Showing posts with label organic chemistry. Show all posts
Showing posts with label organic chemistry. Show all posts

Monday 18 November 2013

Plerixafor , New treatment approaches for patients suffering from frequent bacterial infections


Plerixafor
17 nov 2013
Scientists at A*STAR's Singapore Immunology Network (SIgN) have discovered the exact mode of action by plerixafor, a drug commonly prescribed to stimulate immune responses in patients suffering from neutropenia, which causes them to become prone to oral, skin, genital infections and in worst cases, a fatal whole-body infection. A better understanding of the drug's mechanism can improve its usage to more effectively reduce risk of infections in these patients.
Scientists at SIgN employed cutting-edge imaging techniques to analyze the effects of plerixafor on the white blood cell activity in the study which was published in the Journal of Experimental Medicine (JEM).
Neutrophil Mobilization via Plerixafor
Neutropenia is a condition characterized by the lack of a type of white blood cells, also known as neutrophils, in one's blood circulation. Plerixafor increases the concentration of these white blood cells in the blood by inhibiting a protein called CXCR4. This inhibition prevents neutrophils in the blood stream from returning to the bone marrow, which is the primary compartment where the white blood cells are stored and released. It is therefore commonly accepted that the efficacy of the drug arises only from the release of these white blood cells from the bone marrow.
However, scientists at SIgN found that the inhibition of CXCR4 by the drug actually plays a dual role - It increases the neutrophil count in the blood through their release from the lungs, while simultaneously promoting their retention in the blood stream. Discovery of this additional mode of action not only provides a deeper understanding on the drug's mechanism, it also contributes to a more effective utilization of the drug. The ground-breaking study creates the possibility of using a combined drug treatment to maximise release of white blood cells from both the bone marrow and the lungs. The approach may be more effective in reducing the risk of bacterial infections in neutropenic patients.
The team leader, Dr Ng Lai Guan from SIgN said, "We have identified the precise mechanisms of plerixafor treatment, which has important implications on its usage. We can understand through this study the effectiveness or limitations of the drug on certain patients and thereafter craft new clinical approaches to better treat them. Our study forms a conceptual framework to establish improved therapeutic strategies for neutropenia."
Acting Executive Director of SIgN, Associate Professor Laurent Rénia, said, "Basic research as such is important for us to fully understand how drugs work, so that we can put them to best use. This is a study which can potentially be translated into clinical applications to impact the health and lives of neutropenic patients."
Plerixafor (rINN and USAN, trade name Mozobil) is an immunostimulant used to mobilize hematopoietic stem cells in cancer patients. The stem cells are subsequently transplanted back to the patient. The drug was developed by AnorMED which was subsequently bought by Genzyme.
Three of the four nitrogen atoms of the macrocycle 1,4,8,11-tetraazacyclotetradecan are protected with tosyl groups. The product is treated with 1,4-dimethoxybenzene or 1,4-bis(brommethyl)benzene and potassium carbonate in acetonitrile. After cleaving of the tosyl groups with hydrobromic acid, plerixafor octahydrobromide is obtained.Bridger, G.; et al. (1993). "Linked cyclic polyamines with activity against HIV. WO/1993/012096".
The molecule 1,1′-[1,4-phenylenebis(methylene)]bis [1,4,8,11-tetraazacyclotetradecane], consisting of two cyclam rings linked at the amine nitrogen atoms by a 1,4-xylyl spacer, was first synthesised by Fabbrizzi et al. in 1987 to carry out basic studies on the redox chemistry of dimetallic coordination compounds. Then, it was serendipitously discovered by De Clercq that such a molecule, could have a potential use in the treatment of HIV[2] because of its role in the blocking of CXCR4, a chemokine receptor which acts as a co-receptor for certain strains of HIV (along with the virus's main cellular receptor, CD4).Development of this indication was terminated because of lacking oral availability and cardiac disturbances. Further studies led to the new indication for cancer patients
Plerixafor has orphan drug status in the United States and European Union for the mobilization of hematopoietic stem cells. It was approved by the U.S. Food and Drug Administration for this indication on December 15, 2008. In Europe, the drug was approved after a positive Committee for Medicinal Products for Human Use assessment report on 29 May 2009. The drug was approved for use in Canada by Health Canada on December 8, 2011



Sunday 17 November 2013

DOLASETRON, anti-emetic drug.


DOLASETRON

(3R)-10-oxo-8-azatricyclo[5.3.1.03,8]undec-5-yl 1H-indole-3-carboxylate, 115956-12-2  CAS




Dolasetron (trade name Anzemet) is a serotonin 5-HT3 receptor antagonist used to treat nausea and vomiting following chemotherapy. Its main effect is to reduce the activity of the vagus nerve, which is a nerve that activates the vomiting center in the medulla oblongata. It does not have muchantiemetic effect when symptoms are due to motion sickness. This drug does not have any effect on dopamine receptors or muscarinic receptors.
Dolasetron breaks down slowly, staying in the body for a long time. One dose usually lasts 4 to 9 hours and is usually administered once or twice daily. This drug is removed from the body by the liver and kidneys.

  • Chemotherapy-induced nausea and vomiting
    • 5-HT3 receptor antagonists are the primary drugs used to treat and prevent chemotherapy-induced nausea and vomiting. Many times they are given intravenously about 30 minutes before beginning therapy.
  • Post-operative and post-radiation nausea and vomiting
  • Is a possible therapy for nausea and vomiting due to acute or chronic medical illness or acute gastroenteritis
  • Unlike most other 5HT3 antagonists, data is lacking for use of dolasetron with aprepitant in chemotherapy-induced nausea and vomiting (CINV).
  • It is also sometimes used as an antiemetic (anti-vomiting medication) in veterinary medicine for dogs and cats.

