloro-3-methyl-3H-benzoimidazole-5- carboxylic acid 10 ff

6-(4-Bromo-2-chloro-phenylamino)-7-chloro-3-methyl-3H-benzoimidazole-5- carboxylic acid methyl ester lOee (21 mg, 0.048 mmol) is dissolved into 2:1 THF/water (1.2 mL) and NaOH (0.190 mL, 1.0 M aqueous solution, 0.190 mmol) is added. After stirring for 4 hours the reaction is diluted with water and acidified to pH 2 by addition of 1.0 M HCl. The mixture is then extracted with 3:1 EtOAc/THF (3x), dried (Na2SO ) and concentrated to give quantitative yield of desired prodcut as a white solid. MS APCI (+) m/z 414, 416 (M+, Br pattern) detected.

Step K: 6-(4-Bromo-2’chloro-phenylamino)- 7-chloro-3-methyl-3H-benzoimidazole-5- carboxylic acid (2-vinyloxy-ethoxy) -amide lOgg

6-(4-Bromo-2-chloro-phenylamino)-7-chloro-3-methyl-3H-benzoimidazole-5- carboxylic acid lOff (32 mg, 0.077 mmol), O-(2-vinyloxy-ethyl)-hydroxylamine (0.010 mL, 0.092 mmol), HOBt (13 mg, 0.093 mmol), triethylamine (0.011 mL, 0.077 mmol), and EDCI (19 mg, 0.10 mmol) are dissolved into dimethylformamide (1.0 mL) and allowed to stir under a nitrogen atmosphere at room temperature for 24 hours. The reaction mixture is diluted with EtOAc, washed with water (3x), 10% potassium carbonate (2x), saturated ammonium chloride, brine, dried (Na2SO4), and concentrated under reduced pressure to give 39 mg of 85% pure material. MS APCI (-) m/z 497, 501 (M-, Br pattern) detected.

Step L: 6-(4-Bromo-2-chloro-phenylamino)-7-chloro-3-methyl-3H-benzoimidazole-5- carboxylic acid (2-hydroxy-ethoxy)-amide lOcc

Hydrochloric acid (0.78 mL, 1.0 M aqueous solution, 0.78 mmol) is added to a suspension of 6-(4-bromo-2-chloro-phenylamino)-7-chloro-3-methyl-3H- benzoimidazole-5-carboxylic acid lOgg (2-vinyloxy-ethoxy)-amide (39 mg, 0.078 mmol) in MeOH (1 mL). After one hour, the reaction mixture is neutralized to pH 7 and concentrated under reduced pressure. The solids are dissolved in EtOAc, washed with brine, dried (Na SO4), and concentrated under reduced pressure. Flash chromatography (20:1 CH2Cl2/MeOH) provides 9 mg (23%) of pure product: MS APCI (+) m/z 473, 475 (M+, Br pattern) detected; 1H NMR (400 MHz, CDC13) δ 8.30 (s, IH), 8.08 (s, IH), 7.57

(d, IH), 7.15 (dd, IH), 6.21 (d, IH), 3.97 (s, 3H) 3.86 (m, 2H), 3.57 (m, 2H).

actual is below

Example 18

The following compounds are prepared by methods similar to those described in

Example 10 by using methyl ester 8d and the appropriate alkylating agent (Step A) and

the appropriate hydroxylamine (Step C):

Figure imgf000071_0002

PATENT

WO2014063024

http://patentscope.wipo.int/search/en/detail.jsf;jsessionid=E10680BCA177F821C7FEFA1AFC44A438.wapp2nA?docId=WO2014063024&recNum=6&maxRec=53841&office=&prevFilter=%26fq%3DICF_M%3A%22C07D%22&sortOption=Pub+Date+Desc&queryString=&tab=PCTDescription

COMPD A

Example 1. Preparation of 6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-

In an inertized (N2) reaction vessel at internal temperature 20°C and under exclusion of humidity and air, Compound 1 (1.0 eq.) and Compound 2 (1.2 eq.) are reacted in the presence of cesium carbonate (2.4 eq.), tris(dibenzylidenaceton) dipalladium(O) (0.035 eq.) and Xantphos (0.07 eq.) in a mixture of toluene and 1 ,4-dioxane at internal temperature of 99°C. After 8 hours, the mixture is cooled to internal temperature of 60°C.

Subsequently, dimethylformamide (DMF), filter aid (CEFOK) and activated charcoal (EKNS) are added, and the mixture is stirred and cooled to internal temperature of 35 °C. The solids are filtered off and washed with a mixture of dimethylformamide and toluene. To the filtrate, which contains the product Compound 3, is introduced at internal temperature of

25 °C hydrogen chloride gas (CLC) whereupon the HQ salt of Compound 3 crystallizes. The palladium residue mainly remains in solution. After warming to 60 °C and cooling to 0°C, the solids are filtered using a centrifuge and are washed with a mixture of toluene and dimethylformamide.

The damp Compound 3 HC1 salt is charged to a reactor (equipped with pH probe) together with dimethylformamide and is heated to 60°C. By adding a 4 wt% of aqueous tripotassium phosphate solution, the pH is adjusted to a pH range of 6.8-7.6 (with a target of pH 7.2) while Compound 3 crystallizes as free base. After cooling to 22°C and stirring, the solids are filtered using a centrifuge and are washed with drinking water. The moist solids are dried at 50 °C under vacuum to give dry, crude Compound 3.

In order to remove residual palladium, dry, crude Compound 3 is dissolved in dimethylformamide at internal temperature of 60°C and stirred together with Smopex-234 (commercially available from Johnson Matthey) and activated charcoal for 90 minutes. The solids are filtered off at internal temperature of 60°C and are washed with

dimethylformamide. To the filtrate are added drinking water and Compound 3 seed crystals. More drinking water is added while Compound 3 crystallizes. After cooling to internal temperature of 20 °C, the solids are filtered using a centrifuge and are washed with a mixture of deionized water and dimethylformamide and with deionized water. The moist solids are dried at 50°C under vacuum, providing 6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid methyl ester (Compound 3).

Example 2. Preparation of 6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid-(2-tert-butoxyethoxy)-amide

A. “One-pot” Synthesis


In an inertized reaction vessel at internal temperature 20-25 °C under nitrogen, 6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid methyl ester (Compound 3, 1.0 eq.) is added to a mixture of DMF and THF. To this slurry, a solution of potassium trimethylsilanolate (1.05 eq.) in THF is added to the mixture at internal temperature of 25 °C over a period of about 40 minutes, and the resulting mixture is stirred for about 1 hour, providing a potassium salt solution of Intermediate 1. A THF/methanol mixture is then sequentially distilled off from the mixture at 85-120°C during about 2 hours.

The potassium salt solution is then added to a suspension of CDI (1.25 eq.) and imidazole hydrochloride (1.40 eq.) in THF at internal temperature of 25 °C over a period of about 1 hour. The resulting mixture is then stirred for approximately 1 hour at 50°C, and the following imidazolide intermediate

The imidazolide intermediate is not further isolated.

Subsequently, 1.2 eq. of 0-(2-tert-butoxyethyl)hydroxylamine (Compound 4, CAS No. 1023742-13-3, available from suppliers such as Huhu Technology, Inc.®) is added over a period of about 30 minutes at 50°C and stirred for 1.5 hours. Demineralized water is then added at 50°C, producing a precipitate. After cooling to 20°C and stirring for about 3-16 hours, the slurry is filtered off, washed with THF/ demineralized water (1 :2) in 2 portions and with demineralized water in three portions, and dried at 50°C / <70 mbar for about 17 hours, providing 6-(4-bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid-(2-tert-butoxyethoxy)-amide (Compound 5) as monohydrate.

B. A synthesis method with isolation of the intermediate of step a) from the reaction mixture of step a) prior to the reaction of step b)

Alternatively, 6-(4-bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5 -carboxylic acid-(2-tert-butoxyethoxy)-amide (Compound 5) can be made by the synthesis method as shown below. Compound 3, which is a methyl ester, is first converted to a carboxylic acid, which is then isolated by a crystallization to form Compound

6. Compound 6 is then coupled with Compound 4 to form Compound 5 as monohydrate.

The crystallization step in this method removes starting materials such as Compound 1, process impurities, and the dba ligand from the prior catalyst before the coupling reaction with Compound 4, and at the same time maintains the overall yield of the synthesis.

6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-memy acid In an inertized (N2) reaction vessel at internal temperature of 60°C, Compound 3 (1.0 eq.) is dissolved in DMF and stirred with a fiber, which is sold under the trademark

SMOPEX 234, and activated charcoal for the removal of palladium to not more than 100 ppm. The fiber and activated charcoal are removed by filtration at 60°C and washed with DMF.

The filtrate (containing Compound 3) is transferred to a second inertized (N2) reaction vessel and cooled to an internal temperature of 30°C. A thin suspension can form at this point of time. 30% sodium hydroxide (1.1 eq.) and water (for rinsing) are added, and the resulting reaction mixture is vigorously stirred for 3 hours at an internal temperature of 30 °C. The methyl ester is saponified. Conversion is checked by an IPC (HPLC). As soon as the IPC criterion is met, a filter aid, which is sold under the trademark HYFLO, is added. The mixture is stirred for 15 minutes and then filtered at 30°C via a plate filter and polish filter to a third reaction inertized (N2) vessel.

An aqueous HC1 solution 7.5 % is added to the clear filtrate in the third vessel at an internal temperature of 30 °C until a pH value of 8 is reached. Then the solution is seeded at an internal temperature of 30°C with Compound 6, and an aqueous HC1 solution 7.5 % is added under vigorous stirring until a pH value of pH 2.8 is reached. The product gradually crystalizes. The suspension is cooled over 60 min to an internal temperature of 25 °C and

water is added. The suspension is stirred for at least 4 hours at an internal temperature of 25°C.

The resulting solid is collected by centrifugation or filtration. The filter cake is first washed with DMF/water 1 :1 (w/w) and then with water, discharged and dried in a vacuum at 50°C. The water content is controlled by IPC. The crystalline product Compound 6 is discharged as soon as the IPC criterion is met.

6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid- (2-tert-butoxyethoxy) – amide

An inertized (N2) reaction vessel is charged with Compound 6 (1.0 eq.), DMF, and

THF at room temperature. The suspension is heated to 25 °C under stirring with flow of nitrogen. After CDI (1.13 eq.) is added, the suspension can get thinner and slight evolution of gases can be observed. After the suspension finally becomes a solution, it is then monitored by IPC (HPLC).

As soon as the IPC (HPLC) criterion is met, the reaction mixture is heated to 50°C over 20 minutes and imidazole hydrochloride (0.3 eq.) is added, forming a solution of

Intermediate 2.

To the solution of Intermediate 2, Compound 4 (1.3 eq.) is added over 60 minutes at internal temperature of 50°C under stirring at a speed of 300 rpm with flow of nitrogen. As soon as the IPC (HPLC) criterion is met, the mixture is cooled to 20-25 °C over 30 minutes. The mixture is then stored at ambient temperature overnight under nitrogen without stirring. DMF is added to the mixture followed by heating it to 50 °C over 30 minutes. Complete conversion of Intermediate 2 to Compound 5 is confirmed by IPC (HPLC).

Water is added to the mixture at internal temperature of 50 °C over 20 minutes. Then the solution is seeded with Compound 5. After stirring at 50 °C for 60 minutes, more water is added to the suspension at 50 °C over 90 minutes. After vigorous stirring, the suspension is cooled to 20 °C over 2 hours and filtered. The filter cake is washed twice with THF/water (v/v: 1 :2) at 20 °C, and twice with water at 20 °C. Finally, the filter cake is dried at 50 °C under vacuum to provide 6-(4-bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid-(2-tert-butoxyethoxy)-amide (Compound 5) as monohydrate.

Example 3. Preparation of 6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxyethyoxy)-amide (Compound A)

Compound 5 Compound A

6-(4-Bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid-(2-tert-butoxyethoxy)-amide (Compound 5) monohydrate is added in 3 portions to a premixed solution of Acetonitrile and excess Phosphoric acid (85 % aqueous solution) at internal temperature 20-25 °C. After stirring for about 15 minutes, the suspension is heated to internal temperature 50-53 °C. The suspension is maintained at this temperature for 6 hours, cooled to internal temperature 20-25 °C. The mixture is then heated to internal temperature 35-37°C and diluted with Ethanol- Water (3 :1 v/v). EKNS and CEFOK are added, the reaction mixture is stirred approximately 15 minutes and filtered over a funnel coated with CEFOK. The filtrate is cooled to approximately 30°C. 3 N aqueous potassium hydroxide (ΚΟΗ) is added to the cooled filtrate over a period of 90 minutes until a pH- value of about 8.1 is reached. The suspension is heated to internal temperature 60-63 °C, stirred at this temperature for a period of about 2 hours, cooled to 20-23 °C over a period of about 45 minutes, filtered over a funnel, and dried at 50°C pressure <100 mbar over a period of about 17 hours, providing 6-(4-bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxyethyoxy)-amide (Compound A) as a white powder.

Example 4. Preparation of Crystallized 6-(4-bromo-2-fluorophenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxyethyoxy)-amide (Compound A) In a dry vessel at room temperature, Compound A is added to a premixed solvent solution of methanol/THF/water (35/35/30 w/w). The suspension is heated to internal temperature 53-55°C, and the resulting solution is hot filtered by deep and membrane filtration (via a paper filter and PTFE membrane) at internal temperature 53-56°C. The clear solution is stirred and cooled to 47-48°C, and the seed crystals suspension (i.e., seed crystals of crystallized Compound A in water, 10% m/m) is added (0.2 to 0.5% of crystallized Compound A expected yield mass). After about 20 minutes, water is slowly added within 25 hours (33.3% within 15 hours and 66.6% within 10 hours with at least 10 minute stirring after addition of water) to obtain a final ratio of methanol THF/water (20/20/60 w/w). After the water is added, the suspension is cooled down to internal temperature 3-5 °C within 10 hours and stirred for 0.5 hours. The white suspension is filtered over a sinter glass nutsche (75 ml, diameter = 6 cm, pore 3) suction filter and washed once with ice cold methanol/THF/water (15/15/70 w/w at 2-4 °C), and two times with ice cold water (2-4 °C). Drying takes place in a vacuum oven dryer at 20°C for 10 hours, and then at 40°C for 10 hours, and then at 60°C for at least 12 hours with pressure < lOmbar, providing crystallized Compound A.

CLIP

str1

http://blog.sina.com.cn/s/blog_de171b9b0101dvov.html

CLIP

https://www.pharmacodia.com/yaodu/html/v1/chemicals/675f9820626f5bc0afb47b57890b466e.html

References

  1. Jump up^ “Binimetinib”. Array Biopharma.
  2. Jump up to:a b Koelblinger P, Dornbierer J, Dummer R (August 2017). “A review of binimetinib for the treatment of mutant cutaneous melanoma”. Future Oncology13 (20): 1755–1766. doi:10.2217/fon-2017-0170PMID 28587477.
  3. Jump up to:a b Research, Center for Drug Evaluation and. “Approved Drugs – FDA approves encorafenib and binimetinib in combination for unresectable or metastatic melanoma with BRAF mutations”http://www.fda.gov. Retrieved 2018-07-17.
  4. Jump up^ Wu PK, Park JI (December 2015). “MEK1/2 Inhibitors: Molecular Activity and Resistance Mechanisms”Seminars in Oncology42 (6): 849–62. doi:10.1053/j.seminoncol.2015.09.023PMC 4663016Freely accessiblePMID 26615130.
  5. Jump up^ “Binimetinib”PubChem.
  6. Jump up^ Ascierto PA, Schadendorf D, Berking C, Agarwala SS, van Herpen CM, Queirolo P, Blank CU, Hauschild A, Beck JT, St-Pierre A, Niazi F, Wandel S, Peters M, Zubel A, Dummer R (March 2013). “MEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: a non-randomised, open-label phase 2 study”. The Lancet. Oncology14(3): 249–56. doi:10.1016/S1470-2045(13)70024-XPMID 23414587.
  7. Jump up^ Mehdizadeh A, Somi MH, Darabi M, Jabbarpour-Bonyadi M (February 2016). “Extracellular signal-regulated kinase 1 and 2 in cancer therapy: a focus on hepatocellular carcinoma”. Molecular Biology Reports43 (2): 107–16. doi:10.1007/s11033-016-3943-9PMID 26767647.
  8. Jump up to:a b Woodfield SE, Zhang L, Scorsone KA, Liu Y, Zage PE (March 2016). “Binimetinib inhibits MEK and is effective against neuroblastoma tumor cells with low NF1 expression”BMC Cancer16: 172. doi:10.1186/s12885-016-2199-zPMC 4772351Freely accessiblePMID 26925841.
  9. Jump up^ Clinical trial number NCT01849874 for “A Study of MEK162 vs. Physician’s Choice Chemotherapy in Patients With Low-grade Serous Ovarian, Fallopian Tube or Peritoneal Cancer” at ClinicalTrials.gov
  10. Jump up^ Clinical trial number NCT01909453 for “Study Comparing Combination of LGX818 Plus MEK162 Versus Vemurafenib and LGX818 Monotherapy in BRAF Mutant Melanoma (COLUMBUS)” at ClinicalTrials.gov
  11. Jump up^ Clinical trial number NCT01763164 for “Study Comparing the Efficacy of MEK162 Versus Dacarbazine in Unresectable or Metastatic NRAS Mutation-positive Melanoma” at ClinicalTrials.gov
  12. Jump up^ Hufford A (December 2015). “Array BioPharma Has Successful Trial for Cancer Drug Binimetinib”Wall Street Journal.
  13. Jump up^ “Array BioPharma announces Phase 3 binimetinib trial meets primary endpoint for NRAS-mutant melanoma”Metro Denver. December 2015.
  14. Jump up^ Array Bio submits marketing application in U.S. for lead product candidate in certain type of melanoma. June 2016
  15. Jump up^ House DW (1 September 2016). “FDA accepts Array Bio’s NDA for binimetinib, action date June 30”Seeking Alpha.
  16. Jump up^ House DW (1 April 2016). “Array bags Phase 3 study of binimetinib in ovarian cancer; shares down 4%”Seeking Alpha.
  17. Jump up^ Adams B (20 March 2017). “Losing Nemo: Array pulls skin cancer NDA for binimetinib”Fierce Biotech.
Binimetinib
Binimetinib.svg
Clinical data
ATC code
Legal status
Legal status
  • Investigational
Identifiers
CAS Number
PubChem CID
DrugBank
ChemSpider
KEGG
ChEMBL
Chemical and physical data
Formula C17H15BrF2N4O3
Molar mass 441.23 g/mol
3D model (JSmol)
  1. Koelblinger P, Dornbierer J, Dummer R: A review of binimetinib for the treatment of mutant cutaneous melanoma. Future Oncol. 2017 Aug;13(20):1755-1766. doi: 10.2217/fon-2017-0170. Epub 2017 Jun 7. [PubMed:28587477]
  2. Queirolo P, Spagnolo F: Binimetinib for the treatment of NRAS-mutant melanoma. Expert Rev Anticancer Ther. 2017 Nov;17(11):985-990. doi: 10.1080/14737140.2017.1374177. Epub 2017 Sep 8. [PubMed:28851243]
  3. Dummer R, Schadendorf D, Ascierto PA, Arance A, Dutriaux C, Di Giacomo AM, Rutkowski P, Del Vecchio M, Gutzmer R, Mandala M, Thomas L, Demidov L, Garbe C, Hogg D, Liszkay G, Queirolo P, Wasserman E, Ford J, Weill M, Sirulnik LA, Jehl V, Bozon V, Long GV, Flaherty K: Binimetinib versus dacarbazine in patients with advanced NRAS-mutant melanoma (NEMO): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2017 Apr;18(4):435-445. doi: 10.1016/S1470-2045(17)30180-8. Epub 2017 Mar 9. [PubMed:28284557]
  4. Bendell JC, Javle M, Bekaii-Saab TS, Finn RS, Wainberg ZA, Laheru DA, Weekes CD, Tan BR, Khan GN, Zalupski MM, Infante JR, Jones S, Papadopoulos KP, Tolcher AW, Chavira RE, Christy-Bittel JL, Barrett E, Patnaik A: A phase 1 dose-escalation and expansion study of binimetinib (MEK162), a potent and selective oral MEK1/2 inhibitor. Br J Cancer. 2017 Feb 28;116(5):575-583. doi: 10.1038/bjc.2017.10. Epub 2017 Feb 2. [PubMed:28152546]
  5. Gardner AM, Vaillancourt RR, Lange-Carter CA, Johnson GL: MEK-1 phosphorylation by MEK kinase, Raf, and mitogen-activated protein kinase: analysis of phosphopeptides and regulation of activity. Mol Biol Cell. 1994 Feb;5(2):193-201. [PubMed:8019005]
  6. Wang ZQ, Wu DC, Huang FP, Yang GY: Inhibition of MEK/ERK 1/2 pathway reduces pro-inflammatory cytokine interleukin-1 expression in focal cerebral ischemia. Brain Res. 2004 Jan 16;996(1):55-66. [PubMed:14670631]
  7. Cancer.gov link [Link]
  8. FDA approves encorafenib and binimetinib in combination for unresectable or metastatic melanoma with BRAF mutations [Link]
  9. A phase 1 dose-escalation and expansion study of binimetinib (MEK162), a potent and selective oral MEK1/2 inhibitor [Link]
  10. Binimetinib inhibits MEK and is effective against neuroblastoma tumor cells with low NF1 expression [Link]
  11. Binimetinib [File]
  12. EMA assessment [File]

