AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

What Does 100% of Your Daily Value of Cholesterol Look Like?

 Uncategorized  Comments Off on What Does 100% of Your Daily Value of Cholesterol Look Like?
Feb 262014
 

Healthline just published an interesting infograph that gives a visualization of what your daily value of cholesterol looks like.  In the graphic, you can see what 300 mg of cholesterol looks like for 20 high cholesterol foods: http://www.healthline.com/health/high-cholesterol/daily-value

This is a very informative resource as it helps us visualize what their cholesterol intake look like

What Does 100% of Your Daily Value of Cholesterol Look Like?

It’s no secret that eating fatty foods raises your bad cholesterol level, also known as LDL. An elevated LDL clogs up your arteries and makes it difficult for your heart to do its job. Potentially, it could lead to heart disease.

The USDA recommends consuming no more than 300 mg of cholesterol a day. While a deep-fried Twinkie at the county fair is an obvious no-no, other high cholesterol culprits may be sneaking into your diet. Check out what that number looks like in terms of everyday food items.

Warning: you may need to revise your grocery list—and your eating habits!

Image

Fried Chicken:

4 pieces=300mg cholesterol

Image

Croissants:

6 2/3 rolls=300mg cholesterol

Image

Cheddar Cheese:

12 3/4 slices=300mg cholesterol

Image

Prosciutto:

28 slices=300mg cholesterol

Image

Corned Beef:

14 thin slices=300mg cholesterol

Image

Butter:

1 1/5 sticks=300mg cholesterol

read at

http://www.healthline.com/health/high-cholesterol/daily-value

Share

CANGRELOR

 Uncategorized  Comments Off on CANGRELOR
Feb 242014
 

 

File:Cangrelor.png

Cangrelor, AR-C69931MX

[dichloro-[[[(2R,3S,4R,5R)-3,4-dihydroxy-5-[6-(2-methylsulfanylethylamino)-2-(3,3,3-trifluoropropylsulfanyl)purin-9-yl]oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-hydroxyphosphoryl]methyl]phosphonic acid

N-[2-(Methylthio)ethyl]-2-[(3,3,3-trifluoropropyl)thio]-5¢-adenylic acid monoanhydride with (dichloromethylene)bis[phosphonic acid]

163706-06-7 cas no

Also known as: AR-C69931XX, 163706-06-7, Cangrelor (USAN/INN), Cangrelor [USAN:INN:BAN], UNII-6AQ1Y404U7, cangrelor (AR-C69931MX),
Molecular Formula: C17H25Cl2F3N5O12P3S2
Molecular Weight: 776.359196
Cangrelor

MAR 09, 2013

The Medicines Company said yesterday it will pursue marketing approvals for its anti-clotting drug candidate Cangrelor after it met its primary efficacy endpoint in a Phase III clinical trial of improvement compared with Plavix (clopidogrel).

The intravenous small molecule antiplatelet agent reduced by 22% the likelihood of patients experiencing death, myocardial infarction, ischemia-driven revascularization, or stent thrombosis within 48 hours of taking it—to 4.7% from 5.9% of subjects randomized during CHAMPION PHOENIX. The Phase III trial compared Cangrelor to oral Plavix in 11,145 patients undergoing percutaneous coronary intervention.

Cangrelor also showed a 38% reduction (0.8% compared with 1.4%) over Plavix in the likelihood of patients experiencing the key secondary endpoint, incidence of stent thrombosis at 48 hours.

Cangrelor is designed to prevent platelet activation and aggregation that leads to thrombosis in acute care settings, including in patients undergoing percutaneous coronary intervention. During CHAMPION PHOENIX, Cangrelor made its best showing in patients with Q-wave myocardial infarction (QMI), lowering by 39% (to 0.2% compared with 0.3%) the incidence of QMI. Cangelor’s most disappoint showing was its inability to lower the odds of death compared with Clopidogrel; both drugs showed a likelihood of 0.3%.

“Our next step is to submit for market approvals in the U.S. and Europe. We anticipate submitting these data for a new drug application to the U.S. Food and Drug Administration in the second quarter with findings of prior trials, including the BRIDGE trial in patients awaiting open heart surgery,” Simona Skerjanec, PharmD, senior vp and innovation leader for antiplatelet therapies at The Medicines Company, said in a statement.

Cangrelor is a P2Y12 inhibitor under investigation as an antiplatelet drug[1] for intravenous application. Some P2Y12 inhibitors are used clinically as effective inhibitors of adenosine diphosphate-mediated platelet activation and aggregation.[1] Unlike clopidogrel (Plavix), which is a prodrug, cangrelor is an active drug not requiring metabolic conversion.

Poor interim results led to the abandonment of the two CHAMPION clinical trials in mid 2009.[2] The BRIDGE study, for short term use prior to surgery, continues.[3] The CHAMPION PHOENIX trial was a randomized study of over 11,000 patients published in 2013. It found usefulness of cangrelor in patients getting cardiac stents. Compared with clopidogrel given around the time of stenting, intravenous ADP-receptor blockade with cangrelor significantly reduced the rate of stent thrombosis and myocardial infarction.[4]Reviewers have questioned the methodology of the trial.[5]

One particularly preferred example of a reversible, short-acting P2Y12 inhibitor is cangrelor. Cangrelor is a potent, direct, and reversible antagonist of the platelet P2Y12 receptor. Cangrelor has a half-life of approximately less than 10 minutes, allowing for a return to normal platelet function in a very short period of time upon discontinuation of the drug. By reducing the need for a compound to be metabolized for activity, and by having a relatively short half-life, reversible, short-acting P2Y12 inhibitors are considered “reversible,” meaning that full platelet functionality may return rather quickly as compared to thienopyridines.

The binding of cangrelor to the P2Y12 receptor inhibits platelet activation as well as aggregation when mediated in whole or in part via this receptor. Cangrelor can be derived completely from synthetic materials, and is an analogue of adenosine triphosphate (ATP). ATP is a natural antagonist of the P2Y12 receptor sites and is found in humans.

The chemical structure for cangrelor is depicted below as Formula I.

Figure US20130303477A1-20131114-C00001

Cangrelor is clinically well tolerated and safe and has no drug-drug interaction with aspirin, heparin or nitroglycerin. Unlike orally dosed thienopyridines, cangrelor can be administered intravenously and binds directly to P2Y12 receptor sites of platelets. In each of the embodiments of the present invention, the term “cangrelor” encompasses the compound of Formula I as well as tautomeric, enantiomeric and diastereomeric forms thereof, and racemic mixtures thereof, other chemically active forms thereof, and pharmaceutically acceptable salts of these compounds, including a tetrasodium salt. These alternative forms and salts, processes for their production, and pharmaceutical compositions comprising them, are well known in the art and set forth, for example, in U.S. Pat. No. 5,721,219. Additional disclosure relevant to the production and use of cangrelor may be found in U.S. Pat. Nos. 5,955,447, 6,130,208 and 6,114,313, as well as in U.S. Appln. Publication Nos. 2006/0270607 and 2011/0112030.

Invasive procedures means any technique where entry to a body cavity is required or where the normal function of the body is in some way interrupted by a medical procedure and/or treatment that invades (enters) the body, usually by cutting or puncturing the skin and/or by inserting instruments into the body. Invasive procedures can include coronary artery bypass grafting (CABG), orthopedic surgeries, urological surgeries, percutaneous coronary intervention (PCI), other general invasive procedures, such as endarterectomy, renal dialysis, cardio-pulmonary bypass, endoscopic procedures or any medical, surgical, or dental procedure that could result in excessive bleeding or hemorrhage to the patient.

Perioperative means the period of a patient’s invasive procedure which can occur in hospitals, surgical centers or health care providers’ offices. Perioperative includes admission, anesthesia, surgery, to recovery.

Thrombosis is the formation of a blood clot (thrombus) inside a blood vessel obstructing the flow of blood through the circulatory system. When a blood vessel is injured, the body uses platelets and fibrin to form a blood clot to prevent blood loss. Some examples of the types of thrombosis include venous thrombosis which includes deep vein thrombosis, portal vein thrombosis, renal vein thrombosis, jugular vein thrombosis, Budd-Chiari syndrome, Paget-Schroetter disease, cerebral venous sinus thrombosis, cerebral venous sinus thrombosis and arterial thrombosis which includes stroke and myocardial infarction.

The compound cangrelor from the Medicines Company is represented by the structure

Figure imgf000013_0002

TETRASODIUM SALT
             OR
Cangrelor sodium, AR-C69931MX
Cangrelor Tetrasodium [USAN]
RN: 163706-36-3
Platelet P(2T) receptor antagonist.
5′-O-[[[Dichloro(phosphono)methyl](hydroxy)phosphoryloxy](hydroxy)phosphoryl]-N-[2-(methylsulfanyl)ethyl]-2-(3,3,3-trifluoropropylsulfanyl)adenosine tetrasodium salt
C17-H21-Cl2-F3-N5-O12-P3-S2.4-Na,
864.2899
The Medicines Co. (Proprietary), AstraZeneca Charnwood (Originator)
CARDIOVASCULAR DRUGS, Treatment of Disorders of the Coronary Arteries and Atherosclerosis, P2Y12 (P2T) Antagonists
2-Mercaptoadenosine (I) was S-alkylated with 1-chloro-3,3,3-trifluoropropane (II) in the presence of NaH to give trifluoropropyl sulfide (III). Subsequent acetylation of (III) with Ac2O at 80 C provided (IV), which was N-alkylated with methylthioethyl iodide (V) and NaH yielding (VI).
Further hydrolysis of the resulting (VI) with 0.1 M NaOH in refluxing MeOH furnished adenosine derivative (VII). The 5′-hydroxyl group of (VII) was then phosphorylated by reaction with phosphoryl chloride in cold triethyl phosphate followed by aqueous work-up.
The resulting 5′-monophosphate (VIII) was treated with carbonyl diimidazole and tri-n-butylamine to produce the phosphoryl imidazole intermediate (IX), which was finally condensed with dichloromethylenebis(phosphonic acid) (X).
The target compound was isolated as the tetrasodium salt upon treatment with NaI in methanol-acetone.
Alkylation of mercaptopurine (I) with 3-chloro-1,1,1-trifluoropropane (II) in the presence of NaH gave thioether (III).
After protection of the amino group of (III) as the acetamide (IV) by means of Ac2O and NaOAc, N-alkylation with 2-(methylthio)ethyl iodide (V) yielded (VI),
which was deacetylated by hydrolysis with NaOH in refluxing MeOH. Subsequent treatment with POCl3 produced the intermediate phosphoryl chloride (VIII).
Then, condensation of this acid chloride with dichloromethylene bisphosphonic acid (IX) in the presence of tributylamine in triethyl phosphate yielded the title compound, which was isolated as the tetrasodium salt.
Alternatively, hydrolysis of acid chloride (VIII) in the presence of ammonium bicarbonate gave phosphate salt (X), which was treated with carbonyldiimidazole, and the activated intermediate (XI) was then condensed with bisphosphonate (IX) to furnish the target compound.

…………

J. Med. Chem., 1999, 42 (2), pp 213–220

http://pubs.acs.org/doi/full/10.1021/jm981072s

10l (AR-C69931MX)

N6(2-Methylthioethyl)-2-(3,3,3-trifluoropropylthio)-5-adenylic Acid, Monoanhydride withDichloromethylenebis(phosphonic acid) (10l)Prepared as the triammonium salt in 4% yield from 3l:  1H NMR δ(D2O) 8.30 (1H, s, H8), 5.97 (1H, d, J = 5.5 Hz, H1‘), 4.65 (1H, m, H2‘), 4.47 (1H, m, H3‘), 4.28 (1H, m, H4‘), 4.17 (2H, m, H5‘a and H5‘b), 3.67 (br s, NHCH2), 3.21 (2H, t, J = 7.6 Hz, SCH2), 2.72 (2H, t, J = 6.6 Hz, SCH2CH2CF3), 2.58 (2H, m, NCH2CH2), 2.04 (3H, s, SCH3);31P NMR δ(D2O) 8.80 (d, 1P, J = 18.6 Hz, Pγ), 0.42 (dd, 1P, J1 = 18.9 Hz, J2 = 28.9 Hz, Pβ), −9.41 (d, 1P, J = 29.0 Hz, Pα). Anal. (C17H34Cl2F3N8O12P3S2·3H2O) H, N, S; C:  calcd, 23.16; found, 23.66.

References

  1.  Cangrelor Attenuates Coated-Platelet Formation
  2.  CHAMPION Trials With Cangrelor Stopped for Lack of Efficacy
  3. What Cangrelor Failure Means to Medicines
  4.  Effect of Platelet Inhibition with Cangrelor during PCI on Ischemic Events (2013) Bhatt, DL etal. New England Journal of Medicine March 10, 2013 DOI: 10.1056/NEJMoa1300815 (published initially online).
  5. The Duel between Dual Antiplatelet Therapies (2013) Lange, RA and Hillis, LD. New England Journal of Medicine March 10, 2013 DOI: 10.1056/NEJMe1302504
  6. 15th European Federation for Medicinal Chemistry International Symposium on Medicinal Chemistry (Sept 6 1998, Edinburgh)1998,:Abst P.281
  7.  Specific P2Y12 purinoceptor antagonist; inhibits ADP-induced platelet aggregation. Prepn: A. H. Ingall et al., WO 9418216 (1994 to Fisons); eidemUS 5721219 (1998 to Astra); and in vivo antithrombotic activity: idem et al., J. Med. Chem. 42, 213 (1999).
  8. In vivo antithrombotic effects in canine arterial thrombosis: J. Huang et al., J. Pharmacol. Exp. Ther. 295, 492 (2000).
  9. Mechanism of action study: A. Ishii-Watabe et al., Biochem. Pharmacol. 59, 1345 (2000).
  10. Clinical safety assessment and evaluation in acute coronary syndromes: R. F. Storey et al., Thromb. Haemostasis 85, 401 (2001); in angina pectoris and non-Q-wave myocardial infarction: F. Jacobsson et al., Clin. Ther. 24, 752 (2002).
  11. Clinical pharmacodynamics compared with clopidogrel: R. F. Storey et al., Platelets 13, 407 (2002).
  12. Review of clinical development: S. C. Chattaraj, Curr. Opin. Invest. Drugs2, 250-255 (2001).
  13. WO2013/103567 A2,
  14. Journal of Medicinal Chemistry, 1999 ,  vol. 42,  2  p. 213 – 220
Share
Feb 232014
 

RAMELTEON

ACN-S001714, ZINC00007031

  • HSDB 7787
  • Ramelteon
  • Rozerem
  • TAK-375
  • UNII-901AS54I69
Molecular Formula: C16H21NO2   Molecular Weight: 259.34344
CAS number 196597-26-9 
 (S)-N-[2-(1,6,7,8-tetrahydro-2H-indeno-[5,4-b]furan-8-yl)ethyl]propionamide

(5)-N-[2-(l,6,7,8-tetrahydro-2H-indeno-[5,4-ό]furan-8- yl)ethyl]propionamide

United States US 6034239 1999-07-22 expiry 2019-07-22

EP885210B1 , EP1792899A1 and J. Med Chem. 2002, 45, 4222-4239

NMR

[PPTData Supplement – Drug Metabolism and Disposition

  1. dmd.aspetjournals.org/content/suppl/…/Supplemental_Information.pptx

     May 17, 2010 – Ramelteon NMR Assignments. COSY: Black Arrows. HMBC: Red Arrows. Figure S-1b. 1H NMR Spectrum of Ramelteon. Figure S-1c.

Ramelteon is the first in a new class of sleep agents that selectively binds to the melatonin receptors in the suprachiasmatic nucleus (SCN). It is used for insomnia, particularly delayed sleep onset. Ramelteon has not been shown to produce dependence and has shown no potential for abuse.

Ramelteon, marketed as Rozerem by Takeda Pharmaceuticals North America, is the first in a new class of sleep agents that selectively binds to the MT1 and MT2 receptors in the suprachiasmatic nucleus (SCN), instead of binding to GABA A receptors, such as with drugs like zolpidem,eszopiclone, and zaleplon. Ramelteon is approved by the U.S. Food and Drug Administration (FDA) for long-term use.

Ramelteon does not show any appreciable binding to GABAA receptors, which are associated with anxiolyticmyorelaxant, and amnesic effects.

Rozerem (ramelteon), FDA Approved 07.04.05, can be used for insomnia, particularly delayed sleep onset. Ramelteon has not been shown to produce dependence and has shown no potential for abuse, and the withdrawal and rebound insomnia that is typical with GABA modulators is not present in ramelteon. Some clinicians also use ramelteon for the treatment of Delayed sleep phase syndrome.

Ramelteon

Mechanism of action

Ramelteon is a melatonin receptor agonist with both high affinity for melatonin MT1 and MT2 receptors and selectivity over the MT3 receptor. Ramelteon demonstrates full agonist activity in vitro in cells expressing human MT1 or MT2 receptors, and high selectivity for human MT1 and MT2receptors compared to the MT3 receptor.[1]

The activity of ramelteon at the MT1 and MT2 receptors is believed to contribute to its sleep-promoting properties, as these receptors, acted upon by endogenous melatonin, are thought to be involved in the maintenance of the circadian rhythm underlying the normal sleep-wake cycle. Ramelteon has no appreciable affinity for the GABA receptor complex or for receptors that bind neuropeptidescytokinesserotonindopaminenoradrenaline,acetylcholine, and opiates. Ramelteon also does not interfere with the activity of a number of selected enzymes in a standard panel.

The significance of ramelteon’s lack of affinity for the MT3 receptor is not clear, despite the manufacturer’s emphasis of this fact in commercial advertisements. The MT3 receptor appears almost exclusively in the gut and might not have any relationship to sleep or wakefulness.

The major metabolite of ramelteon, M-II, is active and has approximately one tenth and one fifth the binding affinity of the parent molecule for the human MT1 and MT2 receptors, respectively, and is 17 – 25-fold less potent than ramelteon in in vitro functional assays. Although the potency of M-II at MT1 and MT2 receptors is lower than the parent drug, M-II circulates at higher concentrations than the parent producing 20 – 100 fold greater mean systemic exposure when compared to ramelteon. M-II has weak affinity for the serotonin 5-HT2B receptor, but no appreciable affinity for other receptors or enzymes. Similar to ramelteon, M-II does not interfere with the activity of a number of endogenous enzymes.

All other known metabolites of ramelteon are inactive.

No published studies have indicated whether ramelteon, in humans, is more or less safe or effective than the hormone melatonin which it mimics; melatonin is much less expensive and is widely available over-the-counter in the US and Canada. The biological action of melatonin is similar to that of ramelteon. Ramelteon has been directly compared to melatonin in cats, and Ramelteon had a significant (3x) longer effect and had a more profound effect on the EEG of the sleeping cats.[2]

Introduction

ROZEREM (ramelteon) is an orally active hypnotic, chemically designated as (S)-N-[2- (l,6,7,8-tetrahydro-2H-indeno-[5,4-b]furan-8-yl)ethyl]propionamide, and contains one chiral center. The compound is produced as the (S)-enantiomer, with an empirical formula of C16H21N02, molecular weight of 259.34, and the following chemical structure (I):

 

Figure imgf000002_0001

(I) -Ramelteon

Ramelteon is used to help patients who have sleep-onset insomnia (difficulty falling asleep) to fall asleep more quickly. It is the first in a new class of sleep agents that selectively binds to the MT] and MT2 receptors in the suprachiasmatic nucleus (SCN), in a class of medications called melatonin receptor agonists with both high affinity for melatonin MT! and MT2 receptors and selectivity over the MT3 receptor. It works similarly to melatonin, a natural substance in the brain that is needed for sleep.

Ramelteon was first disclosed in European patent EP 885210, which also disclosed a process for synthesizing ramelteon, as shown in scheme 1 : Scheme 1

 

Figure imgf000003_0001

Ramelteon

The processes of the prior art suffer from many disadvantages, some of which result from the fact that they involve several steps.

For instance, in US patent US 6034239, which is related to EP 885210, there is disclosed a process for preparing an intermediate compound of Formula (IV), which involves conversion of diethylcyano methyl phosphonate in the presence of 60% sodium hydride. Disadvantages of this particular reaction include the need for the highly flammable and corrosive base sodium hydride, the use of toxic triethyl phosphate for the formation of diethylcyano methyl phosphonate (which also has a high boiling point), and low yield of 60%. Such disadvantages mean that the disclosed process is difficult to implement industrially or economically. A further problem associated with prior art preparation techniques is the formation of dimeric impurities at the nitrile reduction stage (i.e. where the intermediate of Formula (IV) is reduced). For instance, US 6034239 discloses reduction of (l,2,6,7-Tetrahydro-8H-indeno-[5,4- b]furan-8-ylidene)-acetonitrile of formula (IV) by means of H2 over Raney nickel in in a solvent medium of ethanol NH3 to provide compound of formula (IIA). The reaction is carried out by applying 5 kg of hydrogen pressure, which results in the formation of the byproduct and impurity Dimer A, which in turn affects the yield and purity of the product of formula (IIA).

 

Figure imgf000004_0001

Dimer A

Similarly, (l,2,6,7-Tetrahydro-8H-indeno-[5,4-b]furan-8-ylidene)-acetonitrile of formula (IV) may be reduced by means of H2 over Raney cobalt in a solvent medium of ethanol/ NH3 to afford compound of formula (IIB). The reaction, which is carried out by applying hydrogen pressure, is not selective, and results in the formation of the by-product and impurity Dimer B, which in turn affects the yield and purity of product of formula (IIB).

 

Figure imgf000004_0002

Dimer B

Repeated purifications are required to remove impurities such as Dimer A and B to obtain ramelteon having the desired purity, which results in the poor yield of ramelteon.

Several other approaches are also described in the literature to make ramelteon and related compounds in WO2006030739, WO208062468, WO2008106179, US 2010152468, WO2009106966 and WO2010/055481. However, all processes of the prior art for the preparation of ramelteon are cumbersome; the processes employ a plurality of reagents and involve multiple steps, which make the overall processes uneconomical. Therefore there is a need for a more economical, efficient and industrially suitable method of making ramelteon, whereby address the problems associated with prior art, some of which are discussed above.

Ramelteon is the active ingredient in trademarked ROZEREM®, and is approved by the United States Food and Drug Administration for the treatment of insomnia characterized by difficulty with sleep onset.

Different processes for preparing (S)-N-[2-(l,6,7,8-tetrahydro-2H-indeno-[5,4- b]furan-8-yl)ethyl]propionamide, i.e. ramelteon, are disclosed in US 6034239, JP 11080106, JP 11140073 and WO 2006/030739. U.S. Patent No. 6034239 describes the following processes for the preparation of ramelteon:

Figure imgf000004_0001

Japan Patent Publication No. 11080106 reports the following process for the preparation of ramelteon:

Ru(OCOCH3)[(R)-BI NAP] IOOatm H2/50 temp

Figure imgf000005_0001
Figure imgf000005_0002

BF3 DEE Complex

 

Figure imgf000005_0003

Japan Patent Publication no. 11140073 reports the following process for the preparation of an intermediate of ramelteon:

 

Figure imgf000005_0004

PCT Publication No. WO/2006/030739 reports the following process for the preparation of ramelteon:

POCI3/DMF (EtO)2P(O)CH2CO2Et Toluene NaH/Toluene

Figure imgf000006_0001
Figure imgf000006_0002
Figure imgf000006_0003

 

Figure imgf000006_0004

 

Figure imgf000006_0005
Figure imgf000006_0006

 

Figure imgf000006_0007

Purification in Ethanol water 95 7%

United States Patent No. 6,034,239 discloses the formation of chiral intermediates (S)-(- )-N-[2-(l,6,7,8,-tetrahydro-2H-indeno[5,4-b]furan-8-yl)ethylamine (sometimes referred to as compound S-2 or intermediate compound S-2) by the catalytic asymmetric hydrogenation of 2- (l,2,6,7,-tetrahydro-8H-indeno[5,4-b]furan-8-ylidene)ethylamine (compound 3 in the reaction scheme shown below) in the presence of a catalytic amount of BINAP-ruthenium complex in approximately 89% e.e. (enantiomeric excess). Following the catalytic reaction, the product is purified by preparing acid salts and acylated with propionyl chloride (compound 4 in the reaction scheme shown below) to obtain ramelteon (compound 1 in the reaction scheme shown below) in its pure (S) isomer form.

 

Figure imgf000003_0001

|S)*2

An alternate process for preparing ramelteon is disclosed in the Journal of Medicinal Chemistry, Vol. 45, pp. 4222-4239 (2002), wherein the exo double bond of intermediates (A) shown below was asymmetrically reduced using (S)-2, 2′-bis-(diphenylphosphino)-l, 1 ‘- binaphthyl (binap)-Ru complex as the catalyst to obtain the enantiomerically pure compound (B). Compound (B) is subsequently converted to ramelteon (1) through the intermediate steps of Claisen condensation, ozonolysis and cyclization.

 

Figure imgf000003_0002

m Both of the above processes uses expensive catalyst and give poor enantioselectivity. Additionally, these processes are expensive due to the need to perform multiple purifications steps in order to achieve an enantioselectivity of at least about 99% or greater of the desired isomer.

PCT Patent Publication No. WO 2008/062468 A2 discloses the following process for the preparation of ramelteon:

 

Figure imgf000004_0001

RAMELTEON

WO 2008/062468 teaches that separation of the enantiomers of intermediate (2) may be accomplished by: i) optical resolution of the racemic amine intermediate (2) by preparing acid salts with chirally pure acids; or ii) chromatographic techniques using chiral and/or achiral stationary phases for batch process, super critical or sub critical chromatography and/or continuous process chromatography. Although WO 2008/062468 mentions the possible use of optical resolution with chirally pure acids, there is no further teaching, discussion or disclosure of this method. WO 2008/062468 does, however, provide detailed descriptions of chromatographic methods for separating the isomers of intermediate compound (2). The disclosed chromatographic process suffers the following disadvantages:

• Preparative chromatography is time consuming & expensive;

• Highly sophisticated instrumentation required; • Not commercially feasible.