Dolasetron is a well-tolerated drug with few side effects. Headache, dizziness, and constipation are the most commonly reported side effects associated with its use. There is a potential for prolonging of the QT interval to occur as well. There have been no significant drug interactions reported with this drug's use. It is broken down by the liver's cytochrome P450 system and it has little effect on the metabolism of other drugs broken down by this system. Intravenous dolasetron is contraindicated in Chemotherapy-induced nausea and vomiting (CINV). Doxorubicin and cyclophosphamide are as emetogenic as cisplatin, and preventive drugs should always be considered. The 5HT3 agonists are the mainstays of prevention and are frequently used in combination with other drugs such as corticosteroids and the NK1 receptor antagonist aprepitant. However, the FDA recently issued a drug communication stating that the injection form of dolasetron, a 5HT3 agonist, should no longer be used in adult or pediatric patients with CINV. Dolasetron injection can increase the risk of developing torsade de pointes, a potentially fatal abnormal heart rhythm. Patients with underlying heart conditions or existing heart rate or rhythm problems are at increased risk. Although the oral form of this agent can still be used, careful monitoring and correction of potassium and magnesium levels should be initiated prior to and during treatment. In addition, in older patients and in patients with heart failure, a slow heart rate, underlying cardiac disease, and those with renal impairment, monitoring with electrocardiography is indicated when this drug is used. Congenital long-QT syndrome and drugs that prolong the PR or QRS interval are contraindications to dolasetron therapy. Dolasetron injection may still be used for the prevention and treatment of postoperative nausea and vomiting. As per Food and Drug Administration.


............
Dolasetron (14), available commercially as Anzement, is used as an anti-emetic that is especially useful to chemotherapy patients. Inventors J. A. P. Andres, P. D. Barjoan, and J. H. Clotet summarize several alternative processes for preparing 14 and state that all of them have shortcomings (e.g., the use of protecting groups or column chromatography) that make them unsuitable for commercial production. They disclose that novel esters 5 (R = Et or Me) are particularly useful in the synthesis of 14. These compounds and their synthesis are the subject of the patent and its claims.
The first sequence in the figure outlines the synthesis of 5 via the formation of 3 and 4. In the case of R = Et, the first step is the base catalyzed condensation of 1 with 2 to form 3 as a colorless oil in 75% yield after evaporating DMF, extracting into toluene, and washing in water. Further condensation of 2 with 3 produces 4, also recovered as an oil in 70% yield. In the final step, 4 is decarboxylated by heating it with NaBr in DMF, and 5is isolated in 80% yield as a colorless oil. For R = Me, 5 is prepared in a one-pot process without isolating intermediate 3.

The preparation of 14 from 5 (R = Me) requires several stages. The HCl salt of 9 is formed in the first stage as shown in the second sequence. Acid hydrolysis of 5 forms 6, which is not isolated but undergoes a Robinson–Schöpf condensation with 7 and 8 in the presence of Na2HPO4 to produce 9. The free base can be isolated by evaporating the solvent; alternatively, the HCl salt is obtained as a monohydrate after being crystallized from i-PrOH–H2O. X-ray diffraction and IR data for polymorph form I of this salt are given. The inventors describe this salt as a key aspect of the patent.
The next stage in the synthesis of 14 is shown in the third sequence; 9 is reduced with NaBH4 to give 10 in 76% isolated yield as a colorless oil. Esterification of 10 with 11 uses (CF3CO)2 and a catalytic amount of 4-dimethylaminopyridine to produce 12. This is isolated in 89% yield after being filtered and dried. It is used directly in the fourth sequence, or it can be converted to its HCl salt.
In the last stage, treating 12 with t-BuOK in a Dieckmann reaction forms 13. Heating with LiCl in DMF produces 14 by dealkoxycarboxylation. The free base is isolated in 83% yield and can be purified by methods previously reported (Anzeveno, P. B. Eur. Pat. EP0339669 [1989]).
1H NMR, 13C NMR, and IR data are given for all of the ethoxy and methoxy intermediates and final products. The patent provides a route to dolasetron that is the inventors claim is commercially viable. (INKE SA [Barcelona, Spain]. US Patent 7,858,821, Dec. 28, 2010;
.............