/////////////BINIMETINIB, FDA 2018, MEK-162, биниметиниб بينيميتينيب , 美替尼 , ビニメチニブ , 606143-89-9 , 9764, ARRY-162, ARRY-438162, NVP-MEK162

CN1C=NC2=C(F)C(NC3=CC=C(Br)C=C3F)=C(C=C12)C(=O)NOCCO

https://cen.acs.org/articles/95/i23/Array-licenses-cancer-compounds-Ono.html

The structure of binimetinib.

Array BioPharma has licensed Japan’s Ono Pharmaceutical the right to develop two late-stage oncology compounds, binimetinib and encorafenib, in Japan and South Korea. Array will get $32 million up front and up to $156 million in milestone payments. The compounds are in Phase III studies of patients with BRAF-mutant cancers. Array recently struck a deal to assess binimetinib with two Bristol-Myers Squibb immuno-oncology agents.

Amfonelic acid, амфонеловая кислота , حمض أمفونيليك , 安福萘酸 , アンホネル酸

$
0
0

Amfonelic acid.pngChemSpider 2D Image | amfonelic acid | C18H16N2O3Amfonelic acid.png

Amfonelic acid

  • Molecular FormulaC18H16N2O3
  • Average mass308.331 Da
1,8-Naphthyridine-3-carboxylic acid, 1-ethyl-1,4-dihydro-4-oxo-7-(phenylmethyl)-
15180-02-6 [RN]
1-Ethyl-1,4-dihydro-4-oxo-7-(phenylmethyl)-1,8-naphthyridine-3-carboxylic Acid
2324
RR302AR19Y
NSC 100638
амфонеловая кислота [Russian] [INN]
حمض أمفونيليك [Arabic] [INN]
安福萘酸 [Chinese] [INN]
Lopac0_000416
MFCD00055095 [MDL number]
NCA
NSC-100638
UNII:RR302AR19Y
UNII-RR302AR19Y
Win 25,978

Amfonelic acid (AFAWIN 25,978) is a research chemical and dopaminergic stimulant with antibiotic properties.[1]

History

The stimulant properties of AFA were discovered serendipitously at Sterling-Winthrop in the midst of research on the antibiotic nalidixic acid.[1] In addition to behaving as antibiotics, it was found that many derivatives of nalidixic acid have either stimulant or depressant effects on the central nervous system.[2] Researchers at Sterling-Winthrop found that AFA had a higher potency and therapeutic indexthan cocaine or amphetamine and so it was singled out for further study.[1][3] A small number of clinical trials were held in the 1970s, but when it was found that AFA exacerbated psychotic symptoms in schizophrenic patients and produced undesirable stimulant properties in geriatric depressives clinical evaluation of AFA was discontinued.[1] AFA remains a widely used pharmacological tool for study of the brain’s reward systemdopamine pathways, and the dopamine transporter.[1] Since 2013 AFA has been sold on the gray market and there are numerous anecdotal reports detailing its non-medical use.[1]

Pharmacology

In studies it proved to be a potent and highly selective dopamine reuptake inhibitor (DRI) in rat brain preparations.[4][5] A study found a moderately long half-life of approximately 12 hours and a dopaminergic potency approximately 50 fold that of methylphenidate in rat brain preparations.[6] Despite lack of direct serotonin activity, rats treated with subchronic doses of amfonelic acid display subsequent decreases in 5HT and 5HIAA.[7] Amfonelic acid displays no activity in the norepinephrine system.[8]

Despite its different mechanism of action, amfonelic acid displays discriminatory substitution with 150% the stimulant potency of dextroamphetamine.[9] Amfonelic acid has been shown to be neuroprotective against methamphetamine damage to dopamine neurons.[10] It also increases the effects of the antipsychotic drugs haloperidoltrifluoperazine and spiperone.[11] Rats are shown to self-administer amfonelic acid in a dose-dependent manner.[12]

Though AFA was discovered in the course of antibiotic research, there is very little data available on the drug’s antimicrobial activity. In 1988 the biologist G.C. Crumplin wrote, “[AFA] is less active against bacteria than are many other 4-quinolones, but studies in our laboratory on selected mammalian cell lines have shown it to be markedly more toxic to these cells than are the 4-quinolones that are more active antibacterial agents. Furthermore, it can be shown that sublethal doses induced marked changes in the pattern of proteins produced by the cell, thus suggesting a possible effect of 4-quinolones on gene transcription in mammalian cells.”[13] When evaluated via broth microdilution the MIC of AFA for Escherichia coli is 125 μg/mL, a concentration thirty times higher than the MIC for nalidixic acid in the same E. coli strain.[1]

References

  1. Jump up to:a b c d e f g Morris, Hamilton (October 2015). “Sad Pink Monkey Blues”. Harper’s Magazine. Retrieved 2015-09-19.
  2. Jump up^ US patent 3590036, “Naphthyridine-3-carboxylic Acids, Their Derivatives and Preparation Thereof”
  3. Jump up^ Aceto, M.A. (1970). “Pharmacologic properties and mechanism of action of amfonelic acid”. European Journal of Pharmacology10: 344–354. doi:10.1016/0014-2999(70)90206-2PMID 4393073.
  4. Jump up^ Fuller, R. W.; Perry, K. W.; Bymaster, F. P.; Wong, D. T. (1978). “Comparative effects of pemoline, amfonelic acid and amphetamine on dopamine uptake and release in vitro and on brain 3,4-dihydroxyphenylacetic acid concentration in spiperone-treated rats”. Journal of Pharmacy and Pharmacology30 (3): 197–198. doi:10.1111/j.2042-7158.1978.tb13201.xPMID 24701.
  5. Jump up^ McMillen, B. A.; Shore, P. A. (1978). “Amfonelic acid, a non-amphetamine stimulant, has marked effects on brain dopamine metabolism but not noradrenaline metabolism: Association with differences in neuronal storage systems”. Journal of Pharmacy and Pharmacology30 (7): 464–466. doi:10.1111/j.2042-7158.1978.tb13293.xPMID 27622.
  6. Jump up^ Izenwasser, S.; Werling, L. L.; Cox, B. M. (1990). “Comparison of the effects of cocaine and other inhibitors of dopamine uptake in rat striatum, nucleus accumbens, olfactory tubercle, and medial prefrontal cortex”. Brain Research520 (1–2): 303–309. doi:10.1016/0006-8993(90)91719-WPMID 2145054.
  7. Jump up^ McMillen, BA; Scott, SM; Williams, HL (1991). “Effects of subchronic amphetamine or amfonelic acid on rat brain dopaminergic and serotonergic function”. Journal of neural transmission. General section83 (1–2): 55–66. doi:10.1007/BF01244452PMID 2018630.
  8. Jump up^ Agmo, A; Belzung, C; Rodríguez, C (1997). “A rat model of distractibility: Effects of drugs modifying dopaminergic, noradrenergic and GABAergic neurotransmission”. Journal of neural transmission (Vienna, Austria : 1996)104 (1): 11–29. doi:10.1007/BF01271291PMID 9085190.
  9. Jump up^ Aceto, MD; Rosecrans, JA; Young, R; Glennon, RA (1984). “Similarity between (+)-amphetamine and amfonelic acid”. Pharmacology Biochemistry and Behavior20 (4): 635–7. doi:10.1016/0091-3057(84)90316-2PMID 6728880.
  10. Jump up^ Pu, C; Fisher, JE; Cappon, GD; Vorhees, CV (1994). “The effects of amfonelic acid, a dopamine uptake inhibitor, on methamphetamine-induced dopaminergic terminal degeneration and astrocytic response in rat striatum”. Brain Research649 (1–2): 217–24. doi:10.1016/0006-8993(94)91067-7PMID 7953636.
  11. Jump up^ Waldmeier, PC; Huber, H; Heinrich, M; Stoecklin, K (1985). “Discrimination of neuroleptics by means of their interaction with amfonelic acid: An attempt to characterize the test”. Biochemical Pharmacology34 (1): 39–44. doi:10.1016/0006-2952(85)90097-8PMID 2857083.
  12. Jump up^ Porrino, LJ; Goodman, NL; Sharpe, LG (1988). “Intravenous self-administration of the indirect dopaminergic agonist amfonelic acid by rats”. Pharmacology Biochemistry and Behavior31 (3): 623–6. doi:10.1016/0091-3057(88)90240-7PMID 2908003.
  13. Jump up^ Crumplin, G.C. (1988). “Aspects of Chemistry in the Development of the 4-Quinolone Antibacterial Agents”. Reviews of Infectious Diseases. 10 Suppl 1 (10): S2–S9. doi:10.1093/clinids/10.Supplement_1.S2PMID 3279494.

External links

Amfonelic acid
Amfonelic acid.png
Clinical data
ATC code
  • none
Legal status
Legal status
  • In general: uncontrolled
Identifiers
CAS Number
PubChem CID
ChemSpider
UNII
KEGG
ChEMBL
Chemical and physical data
Formula C18H16N2O3
Molar mass 308.3329 g/mol
3D model (JSmol)

////////////Amfonelic acid, RR302AR19Y, амфонеловая кислота حمض أمفونيليك 安福萘酸 , アンホネル酸

CCN1C=C(C(=O)C2=C1N=C(C=C2)CC3=CC=CC=C3)C(=O)O


Inotersen sodium, イノテルセンナトリウム

$
0
0

Inotersen sodium.png

2D chemical structure of 1432726-13-0

Inotersen sodium,

UNII: 950736UC77

Data Source for DNA, d(P-thio)((2′-O-(2-methoxyethyl))m5rU-(2′-O-(2-methoxyethyl))m5rC-(2′-O-(2-methoxyethyl))m5rU-(2′-O-(2-methoxyethyl))m5rU-(2′-O-(2-methoxyethyl))rG-G-T-T-A-m5C-A-T-G-A-A-(2′-O-(2-methoxyethyl))rA-(2′-O-(2-methoxyethyl))m5rU-(2′-O-(2-methoxyethyl))m5rC-(2′-O-(2-methoxyethyl))m5rC-(2′-O-(2-methoxyethyl))m5rC)

STR  https://chem.nlm.nih.gov/chemidplus/rn/1432726-13-0

IUPAC Condensed

Thy-MeOEt(-2)Ribf-sP-m5Cyt-MeOEt(-2)Ribf-sP-Thy-MeOEt(-2)Ribf-sP-Thy-MeOEt(-2)Ribf-sP-Gua-MeOEt(-2)Ribf-sP-dGuo-sP-dThd-sP-dThd-sP-dAdo-sP-m5Cyt-dRibf-sP-dAdo-sP-dThd-sP-dGuo-sP-dAdo-sP-dAdo-sP-Ade-MeOEt(-2)Ribf-sP-Thy-MeOEt(-2)Ribf-sP-m5Cyt-MeOEt(-2)Ribf-sP-m5Cyt-MeOEt(-2)Ribf-sP-m5Cyt-MeOEt(-2)Ribf.19Na+

IUPAC

O2′-(2-methoxyethyl)-5-methyl-P-thio-uridylyl-(3′->5′)-O2′-(2-methoxyethyl)-5-methyl-P-thio-cytidylyl-(3′->5′)-O2′-(2-methoxyethyl)-5-methyl-P-thio-uridylyl-(3′->5′)-O2′-(2-methoxyethyl)-5-methyl-P-thio-uridylyl-(3′->5′)-O2′-(2-methoxyethyl)-P-thio-guanylyl-(3′->5′)-2′-deoxy-P-thio-guanylyl-(3′->5′)-P-thio-thymidylyl-(3′->5′)-P-thio-thymidylyl-(3′->5′)-2′-deoxy-P-thio-adenylyl-(3′->5′)-2′-deoxy-5-methyl-P-thio-cytidylyl-(3′->5′)-2′-deoxy-P-thio-adenylyl-(3′->5′)-P-thio-thymidylyl-(3′->5′)-2′-deoxy-P-thio-guanylyl-(3′->5′)-2′-deoxy-P-thio-adenylyl-(3′->5′)-2′-deoxy-P-thio-adenylyl-(3′->5′)-O2′-(2-methoxyethyl)-P-thio-adenylyl-(3′->5′)-O2′-(2-methoxyethyl)-5-methyl-P-thio-uridylyl-(3′->5′)-O2′-(2-methoxyethyl)-5-methyl-P-thio-cytidylyl-(3′->5′)-O2′-(2-methoxyethyl)-5-methyl-P-thio-cytidylyl-(3′->5′)-O2′-(2-methoxyethyl)-5-methyl-cytidine sodium salt

イノテルセンナトリウム
Formula
C230H299N69O121P19S19. 19Na
Cas
1432726-13-0
Antisense oligonucleotide; TTR mRNA
Mol weight
7600.7669
  • ClassAntisense oligonucleotides; Neuroprotectants
  • Mechanism of ActionPrealbumin expression inhibitors
  • Orphan Drug StatusYes – Amyloid polyneuropathy
  • New Molecular Entity Yes

Highest Development Phases

  • RegisteredAmyloid polyneuropathy
  • Phase IIAmyloidosis; Cardiomyopathies

Most Recent Events

  • 07 Aug 2018PTC Therapeutics announces intention to submit regulatory application in Latin America
  • 06 Aug 2018Akcea Therapeutics intends to launch inotersen in Germany after Summer 2018
  • 02 Aug 2018Inotersen licensed to PTC Therapeutics in Latin America

UNII-950736UC77; 950736UC77; Inotersen sodium; Inotersen sodium [USAN]; ISIS 420915 salt; 1432726-13-0

////////////////Inotersen sodium, eu 2018, イノテルセンナトリウム ,

SMILES

[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].COCCO[C@@H]1[C@H](O)[C@@H](COP(=O)([S-])O[C@@H]2[C@@H](COP(=O)([S-])O[C@@H]3[C@@H](COP(=O)([S-])O[C@@H]4[C@@H](COP(=O)([S-])O[C@@H]5[C@@H](COP(=O)([S-])O[C@H]6C[C@@H](O[C@@H]6COP(=O)([S-])O[C@H]7C[C@@H](O[C@@H]7COP(=O)([S-])O[C@H]8C[C@@H](O[C@@H]8COP(=O)([S-])O[C@H]9C[C@@H](O[C@@H]9COP(=O)([S-])O[C@H]%10C[C@@H](O[C@@H]%10COP(=O)([S-])O[C@H]%11C[C@@H](O[C@@H]%11COP(=O)([S-])O[C@H]%12C[C@@H](O[C@@H]%12COP(=O)([S-])O[C@H]%13C[C@@H](O[C@@H]%13COP(=O)([S-])O[C@H]%14C[C@@H](O[C@@H]%14COP(=O)([S-])O[C@H]%15C[C@@H](O[C@@H]%15COP(=O)([S-])O[C@@H]%16[C@@H](COP(=O)([S-])O[C@@H]%17[C@@H](COP(=O)([S-])O[C@@H]%18[C@@H](COP(=O)([S-])O[C@@H]%19[C@@H](COP(=O)([S-])O[C@@H]%20[C@@H](CO)O[C@H]([C@@H]%20OCCOC)N%21C=C(C)C(=O)NC%21=O)O[C@H]([C@@H]%19OCCOC)N%22C=C(C)C(=NC%22=O)N)O[C@H]([C@@H]%18OCCOC)N%23C=C(C)C(=O)NC%23=O)O[C@H]([C@@H]%17OCCOC)N%24C=C(C)C(=O)NC%24=O)O[C@H]([C@@H]%16OCCOC)n%25cnc%26C(=O)NC(=Nc%25%26)N)n%27cnc%28C(=O)NC(=Nc%27%28)N)N%29C=C(C)C(=O)NC%29=O)N%30C=C(C)C(=O)NC%30=O)n%31cnc%32c%31ncnc%32N)N%33C=C(C)C(=NC%33=O)N)n%34cnc%35c%34ncnc%35N)N%36C=C(C)C(=O)NC%36=O)n%37cnc%38C(=O)NC(=Nc%37%38)N)n%39cnc%40c%39ncnc%40N)n%41cnc%42c%41ncnc%42N)O[C@H]([C@@H]5OCCOC)n%43cnc%44c%43ncnc%44N)O[C@H]([C@@H]4OCCOC)N%45C=C(C)C(=O)NC%45=O)O[C@H]([C@@H]3OCCOC)N%46C=C(C)C(=NC%46=O)N)O[C@H]([C@@H]2OCCOC)N%47C=C(C)C(=NC%47=O)N)O[C@H]1N%48C=C(C)C(=NC%48=O)N