PCT Patent Publication No. WO 2008/106179 discloses a process for the preparation of ramelteon that involves the following reaction steps:

 

Figure imgf000005_0001

wherein X= O-alkyl or NH2 and chiral reduction of the compound of formula IV in the presence of Ru-BINAP complex under hydrogen atmosphere in an organic solvent.

 

Figure imgf000005_0002

IV                                                                            V

The process disclosed in WO 2008/106179 is similar to the process disclosed in United States Patent No. 6,034,239 and the Journal of Medicinal Chemistry, Vol. 45 in that a Ru-BINAP complex is employed.

Resolution of racemic mixtures via reaction with optically active acids and the subsequent crystallization of the resulting salts is preferably employed when the chiral carbon of the racemic compound is an alpha carbon {i.e., one carbon removed) to the functional group forming the acid addition salt. As the distance between the chiral carbon of the racemic compound to the functional group of the racemic compound increases to beta (i.e., two carbon removed) & gamma (i.e., three carbon removed), the resolution of the diastereomeric salt becomes more difficult and not very useful.

Ramelteon has a chiral center at the gamma carbon, which makes the separation of the isomer with an optically active acid quite a daunting task. Similarly, N-[2-(l, 6, 7, 8,- tetrahydro-2H-indeno [5, 4-b]furan-8-yl)]ethylamine (compound T), an intermediate useful in the production of ramelteon has a chiral center at the gamma carbon which would lead a skilled artisan to believe that optical resolution with an optically active acid could prove difficult.

Synthesis

Ramelteon synth.png

Chilman-Blair, K.; Castañer, J.; Silvestre, J.S.; Bayés, M. (2003). “TAK-375”. Drugs of the Future 28 (10): 950. doi:10.1358/dof.2003.028.10.763214.

………………..

SYNTHESIS

Scheme 1 :

 

Figure imgf000007_0001

XIV (S)-XII

Figure imgf000007_0002

……………………………………..

SYNTHESIS

WO2012035303A2

Scheme 2

 

Figure imgf000021_0001

0-30°C

Metal salt Propionyl halide/

Propionc anhydride

 

Figure imgf000021_0002

Ramelteon (I)

Synthesis of ramelteon

Preparation 1

N-[2-(l,6,7,8-Tetrahydro-2H-indeno[5,4-b]furan-8-yl)-ethyl]-propionamide (2.0 gm) was dissolved in 50.0 ml (n-Hexane:IPA:DEA) (as used herein, “IPA” stands for isopropyl alcohol, and “DEA” stands for diethylamine)

and optically resolved by high performance column chromatography on CHTRAL PACK IA-3 using Mobile phase : n-Hexane:IPA:DEA Flow rate: 1.0ml/min UV:285 nm; at a column temperature of 25°C;sample concentartion: lmg/ml and, eluted with mobile phase. Both the enantiomers were collected separately and after evaporation of solvent under vacuum, enantiomerically pure ramelteon (I) was obtained. Preparation 2- using Supercritical Fluid Chromatography (SFC)

N-[2-(l56,7,8-Tetrahydro-2H-indeno[5,4-b]furan-8-yl)-ethyl]-propionamide (2.0 gm) was dissolved in 50.0 ml (n-Hexane:Ethanol:DEA) and optically resolved by Supercritical Fluid Chromatography (SFC) on CHIRAL PACK AD-H using a mobile phase : C02/(Methanol/ Diethylamine[DEA]) and eluted with mobile phase. Both the enantiomers were collected separately and after evaporation of solvent under vacuum, enantiomerically pure S- ramelteon of Formula (I) and R-ramelteon were obtained with isomeric purity>99%.

………………………………

SYNTHESIS

WO2010055481A1

synthesis of ramelteon that comprises the step of separating N-[2-(l, 6, 7, 8-tetrahydro-2H-indeno [5, 4-b] furan-8- yl)]ethylamine (compound 2) into its isomers using an optically active acid to achieve high enantioselectivity of the desired isomer. This embodiment may further include the step of acylating the substantially pure enantiomer, (S)-N-[2-(l, 6, 7, 8-tetrahydro-2H-indeno [5, 4-b] furan-8-yl)]ethylamine (compound (S)-2) using a suitable acylating agent, such as propionyl chloride) to provide (S)-7V-[2-(l,6,7,8-tetrahydro-2H-indeno[5,4-b]furan-8-yl]ethyl]propionamide (ramelteon or compound 1) substantially free of the (R)-isomer.

One embodiment of the present invention for the preparation of ramelteon is shown below in Scheme 1.

 

Figure imgf000007_0001

Example 1

Preparation of (S)-N-2-(l, 6, 7, 8-tetrahydro-2H-indeno [5, 4-b] furan-8-yl) ethylamine (Compound (S)-2)

A solution of N-[2-(l, 6, 7, 8-tetrahydro-2H-indeno [5, 4-b] furan-8-yl)ethylamine (45 g; 0.22 mol) in methanol (225 ml) is added to a solution of S-(+)-2-(4-isobutylphenyl)propionic acid (41 g; 0.20 mol) in methanol (205 ml) at 25-300C. The reaction mixture is concentrated to dryness under reduced pressure. The crude salt precipitated is recrystallized in methanol to give a diastereomeric salt of (S)-N-2-(l, 6, 7, 8-tetrahydro-2H-indeno [5, 4-b] furan-8-yl) ethylamine with (S)-(+)-2-(4-isobutylphenyl) propionic acid having a chiral purity of greater than 90% enantioselectivity. The product obtained is recrystallized from methanol to give the pure salt having chiral purity of 99% or greater enantioselectivity.

The purified salt is suspended in water and the pH of the suspension is adjusted to 11-12 using aqueous sodium hydroxide. The reaction mixture is extracted with dichloromethane, washed with water and evaporated to give the pure (S)-N-[2-(l, 6, 7, 8-tetrahydro-2H-indeno [5, 4-b] furan-8-yl)]ethylamine (compound (S)-2), substantially free from its (R) isomer.

Example 2

Preparation of (S)-N-[2-(l, 6, 7, 8-tetrahydro-2H-indeno [5, 4-b] furan-8-yl)ethyl] propionamide (ramelteon)

Triethyl amine (15.15 g, 0.15 mol) and propionyl chloride (13.66 g, 0.15 mol) were added to a solution of S-[2-(l, 6, 7, 8-tetrahydro-2H-indeno [5,4-b]furan-8-yl)]ethylamine (25 g, 0.12 mol) (compound (S)-2) (prepared in Example 1) in dichloromethane and stirred at room temperature for 2 hours. 75 mL water was added to the reaction mixture, and the layers were separated. The dichloromethane layer was concentrated under reduced pressure and purified from a mixture of acetone and hexane to give (S)-N-[2-(l, 6, 7, 8-tetrahydro-2H-indeno [5, 4-b] furan- 8-yl) ethyl] propionamide (compound 1) having a chiral purity of 99% or greater enantioselectivity.

…………
INTERMEDIATES

The intermediate compound of formula Vl, 6,7-dihydro-1 H-indeno[5,4-b]furan-8(2H)-one, can then be subjected to further synthesis steps to yield ramelteon by synthesis route known to or readily devisable by a person skilled in the art, suitably involving the introduction of the side chain having chirality and amide function. The documents mentioned infra are incorporated herein by way of reference. For example, the following synthesis route may be applied:

Figure imgf000016_0001

1 ) NaOH

Vl 2) H2, Ru-BI NAP

3) HCI

4) H2, Pd/C

 

Figure imgf000016_0002

Experimental Procedures

Example 1 :

Preparation of 4-(2-chloroethyl)-2,3-dihydrobenzofuran (II)

 

Figure imgf000016_0003

intermediate FTIR spectra of MeCN (140 ml) was recorded as reference. MeCN was cooled to -200C, oxalyl chloride (16.5 ml) was added at once and waited until temperature re-stabilized at – 200C. DMF (16.6 ml) was then added drop-wise (temperature between -18°C and -22°C, 0.5 ml/min). Reaction was stirred until no oxalyl chloride was visible and DMF level was stable by FTIR. Vilsmeier reagent is thereby formed in situ according to the following reaction:

Figure imgf000017_0001

Product I was then added portion wise (temperature between -18°C and -210C, about 30 min). Formation of intermediate was immediately observed by FTIR. Reaction was stirred for one hour. Et3N was then added drop-wise (temperature between -18°C and -22°C, 50 ml/h). At the end of addition, reaction was stirred 15 min at -200C and temperature was slowly raised to 500C (within about 15 min). Disappearance of intermediate and formation of DMF and product Il was monitored by FTIR. When reaction looked completed by FTIR (about 2h at 50°C), the reaction was cooled down to 200C and quenched with water (45 ml). Solution was transferred to a round bottom flask and MeCN was removed under reduced pressure. Solution was then diluted with MTBE (100 ml) and water (50 ml). Phases were separated and aqueous phase was re-extracted twice with MTBE (50 ml). Combined organic phases were washed twice with 10% H34/10% NaCI solution and stored at 4°C until next step.

List of FTIR bands used to follow the reaction (using 2nd derivative and solvent subtraction): Oxalyl chloride (reactant): Height to two point baseline, peak from 1800 cm“1 to 1770 cm“1, baseline 1800 cm“1 to 1770 cm“1.

Intermediate: Height to single point baseline, peak from 1722 cm“1 to 1712 cm“1, baseline

1722 cm“1.

Compound Il (product): Area to two point baseline, peak from 993 cm“1 to 981 cm“1, baseline 993 cm“1 to 981 cm“1.

DMF: Height to single point baseline, peak from 1694 cm“1 to 1680 cm“1, baseline 1694 cm“1.

Example 2:

Preparation of 4-vinyl-2,3-dihydrobenzofuran (III)

 

Figure imgf000017_0002

M FTIR spectra of MTBE was recorded prior to the reaction as reference. To the solution of 4- (2-chloroethyl)-2,3-dihydrobenzofuran (II) in MTBE (150 ml) obtained at the previous step, was added, water (38 ml), Kl (1.37 g), Bu4NOH 40% (19 ml) and NaOH 50% solution (66 ml). Reaction was vigorously stirred and heated at 500C until reaction looked completed by FTIR (4 to 5 h). Warm reaction mixture was then transferred into an extraction funnel to give three phases. Water phase (bottom) was removed and did not contain product. Medium phase (colored black) was diluted with water (120 ml) and was extracted three times with MTBE. Combined organic phases were washed twice with water, once with 0.5M NaHSO3/10% NaCI solution and once with 1 N NaOH/10% NaCI solution. MTBE solution was dried using MgSO4, filtered, concentrated and used immediately for next step.List of FTIR bands used to follow the reaction (using 2nd derivative and solvent subtraction) Compound Il (reactant): Area to zero, peak from 1440 cm“1 to 1437 cm“1 Compound III (product): Area to zero, peak from 1417 cm“1 to 1412 cm“1. Compound III (product): Area to zero, peak from 1565 cm“1 to 1562 cm“1.

Example 3:

Preparation of 1-(2,3-dihydrobenzofuran-4-yl)ethanone (V)

 

Figure imgf000018_0001

4-vinyl-2,3-dihydrobenzofuran (I I I ) (2.4 g) was dissolved in toluene (2 ml) and were successively added (ITC) (51 mg), PdCI2(30 mg) and H2O2 30% (2 ml). Reaction was vigorously stirred at 55°C until reaction looked completed by FTIR. (for around 24 h). Reaction was cooled down to room temperature, diluted with EtOAc (50 ml) and water (50 ml). Phases were separated and organic phase was washed with 0.5M NaHSO3/10% NaCI solution and twice with 1 M NaHCO3, dried over MgSO4 and concentrated. Purification by flash chromatography gave 1-(2,3-dihydrobenzofuran-4-yl)ethanone (V). 1H NMR δ (CDCI3) 7.35 (dd, 1 H, J = 0.8 Hz, J = 7.8 Hz), 7.19 (t, 1 H, J = 7.9 Hz), 6.95 (d, 1 H, J = 8.0 Hz), 4.57 (t, 2H, J = 8.8 Hz), 3.52 (t, 2H, J = 8.8 Hz), 2.57 (s, 3H). 13C NMR δ (CDCI3) 198.8, 161.0, 133.8, 128.2, 127.9, 121.4, 1 13.4, 71.6, 31.0, 27.6.

List of FTIR bands used to follow the reaction (using 2nd derivative and solvent subtraction) Compound III (reactant): Area to single point baseline, peak from 925 cm“1 to 915 cm“1, baseline 915 cm“1.

Compound V (product): Area to zero, peak from 1730 cm“1 to 1724 cm“1.

Example 4:

Preparation of 6,7-dihydro-1 H-indeno[5,4-b]furan-8(2H)-one (Vl)

 

Figure imgf000019_0001

V Vl1 -(2,3-dihydrobenzofuran-4-yl)ethanone (V) (1 g, 6.2 mmol) was dissolved in dioxane (9 ml). TADCA (dicyclohexylammonium 2,2,2-trifluoroacetate) (1 .82 g, 1 eq) and paraformaldehyde (0.61 1 g, 1.1 eq) were added. The reaction was heated at 1000C for 2 h. A second portion of TADCA (0.91 g, 0.5 eq) and paraformaldehyde (0.333 g, 0.6 eq) were added and the reaction was heated at 1000C for 2 h. Reaction was partitioned between water (20 ml) and pentane (30 ml). Aqueous phase was re-extracted 4 times with pentane (10 ml). Combined pentane phases were washed with water and brine, dried over MgSO4. Solution was diluted to 100 ml with pentane. This solution was added dropwise to a pre-heated solution of sulfuric acid at 67°C (10 ml) under nitrogen stream. At the end of addition, the reaction was stirred for 30 min. Reaction was cooled down to room temperature and poured on iced water (50 ml). Solution was extracted 5 times with MTBE. Combined organic phases were washed with water, NaHCO3 1 M and brine, dried over MgSO4 and concentrated. Purification by flash chromatography furnished pure 6,7-dihydro-1 H-indeno[5,4-b]furan-8(2H)-one (Vl). 1H NMR δ (CDCI3) 7.21 (dd, 1 H, J = 0.9 Hz, J = 9.0 Hz), 7.02 (d, 1 H, J = 8.2 Hz), 4.66 (t, 2H, J = 8.9 Hz), 3.48 (t, 2H, J = 8.9 Hz), 3.08 (dd, 2H, J = 4.9 Hz, J = 6.0 Hz), 2.69 (m, 2H). 13C NMR δ (CDCI3) 207.5, 160.2, 147.1 , 133.6, 125.6, 123.9, 1 15.6, 72.3, 37.1 , 28.4, 25.4.

…………………………………

SYNTHESIS

Improve the synthesis and flow properties of an insomnia drug. 

Ramelteon (1), marketed as Rozerem by Takeda Pharmaceuticals, is used to treat insomnia. V. K. Kansal and co-inventors describe several processes that are used to prepare it, all of which require many steps. The inventors offer no comments about the relative merits of the processes, but they state that a new industrial-scale process is needed. Their main claims are to intermediate acid 2 as a racemic mixture and individual enantiomers; one enantiomer is converted to 1 by the route shown in Figure 1.

The inventors use diastereomeric crystallization to resolve the racemic mixture by forming its (S)-1-phenylethylamine salt. The salt of the (R)-isomer of 2 is recovered first; then the salt of (S)-2 is isolated from the solution and acidified to give the free acid, which is purified by using (R)-1-phenylethylamine. Both enantiomers are isolated with >99.0% purity and >99.0% ee.

The (S)-acid is converted to acid chloride 3 and then to amide 4 by reactions with SOCl2and NH3 gas, respectively. The chloride is not isolated; the amide is recovered in 85–90% yield with 95–98% purity. When aq NH4OH is used instead of NH3 gas, the purity of 4 is slightly lower (93–96%). An alternative method for preparing 4 is to treat 2 with Et3N and ClCO2Et, followed by NH3. This method produces 4 in yields of 80–95% and 97–99% purity.

Amide 4 is reduced to amine 5 with NaBH4 and BF3·Et2O. The amine is purified by forming its chloride or oxalate salt in yields as high as 85% and 96–98% purity. The salts are used to prepare 1 by treating them with EtCOCl in the presence of base: NaOH for the chloride salt and Na2CO3 for the oxalate. In both cases, the yield of 1 is >92%, and the purity is as high as 99.9% after recrystallization from EtOH.

The inventors also recrystallized 1 from toluene to produce what they describe as a “nonelectrostatic” crystalline form, designated as form A. They describe the measurement of the electrostatic charge of the crystals in one of the patent’s examples. The measurements show that the average charge density of form A is ≈15 times lower than crystals obtained from EtOAc. Low electrostatic charge improves the flow characteristics of the solid, which is important in preparing drug formulations.

The inventors report the details of preparing rac-2 by a multistep procedure shown in Figure 2.

In most of the reaction steps, the product is isolated in crude form; the inventors do not indicate whether the product is purified before it is used in the next stage. The synthesis of rac-2 begins with the conversion of benzofuran (6) to aldehyde7 by treatment with POCl3 followed by hydrolysis. The crude product is isolated as a liquid in 85–90% yield and 90–92% purity.

In the next step, 7 is condensed with malonic acid (8) in the presence of piperidine and HOAc; acid 9 is isolated in 92–95% yield and 95% purity. Catalytic hydrogenation of 9produces 10 in 95% yield and 94–96% purity. The hydrogenation also can be carried out in the presence of NaOH and HCO2NH4; the yield and purity of 10 are the same, but the reaction takes 6 h instead of 2 h. [The patent does not state why NaOH and HCO2NH4 would be used.—Ed.] Acid 10 is brominated to produce acid 11, isolated in 50–60% yield and 92–95% purity.

The next stage begins with treating 11 with SOCl2 to activate the carboxyl group by forming acid chloride 12. The chloride is not isolated but cyclized under Friedel–Crafts conditions to give tricyclic compound 13, isolated in yields of 85–92% and 90-95% purity. This reaction also produces two impurities, 14 and 15, but the amounts are not reported. Removing the impurities gives 13 in good yield, but the inventors do not describe how this is done. They do report that the impurities can be isolated, and 1H and 13C NMR data are provided for both.

In the next step, the bromine atoms in 13 are replaced by hydrogen to give 16 in 85–90% yield and 96–97%purity. This reaction produces two impurities, 17 and 18; again, the amounts are not reported, but 1H and 13C NMR data are. After MeOH reflux in the presence active carbon, 16 is isolated in 80–85% yield with 99.3–99.8% purity. It is then converted to ester 20 by treating it with a solution of phosphonate 19 that contains suspended NaH. Crude product 20 is isolated in 80–85% yield and 92–95% purity as a mixture of (E)- and (Z)-isomers. The isomer mixture is hydrogenated, and base hydrolysis gives rac-2 in 90–95% isolated yield and 95–98% purity.

The inventors claim that the overall process is suitable for producing ramelteon on an industrial scale in a crystalline form that has improved flow characteristics. (Teva Pharmaceutical Industries [Petah Tiqva, Israel]. US Patent 8,084,630, Dec. 27, 2011;

RAMELTEON

 

…………………….

SYNTHESIS

http://sat.ecnu.edu.cn/Uploadnews/20120213113859628.pdf

CHINESE CHEMICAL LETTERS 22, 2011, 264 SEE SYN OF KEY INTERMEDIATE

1:(S)-N-(2-(6-Methoxy-2,3-dihydro-1H-inden-1-yl)ethyl)propionamide 1======KEY INTERMEDIATE

[a]D20 10.0 (c, 0.20, EtOH); mp 76–77 8C;

1H NMR (500 MHz, CDCl3): d1.15 (t, 3H, J = 7.5 Hz), 1.60 (m, 1H), 1.70 (m, 1H), 2.02 (m, 1H), 2.19 (q, 2H, J = 7.5 Hz), 2.32 (m, 1H), 2.76 (m, 1H), 2.85 (m, 1H), 3.11 (m,1H), 3.41 (m, 2H), 3.79 (s, 3H), 5.48 (s, 1H), 6.71 (dd, 1H, J = 2.0 Hz, 8.5 Hz), 6.75 (s, 1H), 7.11 (d, 1H, J = 8.0 Hz).

13C NMR (100 MHz,DMSO–d6): d173.7, 158.7, 148.1, 135.8, 124.9, 112.3, 109.2, 55.5, 42.7, 37.9, 34.8, 32.5, 30.6, 29.8, 9.9. EI-MS: 247 ([M]+); HR-MS 247.1572([M]+
, C15H21NO2; Calcd. 247.1571). The enantiomeric excess of (S)-1 was determined by HPLC as >99.9% [column, CHIRALPAK AS-H
(4.6 mm  250 mm), room temperature; eluent, hexane-2-propanol-trifluoroacetic acid (90:10:0.1); flow rate, 1.0 mL/min; detect, 290 nm; tRof (S)-1, 30.7 min; tR of (R)-1 (enantiomer of (S)-1), 37.1 min].

…………………….

NMR

[PPTData Supplement – Drug Metabolism and Disposition

  1. dmd.aspetjournals.org/content/suppl/…/Supplemental_Information.pptx

     May 17, 2010 – Ramelteon NMR Assignments. COSY: Black Arrows. HMBC: Red Arrows. Figure S-1b. 1H NMR Spectrum of Ramelteon. Figure S-1c.

 

References

  1.  Owen RT (April 2006). “Ramelteon: profile of a new sleep-promoting medication”. Drugs Today 42 (4): 255–63. doi:10.1358/dot.2006.42.4.970842PMID 16703122.
  2.  Miyamoto M, Nishikawa H, Doken Y, Hirai K, Uchikawa O, Ohkawa S (November 2004). “The sleep-promoting action of ramelteon (TAK-375) in freely moving cats”. Sleep 27 (7): 1319–25.PMID 15586784.
  3.  Zammit G, Erman M, Wang-Weigand S, Sainati S, Zhang J, Roth T (August 2007). “Evaluation of the Efficacy and Safety of Ramelteon in Subjects with Chronic Insomnia”J Clin Sleep Med 3 (5): 495–504. PMC 1978328PMID 17803013.
  4. Daniel F. Kipke, MD |title=Evidence That New Hypnotics Cause Cancer |journal=University of California |date=March 2008 |url=http://escholarship.org/uc/item/12r2f32g#page-2
  5. http://db.wdc-jp.com/cgi-bin/psj/data/cpb/pdf/201108/c08_1062.pdf
  6. http://sat.ecnu.edu.cn/Uploadnews/20120213113859628.pdf
  7. https://docs.google.com/viewer?url=http%3A%2F%2Fdmd.aspetjournals.org%2Fcontent%2Fsuppl%2F2010%2F05%2F17%2Fdmd.110.034009.DC1%2FSupplemental_Information.pptx
  8. Full-Text PDF – MDPI.com

 

 

WO2008106179A1 Feb 26, 2008 Sep 4, 2008 Teva Pharma Intermediates and processes for the synthesis of ramelteon
WO2008151170A2 Jun 2, 2008 Dec 11, 2008 Teva Pharma Process for the synthesis of ramelteon and its intermediates
EP0885210B1 Mar 5, 1997 Jun 12, 2002 Takeda Chemical Industries, Ltd. Tricylic compounds having binding affinity for melatonin receptors, their production and use
EP1792899A1 Sep 12, 2005 Jun 6, 2007 Takeda Pharmaceutical Company Limited Process for production of optically active amine derivatives
US6034239 * Mar 6, 1997 Mar 7, 2000 Takeda Chemical Industries, Ltd. Tricyclic compounds, their production and use
US20010039286 * Feb 13, 2001 Nov 8, 2001 Kevin Dinnell 2-aryl indole derivatives and their use as therapeutic agents
WO2006030739A1 Sep 12, 2005 Mar 23, 2006 Takeda Pharmaceutical Process for production of optically active amine derivatives
WO2008062468A2 Oct 15, 2007 May 29, 2008 Cadila Healthcare Ltd Process for the preparation of optically pure indeno [5,4-b] furan derivatives
WO2008106179A1 Feb 26, 2008 Sep 4, 2008 Teva Pharma Intermediates and processes for the synthesis of ramelteon
WO2009106966A1 Feb 27, 2009 Sep 3, 2009 Medichem, S.A. Process for preparing ramelteon.
WO2010055481A1 Nov 12, 2009 May 20, 2010 Watson Pharma Private Limited Process for the preparation of ramelteon
WO2010092107A1 * Feb 11, 2010 Aug 19, 2010 Lek Pharmaceuticals D.D. Synthesis of (s)-n-[2-(1,6,7,8-tetrahydro-2h-indeno-[5,4-b]furan-8-yl)ethyl]propionamide
WO2010103553A1 * Mar 10, 2009 Sep 16, 2010 Industriale Chimica S.R.L. Process for the preparation of ramelteon
EP0885210A1 Mar 5, 1997 Dec 23, 1998 Takeda Chemical Industries, Ltd. Tricyclic compounds, their production and use
US6034239 Mar 6, 1997 Mar 7, 2000 Takeda Chemical Industries, Ltd. Tricyclic compounds, their production and use
US20100152468 Oct 16, 2009 Jun 17, 2010 Teva Pharmaceutical Industries Ltd. Process for the synthesis of ramelteon and its intermediates
WO2008062468A2 * Oct 15, 2007 May 29, 2008 Cadila Healthcare Ltd Process for the preparation of optically pure indeno [5,4-b] furan derivatives
US5321154 * Jul 31, 1992 Jun 14, 1994 Nagase & Company, Ltd. Optical resolution of (.+-.)-2-(4-isobutylphenyl)-propionic acid
US6218429 * May 10, 1999 Apr 17, 2001 Takeda Chemical Industries, Ltd. Tricyclic compounds, their production and use
US6348485 * Jun 8, 1999 Feb 19, 2002 Takeda Chemical Industries, Ltd. Method for treating or preventing sleep disorders
US20080214559 * Jan 7, 2008 Sep 4, 2008 Solvay Pharmaceuticals B.V. Compounds with a combination of cannabinoid cb1 antagonism and serotonin reuptake inhibition
US20080242877 * Feb 26, 2008 Oct 2, 2008 Vinod Kumar Kansal Intermediates and processes for the synthesis of Ramelteon
WO2012035303A2 Sep 16, 2011 Mar 22, 2012 Cipla Limited Et Al A novel process for synthesis of ramelteon, and key intermediates for the synthesis of ramelteon

Dedicated to lionel my son

My daughter Aishal

THEY KEEP ME GOING

 

ANTHONY MELVIN CRASTO

THANKS AND REGARD’S
DR ANTHONY MELVIN CRASTO Ph.D

amcrasto@gmail.com

MOBILE-+91 9323115463
GLENMARK SCIENTIST ,  INDIA
web link
http://anthonycrasto.jimdo.com/

Congratulations! Your presentation titled “Anthony Crasto Glenmark scientist, helping millions with websites” has just crossed MILLION views.
アンソニー     安东尼   Энтони    안토니     أنتوني
join my process development group on google
you can post articles and will be administered by me on the google group which is very popular across the world
Share

CAPREOMYCIN

 GENERIC, Uncategorized  Comments Off on CAPREOMYCIN
Feb 082014
 
Structure 

Capreomycin is a peptide antibiotic, commonly grouped with the aminoglycosides, which is given in combination with other antibiotics for MDR-tuberculosis. Adverse effects include nephrotoxicity and 8th cranial auditory vestibular nerve nerve toxicity.