ANZEMET (dolasetron mesylate) is an antinauseant and antiemetic agent. Chemically, dolasetron mesylate is (2α,6α,8α,9aβ)-octahydro-3-oxo-2,6-methano-2H-quinolizin-8-yl-lH- indole-3-carboxylate monomethanesulfonate, monohydrate. It is a highly specific and selective serotonin subtype 3 (5-HT3) receptor antagonist both in vitro and in vivo. Dolasetron mesylate has the following structural formula:
Anzemet (dolasetron mesylate)  structural formula illustration
The empirical formula is C19H20N2O3 • CH3SO3H • H2O, with a molecular weight of 438.50.
Approximately 74% of dolasetron mesylate monohydrate is dolasetron base.
Dolasetron mesylate monohydrate is a white to off-white powder that is freely soluble in water and propylene glycol, slightly soluble in ethanol, and slightly soluble in normal saline.
ANZEMET Injection (dolasetron mesylate injection) is a clear, colorless, nonpyrogenic, sterile solution for intravenous administration. Each milliliter of ANZEMET Injection (dolasetron mesylate injection) contains 20 mg of dolasetron mesylate and 38.2 mg mannitol, USP, with an acetate buffer in water for injection. The pH of the resulting solution is 3.2 to 3.8.
ANZEMET Injection (dolasetron mesylate injection) multidose vials contain a clear, colorless, nonpyrogenic, sterile solution for intravenous administration. Each ANZEMET multidose vial contains 25 mL (500 mg) dolasetron mesylate. Each milliliter contains 20 mg dolasetron mesylate, 29 mg mannitol, USP, and 5 mg phenol, USP, with an acetate buffer in water for injection. The pH of the resulting solution is 3.2 to 3.7.
Synthesis of Dolasetron base is not very widely reported in literature. However, EP0266730/U.S. Pat. No. 4,906,755 describes process for the preparation endo-hexahydro-8-(3-indolylcarbonyloxy)-2,6-methano-2H-quinolizin-3(4H)-one methanesulfonate or Dolasetron mesylate, by the condensation of diethyl malonate with cis-1,4-dichloro-2-butene (2) in presence of lithium hydride in dimethylformamide to give diethyl-3-cyclopentene-1,1-dicarboxylate (3), which on hydrolysis and decarboxylation gave 3-cyclopentene-1-carboxylic acid (4). The compound (4) was further treated with thionyl chloride and pyridine in ethanol to obtain ethyl 3-cyclopentene-1-carboxylate (5). Compound (5) was oxidized to 4-ethoxycarbonyl-1,2-cyclopentanediol (6) by using N-methylmorpholine N-oxide in the presence of osmium tetroxide catalyst. The diol (6) was cleaved to the β-ethoxycarbonylglutaraldehyde (7) using sodium periodate and used directly in the next reaction. Robinson-Schopf cyclisation of the compound (7) with potassium hydrogen phthalate, acetonedicarboxylic acid and glycine ethyl ester hydrochloride resulted in the pseudopelletierine derivative i.e. 7-ethoxycarbonyl-9-(ethoxycarbonylmethyl)-9-azabicyclo-[3.3.1]nonan-3-one (8). The ketone group of compound (8) was reduced with sodiumborohydride in ethanol to give 7-ethoxycarbonyl-9-(ethoxycarbonylmethyl)-9-azabicyclo-[3.3.1]nonan-3-ol (9). The reduced alcohol (9) was treated with dihydropyran to protect the hydroxyl group as a tetrahydropyranyl ether (10). Dieckmann cyclisation of the compound (10) using strong base (potassium t-butoxide) followed by aqueous acid hydrolysis and decarboxylation gave the desired alcohol. The resulting alcohols can exist in two conformations—axial and equatorial. The main product obtained by above procedure was the axial alcohol or endo-hexahydro-8-hydroxy-2,6-methano-2H-quinolizin-3-(4H)-one (11) and it can be separated from the equatorial isomer by crystallization of the camphorsulfonate or tetrafluoroborate salt. The tetrafluoroborate salt of endo-hexahydro-8-hydroxy-2,6-methano-2H-quinolizin-3-(4H)-one (11) was further reacted with 3-indolecarboxylic acid chloride in presence of silver tetrafluoroborate in anhydrous nitroethane at −78° C. to endo-hexahydro-8-(3-indolylcarbonyloxy)-2,6-methano-2H-quinolizin-3(4H)-one or Dolasetron base, which was further converted into Dolasetron mesylate monohydrate (Scheme I) with a yield of 66%. No further purification is described.
The above process uses column chromatography for purification of compounds (9) and (10), which is expensive, time consuming and impractical on an industrial scale. The above patent does not disclose the yield and purity of Dolasetron mesylate obtained and so also for the intermediates. In addition, Osmium tetroxide used for preparation of compound (6) is toxic, has a corrosive action on eyes and hence difficult to use at industrial scale. Also this process uses high volume of water during preparation of the compound (8); preparation of compound (II) from compound (10) is tedious, because the workup involves several extractions with ethyl acetate and preparation of compound (I) in presence of silver tetrafluoroborate involves the use of expensive silver compound.
Figure US20080275241A1-20081106-C00002
Another method described in EP0339669 provides a process for the preparation of endo-hexahydro-8-(3-indolylcarbonyloxy)-2,6-methano-2H-quinolizin-3(4H)-one methanesulfonate or Dolasetron mesylate (1) by the condensation of dimethyl malonate with cis-1,4-dichloro-2-butene (2) in presence of lithium hydride in dimethyl formamide to give dimethyl-3-cyclopentene-1,1-dicarboxylate (12), which was decarbomethylated to obtain methyl-3-cyclopentene-1-carboxylate (13). This compound (13) was treated with m-chloroperbenzoic acid in dichloromethane to obtain 1-methoxycarbonyl-3-cyclopenteneoxide (14). The compound (13) on ozonolysis gave β-methoxycarbonylglutaraldehyde (15) or the epoxide (14) was reacted with periodic acid to obtain the β-methoxycarbonylglutaraldehyde (15), which was used directly in the next reaction. Robinson-Schopf cyclisation of the compound (15) with potassium hydrogen phthalate, acetonedicarboxylic acid and glycine ethyl ester hydrochloride gave the pseudopelletierine derivative i.e. 7-methoxycarbonyl-9-(methoxycarbonylethyl)-9-azabicyclo[3.3.1]nonan-3-one (16). The ketone group of compound (16) was reduced with sodiumborohydride in methanol to give 7-methoxycarbonyl-9-(methoxycarbonylmethyl)-9-azabicyclo-[3.3.1]nonan-3-ol (17). The reduced alcohol (17) was treated with dihydropyran to protect the hydroxyl group as a tetrahydropyranyl ether (18a) or treated with methylal to protect the hydroxyl group to obtain 3-methoxymethoxy-7-methoxycarbonyl-9-(methoxycarbonylmethyl)-9-azabicyclo[3.3.1]nonan-3-ol (18b).
Dieckmann cyclisation of the compound (18) using strong base (potassium t-butoxide) followed by aqueous acid hydrolysis and decarboxylation gave the endo-hexahydro-8-hydroxy-2,6-methano-2H-quinolizin-3-(4H)-one (11). The alcohol (11) was further reacted with 3-indolecarboxylic acid in presence of trifluoroacetic anhydride in dichloromethane to endo-hexahydro-8-(3-indolylcarbonyloxy)-2,6-methano-2H-quinolizin-3(4H)-one or Dolasetron base (A), which was then converted into Dolasetron mesylate (1) (not shown in Scheme II) by treating with methanesulphonicacid in acetone (Scheme II).
Figure US20080275241A1-20081106-C00003
Disadvantages of this process are:
    • (i) use of high volume of water for preparation of compound (16) and
    • (ii) preparation of compound (11) from compound (18) which is tedious because at the time of workup, ethyl acetate extractions take up longer period (20 h).
The process is not only time consuming but also expensive on an industrial scale. The patent does not disclose purity of Dolasetron base obtained nor for any of the intermediates.
The process as described in EP 0266730 involves treatment of endo-hexahydro-8-(3-indolylcarbonyloxy)-2,6-methano-2H-quinolizin-3(4H)-one (Dolasetron base) with a solution of methane sulfonic acid in ethanol to provide Dolasetron mesylate monohydrate. EP 0339669 describes crystallization of crude Dolasetron mesylate by dissolution in aqueous isopropanol and regeneration by adding ether. The polymorphic form obtained by the processes described in U.S. Pat. No. 4,906,755/EP 0266730 and EP 0339669 is designated herein as Dolasetron mesylate Form I.
The ability of the compound to exhibit more than one orientation or conformation of molecule within the crystal lattice is called polymorphism. Many organic compounds including active pharmaceutical ingredients (API's) exhibit polymorphism.
Drug substance existing in various polymorphic forms differs from each other in terms of stability, solubility, compressibility, flowability and spectroscopic properties, thus affecting dissolution, bioavailability and handling characteristics of the substance.
Rate of dissolution of an API's in patient's stomach fluid can have therapeutic consequences since it imposes an upper limit on the rate at which an orally administrated API can reach the patient bloodstream. Flowability affects the ease with which the material is handled while processing a pharmaceutical product.
Investigation of crystal polymorphism is an essential step in pharmaceutical research due to the influence of solid-state properties on dosage form.
As the polymorphs are known to possess different spectroscopic properties, technique such as X-Ray powder diffraction (XRPD), Fourier transformer Infrared (FT-IR) spectroscopy, Solid State 13C-NMR, and thermal method of analysis are keys to identify and characterize the new polymorphs or existing polymorphs.
The discovery of new polymorphs with same or better pharmaceutical equivalence and bioequivalence as that of the known polymorphs provides an opportunity to improve the performance characteristic of the pharmaceutical product.
The prior art describes isolation of endo-hexahydro-8-(3-indolylcarbonyloxy)-2,6-methano-2H-quinolizin-3(4H)-one or Dolasetron base as an oil. It is desirable to have the product in the solid form than oil, as solid is easy to handle and easy to purify.
Dolasetron base is isolated as a solid in EP 0339669. However, there is no evidence of polymorphism.
WO2006056081 discloses purification of Dolasetron base using strong acid especially methanesulphonic acid in presence of acid halide.