CC1=CN(C(=O)NC1=O)C2CC(C(O2)COP(=S)([O-])OC3CC(OC3COP(=S)([O-])OC4CC(OC4COP(=S)([O-])OC5C(OC(C5OCCOC)N6C=NC7=C6N=C(NC7=O)N)COP(=S)([O-])OC8C(OC(C8OCCOC)N9C=C(C(=O)NC9=O)C)COP(=S)([O-])OC1C(OC(C1OCCOC)N1C=C(C(=O)NC1=O)C)COP(=O)(OC1C(OC(C1OCCOC)N1C=C(C(=NC1=O)N)C)COP(=S)([O-])OC1C(OC(C1OCCOC)N1C=C(C(=O)NC1=O)C)CO)[S-])N1C=NC2=C1N=C(NC2=O)N)N1C=C(C(=O)NC1=O)C)OP(=S)([O-])OCC1C(CC(O1)N1C=NC2=C1N=CN=C2N)OP(=S)([O-])OCC1C(CC(O1)N1C=C(C(=NC1=O)N)C)OP(=S)([O-])OCC1C(CC(O1)N1C=NC2=C1N=CN=C2N)OP(=S)([O-])OCC1C(CC(O1)N1C=C(C(=O)NC1=O)C)OP(=S)([O-])OCC1C(CC(O1)N1C=NC2=C1N=C(NC2=O)N)OP(=S)([O-])OCC1C(CC(O1)N1C=NC2=C1N=CN=C2N)OP(=S)([O-])OCC1C(CC(O1)N1C=NC2=C1N=CN=C2N)OP(=S)([O-])OCC1C(C(C(O1)N1C=NC2=C1N=CN=C2N)OCCOC)OP(=S)([O-])OCC1C(C(C(O1)N1C=C(C(=O)NC1=O)C)OCCOC)OP(=S)([O-])OCC1C(C(C(O1)N1C=C(C(=NC1=O)N)C)OCCOC)OP(=S)([O-])OCC1C(C(C(O1)N1C=C(C(=NC1=O)N)C)OCCOC)OP(=S)([O-])OCC1C(C(C(O1)N1C=C(C(=NC1=O)N)C)OCCOC)O.[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+]

Plazomicin sulfate, プラゾマイシン硫酸塩 ,

$
0
0

File:Plazomicin flat.svgPlazomicin structure.svgChemSpider 2D Image | Plazomicin | C25H48N6O10

Plazomicin

  • Molecular FormulaC25H48N6O10
  • Average mass592.683 Da
(2S)-4-Amino-N-[(1R,2S,3S,4R,5S)-5-amino-4-{[(2S,3R)-3-amino-6-{[(2-hydroxyéthyl)amino]méthyl}-3,4-dihydro-2H-pyran-2-yl]oxy}-2-{[3-désoxy-4-C-méthyl-3-(méthylamino)-β-L-arabinopyranosyl]oxy}-3-hyd roxycyclohexyl]-2-hydroxybutanamide [French][ACD/IUPAC Name]
1154757-24-0 [RN]
9522
ACHN-490

1380078-95-4.pngPlazomicin sulfate.png

Image result for Plazomicin sulfateImage result for Plazomicin sulfateImage result for Plazomicin sulfate

Plazomicin Sulfate

Molecular Formula: C25H50N6O14S
Molecular Weight: 690.763 g/mol
Plazomicin Sulfate; UNII-A78L6MT746; Plazomicin Sulfate [USAN]; A78L6MT746; 1380078-95-4; Plazomicin sulfate (USAN),
  • ACHN 490 sulfate

6′-(hydroxylethyl)-1-(haba)-sisomicin

Plazomicin is a neoglycoside antibiotic with activity against a broad range of Gram-positive and Gram-negive pathogens. Plazomicin showed potent in vitro activity against multidrug-resistant Klebsiella pneumoniae and Escherichia coli.

  • Mechanism of ActionProtein synthesis inhibitors
  • Orphan Drug StatusNo
  • New Molecular EntityYes

Highest Development Phases

  • MarketedUrinary tract infections
  • RegisteredPyelonephritis
  • PreregistrationBacteraemia; Nosocomial pneumonia
  • PreclinicalGram-negative infections
  • No development reportedRespiratory tract infections; Tularaemia; Yersinia infections

Most Recent Events

  • 27 Jun 2018Registered for Pyelonephritis (Treatment-resistant) in USA (IV)- First Global Approval
  • 27 Jun 2018Registered for Urinary tract infections (Treatment-resistant) in USA (IV)- First Global Approval
  • 26 Jun 2018Achaogen receives complete response letter from the FDA for Plazomicin in Bloodstream infection
Synonyms:   O-2-Amino-2,3,4,6-tetradeoxy-6-[(2-hydroxyethyl)amino]-α-D-glycero-hex-4-enopyranosyl-(1→4)-O-[3-deoxy-4-C-methyl-3-(methylamino)-β-L-arabinopyranosyl-(1→6)]-N1-[(2S)-4-amino-2-hydroxy-1-oxobutyl]-2-deoxy-D-streptamine; ACHN 490;
CAS Number:   1154757-24-0

Sulfate 1380078-95-4, プラゾマイシン硫酸塩;

Achaogen (USA)Phase II completed
Mol. Formula:   C25H48N6O10
Aminoglycosides, Broad-spectrum,
Mol. Weight:   592.68

FDA

https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/210303Orig1s000lbl.pdf

str1

Developed by Achaogen biopharmaceuticals, plazomicin is a next-generation aminoglycoside synthetically derived from [DB12604]. The structure of plazomicin was established via appending hydroxylaminobutyric acid to [DB12604] at position 1 and 2-hydroxyethyl group at position 6′ [A33942]. It was designed to evade all clinically relevant aminoglycoside-modifying enzymes, which contribute to the main resistance mechanism for aminoglycoside therapy [A33942]. However, acquired resistance of aminoglycosides may arise through over expression of efflux pumps and ribosomal modification by bacteria, which results from amino acid or rRNA sequence mutations [A33942]. Like other aminoglycosides, plazomicin is ineffective against bacterial isolates that produce 16S rRNA methyltransferases [FDA Label]. Plazomicin mediates the antibacterial activity against pathogens including carbapenem-resistant (CRE) and extended-spectrum beta-lactamase (ESBL) producing _Enterobacteriaceae_. It mediates the antibacterial activity by binding to bacterial 30S ribosomal subunit and inhibiting protein synthesis [FDA Label]. On June 28th, 2018, plazomicin sulfate was approved by the FDA for use in adult patients for the treatment of complicated urinary tract infections (cUTI) including Pyelonephritis. It is marketed as Zemdri and is administered via once-daily intravenous infusion.

Developed by Achaogen biopharmaceuticals, plazomicin is a next-generation aminoglycoside synthetically derived from Sisomicin. The structure of plazomicin was established via appending hydroxylaminobutyric acid to Sisomicin at position 1 and 2-hydroxyethyl group at position 6′ [1]. It was designed to evade all clinically relevant aminoglycoside-modifying enzymes, which contribute to the main resistance mechanism for aminoglycoside therapy [1]. However, acquired resistance of aminoglycosides may arise through over expression of efflux pumps and ribosomal modification by bacteria, which results from amino acid or rRNA sequence mutations [1]. Like other aminoglycosides, plazomicin is ineffective against bacterial isolates that produce 16S rRNA methyltransferases [Label]. Plazomicin mediates the antibacterial activity against pathogens including carbapenem-resistant (CRE) and extended-spectrum beta-lactamase (ESBL) producing Enterobacteriaceae. It mediates the antibacterial activity by binding to bacterial 30S ribosomal subunit and inhibiting protein synthesis [Label]. On June 28th, 2018, plazomicin sulfate was approved by the FDA for use in adult patients for the treatment of complicated urinary tract infections (cUTI) including Pyelonephritis. It is marketed as Zemdri and is administered via once-daily intravenous infusion.

Plazomicin (INN,[1] ZEMDRI) is a next-generation aminoglycoside (“neoglycoside”) antibacterial derived from sisomicin by appending a hydroxy-aminobutyric acid (HABA) substituent at position 1 and a hydroxyethyl substituent at position 6′.[2][3]

Plazomicin has been reported to demonstrate in vitro synergistic activity when combined with daptomycin or ceftobiprole versus methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant S. aureus (VRSA) and against Pseudomonas aeruginosawhen combined with cefepimedoripenemimipenem or piperacillin/tazobactam.[3] It also demonstrates potent in vitro activity versus carbapenem-resistant Acinetobacter baumannii.[4]

In 2012, U.S. Food and Drug Administration granted fast track designation for the development and regulatory review of plazomicin.[5]

It is being developed by Achaogen, Inc. to treat serious bacterial infections due to multidrug-resistant Enterobacteriaceae, including carbapenem-resistant Enterobacteriaceae (CRE)[6] and was in Phase III clinical trials as of April 7, 2016.[7]

In June 2018 the FDA approved plazomicin (ZEMDRI) for adults with complicated urinary tract infections (cUTI), including pyelonephritis, caused by Escherichia coliKlebsiella pneumoniaeProteus mirabilis, or Enterobacter cloacae, in patients who have limited or no alternative treatment options. Zemdri is an intravenous infusion, administered once daily.[8][9] The FDA declined approval for treating bloodstream infections due to lack of effectiveness.[10]

To continue the development of plazomicin, the company has received a contract option of US$ 60M from the Biomedical Advanced Research and Development Authority (BARDA) to support a global Phase III clinical study. The study will evaluate plazomicin in treating patients with serious Gram-negative bacterial infections due to carbapenem-resistant Enterobacteriaceae. The study is expected to start in the fourth quarter of 2013 [4].

PATENT

WO 2009067692

WO 2010132770

PAPER

Synthesis and spectrum of the neoglycoside ACHN-490
Antimicrobial Agents and Chemotherapy (2010), 54, (11), 4636-4642

https://aac.asm.org/content/54/11/4636

FIG. 1.

FIG. 2.

FIG. 3.

PAPER

Plazomicin Retains Antibiotic Activity against Most Aminoglycoside Modifying Enzymes
ACS Infectious Diseases (2018), 4, (6), 980-987.

https://pubs.acs.org/doi/abs/10.1021/acsinfecdis.8b00001

PAPER

Effects of the 1-N-(4-Amino-2S-hydroxybutyryl) and 6′-N-(2-Hydroxyethyl) Substituents on Ribosomal Selectivity, Cochleotoxicity, and Antibacterial Activity in the Sisomicin Class of Aminoglycoside Antibiotics
ACS Infectious Diseases (2018), 4, (7), 1114-1120.

https://pubs.acs.org/doi/abs/10.1021/acsinfecdis.8b00052

Abstract Image

Syntheses of the 6′-N-(2-hydroxyethyl) and 1-N-(4-amino-2S-hydroxybutyryl) derivatives of the 4,6-aminoglycoside sisomicin and that of the doubly modified 1-N-(4-amino-2S-hydroxybutyryl)-6′-N-(2-hydroxyethyl) derivative known as plazomicin are reported together with their antibacterial and antiribosomal activities and selectivities. The 6′-N-(2-hydroxyethyl) modification results in a moderate increase in prokaryotic/eukaryotic ribosomal selectivity, whereas the 1-N-(4-amino-2S-hydroxybutyryl) modification has the opposite effect. When combined in plazomicin, the effects of the two groups on ribosomal selectivity cancel each other out, leading to the prediction that plazomicin will exhibit ototoxicity comparable to those of the parent and the current clinical aminoglycoside antibiotics gentamicin and tobramycin, as borne out by ex vivo studies with mouse cochlear explants. The 6′-N-(2-hydroxyethyl) modification restores antibacterial activity in the presence of the AAC(6′) aminoglycoside-modifying enzymes, while the 1-N-(4-amino-2S-hydroxybutyryl) modification overcomes resistance to the AAC(2′) class but is still affected to some extent by the AAC(3) class. Neither modification is able to circumvent the ArmA ribosomal methyltransferase-induced aminoglycoside resistance. The use of phenyltriazenyl protection for the secondary amino group of sisomicin facilitates the synthesis of each derivative and their characterization through the provision of sharp NMR spectra for all intermediates.

https://pubs.acs.org/doi/suppl/10.1021/acsinfecdis.8b00052/suppl_file/id8b00052_si_001.pdf

4 (19 mg, 40%). [α]D 25 = +46.5 (c = 0.01, H2O);

1 H NMR (600 MHz, D2O): δ 5.51 ( s, 1H, H-1ʹ), 5.16 (t, J = 3.5 Hz, H, H-4ʹ), 4.99 (d , J = 4.0 Hz, 1H, H-1ʹʹ), 4.11 (dd , J =9.4 Hz, 3.9 Hz, 1H, CH(OH)CH2CH2), 4.00 (d , J = 12.8 Hz, 1H, H-5ʹʹ), 3.99-3.93 (m, 1H, H-1), 3.84 (dd, J = 11.0 Hz, 4.0 Hz, 1H, H-2ʹʹ), 3.81 (t, J = 9.9 Hz, 1H, H-4), 3.77 (t, J = 5.3 Hz, 1H, H-2ʹ), 3.71 (t, J = 5.1 Hz, 2H, NHCH2CH2O), 3.69 – 3.65 (m, 2H, H-6, H-6ʹ), 3.64 – 3.44 (m , 2H, H-5, H-6ʹ), 3.35 – 3.26 (m , 1H, H-3), 3.24 (d, J = 12.8 Hz, 1H, H-5ʹʹ), 3.15 (d, J = 11.0 Hz, 1H, H-3ʹʹ), 3.09 – 3.06 (m, 2H, NHCH2CH2O), 3.01 (t, J = 7.2 Hz, 2H, CH(OH)CH2CH2), 2.74 (s, 3H, NCH3), 2.58 – 2.52 (m, 1H, H-3ʹ), 2.29 – 2.24 (m, 1H, H-3ʹ), 2.07 (dt, J = 13.2 Hz, 4.4 Hz, 1H, H-2), 2.04 – 1.98 (m, 1H, CH(OH)CH2CH2), 1.84 – 1.79 (m, 1H, CH(OH)CH2CH2), 1.64 (q, 1H, J = 12.5 Hz, H-2), 1.17 (s, 3H, 4ʹʹ-CH3);

13C NMR (151 MHz, D2O): δ 181.2 (s, CH3COOH), 175.4 (s, NHCO), 141.7 (s, C-5ʹ), 102.5 (s, C-4ʹ), 98.0 (s, C-1ʹʹ), 96.9 (s, C-1ʹ), 79.8 (s, C-4), 78.8 (s, C-6), 73.8 (s, C-5), 69.8 (s, C-4ʹʹ), 69.4 (s, CH(OH)CH2CH2), 66.8 (s, C-5ʹʹ), 65.9 (s, C-2ʹʹ), 64.2 (s, C-3ʹʹ), 56.4 (s, NHCH2CH2O), 48.8 (s, C-1), 48.31 (s, NHCH2CH2O), 48.26 (s, C-3), 47.9 (s, C-6ʹ), 45.9 (s, C2ʹ), 36.8 (s, CH(OH)CH2CH2), 34.9 (s, NCH3), 30.7 (s, CH(OH)CH2CH2), 30.4 (s, C-2), 23.1 (s, CH3COOH), 23.0 (s, C-3ʹ), 20.8 (s, 4ʹʹ-CH3).

ESI-HRMS: m/z calcd. for C25H49N6O10 [M+H]+ 593.3510, found: 593.3481.

PATENT

http://www.google.com/patents/US20100099661

Common Intermediates Sisomicin

Figure US20100099661A1-20100422-C00031

Amberlite IRA-400 (OH form) (200 g) was washed with MeOH (3×200 m1). To a stirring suspension of the washed resin in MeOH (150 mL) was added sisomicin sulfate (20.0 g, 0.029 mol) and the mixture was stirred overnight. The resin was then filtered and washed with MeOH (100 mL) and the combined organic layers were concentrated to dryness to yield the desired sisomicin (11.57 g, 0.026 mol, 89.6% yield): MS m/e [M+H]+ calcd 448.3, found 448.1.

Example 1 6′-(2-Hydroxy-ethyl)-1-(4-amino-2(S)-hydroxy-butyryl)-sisomicin

Figure US20100099661A1-20100422-C00074

6′-(2-tert-Butyldimethylsililoxy-ethyl)-2′,3,3″-triBoc-1-(N-Boc-4-amino-2(S)-hydroxy-butyryl)-sisomicin

2′,3,3″-triBoc-1-(N-Boc-4-amino-2(S)-hydroxy-butyryl)-sisomicin (0.10 g, 0.105 mmol) was treated with tert-butyldimethylsilyloxy acetaldehyde following Procedure 1-Method A to yield the desired 6′-(2-tert-butyldimethylsilyloxy-ethyl)-2′,3,3″-triBoc-1-(N-Boc-4-amino-2(S)-hydroxy-butyryl)-sisomicin (MS m/e [M+H]+ calcd 1107.6, found 1107.4), which was carried through to the next step without further purification.

Figure US20100099661A1-20100422-C00075

6′-(2-Hydroxy-ethyl)-1-(4-amino-2(S)-hydroxy-butyryl)-sisomicin

6′ -(2-tert-butyldimethylsililoxy-ethyl)-2′,3,3″-triBoc-1-(N-Boc-4-amino-2(S)-hydroxy-butyryl)-sisomicin (0.105 mmol) was submitted to Procedure 3-Method B for Boc removal to yield a crude, which was purified by RP HPLC Method 1-Column A to yield 6′-(2-hydroxy-ethyl)-1-(4-amino-2(S)-hydroxy-butyryl)-sisomicin: MS m/e [M+H]+ calcd 593.3, found 593.2, [M+Na]+615.3 ; CLND 97.5% purity.