The drug should not be given with streptomycin or other drugs that may damage the auditory vestibular nerve. Patients on this drug will often require audiology tests.

It is a cyclic peptide. Capreomycin is administered intramuscularly and shows bacteriostatic activity.REF 20

Capreomycin is frequently used to treat Mycobacterium tuberculosis infections. Mycobacterium tuberculosis growth has been found to be inhibited at a concentration of 2.5 μg/mL. REF21

This is the basic structure of capreomycin. The table below identifies the various naturally occuring analogues12, 14.

R1
R2
Capreomycin IA
OH
b-Lys
Capreomycin IB

H

b-Lys
Capreomycin IIA

OH

NH2
Capreomycin IIB

H

NH2

Introduction

Capreomycin is a metabolite of Streptomyces capreolus, it is an antimycobacterial agent – and a potent tuberlostatic antibiotic. Capreomycin is effective against a number of Gram-positive and Gram-negative organisms, but is primarily active against mycobacteria. It has been used in the treatment of certain resistant strains of Mycobacterium tuberculosis. The drug was first described in 1960 be Herr, and was subsequently found to contain two components (I and II) and later to be comprised of four (IA, IB, IIA, IIB) as shown on thestructure page.

Tuberculosis

Tuberculosis is a disease of the respiratory system, and is airbourne. The bacilli implant themselves in areas such as the lungs, renal cortex and reticuloendothelial system where there is a high partial pressure of oxygen. This is the Primary infection and does not normally affect the person whilst their immune system is intact as the bacteria lie dormant. When the immune system is depressed, the secondary reactivation occurs, and effects of the disease are seen.

This infectious disease has been known since about 1000B.C., and it stills remains the ‘leading cause of death from a single infectious disease agent’7. It is estimated that around eight million people contract TB every year, of which 95% are in developing countries. Deaths from the disease is estimated at 3 million people per year by the World Health Organisation. The occurance of the disease is related directly to the economic state of the country. This is because the spread of the disease is greatly assisted by poor public and personal hygiene and by overcrowding. New drugs were develoed about forty years ago allowing tuberculosis to be regarded as a curable disease. This is no longer the case, as many multidrug-resistant strains of the disease have emerged. This is where capreomycin has it uses.

There are three groups of drugs used to treat TB, which vary in their effectiveness and potential side effects.

First line drugs include: isoniazid, rifampicin and pyrazinamide. These are most effective and have the fewest potential side effects.

Second line drugs include: ethambutol, streptomycin and p-amino salicyclic acid. These are less effective and have more toxic effects.

Third line drugs include: Capreomycin, cycloserine, viomycin, kanamycin and amikacin. These are least effective and have the most toxic effects.

The third line drugs have to be used for infections with tubercle bacilli, likely to be resistant to first and second line drugs or when first and second line drugs have been abandoned because of unwanted reactions. To decrease the possibility of resistant organisms from emerging, ‘Compound Drug Therapy’ is used where a concoction of several drugs is administered.

General Physical Data

Molecular Weight

653.70
Molecular Formula
C25H43N13O8
CAS Registry number
61394-77-2
Beilstein Registry number
876587
Chemical Name
L-3,6-diamino-hexanoyl->-cyclo-[L-2,3-diamino-propionyl->-L-seryl->-L-alanyl->-2-amino-3-ureido
-acryloyl->-(S)-amino-((R)-2-amino-1(3),4,5,6-tetrahydro-pyrimidin-4-yl)-acetyl-(1->N%3&)]
Auto name
3,6-diamino-hexanoic acid [12-hydroxymethyl-3-(2-imino-hexahydro-pyrimidin-4-yl)-9-methyl-
2,5,8,11,14-pentaoxo-6-ureidomethylene-1,4,7,10,13-pentaaza-cyclohexadec-15-yl]-amide

 

Cpm IA10
Cpm IB10
Cpm IIA14
Cpm IIB14
m.p. / oC
240-5
250-3
250
252
[a]D / o
-22.0
-42.5
+9.3
-24.9
UV / nm
0.1 M HCl
269 (e 23, 400)
268 (22, 000)
H2O
268 (23, 200)
268 (21, 900)
0.1 M NaOH
288 (15, 800)
290 (13, 100)
According to the literature9the following applies to naturally occuring capreomycin:
Ratio of IA to IB    = 1.16
Capreomycin II      = 1.5%

13C NMR Data of Cpm IA

Carbon Number
d /  ppm
1
51.92
2
40.28
4
172.76
10
176.29
11
54.15
5, 14
55.66
56.23
7
168.0
8
105.90
13
172.00
16
176.6
17
135.79
19
155.32
20
18.86
21
68.33
22
49.20
23
23.53
24
49.83
26
157.0 (b)
1′
172.0
2′
36.93
3′
49.26
4′
23.59
5′
29.77
6′
39.77

 

 

 

 

The included NMR data is taken from tables in the literature8, 14

The 13C NMR data is that of Capreomycin IA only, and the carbons are numbered accordingly in red on the structure shown above.

Below are 1H NMR tables for the four different naturally occurring forms of capreomycin, the NH protons and CH protons are given in different tables. The NH protons are again numbered on the Cpm IA structure above, but this time in blue. The CH protons are numbered according to their postion in the amino acid residue. These are also numbered in pink on the above diagram.

Chemical Shifts of CH protons in Capreomycin Analogues

Position of Amino Acid Residue

Cpm IA

Cpm IB
Cpm IIA
Cpm IIB
1
a-CH2
2.63 (dd)
2.5 (dd)
2.85 (dd)
2.81 (dd)
b-CH2
3.8 (m)
3.7 (m)
g-CH2
1.8 (m)
1.8 (m)
d-CH2
1.8 (m)
1.8 (m)
e-CH2
3.10 (m)
3.08 (m)
2
a-CH
4.3-3.5 (m)
4.2-4.5 (m)
4.3-4.6 (m)
4.3-4.6 (m)
b-CH2
3.3 (m)
3.3 (m)
3.3 (m)
3.3 (m)
3.8 (m)
3.8 (m)
4.1 (m)
4.1 (m)
3
a-CH
4.86 (t)
4.67 (q)
4.84 (t)
4.68 (q)
b-CH2
3.84 (d)
3.95 (d)
b-CH3
1.43 (d)
1.45 (d)
4
a-CH
4.3-4.5(m)
4.2-4.5 (m)
4.3-4.5 (m)
4.3-4.5 (m)
b-CH2
3.7-4.2 (m)
3.7-4.2 (m)
3.7-4.2 (m)
3.79 (dd)
3.8-4.2 (m)
5
b-CH
8.04 (s)
8.03 (s)
8.05 (s)
8.04 (s)
6
a-CH
5.01 (d)
4.96 (d)
5.01 (d)
4.95 (d)
b-CH
4.5 (m)
4.5 (m)
4.5 (m)
4.5 (m)
g-CH2
1.6-2.3 (m)
1.6-2.3 (m)
1.6-2.3 (m)
1.6-2.3 (m)
d-CH2
3.3 (m)
3.3 (m)
3.3 (m)
3.3 (m)

 

Chemical Shifts of NH Protons of Capreomycin Analogues

Cpm IA
Cpm IB
Cpm IIA
Cpm IIB
1
9.33 (d)
9.72(d)
9.60 (d)
9.50 (d)
2
9.24 (d)
9.24 (d)
9.33 (d)
9.30 (d)
3
8.82 (s)
8.76 (s)
9.10 (s)
9.10 (s)
4
8.64 (d)
8.68(d)
8.73 (d)
8.73 (d)
6
8.22 (t)
8.15 (t)
7
8.10 (t)
8.15 (t)
8.19 (t)
8.08 (t)
8
7.61 (d)
7.62 (d)
7.50 (d)
7.49 (d)
9
7.46 (s)
7.42 (s)
7.44 (s)
7.44 (s)
10
7.46 (s)
7.42 s)
7.31 (s)
7.18 (s)
11
6.48 (s)
6.49 (s)
6.43 (s)
6.34 (s)
12
6.29 (s)
6.34 (s)
6.29 (s)
6.27 (s)

Using the program gNMR I attempted to plot the above data. However, this was not successful as this program can only cope with molecules with up to 23 protons. As this molecule has Capreomycin IA has 43 hydogens, the generated 1H NMR was lacking many essential peaks, and hence was not included.

IR Spectrum of Capreomycin IA

The same process could have done for any of the other three Capreomycin anlogues. The very broad band around 2000 cm-1 upwards is due to the presence of so many nitrogen and carbonyl groups and hence hydrogen bonding.

Cyclo[3-[[(3S)-3,6-diamino-1-oxohexyl]amino]-L-alanyl-(2Z)-3-[(aminocarbonyl)amino]-2,3-didehydroalanyl-(2S)-2-[(4R)-2-amino-3,4,5,6-tetrahydro-4-pyrimidinyl]glycyl-(2S)-2-amino-b-alanyl-L-seryl]

capreomycinIA;Cyclo[3-[[(3S)-3,6-diamino-1-oxohexyl]amino]-L-alanyl-(2Z)-3-[(aminocarbonyl)amino]-2,3-didehydroalanyl-(2S)-2-[(4R)-2-amino-1,4,5,6-tetrahydro-4-pyrimidinyl]glycyl-(2S)-2-amino-b-alanyl-L-seryl] (9CI);1,4,7,10,13-Pentaazacyclohexadecane, cyclic peptide deriv.

37280-35-6

Formula: C25H44 N14 O8
Molecular Weight: 668.83

Properties:Crystals. Mp: 246–248°C.
Synonyms:capreomycin IA;Cyclo[A2pr*-Ser-N3-[(3S)-3,6-diamino-1-oxohexyl]A2pr-2-[(Z)-aminocarbonylaminomethylene]Gly-2-[(4R)-2-iminohexahydropyrimidine-4-yl]Gly-]
Synthesis
Below is the peptide synthesis of capreomycin IA and IB. This was taken directly from the literature10.

 

 

No chemical synthesis of capreomycin could be found in any of the literature references. However, below is a synthesis devised from the peptide synthesis shown above. This is colour coded depending on the various amino residues. Each of the amino groups is added to the molecule in sequence linked by a peptide bond to eventually form the cyclo-structure. This was designed with some help from general references1,2,3. 

  

 

This synthesis would be identical for capreomycin IB other than the Serine-Bzl is replaced by Alanine and the synthesis works in exactly the same way.

Capreomycin
 

The individual components of the capreomycin were colour coded as follows:
Red
DEA / UDA
b, b  diethoxyalanine / b – ureidodehydroalanine
Green
A2pr
a, b � diaminopropionic acid
Turquoise
Ser
Serine
Blue
Cpd
Capreomycidine
Pink
bLys
bLysine

 

The black components of the synthesis were the various protecting groups involved:

Boc
tert � butoxycarbonyl
 
Z
Benzyloxycarbonyl
 
ONSu
N-hydroxysuccinimide
 
Nps
o-Nitrophenylsulphenyl
 
NO2
Nitro
NO2
Bzl
Benzene
 

 

Abbreviation
Chemical Name
NMM
N-Methylmorpholine
DCC
N,N�-Dicyclohexylcarbodiimine
HOBt
l-Hydroxybenztriazole
HONSu
N-Hydroxysuccinimide
THF
Tetrahydrofuran

 

This is the synthesis of capreomycin IA. The IB form is produced in an identical fashion except that Ser � Bzl , is replaced with Ala. 

……………………..

US8044186

 

 

Capastat Sulfate (capreomycin for injection) is a polypeptide antibiotic isolated from Streptomyces capreolus. It is a complex of 4 microbiologically active components which have been characterized in part; however, complete structural determination of all the components has not been established.

Capreomycin is supplied as the disulfate salt and is soluble in water. In complete solution, it is almost colorless.

Each vial contains the equivalent of 1 g capreomycin activity.

The structural formula is as follows:

Capastat Sulfate Structural Formula Illustration

Biological Action

Capreomycin is part of a group of drugs called aminoglycosides. These act to inhibit bacterial protein synthesis. The oxygen-dependent active transport by a polyamine carrier system affects the penetration of the aminoglycosides through the cell membrane of the bacterium. Minimal action on anaerobic organisms is observed. The effect of the aminoglycosides is bactericidal and is enhanced by agents that interfere with cell wall synthesis.

Very little is known about the mechanism of action of capreomycin specifically, but it is thought to inhibit protein synthesis by binding to the 70s ribosomal unit. Other sources6support this theory by suggesting that capreomycin “prevents protein biosynthesis by inhibiting group I intron splicing of RNA as well as blocking translation on the bacterial ribosome via inhibition of ribosomal subunits.” It has been reported14 that the b-amino group of the A2pr residue promotes biological potency, and that its location within the molecule is of importance.

Side Effects

This powerful antimycobacterial agent can give rise to several side effects, some of which are listed below:

The following Nephrotoxic effects are reversible once treatment is stopped, but capreomycin is not recommended for people with kidney disorders.

  • Polyuria (excess urination)
  • Haematuria (red blood cells in the urine)
  • Proteinuria (protein in the urine)
  • Nitrogen metabolism
  • Electrolyte disturbances
  • Anorexia
  • Anaemia
  • Thirst

 

Capreomycin is also Ototoxic giving the following side effects. The nerve damage is permanent.

  • Deafness
  • Loss of vestibular function
  • Damage to the cranial nerve 8
  • References:1. An Introduction to Peptide Chemistry – P.D. Bailey
    2. Organic Chemistry – Vollhardt and Schore
    3. Peptide Synthesis – M. Bodanszky, Y. Klausner and M. Ondetti
    4. Pharmacology – H.P. Rand, M.M. Dale and J.M. Ritter
    5. http://www.aidsinfonyc.org/network/access/drugs/capr.html
    6. http://rwingo1.chm.colostate.edu/group/duane/duane.html
    7. http://www.hucmlrc.howard.edu/Pharmacology/handouts/TBRCLSIS.html
    8. J. Org.Chem.,1977, 42, 8 – McGahren, Morton, Kunstmann, Ellestad
    9. Bull.W.H.O., 1972, 47(3), 343-56 – Lightbrown et al.
    10. Tetrahedron, 1978, 34(7), 912-7 – Nomoto, Teshima, Wakamiya, Shiba
    11. Tetrahedron Letters, 1976, 43, 3907-10 – Shiba, Nomoto, Teshima, Wakamiya
    12. J.Org.Chem., 1992, 57, 5214-5217 – Gould and Minott
    13. Tetrahedron Letters, 1969, 30, 2549-41 – Bycroft, Cameron, Hassanali-Walji and Johnson
    14. Bull.Chem.Soc.Jpn, 1979, 52(6), 1709-15 – Nomoto and Shiba
    15. Experimentia – 1976, 32(9), 1109-11 – Nomoto and Wakamiya
    16. Pharmazie – 1970, 25(8), 471-2 – Voigt and Maa Bared
    17. Antimicrobial Agents Chemotherapy, 1964, 522-9 – Black, Griffith and Brickler
    18. Antimicrobial Agents Chemotherapy, 1962, 201-12 – Herr
    19. www2.chemie.uni-erlangen.de/services/telespec
  • 20 “Capreomycin binds across the ribosomal subunit interface using tlyA-encoded 2′-O-methylations in 16S and 23S rRNAs”. Mol. Cell 23 (2): 173–82. July 2006. doi:10.1016/j.molcel.2006.05.044PMID 16857584
  • 21   http://www.toku-e.com/Assets/MIC/Capreomycin%20sulfate.pdf
  • CAPREOMYCIN wiki
Systematic (IUPAC) name
(3S)-3,6-diamino-N-[[(2S,5S,8E,11S,15S)-15-amino-11-[(4R)-2-amino-3,4,5,6-tetrahydropyrimidin-4-yl]-8-[(carbamoylamino)methylidene]-2-(hydroxymethyl)-3,6,9,12,16-pentaoxo-1,4,7,10,13-pentazacyclohexadec-5-yl]methyl]hexanamide; (3S)-3,6-diamino-N-[[(2S,5S,8E,11S,15S)-15-amino-11-[(4R)-2-amino-3,4,5,6-tetrahydropyrimidin-4-yl]-8-[(carbamoylamino)methylidene]-2-methyl-3,6,9,12,16-pentaoxo-1,4,7,10,13-pentazacyclohexadec-5-yl]methyl]hexanamide
Clinical data
AHFS/Drugs.com monograph
MedlinePlus a682860
 
Identifiers
CAS number 11003-38-6 
 
Chemical data
Formula C25H44N14O8 
Mol. mass 668.706 g/mol
 
Share

Risk Assessment of Potentially Genotoxic Impurities within the Framework of Quality by Design

 regulatory, Uncategorized  Comments Off on Risk Assessment of Potentially Genotoxic Impurities within the Framework of Quality by Design
Feb 032014
 

Figure

 

A strategy for the risk assessment of potentially genotoxic impurities is described that utilizes Quality by Design in an effort to furnish greater process and analytical understanding, ultimately leading to a determination of impurity criticality. By identifying the risks and parameters that most influence those risks, an enhancement of both product and process control is attained that mitigates the potential impact of these impurities. This approach calls for the use of toxicological testing where necessary, chemical fate arguments when possible, multivariate analyses to develop design space, and use of spiking data to support specifications. Strong analytical support, especially with the development of low-level detection methods, is critical. We believe that this strategy not only aids in the development of a robust API process but also delivers on the identification and subsequent mitigation of risks to a class of impurities that are of high interest in the field.

Risk Assessment of Potentially Genotoxic Impurities within the Framework of Quality by Design

Adam R. Looker, Michael P. Ryan, Bobbianna J. Neubert-Langille and Redouan Naji
Org. Process Res. Dev., 2010, 14 (4), pp 1032–1036
pp 1032–1036
Publication Date (Web): April 7, 2010 (Communication)
DOI: 10.1021/op900338g
Figure
Share
Feb 022014
 

Zanamivir

139110-80-8

APPROVED 26-7-96……. GSK NDA 021036

A guanido-neuraminic acid that is used to inhibit neuraminidase.

Zanamivir INN /zəˈnæmɨvɪər/ is a neuraminidase inhibitor used in the treatment and prophylaxis of influenza caused by influenza A virus andinfluenza B virus. Zanamivir was the first neuraminidase inhibitor commercially developed. It is currently marketed by GlaxoSmithKline under the trade name Relenza as a powder for oral inhalation.

The drug is approved for use for the prevention and treatment of influenza in those over the age of 7 in the United States, Canada, European Union, and many other countries. It is not recommended for people with respiratory problems and ailments.

United States 6294572 APPROVED  1994-12-15 EXPIRY 2014-12-15
United States 5360817                    1993-07-26             2013-07-26
Canada 2291994                    2003-10-14             2011-04-24
Canada 2081356                    2000-02-22             2011-04-24
Patent No PatentExpiry use code
5360817 Jul 26, 2013
5648379 Jul 15, 2014 U-274
5648379 Jul 15, 2014 U-721
5648379 Jul 15, 2014 U-722
6294572 Dec 15, 2014

Zanamivir was discovered in 1989 by scientists led by Peter Malcolm Colman and Joseph Varghese at the CSIRO, in collaboration with theVictorian College of PharmacyMonash University, and scientists at Glaxo, UK. Zanamivir was the first of the neuraminidase inhibitors. The discovery was initially funded by the Australian biotechnology company Biota and was part of Biota’s ongoing program to develop antiviral agents throughrational drug design. Its strategy relied on the availability of the structure of influenza neuraminidase, by X-ray crystallography. It was also known, as far back as 1974, that 2-deoxy-2,3-didehydro-N-acetylneuraminic acid (DANA), a sialic acid analogue, is an inhibitor of neuraminidase. Sialic acid (N-acetyl neuraminic acid, NANA), the substrate of neuraminidase, is itself a mild inhibitor of the enzyme, but the dehydrated derivative DANA, a transition-state analogue, is a better inhibitor.

Computational chemistry techniques were used to probe the active site of the enzyme, in an attempt to design derivatives of DANA that would bind tightly to the amino acid residues of the catalytic site, and so would be potent and specific inhibitors of the enzyme. The GRID software by Molecular Discovery was used to determine energetically favourable interactions between various functional groups and residues in the catalytic site canyon. This investigation showed that there is a negatively charged zone in the neuraminidase active site that aligns with the C4hydroxyl group of DANA. This hydroxyl is, therefore, replaced with a positively charged amino group; the 4-amino DANA was shown to be 100 times better as an inhibitor than DANA, owing to the formation of a salt bridge with a conserved glutamic acid (119) in the active site. It was also noticed that Glu 119 is at the bottom of a conserved pocket in the active site, just big enough to accommodate a more basic functional positively charged group, such as a guanidino group, which was also larger than the amino group. Zanamivir, a transition-state analogue inhibitor of neuraminidase, was the result.

As Biota was a small company, it did not have the resources to bring zanamivir to market by itself. In 1990, zanamivir patent rights were licensed to Glaxo, now GlaxoSmithKline (GSK). In 1999, the product was approved for marketing in the US and subsequently has been registered by GSK in a total of 70 countries (GlaxoSmithKline News release, 2006). Zanamivir is delivered via Glaxo’s proprietary Diskhaler inhalation device. The license agreement entitled Biota to receive a 7% royalty on Glaxo’s sales of zanamivir.

Chemical name:

5- Acetamido- 2, 6- anhydro- 3, 4, 5- trideoxy- 4- guanidino- D- glycero- D- galacto- non- 2- enonic acid
Synonyms: Zanamivir, GG167, 4-guanidino-Neu5Ac2en and 2,3- Didehydro- 2, 4- dideoxy- 4- guanidino- N- acetyl- D- neuraminic acid(2R,3R,4S)-4-guanidino-3-(prop-1-en-2-ylamino)-2-((1R,2R)-1,2,3-trihydroxypropyl)-3,4-dihydro-2H-pyran-6-carboxylic acid
Empirical formula:

C12H20N4O7

Structural formula:
Molecular weight: 332.31g
Beilstein number: 7083099
Normal State: Powder
Colour: White to ‘off white’
Melting point: 325oC
Optical rotary power: Type []Conc: 0.9g/100mlSolvent: H2OOptical rotary power: 41 degWavelength: 589nmTemp: 20oC
 CAS number: 139110-80-8 
Solubility: 18mg/mL in water at 20oC

Zanamivir is used for the treatment of infections caused by influenza A virus and influenza B virus. There is low to moderate evidence that it decreases the risk of one’s getting influenza by 1% to 12% in those exposed. In otherwise-healthy individuals, benefits overall appear to be small.It is unclear whether it affects the risk of one’s need to be hospitalized or the risk of death. An independent analysis of its effects by the Cochrane collaboration was awaiting release of trial data as of 2012. The evidence for a benefit in preventing influenza is weak in children with concerns of publication bias in the literature. As of 2009 no influenza has shown any signs of resistance. Since then genes expressing resistance to were found in patients infected with Influenza A H7N9 and who were treated with corticosteroids.

ZANAMIVIR

Mass

1H NMR
Hydrogen Chemical shift /ppm
(1H, d, 3-H) 5.53
(2H, 2dd, 4- and 6-H) 4.50 – 4.38
(1H, dd, 5-H) 4.21
(2H, dd+ddd, 9-Ha and 8-H) 4.00-3.88
(2H, 2dd, 9-Hb and 7-H) 3.70-3.62
(3H, s,  Ac) 2.05

 

13C NMR
Carbon Shift /ppm
(C=O, Ac) 177.3
(C-1) 172.1
(guanidino) 159.9
(C-2) 152.1
(C-3) 106.8
(C-6) 78.3
(C-8) 72.6
(C-7) 71.0
(C-9) 65.9
(C-4) 54.0
(C-5) 50.6
(Me) 24.8

ref 12

IR spectra:

The following peaks are present in the IR spectra of Relenza: 3332cm-1, 1676cm-1, 1600cm-1, 1560cm-1, 1394cm-1, 1322cm-1 and 1281cm-1.

UV spectra

The maximum peak is 235nm giving E = 199 dm-3 mol-1cm-1

ref 13for above

Although zanamivir was the first neuraminidase inhibitor to the market, it had only a few months lead over the second entrant, oseltamivir (Tamiflu), with an oral tablet formulation.

According to the CDC, Tamiflu, zanamivir’s main competitor, is not as effective at treating the influenza viruses as zanamivir, especially in H1N1 seasonal flu. In fact, tests showed 99.6% of the tested strains of seasonal H1N1 flu and 0.5% of 2009 pandemic flu were resistant to Tamiflu, while no flu samples, seasonal or pandemic, showed any resistance to zanamivir.

When first marketed in the US in 1999/2000, zanamivir captured only 25% of the influenza antiviral market, despite a huge promotional campaign. By the end of that season, Tamiflu was outselling zanamivir 3:1. During that season, zanamivir experienced worldwide safety warnings involving the risk of bronchospasm and death. Glaxo then reduced the marketing of zanamivir, and Tamiflu’s dominance increased. More than US$20 million worth of zanamivir sold by Glaxo in the first US season was returned to the company in the next two seasons because zanamivir’s sales to patients were far less than expected.