home business
back to home for more updates
ANTHONY MELVIN CRASTO
DR ANTHONY MELVIN CRASTO Ph.D
MOBILE-+91 9323115463
GLENMARK SCIENTIST , NAVIMUMBAI, INDIA


Onrigin, Laromustine, VNP40101M, an alkylating agent under investigation for treating high-grade brain tumors.

Cloretazine, Onrigin, VNP40101M, VNP-40101M, 101M, VNP 40101M, Laromustine [USAN], 173424-77-6 casno

108736-35-22, (2-chloroethyl)-N-methyl-1,2-bis(methylsulfonyl)hydrazinecarboxamide
Molecular Formula: C6H14ClN3O5S2   Molecular Weight: 307.77546

Vion Pharmaceuticals Logo.gif
Vion Pharmaceuticals, Inc. was a New HavenConnecticut based pharmaceutical company founded in March 1992 to commercialize several discoveries made in the biomedical laboratories at Yale University.

Two anticancer agents, Onrigin (laromustine), formerly cloretazine (VNP40101M), and Triapine, a ribunucloetide reductase inhibitor similar tohydroxyurea, were in human clinical trials. Onrigin (laromustine), a novel alkylating agent, was evaluated in a Phase 2 trial in elderly de novo poor-riskacute myeloid leukemia (AML). In addition, several trials of Onrigin (laromustine) were conducted in elderly patients with AML and myelodysplastic syndrome (MDS) in combination with cytarabine and in patients with brain tumors in combination with temozolomide. After Onrigin was rejected by the Food and Drug Administration for an AML indication in 2009 due to an unfavorable risk-benefit profile, the company went defunct.