  1. Achaogen. Study for the treatment of complicated urinary tract infection and acute pyelonephritis.Available online: http://www.clinicaltrials.gov/ct2/show/NCT01096849 (accessed on 11 April 2013).
  2. Zhanel, G.G.; Lawson, C.D.; Zelenitsky, S.; Findlay, B.; Schweizer, F.; Adam, H.; Walkty, A.; Rubinstein, E.; Gin, A.S.; Hoban, D.J.; et al. Comparison of the next-generation aminoglycoside plazomicin to gentamicin, tobramycin and amikacin. Expert Rev. Anti-Infect. Ther. 201210, 459–473, doi:10.1586/eri.12.25.
  3. Endimiani, A.; Hujer, K.M.; Hujer, A.M.; Armstrong, E.S.; Choudhary, Y.; Aggen, J.B.; Bonomo, R.A. ACHN-490, a neoglycoside with potent in vitro activity against multidrug-resistant Klebsiella pneumoniae isolates. Antimicrob. Agents Chemother. 200953, 4504–4507.
  4. Achaogen. Achaogen pipeline. Available online: http://www.achaogen.com (accessed on 30 August 2012).
  5. Achaogen. Achaogen Awarded $60M Contract Option by BARDA for the Clinical Development of Plazomicin. Available online: http://www.achaogen.com/news/151/15 (accessed on 19 June 2013).
  6. Achaogen. Achaogen announces all objectives met in Phase 2 Plazomicin complicated urinary tract infections study and start of first-in-human study with ACHN-975. Available online: http://www.achaogen.com/uploads/news/id148/Achaogen_PressRelease_2012–05–15.pdf (accessed on 10 April 2013).
  7. Achaogen. Achaogen Announces Agreement with FDA on a Special Protocol Assessment for a Phase 3 Clinical Trial of Plazomicin to Treat Infections Caused by Carbapenem-Resistant Enterobacteriaceae (CRE); Achaogen: San Francisco, CA, USA, 2013.
  8. Comparison of the next-generation aminoglycoside plazomicin to gentamicin, tobramycin and amikacin
  9. 4-23-2010
    ANTIBACTERIAL AMINOGLYCOSIDE ANALOGS

Patent ID Title Submitted Date Granted Date
US9688711 ANTIBACTERIAL AMINOGLYCOSIDE ANALOGS
2016-01-20
US9266919 ANTIBACTERIAL AMINOGLYCOSIDE ANALOGS
2014-07-17
2015-02-12
Patent ID Title Submitted Date Granted Date
US8383596 ANTIBACTERIAL AMINOGLYCOSIDE ANALOGS
2010-04-22
US8822424 Antibacterial aminoglycoside analogs
2013-01-04
2014-09-02
US2012208781 AMINOGLYCOSIDE DOSING REGIMENS
2011-11-11
2012-08-16
US2012214759 TREATMENT OF KLEBSIELLA PNEUMONIAE INFECTIONS WITH ANTIBACTERIAL AMINOGLYCOSIDE COMPOUNDS
2011-11-11
2012-08-23
US2012214760 TREATMENT OF URINARY TRACT INFECTIONS WITH ANTIBACTERIAL AMINOGLYCOSIDE COMPOUNDS
2011-11-11
2012-08-23
US8318685 Nov 14, 2011 Nov 27, 2012 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8367625 Apr 7, 2011 Feb 5, 2013 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8372813 Apr 7, 2011 Feb 12, 2013 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8377896 Mar 9, 2011 Feb 19, 2013 Isis Pharmaceuticals, Inc Antibacterial 4,6-substituted 6′, 6″ and 1 modified aminoglycoside analogs
US8399419 Mar 9, 2011 Mar 19, 2013 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8481502 Apr 6, 2012 Jul 9, 2013 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8492354 Nov 14, 2011 Jul 23, 2013 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8524675 Nov 14, 2011 Sep 3, 2013 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8524689 Nov 14, 2011 Sep 3, 2013 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8569264 Jan 5, 2012 Oct 29, 2013 Isis Pharmaceuticals, Inc. Antibacterial 4,5-substituted aminoglycoside analogs having multiple substituents
US8653041 Oct 15, 2012 Feb 18, 2014 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8653042 Nov 14, 2011 Feb 18, 2014 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8658606 Nov 14, 2011 Feb 25, 2014 Achaogen, Inc. Antibacterial aminoglycoside analogs

References

  1. Jump up^ “WHO Drug Information, Vol. 26, No. 3, 2012. International Nonproprietary Names for Pharmaceutical Substances (INN). Recommended International Nonproprietary Names: List 68”(PDF). World Health Organization. p. 314. Retrieved 27 April 2016.
  2. Jump up^ Aggen, JB; Armstrong, ES; Goldblum, AA; Dozzo, P; Linsell, MS; Gliedt, MJ; Hildebrandt, DJ; Feeney, LA; Kubo, A; Matias, RD; Lopez, S; Gomez, M; Wlasichuk, KB; Diokno, R; Miller, GH; Moser, HE (30 August 2010). “Synthesis and Spectrum of the Neoglycoside ACHN-490” (PDF). Antimicrobial Agents and Chemotherapy54 (11): 4636–4642. doi:10.1128/AAC.00572-10PMC 2976124Freely accessiblePMID 20805391. Retrieved 27 April2016.
  3. Jump up to:a b Zhanel, GG; Lawson, CD; Zelenitsky, S; Findlay, B; Schweizer, F; Adam, H; Walkty, A; Rubinstein, E; Gin, AS; Hoban, DJ; Lynch, JP; Karlowsky, JA (10 January 2014). “Comparison of the Next-Generation Aminoglycoside Plazomicin to Gentamicin, Tobramycin and Amikacin”. Expert Review of Anti-infective Therapy10 (4): 459–73. doi:10.1586/eri.12.25PMID 22512755.
  4. Jump up^ García-Salguero, C; Rodríguez-Avial, I; Picazo, JJ; Culebras, E (October 2015). “Can Plazomicin Alone or in Combination Be a Therapeutic Option against Carbapenem-Resistant Acinetobacter baumannii?” (PDF). Antimicrobial Agents and Chemotherapy59 (10): 5959–66. doi:10.1128/AAC.00873-15PMC 4576036Freely accessible. Retrieved 27 April 2016.
  5. Jump up^ “Achaogen Announces Plazomicin Granted QIDP Designation by FDA”. GlobeNewswire, Inc. Retrieved 27 April 2016.
  6. Jump up^ “Achaogen — Plazomicin”. Achaogen, Inc. Retrieved 27 April2016.
  7. Jump up^ “Plazomicin — AdisInsight”. Springer International Publishing AG. Retrieved 27 April 2016.
  8. Jump up^ “Medscape Log In”http://www.medscape.com. Retrieved 2018-07-03.
  9. Jump up^ “BioCentury – FDA approves plazomicin for cUTI, but not blood infections”http://www.biocentury.com. Retrieved 2018-06-28.
  10. Jump up^ “Drugs@FDA: FDA Approved Drug Products”http://www.accessdata.fda.gov. Retrieved 2018-06-28.
Plazomicin
Plazomicin structure.svg
Names
IUPAC name
(2S)-4-Amino-N-[(1R,2S,3S,4R,5S)-5-amino-4-[[(2S,3R)-3-amino-6-[(2-hydroxyethylamino)methyl]-3,4-dihydro-2H-pyran-2-yl]oxy]-2-[(2R,3R,4R,5R)-3,5-dihydroxy-5-methyl-4-(methylamino)oxan-2-yl]oxy-3-hydroxycyclohexyl]-2-hydroxybutanamide
Other names
6′-(hydroxylethyl)-1-(HABA)-sisomicin
Identifiers
3D model (JSmol)
ChEMBL
ChemSpider
KEGG
PubChem CID
UNII
Properties
C25H48N6O
Molar mass 592.683 g/mol
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F],

Achaogen is a clinical-stage biopharmaceutical company passionately committed to the discovery, development, and commercialization of novel antibacterials to treat multi-drug resistant, or MDR, gram-negative infections.

Achaogen Inc.jpg

Achaogen (a-KAY-o-jen) is developing plazomicin, its lead product candidate, for the treatment of serious bacterial infections due to MDR Enterobacteriaceae, including carbapenem-resistant Enterobacteriaceae, or CRE. In 2013, the Centers for Disease Control and Prevention identified CRE as a “nightmare bacteria” and an immediate public health threat that requires “urgent and aggressive action.” We expect to initiate a Phase 3 superiority trial of plazomicin in the first quarter of 2014.

CRE are one of many types of MDR gram-negative pathogens threatening patients. Bacteria such as Pseudomonas aeruginosaAcinetobacter baumannii, and extended-spectrum beta-lactamase producing Enterobacteriaceae each pose “serious” resistance threats, according to the CDC, and also drive a great need for new, safe, and effective antibiotics. We have assembled the chemistry and microbiology expertise and capabilities required to develop new agents for the treatment of gram-negative infections. Plazomicin was the first clinical candidate from our gram-negative antibiotic discovery engine. In addition, our research and development pipeline includes two antipseudomonal programs targeting P. aeruginosa—a program to discover and develop small molecule inhibitors of LpxC, which is an enzyme essential for the synthesis of the outer membrane of gram-negative bacteria, and a therapeutic antibody program. We are also pursuing small molecule research programs targeting other essential gram-negative enzymes.

Achaogen has built an exceptional research and development team with deep expertise in the discovery and development of new drugs from research through commercialization. Our executive team has over 60 years of combined industry experience, and a proven track record of leadership, global registration, and lifecycle management for over 20 products. Our facility is located on the shores of the San Francisco Bay, ten minutes from the San Francisco International Airport, and only fifteen minutes from downtown San Francisco.

Image result for Plazomicin sulfate

ZEMDRITM (plazomicin) Approved by FDA for the Treatment of Adults with Complicated Urinary Tract Infections (cUTI)

https://globenewswire.com/news-release/2018/06/26/1529573/0/en/ZEMDRITM-plazomicin-Approved-by-FDA-for-the-Treatment-of-Adults-with-Complicated-Urinary-Tract-Infections-cUTI.html

― ZEMDRI is a new treatment for patients with cUTI, including pyelonephritis, due to certain Enterobacteriaceae ―

― ZEMDRI is the only once-daily aminoglycoside therapy approved for use in cUTI ―


― ZEMDRI has microbiological activity against pathogens designated by the CDC as urgent and serious public health threats, including carbapenem-resistant (CRE) and extended spectrum beta-lactamase (ESBL)- producing Enterobacteriaceae ―

SOUTH SAN FRANCISCO, Calif., June 26, 2018 (GLOBE NEWSWIRE) — Achaogen, Inc. (NASDAQ:AKAO), a biopharmaceutical company developing and commercializing innovative antibacterial agents to address multidrug resistant (MDR) gram-negative infections, today announced that the U.S. Food and Drug Administration (FDA) has approved ZEMDRI™ (plazomicin) for adults with complicated urinary tract infections (cUTI), including pyelonephritis, caused by certain Enterobacteriaceae in patients who have limited or no alternative treatment options. ZEMDRI is an intravenous infusion, administered once daily.

“The approval of ZEMDRI marks a significant milestone for Achaogen and we are excited to offer healthcare practitioners a new treatment option for patients with certain serious bacterial infections. ZEMDRI is designed to retain its potent activity in the face of certain difficult-to-treat MDR infections, including CRE and ESBL- producing Enterobacteriaceae,” said Blake Wise, Achaogen’s Chief Executive Officer. “Today’s milestone was made possible by our employees, by patients and investigators involved in our clinical trials, and by BARDA, who contributed significant funding for the development of ZEMDRI. This marks an important step in our commitment to fighting MDR bacteria and we are excited to launch ZEMDRI, a much needed once-daily antibiotic.”

“Bacteria continue to circumvent existing antibiotics, making certain infections notoriously hard to treat and putting some patients at high risk for mortality,” said James A. McKinnell, Assistant Professor of Medicine at the David Geffen School of Medicine and LA Biomed at Harbor-UCLA. “Aminoglycosides are a familiar and very effective class of antibiotics. I look forward to adding plazomicin to my short list of available treatment options and to its potential impact on patient outcomes.”

Regarding the potential indication for plazomicin for the treatment of bloodstream infection (BSI), the FDA issued a Complete Response Letter (CRL) stating that the CARE study does not provide substantial evidence of effectiveness of plazomicin for the treatment of BSIThe Company intends to meet with the FDA to determine whether there is a feasible resolution to address the CRL.

Achaogen will work with hospitals, providers, and insurers to ensure patients are able to receive this treatment. Patients, physicians, pharmacists, or other healthcare professionals with questions about ZEMDRI should contact 1.833.252.6400 or visit www.ZEMDRI.com.

ZEMDRI Phase 3 Clinical Results
The approval of ZEMDRI is supported in part by data from the EPIC (Evaluating Plazomicin In cUTI) clinical trial, which was the first randomized controlled study of once-daily aminoglycoside therapy for the treatment of cUTI, including pyelonephritis.

In the Phase 3 EPIC cUTI trial, ZEMDRI demonstrated non-inferiority to meropenem for the co-primary efficacy endpoints of composite cure (clinical cure and microbiological eradication) in the microbiological modified intent-to-treat (mMITT; N=388) population at Day 5 and test-of-cure (TOC) visit (Day 17 + 2). Composite cure rates at Day 5 were 88.0% (168/191) for ZEMDRI vs 91.4% (180/197) for meropenem (difference -3.4%, 95% CI, -10.0 to 3.1). Composite cure rates at TOC were 81.7% (156/191) for ZEMDRI vs 70.1% (138/197) for meropenem (difference 11.6%, 95% CI, 2.7 to 20.3). Composite cure at the TOC visit in patients with concomitant bacteremia at baseline was achieved in 72.0% (18/25) of patients in the ZEMDRI group and 56.5% (13/23) of patients in the meropenem group. The most common side effects (≥1% of patients treated with ZEMDRI) were decreased kidney function, diarrhea, hypertension, headache, nausea, vomiting, and hypotension.1

The FDA approved a breakpoint of <= 2 mcg/mL; greater than 99% of Escherichia coliKlebsiella pneumoniae and Enterobacter cloacae in U.S. surveillance are susceptible to Zemdri when applying this breakpoint.2

About cUTI
cUTI is defined as a UTI occurring in a patient with an underlying complicating factor of the genitourinary tract, such as a structural or functional abnormality.3 Patients with pyelonephritis, regardless of underlying abnormalities of the urinary tract, are considered a subset of patients with cUTI.4 An estimated 3 million cases of cUTI are treated in the hospital setting in the US each year.5 Enterobacteriaceae are the most common pathogens causing cUTIs6, and resistance within this family is a global concern. High rates of resistance to previous mainstays of therapy necessitate alternative treatment options. Ineffectively managed cUTI can lead to increased treatment failure rates, recurrence of infection, increased re-hospitalization, and increased morbidity and mortality. cUTI infections place an economic burden on hospitals and payers.6,7

About ZEMDRI
ZEMDRI is an aminoglycoside with once-daily dosing that has activity against certain Enterobacteriaceae, including CRE and ESBL- producing Enterobacteriaceae. Achaogen’s EPIC clinical trial successfully evaluated the safety and efficacy of ZEMDRI in adult patients with cUTI, including pyelonephritis. ZEMDRI was engineered to overcome aminoglycoside-modifying enzymes, the most common aminoglycoside-resistance mechanism in Enterobacteriaceae, and has in vitro activity against ESBL- producing, aminoglycoside- resistant, and carbapenem- resistant isolates. The Centers for Disease Control and Prevention (CDC) has characterized ESBL- producing Enterobacteriaceae as a “serious threat” and CRE as “nightmare bacteria”, which is an immediate public health threat that requires urgent and aggressive action.

Working in the Lab
Working in the Lab
Working in the Lab
Achaogen, Inc.
Blake Wise, Chief Executive Officer at Achaogen
Blake Wise, Chief Executive Officer at Achaogen
Blake Wise, Chief Executive Officer at Achaogen
Achaogen, Inc.
High-Resolution Achaogen company logo
High-Resolution Achaogen company logo
High-Resolution Achaogen company logo
Achaogen, Inc.

/////////Plazomicin, ZEMDRI, FDA 2018, fast track designation, Plazomicin SULFATE, ACHN 490 sulfate, cUTI, Achaogen

CC1(COC(C(C1NC)O)OC2C(CC(C(C2O)OC3C(CC=C(O3)CNCCO)N)N)NC(=O)C(CCN)O)O

CN[C@@H]1[C@@H](O)[C@@H](O[C@H]2[C@@H](C[C@H](N)[C@@H](O[C@H]3OC(CNCCO)=CC[C@H]3N)[C@@H]2O)NC(=O)[C@@H](O)CCN)OC[C@]1(C)O

LAFUTIDINE, ラフチジン

$
0
0

LafutidineChemSpider 2D Image | lafutidine | C22H29N3O4S

Lafutidine.pngLafutidine.svg

LAFUTIDINE

N-[4-[4-(Piperidin-1-ylmethyl)pyridin-2-yloxy]-(Z)-but-2-en-1-yl]-2-(furfurylsulfinyl)acetamide

    • FRG-8813
    • ATC:A02B
  • Use:antisecretory, gastric H2-antagonist
  • (+)-2-[(2-furanylmethyl)sulfinyl]-N-[(2Z)-4-[[4-(1-piperidinylmethyl)-2-pyridinyl]oxy]-2-butenyl]acetamide
  • Formula:C22H29N3O4S
  • MW:431.56 g/mol
  • CAS-RN:118288-08-7
  • (±)-2-(Furfurylsulfinyl)-N-(4-(4-(piperidinomethyl)-2-pyridyl)oxy-(Z)-2-butenyl)acetamide
  • (Z)-2-((2-Furanylmethyl)sulfinyl)-N-(4-((4-(1-piperidinylmethyl)-2-pyridinyl)oxy)-2-butenyl)acetamide
  • 118288-08-7

FRG‐8813、2‐(Furfurylsulfinyl)‐N‐[(Z)‐4‐[[4‐(piperidinomethyl)‐2‐pyridinyl]oxy]‐2‐butenyl]acetamide、ロクチジン、Loctidine、ラフチジン・・・

2-[(furan-2-ylmethyl)sulfinyl]-N-[(2Z)-4-{[4-(piperidin-1-ylmethyl)pyridin-2-yl]oxy}but-2-en-1-yl]acetamide
49S4O7ADLC
7173
Acetamide, 2-[(2-furanylmethyl)sulfinyl]-N-[(2Z)-4-[[4-(1-piperidinylmethyl)-2-pyridinyl]oxy]-2-buten-1-yl]- [ACD/Index Name]
lafutidine [INN] [Wiki]
UNII:49S4O7ADLC
(Z)-2-((Furan-2-ylmethyl)sulfinyl)-N-(4-((4-(piperidin-1-ylmethyl)pyridin-2-yl)oxy)but-2-en-1-yl)acetamide

Lafutidine , also named N-[4-[4-(piperidin-1-ylmethyl)pyridin-2-yloxy]-(Z)-but-2-en-1-yl]-2-(furfurylsulfinyl)acetamide, is a histamine H2 receptor antagonist that was first produced in Japan by Taiho and UCB Japan for the oral treatment of peptic ulcers in 2000. In 2010 it was approved for the treatment of mild gastroesophageal reflux disease, and in 2012 it was approved to help improve symptoms of gastric mucosal lesions due to gastritis

Lafutidine (INN) is a second generation histamine H2 receptor antagonist having multimodal mechanism of action and used to treat gastrointestinal disorders. It is marketed in Japan and India.