Biota commenced legal proceedings in 2004 alleging Glaxo’s reduced marketing of zanamivir to be a breach of contract. Biota claimed approximately A$700m from Glaxo. After Biota spent four years trying to progress its case, and incurring A$50m in legal costs, the company abandoned the claim in July 2008, recovering only A$20 million, including legal costs following settlement at mediation. Biota had refused an earlier tactical offer from Glaxo of A$75 million plus legal costs.

In August 2006, Germany announced it would buy 1.7 million doses of zanamivir, as part of its preparation strategy against bird flu. “Germany’s purchase shows that countries are starting to take a balanced view of influenza preparedness,” says Simon Tucker, head of research at Melbourne-based Biota, where zanamivir was originally developed.

In April 2009, many cases of swine flu (H1N1-type virus) were reported in US and Mexico. Zanamivir is one of only two drugs prescribed to treat it. A study published in June 2009 emphasized the urgent need for augmentation of oseltamivir (Tamiflu) stockpiles, with additional antiviral drugs including zanamivir, based on an evaluation of the performance of these drugs in the scenario that the 2009 H1N1 swine flu neuraminidase (NA) were to acquire the Tamiflu-resistance (His274Tyr) mutation, which is currently widespread in 99.6% of all tested seasonal H1N1 strains.n January 2011, GSK announced that it would commence phase III trials for intravenous zanamivir in a study that will span 20 countries in the Northern and Southern Hemispheres.

Recently, the reported oseltamivir-resistance H5N1 virus neuraminidase still retaining susceptibility to zanamivir indicates that the structure of zanamivir has some advantages over oseltamivir in binding to the active pocket of H5N1 neuraminidase.

As a proven anti-influenza drug target, neuraminidase continues to be attractive for the development of new inhibitors. The crystal structure of H5N1 avian influenza neuraminidase (PDB code: 2HTY) provides the three-dimensional structural information and opportunity for finding new inhibitors in this regard, because the existing inhibitors, such as oseltamivir and zanamivir, were developed based on different structures of neuraminidase, such as subtypes N9 and N2, and type B genus of influenza virus.

ZANAMIVIR

Chemistry

Zanamivir synthesis.png

  1. Scheigetz, J.; Zamboni, R.; Bernstein, M. A.;Roy, B. (December 1995). “A syntheses of 4-a-guanidino-2-deoxy-2,3-didehydro n-acetylneuraminic acid”Organic Letters 27 (6): 637–644.doi:10.1021/ol901511x. Retrieved 2010-11-14.

Zanamivir synthetic process in the world

Together with oseltamivir, zanamivir is the only medicine which can prevent influenza on humans caused by H5N1 and H1N1 virus. Vietnam prepared oseltamivir (Tamiflu) medicine. But there was no zanamivir – the first influenza medicine belonging N1 kind, discovered and commercialized before oseltamivir. The scientific name of zanamivir is acid 5-acetamido-4-guanidino-6-(1,2,3-trihydroxy-propyl)-5,6-dihydro-4H-pyran-2-carboxylic. The discovery of zanamivir opens research possibilities for new medicines which have the same effect on enzyme neuraminidase inhibitor to prevent and treat influenza.

Acid sialic is an input to synthetize zanamivir. The name acid sialic (Neu5Ac2en) is used to indicate derivation at O- and N- positions of acid neuraminic, just for acid N-axetylneuraminic. Acid sialic of carbohydrate groups is on animal cells and microorganism, especially in glycoprotein and gangliosid. The commercial acid sialic is extracted from whey of the cheese and milk process as well as egg yolk, and costs about 5,000 USD per kilo.

In 1994, zanamivir was first synthesized and made public by Von Itzstein and other scientists from the Department of Pharmaceutical Chemistry under Monash University (Australia). Then, Chandler and co-workers of Glaxo company (GSK, Britain) acquired results, improved reaction steps and made them public in 1995. Accordingly, this method produced 8.3% of general output. The synthetic process is described in Figure 1.


Figure 1: Zanamivir synthetic process according to Chandler

Up to now, the research of Chandler has been the only publication about zanamivir synthetic method, the output of which is greater than milligrams, and it reproduces details about reaction conditions and physiochemical properties of the requisite substances.

Recently, a research group of Yao (China) proposed a new approach to synthetize into intermediate compound 5. Researchers started from another material – D-glucono-δ-lactone, which is cheaper than acid sialic. However, the synthetic process is longer and much complicated, including 24 steps, with lower productivity (0.2%).

Researching on synthesizing Zanamivir from Acid sialic by Institute of Chemistry

Synthetizing methyl N-acetylneuraminate (2) and O-pentaacetoxy (3) from acid sialic

Scientists from the Institute of Chemistry used acid sialic (axit N-acetylneuraminic) 98% from China as the input for the zanamivir synthetic process. They decided to use the method of Warner, using ion exchange resin Dowex-H, with the role of catalyst. Reaction was performed in the room in 10 hours. The output was metyl (2) este product of acid N-acetylneuraminic with a productivity of 99%.

Then, to synthetize O-pentaacetoxy (3), scientists applied axetyl effective chemistry method recently published, using BF3.OEt2catalysis at 00C. Productivity in this case exceeded 95%.


Figure 2: The diagram of O-pentaacetoxy 3 derivative making

The use of catalysts which were ion exchange resin Dowex-H (for este chemical reaction) and BF3.OEt2 (for axetyl chemical reaction) had more advantages than the method by scientists from Glaxo.

Synthesizing intermediate compound – oxazoline (4) key from O-pentaacetoxy (3)


Figure 3: Diagram to synthesize oxazoline (4) from O-pentaacetoxy (3) according to a and b methods

Firstly, scientists conducted a survey on oxazoline (4) synthetic process according to Chandler’s process. O-pentaacetoxy (3) compound was separated from two types of OAc and formed oxazoline round thanks to the effect of strong acid Lewis, which was TMSOTf at 520C in 2.5 hour. The productivity of this reaction achieved 40%. The pilot instead of TMSOTf by BF3.OEt2 catalysis in dichloromethane at room temperature at night, the productivity of the reaction to form oxazoline round from penta-acetoxy (5) was similar to the method using TMSOTf (42%). To increase productivity, scientists made a survey on one-pot method, directly from metyl este (2) to oxazoline (4), without passing O-pentaacetoxy (3), gave the highest productivity (73,3%) and was the most economic effectiveness.

Synthesizing zanamivir from oxazoline (4) intermediate compound

The next, scientists successfully conducted reactions from oxazoline (4) intermediate compound to Zanamivir (9) final product (Figure 1). Zanamivir product had IR and NMR data which were compatible with their structure.

Therefore, scientists from the Institute of Chemistry under Vietnam Academy of Science and Technology built a stable process, including seven major steps, synthesizing from acid sialic with the general productivity of 6.6% (the productivity made public in the world was 8.3%). Especially, in the first period, from acid sialic to oxazolin (4) was optimized and gave a general productivity of 74%, higher than the productivity made public by (61.7%). However, the productivity gained in the later period is still low. Now, synthesizing zanamivir influenza medicine still continues to be researched.

……………………

Beau and coworkers assembled the core dihydropyran framework of zanamivir congeners via a combination of PBM reaction and Iron(III)-promoted deprotection-cyclization sequence. A stereochemically-defined α-hydroxyaldehyde 2, diallylamine and a dimethylketal-protected boronic acid 1 is coupled to form the acyclic, stereochemically-defined amino-alcohol 3, which then undergoes an Iron(III)-promoted cyclization to form a bicyclic dihydropyran 4. Selective opening of the oxazoline portion of the dihydropyran intermediate 4 with water or timethylsilyl azide then furnish downstream products that have structures resembling the Zanamivir family members.

zanamivir core_Beau et al.
Reaction scheme part 1:
The commercially available N-acetyl-neuraminic acid 1 is the starting reagent for the most direct approach to the synthesis of 4-guanidino-Neu5Ac2en (Relenza). In reaction scheme 1 the steps for the conversion of N-acetyl-neuraminic acid 1 to its 4-amino analogue is shown. Step 1 is the addition of methanolic HCl (MeOH and HCl gas), which produces the methyl ester of 1, followed by acetic anhydride in pyridine with 4-(dimethylamino)pyridine catalysis, which produces the penta-acetoxy compound, 2. In step 2, 2 is converted into the oxazoline 3 at high yield using trimethylsilyl trifluoromethanesulfonate (TMSOTf) in ethyl acetate at 52oC. In step 3, the azido compound, 4, is produced by the reaction of 3 with trimethylsilyl azide in tert-butyl alcohol at 80oC. In step 4 catalytic sodium methoxide in methanol was used to remove the acetate protecting groups from 4 to give triol 5. The 4-amino analogue, 6 was made in step 5, by hydrolysis using triethylamine in water, hydrogenolysis with a Lindlar catalyst and finally the addition of Dowex 2 * 8 resin. The triethylamine salt of the 6 was made during hydrogenolysis  and the purpose of the Dowex 2 * 8 resin was to desalt this intermediate. The chemical names of the compounds are:
1: N-acetyl-neuraminic acid
2: 5- Acetamido- 3,5- dideoxy- D- glycero- �- D- galacto- 2- nonulo- pyranosonic acid methyl ester
3: Methyl (3aR, 4R, 7aR)- 2- Methyl- 4- [(1’S, 2’R)- 1′, 2′, 3′ – triacet- oxypropyl]- 3a, 7a- dihydro- 4H- pyrano [3, 4-d] oxazole- 6- carboxlate.
4: 5- Acetamido- 7, 8, 9- tri- O- acetyl- 2, 6- anhydro- 4- azido- 3, 4, 5- trideoxy- D- glycero- D- galacto- non- 2- enonic acid methyl ester.
5: 5- Acetamido- 2, 6- anhydro- 4- azido- 3, 4, 5- trideoxy- D- glycero- D- galacto- non- 2- enonic acid methyl ester.
6: 5- Acetamido- 4- amino- 2, 6- anhydro- 3, 4, 5- trideoxy- D- glycero- D- galacto- non- 2- enonic acid.

Part one of reaction scheme

Synthesis of reactant necessary for part 2 of reaction:
Aminoiminomethane-sulfonic acid (AIMSA), 7, which is necessary for the conversion of compound 6 into Relenza, 9, is synthesised in Reaction scheme 2. The oxidizing solution necessary for the reaction is prepared by the addition of peracetic acid to 30% hydrogen peroxide and then conc. sulfuric acid. This is followed by acetic anhydride and, once the reaction has completed, methanol. Thiourea is dissolved in methanol and added slowly to the oxidizing solution.to produce compound 7. Note that any crystals that form are removed and that the reaction needs to be carried out under cooled conditions. See the reference source for more experimental details.

Synthesis of AIMSA

Reaction scheme part 2:
Reaction scheme 3 shows the conversion of compound into Relenza For route A, 3 mol equivalent of AIMSA, 7, and 3 mol equivalent of potassium carbonate are added in a portionwise manner to compound over an eight hour period. A yield of about 48% of the crystalline product should be obtained for this method. An alternative route is to treat compound 6 with 1.1 mol equivalent of cyanogen bromide in the presence of sodium acetate in methanol. Route B step 1 gives compound 9, which can be converted into the final product 8 by treating it with ammonium hydroxide and ammonium formate at 85oC. A 36% yield of the purified product can be obtained after purification with ion-exchange chromatography and crystallisation. The chemical names of the compounds in this scheme are:
8. 5- Acetamido- 2, 6- anhydro- 3, 4, 5- trideoxy- 4- guanidino- D- glycero- D- galacto- non- 2- enonic acid. (Relenza)
9. 5- Acetylamino- 2, 6- anhydro- 4- cyanoamino- 3, 4, 5- trideoxy- D- glycero- D- galacto- non- 2- enonic acid

Part 2 of reaction scheme

 

ref are 13 and 14

,,,,,,,,,,,,,,,,,,,,,,,,,,,,,

SYNTHESIS FROM PATENT  EP2276479A2

ZANAMIVIR AND BOC PROTECTED ZANAMIVIR

The synthesis of zanamivir is shown in Scheme 1. The starting material used for zanamivir synthesis is sialic acid 1, which was converted to the methyl ester 2, in presence of Dowex H+ as described in detail in reference 104. The hydroxyl groups of 2 are protected with acetyl groups to give compound 3, which was then converted to the oxazoline derivative 4 in the presence of trimethyltrifluoromethanesulfonate as described in detail in reference 105. Azide 5 was synthesized from 4 in presence of azidotrimethylsilane as described in detail in reference 105. The azide is reduced to the corresponding amine 6 by using Lindlar’s catalyst, and the amine is in turn converted to the guanidine derivative 7 as described in detail in reference 106. The final step involves the deprotection of the methyl ester and acetyl groups in the presence of methanolic sodium hydroxide to give Boc-protected zanamivir 8 as described in detail in reference 106. 8, 1H NMR (CD3OD) δ (ppm) 5.6 (d, J = 2.0 Hz, IH), 5.01 (dd, J = 9.6, 2.1 Hz, IH), 4.25 (dd, J = 10.8, 1.1 Hz, IH), 4.18 (dd, J = 10.6, 9.6 Hz, IH), 3.89 (ddd, J = 9.4, 6.2, 2.7 Hz, IH), 3.84 (dd, J = 11.3, 2.8 Hz, IH), 3.67 (dd, J = 11.3, 5.8 Hz, IH), 3.57(d, J = 9.3 Hz, IH), 1.9 (s, 3H), 1.55 (s, 9H), 1.50 (s, 9H); ESI-MS: 533 (M+H)+.

Scheme 1

 

Figure imgf000020_0001
Figure imgf000020_0003
Figure imgf000020_0002

a) Dowex H Methanol b) Aceticanhydride DMAP pyridine c) trimethylsilyl tπfluorαmethane sulfonate ethylacetate d) azidotrimethylsilane butanol e) Lindlar’s catalyst ethanol f) N N’-bis-tert-butoxycarbonyMH-pyrazole-i carboxamidine tetrahydrofuran g) sodium hydroxide methanol

104. Martin, R., K.L. Witte, and C-H. Wong, The synthesis and enzymatic incorporation of sialic acid derivatives for use as tools to study the structure, activity, and inhibition of glycoproteins and other glycoconjugates. Bioorganic & Medicinal Chemistry, 1998. 6(8): p. 1283-1292.

105. Malcolm Chandler, M.J.B., Richard Conroy, Brian Lamount, Bina Patel, Vipulkumar K. Patel, Ian P. Steeples, Richard Storer, Naill G. Weir, Michael

Wrightm Christopher Williamson, Synthesis of the potent influenza neuraminidase inhibitor 4-guanidino Neu5Ac2en. X-Ray molecular structure of S-acetamido^-amino^^-anhydro-S^^-trideoxy-D-erythro-L-gluco- nononic acid. J. Chem. Soc, Perkin Trans. 1, 1995: p. 1173 – 1180.

106. Masuda, T., et al., Synthesis and anti-influenza evaluation of polyvalent sialidase inhibitors bearing 4-guanidino-Neu5Ac2en derivatives. Chem Pharm Bull (Tokyo), 2003. 51(12): p. 1386-98

,,,,,,,,,,,,,,,,,,,,,,,,,,,,,

The active component of RELENZA is zanamivir. The chemical name of zanamivir is 5- (acetylamino)-4-[(aminoiminomethyl)-amino]-2,6-anhydro-3,4,5-trideoxy-D-glycero-D-galacto non-2-enonic acid. It has a molecular formula of C12H20N4O7 and a molecular weight of 332.3. It has the following structural formula:

 

RELENZA<br /><br /><br /><br /><br /><br /><br /><br /><br /><br /><br /><br /><br /> (zanamivir) Structural Formula Illustration

 

Zanamivir is a white to off-white powder for oral inhalation with a solubility of approximately 18 mg/mL in water at 20°C.

RELENZA is for administration to the respiratory tract by oral inhalation only. Each RELENZA ROTADISK contains 4 regularly spaced double-foil blisters with each blister containing a powder mixture of 5 mg of zanamivir and 20 mg of lactose (which contains milk proteins). The contents of each blister are inhaled using a specially designed breath-activated plastic device for inhaling powder called the DISKHALER. After a RELENZA ROTADISK is loaded into the DISKHALER, a blister that contains medication is pierced and the zanamivir is dispersed into the air stream created when the patient inhales through the mouthpiece. The amount of drug delivered to the respiratory tract will depend on patient factors such as inspiratory flow. Under standardized in vitro testing, RELENZA ROTADISK delivers 4 mg of zanamivir from the DISKHALER device when tested at a pressure drop of 3 kPa (corresponding to a flow rate of about 62 to 65 L/min) for 3 seconds.

CLIP

On Zanamivir

Total Synthesis of Anti-Influenza Agents Zanamivir and Zanaphosphor via Asymmetric Aza-Henry Reaction

Department of Chemistry, National Taiwan University, Taipei 106, Taiwan
The Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
Org. Lett., 2016, 18 (17), pp 4400–4403
Abstract Image

The potent anti-influenza agents, zanamivir and its phosphonate congener, are synthesized by using a nitro group as the latent amino group at C4 for asymmetric aza-Henry reaction with a chiral sulfinylimine, which is derived from inexpensive d-glucono-δ-lactone to establish the essential nitrogen-containing substituent at C5. This method provides an efficient way to construct the densely substituted dihydropyran core of zanamivir and zanaphosphor without using the hazardous azide reagent.

Zanamivir as the TFA salt (40 mg, 90 %). C14H21F3N4O9; colorless solid, mp 260262 oC;
1H NMR (400 MHz, D2O) δ 5.67 (1 H, d, J = 2.1 Hz), 4.48 (1 H, dd, J = 9.3, 2.1 Hz), 4.41 (1H, d, J = 10.6 Hz), 4.26 (1 H, dd, J = 10.6, 9.3 Hz), 3.98–3.90 (2 H, m), 3.71–3.66 (2 H, m),2.06 (3 H, s);

13C NMR (100 MHz, D2O) δ 174.5, 166.4, 162.9 (CO2 of TFA, q, J = 35.4 Hz ),157.0, 146.2, 116.3 (CF3 of TFA, q, J = 290.2 Hz ), 107.2, 75.8, 69.8, 67.9, 62.9, 50.8, 47.6,21.9;

ESI–HRMS calcd for C12H20N4O7Na: 355.1230, found: m/z 355.1288 [M + Na]+.

str1 str2

Introduction

Relenza (Zanamivir for oral inhalation) is the first in a new generation of influenza virus-specific drugs known as neuraminidase inhibitors, which work by interferring with the life cycles of influenza viruses A and B. It prevents the virus spreading infection to other cells by blocking the neuraminidase enzyme present on the surface of the virus. Relenza is available as a powder that is administered by inhalation of 2 blisters from the rotadisk inside the diskhaler (Fig. 1) twice daily for five daysThis means that 20mg of Relenza is delivered to the principal site of viral replication each day.The main method for preventing influenza since the 1960s is by vaccination and although this and anti-viral drugs such as amantadine and its analogue rimantadine have long been available (since 1976 and 1993 respectively), they are only of limited use because of the constant mutation of the virus. This chameleon-like nature also means that the virus can become unrecognizable to the human immune system and thus repeatedly infect millions of people year after year.

Fig 1: The diskhaler used to administer Relenza. Each blister in the Rotadisk contains 5mg of the drug

Why there is a need for a more effective influenza treatment: At present influenza is basically an uncontrolled disease and an effective method is needed for both the prevention and treatment of it. In the 20th century there were some major pandemics such as the 1918-1919 Spanish ‘flu which killed 20 million people world wide, the 1957 Asian ‘flu, the 1968 Hong Kong ‘flu and the 1977 Russian ‘flu12 These viruses also affect different animals, especially domesticated chickens and turkeys and in Hong Kong in 1997 a virulent bird flu virus, started infecting and killing people for the first time ever. Of the 18 people affected 6 died, although there was no evidence that the virus was able to spread between people. Given the antigenic properties of the influenza virus, in the future the virus may be passed from person to person, and because human immune systems are not prepared for avian viruses the effects on the population could be grave. It would not be possible to prepare vaccines in time and anti-viral drugs are not always adequate.

Advantages of Relenza over previous treatments:

Relenza has a number of advantages over the existing treatments for influenza. It does not cause significant side effects and the development of zanamivir-resistant viruses is not expected to occur readily in patients. This is because selection of drug-resistant mutants characterized by changes in neuraminidase requires prolonged passage in tissue culture and may be a biological cripple. If started within two days of the onset of influenza symptoms and if a fever is present, the duration of illness is decreased by an average of 1.5 days. It appears to decrease the severity of flu symptoms for the remainder of the illness, as well as decreasing the number of complications from the flu. It is also possible that Relenza could be used as a method of ‘flu prevention although it has not yet been approved for this use.

 

Comparison of the symptoms of the ‘flu with that of a common cold:

People infected by an influenza virus suffer a lot more than those with a cold. As you can see from the table below, some of the symptoms are similar, but with a cold they are less severe.Influenza also becomes more serious when it leads to secondary bacterial pneumonia or primary influenza viral pneumonia or when it exacerbates underlying medical conditions such as pulmonary or cardiac disease. In children, the symptoms are similar to those observed in adults, however children often have higher fevers and younger ones may develop gastrointestinal manifestations. It should be noted that Relenza is not effective on people with colds or other viral illnesses.

 

Influenza Cold
Sore throat Mild sore throat
  High fever and chills Low-grade fever
Non-productive cough Cough
Severe muscle aches   Congestion
Headache
Intense fatigue.

The effect of Relenza on patients with respiratory diseases:Relenza is not generally recommended for the treatment of patients with respiratory dieseases such as asthma or chronic obstructive pulmonary disease (COPD) and has carried an approval since its approval in July 1999. Some patients with underlying airway diseases have experienced serious adverse events following treatment, with some fatal outcomes although causality has been difficult to establish. It has been recommended that patients with asthma have a fast-acting bronchodilator inhaler available and use it about 15 minutes before taking Relenza

Successfulness of Relenza:The sialidase inhibitory activities (determined by methods described in reference 7) of Relenza compared to the more recent neuraminidase inhibitor Oseltamivir are shown in the table below9.IC50 is the concentration that reduces enzyme activity by 50%.

Compound Influenza A IC50 (�M) Influenza B IC50 (�M)
Relenza 0.005 0.004
Oseltamivir 0.002 0.032

The results demonstrate that both compounds are good inhibitors of influenza A and B, with Oseltamivir being more selective towards Influenza A and Relenza showing a better overall performance. In phase I and II tests reported by the Lancet5, no important adverse effects were found in healthy patients or those reported to have mild to moderate asthma following an inhaled administration of 40mg/day of Relenza. There was a significant improvement of the symptoms of people taking Relenza compared to those taking the placebo.

1940s: Discovery that the influenza virus’s enzyme was destroying receptors on red blood cellsF.This was discovered by George Hirst, who noticed that when red blood cells were mixed with fluids from influenza infected chicken embryos in cold conditions the cells were very heavily agglutinated by the virus. These red cells dispersed when warmed up and could not be re-agglutinated in the cold with fresh virus. This led him to the conclusion that the influenza virus’s enzyme was destroying receptors on red blood cells.

The finding of sialidase (also known as neuraminidase):Alfred Gottschalk heard of Hirst’s experiment and interpretation of results, and this led him to believe that there was a “split product”. He discovered sialic or neuraminic acid (Fig 2), a type of sugar, and the enzyme on the virus was called neuraminidase (or sialidase). At this time it was thought that it was the neuraminidase which was responsible for the observations made by Hirst, but it was later shown by Robin Valentine, W. Graeme Laver, Norbert Bischofberger and Robert G. Webster that the hemagglutinin (receptor-binding) and neuraminidase (receptor-destroying) activities of the virus resided in two quite different spikes on the surface of the virus.

Fig 2: Sialic Acid

Discovery of how new pandemic strains of ‘flu A occured.

Ed Kilbourne, W. Graeme Laver, Norbert Bischofberger and Robert G. Webster realised that hybrid viruses could be formed by infecting cells simultaneously with two different Type A flu viruses. This was because the RNA pieces coding the various virus proteins reassorted, some of the viruses contained the hemagglutinin from one parent and the neuraminidase from the other. This “mating” of two parent viruses to give a hybrid virus explained how new pandemic strains of ‘flu A occurred, and led to a very good way of producing influenza viruses with any desired combination of hemagglutinin and neuraminidase spikes. This helped towards finding a way of producing pure neuraminidase which was later essential for crystal growth and drug design experiments.

The crystallization of neuraminidase:

Laver, Bischofberger and Webster isolated one type of influenza virus by sucking off the allantoic fluid surrounding the embryo of infected chicken eggs and purifying this. The virus particles were incubated with an enzyme capable of digesting proteins. This enzyme was selected to split the “heads” of the neuraminidase spikes off the virus particle without destroying them and to leave behind or destroy the hemagglutinin spike. The neuraminidase “heads” obtained were concentrated using high-speed centrifugation. The tiny pellet of neuraminidase heads examined had a crystalline appearance, and X-ray diffraction analysis of larger crystals showed that they were made of protein.

Neu5Ac2en (DANA) was shown to inhibit influenza neuraminidase:

Different variants of ‘flu neuraminidase were known to exist, each containing an amino acid sequence that varies between types of neuraminidase apart from one small sequence.It was seen that the conserved amino acids came together when the neuraminidase polypeptide folded up to form the active enzyme. This formed a well conserved cavity which was the active catalytic site of the neuraminidase enzyme. It became apparent that a plug-drug could be made to exactly fit into the active site and inhibit the neuraminidase activity from other influenza viruses. A synthetic analog of sialic acid called Neu5Ac2en (DANA) (Fig 3) was shown to inhibit the influenza virus neuraminidase, but not sufficiently enough to be used treatment for the ‘flu in humans.