Onrigin
  • Generic name: laromustine
  • Company: Vion Pharmaceuticals, Inc.
  • Treatment for: Acute Myeloid Leukemia
Onrigin (laromustine), formerly known as Cloretazine (VNP40101M), is a novel alkylating agent in development for remission induction treatment for patients sixty years of age or older with de novo poor-risk acute myeloid leukemia (AML).
VNP40101M, an anti-neoplasia agent, demonstrates potent anti-tumor activity through alkylation or cross-linking of deoxyribonucleic acid. The compound is classified as a sulfonylhydrazine prodrug, and the proposed mechanism of action suggests formation of a chloroethylating species with a relative specificity for O6-guanine alkylation. An additional decomposition product, methyl isocyanate, may inhibit various DNA repair enzymes (See, Penketh, et al. Biochem Pharmacol 2000, 59(3): 283-291).
A three-step process with a total yield less than 10% has been previously reported published in the following patents and journal articles: U.S. Pat. No. 5,637,619 (Jun. 10, 1997 for VNP40101M); U.S. Pat. No. 4,684,747 (Aug. 4, 1987 for intermediate VNP4090CE); WO97/02029 (Jan. 23, 1997); Journal of Medicinal Chemistry, 1990, 33(8): 2259-2264; Journal of Medicinal Chemistry, 1996, 39(3): 796-801, as illustrated in FIG. 1. 2-Hydroxyethylhydrazine (HEH) was used a starting material and reacted with methanesulfonyl chloride to obtain 1,2-bis(methylsulfonyl)-1-[2-(methylsulfonyloxy)ethyl]hydrazine (BMH) at −20° C. for 18 hours, using pyridine as base. The crude intermediate BMH was treated with lithium chloride in acetone under reflux for 3 days to afford 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine (VNP4090CE). After purification by flash column chromatography, an approximately 20% yield was reached for the two steps. VNP4090CE was condensed with methyl isocyanate at room temperature using triethylamine (TEA) as a base. After crystallization from ethanol, 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)-2-(methylcarbamoyl)hydrazine (VNP40101M) with an approximately 42% yield. The total yield of this process was less than 10%. It is noted that the process used methyl isocyanate, which is extremely dangerous for manufacturing in scaleup kilogram amounts

Onrigin (3; referred to in this patent as VNP40101M) is an alkylating agent under investigation for treating high-grade brain tumors. An existing process for preparing 3 gives a poor overall yield of 10%, and it uses methyl isocyanate (MeNCO), a toxic compound not favored by the pharmaceutical industry.
Inventors X. Lin and I. King report three syntheses of 3; the first is shown in Figure 1. This route proceeds through intermediate 2, which is obtained by treating compound 1with COCl2 in the presence of i-Pr2NEt. The intermediate is not isolated but treated with MeNH2 and additional i-Pr2NEt. The reaction is monitored by TLC and HPLC–UV. After workup and crystallization, 3 is isolated in 94% yield and 97% purity.
A second route to onrigin (Figure 2) starts from methanesulfonate 4, which is converted directly to 3 by the reaction with acyl chloride 5 in the presence of Et3N. After workup and crystallization, 3 is isolated in 67% yield and 97% purity. Another synthesis from 4proceeds through ester intermediate 7, formed by treating 4 with ester 6. Compound 7is isolated in 63% yield and then treated with MeNH2 in the presence of AlCl3 to give 3, isolated in 68% yield and 97% purity after crystallization.
The preparation of 4 and its conversion to 1 are outlined in Figure 3. The reaction of hydroxy hydrazine 8 with MsCl gives 4 in 58% isolated yield. The purity is not reported, but the inventors state that the 1H NMR spectrum is “clean”. Chloro compound 1 is prepared from 4 in a reaction with LiCl that requires 24 h. The product is isolated in 88% yield; again, the NMR spectrum is "clean” and identical to reported spectra.
Some of the experiments are carried out on the 500-g scale, suggesting the process’s advanced stage of development. Although the process avoids the dangers of using MeNCO, it does use extremely hazardous COCl2, which is only safe to use when it is generated onsite. (Nanotherapeutics Inc. [Alachua, FL]. US Patent 8,026,395, Sept. 27, 2011; )http://www.google.nl/patents/US8026395


Laromustine is a sulfonyl hydrazine prodrug with antineoplastic activity. Laromustine releases the DNA chloroethylating agent 90CE after entering the blood stream; 90CE chloroethylates alkylates the 06 position of guanine, resulting in DNA crosslinking, strand breaks, chromosomal aberrations, and disruption of DNA synthesis. Intracellular metabolism of this agent also releases methyl isocyanate which inhibits 06-alkyl-guanine transferase, an enzyme involved with DNA repair. Check for active clinical trialsor closed clinical trials using this agent. (NCI Thesaurus).
   
Current developer:   Vion Pharmaceuticals Inc

Saturday 16 November 2013

BI 201302 a 15-membered macrocyclic hepatitis C virus (HCV) protease inhibitor

Abstract Image

A highly convergent large scale synthesis of a 15-membered macrocyclic hepatitis C virus (HCV) protease inhibitor BI 201302 was achieved, in which the key features are the practical macrocyclization by Ru-catalyzed ring-closing metathesis (0.1 mol % Grela catalyst, 0.1–0.2 M concentration) and the efficient sulfone-mediated SNAr reaction.


Wei X * et al. Boehringer-Ingelheim Pharmaceuticals, Ridgeway, USA
A Highly Convergent and Efficient Synthesis of a Macrocyclic Hepatitis C Virus Protease Inhibitor BI 201302.

Org. Lett. 2013; 15: 1016-1019
Chemical Development, Boehringer-Ingelheim Pharmaceuticals, 900 Ridgebury Road, Ridgefield, Connecticut 06877, United States
Org. Lett.201315 (5), pp 1016–1019
DOI: 10.1021/ol303498m




Significance

The synthesis of the HCV protease inhibitor BI 201302 features an efficient ruthenium-catalyzed ring-closing metathesis reaction (0.1–0.2 M) requiring only 0.1 mol% of the Grela catalyst E to generate the 15-membered macrocycle F. This enhanced efficiency was achieved by installing a Boc substituent on the nitrogen of fragment D.


Comment

The SNAr reaction using a phenylsulfonyl leaving group in quinoline derivative H was more efficient than the reaction with the corresponding chloride (92% vs 40% yield). Potassium 3,7-dimethyl-3-octanoxide (KDMO) was used as a base instead of t-BuOK because transcarbamoylation byproducts (1–2%) were easily removed by crystallization.