Medical use

Lafutidine is used to treat gastric ulcersduodenal ulcers, as well as wounds in the lining of the stomach associated with acute gastritis and acute exacerbation of chronic gastritis.[1][2]

Adverse effects

Adverse events observed during clinical trials included constipationdiarrhea, drug rashnauseavomiting and dizziness.[2]

Mechanism of action

Like other H2 receptor antagonists it prevents the secretion of gastric acid.[2] It also activates calcitonin gene-related peptide, resulting in the stimulation of nitric oxide (NO) and regulation of gastric mucosal blood flow, increases somatostatin levels also resulting in less gastric acid secretion, causes the stomach lining to generate more mucin, inhibits neutrophil activation thus preventing injury from inflammation, and blocks the attachment of Helicobacter pylori to gastric cells.[2]

Image result for LAFUTIDINE SYNTHESIS

Trade names

It is marketed in Japan as Stogar by UCB[1] and in India as Lafaxid by Zuventus Healthcare.[2]

N-[4-[4-(Piperidin-1-ylmethyl)pyridin-2-yloxy]-(Z)-but-2-en-1-yl]-2-(furfurylsulfinyl)acetamide 1 as a white solid (15.8 kg, 91.3%).(2,3)

1H NMR (600 MHz, CDCl3): δ 1.43 (m, 2H), 1.56–1.60 (m, 4H), 2.36 (m, 4H), 3.34 (d, 1H, J = 14.4 Hz), 3.40 (s, 2H), 3.59 (d, 1H, J = 14.4 Hz), 4.10 (t, 2H, J = 6.6 Hz), 4.17 (d, 1H, J = 13.8 Hz), 4.31 (d, 1H, J = 13.8 Hz), 4.93 (d, 2H, J = 6.6 Hz), 5.67–5.69 (m, 1H), 5.83–5.87 (m, 1H), 6.39 (dd, 1H, J = 1.8, 3.0 Hz), 6.47 (d, 1H, J = 3.0 Hz), 6.72 (s, 1H), 6.87 (d, 1H, J = 5.4 Hz), 7.19 (s, 1H), 7.43 (d, 1H, J = 1.8 Hz), 8.03 (d, 1H, J = 5.4 Hz).

13C NMR (150 MHz, CDCl3): δ 24.2, 26.0, 26.0, 37.2, 50.2, 53.4, 54.6, 54.6, 61.4, 62.4, 110.8, 111.3, 112.2, 117.7, 128.4, 128.9, 143.3, 143.9, 146.3, 151.5, 163.6, 163.6.

IR (KBr): 3325, 2935, 1638, 1613, 1041 cm–1.

ESI-MS: m/z 431.1.

Increasing the Purity of Lafutidine Using a “Suicide Substrate”

Chengjun Wu Zhen LiChunchao WangYanan Zhou, and Tiemin Sun* 

Key Laboratory of Structure-Based Drug Design and DiscoveryShenyang Pharmaceutical University, Ministry of Education, Shenyang 110016, P. R. China
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.8b00070

https://pubs.acs.org/doi/suppl/10.1021/acs.oprd.8b00070/suppl_file/op8b00070_si_001.pdf

CLIP

http://www.drugfuture.com/synth/syndata.aspx?ID=145925

EP 0282077; JP 1988225371; JP 1989230556; JP 1989230576; US 4912101

1) The reaction of 2-bromo-4-(piperidin-1-ylmethyl)pyridine (I) with 4-amino-2(Z)-buten-1-ol (II) by means of NaH in THF gives 4-[4-(piperidin-1-ylmethyl)pyridin-2-yloxy]-2(Z)-buten-1-amine (III), which is then condensed with 2-(2-furylmethylsulfinyl)acetic acid (IV) by means of 1-ethyl-3-[3-(dimethylamino)propyl]carbodiimide (EDCD) in dichloromethane.

EP 0582304; JP 1994192195

The condensation of 2-chloro-4-(piperidin-1-ylmethyl)pyridine (V) with 4-(tetrahydropyranyloxy)-2(Z)-buten-1-ol (VI) by means of NaH in THF gives 4-(piperidin-1-ylmethyl)-2-[4-(tetrahydropyranyloxy)-2(Z)-butenyloxy)pyridine (VII), which is deprotected with 4-methylbenzenesulfonic acid in methanol, yielding the free butenol (VIII). The acylation of (VIII) with methanesulfonyl chloride in toluene affords the corresponding mesylate (IX), which is finally condensed with 2-(2-furylmethylsulfonyl)acetamide (X) (obtained from the corresponding 4-nitrophenyl ester (XI) with ammonia) by means of potassium tert-butoxide in toluene.

Chem Pharm Bull 1998,46(4),616

A new synthesis of lafutidine has been described: The condensation of 2-bromopyridine-4-carbaldehyde ethylene ketal (I) with 4-(tetrahydropyranyloxy)-2(Z)-buten-1-ol (II) by means of NaOH, K2CO3 and tetrabutylammonium bisulfate in refluxing toluene gives the corresponding substitution product (III), which by treatment with pyridinium p-toluenesulfonate (PPTS) in hot ethanol yields the 2(Z)-butenol (IV). The reaction of (IV) with SOCl2 and then with potassium phthalimide (V) affords the substituted phthalimide (VI), which by treatment with hydrazine hydrate in refluxing methanol gives the 2(Z)-butenamine (VII). The condensation of (VII) with 2-(2-furylmethylsulfinyl)acetic acid 4-nitrophenyl ester (VIII) in THF yields the expected amide (IX), which is treated with p-toluenesulfonic acid in refluxing acetone/water to eliminate the ethylene ketal protecting group yilding the aldehyde (X). Finally, this compound is reductocondensed with piperidine (XI) by means of NaBH4 in ethanol.

CLIP

Synthesis Path

Lafutidine
CAS Registry Number: 118288-08-7
CAS Name: 2-[(2-Furanylmethyl)sulfinyl]-N-[(2Z)4-[[4-(1-piperidinylmethyl)-2-pyridinyl]oxy]-2-butenyl]-acetamide
Additional Names: 2-(furfurylsulfinyl)-N-[(Z)-4-[[4-(piperidinomethyl)-2-pyridyl]oxy]-2-butenyl]acetamide
Manufacturers’ Codes: FRG-8813
Trademarks: Protecadin (Taiho); Stogar (Fujirebio)
Molecular Formula: C22H29N3O4S
Molecular Weight: 431.55
Percent Composition: C 61.23%, H 6.77%, N 9.74%, O 14.83%, S 7.43%
Literature References: Second generation histamine H2-receptor antagonist. Prepn of racemate: N. Hirakawa et al., EP 282077eidem, US 4912101 (1988, 1990 both to Fujirebio); and pharmacology: eidem, Chem. Pharm. Bull. 46, 616 (1998). Pharmacology: S. Onodera et al., Jpn. J. Pharmacol. 68, 161 (1995). Mode of action study: M. Umeda et al., J. Gastroenterol. Hepatol. 14, 859 (1999). Gastroprotective effects in rats: H. Ajioka et al., Pharmacology 61, 83 (2000). Clinical pharmacokinetics: S. Haruki et al.,Yakuri to Chiryo 23, 3049 (1995). Toxicology study: A. Broadmeadow et al., Oyo Yakuri 50, 167 (1995).
Properties: Prepd as the (±) mixture, crystals from benzene-hexane, mp 92.7-94.9°. Slightly bitter taste. Freely sol in DMF, glacial acetic acid; sol in methanol; sparingly sol in dehydrated ethanol; very slightly sol in ether. Practically insol in water.
Melting point: mp 92.7-94.9°
Therap-Cat: Antiulcerative.
Keywords: Antiulcerative; Histamine H2-Receptor Antagonist.

References

References

  1. Jump up to:a b UCB Japan Revised: April 2005 Stogar tablets
  2. Jump up to:a b c d e Zuventus Healthcare Ltd. India Lafaxid tablets
    • a EP 582 304 (Fujirebio; 5.8.1993; J-prior. 7.8.1992).
  • preparation of 2-benzenesulfonyl-4-methylpyridine:

    • EP 931 790 (Kuraray; 26.1.1999; J-prior. 26.1.1998).
  • chlorination of 2-benzenesulfonyl-4-methylpyridine:

    • JP 10 231 288 (Kuraray; 2.9.1998; J-prior. 21.2.1997).
    • WO 9 626 188 (Sagami Res. Center; 21.2.1996; J-prior. 22.2.1995).
    • b EP 282 077 (Fujirebio; 11.3.1988; J-prior. 13.3.1987).
    •  US 4 912 101 (Fujirebio; 27.3.1990; J-prior. 13.3.1987).
  • preparation of I:

    • JP 10 231 288 (Kuraray; 2.9.1998; J-prior. 21.2.1997).
  • chlorination of 2-chloromethylpyridines forming 2-chloro-4-trichloromethylpyridine:

    • EP 557 967 (Central Glass Co.; 1.9.1993; J-prior. 24.2.1993).
  • treatment of I with (Z)-4-(tetrahydro-2H-pyran-2-yloxy)-2-buten-1-ol:

    • US 5 382 589 (Fujirebio; 17.1.1995; J-prior. 27.1.1992).
  • preparation of furfuryl acetate and derivatives:

    • JP 8 198 844 (Fujirebio; 6.8.1996; J-prior. 23.1.1995).
    • JP 8 198 843 (Fujirebio; 6.8.1996; J-prior. 23.1.1995).
    • JP 07 010 860 (Central Glass Co.; 13.1.1995; J-prior. 25.6.1993).
    • JP 07 010 864 (Central Glass Co.; 13.1.1995; J-prior. 25.6.1993).
  • 2-(furfurylsulfinyl)acetic acid nitrophenyl ester:

    • JP 07 010 862 (Central Glass Co.; 13.1.1995; J-prior. 25.6.1993).
  • 4-(tetrahydro-2-pyranyloxy)-2(Z)-buten-1-ol from 2(Z)-butene-1,4-diol:

    • Nishiguchi, T. et al.: J. Org. Chem. (JOCEAH) 63, 23, 8183 (1998).
    • Davis, K. J. et al.: Synth. Commun. (SYNCAV) 29, 10, 1679 (1999).
    • Nishiguchi, T. et al.: J. Chem. Soc., Perkin Trans. 1 (JCPRB4) 1995, 24, 2491.

/////////////////LAFUTIDINE, ラフチジン , FRG-8813, ATC:A02B

FRG‐8813
2‐(Furfurylsulfinyl)‐N‐[(Z)‐4‐[[4‐(piperidinomethyl)‐2‐pyridinyl]oxy]‐2‐butenyl]acetamide
ロクチジン
Loctidine
ラフチジン
Laftidine
2‐[[(2‐Furyl)methyl]sulfinyl]‐N‐[(Z)‐4‐[[4‐(piperidinomethyl)‐2‐pyridyl]oxy]‐2‐butenyl]acetamide
N‐[(Z)‐4‐[4‐(Piperidinomethyl)‐2‐pyridyloxy]‐2‐butenyl]‐2‐(furfurylsulfinyl)acetamide
(Z)‐2‐フルフリルスルフィニル‐N‐[4‐(4‐ピペリジノメチル‐2‐ピリジルオキシ)‐2‐ブテニル]アセトアミド
ストガー
Stogar
プロテカジン
Protecadin
(+)‐ラフチジン
(+)‐Laftidine
ラフルチジン
Laflutidine
(Z)‐2‐Furfurylsulfinyl‐N‐[4‐(4‐piperidinomethyl‐2‐pyridyloxy)‐2‐butenyl]acetamide
2‐[[(2‐フリル)メチル]スルフィニル]‐N‐[(Z)‐4‐[[4‐(ピペリジノメチル)‐2‐ピリジル]オキシ]‐2‐ブテニル]アセトアミド
N‐[(Z)‐4‐[4‐(ピペリジノメチル)‐2‐ピリジルオキシ]‐2‐ブテニル]‐2‐(フルフリルスルフィニル)アセトアミド
2‐(フルフリルスルフィニル)‐N‐[(Z)‐4‐[[4‐(ピペリジノメチル)‐2‐ピリジニル]オキシ]‐2‐ブテニル]アセトアミド

C1CCN(CC1)CC2=CC(=NC=C2)OCC=CCNC(=O)CS(=O)CC3=CC=CO3

Ferric Maltol, マルトール第二鉄

$
0
0

Ferric maltol.png

Ferric Maltol

Iron, tris(3-hydroxy-2-methyl-4H-pyran-4-onato-O3,O4)-

Molecular Formula: C18H15FeO9
Molecular Weight: 431.154 g/mol

iron(3+);2-methyl-4-oxopyran-3-olate

RN: 33725-54-1
UNII: MA10QYF1Z0

Feraccru

Ferric maltol; UNII-MA10QYF1Z0; MA10QYF1Z0; Ferric maltol (INN); Ferric maltol [INN]; 33725-54-1

Shield Therapeutics, under license from Vitra Pharmaceuticals

NDA filing expected in US in 2H 2018, Ph 3 trial is planned in 2018/19 for treatment of iron deficiency anemia (IDA) in children., Expected dose form: Oral Capsule; 30 mg

Treatment of iron deficiency anemia (IDA) associated with inflammatory bowel disease (IBD) and Chronic Kidney disease.

Iron deficiency anaemia (IDA) occurs when iron levels are insufficient to support red blood cell production and is defined – according to the WHO – as haemoglobin levels below 13 g/dL in men over 15 years, below 12 g/dL in non-pregnant women over 15 years, and below 11 g/dL in pregnant women. Iron is absorbed at the apical surface of enterocytes to be transported by ferroportin, the only known iron exporter, across the basolateral surface of the enterocyte into circulation. Inflammation from IBD interferes with iron absorption by causing an increase in hepcidin, a peptide hormone synthesized in the liver that inhibits ferroportin activity. Anaemia is the most common extra-intestinal complication of inflammatory bowel disease (IBD) and although it often involves a combination of IDA and anaemia of chronic disease, IDA remains an important contributor in this condition due to chronic intestinal bleeding and decreased iron intake (from avoidance of foods that may exacerbate symptoms of IBD). In a variety of populations with IBD, the prevalence of iron deficiency anaemia ranges from 36%-76%. The serum markers of iron deficiency are low ferritin, low iron, raised total iron binding capacity, raised red cell protoporhyrin and increased transferrin binding receptor (sTfR). Serum ferritin is the most powerful test for iron deficiency. The cut-off level of ferritin which is diagnostic varies between 12-15 µg/L. Higher levels of serum ferritin do not exclude the possibility of iron deficiency, and a serum ferritin level of <100 μg/L may still be consistent with iron deficiency in patients with IBD. A transferrin saturation of <16% is indicative of iron deficiency, either absolute or functional. Other findings on a complete blood count panel that are suggestive of iron deficiency anaemia, but are not considered diagnostic, include microcytosis, hypochromia, and elevation of red cell distribution width.

A deficiency of iron can have a significant impact on a patient’s quality of life. Appropriate diagnosis and treatment of iron deficiency anaemia are important to improve or maintain the quality of life of patients. The goals of treatment are to treat the underlying cause, limit further blood loss or malabsorption, avoid blood transfusions in haemodynamically stable patients, relieve symptoms, and improve quality of life. More specifically, therapeutic goals of treatment include normalizing haemoglobin levels within 4 weeks (or achieving an increase of >2 g/dL) and replenishing iron stores (transferrin saturation >30%). Oral iron supplementation has been considered standard treatment because of an established safety profile, lower cost, and ease of administration. It has been shown to be effective in correcting anaemia and repleting iron stores. One concern with higher doses of daily oral iron is intolerance due to GI side effects. Symptoms include nausea, vomiting, diarrhea, abdominal pain, constipation, and melena-like stools. Guidelines on the Diagnosis and Management of Iron Deficiency and Anaemia in Inflammatory Bowel Diseases recommend IV iron therapy over oral iron supplementation in the treatment of iron deficiency anaemia in patients with IBD, citing faster and prolonged response to treatment, decreased irritation of existing GI inflammation, improved patient tolerance, and improved quality of life. Patients with severe anaemia (haemoglobin level of <10 g/dL), failure to respond or intolerance to oral iron therapy, severe intestinal disease or patients receiving concomitant erythropoietin are recommended indications for IV iron therapy. Other conditions where patients should be considered for first-line IV therapy over oral therapy include congestive heart failure, upper GI bleeding, and in situations where rapid correction of anaemia may be required.

Across EU there are several iron (Fe+2) oral preparations as ferrous fumarate, ferrous gluconate, ferrous sulphate and ferrous glycine sulfate, formulated as tablet, solution or gastroresistent capsules. All ferrous compounds are oxidised in the lumen of the gut or within the mucosa with release of activated hydroxyl radicals, which may attack the gut wall and can effect a range of gastrointestinal symptoms and discomfort. Ferric preparations also exist but with less bioavailability. Across EU there are also several IV products on the market: iron (III) hydroxide dextran complex, iron sucrose, ferric carboxymaltose, iron isomaltoside. IV iron therapy, however, is inconvenient, invasive and associated with the risk of rare but serious hypersensitivityreactions; it is used in those situations when oral preparations cannot be used or when there is a need to deliver iron rapidly. Feraccru is a trivalent iron, oral iron replacement preparation. The active substance of Feraccru is ferric maltol (also known as 3-hydroxy-2-methyl-4H-pyrane-4-one iron (III) complex, or ST10, or ferric trimaltol or ferric maltol) an oral ferric iron/maltol complex. It is presented as red hard gelatine capsules containing 30 mg iron (ferric iron). Maltol is a sugar derivative that strongly chelates iron in the ferric form (FeIII) rendering the iron stable and available for absorption. Upon dissociation of the ferric maltol complex, the maltol molecules are absorbed and glucuronidated in the intestinal wall, and within the liver during first pass metabolism, and subsequently eliminated from the body in the urine. The iron is absorbed via the endogenous dietary iron uptake system. The indication finally agreed with the CHMP was: Feraccru is indicated in adults for the treatment of iron deficiency anaemia (IDA) in patients with inflammatory bowel disease (IBD) (see section 5.1). The proposed dosage is one 30 mg capsule twice daily on an empty stomach, corresponding to 60 mg ferric iron per day. There was agreement in the paediatric investigation plan to grant a deferral and a waiver for iron as iron (III)-maltol complex (EMEA-001195-PIP01-11).The PDCO granted a waiver in infants under 6 months of age and a referral for the completion of the planned paediatric studies (ST10-021 PK-PED/ST10-01-102, an open label, randomised, multiple-dose, parallel PK study and ST10-01-303, a randomised, open label comparative safety and efficacy study of ST10 and oral ferrous sulphate as comparator) until the adult studies are completed.

http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Public_assessment_report/human/002733/WC500203504.pdf

SYN

Patent

https://patents.google.com/patent/WO2017167963A1/en

The sugar derivative maltol is a hydroxypyrone (IUPAC name: 3- hydroxy-2-methyl-4£f-pyran-4-one) and it strongly chelates iron and the resulting complex (ferric trimaltol) is well absorbed, unlike many other ferric iron therapies. Ferric trimaltol appears well tolerated even in populations highly susceptible to gastrointestinal side-effects, such as IBD patients (Harvey et al . , 1998), and as such it provides a valuable alternative to patients who are intolerant of oral ferrous iron products, notably in place of intravenous iron. Clinical trials using ferric trimaltol have been carried out, see for example, Gasche et al., 2015.