Fig 3: Neu5Ac2en (DANA)

Fig 3: Neu5Ac2en (DANA)

The plug drug.Mark von Itzstein and colleagues discovered that replacing the OH at the 4 position of sialic acid with a positively charged amino group made a better inhibitor than sialic acid or its analogue, DANA. Replacing the OH at the 4 position of sialic acid with a guanidino group led to a potent inhibitor of ‘flu neuraminidase. This compound was given the names GG167 and Zanamivir and is now more commonly known as Relenza. Peter Colman soaked the substrate for sialic acid in neuraminidase crystals and used X-ray crystallography to determine the three-dimensional structure of the crystals. The strong binding of Relenza by ‘flu neuraminidase which was seen is due to the positively charged guanidino group being anchored by the negatively charged glutamic acids. More details about this are provided in the immunology section.

Immunology

Fig 4: The influenza viruses as seen under the electron microscope. Neuraminidase and haemagglutin spikes are visible.

Structure of the flu virus:Influenza (Fig 4) is an RNA virus which may exist as any shape from round balls to long, spaghetti-like filaments. The genome of this virus is associated with five different viral proteins and is surrounded by a lipid membrane, which means that influenza belongs to the “enveloped” group of viruses. Eight separate pieces of ribonucleic acid (RNA) make up the influenza virus genome and each piece of RNA specifies the amino acid sequence of one and sometimes two of the virus’s proteins. The segmented nature of the RNA allows differenet flu viruses to easily “mate” with each other to form hybrid progeny viruses with bits of RNA from each parent virus.Two glycoprotein molecules, known as hemagglutinin (HA) and neuraminidase (NA) (Fig 5) are stuck onto the lipid envelope of the virus and both play a crucial role in the infection of the epithelial cells of the upper respiratory tract. HA is a rod-shaped triangular molecule.and NA exists as a mushroom shaped spike with a box-like head on top of a long stalk, containing a hydrophobic region by which it is embedded in the viral membrane..

Fig 5: The Neuraminidase enzyme

The enzyme Neuraminidase, also known as sialidase, is a tetramer with C-4 symmetry and an approximate molecular weight of 250 000. It contains a symmetrical folding pattern of six four-stranded antiparallel �-sheets arranged like propeller blades. Nine types of neuraminidase have been identified for influenza A and only one subtype for influenza B, and only 30% of the overall amino acid sequence is conserved between all known types of neuraminidase8  – these are the amino acids which line and surround the walls of the binding pocket. If they mutate, the enzyme is inactivated, so the virus could not mutate to escape from a drug which interfered with this site. So neuraminidase offers an attractive site for therapeutic intervention in influenza infections.

 

How the influenza virus works:The influenza virus (like all viruses) can only replicate after invading selected living cells and growing inside them. It makes thousands of new virus particles from the cellular machinery and then goes on to infect other cells.. Hemagglutinin allows the virus to infect the epithelial cells of the upper respiratory tract by attaching it to cells through receptors on the cell containing sialic acid, it fuses the cell membrane with the membrane of the virus, allowing the RNA of the virus to get inside the cell and thus instruct the cell to make thousands of new virus particles. After this viral replication, the progeny virions must be released from the cell to repeat the cell cycle of infection.Neuraminidase removes the sialic acid receptors from the host cell and other newly made virus particles by cleavage of -glycosidic bonds. This enables the virus to escape from the cell in which it grew and spread in the body to infect other cells. The action of NA may also facilitate viral mobility through the mucus of the respiratory tract. virusattack.gif (46720 bytes)

Fig. 6: The life cycle of the influenza virus. Click once on this image to see a larger version

The life cycle of the influenza virusG begins with the individual virus entering the cell lining of the respiratory tract (letter a in Fig. 6), and the cell being induced to take up the virus because hemagglutinin on the virus binds to the sialic acid (b and c in Fig 6). The virus then dispatches its genetic material (made up of RNA) and its internal proteins to the nucleus of the cell (e and f). Messenger RNA is produced when some of the internal proteins duplicate the RNA (f). This messenger RNA is used by the cell as a template for making viral proteins (g and h) and genes which become new viral particles and leave the cell covered in sialic acid. This sialic acid needs to be removed so that the hemagglutinin molecules on one particle don’t attach to the sialic acid on other ones, thus causing the new viruses to clump together and stick to the cell. The sialic acid is removed from the surface of the new viral particle by neuraminidase (j) and the new viral particles are able to travel and invade other cells (k).

How Relenza works:

Relenza adopts a position within the active site of the enzyme and copies the geometry of the sialoside hydrolysis transition state9. It can achieve very good binding through appropriate presentation of its four pendent substituents and contains a hydrogen bonding glycerol sidechain. The guanidino group in Relenza is believed to form salt bridges with Glu 119 in the neuraminidase active site and add a strong charge interaction with Glu 2278.

Two hydroxyl groups of the 6-glycerol side chain are hydrogen bonded to Glu276 and the 4-hydroxyl is oriented towards Glu119. The NH group of the 5-N acetyl side chain interacts with a bound water molecule on the floor of the active site. The carbonyl oxygen of the same side chain is hydrogen bonded to Arg152 and the methyl group enters a hydrophobic pocket lined by Ile222 and Trp178. The glycosidic oxygen projects into bulk solvent.

Fig 7. Relenza bound to neuraminidase

The binding involved in Fig 7 is shown more clearly in Fig 8 below. Neuraminidase can no longer remove the sialic acid receptors from the host cell and newly made virus particles because of this binding. Therefore the virsuse ‘clump’ together or to the host cell and cannot go on to effect new cells.

 

Fig 8: Depiction of interaction of Relenza (GG 167) in the neuraminidase binding site6

References

1): K. J. Lui and A. P. Kendal, Am. J. Public Health, 1987, 77, 712
2): Scheiget, Zambonis, Bernstein and Roy, Org. Prep. Proced. Int., 1995, 27, 637- 644
3): Glaxo Wellcome Inc. Relenza� (zanamivir for inhalation) [package insert]. Research Triangle Park, NC: Glaxo Wellcome, Inc., 1999
4): N Seppa, Scientific American, July 10th 1999, Volume 156
5): L. Gubareva, Lancet, March 4th 2000, 355: 827-35
6): J. Medicinal Chemistry. 1999, 42, 2332-2343
7):P Smith, S Sollis, P Howes, P Cherry, I Starkey, K Cobley, H Weston, J Scicinski, A Merritt, A Whittington, P Wyatt, N Taylor, D Green, R Bethall, S Madar, R Fenton, P Morley, T Pateman, A Beresford. A. J. Med. Chem, 41, 1998, 787-797
8): C Kim, W Lew, M Williams, H Liu, L Zhang, S Swaminathan, N Bischofberger, M Chen, D Mendel, C Tai, G Laver, R Stevens, J Am Chem Soc, 1997119, 681-690
9): P Smith, J Robinson, D Evans, S Sollis, P Howes, N Trivedi and R Bethell, Bioorganic and Medicinal Chemistry Letters 9, 1999, 601-604
10): A. J. Hay, A. J. Wolstenholme, J. J. Skehel and M. H. Smith. EMBO J,. 1985, 4, 3021: L. J. Holsinger and R. A. Lamb, Cell, 1992, 69, 517
11): J. C. Stoof, J. Booij, B. Drukarch and E. C. Wolters, Eur. J. Pharmacol., 1992, 213, 439
12):  W. Graeme Laver, Norbert Bischofberger, and Robert G. Webster, Perspectives in Biology and Medicine 43.2 (2000) 173-192. This can be seen by visitinghttp://www.press.jhu.edu/journals/perspectives_in_biology_and_medicine/v043/43.2laver.html   nmr
13): M. Chandler, M. J. Bamford, R. Conroy, B. Lamont, B. Patel, V. K. Patel, I. P. Steeples, R. Storer, N. G. Weir, M. Wright, C. Williamson, J. Chem. Soc. Perkin Trans. 1, 1995, 1173- 1180    nmr  synth
14): A. E. Miller, J. J. Bischoff, Synthesis, 1986, 777- 779
15): G. D. Allena, S. T. Brookesa, A. Barrow, b, J. A. Dunnc and C. M. Grossec, Journal of Chromatography B: 1999, 732, 383-393 
Share

The Magic of Cubane!

 PROCESS, Uncategorized  Comments Off on The Magic of Cubane!
Feb 012014
 

 

File:Cuban.svgCubane[1]
Pentacyclo[4.2.0.02,5.03,8.04,7]octane
CAS 277-10-1

Cubane (C8H8) is a synthetic hydrocarbon molecule that consists of eight carbon atoms arranged at the corners of a cube, with one hydrogen atom attached to each carbon atom. A solid crystalline substance, cubane is one of the Platonic hydrocarbons. It was first synthesized in 1964 by Philip Eaton, a professor of chemistry at the University of Chicago.[2] Before Eaton and Cole’s work, researchers believed that cubic carbon-based molecules could not exist, because the unusually sharp 90-degree bonding angle of the carbon atoms was expected to be too highly strained, and hence unstable. Once formed, cubane is quite kinetically stable, due to a lack of readily available decomposition paths.

The other Platonic hydrocarbons are dodecahedrane and tetrahedrane.

Cubane and its derivative compounds have many important properties. The 90-degree bonding angle of the carbon atoms in cubane means that the bonds are highly strained. Therefore, cubane compounds are highly reactive, which in principle may make them useful as high-density, high-energyfuels and explosives (for example, octanitrocubane and heptanitrocubane).

Cubane also has the highest density of any hydrocarbon, further contributing to its ability to store large amounts of energy, which would reduce the size and weight of fuel tanks in aircraft and especially rocket boosters. Researchers are looking into using cubane and similar cubic molecules inmedicine and nanotechnology.

Synthesis

The original 1964 cubane organic synthesis is a classic and starts from 2-cyclopentenone (compound 1.1 in scheme 1):[2][3]

Scheme 1. Synthesis of cubane precursor bromocyclopentadienone

Reaction with N-bromosuccinimide in carbon tetrachloride places an allylic bromine atom in 1.2 and further bromination with bromine in pentane –methylene chloride gives the tribromide 1.3. Two equivalents of hydrogen bromide are eliminated from this compound with diethylamine in diethyl ether to bromocyclopentadienone 1.4

Scheme 2. Synthesis of cubane 1964

In the second part (scheme 2), the spontaneous Diels-Alder dimerization of 2.1 to 2.2 is analogous to the dimerization of cyclopentadiene to dicyclopentadiene. For the next steps to succeed, only the endo isomer should form; this happens because the bromine atoms, on their approach, take up positions as far away from each other, and from the carbonyl group, as possible. In this way the like-dipole interactions are minimized in the transition state for this reaction step. Both carbonyl groups are protected as acetals with ethylene glycol and p-toluenesulfonic acid inbenzene; one acetal is then selectively deprotected with aqueous hydrochloric acid to 2.3

In the next step, the endo isomer 2.3 (with both alkene groups in close proximity) forms the cage-like isomer 2.4 in a photochemical [2+2] cycloaddition. The bromoketone group is converted to ring-contracted carboxylic acid 2.5 in a Favorskii rearrangement with potassium hydroxide. Next, the thermal decarboxylation takes place through the acid chloride (with thionyl chloride) and thetert-butyl perester 2.6 (with t-butyl hydroperoxide and pyridine) to 2.7; afterward, the acetal is once more removed in 2.8. A second Favorskii rearrangement gives 2.9, and finally another decarboxylation gives 2.10 and 2.11.

The cube motif occurs outside of the area of organic chemistry. Prevalent non-organic cubes are the [Fe4-S4] clusters found pervasively iron-sulfur proteins. Such species contain sulfur and Fe at alternating corners. Alternatively such inorganic cube clusters can often be viewed as interpenetrated S4 and Fe4 tetrahedra. Many organometallic compounds adopt cube structures, examples being (CpFe)4(CO)4, (Cp*Ru)4Cl4, (Ph3PAg)4I4, and (CH3Li)4.

 

It was mentioned previously that cubane was first prepared in 1964 by Dr. Philip E. Eaton. He was partnered by Thomas W. Cole and together they successfully completed the first synthesis, shown schematically below:

N-bromosuccinimide acts as the reagent for a radical mediated allylic bromination reaction which is carried out in tetrachloromethane with heat as the initiatorBromine is added......and 2 moles of HBr are eliminated......and 2 moles of HBr are eliminated...

Reactive enough to undergo dimerisation via a [4+2] cycloaddition reaction to give the ENDO cycloadductThe more reactive, bridgehead ketone group is protected by Ketal formation.Photochemical energy is required to promote the [2+2] intramolecular cycloaddition reaction.

The acid mediated oxidation of the ktone group to a carboxylic acid.

 

 

The first occurance of a Hunsdiecker decarboxylation, firstly substitutes the caroxylic acid group and then removes it.The first occurance of a Hunsdiecker decarboxylation, firstly substitutes the caroxylic acid group and then removes it.

 

Acid hydrolysis releases the protected ketoneThe second instance of a Hunsdiecker decarboxylation.

 

The second instance of a Hunsdiecker decarboxylation.The second instance of a Hunsdiecker decarboxylation.

Decarboxylation via thermal degradation of di-t-butyl perester

 

This, however, was soon simplified by N.B.Chapman who condensed the process to give cubane-1,4-dicarboxylic acid in five steps and so cubane in six:

n 1966 J C Barborak et al discovered yet another new synthesis of cubane. It was slightly unconventional in the fact that it utilised cyclobutadiene as a key substance to the process. Before this,cyclobutadiene was usually unavailable for the purposes of organic chemistry due to it’s instability. The shorter synthesis is shown below:

Decomposition in presences of 2,5-dibromobenzoquinone gives......the endo adduct.

 

Irradiation, in benzene, with a mercury lamp initiates the intramolecular [2+2] cycloaddition reaction.

Treatment with KOH at 100 ºC gives the cubane-1,3-dicarboxylic acid

Decarboxylation via thermal degradation of di-t-butyl perester

Since the synthesis of the cubane-1,4-dicarboxylic acid has become shorter and easier, a new decarboxylation method has also devised to give increased yields of the final cubane product. This has allowed the scale of production reach multikilogram batches in places (Fluorochem in California and EniChem Synthesis in Milan) eventhough cubane and its derivatives remain expensive to purchase.

Cuneane may be produced from cubane by a metal-ion-catalyzed σ-bond rearrangement.[4][5]

Cubane is a unique molecule for its extraordinary C8 cage, very high symmetry,exceptional strain and unusual kinetic stability. The particular appeal of cubane,referred to as a landmark in the world of impossible compounds, stems from therehybridization of the carbon atoms away from the canonical sp3 configuration,that is required to bound together eight CH units in a cubic framework.There is now a revival of interest on the chemistry of cubane and its functionalized derivatives,triggered by potential applications as high-energy fuels, explosives and propellantsand intermediates in pharmaceuticalpreparations.Let us now discover the synthesis and properties of this landmark molecule of impossible chemistry
Cubanehas the highest strain energy (166kcal/mol) of any organiccompounds available in multi gram amount. It is a kineticallystable compound and only decomposite above 220 Celsius Degree.It is also one of the most dense hydrocarbons ever know.However, although many physical properties of cubane have been measured, in1980 and before, cubane was considered just a laboratory curiosity of interest only to academics.It changed, in early 1980s when Gilbert of U.S ArmyArmament and Development Command (now ARDEC) pointed out that cubane’svery high heat of formation and its exceptionally high density could make certain cubanederivatives important explosives.The effectiveness of an explosive is dependent on the energentics of the decomposition reaction,the number of moles and molecular weight of the gaseous products and also the density.

The more mols of of an explosive that can be packed into the limited volume the better. .

Highly nitrated cubanes can be predicted to be very dense and very powerful explosives.

Octanitrocubane is calculated to be 15~30%more powerful than HMX.

 

Cubane, which CA index name is Pentacyclo[4.2.0.02,5.03,8.04,7]octane (7CI,8CI,9CI),has exceptional structure, strain and symmetry and it is a benchmark in organic chemistry.It has been studied extensively and much of its properties has been published.Some of the physical properties are given at right hand table.

The C-C bond length is a bit longer than obtained in the original X-ray structure determination by

Fleischer in 1964. There is not much difference between this bond length and the

C-C bond length in a simple cyclobutane.

 

SYNTHESIS

The cubane system was first synthesized over 35 years ago by Philip Eaton and Tom Cole.
It is a highly symmetric cubic cage structure having carbon atoms at the vertices of a cube.
The synthesis needs to go through brombromocyclopentadienone
dimer I and cubane-1,4,dicarboxylic acid. It is a marvel scheme of economy and simplicity.
With only minor modification, this procedure remains to this day the best available

method for large-scale synthesis of cubane-1,4,dicarboxylic acid.

 

 

 

The stereospecific in situ [4 + 2] (Diels-Alder) cyclodimerization of 4-bromocyclo-pentadienone
is the key in this kinetically controlled synthesis. However, it is still a tricky matter
and a few years later after this synthesis is published, N.B.Chapman et al in England following up
this work and improved this synthesis.

Why cubane is stable?

The reason for this, unappreciated at the time of the early predictions of instability,

 is that there are no kinetically viable paths along which cubane can rearrange thermally.

 On one hand, orbital symmetry considerations raise the energy of concerted two-bond ring

opening reactions. On the

other, there is little to be gained by breaking just one bond as there is concomitantly

only a small change in geometry, and the resulting biradical is still very strained.

Functional group transformation

Functional groups on the cubane system generally behaves very well.Functional group transformation can be applied successfully.For example, the preparation of 1,4-dinitrocubane from cubane-1,4-dicarboxylic acid.(The mechanism is provided on the right hand side.) Classical methodology is used here.

Substitution on the cubane framework is fairly easy done by the cubyl radical.
However, the problem is such that a mixture of products are obtained.
Thus, to achieve controlled substitution on the cubane framework,
we need to carefully study the chemistry of the cubane system.

 

The improvement in synthesis of

cubane-1,4-dicarboxylic acid

 

 

This is the improved synthesis by N.B Chapman et al in England.

 Basically the improvement is such that the

2-bromocyclopentadienone could be made easily and undergoes spontaneous dimerization.

The rest of the reaction is the same as the original one.

 

This synthesis now is scaled up and is conducted in small pilot plants by

Flurochem in California and EniChem Synthesis in Milan.

This method is much more superior than the old method. It is introduced by

Derek Barton et al and use the radical-induced decomposition of diester which can be

prepared easily from cubane-1,4-dicarboxylic acid.

IMPROVEMENT

This method is much more superior than the old method. It is introduced by

Derek Barton et al and use the radical-induced decomposition of diester which can be

prepared easily from cubane-1,4-dicarboxylic acid

 

IR

 

Cubane is a colorless solid. It melts at 130- 131°C, and decomposes above the melting point.

 It is soluble in CS2, CC14, CHC13, and benzene.

Spectra were obtained from 400 to 3600 cm-l with a Beckman IR-12 spectrophotometer.

The lower limit was set by KBr cell windows. In addition a thick deposit of do was

 measured down to 200 cm-lin a Csl cell. Since no infrared bands were found, the range

200-400 cm-l was not examined for the other compounds.

The spectral slit widths were 1-2 cm-l in all cases.

In the infrared spectrum, there are only noticeable absorptions in the region from

 4000 to 660 cm-1appear at 300,1231, and 851 cm-1.

Generally, for single-line proton magnetic resonance spectrum, the one

and only absorption appears at chemical shift=6.0ppm.

Originally there was doubt whether cubane does exist.

The geometry at each carbon atom is far from tetrahedral.

Only later, we found out that there is no kinetically viable paths exist for

the thermal rearrangement of cubane.

At same time, orbital symmetry considerations shows that

the energy of concerted two-bond ring-opening reactions is very high.

There will be very little gain in energy by breaking just one bond, as the

concomitant change in geometry is small, and the resulting biradical is still very strained

In 1964 Fleischer showed that cubane forms a stable solid at room temperature with a

crystalline structure composed of cubane molecules occupying corners of the rhombohedral

primitive unit cell (space group R3). The cubic molecular geometry gives the solid many unusual

electronic,structural, and dynamical properties compared to the other hydrocarbons.

For example, solid cubane has a relatively high melting point temperature about 405 K! and a

very high frequency for the lowest-lying intramolecular vibrational

mode (617 cm-1). Recent work related to cubane has focused on solid cubane and cubane based

derivatives.Because of relatively weak intermolecular interaction the cohesive energy relative

to the constituent C8H8 is expected to be small, and most of the physical properties of

solid cubane are dominated by the properties of the C8H8molecule.

Pharmaceutical aspect of cubane

Because the cubane frame is rigid, substituent have precise spatial relationships to each another.

The distance across the cubane (the body diagonal) is almost the same as that between the para

positions of the benzene ring. On cubane, on can add substituents in the “benzene plane”, as

well as above and below it, so to speak. This offers fascinating position possibilities for

the synthesis of new pharmaceuticals. A number of cubane derivatives have already

been obtained which shows interesting activity in anti-AIDS and anti-tumor screens.

Although the activity or the toxicity balance of cubane is yet not know, the cubane

system is not inherently toxic. Most of cubanes are biologically innocuous.

The research of cubane pharmaceutical has just began. At least now,

cubane is a biologically stable, lipophilic platform on which the chemist

can install a wide range of substituents in a variety of well defined special relationships.

Developments in drug design programs should allow the judicious choice.

 

Dipivaloylcubane: a cubane derivatized with keto, cyano, and amide groups,

shown on the left- exhibits moderate activity against human immunodeficiency virus (HIV),

which causes AIDS, without impairing healthy cells.

Polymers of cubane:

Optically transparent cubanes and cubylcubanes have been proposed as building

blocks for rigid liquid-crystal compounds. UV active cubanes, for example cubyl ketones,

are readily transformed photochemically into coloured cyclooctatetraenes;this transformation

can be used to permanent information storage.

Another example of UV active cubane, which can be used to synthesis liquid crystals.

Polymers with cubane in the backbone or as a pendant group along a polymer chain is

focused now.

The cubane subunits in these polymers can be rearranged easily to cycloctatetraenes.

It is expected that polycyclooctatetra can be converted in to polyacetylenes by

the way of ring-opening metathesis polymerization. The polyacetylenes will have properties

which are enhanced by the chain being intrinsically part of another polymer.

These properties including stability and extrudability and etc. A example is shown below:

 

Cubane derivative could be the structural basis for a class of intrinsic small gap polymers.The small gap polymer could present intrinsic good conductivity without doping,good nonlinear optical and photoelectric properties.Investigation of oligamers with up to six units of a conjugated unsaturated cubane derivative,where all the hydrogen were removed, is carried out.The table below shows that the gap values in eV by EHT and PM3.These values suggest to us that these structures could be used to design a newclass of polymers with very small gap.

Explosive and fuels:

In the early 1980s Everett Gilbert of the U.S. Army Armament Research and Development

Command (now ARDEC) pointed out that the nitrocarbon octanitrocubane (ONC),

then unknown, has a perfect oxygen balance, and in light of the properties of the

parent hydrocarbon cubane should have a very high heat of formation per CNO2 unit

and an exceptionally high density as well. His colleagues Jack Alster, Oscar Sandus

and Norman Slagg at ARDEC provided theoretical support for Gilbert’s

brilliant insight and estimated that ONC would have a detonation pressure

significantly greater than HMX. Later, both statistical and computational

approaches predicted a density of 2.1 ± 2.2 g /cm3 for octanitrocubane,

greater than any other C, N, O compound.

Is Cubane a really good explosives?

Quantitative evaluation of the potential of a candidate explosive before synthesis is very difficult.

Currently, estimation of energetic properties relies on the empirically derived Kamlet and Jacobs

equations:

In these equations the heat released by the decomposition, the number of moles of gas produced,

and the molecular

weight of these gases are all critical factors. Density too is crucial.

Obviously, the more molecules of a high-energy material that can be packed into the limited

volume of a shell or rocket the better. Less obvious, but more important, density affects the

detonation velocity of an explosive.

This is a specialized “linear” rate of reaction that ranges from 5 to 10 km/s in

explosives and affects the maximum detonation pressure, a direct measure of the

power of an explosive. For a given explosive, the detonation pressure is proportional

to the square of its density, so great effort is made to obtain the highest density form

of any particular explosive.

Quantitative evaluation of the potential of a candidate explosive before synthesis is very difficult.

Currently, estimation of energetic properties relies on the empirically derived Kamlet and Jacobs

equations:

In these equations the heat released by the decomposition, the number of moles of gas produced,

and the molecular

weight of these gases are all critical factors. Density too is crucial.

Obviously, the more molecules of a high-energy material that can be packed into the limited

volume of a shell or rocket the better. Less obvious, but more important, density affects the

detonation velocity of an explosive.

This is a specialized “linear” rate of reaction that ranges from 5 to 10 km/s in

explosives and affects the maximum detonation pressure, a direct measure of the

power of an explosive. For a given explosive, the detonation pressure is proportional

to the square of its density, so great effort is made to obtain the highest density form

of any particular explosive.

Numerous nitro compounds are employed commonly as military and commercial explosives.

There is a continuing search for more powerful and less shock-sensitive examples.

Such materials are also sought as potentially useful fuels and propellants.

Most interest is focused on high-density organic compounds that contain all of the

elements needed for combustion to gaseous products in the absence of air.

Nitrocubanes carrying five or more nitro groups contain enough oxygen to oxidize

all constituent carbon and hydrogen atoms to gaseous CO, CO2, or H2O.

Each of these, along with N2, “explodes” from the solid to 12 gaseous molecules.

The expansion from the dense solid to a lot of gas (much expanded by the released heat)

produces the desired effect in propellants and explosives. ONC has a “perfect”

oxygen balance and would produce (were the detonation completely efficient)

eight molecules of carbon dioxide and four of dinitrogen. As ONC has no

hydrogen, no water forms when it burns; when used as propellants such zero-hydrogen

compounds leave little or no visible smoke (steam) in the plume behind the rocket;

such “low-signature” rockets are difficult to track.

On application of the Kamlet and Jacobs equations led ARDEC to predict that

octanitrocubane would be a very much better explosive (Table 1) than the classic

C-nitro compound trinitrotoluene (TNT), perhaps 15±30% better than the nitramine

HMX (the most powerful, commonly used military explosive), and at least competitive

with (and perhaps less shock-sensitive than) the newest experimental explosive CL-20

 

 

SYNTHESIS:(1)

The high strain that the cubane framework is under has already been highlighted. The researchers had to very cautiously attach a nitro group to each of the corners of the cube in order to make the desired product. The insertion of the first four nitro groups could be done by manipulating functional groups:

The key intermediate, cubane-1,3,5,7- tetracarboxylic acid (TNC), was obtained by clever application of the Brown-Kharasch photochlorocarbonylation to cubane mono-acid.