Friday 1 November 2013

Gossypol a male antifertility agent with antispermatogenic activity and has been shown to contain anitumor, anitviral, and antioxidant properties

Gossypol

2,2-Bis(1,6,7-trihydroxy-3-methyl-5-isopropyl-8-formylnaphthalene)

This ridiculously named molecule is found in cotton seeds. It was used as a male contraceptive in China, but was never used in the West (and may have since been banned in China as well), since its effects were permanent in about 20% of patients! Its name originated from being present in the flowers of the Indian cotton plant Gossypium herbaceum L. Apart from its contraceptive effects, gossypol has properties that might make it useful in treating a number of ailments, including cancer, HIV, malaria and some bacterial/viral illnessness. Related to this molecule are the equally strangely named gossypetin and gossypin.

Gossypol is a natural phenol derived from the cotton plant (genus Gossypium). Gossypol is a phenolic aldehyde that permeates cells and acts as an inhibitor for several dehydrogenase enzymes. It is a yellow pigment.
Among other things, it has been tested as a male oral contraceptive in China. In addition to its contraceptive properties, gossypol has also long been known to possess antimalarial properties. Other researchers are investigating the anticancer properties of gossypol.
Biological properties

It has proapoptotic properties, probably due to the regulation of the Bax and Bcl2. It also reversibly inhibits calcineurin and binds to calmodulin. It inhibits replication of the HIV-1 virus.[1] It is an effective protein kinase C inhibitor.[2] It also causes low potassium levels, and thus causes temporary paralysis.

Biosynthesis

Gossypol is a terpenoid aldehyde, which is formed metabolically through acetate via the isoprenoid pathway.[3] Sesquiterpene dimer undergoes a radical coupling reaction to form gossypol.[4] Geranyl pyrophosphate (GPP) and IPP make sesquiterpene precursor, farnesyl diphosphate (FPP), for gossypol. The biosynthesis of gossypol is summarized in figure below. The biosynthesis begins with the 0 compound derived from GPP and IPP. Cadinyl cation (1) is oxidized to 2 by (+)-d-cadinene synthase. The (+)-d-cadinene (2) is involved in making the basic aromatic sesquiterpene unit, homigossypol, by oxidation, which generates the 3 (8-hydroxy-d-cadinene) with the help of (+)-d-cadinene 8-hyroxylase. At 5, the 3 goes through various oxidative processes to make 4 (deoxyhemigossypol), which is oxidized by one electron into hemigossypol (5,6,7) and then undergoes a phenolic oxidative coupling, ortho to the phenol groups, to form 8 (gossypol).[5] The coupling is catalyzed by a hydrogen peroxide-dependent peroxidise enzyme, which results in the final product.[5]
Gossypol biosyn.jpg

Contraceptive use

A 1929 investigation in Jiangxi showed correlation between low fertility in males and use of crude cottonseed oil for cooking. The compound causing the contraceptive effect was determined to be gossypol.
In the 1970s, the Chinese government began researching the use of gossypol as a contraceptive. Their studies involved over 10,000 subjects, and continued for over a decade. They concluded gossypol provided reliable contraception, could be taken orally as a tablet, and did not upset men's balance of hormones.
However, gossypol also had serious flaws. The studies also discovered an abnormally high rate of hypokalemia among subjects.[6] Hypokalemia — low blood potassium levels — causes symptoms of fatigue, muscle weakness, and at its most extreme, paralysis. In addition, about 7% of subjects[citation needed] reported effects on their digestive systems, and about 12%[citation needed] had increased fatigue. Most subjects recovered after stopping treatment and taking potassium supplements. The same study showed taking potassium supplements during gossypol treatment did not prevent hypokalemia in primates.[7] The potassium deficiency may also be a result of the Chinese diet or genetic predisposition. [8]
In the mid-1990s, the Brazilian pharmaceutical company Hebron announced plans to market a low-dose gossypol pill called Nofertil, but the pill never came to market. Its release was indefinitely postponed due to unacceptably high rates of permanent infertility[]. 5-25% of the men remained azoospermic up to a year after stopping treatment.[9] The longer the men had taken the drug and the higher their overall dosages, the more likely[citation needed] they were to have lowered fertility or to become completely infertile.
Researchers have suggested gossypol might make a good noninvasive alternative to surgical vasectomy.[10]
In 1986, the Chinese stopped research because of these side effects.
In 1998, the World Health Organization's Research Group on Methods for the Regulation of Male Fertility recommended the research should be abandoned. In addition to the other side effects, the WHO researchers were concerned about gossypol's toxicity: the toxic dose in primates is less than 10 times the contraceptive dose.[11]This report effectively ended further studies of gossypol as a temporary contraceptive, but research into using it as an alternative to vasectomy continues in AustriaBrazil,ChileChina, the Dominican Republic, and Nigeria.

First identified as an anti-fertility agent in China in the 1950s, gossypol is also a component of cottonseed oil, which is used for cooking. It's derived from the stems, roots, and seeds of plants from the Malvaceae family. The cotton plant (Gossypium species) is the most common source. Gossypol is the active ingredient found in seeds and other parts of the plant; however, content varies significantly from specjes to species.
Gossypol exerts antifertility action by inhibiting sperm production and motility. It possesses antitumorigenic activity and may also have anti-human immunodeficiency virus properties. It's available as liquid extracts and tinctures.
Reported uses
Gossypol is used in China as a male contraceptive. It's also used topically as a spermicide.
Administration
Dosage is 20 mg by mouth every day for 2 to 3 months until the sperm count is decreased to less than 4 million/ml. The dosage is reduced to a maintenance ranging from 50 mg weekly to 75 to 100 mg twice a month.
Hazards
Adverse effects associated with gossypol include paralysis, circulatory problems, diarrhea, malnutrition, hypokalemia, muscle fatigue, muscle weakness, and hair discoloration.
Pregnant and breast-feeding patients shouldn't use this herb. Patients with renal insufficiency should use with caution.
Safety Risk Gossypol has been associated with heart failure and nephrotoxicity. There's increased risk of nephrotoxicity when gossypol is given with nephrotoxic drugs. Administration with potassium-wasting diuretics could lead to hypokalemia.
Clinical considerations
The contraceptive effect of gossypol in men is higher than 99%. Fertility usually returns to normal within 3 months of discontinuation; however, inhibition of spermatogenesis may persist in up to 20% of men 2 years after discontinuation.
Monitor serum electrolyte levels, especially potassium, creatinine, and BUN levels.
Monitor patient for muscle weakness and fatigue.
If using formulation containing alcohol, avoid using in patients taking disulfiram, metronidazole, cephalosporins, or any CNS depressants.
If patient is pregnant or breast-feeding, advise her not to use gossypol.
Inform men of the potential for permanent sterility after using oral gossypol.

Food and animal agricultural industries must manage cotton-derivative product levels to avoid toxicity. For example, only ruminant microflora can digest gossypol, but only to a certain level, and cottonseed oil must be refined.
A research team at Texas A&M University has genetically engineered cotton plants that contain very little gossypol in the seed, but still contain the compound in the stems and leaves. This provides protection against pests and diseases, while allowing the seed to be used for oil and meal for human consumption. The plants are modified by RNA interference, shutting down the genes for gossypol production in the seed, while leaving them unaffected in the rest of the plant. The resulting gossypol-free cottonseed is then suitable as a high-quality protein source suitable for consumption not only by cattle, but also by humans. Protein makes up 23% of the cottonseed. [12][13]
Gossypol is toxic to erythrocytes in vitro by stimulating cell death contributing to the side effect of hemolytic anemia.[14]

References[edit]

  1. Jump up^ Polsky, B; Segal, SJ; Baron, PA; Gold, JW; Ueno, H; Armstrong, D (1989). "Inactivation of human immunodeficiency virus in vitro by gossypol". Contraception 39 (6): 579–87.doi:10.1016/0010-7824(89)90034-6PMID 2473865.
  2. Jump up^ Gossypol (Gossipol). Bioscreening.net (last updated 2008-07-09). Retrieved on 2012-06-09.
  3. Jump up^ Burgos, M.; Ito, S.; Segal, J. S.; Tran, T. P. (1997). "Effect of Gossypol on Ultrastructure of Spisula Sperm"Biol. Bull. 193: 228–229.
  4. Jump up^ Heinstein, P. F. ; Herman, L. D.; Tove, B. S.; Smith, H. F. (1970). "Biosynthesis of Gossypol". J. Biol. Chem. 245 (18): 4658–4665. PMID 4318479.
  5. Jump up to:a b Dewick, P. M. Medicinal Natural Product: A Biosynthetic approach. 3rd ed., 2008 ISBN 0-470-74167-8
  6. Jump up^ Gossypol. Malecontraceptives.org (2011-07-27). Retrieved on 2013-03-30.
  7. Jump up^ Gossypol. Malecontraceptives.org (2011-07-27). Retrieved on 2013-03-30.
  8. Jump up^ Gossypol. Malecontraceptives.org (2011-07-27). Retrieved on 2013-03-30.
  9. Jump up^ Gossypol. Malecontraceptives.org (2011-07-27). Retrieved on 2013-03-30.
  10. Jump up^ Coutinho, F. M. (2002). "Gossypol: a contraceptive for men". Contraception 65 (4): 259–263. doi:10.1016/S0010-7824(02)00294-9PMID 12020773.
  11. Jump up^ Gossypol. Malecontraceptives.org (2011-07-27). Retrieved on 2013-03-30.
  12. Jump up^ Cottonseed Protein: From Farmers to Your Family Table. Medgadget.com (2006-11-22). Retrieved on 2012-06-09.
  13. Jump up^ Walsh, Brian. Hungry? How About Some Protein-Rich Cotton..., Time Magazine, September 14, 2009, p. 54
  14. Jump up^ Zbidah, M; Lupescu, A; Shaik, N; Lang, F (2012). "Gossypol-induced suicidal erythrocyte death". Toxicology 302 (2–3): 101. doi:10.1016/j.tox.2012.09.010PMID 23041711.

  • Ibragimov, B. T.; Talipov, S. A.; Aripov, T. F.; Sadykov, A. S. (1990). "Inclusion complexes of the natural product gossypol. Crystal structure of the 2:1 complex of gossypol withm-Xylene". Journal of Inclusion Phenomena and Molecular Recognition in Chemistry 8 (3): 323. doi:10.1007/BF01041888.

Biosynthesis of gossypol from hemigossypol (Stipanovic et. al. 2005)

the compound is derived from hemigossypol through a free radical coupling reaction (Stipanovic et. al. 2005). This coupling reaction produces the two enantiomers (+)-gossypol and (-)-gossypol, depending on the orientation around the binaphthyl bond.
The toxicity of gossypol has made it an area of interest in plant research. When ingested orally, its toxicity can lead to anorexia, severe weight loss, and even death (Eagle 1949). The (-)-gossypol enantiomer will enter the cell and act as an inhibitor of dehydrogenase enzymes involved electron transport (Benz et. al. 1990), which ultimately leads to necrosis of the tissue.
There are also advantageous properties of gossypol to the cotton plant, especially in medicinal uses. (-)-gossypol is a potential aid in cancer research due to its inhibition of cell growth (Band et. al. 1989; Blackstaffe et. all 1997; Shelley et. al. 1999). This is accomplished by reduction of ATP by the uncoupling of oxidative phosphorylation in electron transport (Benz et. al. 1990). HIV research has shown that (-)-gossypol shows anti-HIV-1 activity (Lin et. al. 1993).This enantiomer can be used as an antiamoebic agent (Gonzales-Graza et. al. 1992). There have also been studies in China to suggest its use as a male contraceptive (Matlin et. al. 1985; Lindberg et. al. 1987; Wang et. al. 1987; Yao et. al. 1987).
Research shows that (+)-gossypol is useful to cotton as a natural protection against insects and pathogens. For example, Helicoverpa armigera larvae mature slowly and have a lower survival rate on cotton plants with (+)-gossypol (Yang et. al. 1999). It can also be consumed by nonruminant animals without expressing any signs of toxicity (Stipanovic et. al. 2005).
Cotton is well known as the leading fiber crop in the world (Lusas and Jividen 1987), but it's potential as a food crop has often been overlooked (Alford et. al. 1996). It has actually been classified as the second best potential source for plant proteins and the fifth best oil-producing plant (Texier 1993). Gossypol's toxicity restrains the potential of using cotton as a food crop, which is why emphasis has been placed on breeding for cotton with reduced amounts of gossypol in its seed (Vroh et. al. 1999). With low gossypol in the seed it can be safely used to produce cotton oil.


Zerp et al. Radiation Oncology 2009 4:47   doi:10.1186/1748-717X-4-47

Synonym: AT 101; 1,1',6,6',7,7'-hexahydroxy-5,5'-diisopropyl-3,3'-dimethyl-2,2'-binaphthyl-8,8'-dicarbaldehyde
Application: An inhibitor of PKC, Bcl-2, 5-LO, and 12-LO
CAS Number: 303-45-7
Purity: ≥90%
Molecular Weight: 518.56
Molecular Formula: C30H30O8
Set Currency Add Selected

Description
Gossypol is a male antifertility agent with antispermatogenic activity and has been shown to contain anitumor, anitviral, and antioxidant properties. Gossypol is a reversible inhibitor of PP2B (protein phosphatase 2B), mitotic kinesin Eg5, and PKC (protein kinase C). Gossypol also potentially inhibits PAF-R, Bcl-2, 5-LO (5-lipoxygenase), 12-LO, and leukotriene-induced guinea pig parenchyma contractions. Gossypol is an inhibitor of Bcl-xl, PKD and p107.
Technical Information
Appearance: Off-white to yellow crystalline solid
Physical State: Solid
Derived from: Gossypium genus, Malvaceae
Solubility: Soluble in 100%ethanol (25 mg/ml), DMF (25 mg/ml), acetone, DMSO (25 mM), methanol (2 mg/ml), ether, chloroform, sodium carbonate, and dilute aqueous solutions of ammonia . Insoluble in water.
Storage: Store at -20° C
Melting Point: 191-193 ºC
Boiling Point: 707.89 °C at 760 mmHg
Density: 1.40 g/cm3
Refractive Index: n20D 1.74
IC50: mitotic kinesin Eg5: IC50 = 10.8 µM; RBL-1, 5-lipoxygenase: IC50 = 0.3 µM; 12-lipoxygenase: IC50 = 0.7 µM; PC-3 (Prostate carcinoma cells) : EC5050 = 17.8 µM (Homo sapiens ); PC-3 (Prostate carcinoma cells) : EC5050 = 3.3 µM (Homo sapiens ); Transitional endoplasmic reticulum ATPase : IC50 = 5.39 µM (Homo sapiens ); Induced myeloid leukemia cell differentiation protein Mcl-1 : IC50 = 1.36 µM (Homo sapiens ); Estrogen receptor beta : IC50 >50 µM (Homo sapiens ); Apoptosis regulator Bcl-2 : IC50 = 0.5 µM (Homo sapiens ); HCT-116 (Colon carcinoma cells) : IC50 = 3.3 µM (Homo sapiens ); Malate dehydrogenase mitochondrial Malate dehydrogenase mitochondrial : IC50 = 2.8 µM (Sus scrofa ); MDA-MB-231 (Breast adenocarcinoma cells) : IC50 = 1.9 µM (Homo sapiens ); Lactate dehydrogenase : IC50 = 2.64 µM (Plasmodium falciparum ); Malate dehydrogenase : IC50 = 2.03 µM (Plasmodium falciparum )
Ki Data: Induced myeloid leukemia cell differentiation protein Mcl-1 : Ki= 0.18 µM (Homo sapiens ); Apoptosis regulator Bcl-X : Ki= 0.48 µM (Homo sapiens ); Apoptosis regulator Bcl-2 : Ki= 0.32 µM (Homo sapiens ); Aldose reductase : Ki= 0.5 µM (Homo sapiens ); Apoptosis regulator Bcl-2 : Ki= 0.17 µM (Homo sapiens ); Apoptosis regulator Bcl-W : Ki= 17.7 µM (Homo sapiens )


3,4-Dimethoxy-2-isopropylbenzaldehyde (I) could be condensed with diethyl succinate (II) to produce 2-(2-isopropyl-3,4-dimethoxybenzylidene)succinic acid monoethyl ester (III) in the presence of NaH in refluxing benzene, and the yielding product is cyclized by treatment with acetic anhydride and sodium acetate in refluxing acetic acid and hydrolyzed with NaOH in refluxing methanol affording acid (IV). Next, (IV) is reduced with LiAlH4 in ether to give 3-hydroxymethyl-5-isopropyl-6,7-dimethoxy-1-naphthol (V), and the compound produced is hydrogenated with H2 over Pd/C in methanol with some HCl affording 3-methyl-5-isopropyl-6,7-dimethoxy-1-naphthol (VI). Thermal dimerization of (VI) in the condition of heating at 215 C yields 2,2-bis(3-methyl-5-isopropyl-6,7-dimethoxy-1-naphthol) (VII).