However, despite the evidence of bioavailability and tolerability for ferric trimaltol, its clinical development has been limited by the absence of adequate synthetic routes. In particular, most manufacturing processes require the use of organic solvents, which increase manufacturing costs, for example to deal with post-synthesis solvent removal, and require additional safety measures, for example to deal with flammability . Critically, solvent-based syntheses are not robust and often generate ferric hydroxide, described in the prior art to be an unwanted impurity of the synthesis.

WO 03/097627 (Vitra Pharmaceuticals Limited) describes the synthesis of ferric trimaltol from iron salts of carboxylic acids in aqueous solution at a pH greater than 7. In a first

synthesis, ferric citrate is added to a solution of sodium hydroxide at room temperature and maltol is added to a second solution of sodium hydroxide at pH 11.6. The ferric citrate solution is added to the maltol solution, leading to the

production of a deep red precipitate. This composition is then evaporated until dryness and the material is powdered and dried. Alternative syntheses are described using ferrous fumarate or ferrous gluconate as the iron carboxylate salt starting material, and by dissolving maltol in sodium carbonate solution in place of sodium hydroxide. However, despite the fact that this process is fully aqueous, several of the iron carboxylate salts employed are expensive, especially as they need to be pharmaceutical grade if the ferric trimaltol is to be suitable for human administration. More importantly, this process introduces high levels of

carboxylates (equimolar to iron or greater) to the synthesis that are not easily removed by filtration or centrifugation of the ferric trimaltol cake. Instead these water soluble contaminants must be washed off (e.g. water washed), but this would result in considerable losses of the product due to the amphipathic nature of ferric trimaltol.

WO 2012/101442 (Iron Therapeutic Holdings AG) describes the synthesis of ferric trimaltol by reacting maltol and a non- carboxylate iron salt in an aqueous solution at alkaline pH .

However, despite the lower cost of non-carboxylate iron salts, pharmaceutically appropriate grades are still required if the ferric trimaltol is to be suitable for human administration and hence are comparatively expensive starting materials.

Importantly, the use of non-carboxylate iron salts (e.g. ferric chloride) results in the addition of considerable levels of the respective counter-anion (e.g. three moles of chloride per every mole of iron) of which a significant part is retained in the filtration (or centrifugation) cake and thus must be washed off. As such, WO 2012/101442 does not address the problem of product losses in WO 03/097627. Furthermore, the addition of a non- carboxylate iron salt (e.g. ferric chloride) to a very alkaline solution, as described in WO 2012/101442, promotes the formation of stable iron oxides, which is an unwanted contaminant in ferric trimaltol . As a consequence, further costly and time-consuming processing of the material would be required for manufacturing .

Overall, the cost of the current aqueous syntheses is driven by regulatory demands for low levels of toxic heavy metals and residual reagents in the final pharmaceutical formulation, which force the use of highly purified, and thus expensive, iron salts as well as thorough washing of the final product (resulting in significant losses of product) . This will impact on the final price of ferric trimaltol and potentially limits patient access to this therapy. As such, there is a need for a process that can use lower iron grades and limited wash cycles, whilst producing ferric trimaltol of adequate purity.

Ferric maltols are a class of compounds that include ferric trimaltol, a chemical complex formed between ferric iron (Fe3+) and the hydroxypyrone, maltol (IUPAC name: 3-Hydroxy-2-methyl-4£f- pyran-4-one) , in a molar ratio of ferric iron to maltol of 3:1. Maltol strongly chelates the ferric iron and the resulting complex (ferric trimaltol which may also be written as ferric tri-maltol) is well absorbed, in contrast to some other ferric iron supplements, fortificants and therapies. Maltol binds metal cations mainly in the form of a dioxobidentate ligand in a similar manner proposed for other 4 ( 1H) -pyranones :

Figure imgf000010_0001

Structure of maltol (3-hydroxy-2-methyl-4 (H) -pyran-4-one) and dioxo-chelation to metal cations (M) such as iron. For ferric trimaltol three maltol groups surround one iron.

Examples

Example 1: Ferric trimaltol from L-lyslne coated ferric hydroxide

Synthesis of lysine-coated ferric hydroxide colloid

14.87g FeCl3. 6H20 was added to 25 mL UHP water and stirred until dissolved. 14.9g NaOH 5M was then added drop-wise to this solution with constant stirring, during which a ferric hydroxide colloid was gradually produced. This colloidal suspension was then added to a L-Lysine suspension (5.02g in 25mL ddH2<D) .

Ferric trimaltol synthesis

7 g NaOH pellets was added to 25 mL UHP water and stirred until dissolved. Next, 24.5g maltol was added and stirred until dissolved. Then, the suspension of lysine-coated ferric

hydroxide colloids was gradually added to the maltol with vigorous stirring, producing a dark red precipitate (with a significant brown hue) . This suspension was incubated overnight during which time it became lighter and the brown hue

disappeared. This precipitate was then recovered by

centrifugation (4500 rpm x 5min) and dried overnight (50°C) .

Example 2: Ferric trimaltol from L-lysine modified ferric hydroxide

Synthesis of lysine-modified ferric hydroxide gel

14.87g FeCl3.6H20 and 5.02g L-Lysine were added to 25 mL UHP water and stirred until dissolved. 32 mL NaOH 5M was then gradually added to this solution producing a ferric hydroxide gel .

Ferric trimaltol synthesis

7 g NaOH pellets was added to 25 mL UHP water and stirred until dissolved. Next, 24.5g maltol was added and stirred until dissolved. Next, the lysine-modified ferric hydroxide gel was gradually added to this solution with vigorous stirring. A 1.2 M HC1 solution was then used to drop the pH of the solution to 10, which was then incubated for 70 min. Finally, a dark red precipitate (i.e., ferric trimaltol) was recovered by

centrifugation (4500 rpmx5min) and dried overnight (45°C) .

Example 3: Absence of ferric hydroxide in ferric trimaltol

Ferric trimaltol is soluble in ethanol whereas ferric hydroxide (a potential contaminant) is not. As such ferric trimaltol powders produced as per Examples 1 and 2 were dissolved in ethanol. The material from Example 2 dissolved completely confirming the absence of iron hydroxides whereas the material from Example 1 did not. This supported the preference in the present invention for ligand modification, rather just surface coating, to ensure full conversion to ferric trimaltol .

Example 4: Ferric trimaltol from tartrate-modified ferric hydroxide

Synthesis of tartrate-modified ferric hydroxide gel

14.87g FeCl3.6H20 (0.055 mol) was added to 25 mL UHP water and stirred until dissolved. 4.12 g tartaric acid (0.0275 mol) was added to this solution and stirred until dissolved. 38 mL NaOH 5M was then gradually added to this solution producing a ferric hydroxide gel .

Ferric trimaltol synthesis

2 g NaOH pellets was added to 25 mL UHP water and stirred until dissolved. Next, 24.5g maltol was added and stirred. This produced a slurry in which most of the maltol remained

undissolved. Next, the tartrate-modified ferric hydroxide gel was gradually added to this solution with vigorous stirring during which the remainder of maltol dissolved. After 15 min a dark red precipitate (i.e. ferric trimaltol) had been formed and pH had stabilised at 8.5. The material was then washed by (1)

centrifuging, (2) disposing of the supernatant and (3)

resuspending in water back to its original volume. Finally, the material was recovered by centrifugation (4500 rpm x 5min) and dried overnight (50°C) . Previously disclosed synthetic processes for the production of ferric trimaltol under aqueous conditions require the addition of NaOH (or other suitable bases) for conversion of maltol from its protonated form to its deprotonated form prior to complexation of iron. However this results in the formation of unwanted sodium ions which must be washed off. In contrast, the use of ferric hydroxides according to the methods of the present invention reduces the requirements for base and associated counter cation (e.g. sodium), which is a favourable feature. Note that ferric hydroxides are represented above as Fe (OH) 3 for illustrative purposes only. Different iron hydroxides possess different structures and elemental compositions (see Cornell & Schwertmann, The Iron Oxides Structure, Properties, Reactions, Occurrence and Uses. 2nd edition, 1996, VCH Publishers, New York) . Example 5: Ferric trimaltol from tartrate-modified ferric hydroxide (with removal of contaminants from ferric hydroxide)

Material prepared as in Example 4, except excess reactants and reaction products (e.g. unbound tartaric acid, sodium chloride) were removed from the ferric hydroxide gel. This was achieved by centrifuging the ferric hydroxide gel after its synthesis and discarding the supernatant, which contained unwanted soluble species. Finally, the ferric hydroxide gel was re-suspended in water back to its original volume prior to being added to a maltol slurry.

Example 6: Ethanolic clean up for ferric trimaltol produced from ligand coated ferric hydroxide

Ferric trimaltol precipitate was purified as it contained an unwanted iron oxide fraction. Part of the wet pellet recovered by centrifugation (4.5 g) was dissolved in 1L ethanol. The iron oxide fraction (which remained undissolved) was then removed by filtration, producing a turbidity-free solution. Next, ethanol was evaporated (40°C in a rotavapor under vacuum) producing a concentrated ferric trimaltol slurry. This was then recovered and oven dried overnight at 50°C.

PATENT

https://patents.google.com/patent/WO2012101442A1/en

Comparative Example 1

Preparation of Iron Trimaltol from Pure Maltol Maltol was dissolved in an aqueous solution of ferric chloride and ferric trimaltol was precipitated upon the addition of sodium hydroxide.

An accurate mass of ferric chloride hexahydrate granules (330g) was dissolved in distilled water to yield a pH of 0.6. To this solution, an equimolar amount of maltol was added (490g in total, initially 250g) and allowed to dissolve with continuous stirring. The pH of this solution was found to be zero and the colour of this solution was deep- purple. Spectroscopy showed that the initial solution was mainly a 1 :1 Fe/maltol mixture with some 1 :2 component. The remaining maltol was added. After an hour of stirring, sodium hydroxide (147g NaOH in 750 ml water) was added dropwise to the solution until a pH of 8.3 was achieved. The solution and precipitate were red. The precipitate was collected using a Buchner funnel under vacuum. The precipitate was dried at 40°C under vacuum.

Maltol is only slightly soluble in an aqueous acidic reaction medium. After an hour of stirring, traces of undissolved maltol were visible on the surface of the ferric chloride/maltol solution, on the walls of the reaction vessel and on the stirrer. Upon addition of sodium hydroxide, there appeared to be lumps of a brownish-black substance on the walls of the reaction vessel and on the stirrer which seemed to add to the impurities in the desired product.

An attempt to heat the ferric chloride/maltol solution so as to assist the maltol to dissolve in the ferric chloride solution resulted in a burnt, off spec, colour iron maltol sample. This method also produces two by-products which consume expensive maltol namely Fe(OH)2 (Maltol) and Fe (OH) (Maltol)2.

The sodium hydroxide solution has to be added extremely slowly to prevent “gumming up” and formation of undesirable lumps at the bottom of the reaction vessel. A yield of about 78% ferric trimaltol was obtained using this method of preparation.

When maltol is added to a ferric chloride solution at a low pH, no ferric trimaltol is formed and ferric hydroxide is generated with ferric monomaltol and a small percentage of ferric dimaltol species. The charge neutralisation of these complexes is either the hydroxy! functional group or the chloride anion. This addition also results in the formation of black deposits and gums consisting of ferric chloride/ferric hydroxide polymers. These black deposits are also produced if the solutions are heated. Therefore it is not possible to obtain the correct stoichiometry for the formation of ferric trimaltol and manufacture a pharmaceutically acceptable product using this method.

The addition of maltol to an aqueous solution of ferric or ferrous chloride was deemed impractical for scale up and manufacturing purposes and Examples 2 to 4 investigate the addition of the iron chlorides to maltol in solution.

The problem of working in an aqueous environment

Ferric chloride as a hydrated ion in aqueous solution is a strong Lewis acid with a Ka of 7x 103 and ferrous chloride as a hydrated ion in aqueous solution is also a strong Lewis acid with a Ka of 5 x 10“9. Over the desired range for using iron chlorides as starting materials for the synthesis of ferric trimaltol, ferric chloride in aqueous solution has a pH value in the range of 1-3 and ferrous chloride has a pH in the range of 3-5. Furthermore, commercial solutions of iron chlorides have a pH circa 1 because they are stabilised by the addition of hydrochloric acid to prevent the precipitation of ferric hydroxide species.

The present invention recognises that maltol is virtually insoluble at these low pH values and has limited solubility when dissolved in water in the pH range 6-8. The maximum aqueous solubility is 1g/100m! at 20°C. However, the solubility of maltol can be increased to 10g/100ml by heating to near boiling temperatures. Maltol is stable in aqueous solution at these temperatures and this property has been employed in Example 4 to synthesise ferric trimaltol. At low pH values ferric trimaltol is not the preferred species due to disproportionation. In order to obtain significant amounts of ferric trimaltol using a stoichiometric ratio of iron salt to hydroxypyrone of 1 :3, the eventual pH of the solution must exceed 7 since below that pH ferric dimaltol and monomaltol species will exist. Therefore two methods of increasing the pH were researched 1) using sodium carbonate and 2) using sodium hydroxide. Other alkali hydroxides could be used such as potassium hydroxide. The sodium carbonate neutralisation was found to be less preferable due to C02 generation. This research lead to an improved synthesis of ferric trimaltol.

Example 2

Maltol was dissolved in an aqueous solution of sodium hydroxide and iron maltol was precipitated upon the addition of ferric chloride.

In view of some of the difficulties experienced in Example 1 , and the fact that maltol is very soluble in aqueous alkali hydroxide solutions, it was decided to change the manufacturing procedure.

The initial work using this method of preparation showed that a 90% yield was achieved. Various operating parameters were then optimised and the following procedure outlines the final method chosen. A yield of 95% was then achieved. An accurate mass of sodium hydroxide pellets (20g) was dissolved in distilled water to yield a pH of 13.50. An equimolar amount of maltol (63g) was added to this aqueous solution of NaOH to give a clear yellow coloured solution with a pH of 11.6. Almost immediately a stoichiometric amount of ferric chloride (45g) was added slowly to this solution to give a pH of 7.1 and a red precipitate formed, which was then collected using a Buchner funnel under vacuum. The precipitate was then dried at 40°C under vacuum.

Adding the maltol solution in sodium hydroxide to ferric chloride as in method 1 is not preferred since it gives an off spec product and gums and a black precipitate.

Maltol is very soluble in aqueous alkali hydroxide solutions giving a yellow solution. The concentration of the hydroxide solution preferably does not exceed 20%.

This method is advantageous since it has the potential to produce only one by-product viz, ferric hydroxide Fe(OH)3 which consumes some of the iron intended to complex with the maltol. This is not easily measurable in the presence of iron maltol and so the following method was used to measure the ferric hydroxide. Fe(OH)3 is insoluble in ethanol and so the iron maltol product was dissolved in ethanol. It was found that small amounts of Fe(OH)3 may be present in the batches of iron maltol synthesized according to Example 2.

Taking the extremes of the specification, in one embodiment, the amount of Fe(OH)3 present in the active material may not exceed 2 wt. % Fe(OH)3 based on the total weight of the composition. In view of its well known inert characteristics the level of this compound is adequately controlled and a final specification including controlled ferric hydroxide should be acceptable.

The mass balance for maltol and iron was closed at 99%.

A yield of 95% iron maltol was obtained using this method of preparation.

Example 3

Maltol was dissolved in an aqueous solution of Sodium Carbonate and Iron Maltol was precipitated upon the addition of Ferric Chloride.

An accurate mass of sodium carbonate (Na2C03) (53g) was dissolved in distilled water to give a solution having pH = 11.5. An equimolar amount of maltol (65g) was added to this aqueous alkali solution to give a murky creme coloured solution of pH = 9.9. A stoichiometric amount of a ferric chloride solution was added drop wise to this solution to a pH of 8.00. A further 15 grams of Na2C03 was added to this solution to increase the pH to 9.00. The remainder of the ferric chloride solution was then added to give a solution pH = 8.77 and a red coloured precipitate appeared.

The precipitate was collected using a Buchner funnel under vacuum. The precipitate was then dried at 40°C under vacuum. The release of C02 during the reaction tends to make this process less desirable due to foaming on the surface. The final product is a gellike solid when wet and the removal of moisture during drying can therefore be time consuming. The process may not be preferred but the ferric trimaltol produced could be acceptable.

Example 4 Maltol was dissolved in water and heated to a near boiling temperature and ferric or ferrous chloride was added to form a 1 :1/1:2 mixture of ferric maltol. The solution was allowed to cool and was added to maltol dissolved in sodium hydroxide. Stage 1

Depending on the batch size required, the ferric chloride was added slowly to a maltol solution in water at a pH of 6-7. The solubility of maltol is greatly enhanced up to 10g/100ml by heating to temperatures above 60°C. Addition of ferric chloride or ferrous chloride and monitoring the pH of the solution and maintaining the pH> 3 mainly produces ferric dimaltol species but very little ferric trimaltol. Above pH 3, no ferric hydroxide appeared to be generated. Ferric monomaltol and dimaltol species either with hydroxy or chloride giving the charge neutralisation are very soluble and a concentrated solution in excess of 30g/100ml can be generated. In order to obtain the correct stoichiometry for the formation of ferric trimaltol, further maltol is required and the pH needs to be corrected to values higher than 7.

As anhydrous ferrous or ferric chloride either 126g or 162g in 200ml of water can be added to a litre of water containing 120g of maltol. This ratio of iron to maltol does not provide sufficient maltol to produce any significant amounts of ferric trimaltol which does not precipitate at this stage.

Stage 2 Maltol in alkaline solution has been described as set out above. Conveniently, because maltol solutions up to 20% in sodium hydroxide have a pH circa 11.6, mixing of this solution with the ferric mono/dimaltol solutions from stage 1 yields a precipitate of ferric trimaltol with a deep characteristic burgundy red colour of high purity as determined by UV-vis spectroscopy. The filtrate yields product which is suitable for a GMP (good manufacturing process). The sodium chloride which is generated by this process is found in the supernatant since it has a much higher solubility at 35g/100ml than ferric trimaltol. The small amounts of sodium chloride in the ferric trimaltol can be reduced, if required, by washing in water. A further, surprising feature of the research resulted from work on ferrous chloride. Ferrous chloride may be substituted in stage 1 to form ferric dimaltol since the maltol was found to auto-oxidise the ferrous to ferric during the process of chelation. One aspect of this work which was considered to be potentially very useful if larger batch sizes were required arose from the finding that being a weaker Lewis acid than ferric chloride the pH of the starting solution was in excess of 3. Therefore the risk of generating ferric hydroxide was lower than with the use of ferric chloride at higher concentrations.

Ferrous and ferric chloride in solution or as a solid may be added to an alkaline solution of maltol in sodium hydroxide, combining stages 1 & 2. Providing a small excess of maltol up to about 10% is added then a precipitate of ferric trimaltol with a small amount of maltol is obtained. Such a preparation would be satisfactory as a GMP ferric trimaltol product.

 PATENT

https://patents.google.com/patent/WO2017167963A1/en

AMPLE 1

Synthesis of ferric trimaltol using ferric citrate

NaOH (12g, 0.3 moles) is dissolved in water (50 ml) to form a sodium hydroxide solution. 20 ml of the sodium hydroxide solution is placed in a separate vessel.

Ferric citrate (30g, 0.11 moles) is slowly added to the sodium hydroxide solution in the separate vessel at room temperature with gentle stirring. Further portions of the sodium hydroxide solution are added to the solution of ferric citrate, as necessary, in order to ensure that all of the ferric citrate is dissolved.

Maltol (49g, 0.39 moles) is added to the remaining volume of sodium hydroxide solution and dissolved. The pH of the maltol solution is 11.6.

The ferric citrate solution is slowly added to the maltol solution with gentle stirring. A deep red precipitate forms; the supernatant is a deep red colour.

The solution is slowly evaporated to dryness at 60 to 80° C until the material is suitable for powdering. The material is powdered and the powder is then dried to a constant weight.

The yield of the final product is 87g. The final product comprises ferric trimaltol and sodium citrate. The product was assayed, using elemental analysis, for iron and sodium content. The iron content is 7.89% (theoretical 7.8%) and the sodium content is 13.45%.

The pH of a solution of the final product in water was measured. The pH of a 1% solution of the product by total weight of aqueous solution is 9.9 at 20°C.

EXAMPLE 2

Synthesis of ferric trimaltol using ferrous fumarate

NaOH (40g, 1 mole) is dissolved in water (100 ml) to form a sodium hydroxide solution. The pH of the solution is approximately 13.0.

Ferrous fumarate (170g, 1 mole) is slowly added to the sodium hydroxide solution at room temperature with gentle stirring.

Maltol (408g, 3.23 moles) is added to a separate volume of sodium hydroxide (40g, 1 mole) dissolved in water (100 ml) and dissolved. The pH of the solution is approximately 11.

The ferrous fumarate solution is slowly added to the maltol solution with gentle stirring. A deep red precipitate forms; the supernatant is a deep red colour.

The solution is slowly evaporated to dryness at 60 to 80° C until the material is suitable for powdering. The material is powdered and the powder is then dried to a constant weight. The yield of the final product is 615g.

The final product comprises ferric trimaltol and sodium fumarate.

EXAMPLE 3

Synthesis of ferric trimaltol using sodium carbonate to vary pH

Sodium carbonate (2.5g) is dissolved in 10ml of distilled water at room temperature. The pH of the solution is 11.6. Maltol (9.6g – three molar equivalents of sodium carbonate) is added to the sodium carbonate solution to give a cream coloured solution having a pH of 10.0.

A stoichiometric amount of ferric citrate (5g, allowing for a small excess of maltol) in an aqueous solution of sodium hydroxide (lg in 5ml of distilled water) is added slowly to the solution of maltol. The pH of the combined solutions is about 9. A red precipitate appears which is separated by decantation and dried at 80°C in an oven.

The red precipitate is ferric trimaltol, as confirmed by UV-Vis spectrometry.

EXAMPLE 4

Synthesis of ferric trimaltol using ferrous gluconate

Potassium hydroxide (5.5g) is dissolved in 50ml of distilled water at room temperature. To 25ml of this solution, maltol (16.5g, 0.13 moles) is added and gently heated to form a clear solution. To the other 25ml aliquot of the potassium hydroxide solution ferrous gluconate (22.5g) is added. This is gently heated to form a dark green saturated solution. The ferrous gluconate solution is added to the maltol solution and immediately a colour change to dark brown is noted.

On cooling, a deep brown precipitate forms (which is ferric trimaltol). The supernatant is a deep brown solution containing ferric trimaltol and potassium gluconate. The precipitate and the supernatant are dried separately at 80°C in an oven. The ferric trimaltol is a deep red brown powder with a characteristic caramel odour and UV-vis spectrum in aqueous solution.

EXAMPLE 5

Synthesis of ferric trimaltol using solid ferrous gluconate

Example 4 was repeated with the modification that the maltol is added to all of the 50 ml solution of potassium hydroxide and then solid ferrous gluconate is added directly to the maltol solution. This method gives similar end products to Example 4.

EXAMPLE 6

Synthesis of ferric trimaltol using sodium ferrous citrate

A 20% solution w/v of sodium ferrous citrate in distilled water is prepared from 7.5g of sodium ferrous citrate in 37.5ml of water. The solution of sodium ferrous citrate is dark green with an iron content of about 20%. A solution of maltol (containing 10g/50ml) in 20% sodium hydroxide is added to the solution of sodium ferrous citrate. A characteristic deep red/brown iron complex of ferric trimaltol is formed.

EXAMPLE 7

Synthesis of ferric trimaltol using solid sodium ferrous citrate

Example 6 was repeated using the same amounts and concentrations of components but the method is varied in that solid sodium ferrous citrate (7.5g) is added directly to the maltol solution (containing lOg of maltol in 50ml). Ferric trimaltol is formed using this alternative method.

EXAMPLE 8

Synthesis of ferric trimaltol using sodium ferric citrate

A 20% solution w/v of sodium ferric citrate in distilled water is prepared from 7.5g of sodium ferric citrate in 37.5ml of water. The solution of sodium ferric citrate is dark brown with an iron content of about 20%.

A solution of maltol (containing 10g/50ml) in 20% sodium hydroxide is added to the solution of sodium ferric citrate. A characteristic deep red/brown iron complex of ferric trimaltol is formed. EXAMPLE 9

Example 8 was repeated using the same amounts and concentrations of components but the method is varied in that solid sodium ferric citrate (7.5g) is added directly to the maltol solution (containing lOg of maltol in 50ml). Ferric trimaltol is formed using this alternative method.

If any of Examples 3 to 9 are repeated using maltol in a neutral or acidic aqueous medium, such as for example in buffered citric acid, brown/black impurities appear and insoluble fractions are formed (probably of ferric hydroxide) and the UN-vis spectra of the solutions are not correct. In particular, there is a peak shift towards 510nm indicating the formation of mono or dimaltol complexes or compounds.

PATENT

WO 2017167970

POLYMORPH

GB 2531742

PATENT

WO 2016066555

https://patents.google.com/patent/WO2016066555A1/en

An adequate supply of iron to the body is an essential requirement for tissue growth and the maintenance of good health in both man and animals. Moreover, in certain pathological conditions where there is an insidious blood loss, or where there is a mal-distribution of iron in the body, there may be a state of low iron stores in the body leading to an iron deficiency and a concomitant chronic anaemia. This is seen in inflammatory diseases of the gastrointestinal tract, such as gastric and peptic ulcers, reflux oesophagitis, ulcerative colitis and Crohn’s disease.

Anaemia can also follow operations that result in serious blood loss and can be associated with gastrointestinal infections, such as those caused by Helicobacter pylori.

Ferric maltol comprises a complex of one ferric iron and three maltol anions and has the following molecular formula: (C6H503)3Fe. Maltol is also known as 3-hydroxy-2-methyl-4- pyrone.

Polymorphic forms occur where the same composition of matter crystallises in a different lattice arrangement, resulting in different thermodynamic properties and stabilities specific to the particular polymorphic form. WO 03/097627 A1 discloses a method of forming iron hydroxypyrone compounds.

EP 0 159 917 A3 describes a pharmaceutical composition containing a hydroxypyrone-iron complex. WO 2012/101442 A1 discloses a method of forming iron hydroxypyrone compounds.

Schlindwein et al (Dalton Transactions, 2006, Vol. 10, pages 1313-1321) describes lipophilic 3-hydroxy-4-pyridinonate iron(lll) complexes. Ferric maltol has been known for about 100 years but no polymorphs have been identified or studied prior to this invention.

We have now found that it is possible to produce different polymorphs of ferric maltol, which crystalline forms may be referred to herein as the “compounds of the invention”. One polymorph form can be preferable in some circumstances when certain aspects, such as ease of preparation and stability, such as thermodynamic stability are required. In other situations, a different polymorph may be preferred for greater solubility and/or superior pharmacokinetics. The polymorphs of the invention can provide advantages in terms of improved or better bioavailability or improved or better stability or solubility.

The term “ferric maltol” as used herein refers to both ferric trimaltol and the designation INN ferric maltol. In one aspect of the invention there is provided a Form I polymorph of ferric maltol characterized by a powder X-ray diffraction pattern comprising characteristic crystalline peaks expressed in degrees 2-theta at each of 15.6 and 22.5 ± 0.25 or 0.2 degrees, optionally wherein the Form I polymorph comprises greater than about 92 wt.% ferric maltol based on the weight of the polymorph, such as greater than about 95 wt.%, preferably greater than about 96 wt.%, or about 98 wt.%, or about 99 wt.% such as about 99.8 wt.%.

In a further aspect of the invention there is provided a Form II polymorph of ferric maltol characterized by a powder X-ray diffraction pattern comprising a peak expressed in degrees 2-theta at 8.3 ± 0.25 degrees.

In a yet further aspect of the invention there is provided a Form III polymorph of ferric maltol characterized by a powder X-ray diffraction pattern comprising a peak expressed in degrees 2-theta at 7.4 ± 0.25 degrees. In a still further aspect of the invention there is provided a Form IV polymorph of ferric maltol characterized by a powder X-ray diffraction pattern comprising peaks expressed in degrees 2-theta at 9.5 and 14.5 ± 0.2 degrees.

The measurements of degrees 2-theta generally refer to measurements at ambient temperature, such as from about 5 to about 40°C, preferably about 10 to about 30°C. The relative intensities of the peaks can vary, depending on the sample preparation technique, the sample mounting procedure, the particular instrument employed, and the morphology of the sample. Moreover, instrument variation and other factors can affect the 2-theta values. Therefore, XRPD peak assignments for the polymorphs of the invention, as defined herein in any embodiment, can vary by, for example, ± 0.2, such as ±0.1 or ±0.05. The term “about” in relation to XRPD peak values may include for example, ±0.25 or ± 0.2, such as ±0.1 or ±0.05. These ranges may apply to any of the peak values in degrees referred to herein.

In another embodiment of the invention, there is provided a process for the preparation of a ferric maltol polymorph, such as Form I or Form II polymorph, which comprises combining ferric citrate with maltol anions to form a mixture comprising ferric maltol and wherein the process comprises the use of a ferric maltol seed crystal. The seed crystal may comprise a Form I and/or Form II polymorph as described herein and these polymorphs may be prepared using the methods described herein.

In another aspect of the invention, there is provided a process for the preparation of Form I polymorph, which comprises combining ferric citrate with maltol anions to form a mixture comprising ferric maltol polymorph Form I wherein the process comprises the use of a ferric maltol seed crystal comprising Form I and/or Form II polymorph and preferably wherein the polymorph formed is washed (typically with water) prior to drying.

In a further aspect of the invention, there is provided a process for the preparation of Form II polymorph, which comprises combining ferric citrate with maltol anions in solution to form a mixture comprising ferric maltol polymorph Form II, wherein the process preferably comprises the use of a ferric maltol seed crystal comprising Form I and/or Form II polymorph and preferably wherein the polymorph formed is washed (typically with water) prior to drying.

The invention also provides a pharmaceutical composition comprising a polymorph according to the invention, or mixtures thereof, and a pharmaceutically acceptable adjuvant, diluent or carrier. In addition, the invention provides a composition comprising Form I and Form II polymorphs as defined herein.

In an aspect of the invention, the polymorph of the invention is for use in the prevention or treatment of iron deficiency with or without anaemia in a subject. The anaemia is preferably iron deficiency anaemia.

In a further aspect of the invention there is provided the use of a polymorph of the invention for the manufacture of a medicament for the prevention or treatment of iron deficiency with or without anaemia in a subject. The anaemia is preferably iron deficiency anaemia.

The invention further provides a method for the prevention or treatment of iron deficiency with or without anaemia which method comprises the administration of a polymorph according to the invention to a subject in need of such treatment. The anaemia is preferably iron deficiency anaemia.

Preferably the polymorphs of the invention are obtained in forms that are greater than about 90%, such as greater than about 95%, crystalline (e.g. greater than about 98% crystalline and, particularly, 100%, or nearly 100%, crystalline). By “substantially crystalline” we include greater than about 60%, preferably greater than about 75%, and more preferably greater than about 80% (such as about 90%) crystalline. The degree (%) of crystallinity may be determined by the skilled person using X-ray powder diffraction (XRPD). Other techniques, such as solid state NMR, FT-IR, Raman spectroscopy, differential scanning calorimetry (DSC) microcalorimetry and calculations of the true density may also be used.

The polymorphs of the invention may be characterised by an X-ray powder diffraction pattern comprising the following characteristic crystalline peaks with approximate 2-Theta values (in degrees) as well as an indication of the relative intensity of those peaks in brackets, where a percentage relative intensity of approximately 25- 00% is termed “vs” (very strong), approximately 10-25% is termed “s” (strong), approximately 3-10% is termed “m” (medium) and approximately 1-3% is termed “w” (weak).

Form I: The Form I polymorph preferably comprises characteristic crystalline peaks with 2-Theta values (in degrees) of around (i.e. at about or at approximately) 15.6 and 22.5 ± 0.25, or 0.2 degrees. The diffraction pattern typically does not comprise peaks at one or more, or all, or each of, about 6.9, 7.4, 8.3, 9.3, 10.5, or about 11.8 degrees, such as 8.3 or 11.8 ± 0.25, or ± 0.2, or ±0.1 such as about ±0.05 degrees.

Form II:

The form II polymorph preferably comprises a characteristic crystalline peak with 2-Theta value (in degrees) of around (i.e. at about or at approximately) 8.3 ± 0.25, or ± 0.2, or +0.1 such as about ±0.05 degrees. The diffraction pattern typically does not comprise peaks at one or more, or all, or each of, about 6.9, 7.4, 9.3, 9.5, 10.5, 11.4 or about 13.7 degrees, such as 11.4 or 13.8 ±0.25, or ±0.2, or ±0.1 such as about ±0.05 degrees.

The Form III polymorph preferably comprises a characteristic crystalline peak with 2-Theta value (in degrees) of around (i.e. at about or at approximately) 7.4 ±0.3, ±0.25, or 0.2, or ±0.1 such as about ±0.05 degrees. The diffraction pattern typically does not comprise peaks at one or more, or two or more, or three or more or each of, about 6.9, 8.3, 9.5, 11.3, 12.0, 12.5, 12.9, 14.5, or about 15.8 degrees, such as 6.9, 9.5, 11.3 ±0.25, or ±0.2, or ±0.1 such as about ±0.05 degrees.

The form IV polymorph preferably comprises a characteristic crystalline peaks with 2-Theta values (in degrees) of around (i.e. at about or at approximately) 9.5 and 14.5 +0.2, or ±0.1 such as about ±0.05 degrees. The diffraction pattern typically does not comprise peaks at one or more, or two or more, or three or more or each of, about 6.9, 8.3, 10.5, 11.7, 12.0, 12.2, 12.5, 13.0, 13.4, and about 15.8 degrees, such as 6.9, 8.3, 11.7 ±0.25, or ±0.2, or ±0.1 such as about ±0.05 degrees.

Example 1 : Form I 9.04 kg ferric citrate was combined with 29 litres of purified water. Separately, 12.2 kg of maltol was combined with 15.2 litres of sodium hydroxide solution (20 % w/w). The ferric citrate and sodium hydroxide were charged into a vessel with the addition of 4 litres of water and then stirred at 20 to 25°C. A seed was then added. The seed was 65g of ferric maltol polymorph in 12 litres of water. The seed crystal was prepared by the same process as described in Example 1 but without the use of a seed crystal. The seed was added to the vessel to aid a consistent crystallisation/precipitation. The mixture was held in the vessel, as a suspension, to allow crystal growth and then filtered and washed three times, each time with 13 litres of water. The resulting solid was dried at less than 80°C and produced 13.25 kg of dried ferric maltol.

The ferric maltol in Example 1 was produced on a scale of 12 to 15 kg in different batches. The analysis of the ferric maltol produced showed the % w/w of iron present was about 12.8 to 13.0 and the % w/w of maltol present was about 87.6 to 89.3.

Patent

Publication numberPriority datePublication dateAssigneeTitle
EP0159917A2 *1984-04-191985-10-30National Research Development CorporationPharmaceutical composition containing a hydroxypyrone-iron complex
WO2003097627A1 *2002-05-182003-11-27Vitra Pharmaceuticals LimitedMethod of forming iron hydroxypyrone compounds
WO2012101442A1 *2011-01-272012-08-02Iron Therapeutics Holdings AgProcess
Family To Family Citations
EP0107458B1 *1982-10-221987-07-29National Research Development CorporationPharmaceutical compositions
GB2531742B2014-10-282016-10-05Iron Therapeutics Holdings AgPolymorphs of ferric maltol
* Cited by examiner, † Cited by third party

Publication numberPriority datePublication dateAssigneeTitle
Family To Family Citations
GB2531742B2014-10-282016-10-05Iron Therapeutics Holdings AgPolymorphs of ferric maltol
WO2003097627A1 *2002-05-182003-11-27Vitra Pharmaceuticals LimitedMethod of forming iron hydroxypyrone compounds
US20080188555A1 *2007-02-062008-08-07Jonathan Joseph PowellLigand modified poly oxo-hydroxy metal ion materials, their uses and processes for their preparation
WO2012101442A1 *2011-01-272012-08-02Iron Therapeutics Holdings AgProcess
REFERENCES
Inorganica Chimica Acta (1990), 170(2), 241-3
Dalton Transactions (2006), (10), 1313-1321.
EP 0,107,458 [ 1984, to National Research Development Corporation]
Journal of Chemical Research, Synopses (1980), (9), 314.
Chemistry for Sustainable Development (2007) 15(4), PP- 448 – 458
US 5,028,411 [ 1991, to National Research Development Corporation]
Journal of Coordination Chemistry (1978), 8(1), 27-33
Polyhedron (1988), 7(19-20), 1973-9.
US 7,459,569 [2008, to Vitra Pharmaceuticals Limited].
Journal of Pharmaceutical Sciences (1972), 61(8), 1209-12
WO 2012 / 101,442 [ 2012, to Iron Therapeutics Holdings Ag]
Chemistry Letters (1975), (4), 339-42

//////////////Ferric Maltol, マルトール第二鉄 , Feraccru

CC1=C(C(=O)C=CO1)[O-].CC1=C(C(=O)C=CO1)[O-].CC1=C(C(=O)C=CO1)[O-].[Fe+3]

Vericiguat, ベルイシグアト

$
0
0

Vericiguat.pngImage result for vericiguatImage result for vericiguat

Vericiguat

BAY 102; BAY-1021189; MK-1242

1350653-20-1
Chemical Formula: C19H16F2N8O2

Molecular Weight: 426.3878

Vericiguat; 1350653-20-1; UNII-LV66ADM269; Methyl (4,6-diamino-2-(5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl)pyrimidin-5-yl)carbamate; BAY-1021189; LV66ADM269

Methyl (4,6-diamino-2-(5-fluoro-1-((2-fluorophenyl)methyl)-1H-pyrazolo(3,4-b)pyridin-3-yl(pyrimidin-5-yl)carbamate

methyl N-[4,6-diamino-2-[5-fluoro-1-[(2-fluorophenyl)methyl]pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl]carbamate

Methyl{4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridi- n-3-yl]pyrimidin-5-yl}carbamate

  • Originator Bayer HealthCare Pharmaceuticals
  • Developer Bayer HealthCare Pharmaceuticals; Merck & Co
  • Mechanism of Action Guanylate cyclase stimulants
  • Phase III Chronic heart failure
  • Phase I Coronary artery disease
  • 28 May 2018 Phase II VITALITY HFpEF trial for Chronic heart failure in Austria, USA, Belgium, Portugal, Canada, Spain, Hungary and Greece (PO) (EudraCT2018-000298-65) (NCT03547583)
  • 17 May 2018 Phase-I clinical trials in Coronary artery disease (In adults, In the elderly) in Moldova and Germany (PO) (NCT03504982)
  • 20 Apr 2018 Bayer in collaboration with Merck Sharp & Dohme Corp. plans a phase I trial for Coronary Artery Disease in the Netherlands, Moldova and Germany (NCT03504982)

Vericiguat, also known as BAY1021189 or BAY10-21189, is a potent and orally active sGC stimulator (Soluble Guanylate Cyclase Stimulator). Direct stimulation of soluble guanylate cyclase (sGC) is emerging as a potential new approach for the treatment of renal disorders. sGC catalyzes the formation of cyclic guanosine monophosphate (cGMP), deficiency of which is implicated in the pathogenesis of chronic kidney disease (CKD).

Vericiguat, discovered at Bayer, is the first soluble guanylate cyclase (sGC) stimulator. Vericiguat is currently being studied in a Phase III clinical program for the treatment of heart failure with reduced ejection fraction (HFrEF)

ベルイシグアト
Vericiguat

C19H16F2N8O2 : 426.38
[1350653-20-1]

Vericiguat hydrochloride.png

Vericiguat hydrochloride

cas 1350658-96-6

PHASE 3 MERCK/BAYER

Chemical Names: UNII-5G76IGF54K; 5G76IGF54K; ; 1350658-96-6; Carbamic acid, N-(4,6-diamino-2-(5-fluoro-1-((2-fluorophenyl)methyl)-1H-pyrazolo(3,4-b)pyridin-3-yl)-5-pyrimidinyl)-, methyl ester, hydrochloride (1:1); Methyl (4,6-diamino-2-(5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo(3,4-b)pyridin-3-yl)pyrimidin-5-yl)carbamate hydrochloride
Molecular Formula: C19H17ClF2N8O2
Molecular Weight: 462.846 g/mol

Image result for DRUG FUTURE Vericiguat

Clip

https://www.thieme-connect.com/products/ejournals/pdf/10.1055/s-0036-1590758.pdf

Image result for vericiguat

Significance: Vericiguat (BAY 1021189) is an orally available soluble guanylate cyclase (sGC) stimulator that has entered phase-three trials for the once-daily treatment of chronic heart failure. Key steps in the synthesis depicted are (1) construction of the 5-fluoro-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridine-3-carboxylate C by condensation of the 5-amino-1H-pyrazole-3-carboxylate A with the aldehyde B and (2) construction of the pyrimidine-4,5,6-triamine derivative H through reaction of [(E)-phenyldiazenyl]malononitrile (G) with amidine F.

Comment: Experimental details are provided for the noteworthy four-step synthesis (not shown) of the crystalline 2-fluoro-(3-morpholin-4-yl)acrylaldehyde B from commercially available 2,2,3,3- tetrafluoro-1-propanol. The synthesis of pyrazole A is described in a patent (A. Straub et al. WO 2000/006569 A1). The [(E)-phenyldiazenyl]malononitrile (G) was generated in situ by reaction of phenyldiazonium chloride with malononitrile.

M. FOLLM ANN * E T AL. (BAYER AG, WUPPERTAL , GE RMANY) Discovery of the Soluble Guanylate Cyclase Stimulator Vericiguat (BAY 1021189) for the Treatment of Chronic Heart Failure J. Med. Chem. 2017, 60, 5146–5161
Clip
Image result for vericiguat
Image result for vericiguat
Image result for vericiguat
Image result for vericiguatImage result for vericiguatImage result for vericiguat
24. Yield 2.2 g (70%). 1 H NMR (400 MHz, DMSO-d6): δ = 8.89 (dd, J = 9.0, 2.8 Hz, 1H), 8.66 (m, 1H), 7.99 and 7.67 (2 br s, 1H), 7.32−7.40 (m, 1H), 7.19−7.26 (m, 1H), 7.10−7.19 (m, 2H), 6.22 (br s, 4H), 5.79 (s, 2H), 3.62 (br s, 3H). LC-MS (method d): tR (min) = 0.79. MS (ESI +): m/z = 427 [M + H]+
PATENT
US 8,802,847

Example 13

Methyl{4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridi- n-3-yl]pyrimidin-5-yl}carbamate

Method A:

4.0 g (77.0% by weight, 8.36 mmol) of the compound from Example 12 in 37.9 ml of isopropanol were heated to 35.degree. C. and then 0.84 ml (10.87 mmol) of methyl chloroformate was added dropwise. The mixture was stirred at 35.degree.-40.degree. C. for 20 h and heated to 50.degree. C., and 9.5 ml of methanol were added. Subsequently, 1.9 ml of triethylamine were added dropwise within 0.5 h and rinsed in with 1.3 ml of methanol, and the mixture was stirred at 50.degree. C. for 1 h. Thereafter, the reaction mixture was cooled to RT and stirred at RT for 1 h, and the solids were filtered off with suction, washed three times with 8 ml each time of ethanol, suction-dried and dried in a vacuum drying cabinet at 50.degree. C. under a gentle nitrogen stream. This gave 3.4 g of crude product. 3.0 g of the crude product were stirred in 8 ml of DMSO for 5 min, 13.0 ml of ethyl acetate and 50 mg of activated carbon were added, and the mixture was heated at reflux (84.degree. C.) for 15 min. The suspension was hot-filtered and the filter residue was washed with 1.9 ml of ethyl acetate.sup.1). 60 ml of ethyl acetate and 16 ml of ethanol were heated to 60.degree. C., and the combined filtrates were added dropwise and stirred at 60.degree. C. for 1.5 h. The suspension was cooled to RT within 25 min, stirred for a further 1.5 h, cooled further to 0.degree.-5.degree. C. and stirred for a further 1 h. The solids were filtered off with suction, washed twice with 6.4 ml each time of ethyl acetate, suction-dried and dried in a vacuum drying cabinet at 50.degree. C. under a gentle nitrogen stream. This gave 2.2 g (70.0% of theory) of the title compound. 1) According to the preparation process described, the di-dimethyl sulphoxide solvate is obtained at this point, and this is characterized in Tables 2 and 4 by the reflections in the x-ray diffractogram and bands in the IR spectrum.

MS (ESIpos): m/z=427 (M+H).sup.+

.sup.1H NMR (400 MHz, DMSO-d.sub.6): .delta.=3.62 (br s, 3H), 5.79 (s, 2H), 6.22 (br s, 4H), 7.10-7.19 (m, 2H), 7.19-7.26 (m, 1H), 7.32-7.40 (m, 1H), 7.67 and 7.99 (2 br s, 1H), 8.66 (m, 1H), 8.89 (dd, 1H) ppm.

The di-dimethyl sulphoxide solvate of the compound of the formula (I) has the advantage of much better filterability than the substance in the prior art. Furthermore, the preparation process via the di-dimethyl sulphoxide solvate of the compound of the formula (I) leads to a very high purity of the compound of the formula (I).

Method B:

4.0 g (10.8 mmol) of the compound from Example 12 Method B in 37.9 ml of isopropanol were heated to 35.degree. C. and then 1.1 ml (14.1 mmol) of methyl chloroformate were added dropwise. The mixture was stirred at 35.degree.-40.degree. C. for 16.5 h and cooled to RT, and 2.1 ml of aqueous ammonia (28%) were added. Subsequently, 4.2 ml of water were added and the mixture was stirred for 2.5 h. The solids were filtered off with suction, washed twice with 5 ml each time of water, suction-dried and dried in a vacuum drying cabinet at 50.degree. C. under a gentle nitrogen stream. This gave 4.4 g of crude product.

Method C:

4.0 g (10.8 mmol) of the compound from Example 12 Method B in 37.9 ml of isopropanol were heated to 35.degree. C. and then 1.1 ml (14.1 mmol) of methyl chloroformate were added dropwise. The mixture was stirred at 35.degree.-40.degree. C. for 16.5 h, and 9.5 ml of methanol were added at 50.degree. C. Subsequently, 2.42 ml of triethylamine were added dropwise within 20 min and rinsed in with 1.3 ml of methanol, and the mixture was stirred at 50.degree. C. for 1 h. Thereafter, the reaction mixture was cooled to RT and stirred at RT for 1 h, and the solids were filtered off with suction, washed three times with 8 ml each time of methanol, suction-dried and dried in a vacuum drying cabinet at 50.degree. C. under a gentle nitrogen stream. This gave 4.3 g of crude product.

Method D:

6.9 g of the crude product were stirred in 18.4 ml of DMSO for 5 min, 30.0 ml of ethyl acetate and 115 mg of activated carbon were added, and the mixture was heated at reflux (84.degree. C.) for 15 min. The suspension was hot-filtered and the filter residue was washed with 4.4 ml of ethyl acetate. 138 ml of ethyl acetate were heated to 50.degree. C., and the combined filtrates were added dropwise and stirred at 45-50.degree. C. for 1 h. The suspension was cooled to 0.degree.-5.degree. C. within 1.5 h and stirred for a further 1 h. The solids were filtered off with suction, washed twice with 14.8 ml each time of ethyl acetate and suction-dried for 1 h. 6.4 g of the di-dimethyl sulphoxide solvate were obtained as a moist product.sup.1).

Method E:

2.0 g of the di-dimethyl sulphoxide solvate were stirred at reflux temperature in 40 ml of ethyl acetate and 11.1 ml of ethanol for 17 h, cooled to RT and stirred for a further 1 h. The solids were filtered off with suction, washed four times with 1.4 ml each time of ethyl acetate and dried in a vacuum drying cabinet at 50.degree. C. under a gentle nitrogen stream. This gave 1.4 g of the title compound present in polymorph I.

Method F:

0.5 g of the di-dimethyl sulphoxide solvate were stirred at reflux temperature in 12.5 ml of solvent for 17 h, cooled to RT and stirred for a further 1 h. The solids were filtered off with suction, washed with 2 ml of solvent and suction-dried for 30 min. This gave 0.3 g of the title compound present in polymorph I.

The following solvents were used:

1.) 9 ml of ethyl acetate/3.5 ml of ethanol/0.3 ml of water

2.) 12.5 ml of isopropanol

3.) 12.5 ml of isopropanol/0.3 ml of water

4.) 12.5 ml of methanol

5.) 12.5 ml of methanol/0.3 ml of water

6.) 12.5 ml of acetonitrile

7.) 12.5 ml of acetone

8.) 12.5 ml of tetrahydrofuran,

9.) 12.5 ml of methyl tert-butyl ether

Table 1 indicates the reflections of the x-ray diffractogram. Table 3 shows the bands of the IR spectrum.

The compound (I) in crystalline polymorph I is notable for higher stability and more particularly for the fact that it is stable in the micronization process and hence no conversion and recrystallization takes place.

The compound of the formula (I) can be prepared by processes described above. This affords the compound of the formula (I) in a crystal polymorph referred to hereinafter as polymorph I. Polymorph I has a melting point of 257.degree. C. and a characteristic x-ray diffractogram featuring the reflections (2 theta) 5.9, 6.9, 16.2, 16.5, 24.1 and 24.7, and a characteristic IR spectrum featuring the band maxima (in cm.sup.-1) 1707, 1633, 1566, 1475, 1255 and 1223 (Tables 1 and 3, FIGS. 1 and 5).

Surprisingly, four further polymorphs, a monohydrate, a dihydrate, a DMF/water solvate and a di-dimethyl sulphoxide solvate, and also a triacetic acid solvate of the compound of the formula (I) were found. The compound of the formula (I) in polymorph II melts at approx. 253.degree. C.; the compound of the formula (I) in polymorph III has a melting point of approx. 127.degree. C. Polymorph IV of the compound of the formula I melts at a temperature of 246.degree. C., while polymorph V has a melting point of 234.degree. C. The monohydrate contains approx. 4.1% water, the dihydrate contains 7.8% water, the DMF/water solvate contains 13.6% dimethylformamide and 0.9% water, the di-DMSO solvate contains 26.8% dimethyl sulphoxide and the triacetic acid solvate contains 29.7% acetate. Each of the crystalline forms mentioned has a characteristic x-ray diffractogram and IR spectrum (Tables 2 and 3, FIGS. 1-4, 6-14).

TABLE 1
X-ray diffractometry for polymorphs I to V

FIGURES

FIG. 1: IR spectrum of the compound of the formula (I) in polymorphs I, II and III

FIG. 2: IR spectrum of the compound of the formula (I) in polymorphs IV, V and as the triacetic acid solvate

FIG. 3: IR spectrum of the compound of the formula (I) as the di-DMSO solvate, DMF/water solvate and monohydrate

FIG. 4: IR spectrum of the compound of the formula (I) as the dihydrate

FIG. 5: X-ray diffractogram of the compound of the formula (I) in polymorph I

FIG. 6: X-ray diffractogram of the compound of the formula (I) in polymorph II

FIG. 7: X-ray diffractogram of the compound of the formula (I) in polymorph III

FIG. 8: X-ray diffractogram of the compound of the formula (I) in polymorph IV

FIG. 9: X-ray diffractogram of the compound of the formula (I) in polymorph V

FIG. 10: X-ray diffractogram of the compound of the formula (I) as the triacetic acid solvate

FIG. 11: X-ray diffractogram of the compound of the formula (I) as the di-DMSO solvate

FIG. 12: X-ray diffractogram of the compound of the formula (I) as the DMF-water solvate

FIG. 13: X-ray diffractogram of the compound of the formula (I) as the monohydrate

FIG. 14: X-ray diffractogram of the compound of the formula (I) as the dihydrate

PATENT

Example 11A

2-[5-Fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidine-4,5,6-triamine

      Variant A: Preparation Starting from Example 7A:
      In pyridine (30 ml), 378 mg (0.949 mmol) of the compound from Example 7A were introduced and then 143 mg (0.135 mmol) of palladium (10% on carbon) were added. The mixture was hydrogenated overnight at RT under standard hydrogen pressure. The suspension was then filtered through kieselguhr and the filtercake was washed with ethanol. The filtrate was concentrated and yielded 233 mg (81% purity, 51% of theory) of the desired compound, which was reacted without further purification.
      Variant B: Preparation Starting from Example 10A:
      In DMF (800 ml), 39.23 g (85.75 mmol) of the compound from Example 10A were introduced and then 4 g of palladium (10% on carbon) were added. The mixture was hydrogenated with stirring overnight under standard hydrogen pressure. The batch was filtered over kieselguhr and the filter product was washed with a little DMF and then with a little methanol, and concentrated to dryness. The residue was admixed with ethyl acetate and stirred vigorously, and the precipitate was filtered off with suction, washed with ethyl acetate and diisopropyl ether and dried under a high vacuum over Sicapent.
      Yield: 31.7 g (100% of theory)
      LC-MS (method 2): R t=0.78 min
      MS (ESIpos): m/z=369 (M+H) +

Working Examples

Example 1

Methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate

      In pyridine (600 ml), 31.75 g (86.20 mmol) of the compound from Example 11A were introduced under argon and cooled to 0° C. Then a solution of 6.66 ml (86.20 mmol) of methyl chloroformate in dichloromethane (10 ml) was added dropwise and the mixture was stirred at 0° C. for 1 h. Thereafter the reaction mixture was brought to RT, concentrated under reduced pressure and co-distilled repeatedly with toluene. The residue was stirred with water/ethanol and then filtered off on a frit, after which it was washed with ethanol and ethyl acetate. Subsequently the residue was again stirred with diethyl ether, isolated by filtration with suction and then dried under a high vacuum.
      Yield: 24.24 g (65% of theory)
      LC-MS (method 2): R t=0.79 min
      MS (ESIpos): m/z=427 (M+H) +
       1H NMR (400 MHz, DMSO-d 6): δ=3.62 (br. s, 3H), 5.79 (s, 2H), 6.22 (br. s, 4H), 7.10-7.19 (m, 2H), 7.19-7.26 (m, 1H), 7.32-7.40 (m, 1H), 7.67 and 7.99 (2 br. s, 1H), 8.66 (m, 1H), 8.89 (dd, 1H).
Patent ID Title Submitted Date Granted Date
US2016324856 USE OF SGC STIMULATORS FOR THE TREATMENT OF NEUROMUSCULAR DISORDERS
2015-01-13
US2016158233 SGC STIMULATORS OR SGC ACTIVATORS AND PDE5 INHIBITORS IN COMBINATION WITH ADDITIONAL TREATMENT FOR THE THERAPY OF CYSTIC FIBROSIS
2014-07-21
2016-06-09
US2013158028 USE OF STIMULATORS AND ACTIVATORS OF SOLUBLE GUANYLATE CYCLASE FOR TREATING SICKLE-CELL ANEMIA AND CONSERVING BLOOD SUBSTITUTES
2011-06-21
2013-06-20
US9845300 PROCESS FOR PREPARING SUBSTITUTED 5-FLUORO-1H-PYRAZOLOPYRIDINES
2017-02-17
US9604948 PROCESS FOR PREPARING SUBSTITUTED 5-FLUORO-1H-PYRAZOLOPYRIDINES
2015-07-10
2016-01-14
Patent ID Title Submitted Date Granted Date
US2017273977 SUBSTITUTED 5-FLUORO-1H-PYRAZOLOPYRIDINES AND THEIR USE
2016-11-10
US8921377 Substituted 5-fluoro-1H-pyrazolopyridines and their use
2013-03-27
2014-12-30
US8420656 Substituted 5-fluoro-1H-pyrazolopyridines and their use
2012-01-26
US9096592 BICYCLIC AZA HETEROCYCLES, AND USE THEREOF
2011-08-31
2014-05-29
US2014038956 Use of sGC stimulators, sGC activators, alone and combinations with PDE5 inhibitors for the treatment of systemic sclerosis (SSc).
2011-05-24
2014-02-06

////////////////Vericiguat,  BAY 102, BAY-1021189, MK-1242, ベルイシグアト , PHASE 3,  MERCK, BAYER

COC(=O)NC1=C(N=C(N=C1N)C2=NN(C3=NC=C(C=C23)F)CC4=CC=CC=C4F)N

Viewing all 2871 articles
Browse latest View live


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