The addition of four further nitro groups proved far more difficult and new methodologies had to be developed, specifically the process of interfacial nitration. This method was used successfully to convert the sodium salt of TNC to pentanitrocubane (PNC) and then hexanitrocubane (HNC), both are stable materials.

Interfacial nitration, however, proved deficient for further nitration of HNC and again new experimental methodology had to be developed for its successful conversion to heptanitrocubane (HpNC):

Addition of excess NOCl to a solution of the lithium salt of HpNC in dichloromethane at -78°C gave the long-sought ONC:

 DIFF TYPES

For the last planned post in my Unnatural Products series, I’m going to write about Eaton’s 1981 synthesis of pentaprismane.[A] At the time, unnatural hydrocarbons were hot targets, and as the next largest prismane on the list this target was the subject of much research by groups around the world. Perhaps Eaton’s biggest rivals were the groups of Paquette and Petit, and in fact all three had, at various times, synthesised hypostrophene as an intended precursor to the target.

Unfortunately, the ‘obvious’ [2 + 2] disconnection from pentaprismane turned out to be a dead end and the photochemical ring closure was unsuccessful. The 1970s and early 1980s saw the publication of a number of other similarly creative, but sadly ill-fated, approaches based on various ring contractions, and the compound gained a well-earned reputation for extraordinary synthetic inaccessibility.

Eaton’s route began, as with the cubane and dodecahedrane syntheses previously covered in this series, with a Diels-Alder reaction. The diene used was the known tetrachlorocyclopentadienone acetal shown that upon heating neat with benzoquinone produced the endo adduct shown in excellent yield. Next, an even higher yielding photochemical [2 + 2] reaction was used to close the cage-like structure by cyclobutane formation. Treatment with lithium in liquid ammonia simultaneously reduced both ketones and removed all four chlorine atoms. The resulting diol was converted to the ditosylate, which, under carefully controlled conditions with sodium iodide in HMPA, underwent a mono-Finkelstein reaction to give the iodotosylate shown. When this was treated with t-BuLi halogen-lithium exchange, followed by an extraordinary fragmentation, gave a diene reminiscent of hypostrophene shown above. However, the extra carbon atom in the skeleton made all the difference, and unlike the parent compound, this did undergo a [2 + 2] cycloaddition when exposed to UV light. Finally, acetal hydrolysis gave homopentaprismane in 34% yield from benzoquinone, putting the group a single ring contraction from victory.[B]

With significant amounts of homopentaprismanone in hand, the group now intended to employ the transformation that had been the cornerstone of their cubane synthesis – the Favorskii rearrangement. Unfortunately, this required the introduction of a leaving group in the ketone α-position, a transformation made incredibly difficult due to the strained system and Bredt’s rule, which prevented enolisation.[C] Eventually a six-step sequence (!) to introduce a tosyloxy group was devised, beginning with a Baeyer-Villiger reaction using m-CPBA. A remarkable CH oxidation with RuO4, generated in situ, then gave the hydroxylactone. Treatment of this with diazomethane gave the corresponding δ-ketoester in almost quantitative yield. The group then reformed the starting norbornane-like bridge through use of an unusual acyloin type reaction effected by treatment with sodium in liquid ammonia. Finally, oxidation of the secondary alcohol and tosylation gave the Favorskii precursor, apparently preparable in muti-gram quantities.

Treatment with aqueous potassium hydroxide solution effected Favorskii rearrangement in excellent yield, especially considering that this was the first time the elusive pentaprimane ring system had been prepared. Finally, Eaton used the three-step decarboxylation he had developed for cubane to remove the extraneous acid and give pentaprismane in 18 steps. Awesome.[D]

References and suchlike

  1. A    J. Am. Chem. Soc., 1981, 103, 2134. Much like Eaton’s seminal cubane paper, the title is a single word, ‘Pentaprismane’. I love the lack of hype.
  2.  B   Although Petit had prepared this compound a full decade earlier, his approach relied on a cycloaddition of the difficult to prepare cyclobutadieneiron tricarbonyl with the acetal of tropone, and proved difficult to scale  up. In fact, in his own paper Eaton rather directly described it as ‘conceptually fascinating [but] useless synthetically’.
  3. C   Eaton uses the phrase ‘invasion at the bridgehead’, which I find delightfully evocative. Makes it sound like a second world war campaign. Apparently the group initially planned, in spite of Bredt’s rule, to deprotonate the bridgehead position, relying on inductive stabilisation of the anion rather than enolate formation, but were unable to do so.
  4. D  Pentaprismane is the most recent of three prismanes synthesised to date, the other two being cubane, and triprismane. Although I think triprismane looks quite silly, it was actually synthesised some 8 years previouslyby T. J. Katz in far fewer steps. Go figure.

 

The Amide Activating Group

 

The very first step of cubane frame substitution will be the activation of the cubane frame.

This can be done by amides. The idea is derived from the similarities between cubane and arenes.

 Both of them have C-H bonds with enhanced s character ( see structure),

 and in both the adjacent (ortho) substituents are forced to be coplanar.

A more specific example is the cubane-N,N-diisopropyl carboxamide

 reacts with excess lithium tetramethylpiperidide (LiTMP) in THF solvent.

About 3% of the deuteriation products obtained.

The diisopropyl amide activating group is used because it is inert to the amide

bases employed for ortho metalation. Although there is a problem, there is

 difficulty in hydrolyzed it the corresponding carboxylic acid.

The problem is finally solved by using borane reduction followed by the oxidation

 of the amine so produced with dimethyldioxirane or potassium permanganate (in large scale).

Transmetalation is the basis of a complete synthetic methodology for the preparation

of a great variety of the substituted cubanes.

In order to make the substitution productively, a way must be found to

make use of the small amount of anion in the equilibrium with the starting material.

Mercury salt is used here as an effective anion trap and very little starting material remain unreacted.

The mercury for lithium transmetalation resulted in nearly complete conversion of the

starting material by drawing the lithiation equilibrium to the right.

 

The amide group is important in stabilizing the intermediate lithiated cubane,

but not the mercuriated compound. Once the lithium is replaced by mercury,

 the amide group is again able to assist removal of another ortho-hydrogen atom.

In the end, the complex ortho-mercurated product mixture obtained was

 simplified by treatment with elemental iodine.

The iodine cleavage of the carbon-mercury bonds 2-iodo and 2,6-diiodo derivatives

of the starting amide in72% and 15% respectively

Cubyl Grignard Reagents

From transmetalation, a reverse transmetalation was also developed, which is basically adding Grignard reagent to the mercuriated cubane instead of the iodine. However, these processes have a great main disadvantage, the mercury is highly toxic. Thus, scale up of this method was limited.

In 1988, Bashir-Hashemi introduced transmetaltion with magnesium salts and thereby provided easy access to cubyl mono-and bis-Grignard reagents. It is a reaction of cubane diamide with an excess of LiTMP/MgBRin THF and quenching with I2 gave diiodocubane diamide of 72% yield.

The effect of the presence of electron withdrawing group     –  Cyanide

When electron withdrawing group such as cynate present, they stabilize both intermediate lithiated cubanes very well. As a result, only a small amount of LiTMP is need to achieve fairly complete deprotonation even at -78°C.

The inductive effect of the cyano group clearly enhances the reaction. However, the adding of cyano groups results in competitive lithiation and a mixture of products. However, this problem can be well trackled by adding MgBr2.The product ratio was improved to 9:1 favoring carboxyliation ortho to the amide function.

A mixture of product formed.

Increased selectivity by adding MgBr2

Since the reactivity of cubane metalation is enhanced greatly with presence of cyano groups, it is possible to substitute all three positions ortho to the amide in a simple reaction. For instance, 4-cyanocubanamide can be converted directly into the tri(tert-butylcarbonyl)derivative as shown below.

Through Baeyer-Villiger oxidation, ter-butyl cuybl ketones can be converted easily to the polycarboxyliated cubane.

PHENYL CUBANE

 

From the basis idea of cubyl Grignard Reagent, phenyl cubane can be synthesised. The reaction of cubane diamide with 10 equiv of LiTMP and 4.0 equivalents of MgBr2 etherate in THF at 0°C followed by the addition of 10.0 equiv of bromobenzene, gave diphenylcubane diamide in 53% yield.

The mechanism is shown below:

 

The benzyne intermediate was formed in situ from the reaction of excess of LiTMP with bromobenzene. For a similar reaction, MeMgBr is used and give 30% yield of bromo-phenylcubane diamide, the first cubane derivative containing 3 different substituents.

Now, let us look the main concern of the cubane derivatives–the nitrocubanes.

Nitrocubanes are sought to be powerful, shock-insensitive, high-density explosives. They are stable compounds with decomposition points above 200°C. Simple nitrocubane can be made from simple oxidation of amines( See Functional Group Transformation.)

If we want to add more nitro groups into the cubane nucleus, we cannot do it though transmetalation because there is unstoppable cage cleavage reactions when make adjacent nitro groups. The ab initio calculation has confirmed this destabilising effect.

We are going to discuss how to make more and more substituted nitrocubane until octanitrocubane(ONC), the ultimate power house, is synthesised.

 

1,3,5 trinitrocubane and 1,3,5,7 tetranitrocubane(TNC)

As we mention early, addition of nitro groups cannot be done through direct transmetalation. Thus, we need found some indirect route.

This is done by introducing a substituent on each of 3 ortho carbons and remove the ortho-activating group in the end.

By adding a electron-withdrawing group such as a cyano group will help the case here. This choice of original substituent is important here and when cyano group is chosen, it activates the cubane nucleus without affecting the ortho directing by the diamide (for details please refer to electron-withdrawing group-cyanite).

When the dicyano amide was treated with TMPMgBr in THF and quenched with CO2. The ortho (to amide) carboxylic acid was the only product.

Even when the much activated tricyanoamide is treated with TMPMgBr and CO2 ,again, the ortho position ( to amide) carboxylic acid was formed.

The removal of the carboxamido group is done through a smart yet tedious process. The cyano group is converted to acid group first. Then, it is reduced to alcohol by lithium aluminium hydride. At same time, the carboxamido is reduced to aminotetrol. The alcohols are protected as acetates and amino tetrol is converted to carboxylic acid. The carboxylic is then removed through Barton Decarboxylatio. A detail mechanism is provided below.

The cubane-1,3,5,7-teracarboxylic acid is converted to TNC on the mechanism as follow:

The whole process is very clever, but it is very long. Thus, in 1997, a improved synthesis method for TNC was proposed by making use of the photochemsitry.

Improved synthesis for TNC

In 1993, Bashir-Hashemi showed the cubane-1,3,5,7-tetracarboxylic acid chloride can be formed by applying photochemically induced chlorocarbonyl cation( the Kharasch_Brown Reaction).

For a fast reaction, a high power Hanovia of 450 watts, medium pressure Hg was used. The favoured products are cubane tetraacid chloride shown on the right hand side. The first one, cubane-1,3,5,7-tetracarboxylic acid, made up 30% overall. This reaction conveniently prepare us the important versatile intermediate .

A detail conversion process is provided below:

 

A catalyst TMSN3 is used in converting tetraacid chloride to tetracylazide. The rest is the same as the orginal reaction.

TNC is a thermodynamic powerhouse but remarkly stable kinetically. Figure 1 shows that rapid thermal decomposition doesnot start until over 250°C.

The literature was unsupportive of this optimistic view. Poor results were also obtained initially with nitrating agent such as NO2BF4, acetyl nitrate, amyl nitrate etc.

Tetranitrocubylsodium can be formed directly on treatment of TNC with sodium bis(trimethylsilyl) amide in THF at -75°C. It can react with electrophiles to provide a useful and convenient way to achieve further functionalization of cubane nucleus.

More substituted nitrocubanes-

Pentanitrocubane(PNC) and Hexanitrocubane(HNC)

PNC

Base on the property of tetranitrocubylsodium, nitryl chloride(NO2Cl) was used to further nitrate the cubane nucleus. Treatment of NO2Cl with tetranitrocubylsodium in THF at -75°C works out 10-15% yield of pentanitrocubane(PNC). The yield increased to 30% when the solution was frozen to-180°C and allowed to warm slowly. This is called the interfacial nitration process. It is suggested that NO2Cl oxidized tetranitrocubylsodium to a radical, which made the whole reaction worked.

Base on the property of NO2Cl , N2O4 should be a better choice. The results showed that it is actually a better with 60:40 PNC to TNC ratio. The reaction is extremely clean.

PNC is colourless and highly crystalline. It is the first nitrated cubane to contain adjacent nitro groups. It behaves just TNC and other nitrocubanes, remarkly stable kinetically.

HNC

Although HNC can be prepared the same way as PNC, but the separation between PNC and HNC is extremely difficult.

However, if TIPS-substituted PNC( by N2O4 nitration from TIPS-sub TNC) react with potassium base (K(TMSN)2and the nitration with N2O4 gave a mixture of (triisopropyl) HNC and PNC in 60:40 ratio. This step is important and crucial. The separation is now possible by column chromatography on silica gel. 30% isolated yield of PURE HNC could be obtained when further treated with SiO2.

Synthesis for the last two nitro cubanes- heptanitrocubane and octanitrocubane

Interfacial nitration is not sufficient to further nitration for heptanitrocubane. Al though it is very good in deed, we need to find something which can successfully convert heptanitrocubane (HpNC).

HpNC

In this procedure TNC was treated with at least 4 equivalents of the base NaN(TMS)2 (where TMS = trimethylsilyl) at ±78 C in 1:1 THF/MeTHF. After the mono sodium salt had formed, the solution was cooled to between ±125 and ±130°C giving a clear, but very viscous fluid. This was stirred vigorously as excess N2O4 in cold isopentane was added. After one minute, the base was quenched, and the whole mixture was added to water. This resulted reproducibly in almost complete conversion of TNC (1 g scale) to HpNC (95% by NMR), isolated crystalline in 74% yield!

ONC

However, even in the presence of excess nitrating agent (N2O4 or many others) no indication
of any formation of ONC was ever seen. It is suspected that anion nitration with N2O4 proceeds by oxidation of the carbanion to the corresponding radical.Perhaps the anion of HpNC is too stabilized for this to occur. (HpNC is significantly ionized in neutral methanol.) This concept led to the use of the more powerful oxidant nitrosyl chloride. Addition of excess NOCl to a solution of the lithium salt of HpNC in dichloromethane at 78° C followed by ozonation at 78° C gave the long-sought ONC in 45±55% isolated yield on millimole scale. The intermediate product prior to oxidation is thought to be nitrosoheptanitrocubane.

Finally, the magic molecule, the so called the impossible molecule, octanitrocubane was synthesised. But, how good are they and how useful are they? Let us discuss about it in the following section.

Properties of nitrocubane:

Neither HpNC nor ONC is detonated by hammer blows!
Both have decomposition points well above 200 C. Octanitrocubane
sublimes unchanged at atmospheric pressure at 200 C. HpNC forms beautiful, colorless, solvent-free crystals when
its solution in fuming nitric acid is diluted with sulphuric acid. Single-
crystal X-ray analysis confirmed the assigned structure and
provided an accurate density at 21 C of 2.028 g cm±3, impressively
high for a C, H, N, O compound. Although octanitrocubane
catches the imagination with its symmetry, heptanitrocubane
currently is significantly easier to make than ONC. It is
denser, and it may be a more powerful, shock-insensitive explosive
than any now in use. According to page 41 of a 2004 IUPAC guide, cubane is the “preferred IUPAC name.”

  1.  ‘ ‘Cubaneand Thomas W. Cole. Philip E. Eaton and Thomas W. Cole J. Am. Chem. Soc.1964; 86(15) pp 3157 – 3158; doi:10.1021/ja01069a041.
  2.  The Cubane System Philip E. Eaton and Thomas W. Cole J. Am. Chem. Soc.1964; 86(5) pp 962 – 964; doi:10.1021/ja01059a072
  3.  Michael B. Smith, Jerry March, March’s Advanced Organic Chemistry, 5 th Ed., John Wiley & Sons, Inc., 2001, p. 1459. ISBN 0-471-58589-0
  4.  K. Kindler, K. Lührs, Chem. Ber., vol. 99, 1966, p. 227.

 

 

Dimethyl cubane-1,4-dicarboxylate

dimethyl 1,4-cubanedicarboxylate;

1,4-cubanedicarboxylic acid dimethyl*ester;

methyl 4-(methoxycarbonyl)pentacyclo[4.2.0.0<2,5>.0<3,8>.0<4,7>]octanecarboxylate

Pentacyclo(4.2.0.0(2,5).0(3,8).0(4,7))octane-1,4-dicarboxylic acid dimethyl ester

CAS 29412-62-2

Molecular Weight: 220.2213
Molecular Formula: C12H12O4
Density: 1.684g/cm3
Boiling Point(℃): 270°C at 760 mmHg
Flash Point(℃): 131.3°C
refractive_index: 1.704

An interesting   OPRD paper on the scale up of dimethyl cubane -1,4-dicarboxylate.

cubane

The work appeared in Organic Process Research and development, 2013, doi.org/10.1021/op400181g . It was carried out by an Australian group, John Tsanaktsidis, Michael Falkiner, Stuart Littler, Kenneth McRae and Paul Savage from CSIROand features a large-scale photochemical reaction which is very unusual to see in a scaled chemical process.

Extending their previous work from 1997, they scaled the following reaction.

photo

As is the norm with such reactions the reaction requires high dilution to be successful. In this case they used a tailor made photochemical  reactor. A solution of 1 in methanol/water was pumped through the reactor at 4 L/minute and the conversion of 1 to 2 was noted as 1g/4 minutes of irradiation.

This meant a total time of 173 hours. Further processing of 2 through the double Favourskii ring contraction required significant development but eventually delivered the di-sodium salt corresponding to the di-ester of cubane.

One needs to be careful with these cubanes as they are, due to the highly strained nature of the system quite energetic materials, the do-acid and ester being more stable than the parent hydrocarbon. However the energy released upon warming above the melting point is not insignificant.

This paper represents a good demonstration of the scale-up of several very difficult chemical reactions, including excellent descriptive paragraphs of the problems and solutions. They are to be congratulated on a very nice piece of  work.

See below
Abstract Image

A scalable process for the preparation of high purity dimethyl 1,4-cubanedicarboxylate (3) is reported.

The work described herein builds on previous synthetic work from this and other laboratories, to provide a reliable process that can be used to prepare multigram quantities of 3 in a partially telescoped, 8 step process, with minimal purification of intermediates.

CSIRO Materials Science & Engineering, Ian Wark Laboratory, Bayview Avenue, Clayton Victoria 3168,Australia
Org. Process Res. Dev., 2013, 17 (12), pp 1503–1509
DOI: 10.1021/op400181g
Publication Date (Web): November 8, 2013
Figure
Scheme 5. Pilot-Scale Synthesis of Dimethyl 1,4-Cubanedicarboxylate (3)
figure

Figure 1. Cubane nucleus.

Step 5

A dry 100 L glass reactor was charged with the crude diacid 2 (1287 g), dry methanol (36 L), and Dowex ion-exchange resin 50WX8–100 (176 g) that was prewashed with 1 L of methanol. This mixture was then stirred (150 rpm), and heated under reflux for 18 h under an atmosphere of nitrogen. The mixture was then cooled to room temperature and filtered to remove the resin. The methanol solution mixture was then evaporated to dryness using a rotary evaporator (45 °C at 45 mmHg) leaving behind the crude diester 3 (863 g) as a dark brown solid. Purification by sublimation (100–120 °C/0.01 mmHg), followed by recrystallization from acetonitrile furnished the diester 3 (560 g, 30%), as a colorless solid,
mp 164.5 °C (lit. 161–162 °C).(47)
 1H NMR δ: 3.7, s, 6H 4.24, s, 6H, ring protons.
13C NMR δ: 47.03, 51.55, 55.77, 171.89.

………………………………..

http://cst-www.nrl.navy.mil/lattice/struk/c8h8.html.

http://www.ch.ic.ac.uk/local/projects/b_muir/Enter.html.

http://www.sciencedirect.com.

http://www.sciencenews.org/.

http://www.winbmdo.com/.

Bashir-Hashemi, A., New developments in cubane chemistry: phenylcubanes.

J. Am. Chem. Soc.;1988;110(21);7234-7235, 110(21), 7234-7235.

D.S.Calvao, p. m. v. b. B. A. C. J. a., Theooretical Characterization of oligocubane.

Synthetic Metals 102 (1999) 1410.

E. W. Della, E. F. M., H. K. Patney,Gerald L. Jones,; Miller, a. F. A.,

Vibrational Spectra of Cubane and Four

of Its Deuterated Derivatives.

Journal of the American Chemical Society / 101.25 / December 5, I979,7441-7457.

Galasso, V., Theoretical study of spectroscopic properties of cubane.

Chemical Physics 184 (1994) 107-114.

Kirill A. Lukin, J. L., Philip E. Eaton,*,Nobuhiro Kanomata,Juirgen Hain,Eric Punzalan,and

Richard Gilardi, Synthesis and Chemistry of 1,3,5,7-Tetranitrocubane Including

Measurement of Its Acidity, Formation of o-Nitro Anions, and

the First Preparations of Pentanitrocubane and Hexanitrocubane.

J. Am. Chem. Soc., Vol. 119, No. 41, 1997,9592-9602.

P.E.Eaton, Cubanes: starting Materials For the chemistry of 1990s and the New Century.

J. Am. Chem. Soc.;1992;31;1421-1436, 31, 1421-1436.

Philip E. Eaton, t. Y. X., t and Richard Gilardi*, Systematic Substitution on the Cubane Nucleus.

Synthesis and

Properties of 1,3,5-Trinitrocubane and 1,3,5,7-Tetranitrocubane

. J. Am. Chem. SOC.1993,115, 10195-10202.

Philip E. Eaton, R. L. G.; Zhang, a. M.-X., Polynitrocubanes: Advanced High-Density,

High-Energy Materials**. Adv. Mater. 2000, 12, No. 15, August 2.

Philip E. Eaton, Cubane: Starting Materials for the chemistry of the 1990s and the new century.

Angew.Chem.Int.Ed.Engl.1992,31,1421-1436.

Philip E. Eaton, t. Y. X., t and Richard Gilardi*, Systematic Substitution on the Cubane Nucleus.

Synthesis and

Properties of 1,3,5-Trinitrocubane and 1,3,5,7-Tetranitrocubane.

J. Am. Chem. SOC., Vol. 115, No. 22, 1993,10196-10202.

T. YILDIRIM, P. M. G., D. A. NEUMANN, P. E. EATONC and ‘T. EMRICK’, SOLID

CUBANE: A BRIEF REVIEW. Carbon Vol. 36, No. 5-6, pp. 809-815,1998.

Zhang, P. E. E. a. M.-X., Octanitrocubane: A New Nitrocarbon.

Propellants, Explosives, Pyrotechnics 27, 1 – 6 (2002).

Share
Jan 312014
 

VARDENAFIL

224785-90-4  CAS NO

Vardenafil hydrochloride (CAS NO.224785-91-5)

Formula C23H32N6O4S 
Mol. mass 488.604 g/mol

4-[2-Ethoxy-5-(4-ethylpiperazin-1-yl)sulfonyl-phenyl]-9-methyl-7-propyl-3,5,6,8-tetrazabicyclo[4.3.0]nona-3,7,9-trien-2-one

Vivanza, Vardenafil (INN), Levitra (TN),  STK642629, , LEVITRA

Vardenafil (INN) is a PDE5 inhibitor used for treating erectile dysfunction that is sold under the trade names Levitra (Bayer AGGSK, and SP) andStaxyn.

Vardenafil was co-marketed by Bayer PharmaceuticalsGlaxoSmithKline, and Schering-Plough under the trade name Levitra. As of 2005, the co-promotion rights of GSK on Levitra have been returned to Bayer in many markets outside the U.S. In Italy, Bayer sells vardenafil as Levitra and GSK sells it as Vivanza. Thus, because of European Union trade rules, parallel imports might result in Vivanza sold next to Levitra in the EU.

Vardenafil (Levitra) is an oral therapy for the treatment of erectile dysfunction. It is a selective inhibitor of cyclic guanosine monophosphate (cGMP)-specific phosphodiesterase type 5 (PDE5). Penile erection is a hemodynamic process initiated by the relaxation of smooth muscle in the corpus cavernosum and its associated arterioles. During sexual stimulation, nitric oxide is released from nerve endings and endothelial cells in the corpus cavernosum. Nitric oxide activates the enzyme guanylate cyclase resulting in increased synthesis of cyclic guanosine monophosphate (cGMP) in the smooth muscle cells of the corpus cavernosum. The cGMP in turn triggers smooth muscle relaxation, allowing increased blood flow into the penis, resulting in erection. The tissue concentration of cGMP is regulated by both the rates of synthesis and degradation via phosphodiesterases (PDEs). The most abundant PDE in the human corpus cavernosum is the cGMPspecific phosphodiesterase type 5 (PDE5); therefore, the inhibition of PDE5 enhances erectile function by increasing the amount of cGMP.

An orally disintegrating form, marketed as Staxyn, has been gaining approvals in countries such as the United States[1] and Canada.[2]

Vardenafil’s indications and contra-indications are the same as with other PDE5 inhibitors; it is closely related in function to sildenafil citrate (Viagra) and tadalafil (Cialis). The difference between the vardenafil molecule and sildenafil citrate is a nitrogen atom’s position and the change of sildenafil’spiperazine ring methyl group to an ethyl group. Tadalafil is structurally different from both sildenafil and vardenafil. Vardenafil’s relatively short effective time is comparable to but somewhat longer than sildenafil’s.

Beyond its indications for erectile dysfunction, vardenafil may be effective in the treatment of premature ejaculation, where it may significantly increase the time from vaginal penetration to ejaculation.[3]

The common, adverse drug reactions (side-effects) are the same as with other PDE5 inhibitors. The frequent vardenafil-specific side-effect is nausea; the infrequent side-effects are abdominal pain, back pain, photosensitivity, abnormal vision, eye pain, facial edemahypotension, palpitation,tachycardiaarthralgiamyalgia, rash, itch, and priapism.

One possibly serious, but rare, side-effect with vardenafil is heart attack. Also, in rare cases, vardenafil use may cause priapism, a very painful emergency condition that can cause impotence if left untreated.[4]

On 18 October 2007, the U.S. Food and Drug Administration (FDA) announced that a warning about possible deafness (sudden hearing loss) would be added to the drug labels of Vardenafil, and other PDE5 inhibitors.[5]

Vardenafil, as with all PDE5 inhibitors, should not be used by men taking nitrate medications, because combining them with vardenafil might provoke potentially life-threatening hypotension (low blood pressure).

Further, Vardenafil causing lengthening of the QT interval. Therefore it should not be taken by men taking other medications that affect the QT interval (such as amiodarone).

Levitra 20mg Oral Tablet

It is available in 2.5 mg, 5 mg, 10 mg, and 20 mg doses in round orange tablets. The normal starting dose is 10 mg (roughly equivalent to 50 mg of sildenafil). Vardenafil should be taken 1 to 2 hours prior to sexual activity, with a maximum dose frequency of once per day. In some territories, such as the UK, only certain doses may be available.

Vardenafil is also available under the name Staxyn as a tablet which dissolves on the tongue rather than being swallowed in the form of a pill.

STAXYN is an oral therapy for the treatment of erectile dysfunction. This monohydrochloride salt of vardenafil is a selective inhibitor of cyclic guanosine monophosphate (cGMP)-specific PDE5.

Vardenafil HCl is designated chemically as piperazine, 1-[[3-(1,4-dihydro-5-methyl-4-oxo-7-propylimidazo[5,1f][1,2,4]triazin-2-yl)-4-ethoxyphenyl]sulfonyl]-4-ethyl-, monohydrochloride and has the following structural formula:

 

STAXYN (vardenafil hydrochloride) Structural Formula Illustration

 

Vardenafil HCl is a nearly colorless, solid substance with a molecular weight of 579.1 g/mol and a solubility of 0.11 mg/mL in water.

LEVITRA

TRIHYDRATE, HCL SALT

 

 

Abbreviated New Drug Application (ANDA)

 

 



US2002137930A

 

 

 

vardenafil hydrochloride is piperazine, 1-[[3-(1,4-dihydro-5-methyl-4-oxo-7-propylimidazo[5,1-f][1,2,4]triazin-2-yl)-4-ethoxyphenyl]sulfonyl]-4-ethyl-, mono -hydrochloride and can be structurally represented by Formula I.

 

Figure US07977478-20110712-C00001

 

The monohydrochloride salt of vardenafil is a selective inhibitor of cyclic guaosine monophosphate (cGMP)-specific phosphodiesterase type 5 (PDE5). It is commercially available in products sold under the brand name LEVITRA formulated as 2.5 mg, 5 mg, 10 mg, 20 mg film-coated tablets.

U.S. Pat. No. 6,362,178 B1 discloses vardenafil, its related compounds and processes for their preparation. The patent describes a process in which vardenafil is obtained by recrystallization in ether in Example 19. Vardenafil produced as per Example 19 is hereinafter referred as “crystalline Form I” of vardenafil. The patent also describes processes for the preparation of its monohydrochloride and dihydrochloride salts, which are formed in a combination of ether and dichloromethane. The patent also describes a process for the preparation of vardenafil monohydrochloride trihydrate.

U.S. Patent Application Publication No. 2005/0203298 also describes a process for the preparation of vardenafil, and its monohydrochloride trihydrate.

Chemical synthesis of vardenafil has mostly been directed to the preparation of the trihydrate of monohydrochloride of vardenafil.

In WO 99/24433, sulphonamide-substituted imidazotriazinones are described as potent inhibitors of either one or more of the cyclic guanosine 3′,5′-monophosphate-metabolizing phosphodiesterases (cGMP PDEs). According to the nomenclature of Beavo and Reifsnyder (Trends in Pharmacol. Sci. 11, 150-155, 1990), these cGMP PDEs are the phosphodiesterase isoenzymes PDE-I, PDE-II and PDE-V.

According to WO 99/24433, the sulphonamide-substituted imidazotriazinones described therein are prepared from corresponding 2-ethoxyphenyl-substituted imidazotriazinones by reaction with chlorosulphonic acid and subsequent reaction with an appropriate amine, as is illustrated by the following scheme (Rto Rhere have the meanings indicated in WO 99/24433):

Figure US20050203298A1-20050915-C00003

 

In this process, highly reactive chlorosulphonic acid has to be used as a reagent. Moreover, the imidazotriazinonesulphonyl chlorides formed as intermediates are sensitive to hydrolysis, which, in particular in the conversion of this preparation process to the industrial scale, can lead to not inconsiderable yield variations.

It was therefore the object of the present invention to make available a process for the preparation of sulphonamide-substituted imidazotriazinones in which the disadvantages of the above process known from the prior art are avoided.

This object is achieved according to the present invention by a process as in claim 1. In particular, in the process according to the invention as in claim 1 the use of chlorosulphonic acid is avoided by introduction of the sulphonic acid via a reaction with sulphuric acid and subsequent reaction with thionyl chloride. Moreover, the reaction with thionyl chloride and the subsequent reaction with an amine is carried out in a one-pot process, so that the imidazotriazinonesulphonyl chloride intermediate, which is sensitive to hydrolysis, does not need to be isolated. By means of this, yield variations on account of partial hydrolysis of this intermediate can be excluded. As a result of these advantages, the process according to the invention is much simpler to carry out on the industrial scale than the process described in WO 99/24433.

………………….

SYNTHESIS

US6362178

2-butyrylamino-propionic acid

EXAMPLE 1A 2-Butyrylaminopropionic acid

Figure US06362178-20020326-C00052

 

22.27 g (250 mmol) of D,L-alanine and 55.66 g (550 mmol) of triethylamine are dissolved in 250 ml of dichloromethane, and the solution is cooled to 0° C. 59.75 g (550 mmol) of trimethylsilyl chloride are added dropwise, and the solution is stirred for 1 hour at room temperature and for 1 hour at 40° C. After cooling to −10° C., 26.64 g (250 mmol) of butyryl chloride are added dropwise, and the resulting mixture is stirred for 2 hours at −10° C. and for one hour at room temperature.

With ice-cooling, 125 ml of water are added dropwise and the reaction mixture is stirred at room temperature for 15 minutes. The aqueous phase is evaporated to dryness, the residue is titrated with acetone and the mother liquor is filtered off with suction. The solvent is removed and the residue is chromatographed. The resulting product is dissolved in 3N aqueous sodium hydroxide solution and the resulting solution is evaporated to dryness. The residue is taken up in conc. HCl and once more evaporated to dryness. The residue is stirred with acetone, precipitated solid is filtered off with suction and the solvent is removed under reduced pressure. This gives 28.2 g (71%) of a viscous oil which crystallizes after some time.

200 MHz 1H-NMR (DMSO-d6): 0.84, t, 3H; 1.22, d, 3H; 1.50, hex, 2H; 2.07, t, 2H; 4.20, quin., 1H; 8.09, d, 1H.

EXAMPLE 3A 2-Ethoxybenzonitrile

Figure US06362178-20020326-C00054

 

25 g (210 mmol) of 2-hydroxybenzonitrile are refluxed with 87 g of potassium carbonate and 34.3 g (314.8 mmol) of ethyl bromide in 500 ml of acetone overnight. The solid is filtered off, the solvent is removed under reduced pressure and the residue is distilled under reduced pressure. This gives 30.0 g (97%) of a colourless liquid.

200 MHz 1H-NMR (DMSO-d6): 1.48, t, 3H; 4.15, quart., 2H; 6.99, dt, 2H; 7.51, dt, 2H.

 2-ethoxybenzamidine hydrochloride

EXAMPLE 4A 2-Ethoxybenzamidine hydrochloride

Figure US06362178-20020326-C00055

 

21.4 g (400 mmol) of ammonium chloride are suspended in 375 ml of toluene, and the suspension is cooled to 0° C. 200 ml of a 2M solution of trimethylaluminium in hexane are added dropwise, and the mixture is stirred at room temperature until the evolution of gas has ceased. After addition of 29.44 g (200 mmol) of 2-ethoxybenzonitrile, the reaction mixture is stirred at 80° C. (bath) overnight.

With ice-cooling, the cooled reaction mixture is added to a suspension of 100 g of silica gel and 950 ml of chloroform, and the mixture is stirred at room temperature for 30 minutes. The mixture is filtered off with suction, and the filter residue is washed with the same amount of methanol. The mother liquor is concentrated, the resulting residue is stirred with a mixture of dichloromethane and methanol (9:1), the solid is filtered off with suction and the mother liquor is concentrated. This gives 30.4 g (76%) of a colourless solid.

200 MHz 1H-NMR (DMSO-d6): 1.36, t, 3H; 4.12, quart., 2H; 7.10, t, 1H; 7.21, d, 1H; 7.52, m, 2H; 9.30, s, broad, 4H.

EXAMPLE 10A 2-(2-Ethoxy-phenyl)-5-methyl-7-propyl-3H-imidazo[5,1-f][1,2,4]triazin-4-one

Figure US06362178-20020326-C00061

 

7.16 g (45 mmol) of 2-butyrylamino-propionic acid and 10.67 g of pyridine are dissolved in 45 ml of THF and, after addition of a spatula tip of DMAP, heated to reflux. 12.29 g (90 mmol) of ethyl oxalyl chloride are slowly added dropwise, and the reaction mixture is refluxed for 3 hours. The mixture is poured into ice-water and extracted three times with ethyl acetate and the organic phase is dried over sodium sulphate and concentrated using a rotary evaporator. The residue is taken up in 15 ml of ethanol and refluxed with 2.15 g of sodium bicarbonate for 2.5 hours. The cooled solution is filtered.

With ice-cooling, 2.25 g (45 mmol) of hydrazine hydrate are added dropwise to a solution of 9.03 g (45 mmol) of 2-ethoxybenzamidine hydrochloride in 45 ml of ethanol, and the resulting suspension is stirred at room temperature for another 10 minutes. The ethanolic solution described above is added to this reaction mixture, and the mixture is stirred at a bath temperature of 70° C. for 4 hours. After filtration, the mixture is concentrated, the residue is partitioned between dichloromethane and water, the organic phase is dried over sodium sulphate and the solvent is removed under reduced pressure.

This residue is dissolved in 60 ml of 1,2-dichloroethane and, after addition of 7.5 ml of phosphorus oxychloride, refluxed for 2 hours. The mixture is diluted with dichloromethane and neutralized by addition of sodium bicarbonate solution and solid sodium bicarbonate. The organic phase is dried and the solvent is removed under reduced pressure. Chromatography using ethyl acetate and crystallization afford 4.00 g (28%) of a colourless solid, Rf=0.42 (dichloromethane/methanol=95:5)

200 MHz 1H-NMR (CDCl3): 1.02, t, 3H; 1.56, t, 3H; 1.89, hex, 2H; 2.67, s, 3H; 3.00, t, 2H; 4.26, quart., 2H; 7.05, m, 2H; 7.50, dt, 1H; 8.17, dd, 1H; 10.00, s, 1H.

EXAMPLE 15A 4-Ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydro-imidazo[5,1-f][1,2,4]triazin-2-yl)-benzenesulphonyl chloride

 

Figure US06362178-20020326-C00066

 

At 0° C., 2.00 g (6.4 mmol) of 2-(2-ethoxy-phenyl)-5-methyl-7-propyl-3H-imidazo[5,1-f][1,2,4]triazin-4-one are slowly added to 3.83 ml of chlorosulphonic acid. At room temperature, the reaction mixture is stirred ovemight, and then poured into ice-water and extracted with dichloromethane. This gives 2.40 g (91%) of a colourless foam.

200 MHz 1H-NMR (CDCl3): 1.03, t, 3H; 1.61, t, 2H; 1.92, hex, 2H; 2.67, s, 3H; 3.10, t, 2H; 4.42, quart., 2H; 7.27, t, 1H; 8.20, dd, 1H; 8.67, d, 1H; 10.18, s, 1H.

Example 19 2-[2-Ethoxy-5-(4-ethyl-piperazine-1-sulphonyl)-phenyl]-5-methyl-7-propyl-3H-imidazo[5,1-f][1,2,4]triazin-4-one

 

Figure US06362178-20020326-C00093

 

470 mg (1.14 mmol) of 4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydro-imidazo[5,1-f][1,2,4]triazin-2-yl)-benzenesulphonyl chloride are dissolved in 20 ml of dichloromethane and cooled to 0° C. 390 mg (3.42 mmol) of N-ethylpiperazine are added, and the reaction mixture is stirred at room temperature overnight. The mixture is diluted with dichloromethane, the organic phase is washed twice with water and dried over sodium sulphate and the solvent is removed under reduced pressure. Crystallization from ether gives 370 mg (66%) of a colourless solid.

400 MHz 1H-NMR (CDCl3): 1.01, t, 3H; 1.59, t, 3H; 1.88, hex, 2H; 2.42, quart., 2H; 2.56, m, 4H; 2.63, s, 3H; 3.00, t, 2H; 3.10, m, 4H; 4.33, quart., 2H, 7.17, d, 1H; 7.88, dd, 1H; 8.44, d, 1H; 9.75, s, 1H.

…………………….

US7977478

EXAMPLE 7 Preparation of the Trihydrate of Vardenafil Monohydrochloride

14 g of vardenafil hydrochloride was taken into a round bottom flask followed by the addition of 70 ml water and the pH of the reaction mass was adjusted using sodium hydroxide to 11 at 30° C. 280 ml of dichloromethane was added to the above reaction mass and the layers were separated. The organic layer was dried over sodium sulfate and the organic layer was transferred into a round bottom flask and subjected to heating for distillation at 40° C. for 1.5 hours. The solid material was transferred into a round bottom flask and 36 ml of a mixture of acetone and water in 12:1 ratio was added with stirring, then 2.2 ml of 36% aqueous hydrochloric acid was added with stirring. The reaction mass was heated to a temperature of about 45° C. and the undissolved particles were removed by filtration. The filtrate was taken into a round bottom flask and cooled to 5° C., maintained for 45 minutes at 3 to 5° C. followed by the filtration of the solid which was then subjected to suction drying and finally dried at 40° C. to yield 9.0 g of the trihydrate of vardenafil monohydrochloride.

……………………..

US20050203298

STARTING COMPOUNDS

Example I Preparation of 2-(2-ethoxyphenyl)-5-methyl-7-propyl-3H-imidazo-[5,1-f][2,4]triazin-4-oneIa) Preparation of 2-butyrylaminopropionic acid

 

Figure US20050203298A1-20050915-C00021

 

A solution of 100 kg of D,L-alanine in aqueous sodium hydroxide solution is reacted in the cold with 119 kg of butyryl chloride. After addition of butyl acetate, the mixture is acidified with hydrochloric acid, the organic phase is separated off and the aqueous phase is re-extracted. The organic phase is dried by azeotropic distillation. The crystallizate is isolated, washed with butyl acetate and dried.

Yield: 132.6 kg (68%)

1H-NMR: δ=0.8 (t, 3H), 1.25 (d, 3H), 1.5 (m, 2H), 2.1 (t, 2H), 4.2 (q, 1H), 8.1 (d, NH), 12.0-12.7 (s, COOH)

MS: 336 (2M+NH4, 40), 319 (2M+H, 15), 177 (M+NH4, 100), 160 (M+H, 20)

Ib) Preparation of 2-ethoxybenzonitrile

 

Figure US20050203298A1-20050915-C00022

 

260 kg of thionyl chloride are added at 85-95° C. to a suspension of 250 kg of 2-ethoxybenzamide in toluene under metering control. The reaction mixture is stirred in the presence of heat. Thionyl chloride and toluene are then distilled off in vacuo. The product is employed in the subsequent stage as a crude product.

Yield: 228.5 kg (crude product)

1H-NMR: δ=1.45 (t, 3H), 4.15 (q, 2H), 7.0 (m, 2H, phenyl), 7.5 (m, 2H, phenyl)

MS: 312 (2M+N4, 35), 165 (M+NH4, 100), 147 (5)

Ic) Preparation of 2-ethoxy-N-hydroxybenzamidine

 

Figure US20050203298A1-20050915-C00023

 

111 kg of 2-ethoxybenzonitrile (crude product) from Example Ib are heated under reflux with 164 1 of triethylamine and 73 kg of hydroxylamine hydrochloride in isopropanol. The reaction mixture is treated with water and cooled. The crystallizate is isolated, washed and employed in the subsequent stage as a moist product.

Yield: 92.6 kg (moist product)

1H-NMR: δ=1.35 (t, 3H), 4.1 (q, 2H), 5.6 (s, 2H), 6.9-7.4 (4H, phenyl), 9.4 (s, 1H, OH)

MS: 361 (2M+H, 30), 198 (M+N, 30), 181 (M+H, 100)

Id) Preparation of 2-ethoxybenzamidine hydrochloride

 

Figure US20050203298A1-20050915-C00024

 

135 kg of 2-ethoxy-N-hydroxybenzamidine (moist product) from Example Ic are hydrogenated at 50-60° C. in acetic acid using palladium on carbon as a catalyst. For the work-up, the hydrogenation reaction is freed from the catalyst, treated with hydrochloric acid and concentrated. Residual acetic acid and water are removed by azeotropic distillation with toluene. The crystallizate is isolated and dried in vacuo.

Yield: 136.4 kg

H-NMR: 1.35 (t, 3H), 4.15 (q, 2H), 7.1-7.7 (m, 4H, phenyl), 9.1-9.4 (2×s, 3H), 10.5-10.7 (s, 1H)

MS: 329 (2M+H, 10), 165 (M+H, 100)

Ie) Preparation of 2-(2-ethoxyphenyl)-5-methyl-7-propyl-3H-imidazo[5,1-f][1,2,4]-triazin-4-one

 

Figure US20050203298A1-20050915-C00025

 

231 kg of 2-butyrylaminopropionic acid from Example Ia are treated in tetrahydrofuran with 341 kg of pyridine, catalytic amounts of 4-N,N-dimethylaminopyridine and 392 kg of ethyl chloroxalate and stirred with heating under reflux. The reaction mixture is taken up in ethyl acetate, washed with water and the ethyl acetate phase is concentrated. The distillation residue is taken up in methanol and reacted with the following solution.

192 kg of 2-ethoxybenzamidine hydrochloride from Example Id are treated in methanol with 47.5 kg of hydrazine hydrate and the mixture is stirred at room temperature. The solution is combined with the solution of 2-butyrylamino-1-ethoxycarbonylpropenyl ethyl oxalate prepared above. The reaction mixture thus obtained is stirred with heating under reflux. Methanol is removed by distillation and replaced by acetic acid.

Option A:

138.6 kg of phosphorus oxychloride are added and stirred in the presence of heat.

Acetic acid is distilled off in vacuo. The residue is treated with water and dichloromethane or optionally methyl isobutyl ketone and rendered neutral using sodium hydroxide solution. The organic phase is concentrated, and the residue is dissolved in acetone and crystallized with cooling. The crystallizate is isolated, washed and dried.

Option B:

At least 190 kg of acetyl chloride are added and stirred in the presence of heat. Acetic acid is distilled off in vacuo. The distillation residue is treated with acetone and water, and the product is crystallized by rendering neutral with sodium hydroxide solution. The product is isolated, washed and dried.

Yield: 90-160 kg

1H-NMR: δ=1.0 (t, 3H), 1.6 (t, 3H), 1.9 (m, 2H), 2.8 (s, 3H), 3.3 (t, 2H), 4.3 (q, 2H), 7.0-8.2 (Ar, 4H), 10.3 (CONH, 1H)

MS: 313 (M+H, 100), 149 (25), 151 (40), 121 (15)

HPLC: Kromasil C-18 phase, neutral phosphate buffer, acetonitrile, 233 nm, linear gradient of 30% acetonitrile ->80% acetonitrile (30 min.): 99 area % (R19.1)

PREPARATION EXAMPLES Example 1a 4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-fl-][1,2,4]triazin-2-yl)benzenesulphonic acid

 

Figure US20050203298A1-20050915-C00026

 

194 kg of 2-(2-ethoxyphenyl)-5-methyl-7-propyl-3H-imidazo[5,1-f][1,2,4]triazin-4-one from Example Ie are reacted with 504 kg of concentrated sulphuric acid. The reaction mixture is added to water, cooled, and the crystallizate is isolated and dried in vacuo.

Yield: 195.2 kg

1H-NMR: δ=0.95 (t, 3H), 1.3 (t, 3H), 1.8 (m, 2H), 2.6 (s, 3H), 3.05 (t, 2H), 4.1 (q, 2H), 7.15 (Ar, 1H), 7.75 (m, 2H), 12.3 (SO2OH)

MS: 393 (M+H, 100), 365 (25), 151 (40)

HPLC: X-Terra C-18 phase, aqueous phosphoric acid, acetonitrile, 242 nm, linear gradient of 10% acetonitrile ->90% acetonitrile (20 min.):

98 area % (R, 9.2)

Example 1b) 2-[2-ethoxy-5-(4-ethlylpiperazin-1-sulphonyl)phenyl]-5-methyl-7-propyl-3H-imidazo[5,1-f][1,2,4]triazin-4-one

 

Figure US20050203298A1-20050915-C00027

 

22.5 kg of 4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydro-imidazo[5,1-f][1,2,4]-triazin-2-yl)benzenesulphonic acid from Example 1a are reacted with 74 kg of thionyl chloride and catalytic amounts of dimethylformamide until the evolution of gas has ended. Xylene is repeatedly added to the reaction mixture and thionyl chloride is distilled off. 15.1 kg of N-ethylpiperazine are added to the suspension and it is stirred. After the addition of water, it is adjusted to pH 1 using hydrochloric acid, and the phases are separated. The aqueous phase is treated with acetone and rendered neutral by addition of sodium hydroxide solution. The mixture is cooled, and the crystallizate is isolated, washed and dried in vacuo.

Yield: 26.1 kg

1H-NMR: δ=1.0 (2×t, 6H), 1.6 (t, 3H), 1.9 (m, 2H), 2.45 (q, 2H), 2.55 (m, 4H), 2.65 (s, 3H), 3.0 (t, 2H), 3.1 (m, 4H), 4.35 (q, 2H), 7.15 (Ar, 1H), 7.9 (Ar, 1H), 8.4 (Ar, 1H), 9.8 (CONH)

MS: 489 (M+H, 100), 345 (10), 313, (10), 285 (10), 113 (20)

HPLC: X-Terra C-18 phase, neutral phosphate buffer, acetonitrile, 242 nm, linear gradient of 20% acetonitrile ->75% acetonitrile (20 min.): 98 area % (R16.3)

1 c) 2-[2-ethoxy-5-(4-ethylpiperazin-1-sulphonyl)phenyl]-5-methyl-7-propyl-3H-imidazo[5,1-fl][1,2,4]triazin-4-one hydrochloride trihydrate

 

Figure US20050203298A1-20050915-C00028

 

22.5 kg of 2-[2-ethoxy-5-(4-ethylpiperazin-1-sulphonyl)phenyl]-5-methyl-7-propyl-3H-imidazo[5,1-f][1,2,4]triazin-4-one from Example 1b are dissolved in 5.1 kg of concentrated hydrochloric acid and acetone/water (12:1 v/v) in the presence of heat. The clear solution is filtered hot and crystallized by cooling and seeding. The crystallizate is isolated, washed and dried in vacuo at about 30° C. and about 300 mbar.

Yield: 25.4 kg

M.p. (DSC): 192° C.

HPLC: X-Terra C-18 phase, neutral phosphate buffer, acetonitrile, 242 nm, linear gradient of 20% acetonitrile ->75% acetonitrile (20 min.): 99 area % (R16.3)

 

  1.  http://www.pharmpro.com/News/Feeds/2010/06/pharmaceutical-companies-bayer-new-erectile-dysfunction-treatment-staxyn-approve/
  2.  http://www.newswire.ca/en/story/832217/staxyn-new-innovation-in-erectile-dysfunction-helps-younger-men-rise-to-the-occasion
  3.  A Aversa et al. “Effects of vardenafil administration on intravaginal ejaculatory latency time in men with lifelong premature ejaculation”. Retrieved 2010-12-14.
  4.  Schools of Pharmacy (Glen L. Stimmel, Pharm.D., and Mary A. Gutierrez, Pharm.D.) and Medicine (Glen L. Stimmel, Pharm.D.), University of Southern California, Los Angeles, California. “Counseling Patients About Sexual Issues: Drug-Induced Priapism”. Medscape. Retrieved 2010-12-06.
  5.  “FDA Announces Revisions to Labels for Cialis, Levitra and Viagra”Food and Drug Administration. 2007-10-18. Retrieved 2009-08-06.

 

PATENTS

US6362178 * Oct 31, 1998 Mar 26, 2002 Bayer Aktiengesellschaft 2-phenyl substituted imidazotriazinones as phosphodiesterase inhibitors
US20050203298 * May 5, 2005 Sep 15, 2005 Bayer Healthcare Aktiengesellschaft Process for the preparation of sulphonamide-substituted imidazotriazinones
US20060111354 * Jul 3, 2003 May 25, 2006 Peter Serno Medicaments containing vardenafil hydrochloride trihydrate
WO2004006894A1 * Jul 3, 2003 Jan 22, 2004 Bayer Healthcare Ag Medicaments containing vardenafil hydrochloride trihydrate

 

11-4-2011
ROFLUMILAST FOR THE TREATMENT OF DIABETES MELLITUS
9-14-2011
Roflumilast for the Treatment of Diabetes Mellitus
8-5-2011
N-BUTYRAMIDE, THE PREPARATION METHOD AND USE THEREOF
3-4-2011
Fatty Acid Oxidation Inhibitors Treating Hyperglycemia and Related Disorders
1-14-2011
2-PHENYL SUBSTITUTED IMIDAZOTRIAZINONES AS PHOSPHODIESTERASE INHIBITORS
9-17-2010
SUBSTITUTED PDE5 INHIBITORS
7-16-2010
Combination treatment for diabetes mellitus
4-28-2010
2-Phenyl substituted imidazotriazinones as phosphodiesterase inhibitors
4-14-2010
2-PHENYL SUBSTITUTED IMIDAZOTRIAZINONES AS PHOSPHODIESTERASE INHIBITORS
2-5-2010
Heterocyclic Compounds And Uses Thereof In The Treatment Of Sexual Disorders

 

12-25-2009
Therapeutic Compositions Comprising a Specific Endothelin Receptor Antagonist and a PDE5 Inhibitor
11-27-2009
Substituted PDE5 inhibitors
9-4-2009
Uses of 2-Phenyl-Substituted Imidazotriazinone Derivatives for Treating Pulmonary Hypertension
8-28-2009
Roflumilast for the Treatment of Pulmonary Hypertension
8-7-2009
Use of Phosphodiesterase Inhibitor as a Component of Implantable Medical Devices
6-26-2009
Method for healing a wound using a phosphodiesterase type five inhibitor
3-20-2009
Pde5 inhibitor compositions and methods for immunotherapy
3-6-2009
Pde5 inhibitor compositions and methods for treating cardiac indications
10-31-2008
Formulations with Controlled Release of Active Ingredient
8-15-2008
HIGHLY SELECTIVE and LONG-ACTING PDE5 MODULATORS

 

8-8-2008
Formulations With Controlled Release Of Active Ingredient
4-11-2008
Use of 2-alkoxyphenyl-substituted imidazotriazinones
1-2-2008
2-phenyl substituted imidazotriazinones as phosphodiesterase inhibitors, for treatment of hypertension
12-28-2007
Novel Uses of 2-Phenyl-Substituted Imidazotriazinone Derivatives
10-3-2007
Use of 2-alkoxyphenyl-substituted imidazotriazinones
11-24-2006
Methods for synthesizing imidazotriazinones
10-18-2006
2-Phenyl substituted imidazotriazinones as phosphodiesterase inhibitors
2-15-2006
Process for the preparation of sulphonamide-substituted imidazotriazinones
8-17-2005
Use of 2-alkoxyphenol-substituted imidazotriazinones
5-11-2005
2-phenyl substituted imidazotriazinones as phosphodiesterase inhibitors

 

1-21-2005
Process for the preparation of sulphonamide-substituted imidazotriazinones
8-18-2004
Process for the preparation of sulphonamide-substituted imidazotriazinones
8-6-2004
Novel use of 2-phenyl-substituted imidazotriazinones
7-32-2003
Daily treatment for erectile dysfunction using a PDE5 inhibitor
5-21-2003
2-phenyl substituted imidatriazinones as phosphodiesterase inhibitors
3-27-2002
2-phenyl substituted imidazotriazinones as phosphodiesterase inhibitors
12-21-2001
Daily treatment for erectile dysfunction using a PDE5 inhibitor
5-21-1999
2-PHENYL SUBSTITUTED IMIDAZOTRIAZINONES AS PHOSPHODIESTERASE INHIBITORS

 ////////////

Share
Jan 312014
 

 

Lodenafil carbonate

UNII-29X84F932D, CRIS-031  

bis-(2-{4-[4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)-benzenesulfonyl]piperazin-1-yl}-ethyl)carbonate

5-{2-ethoxy-5-[(4-hydroxyethyl-1-piperazinyl)sulfonyl]phenyl}-1-methyl-3-n-propyl-1,6-dihydro-7H-pyrazole[4,3-d]pyrimidin-7-one. IS THE NAME OF MONOMER

398507-55-6  CAS

Cristalia (Originator)

C47 H62 N12 O11 S2= MF
 Molecular Weight 1035.199

Lodenafil is a drug belonging to a class of drugs called PDE5 inhibitor, which many other erectile dysfunction drugs such as sildenafiltadalafil, and vardenafil also belong to. Like udenafil and avanafil it belongs to a new generation of PDE5 inhibitors.

Lodenafil is formulated as a dimerlodenafil carbonate, which breaks down in the body to form two molecules of the active drug lodenafil. This formulation has higher oral bioavailability than the parent drug.[1]

It is manufactured by Cristália Produtos Químicos e Farmacêuticos in Brazil and sold there under the brand-name Helleva.[2]

Helleva (Lodenafil Carbonate) - 80mg (4 Tablets)

Helleva (Lodenafil Carbonate) is an oral PDE5 inhibitor prescribed to treat men suffering from erectile dysfunction. It operates by relaxing muscles and dilating blood vessels in the penis to increase circulation making it easier to attain and maintain an erection.

It has undergone Phase III clinical trials,[3][4][5] but is not yet approved for use in the United States by the U.S. Food and Drug Administration.

lodenafil

………..

SYNTHESIS

WO 2002012241 OR US7148350

MONOMER synthesis

PIPERAZINE

AND

ETHYL CHLORO ACETATE

WILL GIVE

Ethyl 1-piperazinylacetateChemSpider 2D Image | Ethyl 1-piperazinylacetate | C8H16N2O2

SEE RXN 1 BELOW

Reaction 1:

Synthesis of Piperazine Ethyl Acetate

To a reaction blend containing 100 g (3 Eq, 0.515 mol, MW=194) of piperazine, 26.3 mL (1.1 Eq, 0.189 mol, MW=101, d=0.726) of triethylamine in 200 mL of isopropanol, add to a solution previously prepared of 18.4 mL (1 Eq., 0.172 mol, MW=122.55, d=1.15) of chloroacetate of ethyl in 140 mL of isopropanol under stirring, at room temperature. Keep the reaction medium under stirring, monitoring the reaction termination by means of a chromatography of the thin layer (about 2–3 hours). Add a solution of 40.6 g (0.344 mol) of succinic acid in 140 mL of isopropanol. Keep the system under stirring for about 30 minutes to assure total precipitation of the succinate salt of piperazine formed. Filter this salt and concentrate the filtrate containing the mono and dialkyled derivatives. We obtain a slightly yellowish oil, which is used in later phases without purification.

Mass obtained=33 g

GC/MS: Monoalkylated derivative 72%, and dialkylated 22%.

NEXT

ChemSpider 2D Image | Ethyl 1-piperazinylacetate | C8H16N2O2Piperazine Ethyl Acetate

AND

5-(5-Chlorosulfonyl-2-ethoxyphenyl)-1-methyl-3-propyl-1,6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one Structure

5-(5-chlorosulfonyl-2-ethoxyphenyl)-1-methyl-3n-propyl-1,6-dihydro-7H-pyrazole[4,3-d]pyrimidin-7-one

WILL REACT TO GIVE… 5-{2-ethoxy-5-[(4-ethyl acetate 1-piperazinyl)sulfonyl]phenyl}-1-methyl-3-n-propyl-1,6-di-hydro-7H-pyrazole[4,3-d]pyrimidin-7-one AS IN RXN 4 BELOW

Reaction 4:

Synthesis of 5-{2-ethoxy-5-[(4-ethyl acetate 1-piperazinyl)sulfonyl]phenyl}-1-methyl-3-n-propyl-1,6-di-hydro-7H-pyrazole[4,3-d]pyrimidin-7-one.

Suspend 24.6 g (60 mmol, MW=410.9) of 5-(5-chlorosulfonyl-2-etoxyphenyl)-1-methyl-3n-propyl-1,6-dihydro-7H-pyrazole[4,3-d]pyrimidin-7-one in 900 mL of ethanol absolute. Under stirring and at room temperature, add at only one time, a solution containing 31.0 g (3 Eq., 180 mmol MW=172) of N-piperazine ethyl acetate (Reaction 1) dissolved in 150 mL of ethanol absolute. In an interval of 2–10 minutes, all solid is consumed, forming a clean and homogeneous solution, and after that starts the precipitation of the expected product. At the end of the reaction, which lasts 2–3 hours (monitored by chromatography of thin layer), the product is vacuum filtered and the solid is washed with two portions of 50 mL of iced absolute ethanol. 29 g are obtained (yielding=89%) from the product as a white solid of MP=165.5–166.5° C.

Reaction 7:

Intermediate 1

5-{2-ethoxy-5-[(4-hydroxyethyl-1-piperazinyl)sulfonyl]phenyl}-1-methyl-3-n-propyl-1,6-dihydro-7H-pyrazole[4,3-d]pyrimidin-7-one.  IS MONOMER

please note during LAH redn  …………. the PIP CH2-C=O-O CH2 CH3     BECOMES        PIP-CH2CH2-OH

To a suspension of lithium aluminum hydride (0.74 g 2.2 Eq. MW=37.9) in 25 mL of THF, slowly add, under stirring and at room temperature, a suspension of 5.0 g (9.1 mmol, MW=546.6) of 5-{2-ethoxy-5-[(4-ethyl acetate 1-piperazinyl)sulfonyl]phenyl}-1-methyl-3-n-propyl-1,6-di-hydro-7H-pyrazole[4,3-d]pyrimidin-7-one in 50 mL of THF. The system is maintained under stirring, monitoring the consumption of the product by chromatography of thin layer, until the complete consumption of the starting reagent (about 5–6 hours). Slowly add water to the reaction medium and, when there is no longer release of H2, add HCl 1M regulating pH for 7. Extract the product with 3 200 mL-portions of chloroform, dry with anhydrous sodium sulfate and vacuum concentrate the product. It is obtained 3.8 g of the product as a cream solid MP=183–187° C. yielding 83%. The same was crystallized from methanol and DMF yielding a slightly yellowish solid with melting point at 189–192° C.

 

note …………. the PIP CH2-C=O-O CH2 CH3 BECOMES  PIP-CH2CH2-OH

 

HOMODIMER CARBONATE

 

EXAMPLE 1B

Homodimer Carbonate of Intermediate 1—Alternative Method

A phosgene solution (3.5 g, 35 mmol) dissolved in 20 mL of toluene was added dropwise to a solution of 2.02 g (4 mmol) of 5-{2-ethoxy-5-[(4-hydroxyethyl-1-piperazinyl)sulfonyl]phenyl}-1-methyl-3-n-propyl-1,6-dihydro-7H-pyrazole[4,3-d]pyrimidin-7-one, suspended in 44 mL of toluene. The reaction mixture resulting is stirred and followed by chromatography analysis of thin layer every hour until the reagent conversion in its chloroformate was completed. When the analysis indicates the complete consumption of 5-{2-ethoxy-5-[(4-hydroxyethyl-1-piperazinyl)sulfonyl]phenyl}-1-methyl-3-n-propyl-1,6-dihydro-7H-pyrazole[4,3-d]pyrimidin-7-one, the volatile compounds of the reaction are vacuum removed (solvents and phosgene), yielding the esther chloroformate raw derivative of 5-{2-ethoxy-5-[(4-hydroxyethyl-1-piperazinyl)sulfonyl]phenyl}-1-methyl-3-n-propyl-1,6-dihydro-7H-pyrazole[4,3-d]pyrimidin-7-one.

The raw chloroformate obtained above (4.0 mmol, 2.27 g) is dissolved in about 30 mL of dichloromethane, to which is added 2.07 g (4.1 mmol) of 5-{2-ethoxy-5-[(4-hydroxyethyl-1-piperazinyl)sulfonyl]phenyl}-1-methyl-3-n-propyl-1,6-dihydro-7H-pyrazole[4,3-d]pyrimidin-7-one, followed by the addition of 4 mL of dichloromethane containing 450 mg of triethylamine. The reaction mixture is maintained under stirring, being followed by chromatography of thin layer every hour until this indicates the end of the reaction (disappearing of chloroformate derivative). The reaction mixture is then diluted with 60 mL of dichloromethane, washed with NaCl saturated solution, after with sodium bicarbonate saturated solution and again with NaCl saturated solution. Organic phase is separated and dry with anhydrous sodium sulfate. The solvent is then evaporated to dry, yielding the dimer carbonate as a slightly yellowish solid.

This compound is re-crystallized from ethanol:DMF, yielding a pale white solid. Yielding m=3.2 g (76%)

Microanalysis: Theoretical C, (54.53%); H, (6.04%); N, (16.24%);

Obtained C, (54.45%); H, (6.02%); N, (16.17%).

 

INFO ABOUT INTERMEDIATE

5-(5-Chlorosulfonyl-2-ethoxyphenyl)-1-methyl-3-propyl-1,6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one Structure

5-(5-chlorosulfonyl-2-ethoxyphenyl)-1-methyl-3n-propyl-1,6-dihydro-7H-pyrazole[4,3-d]pyrimidin-7-one

CAS No. 139756-22-2
Chemical Name: 5-(5-Chlorosulfonyl-2-ethoxyphenyl)-1-methyl-3-propyl-1,6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one
Synonyms: Sildenafil Chlorosulfone IMpurity;Sildenafil Chlorosulfonyl IMpurity;5-(5-CHLOROSULFONYL-2-ETHOXY PHENYL)-1-METHYL-3-N-PROPYL-1;3-(6,7-dihydro-1-methyl-7-oxo-3-propyl-1 H-pyrazolo-(4-3-d)-pyrimidine-5;5-(5-Chlorosulfonyl-2-ethoxyphenyl)-1-methyl-3-propyl-1,6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one;3-(4,7-Dihydro-1-methyl-7-oxo-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-5-yl)-4-ethoxy-benzenesulfonyl Chloride;4-Ethoxy-3-(1-Methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyriMidin-5-yl)benzene-1-sulfonyl chloride
CBNumber: CB11175931
Molecular Formula: C17H19ClN4O4S

http://www.chemicalbook.com/ChemicalProductProperty_EN_CB11175931.htm

…………..

SYNTHESIS OF

Figure US06362178-20020326-C00096

http://www.google.co.in/patents/US6362178

2-butyrylamino-propionic acid
EXAMPLE 1A 2-Butyrylaminopropionic acid 

Figure US06362178-20020326-C00052

 

22.27 g (250 mmol) of D,L-alanine and 55.66 g (550 mmol) of triethylamine are dissolved in 250 ml of dichloromethane, and the solution is cooled to 0° C. 59.75 g (550 mmol) of trimethylsilyl chloride are added dropwise, and the solution is stirred for 1 hour at room temperature and for 1 hour at 40° C. After cooling to −10° C., 26.64 g (250 mmol) of butyryl chloride are added dropwise, and the resulting mixture is stirred for 2 hours at −10° C. and for one hour at room temperature.

With ice-cooling, 125 ml of water are added dropwise and the reaction mixture is stirred at room temperature for 15 minutes. The aqueous phase is evaporated to dryness, the residue is titrated with acetone and the mother liquor is filtered off with suction. The solvent is removed and the residue is chromatographed. The resulting product is dissolved in 3N aqueous sodium hydroxide solution and the resulting solution is evaporated to dryness. The residue is taken up in conc. HCl and once more evaporated to dryness. The residue is stirred with acetone, precipitated solid is filtered off with suction and the solvent is removed under reduced pressure. This gives 28.2 g (71%) of a viscous oil which crystallizes after some time.

200 MHz 1H-NMR (DMSO-d6): 0.84, t, 3H; 1.22, d, 3H; 1.50, hex, 2H; 2.07, t, 2H; 4.20, quin., 1H; 8.09, d, 1H.

EXAMPLE 3A 2-Ethoxybenzonitrile 

Figure US06362178-20020326-C00054

 

25 g (210 mmol) of 2-hydroxybenzonitrile are refluxed with 87 g of potassium carbonate and 34.3 g (314.8 mmol) of ethyl bromide in 500 ml of acetone overnight. The solid is filtered off, the solvent is removed under reduced pressure and the residue is distilled under reduced pressure. This gives 30.0 g (97%) of a colourless liquid.

200 MHz 1H-NMR (DMSO-d6): 1.48, t, 3H; 4.15, quart., 2H; 6.99, dt, 2H; 7.51, dt, 2H.

 2-ethoxybenzamidine hydrochloride
EXAMPLE 4A 2-Ethoxybenzamidine hydrochloride 

Figure US06362178-20020326-C00055

 

21.4 g (400 mmol) of ammonium chloride are suspended in 375 ml of toluene, and the suspension is cooled to 0° C. 200 ml of a 2M solution of trimethylaluminium in hexane are added dropwise, and the mixture is stirred at room temperature until the evolution of gas has ceased. After addition of 29.44 g (200 mmol) of 2-ethoxybenzonitrile, the reaction mixture is stirred at 80° C. (bath) overnight.

With ice-cooling, the cooled reaction mixture is added to a suspension of 100 g of silica gel and 950 ml of chloroform, and the mixture is stirred at room temperature for 30 minutes. The mixture is filtered off with suction, and the filter residue is washed with the same amount of methanol. The mother liquor is concentrated, the resulting residue is stirred with a mixture of dichloromethane and methanol (9:1), the solid is filtered off with suction and the mother liquor is concentrated. This gives 30.4 g (76%) of a colourless solid.

200 MHz 1H-NMR (DMSO-d6): 1.36, t, 3H; 4.12, quart., 2H; 7.10, t, 1H; 7.21, d, 1H; 7.52, m, 2H; 9.30, s, broad, 4H.

EXAMPLE 10A 2-(2-Ethoxy-phenyl)-5-methyl-7-propyl-3H-imidazo[5,1-f][1,2,4]triazin-4-one

 

Figure US06362178-20020326-C00061

 

7.16 g (45 mmol) of 2-butyrylamino-propionic acid and 10.67 g of pyridine are dissolved in 45 ml of THF and, after addition of a spatula tip of DMAP, heated to reflux. 12.29 g (90 mmol) of ethyl oxalyl chloride are slowly added dropwise, and the reaction mixture is refluxed for 3 hours. The mixture is poured into ice-water and extracted three times with ethyl acetate and the organic phase is dried over sodium sulphate and concentrated using a rotary evaporator. The residue is taken up in 15 ml of ethanol and refluxed with 2.15 g of sodium bicarbonate for 2.5 hours. The cooled solution is filtered.

With ice-cooling, 2.25 g (45 mmol) of hydrazine hydrate are added dropwise to a solution of 9.03 g (45 mmol) of 2-ethoxybenzamidine hydrochloride in 45 ml of ethanol, and the resulting suspension is stirred at room temperature for another 10 minutes. The ethanolic solution described above is added to this reaction mixture, and the mixture is stirred at a bath temperature of 70° C. for 4 hours. After filtration, the mixture is concentrated, the residue is partitioned between dichloromethane and water, the organic phase is dried over sodium sulphate and the solvent is removed under reduced pressure.

This residue is dissolved in 60 ml of 1,2-dichloroethane and, after addition of 7.5 ml of phosphorus oxychloride, refluxed for 2 hours. The mixture is diluted with dichloromethane and neutralized by addition of sodium bicarbonate solution and solid sodium bicarbonate. The organic phase is dried and the solvent is removed under reduced pressure. Chromatography using ethyl acetate and crystallization afford 4.00 g (28%) of a colourless solid, Rf=0.42 (dichloromethane/methanol=95:5)

200 MHz 1H-NMR (CDCl3): 1.02, t, 3H; 1.56, t, 3H; 1.89, hex, 2H; 2.67, s, 3H; 3.00, t, 2H; 4.26, quart., 2H; 7.05, m, 2H; 7.50, dt, 1H; 8.17, dd, 1H; 10.00, s, 1H.

EXAMPLE 15A 4-Ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydro-imidazo[5,1-f][1,2,4]triazin-2-yl)-benzenesulphonyl chloride

 

Figure US06362178-20020326-C00066

 

At 0° C., 2.00 g (6.4 mmol) of 2-(2-ethoxy-phenyl)-5-methyl-7-propyl-3H-imidazo[5,1-f][1,2,4]triazin-4-one are slowly added to 3.83 ml of chlorosulphonic acid. At room temperature, the reaction mixture is stirred ovemight, and then poured into ice-water and extracted with dichloromethane. This gives 2.40 g (91%) of a colourless foam.

200 MHz 1H-NMR (CDCl3): 1.03, t, 3H; 1.61, t, 2H; 1.92, hex, 2H; 2.67, s, 3H; 3.10, t, 2H; 4.42, quart., 2H; 7.27, t, 1H; 8.20, dd, 1H; 8.67, d, 1H; 10.18, s, 1H.

Example 22 2-[2-Ethoxy-5-(4-hydroxyethyl-1-amino-piperazine-1-sulphonyl)-phenyl]-5-methyl-7-propyl-3H-imidazo[5,1-f][1,2,4]triazin-4-one

 

Figure US06362178-20020326-C00096

 

By the same method, starting with 0.04 g (0.097 mmol) of 4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydro-imidazo[5,1-f][1,2,4]triazin-2-yl)-benzenesulphonyl chloride and 0.04 g (0.29 mmol) of 1-amino-4-hydroxyethylpiperazine, 46 mg (91%) of 2-[2-ethoxy-5-(4-hydroxyethyl-1-amino-piperazine-1-sulphonyl)-phenyl]-5-methyl-7-propyl-3H-imidazo[5,1-f][1,2,4]triazin-4-one are obtained.

Rf=0.08 (dichloromethane/methanol=19:1)

200 MHz 1H-NMR (CDCl3): 1.02, t, 3H; 1.59, t, 3H; 1.90, sex., 2H; 2.49, m, 6H; 2.62, s, 3H; 2.71, m, 4H; 3.00, t, 2H; 3.55, t, 2H; 4.31, quart., 2H; 7.14, d, 1H; 8.05, dd, 1H; 8.60, d, 1H.

…………..

Methods of analysis

The development of lodenafil carbonate was reported by Toque et al. (2008). They observed the effects of lodenafil carbonate on rabbit and human corpus cavernosum relaxation, activity of PDE5 in human platelets, stability and metabolic studies in comparison with sildenafil and lodenafil, as well as the pharmacological evaluation of lodenafil carbonate after intravenous and oral administration in male beagles.

The determination of PDE activity, stability of lodenafil carbonate in human, dog and rat plasma and the pharmacokinetic parameters after a single intravenous or oral dose was carried out by LC-MS/MS analysis

Codevilla et al. (2011a) developed a stability-indicating reversed-phase liquid chromatography method using ultraviolet (UV) detection for the quantitative determination of lodenafil carbonate in tablets. The method can be useful for routine quality control assay and stability studies.

Another study for the determination of lodenafil carbonate in tablets was developed by Codevilla et al. (2011b). As an alternative to the LC method the authors suggested a UV-spectrophotometric method for the analysis of lodenafil carbonate in pharmaceutical form. The UV method offers advantages over other analytical methods due to its rapidity, simplicity, and lower cost. Recently, Codevilla et al. (2012) developed and validated a capillary zone electrophoresis (CZE) method for determination of lodenafil carbonate in drug products. There are some advantages to use the CZE method, such as rapid analysis, small sample and reagent consumption, high separation efficiency (Furlanetto et al., 2001; Yang et al., 2010). The results obtained from the UV-spectrophotometric method and CZE method were compared statistically with the LC method (Codevilla et al., 2011a) and the results showed no significant difference between these methods.

 

References

  1.  Toque HA, Teixeira CE, Lorenzetti R, Okuyama CE, Antunes E, De Nucci G (September 2008). “Pharmacological characterization of a novel phosphodiesterase type 5 (PDE5) inhibitor lodenafil carbonate on human and rabbit corpus cavernosum”. European Journal of Pharmacology 591 (1–3): 189–95. doi:10.1016/j.ejphar.2008.06.055PMID 18593576.
  2.  Cristália Product page. Retrieved on September 16, 2009.
  3.  ukmedix Lodenafil article. Retrieved on September 16, 2009.
  4.  Glina S, Toscano I, Gomatzky C, de Góes PM, Júnior AN, Claro JF, Pagani E (February 2009). “Efficacy and tolerability of lodenafil carbonate for oral therapy in erectile dysfunction: a phase II clinical trial”. The Journal of Sexual Medicine 6 (2): 553–7. doi:10.1111/j.1743-6109.2008.01079.x.PMID 19040623.
  5.  Glina S, Fonseca GN, Bertero EB, Damião R, Rocha LC, Jardim CR, Cairoli CE, Teloken C, Torres LO, Faria GE, da Silva MB, Pagani E (February 2010). “Efficacy and Tolerability of Lodenafil Carbonate for Oral Therapy of Erectile Dysfunction: A Phase III Clinical Trial”. The Journal of Sexual Medicine 7 (5): 1928–1936. doi:10.1111/j.1743-6109.2010.01711.xPMID 20214718.
  6. Toque H A et al., (2008) European Journal of Pharmacology, 591(1-3):189-95.
  7. Exploring the role of PDE5 inhibition in the treatment of muscular dystrophy
    Drugs Fut 2011, 36(4): 321

 

 

 

Share

Prediction of Drug Degradation Pathways leading to Structural Alerts for Potential Genotoxic Impurities

 regulatory, Uncategorized  Comments Off on Prediction of Drug Degradation Pathways leading to Structural Alerts for Potential Genotoxic Impurities
Jan 302014
 

Figure

An in-depth analysis of the web-based CambridgeSoft Pharmaceutical Drug Degradation Database, Pharma D3, was conducted in two phases in an attempt to generate some general rules for the prediction of alerting structures for genotoxicity that may arise as a result of degradation. The first phase involved interrogation of the database to determine the nature and frequency of alerting structures present in the degradants. This analysis revealed five functional groups, which account for approximately 70% of the alerting structures found in the degradants within the database: (1) aldehydes; (2) α,β unsaturated carbonyls; (3) aromatic amines, hydroxylamine and its derived esters; (4) epoxides; and (5) polyaromatic hydrocarbons. The second phase of the analysis involved categorizing the major chemical reactions responsible for the generation of the five most prevalent alerting structures. This two-step approach led, in turn, to a proposal for the prediction of functional groups that may have a propensity to degrade to alerting structures not necessarily present in the parent molecule.

Prediction of Drug Degradation Pathways leading to Structural Alerts for Potential Genotoxic Impurities

Stephen P. Raillard, Joel Bercu, Steven W. Baertschi and Christopher M. Riley
Org. Process Res. Dev., 2010, 14 (4), pp 1015–1020
Publication Date (Web): April 21, 2010 (Article)
DOI: 10.1021/op100007q
READ ALL AT
Share
Follow

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

Join other followers: