AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

Antimicrobial resistance

 Uncategorized  Comments Off on Antimicrobial resistance
Aug 182014
 

 

Antimicrobials are medicines that kill or inactivate microbes, small disease-causing organisms. They include antibiotics, which are used against bacteria. After being exposed to an antimicrobial repeatedly, microbes can undergo changes that stop them being killed or inactivated by the treatments. This is known as antimicrobial resistance.

The European Medicines Agency is concerned about the development of antimicrobial resistance, particularly resistance to antibiotics. A well-known example of a bacterium that is resistant to a number of antibiotics is meticillin-resistant Staphylococcus aureus(MRSA), which has caused infections that are difficult to treat across the European Union (EU).

 

This problem is being made worse by the fact that few new antimicrobials have been authorised over the past few years. This may lead to infections becoming more difficult to treat in the future.

Antimicrobial resistance is a growing problem in humans and in animals. Resistance can also spread from animals to humans through the food chain or direct contact.

The role of the Agency

The Agency works in collaboration with its EU and international partners in a number of initiatives aiming to limit the development of resistance. It is also monitoring and evaluating the risks to human and animal health.

A major such initiative is the Transatlantic Task Force on Antimicrobial ResistanceExternal link icon(TATFAR), which was established following the EU-United States summit in November 2009. The Task Force aims to increase levels of communication, coordination and co-operation between the EU and the United States on human and veterinary antimicrobials.

 

Human health

In human medicine, the availability of medicines to treat infections with resistant organisms has become a major problem in recent years.

In September 2009, the Agency published a joint report together with the European Centre for Disease Prevention and ControlExternal link icon (ECDC) and the international network ReAct – Action on Antibiotic ResistanceExternal link icon. This report highlights the gap between infections due to resistant bacteria and the development of new antibiotics.

The report states that at least 25,000 patients in the EU die each year from infections due to bacteria that are resistant to many medicines, and that infections due to these bacteria in the EU result in extra healthcare costs and productivity losses of at least €1.5 billion each year. Although it identified 15 antibiotics under development, most of these were early in development and were targeted against bacteria for which treatment options were already available.

 

Authorisation of new antibiotics

The Agency plays a key role in the authorisation of new antibiotics, because medicines with a significant therapeutic innovation or that are in the interest of public or animal health are authorised centrally in the EU, on the recommendation of the Agency.

In January 2012, the Agency updated its guidance to companies developing antibiotics, covering how they should carry out studies to test these medicines’ benefits and risks:

This is accompanied by an addendum giving information on how to study medicines for specific indications. The final addendum was published in November 2013 following a public consultation:

 

Animal health

The Agency is focused on promoting the prudent use of antimicrobials in animals, to limit the development of resistance. This goal is addressed in this document:

In line with this strategy, the Agency published a revised version of its guideline onefficacy for public consultation in May 2013. This draft guideline provides detailed recommendations for the design and conduct of pre-clinical and clinical studies to support clinical efficacy for antimicrobial veterinary products:

Since early 2010, the Agency has been leading a project collecting information on the sale of veterinary antimicrobials across the EU:

The CVMP has also published a large number of documents on microbial resistance in animals and its risks for humans.

Reports published by the Agency together with other European bodies, including ECDC, EFSA and the European Commission’s Scientific Committee on Emerging and Newly Identified Health RisksExternal link icon (SCENIHR) have emphasised the need for the prudent use of antibiotics in animals and the role of basic hygiene, and called for strengthened surveillance of resistance, the development of new antimicrobials and new strategies to combat the spread of resistance:

In 2013 and 2014, the Agency carried out a large body of work to provide advice to the European Commission on the use of antibiotics in animals and the impact on public health and animal health.

Share

Turkish man pleads guilty to importing illegal cancer drugs

 cancer, Uncategorized  Comments Off on Turkish man pleads guilty to importing illegal cancer drugs
Aug 182014
 

 

August 15, 2014

Release

Sabahaddin Akman, owner of the Istanbul, Turkey, firm Ozay Pharmaceuticals, has pleaded guilty to charges of smuggling misbranded and adulterated cancer treatment drugs into the United States.

Akman pleaded guilty in the U.S. District Court for the Eastern District of Missouri, in St. Louis, Missouri, where he initially shipped his illegal drugs. The drugs did not meet the FDA’s standards and had not been approved for distribution in the United States.

The FDA’s Office of Criminal Investigations coordinated a complex, multi-layered international investigation that led to Akman’s arrest in Puerto Rico in January 2014. The investigation identified Akman and his company as a source of Altuzan, the Turkish version of the cancer treatment drug Avastin.

“These criminals exploited our most vulnerable patients when they arranged for their illicit drugs to be brought into the United States and used to treat cancer patients. We will continue to investigate and bring to justice those who prey on our ill, susceptible patients,” said Philip J. Walsky, acting director of the FDA’s Office of Criminal Investigations. “We commend our colleagues – international, national, state, and local – whose contributions helped bring this case to a successful conclusion.”

Akman, along with his employee, Ozkan Semizoglu, obtained the illicit drugs and then used shipping labels to conceal the illegal nature of the shipments, including customs declarations falsely describing the contents as gifts. They also broke large drug shipments into several smaller packages to reduce the likelihood of seizures by U.S. Customs and Border Protection authorities.

Along with the FDA and Europol, the international operation involved several German government offices: the Bonn prosecutor; the Federal Criminal Police, the Dusseldorf police, and the German State Criminal Police.  Special agents of the U.S. Department of State’s Diplomatic Security Service assigned to the U.S. Embassy’s Regional Security Office in Ankara, Turkey, and the U.S. Consulate General’s Overseas Criminal Investigations Branch in Istanbul, Turkey also played key roles in the successful resolution of this case.

Share

Road map to 2015, The European Medicines Agency’s contribution to science, medicines and health

 EU, regulatory, Uncategorized  Comments Off on Road map to 2015, The European Medicines Agency’s contribution to science, medicines and health
Aug 182014
 

 

One of the European Medicines Agency’s long-term strategic goals is to foster researchand the uptake of innovative methods in the development of medicines.

READ………….Road map to 2015

The European Medicines Agency’s
contribution to science, medicines and health……………..http://www.ema.europa.eu/docs/en_GB/document_library/Report/2011/01/WC500101373.pdf

This helps the Agency to meet its objective of making safe and effective medicines available to patients in a timely manner, following evaluation using state-of-the-art methods.

The Agency also supports the development of new therapies and technologies by working with interested parties in the European Union (EU).

Activities at the Agency

In 2004, the Agency set up the European Medicines Agency/Committee for Medicinal Products for Human Use (CHMP) Think-Tank Group on Innovative Drug Development.

This group included Agency staff and members of the CHMP and its working parties. Its work focused on identifying scientific bottlenecks and emerging science in the development of medicines, both in industry research and development and in academia, and on generating recommendations for future activities at the Agency:

In 2008 the EMA and its Scientific Committees integrated the recommendations made by the Think Tank in its strategy for supporting innovative medicines developments. Key areas of action included the strengthening of the EU scientific network model, emphasis on communication during the lifecycle of medicinal products development and international activities. Overview of measures implemented in the period 2008-2010.

The recently published Road Map to 2015 further expands on the role the Agency plays to promote innovation in pharmaceuticals.

The Agency also contributes to the Innovative Medicines InitiativeExternal link icon (IMI). This is a public-private initiative that aims to speed up the development of better and safer medicines for patients:

Support for business

The Agency provides support for business on issues related to innovative medicines:

Share

Lupin launches insulin glargine in India

 diabetes, Uncategorized  Comments Off on Lupin launches insulin glargine in India
Aug 182014
 

lupin ltd biosimilarnews Lupin launches insulin glargine in India

Lupin launches insulin glargine in India:

Indian pharma company, Lupin Limited announced a strategic distribution agreement with LG Life Sciences of South Korea to launch Insulin Glargine, a novel insulin analogue under the brand name Basugine™.

According to the agreement, Lupin would be responsible for marketing and sales of Basugine™ in India.

READ MORE

http://www.biosimilarnews.com/lupin-launches-insulin-glargine-in-india?utm_source=Biosimilar%20News%20%7C%20Newsletter&utm_campaign=0b76af10ab-15_08_2014_Biosimilar_News&utm_medium=email&utm_term=0_9887459b7e-0b76af10ab-335885197

Share

Celltrion files Remsima in the United States

 Uncategorized  Comments Off on Celltrion files Remsima in the United States
Aug 182014
 

 

Celltrion files Remsima in the United States:

Celltrion announced that the company, on August 8, 2014, completed the filing procedure to obtain US FDA approval for its infliximab biosimilar. This marks the first 351(k) biosimilar mAb application to be filed in the U.S.A. and the second application for a biosimilar to be filed through the US BPCIA.

READ MORE

http://www.biosimilarnews.com/celltrion-files-remsima-in-the-us?utm_source=Biosimilar%20News%20%7C%20Newsletter&utm_campaign=0b76af10ab-15_08_2014_Biosimilar_News&utm_medium=email&utm_term=0_9887459b7e-0b76af10ab-335885197

Share

Zopolrestat

 Uncategorized  Comments Off on Zopolrestat
Aug 182014
 

 

Chemical structure for zopolrestat

 

Zopolrestat

Zopolrestat
CAS : 110703-94-1
110765-49-6 (Na salt)
3,4-Dihydro-4-oxo-3-[[5-(trifluoromethyl)-2-benzothiazolyl]methyl]-1-phthalazineacetic acid
2- [4-Oxo-3- [5- (trifluoromethyl) benzothiazol-2-ylmethyl] -3,4-dihydrophthalazin-1-yl] acetic acid
3-(5-trifluoromethylbenzothiazol-2-ylmethyl)-4-oxo-3H-phthalazin-1-ylacetate
Pfizer Inc. INNOVATOR
2-[4-oxo-3-[5-(trifluoromethyl)benzothiazol-2-ylmethyl]-3,4-dihydrophthalazin-1-yl]acetic acid
Manufacturers’ Codes: CP-73850
MF: C19H12F3N3O3S
MW: 419.38
C 54.41%, H 2.88%, F 13.59%, N 10.02%, O 11.45%, S 7.65%
 Crystals, mp 197-198°. pKa (dioxane/water): 5.46 (1:1); 6.38 (2:1). Log P (n-octanol/water): 3.43.
 mp 197-198°
pKa: pKa (dioxane/water): 5.46 (1:1); 6.38 (2:1)
Log P: Log P (n-octanol/water): 3.43
Therap-Cat: Treatment of diabetic complications.
Keywords: Aldose Reductase Inhibitor.
…………………………..
synthesis
2-(8-oxo-7-((5-trifluromethyl)-1H-benzo[d]imidazol-2-yl)methyl)7,8-dihydropyrazin[2,3-d]pyridazin-5-yl)acetic acid and [4-oxo-(5-trifluoromethyl-benzothaiazol-2-ylmethyl)-3,4-dihydro-phthalazin-1-yl]-acetic acid (also known as zopolrestat), pharmaceutical compositions thereof and methods of treating diabetic complications in mammals comprising administering to mammals these salt and compositions. 2-(8-oxo-7-((5-trifluromethyl)-1H-benzo[d]imidazol-2-yl)methyl)8-dihydropyrazin[2,3-d]pyridazin-5-yl) acetic acid (formula II), is disclosed in WO 2012/009553 A1. Zopolrestat (formula III) is disclosed in U.S. Pat. No. 4,939,140.
Each of the patents, applications, and other references referred to herein are incorporated by reference. The diabetic complications include neuropathy, nephropathy, retinopathy, cataracts and cardiovascular complications, including myocardial infarction and cardiomyopathy. This invention is also directed to combinations of these salts and antihypertensive agents. These combinations are also useful in treating diabetic complications in mammals.
2-(8-oxo-7-((5-trifluoromethyl)-1H-benzo[d]imidazol-2-yl)methyl)8-dihydropyrazin[2,3-d]pyridazin-5-yl)acetic acid is prepared as disclosed in WO 2012/009553 A1, which is incorporated herein by reference. Zopolrestat is prepared as disclosed in U.S. Pat. No. 4,939,140.
…………………………
Zopolrestat can be obtained by several different ways: 1) The reaction of 2- (4-oxo-3,4-dihydrophthalazin-1-yl) acetic acid ethyl ester (I) with 2-chloroacetonitrile by means of potassium tert-butoxide in DMF gives 2- [3- (cyanomethyl) -4-oxo-3,4-dihydrophthalazin-1-yl] acetic acid ethyl ester (II), which is cyclized with 2-amino-4- (trifluoromethyl) thiophenol (III) in refluxing ethanol yielding zopolrestat ethyl ester (IV). Finally, this compound is hydrolyzed with KOH in methanol / water / THF. 2) Compound (IV) can also be obtained by cyclization of (II) with 4-chloro-3-nitrobenzotrifluoride . (V) in hot DMF saturated with H2S 3) Compound (II) can also be obtained as follows: The reaction of phthalazine (I) with aqueous formaldehyde gives 2- [3- (hydroxymethyl) -4-oxo-3,4 -dihydrophthalazin-1-yl] acetic acid ethyl ester (VI), which is treated with PBr3 in ethyl ether yielding the bromomethyl derivative (VII). Finally, this compound is treated with potassium cyanide and KI in acetone / water.
……………………….
5=CF3 IS SUBS
EXAMPLE 7

  • [0051]
    In accordance with Example 6, the following compounds are prepared:

    Figure imgb0011
    Figure imgb0012
    Figure imgb0013
……………………..
EXAMPLE 18 Sodium 3-(5-trifluoromethylbenzothiazol-2-ylmethyl)-4-oxo-3H-phthalazin-1-ylacetateSodium methoxide (54 mg) was added to 3-(5-trifluoromethylbenzothiazol-2-ylmethyl)-4-oxo-phthalazin-1-ylacetic acid (0.4 g) in methanol 10 ml) at room temperature. After the addition was complete, a clear solution was obtained which was stirred for 15 minutes at room temperature. The excess methanol was evaporated. The residue was triturated with ether (20 ml) and filtered to obtain the product (0.43 g; m.p. 300° C.).EXAMPLE 19 3-(5-Trifluoromethylbenzothiazol-2-ylmethyl)-4-oxo-3H-phthalazin-1-ylacetate, dicyclohexylamine saltTo a mixture of 3-(5-trifluromethylbenzothiazol-2ylmethyl)-4-oxo-phthalazin-1-ylacetic acid (0.42 g) in methanol (10 ml) was added dicyclohexylamine (0.2 g) in methanol (5 ml). The resulting clear solution was stirred at room temperature for 15 minutes and then evaporated to dryness. Trituration of the residue with ether (30 ml) gave a white solid (0.38 g; m.p. 207° C.).EXAMPLE 20 3-(5-Trifluoromethylbenzothiazol-2ylmethyl)-4-oxo-3H-phthalazin-1-ylacetic acid, meglumine saltA solution of 3-(5-trifluoromethylbenzothiazol-2-ylmethyl)-4-oxo-phthalazin-1-ylacetic acid (419 mg) and meglumine (196 mg) in methanol (50 ml) was stirred at room temperature for an hour and then evaporated to dryness. The residue was triturated with ether (25 ml), filtered and the collected solid was air dried (610 mg; m.p. 157° C.)……………………………

J. Med. Chem., 1991, 34 (1), pp 108–122
DOI: 10.1021/jm00105a018

http://pubs.acs.org/doi/abs/10.1021/jm00105a018

……………………………………

Mylari, Banavara L.; Zembrowski, William J.; Beyer, Thomas A.; Aldinger, Charles E.; Siegel, Todd W.
Journal of Medicinal Chemistry, 1992 ,  vol. 35,   12  p. 2155 – 2162

………………………………..

Mylari; Beyer; Scott; Aldinger; Dee; Siegel; Zembrowski
Journal of Medicinal Chemistry, 1992 ,  vol. 35,   3  p. 457 – 465

…………………………….

Literature References:
Aldose reductase inhibitor. Prepn: B. L. Mylari et al., EP 222576; E. R. Larson, B. L. Mylari, US 4939140(1987, 1990 both to Pfizer);
B. L. Mylari et al. J. Med. Chem. 34, 108 (1991).
Pharmacology: B. Tesfamariam et al., J. Cardiovasc.Pharmacol. 21, 205 (1993); B. Tesfamariam et al., Am. J. Physiol. 265, H1189 (1993).
Clinical pharmacokinetics: P. B. Inskeep et al., J. Clin. Pharmacol. 34, 760 (1994).
Zopolrestat < Rec INN; BAN; USAN >
Drugs Fut 1995, 20(1): 33
Synthesis of aldose reductase inhibitor, 3, 4-dihydro-4-oxo-3-[[5-(trifluoromethyl)-2 14C benzothiazolyl]methyl]-1-phthalazineacetic acid
J Label Compd Radiopharm 1991, 29(2): 143
3-19-1992
HETEROCYCLIC OXOPHTHALAZINYL ACETIC ACIDS
3-6-1992
3-(5-TRIFLUOROMETHYLBENZOTHIAZOL-2-YLMETHYL)-4-OXO-3H-PHYTHALAZIN-1-YLACETIC ACID MONOHYDRATE
7-4-1990
Heterocyclic oxophthalazinyl acetic acids
3-24-2006
Medical devices to treat or inhibit restenosis
12-30-2004
N-[(SUBSTITUTED FIVE-MEMBERED DI- OR TRIAZA DIUNSATURATED RING)CARBONYL]GUANIDINE DERIVATIVES FOR THE TREATMENT OF ISCHEMIA
10-7-2004
COMBINATION OF AN ALDOSE REDUCTASE INHIBITOR AND A GLYCOGEN PHOSPHORYLASE INHIBITOR COMBINATION OF AN ALDOSE REDUCTASE INHIBITOR AND A GLYCOGEN PHOSPHORYLASE INHIBITOR
9-30-2004
Aldose reductase inhibition in preventing or reversing diabetic cardiomyopathy
5-27-2004
SUBSTITUTED FUSED HETEROCYCLIC COMPOUNDS
4-15-2004
Compounds for treating and preventing diabetic complications
3-32-2004
IMPROVED MUTANTS OF (2,5-DKG) REDUCTASE A
12-18-2003
Pharmaceutical composition for use in treatment of diabetes
11-14-2003
Salts of zopolrestat
4-18-2002
Use of an aldose reductase inhibitor for reducing non-cardiac tissue damage
Share

REFLECTION PAPER ON NANOTECHNOLOGY-BASED MEDICINAL PRODUCTS FOR HUMAN USE

 nanotechnology, Uncategorized  Comments Off on REFLECTION PAPER ON NANOTECHNOLOGY-BASED MEDICINAL PRODUCTS FOR HUMAN USE
Aug 182014
 

Nanotechnology

Nanotechnology is the use of tiny structures – less than 1,000 nanometres across – that are designed to have specific properties. Nanotechnology is an emerging field in science that is used in a wide range of applications, from consumer goods to health products.

 

In medicine, nanotechnology has only partially been exploited. It is being investigated as a way to improve the properties of medicines, such as their solubility or stability, and to develop medicines that may provide new ways to:

  • deliver medicines to the body;
  • target medicines in the body more accurately;
  • diagnose and treat diseases;
  • support the regeneration of cells and tissues.

Activities at the European Medicines Agency 

The European Medicines Agency follows the latest developments in nanotechnology that are relevant to the development of medicines. Recommendations from the Agency’sCommittee for Medicinal Products for Human Use (CHMP) have already led to the approval of a number of medicines based on nanotechnology. These include medicines containing:

 

  • liposomes (microscopic fatty structures containing the active substance), such asCaelyx (doxorubicin), Mepact (mifamurtide) and Myocet (doxorubicin);
  • nano-scale particles of the active substance, such as Abraxane (paclitaxel), Emend(aprepitant) and Rapamune (sirolimus).

The development of medicines using newer, innovative nanotechnology techniques may raise new challenges for the Agency in the future. These include discussions on whether the current regulatory framework is appropriate for these medicines and whether existing guidelines and requirements on the way the medicines are assessed and monitored are adequate.

The Agency also needs to consider the acceptability of new testing methods and the availability of experts to guide the Agency’s opinion-making.

 

An overview of the initiatives taken by European Union (EU) regulators in relation to the development and evaluation of nanomedicines and nanosimilars was published in the scientific journal Nanomedicines. The article describes the regulatory challenges and perspectives in this field:

Ad hoc expert group on nanomedicines

In 2009, the CHMP established an ad hoc expert group on nanomedicines.

This group includes selected experts from academia and the European regulatory network, who support the Agency’s activities by providing specialist input on new scientific knowledge and who help with the review of guidelines on nanomedicines. The group also helps the Agency’s discussions with international partners on issues concerning nanomedicines.

The group held the first ad hoc expert group meeting on nanomedicines on 29 April 2009.

 

Reflection papers on nanomedicines

In 2011, the CHMP began to develop in 2011 a series of four reflection papers on nanomedicines to provide guidance to sponsors developing nanomedicines.

These documents cover the development both of new nanomedicines and of nanosimilars (nanomedicines that are claimed to be similar to a reference nanomedicine), since the first generation of nanomedicines, including liposomal formulations, iron-based preparations and nanocrystal-based medicines, have started to come off patent:

The fourth document, a draft reflection paper on the data requirements for intravenous iron-based nanocolloidal products developed with reference to an innovator medicine, will be released for a six-month public consultation in 2013.

International workshops on nanomedicines

The Agency organises workshops on nanomedicines to explore the scientific aspects of nanomedicines and enable the sharing of experience at an international level, in order to assist future developments in the field:

REFLECTION PAPER ON NANOTECHNOLOGY-BASED MEDICINAL PRODUCTS FOR
HUMAN USE

http://www.ema.europa.eu/docs/en_GB/document_library/Regulatory_and_procedural_guideline/2010/01/WC500069728.pdf

Related information

 

Share

Кальцитонин, Calcitonin

 Uncategorized  Comments Off on Кальцитонин, Calcitonin
Aug 162014
 

 

 

Molecular formula of calcitonin is C145H241N43O49S2
• Molecular weight is 3434.8 g/mol

Calcitonin-related polypeptide alpha
Calcitonin.png
NMR solution structure of salmon calcitonin in SDS micelles.[1]
Calcitonin
CAS Registry Number: 9007-12-9
Additional Names: Thyrocalcitonin; TCA; TCT
Therap-Cat: Calcium regulator.

 

The structural formula

 

Calcitonin (also known as thyrocalcitonin) is a 32-amino acid linear polypeptide hormone that is produced in humansprimarily by the parafollicular cells (also known as C-cells) of the thyroid, and in many other animals in the ultimobranchial body.[2] It acts to reduce blood calcium (Ca2+), opposing the effects of parathyroid hormone (PTH).[3]

Calcitonin has been found in fishreptilesbirds, and mammals. Its importance in humans has not been as well established as its importance in other animals, as its function is usually not significant in the regulation of normal calcium homeostasis.[4] It belongs to the calcitonin-like protein family.

UV – range

Conditions : Concentration – 53 mg / 100 ml
Solvent designation schedule
Methanol
Water
0.1М HCl
0.1M NaOH
The absorption maximum 278 nm 275 nm
4.9 4.4
with 1670 1500

 

 

 

IR – spectrum

Wavelength (μm)
Wavenumber (cm -1 )

 

Links

  • UV and IR Spectra. H.-W. Dibbern, R.M. Muller, E. Wirbitzki, 2002 ECV
  • NIST/EPA/NIH Mass Spectral Library 2008
  • Handbook of Organic Compounds. NIR, IR, Raman, and UV-Vis Spectra Featuring Polymers and Surfactants, Jr., Jerry Workman. Academic Press, 2000.
  • Handbook of ultraviolet and visible absorption spectra of organic compounds, K. Hirayama. Plenum Press Data Division, 1967.
Calcitonin-related polypeptide alpha
Calcitonin.png
NMR solution structure of salmon calcitonin in SDS micelles.[1]
Available structures
PDB Ortholog search: PDBeRCSB
[show]List of PDB id codes
Identifiers
Symbols CALCA ; CALC1; CGRP; CGRP-I; CGRP1; CT; KC
External IDs OMIM114130 MGI2151253HomoloGene88401 ChEMBL5293GeneCardsCALCA Gene
[show]Gene ontology
RNA expression pattern
PBB GE CALCA 210728 s at tn.png
PBB GE CALCA 210727 at tn.png
PBB GE CALCA 217495 x at tn.png
More reference expression data
Orthologs
Species Human Mouse
Entrez 796 12310
Ensembl ENSG00000110680 ENSMUSG00000030669
UniProt P01258 P70160
RefSeq (mRNA) NM_001033952 NM_001033954
RefSeq (protein) NP_001029124 NP_001029126
Location (UCSC) Chr 11:
14.99 – 14.99 Mb
Chr 7:
114.63 – 114.64 Mb
PubMedsearch [1] [2]

Biosynthesis and regulation

Calcitonin is formed by the proteolytic cleavage of a larger prepropeptide, which is the product of the CALC1 gene (CALCA). The CALC1 gene belongs to a superfamily of related protein hormone precursors including islet amyloid precursor proteincalcitonin gene-related peptide, and the precursor of adrenomedullin.

Secretion of calcitonin is stimulated by:

Effects

The hormone participates in calcium (Ca2+) and phosphorus metabolism. In many ways, calcitonin counteracts parathyroid hormone (PTH).

More specifically, calcitonin lowers blood Ca2+ levels in three ways:

However, effects of calcitonin that mirror those of PTH include the following:

  • Inhibits phosphate reabsorption by the kidney tubules[11]

In its skeleton-preserving actions, calcitonin protects against calcium loss from skeleton during periods of calcium mobilization, such as pregnancy and, especially, lactation.

Other effects are in preventing postprandial hypercalcemia resulting from absorption of Ca2+. Also, calcitonin inhibits food intake in rats and monkeys, and may have CNS action involving the regulation of feeding and appetite.

Receptor

The calcitonin receptor, found on osteoclasts,[12] and in kidney and regions of the brain, is a G protein-coupled receptor, which is coupled by Gs to adenylate cyclase and thereby to the generation of cAMP in target cells. It may also affect the ovaries in women and the testes in men.

Discovery

Calcitonin was purified in 1962 by Copp and Cheney.[13] While it was initially considered a secretion of the parathyroid glands, it was later identified as the secretion of the C-cellsof the thyroid gland.[14]

Pharmacology

Salmon calcitonin is used for the treatment of:

It has been investigated as a possible non-operative treatment for spinal stenosis.[16]

The following information is from the UK Electronic Medicines Compendium[17]

General characteristics of the active substance

Salmon calcitonin is rapidly absorbed and eliminated. Peak plasma concentrations are attained within the first hour of administration.

Animal studies have shown that calcitonin is primarily metabolised via proteolysis in the kidney following parenteral administration. The metabolites lack the specific biological activity of calcitonin. Bioavailability following subcutaneous and intramuscular injection in humans is high and similar for the two routes of administration (71% and 66%, respectively).

Calcitonin has short absorption and elimination half-lives of 10–15 minutes and 50–80 minutes, respectively. Salmon calcitonin is primarily and almost exclusively degraded in the kidneys, forming pharmacologically inactive fragments of the molecule. Therefore, the metabolic clearance is much lower in patients with end-stage renal failure than in healthy subjects. However, the clinical relevance of this finding is not known. Plasma protein binding is 30% to 40%.

Characteristics in patients

There is a relationship between the subcutaneous dose of calcitonin and peak plasma concentrations. Following parenteral administration of 100 IU calcitonin, peak plasma concentration lies between about 200 and 400 pg/ml. Higher blood levels may be associated with increased incidence of nausea, vomiting, and secretory diarrhea.

Preclinical safety data

Conventional long-term toxicity, reproduction, mutagenicity, and carcinogenicity studies have been performed in laboratory animals. Salmon calcitonin is devoid of embryotoxic, teratogenic, and mutagenic potential.

An increased incidence of pituitary adenomas has been reported in rats given synthetic salmon calcitonin for 1 year. This is considered a species-specific effect and of no clinical relevance. Salmon calcitonin does not cross the placental barrier.

In lactating animals given calcitonin, suppression of milk production has been observed. Calcitonin is secreted into the milk.

Pharmaceutical manufacture

Calcitonin was extracted from the ultimobranchial glands (thyroid-like glands) of fish, particularly salmon. Salmon calcitonin resembles human calcitonin, but is more active. At present, it is produced either by recombinant DNA technology or by chemical peptide synthesis. The pharmacological properties of the synthetic and recombinant peptides have been demonstrated to be qualitatively and quantitatively equivalent.[17]

Uses of calcitonin

Treatments

Calcitonin can be used therapeutically for the treatment of hypercalcemia or osteoporosis.

Oral calcitonin may have a chondroprotective role in osteoarthritis (OA), according to data in rats presented in December, 2005, at the 10th World Congress of the Osteoarthritis Research Society International (OARSI) in Boston, Massachusetts. Although calcitonin is a known antiresorptive agent, its disease-modifying effects on chondrocytes and cartilage metabolisms have not been well established until now.

This new study, however, may help to explain how calcitonin affects osteoarthritis. “Calcitonin acts both directly on osteoclasts, resulting in inhibition of bone resorption and following attenuation of subchondral bone turnover, and directly on chondrocytes, attenuating cartilage degradation and stimulating cartilage formation,” says researcher Morten Karsdal, MSC, PhD, of the department of pharmacology at Nordic Bioscience in Herlev, Denmark. “Therefore, calcitonin may be a future efficacious drug for OA.”[18]

Subcutaneous injections of calcitonin in patients suffering from mania resulted in significant decreases in irritability, euphoria and hyperactivity and hence calcitonin holds promise for treating bipolar disorder.[19] However no further work on this potential application of calcitonin has been reported.

Diagnostics

It may be used diagnostically as a tumor marker for medullary thyroid cancer, in which high calcitonin levels may be present and elevated levels after surgery may indicate recurrence. It may even be used on biopsy samples from suspicious lesions (e.g., lymph nodes that are swollen) to establish whether they are metastasis of the original cancer.

Cutoffs for calcitonin to distinguish cases with medullary thyroid cancer have been suggested to be as follows, with a higher value increasing the suspicion of medullary thyroid cancer:[20]

  • females: 5 ng/L or pg/mL
  • males: 12 ng/L or pg/mL
  • children under 6 months of age: 40 ng/L or pg/mL
  • children between 6 months and 3 years of age: 15 ng/L or pg/mL

When over 3 years of age, adult cutoffs may be used

Increased levels of calcitonin have also been reported for various other conditions. They include: C-cell hyperplasia, Nonthyroidal oat cell carcinoma, Nonthyroidal small cell carcinoma and other nonthyroidal malignancies, acute and chronic renal failure, hypercalcemia, hypergastrinemia and other gastrointestinal disorders, and pulmonary disease.[21]

Structure

Calcitonin is a polypeptide hormone of 32 amino acids, with a molecular weight of 3454.93 daltons. Its structure comprises a single alpha helix.[1] Alternative splicing of the gene coding for calcitonin produces a distantly related peptide of 37 amino acids, called calcitonin gene-related peptide (CGRP), beta type.[22]

The following are the amino acid sequences of salmon and human calcitonin:[23]

  • salmon:
      Cys-Ser-Asn-Leu-Ser-Thr-Cys-Val-Leu-Gly-Lys-Leu-Ser-Gln-Glu-Leu-His-Lys-Leu-Gln-Thr-Tyr-Pro-Arg-Thr-Asn-Thr-Gly-Ser-Gly-Thr-Pro
  • human:
      Cys-Gly-Asn-Leu-Ser-Thr-Cys-Met-Leu-Gly-Thr-Tyr-Thr-Gln-Asp-Phe-Asn-Lys-Phe-His-Thr-Phe-Pro-Gln-Thr-Ala-Ile-Gly-Val-Gly-Ala-Pro

Compared to salmon calcitonin, human calcitonin differs at 16 residues.

Description: Cellular and molecular coordination of tissues which secrete chemical compounds to regulate growth, reproduction, metabolism, and ion homeostasis.

 

 

 

 

References

  1. Jump up to:a b PDB 2glhAndreotti G, Méndez BL, Amodeo P, Morelli MA, Nakamuta H, Motta A (August 2006). “Structural determinants of salmon calcitonin bioactivity: the role of the Leu-based amphipathic alpha-helix”. J. Biol. Chem. 281 (34): 24193–203.doi:10.1074/jbc.M603528200PMID 16766525.
  2. Jump up^ Costoff A. “Sect. 5, Ch. 6: Anatomy, Structure, and Synthesis of Calcitonin (CT)”.Endocrinology: hormonal control of calcium and phosphate. Medical College of Georgia. Retrieved 2008-08-07.
  3.  Boron WF, Boulpaep EL (2004). “Endocrine system chapter”. Medical Physiology: A Cellular And Molecular Approach. Elsevier/Saunders. ISBN 1-4160-2328-3.
  4. Jump up^ Costoff A. “Sect. 5, Ch. 6: Biological Actions of CT”. Medical College of Georgia. Retrieved 2008-08-07.
  5. Costanzo, Linda S. (2007). BRS Physiology. Lippincott, Williams, & Wilkins. p. 263.ISBN 978-0-7817-7311-9.
  6. Jump up^ Erdogan MF, Gursoy A, Kulaksizoglu M (October 2006). “Long-term effects of elevated gastrin levels on calcitonin secretion”J Endocrinol Invest. 29 (9): 771–775.PMID 17114906.
  7.  Costoff A. “Sect. 5, Ch. 6: Effects of CT on the Small Intestine”. Medical College of Georgia. Retrieved 2008-08-07.
  8.  Costoff A. “Sect. 5, Ch. 6: Effects of CT on Bone”. Medical College of Georgia. Retrieved 2008-08-07.
  9. Jump up^ Potts, John; Jüppner, Harald (2008). “Chapter 353. Disorders of the Parathyroid Gland and Calcium Homeostasis”. In Dan L. Longo, Dennis L. Kasper, J. Larry Jameson, Anthony S. Fauci, Stephen L. Hauser, and Joseph Loscalzo. Harrison’s Principles of Internal Medicine (18 ed.). McGraw-Hill.
  10.  Rhoades, Rodney (2009). Medical Physiology: Principles for Clinical Medicine. Philadelphia: Lippincott Williams & Wilkins. ISBN 978-0-7817-6852-8.
  11. Jump up^ Carney SL (1997). “Calcitonin and human renal calcium and electrolyte transport”.Miner Electrolyte Metab 23 (1): 43–7. PMID 9058369.
  12. Jump up^ Nicholson GC, Moseley JM, Sexton PM, et al (1986). “Abundant calcitonin receptors in isolated rat osteoclasts. Biochemical and autoradiographic characterization”J Clin Invest 78 (2): 355–60. doi:10.1172/JCI112584PMC 423551PMID 3016026.
  13. Jump up^ Copp DH, Cheney B (January 1962). “Calcitonin-a hormone from the parathyroid which lowers the calcium-level of the blood”. Nature 193 (4813): 381–2.doi:10.1038/193381a0PMID 13881213.
  14. Jump up^ Hirsch PF, Gauthier GF, Munson PL (August 1963). “Thyroid hypocalcemic principle and recurrent laryngeal nerve injury as factors affecting the response to parathyroidectomy in rats”. Endocrinology 73 (2): 244–252. doi:10.1210/endo-73-2-244.PMID 14076205.
  15. Jump up^ Wall GC, Heyneman CA (April 1999). “Calcitonin in phantom limb pain”. Ann Pharmacother 33 (4): 499–501. doi:10.1345/aph.18204PMID 10332543.
  16. Jump up^ Tran de QH, Duong S, Finlayson RJ (July 2010). “Lumbar spinal stenosis: a brief review of the nonsurgical management”. Can J Anaesth 57 (7): 694–703. doi:10.1007/s12630-010-9315-3PMID 20428988.
  17. Jump up to:a b “Electronic Medicines Compendium”. Retrieved 2008-08-07.
  18. Jump up^ Kleinman DM (2006-01-04). “Oral Calcitonin May Delay Onset of Joint Disease and Relieve Pain of OA”Musculoskeletal Report. Musculoskeletal Report, LLC. Retrieved 2008-08-07.
  19. Jump up^ Vik A, Yatham LN (March 1998). “Calcitonin and bipolar disorder: a hypothesis revisited”J Psychiatry Neurosci 23 (2): 109–17. PMC 1188909PMID 9549251.
  20. Jump up^ Basuyau, J. -P.; Mallet, E.; Leroy, M.; Brunelle, P. (2004). “Reference Intervals for Serum Calcitonin in Men, Women, and Children”. Clinical Chemistry 50 (10): 1828–1830.doi:10.1373/clinchem.2003.026963PMID 15388660edit
  21. Jump up^ Burtis CA, Ashwood ER, Bruns DE. Tietz Textbook of Clinical Chemistry and Molecular Diagnostics, 5th edition. Elsevier Saunders. p. 1774. ISBN 978-1-4160-6164-9.
  22. Jump up^ “calcitonin domain annotation”SMART (a Simple Modular Architecture Research Tool). embl-heidelberg.de. Retrieved 2009-02-22.
  23. Jump up^ http://www.newworldencyclopedia.org/entry/Calcitonin

Further reading

External links

Literature References: 
Calcium regulating hormone secreted from the mammalian thyroid gland and in non-mammalian species from the ultimobranchial gland. Postulation of a plasma-calcium lowering substance: Copp et al., Endocrinology 70, 638 (1962).
Recognition as a hormone: Hirsch et al., ibid. 73, 244 (1963); of thyroid origin: Foster et al., Nature 202, 1303 (1964).
Over-all action is to oppose the bone and renal effects of parathyroid hormone, q.v.; inhibits bone resorption of Ca2+, with accompanying hypocalcemia and hypophosphatemia and decreased urinary Ca2+ concentrations. Also abolishes the osteolytic effect of toxic doses of vitamins A and D. Calcitonin is highly active biologically, e.g. 50 mg/min infused into a 100 g rat leads to a significant (1 mg/100 ml) decrease in the concn of the plasma calcium within 60 min (together with a corresponding fall in plasma phosphate). Activity is destroyed by trypsin, chymotrypsin, pepsin, polyphenol oxidase; also by hydrogen peroxide oxidation, photooxidation, and treatment with N-bromosuccinimide. Calcitonin structures are single polypeptide chains containing 32 amino acid residues. Structure of porcine: Neher et al., Helv. Chim. Acta 51, 917 (1968); Potts et al., Proc. Natl. Acad. Sci. USA 59, 1321 (1968); Bellet al., J. Am. Chem. Soc. 90, 2704 (1968); eidem, Biochemistry 9, 1665 (1970).
Synthesis of porcine: Rittel et al., Helv. Chim. Acta 51, 924 (1968); Guttmann et al., ibid. 1155.
Isoln of human calcitonin from non-pathological thyroid glands: Haymovits, Rosen, Endocrinology 81, 993 (1967); from medullary carcinoma of the thyroid: Neher et al., Nature 220, 984 (1968); Helv. Chim. Acta 51, 1738 (1968); Neher, Riniker, DE 1929957 (1970 to Ciba), C.A. 73, 28902b (1970).
Structure of human: Neher et al., Helv. Chim. Acta 51, 1900 (1968). Synthesis of human: Sieber et al., ibid. 2057; J. Hirt et al., Rec. Trav. Chim. 98, 143 (1979).
Biosynthetic studies: J. W. Jacobs et al., J. Biol. Chem. 254, 10600 (1979); S. G. Amara et al., ibid. 255, 2645 (1980).
Amino acid sequence differs among mammalian species, salmon calcitonin showing a marked difference from that of the higher vertebrae as well as a more potent biological activity. Mechanism of action: E. M. Brown, G. D. Aurbach, Vitam. Horm. 38, 236 (1980). Anorectic activity in rats: W. J. Freed et al., Science 206, 850 (1979).
Growth inhibition of human breast cancer cells in vitro: Y. Iwasaki et al., Biochem. Biophys. Res. Commun. 110, 235 (1983).
Review of early literature: Munson, Hirsch, Clin. Orthop. 49, 209 (1966).
Review of isoln, structure, synthesis: Behrens, Grinnan, Annu. Rev. Biochem. 38, 83 (1969); Potts et al., Vitam. Horm. 29,41 (1971).
Comprehensive review: Calcitonin, Proc. Symp. on Thyrocalcitonin and the C Cells, S. Taylor, Ed. (Springer-Verlag, New York, 1968); Foster et al., “Calcitonin” in Clinics in Endocrinology and Metabolism, I. MacIntyre, Ed. (W. B. Saunders, Philadelphia, 1972) pp 93-124.
Review of pharmacology and therapeutic use: J. C. Stevenson, I. M. A. Evans, Drugs 21, 257-272 (1981).
Derivative Type: Calcitonin, porcine
CAS Registry Number: 12321-44-7
Trademarks: Calcitar(e) (RPR); Staporos (Cassenne)
Derivative Type: Calcitonin, human synthetic
CAS Registry Number: 21215-62-3
Trademarks: Cibacalcin (Novartis)
Derivative Type: Calcitonin, salmon synthetic
CAS Registry Number: 47931-85-1
Additional Names: Salcatonin
Trademarks: Calciben (Firma); Calcimar (RPR); Calsyn (RPR); Calsynar (RPR); Catonin (Magis); Karil (Novartis); Miacalcic (Novartis); Miacalcin (Novartis); Miadenil (Francia); Osteocalcin (Tosi); Prontocalcin (Domp?; Rulicalcin (HMR); Salmotonin (Yamanouchi); Stalcin (Locatelli); Tonocalcin (Searle)
Literature References: Clinical trial in postmenopausal osteoporosis: C. H. Chesnut et al., Am. J. Med. 109, 267 (2000). LC determn in biological fluids: M. Aguiar et al.J. Chromatogr. B 818, 301 (2005).
Properties: See also Elcatonin.
eel
cas   57014-02-5
Share

HALOPERIDOL

 Uncategorized  Comments Off on HALOPERIDOL
Aug 152014
 

Haloperidol3DanJ.gif

 

 

Haloperidol /hælpɛridɒl/ (INNBANUSANAAN; most common brand names: HaldolSerenace) is an antipsychotic medication used in the treatment of schizophrenia, acute psychosismaniadelirium, tics in Tourette syndromechoreas, nausea and vomiting inpalliative care, intractable hiccups, agitation and severe anxiety.[3][4][5] Haloperidol is a butyrophenone derivative and functions as aninverse agonist of dopamine. It is classified as a typical antipsychotic and has pharmacological effects similar to the phenothiazines.[4]

A long-acting decanoate ester of haloperidol is used as an injection given every four weeks to people with schizophrenia or related illnesses who have poor adherence to medication regimens (most commonly due to them forgetting to take their medication, or due to poor insight into their illness) and suffer frequent relapses of illness, or to overcome the drawbacks inherent to its orally administered counterpart.[6] Such long acting injections are controversial because it can be seen as denying people their right to stop taking their medication.

It is on the World Health Organization’s List of Essential Medicines, a list of the most important medication needed in a basic health system.[7]

Skeletal formula of haloperidol decanoate: The decanoate group is highlighted in blue.

 

Brief background information

Salt ATC Formula MM CAS
N05AD01 21 H 23 ClFNO 2 375.87 g / mol 52-86-8

Application

  • neuroleptic
  • antidiskinetik
  • antipsychotic
  • dopamine antagonists

Classes of substances

  • Chloro alcohols
    • p-Ftorbutirofenony 4-piperidinyl derivatives
      • Piperidinol

Synthesis pathway

Synthesis a)


Trade Names

Country Trade name Manufacturer
Germany Haldol-Janssen Janssen-Cilag
various generic drugs
France Haldol Janssen-Cilag
United Kingdom – “- – “-
Serenak Ivax
Italy Haldol Janssen-Cilag
Serenas Lusofarmaco
Japan Galomont Janssen – Dainippon Sumitomo
Neoperidol Janssen
Serenak Dainippon Sumitomo
USA various generic drugs
Ukraine Haloperidol Ltd. “Gedeon Richter”, Hungary
various generic drugs

Formulations

  • ampoules of 5 mg / 1 ml, 100 mg / ml, 50 mg / ml;
  • drops of 2 mg to 20 mg / ml, 2 mg / ml, 0.5 mg / ml;
  • garnuly 1%;
  • Powder 1%;
  • 0.2% solution, 10 mg;
  • oral solution 2 mg / ml, 10 mg / ml;
  • Tablets of 0.75 mg, 1 mg, 1.5 mg, 2 mg, 3 mg, 5 mg, 10 mg, 20 mg

Links

  • Janssen, PAJ et al .: J. Med. Pharm. Chem. (JMPCAS) 1, 281 (1959).
  • DE 1289845 (Janssen; appl. 18/4/1959; GB -prior. 4.22.1958).
  • US 3,438,991 (Janssen; 4.15.1969; GB -prior. 18.11.1959).

 

1H NMR

13 C NMR

IR

 

 

 

MASS

http://www.scielo.br/scielo.php?script=sci_arttext&pid=S0100-40422012000200028

Systematic (IUPAC) name
4-[4-(4-Chlorophenyl)-4-hydroxy-1-piperidyl]-1-(4-fluorophenyl)-butan-1-one
Clinical data
Trade names Haldol
AHFS/Drugs.com monograph
MedlinePlus a682180
Pregnancy cat. (AU) C (US)
Legal status Prescription Only (S4) (AU) -only (CA) POM (UK) -only (US)
Routes Oral, IMIVdepot (asdecanoate ester)
Pharmacokinetic data
Bioavailability 60-70% (Oral)[1]
Protein binding ~90%[1]
Metabolism Liver-mediated[1]
Half-life 14-26 hours (IV), 20.7 hours (IM), 14-37 hours (oral)[1]
Excretion Biliary (hence in faeces) and in urine[1][2]
Identifiers
CAS number 52-86-8 Yes
ATC code N05AD01
PubChem CID 3559
IUPHAR ligand 86
DrugBank DB00502
ChemSpider 3438 Yes
UNII J6292F8L3D Yes
KEGG D00136 Yes
ChEBI CHEBI:5613 Yes
ChEMBL CHEMBL54 Yes
Chemical data
Formula C21H23ClFNO2 
Mol. mass 375.9 g/mol

History

Haloperidol was discovered by Paul Janssen.[70] It was developed in 1958 at the Belgian company Janssen Pharmaceutica and submitted to the first of clinical trials in Belgiumlater that year.[71]

Haloperidol was approved by the U.S. Food and Drug Administration (FDA) on April 12, 1967; it was later marketed in the U.S. and other countries under the brand name Haldol byMcNeil Laboratories.[citation needed]

Society and culture

Coincident with civil unrest in the United States in the 1960s and 1970s, schizophrenia was racialized to match the behavior of angry/violent black men. Haldol was promoted as a way to pacify them, and was marketed to appeal to feelings of racial unease. (cf. Metzl 2010. The Protest Psychosis)

Soviet dissidents, including medical staff, have reported several times on the use of haloperidol in the Soviet Union for punitive purposes or simply to break the prisoners’ will.[72][73][74] Notable dissidents who were administered haloperidol as part of their court-ordered treatment were Sergei Kovalev and Leonid Plyushch.[75] The accounts Plyushch gave in the West, after he was allowed to leave the Soviet Union in 1976, were instrumental in triggering Western condemnation of Soviet practices at the World Psychiatric Association‘s 1977 meeting.[76] The use of haloperidol in the Soviet Union’s psychiatric system was prevalent because it was one of the few psychotropic drugs produced in quantity in the USSR.[77]

Haloperidol has been used for its sedating effects during the deportations of immigrants by the United States Immigration and Customs Enforcement (ICE). During 2002-2008, federal immigration personnel used haloperidol to sedate 356 deportees. By 2008, following court challenges over the practice, it was given to only three detainees. Following lawsuits, U.S. officials changed the procedure so the drug is administered only by the recommendation of medical personnel and under court order.[78][79]

Brand names

Haloperidol is sold under the tradenames AloperidinBioperidoloBrotoponDozicDuraperidol (Germany), Einalon SEukystolHaldol (common tradename in the US and UK), HalostenKeselanLintonPelucesSerenace and Sigaperidol.

Veterinary use

Haloperidol is also used on many different kinds of animals. It appears to be particularly successful when given to birds, e.g., a parrot that will otherwise continuously pluck its feathers out.[80]

References

  1. Jump up to:a b c d e f g h i Kudo, S; Ishizaki T (December 1999). “Pharmacokinetics of haloperidol: an update”. Clinical pharmacokinetics 37 (6): 435-456. doi:10.2165/00003088-199937060-00001PMID 10628896.
  2. Jump up^ “PRODUCT INFORMATION Serenace” (PDF). TGA eBusiness Services. Aspen Pharma Pty Ltd. 29 September 2011. Retrieved 29 May 2014.
  3. Jump up^ Joint Formulary Committee (2013). British National Formulary (BNF) (65 ed.). London, UK: Pharmaceutical Press. p. 229-230. ISBN 978-0-85711-084-8edit
  4. Jump up to:a b Brayfield, A, ed. (13 December 2013). “Haloperidol”Martindale: The Complete Drug Reference. London, UK: Pharmaceutical Press. Retrieved 29 May 2014.
  5. Jump up^ “TGA Approved Terminology for Medicines” (PDF). Therapeutic Goods Administration. Australian Government, Department of Health and Ageing. July 1999. p. 66. Retrieved 29 May 2014.
  6. Jump up^ Rossi, S, ed. (2013). Australian Medicines Handbook (2013 ed.). Adelaide: The Australian Medicines Handbook Unit Trust. ISBN 978-0-9805790-9-3edit
  7. Jump up^ “WHO Model List of Essential Medicines” (PDF). World Health Organization. October 2013. p. 7, 35. Retrieved 22 April 2014.
  8. Jump up^ “Haldol Official FDA information, side effects and uses”. Drugs.com. Retrieved 2013-10-03.
  9. Jump up^ Giannini, A. James; Underwood, Ned A.; Condon, Maggie (2000). “Acute Ketamine Intoxication Treated by Haloperidol”. American Journal of Therapeutics 7 (6): 389–91.doi:10.1097/00045391-200007060-00008PMID 11304647.
  10. Jump up^ Giannini, A. James; Eighan, Michael S.; Loiselle, Robert H.; Giannini, Matthew C. (1984). “Comparison of Haloperidol and Chlorpromazine in the Treatment of Phencyclidine Psychosis”. The Journal of Clinical Pharmacology 24 (4): 202–4.doi:10.1002/j.1552-4604.1984.tb01831.xPMID 6725621.
  11. Jump up^ Cavanaugh, SV (1986). “Psychiatric emergencies”. The Medical clinics of North America 70 (5): 1185–202. PMID 3736271.
  12. Jump up^ Currier, Glenn W. (2003). “The Controversy over ‘Chemical Restraint’ In Acute Care Psychiatry”. Journal of Psychiatric Practice 9 (1): 59–70. doi:10.1097/00131746-200301000-00006PMID 15985915.
  13. Jump up^ Irving, Claire B; Adams, Clive E; Lawrie, Stephen (2006). “Haloperidol versus placebo for schizophrenia”. In Irving, Claire B. Cochrane Database of Systematic Reviews (4): CD003082. doi:10.1002/14651858.CD003082.pub2PMID 17054159.
  14. Jump up^ Allen, MH; Currier, GW; Hughes, DH; Reyes-Harde, M; Docherty, JP; Expert Consensus Panel for Behavioral Emergencies (2001). “The Expert Consensus Guideline Series. Treatment of behavioral emergencies”. Postgraduate Medicine (Spec No): 1–88; quiz 89–90. PMID 11500996.
  15. Jump up^ Allen, Michael H.; Currier, Glenn W.; Hughes, Douglas H.; Docherty, John P.; Carpenter, Daniel; Ross, Ruth (2003). “Treatment of Behavioral Emergencies: A Summary of the Expert Consensus Guidelines”. Journal of Psychiatric Practice 9 (1): 16–38. doi:10.1097/00131746-200301000-00004PMID 15985913.
  16. Jump up^ Allen, Michael H.; Currier, Glenn W.; Carpenter, Daniel; Ross, Ruth W.; Docherty, John P. (2005). “Introduction: Methods, Commentary, and Summary”. Journal of Psychiatric Practice 11: 5. doi:10.1097/00131746-200511001-00002.
  17. Jump up^ Ballard, Clive; Lana, Marisa Margallo; Theodoulou, Megan; Douglas, Simon; McShane, Rupert; Jacoby, Robin; Kossakowski, Katja; Yu, Ly-Mee; Juszczak, Edmund; on behalf of the Investigators DART AD (2008). “A Randomised, Blinded, Placebo-Controlled Trial in Dementia Patients Continuing or Stopping Neuroleptics (The DART-AD Trial)”. In Brayne, Carol. PLoS Medicine 5 (4): e76. doi:10.1371/journal.pmed.0050076.PMC 2276521PMID 18384230Lay summary – BBC News (April 1, 2008). “Neuroleptics provided no benefit for patients with mild behavioural problems, but were associated with a marked deterioration in verbal skills”
  18. Jump up to:a b c d e “Haldol Official FDA information, side effects and uses”. Drugs.com. Retrieved 2013-10-03.
  19. Jump up^ “Haloperidol at Chemeurope”.
  20. Jump up to:a b Work Group on Schizophrenia. “Practice Guideline for the Treatment of Patients With Schizophrenia Second Edition”. Retrieved 21 April 2014.
  21. Jump up^ Oosthuizen, P.; Emsley, R. A.; Turner, J.; Keyter, N. (2001). “Determining the optimal dose of haloperidol in first-episode psychosis”. Journal of Psychopharmacology 15 (4): 251–5. doi:10.1177/026988110101500403PMID 11769818.
  22. Jump up^ Tauscher, Johannes; Kapur, Shitij (2001). “Choosing the Right Dose of Antipsychotics in Schizophrenia”. CNS Drugs 15 (9): 671–8. doi:10.2165/00023210-200115090-00001.PMID 11580306.
  23. Jump up^ Goodman and Gilman’s Pharmacological Basis of Therapeutics, 10th edition (McGraw-Hill, 2001).[page needed]
  24. Jump up^ American Academy of Hospice and Palliative Medicine“Five Things Physicians and Patients Should Question”Choosing Wisely: an initiative of the ABIM Foundation(American Academy of Hospice and Palliative Medicine). Retrieved August 1, 2013., which cites
    • Smith, Thomas J.; Ritter, Joseph K.; Poklis, Justin L.; Fletcher, Devon; Coyne, Patrick J.; Dodson, Patricia; Parker, Gwendolyn (2012). “ABH Gel is Not Absorbed from the Skin of Normal Volunteers”. Journal of Pain and Symptom Management 43(5): 961–6. doi:10.1016/j.jpainsymman.2011.05.017PMID 22560361.
    • Weschules, Douglas J. (2005). “Tolerability of the Compound ABHR in Hospice Patients”. Journal of Palliative Medicine 8 (6): 1135–43.doi:10.1089/jpm.2005.8.1135PMID 16351526.
  25. Jump up^ PRODUCT INFORMATION [Internet]. 2011 [cited 2013 Sep 29]. Available from:https://www.ebs.tga.gov.au/ebs/picmi/picmirepository.nsf/pdf?OpenAgent&id=CP-2011-PI-03532-3
  26. Jump up^ HALDOL® Injection FOR INTRAMUSCULAR INJECTION ONLY PRODUCT INFORMATION [Internet]. Janssen; 2011 [cited 2013 Sep 29]. Available from:https://www.ebs.tga.gov.au/ebs/picmi/picmirepository.nsf/pdf?OpenAgent&id=CP-2009-PI-00998-3
  27. Jump up^ Truven Health Analytics, Inc. DrugPoint® System (Internet) [cited 2013 Sep 29]. Greenwood Village, CO: Thomsen Healthcare; 2013.
  28. Jump up^ Joint Formulary Committee. British National Formulary (BNF) 65. Pharmaceutical Pr; 2013.
  29. Jump up to:a b Leucht, Stefan; Cipriani, Andrea; Spineli, Loukia; Mavridis, Dimitris; Örey, Deniz; Richter, Franziska; Samara, Myrto; Barbui, Corrado; Engel, Rolf R; Geddes, John R; Kissling, Werner; Stapf, Marko Paul; Lässig, Bettina; Salanti, Georgia; Davis, John M (2013). “Comparative efficacy and tolerability of 15 antipsychotic drugs in schizophrenia: A multiple-treatments meta-analysis”. The Lancet 382 (9896): 951–62.doi:10.1016/S0140-6736(13)60733-3PMID 23810019.
  30. Jump up to:a b Silvestri, Simone; Seeman, Mary V.; Negrete, Juan-Carlos; Houle, Sylvain; Shammi, C.M.; Remington, Garry J.; Kapur, Shitij; Zipursky, Robert B.; Wilson, Alan A.; Christensen, Bruce K.; Seeman, Philip (2000). “Increased dopamine D 2 receptor binding after long-term treatment with antipsychotics in humans: A clinical PET study”.Psychopharmacology 152 (2): 174–80. doi:10.1007/s002130000532.PMID 11057521.
  31. Jump up^ Dorph-Petersen, Karl-Anton; Pierri, Joseph N; Perel, James M; Sun, Zhuoxin; Sampson, Allan R; Lewis, David A (2005). “The Influence of Chronic Exposure to Antipsychotic Medications on Brain Size before and after Tissue Fixation: A Comparison of Haloperidol and Olanzapine in Macaque Monkeys”. Neuropsychopharmacology 30 (9): 1649–61. doi:10.1038/sj.npp.1300710PMID 15756305.
  32. Jump up^ Konopaske, Glenn T.; Dorph-Petersen, Karl-Anton; Sweet, Robert A.; Pierri, Joseph N.; Zhang, Wei; Sampson, Allan R.; Lewis, David A. (2008). “Effect of Chronic Antipsychotic Exposure on Astrocyte and Oligodendrocyte Numbers in Macaque Monkeys”Biological Psychiatry 63 (8): 759–65.doi:10.1016/j.biopsych.2007.08.018PMC 2386415PMID 17945195.
  33. Jump up^ Vernon, Anthony C.; Natesan, Sridhar; Modo, Mike; Kapur, Shitij (2011). “Effect of Chronic Antipsychotic Treatment on Brain Structure: A Serial Magnetic Resonance Imaging Study with Ex Vivo and Postmortem Confirmation”. Biological Psychiatry 69 (10): 936–44. doi:10.1016/j.biopsych.2010.11.010PMID 21195390.
  34. Jump up^ Breggin, Peter R. (2007). “Neuroleptic-Induced Neurotoxicity, Brain Damage, Persistent Cognitive Deficits, Dementia, and Psychosis”Brain disabling treatments in psychiatry. Springer. pp. 85–114. ISBN 0-8261-2934-X.
  35. Jump up^ Halbreich, U; Shen, J; Panaro, V (1996). “Are chronic psychiatric patients at increased risk for developing breast cancer?”The American Journal of Psychiatry 153 (4): 559–60. PMID 8599407.
  36. Jump up^ Dalton, Susanne Oksbjerg; Mellemkjaer, Lene; Thomassen, Lars; Mortensen, Preben B.; Johansen, Christoffer (2005). “Risk for cancer in a cohort of patients hospitalized for schizophrenia in Denmark, 1969–1993”. Schizophrenia Research 75 (2–3): 315–24.doi:10.1016/j.schres.2004.11.009PMID 15885523.
  37. Jump up^ Grinshpoon, Alexander; Barchana, Micha; Ponizovsky, Alexander; Lipshitz, Irena; Nahon, Daniella; Tal, Orna; Weizman, Abraham; Levav, Itzhak (2005). “Cancer in schizophrenia: Is the risk higher or lower?”. Schizophrenia Research 73 (2–3): 333–41.doi:10.1016/j.schres.2004.06.016PMID 15653279.
  38. Jump up^ Szarfman, Ana; Tonning, Joseph M; Levine, Jonathan G; Doraiswamy, P. Murali (2006). “Atypical Antipsychotics and Pituitary Tumors: A Pharmacovigilance Study”.Pharmacotherapy 26 (6): 748–58. doi:10.1592/phco.26.6.748PMID 16716128.
  39. Jump up^ Hippisley-Cox, Julia; Vinogradova, Y; Coupland, C; Parker, C (2007). “Risk of Malignancy in Patients with Schizophrenia or Bipolar Disorder”. Archives of General Psychiatry 64 (12): 1368–76. doi:10.1001/archpsyc.64.12.1368PMID 18056544.
  40. Jump up^ Levav, Itzhak; Kohn, Robert; Barchana, Micha; Lipshitz, Irena; Pugachova, Inna; Weizman, Abraham; Grinshpoon, Alexander (2009). “The risk for cancer among patients with schizoaffective disorders”. Journal of Affective Disorders 114 (1–3): 316–20.doi:10.1016/j.jad.2008.06.010PMID 18675461.
  41. Jump up^ De Hert, M; Correll, CU; Bobes, J; Cetkovich-Bakmas, M; Cohen, D; Asai, I; Detraux, J; Gautam, S; Möller, HJ; Ndetei, DM; Newcomer, JW; Uwakwe, R; Leucht, S (2011).“Physical illness in patients with severe mental disorders. I. Prevalence, impact of medications and disparities in health care”World psychiatry 10 (1): 52–77.PMC 3048500PMID 21379357.
  42. Jump up^ Leentjens, Albert FG; Van Der Mast, Rose C (2005). “Delirium in elderly people: An update”. Current Opinion in Psychiatry 18 (3): 325–30.doi:10.1097/01.yco.0000165603.36671.97PMID 16639157.
  43. Jump up^ Sandyk, R; Hurwitz, MD (1983). “Toxic irreversible encephalopathy induced by lithium carbonate and haloperidol. A report of 2 cases”. South African medical journal 64 (22): 875–6. PMID 6415823.
  44. Jump up^ Bush, S. E.; Hatton, R. C.; Winterstein, A. G.; Thomson, M. R.; Woo, G. W. (2008). “Effects of concomitant amiodarone and haloperidol on Q-Tc interval prolongation”.American Journal of Health-System Pharmacy 65 (23): 2232–6.doi:10.2146/ajhp080039PMID 19020191.
  45. Jump up^ Igarashi, K.; Kasuya, F.; Fukui, M.; Usuki, E.; Castagnoli Jr, N. (1995). “Studies on the metabolism of haloperidol (HP): The role of CYP3A in the production of the neurotoxic pyridinium metabolite HPP+ found in rat brain following ip administration of HP”. Life Sciences 57 (26): 2439–46. doi:10.1016/0024-3205(95)02240-5PMID 8847965.
  46. Jump up^ Usuki, Etsuko; Pearce, Robin; Parkinson, Andrew; Castagnoli, Neal (1996). “Studies on the Conversion of Haloperidol and Its Tetrahydropyridine Dehydration Product to Potentially Neurotoxic Pyridinium Metabolites by Human Liver Microsomes”. Chemical Research in Toxicology 9 (4): 800–6. doi:10.1021/tx960001yPMID 8831826.
  47. Jump up^ Avent, Kathryn M.; Devoss, J. J.; Gillam, Elizabeth M. J. (2006). “Cytochrome P450-Mediated Metabolism of Haloperidol and Reduced Haloperidol to Pyridinium Metabolites”.Chemical Research in Toxicology 19 (7): 914–20. doi:10.1021/tx0600090.PMID 16841959.
  48. Jump up^ Avent, Kathryn M.; Riker, Richard R.; Fraser, Gilles L.; Van Der Schyf, Cornelis J.; Usuki, Etsuko; Pond, Susan M. (1997). “Metabolism of haloperidol to pyridinium species in patients receiving high doses intravenously: Is HPTP an intermediate?”. Life Sciences61 (24): 2383–90. doi:10.1016/S0024-3205(97)00955-7PMID 9399630.
  49. Jump up^ Kawashima, Hidekazu; Iida, Yasuhiko; Kitamura, Youji; Saji, Hideo (2004). “Binding of 4-(4-chlorophenyl)-1-[4-(4-fluorophenyl)-4-oxobutyl]pyridinium ion (HPP+), a metabolite of haloperidol, to synthetic melanin: Implications for the dopaminergic neurotoxicity of HPP+“. Neurotoxicity Research 6 (7–8): 535–42. doi:10.1007/BF03033449.PMID 15639785.
  50. Jump up to:a b Rollema, H; Skolnik, M; d’Engelbronner, J; Igarashi, K; Usuki, E; Castagnoli Jr, N (1994). “MPP(+)-like neurotoxicity of a pyridinium metabolite derived from haloperidol: In vivo microdialysis and in vitro mitochondrial studies”The Journal of Pharmacology and Experimental Therapeutics 268 (1): 380–7. PMID 8301579.
  51. Jump up^ Mythri, Rajeswara Babu; Jagatha, Balusamy; Pradhan, Nityananda; Andersen, Julie; Bharath, M. M. Srinivas (2006). “Mitochondrial Complex I Inhibition in Parkinson’s Disease: How Can Curcumin Protect Mitochondria?”. Antioxidants & Redox Signaling 9(3): 399–408. doi:10.1089/ars.2007.9.ft-25PMID 17184173.
  52. Jump up^ Beal, M. Flint (2006). “Mitochondria, Oxidative Damage, and Inflammation in Parkinson’s Disease”. Annals of the New York Academy of Sciences 991: 120–31.doi:10.1111/j.1749-6632.2003.tb07470.xPMID 12846981.
  53. Jump up^ Bishnoi, Mahendra; Chopra, Kanwaljit; Kulkarni, Shrinivas K. (2008). “Activation of striatal inflammatory mediators and caspase-3 is central to haloperidol-induced orofacial dyskinesia”. European Journal of Pharmacology 590 (1–3): 241–5.doi:10.1016/j.ejphar.2008.06.033PMID 18590723.
  54. Jump up^ Eyles, Darryl W.; Avent, Kathryn M.; Stedman, Terry J.; Pond, Susan M. (1997). “Two pyridinium metabolites of haloperidol are present in the brain of patients at post-mortem”.Life Sciences 60 (8): 529–34. doi:10.1016/S0024-3205(96)00656-XPMID 9042387.
  55. Jump up^ Ulrich, Sven; Neuhof, Sabine; Braun, Verena; Danos, Peter; Pester, Uwe; Hoy, Ludwig (2000). “Disposition of Haloperidol Pyridinium and Reduced Haloperidol Pyridinium in Schizophrenic Patients: No Relationship with Clinical Variables During Short-Term Treatment”. Journal of Clinical Psychopharmacology 20 (2): 210–9.doi:10.1097/00004714-200004000-00014PMID 10770460.
  56. Jump up^ Ulrich, S.; Sandmann, U.; Genz, A. (2005). “Serum Concentrations of Haloperidol Pyridinium Metabolites and the Relationship with Tardive Dyskinesia and Parkinsonism: A Cross-Section Study in Psychiatric Patients”. Pharmacopsychiatry 38 (4): 171–7.doi:10.1055/s-2005-871240PMID 16025420.
  57. Jump up^ “Haloperidol at Drugs.com”.
  58. Jump up^ Seeman, P; Tallerico, T (1998). “Antipsychotic drugs which elicit little or no Parkinsonism bind more loosely than dopamine to brain D2 receptors, yet occupy high levels of these receptors”. Molecular Psychiatry 3 (2): 123–34.doi:10.1038/sj.mp.4000336PMID 9577836.
  59. Jump up^ Schotte, A; Janssen PF; Megens AA; Leysen JE (1993). “Occupancy of central neurotransmitter receptors by risperidone, clozapine and haloperidol, measured ex vivo by quantitative autoradiography”Brain Research 631 (2): 191–202. doi:10.1016/0006-8993(93)91535-zPMID 7510574. Retrieved 21 April 2014.
  60. Jump up^ Leysen, JE; Janssen, PM; Gommeren, W; Wynants, J; Pauwels, PJ; Janssen, PA (1992). “In vitro and in vivo receptor binding and effects on monoamine turnover in rat brain regions of the novel antipsychotics risperidone and ocaperidone”Molecular Pharmacology 41 (3): 494–508. PMID 1372084.
  61. Jump up^ Malmberg, Åsa; Mikaels, Åsa; Mohell, Nina (1998). “Agonist and Inverse Agonist Activity at the Dopamine D3 Receptor Measured by Guanosine 5′-[γ-Thio]Triphosphate-[35S] Binding”The Journal of Pharmacology and Experimental Therapeutics 285 (1): 119–26. PMID 9536001.
  62. Jump up^ Leysen, JE; Janssen, PM; Megens, AA; Schotte, A (1994). “Risperidone: A novel antipsychotic with balanced serotonin-dopamine antagonism, receptor occupancy profile, and pharmacologic activity”. The Journal of Clinical Psychiatry 55 (Suppl): 5–12.PMID 7520908.
  63. Jump up^ Cobos, Enrique J.; Del Pozo, Esperanza; Baeyens, José M. (2007). “Irreversible blockade of sigma-1 receptors by haloperidol and its metabolites in guinea pig brain and SH-SY5Y human neuroblastoma cells”. Journal of Neurochemistry 102 (3): 812–25.doi:10.1111/j.1471-4159.2007.04533.xPMID 17419803.
  64. Jump up^ Colabufo, Nicolaantonio; Berardi, Francesco; Contino, Marialessandra; Niso, Mauro; Abate, Carmen; Perrone, Roberto; Tortorella, Vincenzo (2004). “Antiproliferative and cytotoxic effects of some σ2 agonists and σ1 antagonists in tumour cell lines”. Naunyn-Schmiedeberg’s Archives of Pharmacology 370 (2): 106–13. doi:10.1007/s00210-004-0961-2PMID 15322732.
  65. Jump up to:a b c d e f g h i j k Kroeze, Wesley K; Hufeisen, Sandra J; Popadak, Beth A; Renock, Sean M; Steinberg, Seanna; Ernsberger, Paul; Jayathilake, Karu; Meltzer, Herbert Y; Roth, Bryan L (2003). “H1-Histamine Receptor Affinity Predicts Short-Term Weight Gain for Typical and Atypical Antipsychotic Drugs”. Neuropsychopharmacology 28 (3): 519–26.doi:10.1038/sj.npp.1300027PMID 12629531.
  66. Jump up^ Ilyin, VI; Whittemore, ER; Guastella, J; Weber, E; Woodward, RM (1996). “Subtype-selective inhibition of N-methyl-D-aspartate receptors by haloperidol”Molecular Pharmacology 50 (6): 1541–50. PMID 8967976.
  67. Jump up^ “drugs.com”.
  68. Jump up to:a b Kornhuber, Johannes; Schultz, Andreas; Wiltfang, Jens; Meineke, Ingolf; Gleiter, Christoph H.; Zöchling, Robert; Boissl, Karl-Werner; Leblhuber, Friedrich; Riederer, Peter (1999). “Persistence of Haloperidol in Human Brain Tissue”The American Journal of Psychiatry 156 (6): 885–90. PMID 10360127.
  69. Jump up^ Kornhuber, Johannes; Wiltfang, Jens; Riederer, Peter; Bleich, Stefan (2006). “Neuroleptic drugs in the human brain: Clinical impact of persistence and region-specific distribution”. European Archives of Psychiatry and Clinical Neuroscience 256 (5): 274–80. doi:10.1007/s00406-006-0661-7PMID 16788768.
  70. Jump up^ Healy, David (1996). The psychopharmacologists 1. London: Chapman and Hall.ISBN 978-1-86036-008-4.[page needed]
  71. Jump up^ Granger, Bernard; Albu, Simona (2005). “The Haloperidol Story”. Annals of Clinical Psychiatry 17 (3): 137–40. doi:10.1080/10401230591002048PMID 16433054.
  72. Jump up^ Podrabinek, Aleksandr (1980). Punitive Medicine. Ann Arbor Mich.: Karoma Publishers. pp. 15–20. ISBN 0-89720-022-5.
  73. Jump up^ Kosserev, I.; Crawshaw, R. (1994). “Medicine and the Gulag”BMJ 309 (6970): 1726–30. doi:10.1136/bmj.309.6970.1726PMC 2542687PMID 7820004.
  74. Jump up^ de Boer, S. P.; E. J. Driessen; H. L. Verhaar (1982). Biographical Dictionary of Dissidents in the Soviet Union, 1956-1975. The Hague: Martinus Nijhoff Publishers.ISBN 90-247-2538-0.[page needed]
  75. Jump up^ Wade, N. (1976). “Sergei Kovalev: Biologist Denied Due Process and Medical Care”.Science 194 (4265): 585–7. doi:10.1126/science.194.4265.585PMID 17818411.
  76. Jump up^ “Censuring the Soviets”TIME (CNN). 1977-09-12. Retrieved 2009-06-21.
  77. Jump up^ The Children of Pavlov, TIME, Jun. 23, 1980
  78. Jump up^ “Fewer US deportees being sedated for removal”. Epilepsy.com. Associated Press. 2008-12-30. Retrieved 2009-06-21.
  79. Jump up^ Solis, Dianne (2009-01-05). “U.S. cuts back on sedating deportees with Haldol”. Seattle Times. Retrieved 2009-06-21.
  80. Jump up^ “Veterinary:Avian at Lloyd Center Pharmacy”.

External links

 

Share

Pantoprazole sodium

 Uncategorized  Comments Off on Pantoprazole sodium
Aug 132014
 

 

Chemical structure for pantoprazole

Protonix; Pantoprazolum; Pantoprazol; Pantozol; 102625-70-7; Pantoprazole Sodium; BY-1023; Pantoloc
Molecular Formula: C16H15F2N3O4S   Molecular Weight: 383.369806

 

Pantoprazole
Pantoprazole.svg
Systematic (IUPAC) name
(RS)-6-(Difluoromethoxy)-2-[(3,4-dimethoxypyridin-2-yl)methylsulfinyl]-1H-benzo[d]imidazole
Clinical data
Trade names Protonix
AHFS/Drugs.com monograph
MedlinePlus a601246
Licence data US FDA:link
Pregnancy cat. B3 (AU) B (US)
Legal status ℞ Prescription only
Routes Oral and intravenous
Pharmacokinetic data
Bioavailability 77%
Metabolism Hepatic (CYP3A4)
Half-life 1 hour
Excretion Renal
Identifiers
CAS number 102625-70-7 Yes
ATC code A02BC02
PubChem CID 4679
DrugBank DB00213
ChemSpider 4517 Yes
UNII D8TST4O562 Yes
KEGG D05353 Yes
ChEBI CHEBI:7915 Yes
ChEMBL CHEMBL1502 Yes
Chemical data
Formula C16H15F2N3O4S 
Mol. mass 383.371 g/mol

Pantoprazole is a proton pump inhibitor drug that inhibits gastric acid secretion.

Pantoprazole is a proton pump inhibitor drug used for short-term treatment of erosion and ulceration of the esophagus caused by gastroesophageal reflux disease.

Use

Pantoprazole is used for short-term treatment of erosion and ulceration of the oesophagus caused by gastroesophageal reflux disease. Initial treatment is generally of eight weeks’ duration, after which another eight week course of treatment may be considered if necessary. It can be used as a maintenance therapy for long term use after initial response is obtained.

Adverse effects

Antacid preparations such as pantoprazole work by suppressing the acid-mediated breakdown of proteins. This leads to an elevated risk of developing food and drug allergies due to undigested proteins passing into the gastrointestinal tract where sensitisation occurs. It is unclear whether this risk occurs with short-term or only long-term use.[1]

Common

  • Gastrointestinal: Abdominal pain (3%), diarrhea (4%), flatulence (4%)
  • Neurologic: Headache (5%)

Serious

  • Gastrointestinal: Atrophic gastritisclostridium difficile diarrhea
  • Hematologic: Thrombocytopenia (less than 1%)
  • Immunologic: Stevens-Johnson syndrometoxic epidermal necrolysis
  • Musculoskeletal: Muscle disorders, bone fracture and infection, Clostridium difficile, osteoporosis-related, hip fracture,rhabdomyolysis
  • Renal: Interstitial nephritis (rare)
  • Nutrition: May reduce the absorption of important nutrients, vitamins and minerals, as well as medications, leaving users at increased risk for pneumonia.[2]
  • Cardiovascular: Increase in a chemical that suppresses the production of nitric oxide by 25% in humans, which have proven to relax and protect arteries and veins. Causes blood vessels to constrict, a development that could lead to a number of cardiovascular problems if continued for a prolonged period of time.[2]

Pharmacology

Wyeth pantoprazole 20mg.

Pantoprazole is metabolized in the liver by the cytochrome P450 system.[3] Metabolism mainly consists of demethylation by CYP2C19followed by sulfation. Another metabolic pathway is oxidation by CYP3A4. Pantoprazole metabolites are not thought to have any pharmacological significance. Pantoprazole is relatively free of drug interactions;[4] however, it may alter the absorption of other medications that depend on the amount of acid in the stomach, such as ketoconazole or digoxin. Generally inactive at acidic pH of stomach, thus it is usually given with a pro kinetic drug. Pantoprazole binds irreversibly to H+K+ATPase (proton pumps) and suppresses the secretion of acid. As it binds irreversibly to the pumps, new pumps have to be made before acid production can be resumed. The drug’s plasma half-life is about 2 hours.[5]

Pharmacokinetics

Absorption

  • Bioavailability: (oral, delayed release tablets), approximately 77%
  • Effect of food: (oral, delayed-release tablets), AUC and Cmax no effect, Tmax variable, absorption delayed, no net effect
  • Effect of food: (oral, for-delayed-release suspension), administer 30 minutes before a meal
  • Tmax, Oral, delayed-release suspension: 2 to 2.5 h
  • Tmax, Oral, delayed-release tablets: 2.5 h
  • Tmax, Oral, delayed-release tablets: 1.5 to 2 hours (pediatrics)

Distribution

  • Protein binding: about 98% to primarily albumin
  • Vd, extensive metabolizers (IV): approximately 11 L to 23.6 L
  • Vd, pediatrics (oral): 0.21 to 0.43 L/kg.

Metabolism

  • Hepatic; cytochrome P450 CYP2C19; minor metabolism from CYP3A4, 2D6, and 2C9

Excretion

  • Fecal: (oral or IV, normal metabolizers), 18%
  • Renal: (oral or IV, normal metabolizers), approximately 71%, none as unchanged
  • Dialyzable: no (hemodialysis)
  • Total body clearance: (IV) 7.6 to 14 L/hour.
  • Total body clearance: (oral, pediatrics) 0.18 to 2.08 L/h/kg

Elimination Half Life

  • Oral or IV, 1 hour
  • Oral or IV, slow metabolizers, 3.5 to 10 hours
  • Pediatrics, 0.7 to 5.34 hours

Availability

Pantoprazole was developed by Altana (owned by Nycomed) and was licensed in the USA to Wyeth (which was taken over by Pfizer). It was initially marketed under the brand name Protonix by Wyeth-Ayerst Laboratories and now is available as a generic. It is available by prescription in delayed-release tablets. It is also available for intravenous use.

On 24 December 2007, Teva Pharmaceutical released an AB-rated generic alternative to Protonix.[6] This was followed by generic equivalents from Sun Pharma and Kudco Pharma. Wyeth sued all three for patent infringement and launched its own generic version of Protonix with Nycomed.[7][8]

On October 18, 2010 the U.S. Food and Drug Administration (FDA) accepted the filing of an ANDA for a delayed release generic version of Protonix by Canadian companyIntelliPharmaCeutics.[9]

Brand names

Pantoprazole is available from a range of international suppliers under brand names including Pantazone, Pantop-D, Pantasan, Pantrol, Prazolin, Pantochem, Pansev, Pantec, Somac, API, Tecta, Protium, Pantodac, Perizole, Pansped, Percazole, Astropan, Fenix, Pantecta, Pantoloc, Controloc, Somac, Tecta, Protium, Inipomp, Eupantol, Pantozol, Pantodac, Perizole, Pansped, Zurcazol, Protonex, Pantup,Pantomed, TopZole, Nolpaza, Controloc, UXL-D, Pantid, Pantogen, Pantpas and Prazolin.

Pantoprazole sodium salt

The structural formula

Brief background information

Salt ATC Formula MM CAS
A02BC02
A02BD04
16 H 14 F 2 N 3 NaO 4 S 405.36 g / mol 138786-67-1
hydrate A02BC02
A02BD04
16 H 14 F 2 N 3 NaO 4 S · 3 / 2H 2 O 864.76 g / mol 164579-32-2
(+) – Isomer A02BC02
A02BD04
16 H 14 F 2 N 3 NaO 4 S 405.36 g / mol 160098-11-3
(-) – Isomer A02BC02
A02BD04
16 H 14 F 2 N 3 NaO 4 S 405.36 g / mol 160488-53-9
racemate A02BC02
A02BD04
16 H 14 F 2 N 3 NaO 4 S 405.36 g / mol 142678-34-0

Application

  • agent for the treatment of gastric ulcer
  • inhibitor of gastric H + / K + ATPase

Classes of substances

  • Benzimidazoles, 2 (alkylsulfinyl) benzimidazoles
    • Fluoro-ethers
      • Pyridines

 

Country Patent Number Approved Expires (estimated)
Canada 2428870 2006-05-23 2021-11-17
Canada 2092694 2005-04-05 2011-09-06
Canada 2341031 2006-04-04 2019-08-12
United States 7544370 2006-12-07 2026-12-07
United States 4758579 1993-07-19 2010-07-19

 

Synthesis pathway

Synthesis a)





 

http://www.google.com/patents/EP1335913A1?cl=en

Pantoprazole is the international non-proprietary name of the chemical product 5-(difluoromethoxy)-2-[[(3,4-dimethoxy-2- pyridinyl)methyl]sulfmyl]-lH-benzimidazole of formula

 

Figure imgf000002_0001

Pantoprazole This product is an active ingredient used in the treatment of gastric ulcers, usually in the form of its sodium salt.

The product was described for the first time in European patent application EP-A-0166287 that also describes several processes for the preparation of products assignable to a general formula among which pantoprazole is to be found. The reaction sequences of these processes, applied precisely to the preparation of pantoprazole, are given in Scheme 1.

Figure imgf000003_0001

Scheme 1

In Scheme 1, the variables Y, Z, Z’ and Z” are leaving groups, for example atoms of halogen, and the variables M and M’ are atoms of alkali metals.

Austrian patent AT-B-394368 discloses another process based on a different route of synthetis, the reaction sequence of which is given in Scheme 2.

 

Figure imgf000004_0001

Pantoprazole Scheme 2

Nevertheless, this process has obvious drawbacks, since the methylation can take place not only in OH in the 4-position of the pyridine ring, but also in the nitrogen linked to a hydrogen of the benzimidazole ring, which can give place to mixtures of the desired product with the two possible methylated isomers of the benzimidazole compounds obtained, 3- methyl or 1 -methyl, which means that additional chromatographic purification steps are needed and the yields obtained are low.

PCT application WO97/29103 discloses another process for the preparation of pantoprazole, the reaction sequence of which is given in Scheme 3.

 

Figure imgf000004_0002

Scheme 3 As may be seen, different synthesis strategies have been proposed for the preparation of pantoprazole, some of them recently, which is an indication that the preparation of the product is still not considered to be sufficiently well developed, whereby there is still a need for developing alternative processes that allow pantoprazole to be prepared by means of simpler techniques and more accessible intermediate compounds and with good chemical yields.

EXAMPLES

Example 1. – Preparation of compound (IX)

 

Figure imgf000009_0002

47.5 ml (0.502 mol) of acetic anhydride were mixed with 1.65 g (0.0135 mol) of 4-dimethylaminopyridine, giving a transparent yellow solution which was heated to 65° – 70°C. This temperature was held by cooling since the reaction is exothermic. 25 g (0.1441 mol) of 2-methyl-3- methoxy-4-chloropyridine N-oxide (X) were added over a period of about 70 minutes. Once the addition was completed, the reaction was held at 65° – 70°C for a further 2h 20 minutes and after this time it was allowed to cool down to below 65°C and 90 ml of methanol were added gradually, while holding the temperature below 65°C. The resulting reaction mass was distilled at reduced pressure in a rotavap to remove the volatile components and the residue containing compound (IX) was used as such for the following reaction. Thin layer chromatography on silica gel 60 F254, eluting with CHCl3/MeOH (15: 1), showed a single spot at Rf – 0.82, indicating that the reaction has been completed.

Example 2. – Preparation of compound fVIII

 

Figure imgf000010_0001

(IX) (VIII)

11.5 ml methanol and 11.5 ml of water were added over the crude residue from Example 1 containing compound (IX), and thereafter, while holding the temperature to between 25° and 30°C with a water bath, the residual acetic acid contained in the crude residue was neutralized by the addition of 33% aqueous NaOH. Once the residual acid had been neutralized, 19 ml (0.2136 mol) of the 33% aqueous NaOH were added over 20 minutes, while holding the temperature to between 25° and 30°C, and, on completion of the addition, the hydrolysis reaction at pH 11.7 – 11.8 was held for 2h 30 minutes, to between 25° and 30°C. On completion of the reaction, the pH was adjusted to 7.0 – 7.5 by the addition of HC1 35%, while holding the temperature to 25°C. Thereafter, 50 ml of methylene chloride were added and, after stirring and allowing to rest, the phases were decanted. A further five extractions were carried out with 30 ml methylene chloride each and the pooled organic phases were dried with anhydrous sodium sulfate, were filtered and washed, and were evaporated at reduced pressure in a rotavap, providing a solid residue having a melting point around 73°C and containing compound (VIII). Thin layer chromatography on silica gel 60 F254, eluting with CHCl3/MeOH (15: 1), gave a main spot at Rf = 0.55, showing that the reaction was complete. The thus obtained crude residue was used as such in the following reaction.

Example 3. – Preparation of compound (VI)

 

Figure imgf000011_0001

24.5 g of the residue obtained in Example 2, containing approximately 0.142 mol of the compound 2-hydroxymethyl-3-methoxy-4-chloropyridine (VIII), were mixed with 0.5 ml of DMF and 300 ml of anhydrous methylene chloride, to give a brown solution which was cooled to 0° – 5°C in an ice water bath. Thereafter, a solution of 11.5 ml (0.1585 mol) of thionyl chloride in 50 ml of anhydrous methylene chloride was added over 20 minutes, while holding the above-mentioned temperature,. Once the addition was complete, the reaction was held at 0° – 5°C for a further 90 minutes and then 120 ml of water and NaOH 33% were added to pH 5 – 6, requiring approximately 29 ml of NaOH. The phases were then decanted and separated. The organic phase was extracted with a further 120 ml of water and the pooled aqueous phases were extracted with a further 4×25 ml of methylene chloride, in order to recover the greatest possible amount of product. The pooled organic phases were dried over anhydrous sodium sulfate, filtered and washed, and evaporated at reduced pressure in a rotavap, to give a residue containing the compound 2-chloromethyl-3- methoxy-4-chloropyridine (VI). Thin layer chromatography on silica gel 60 F254, eluting with CHCl3/MeOH (15:1), showed a main spot at Rf = 0.83, indicating that the reaction was complete. The thus obtained crude residue was used as such in the following reaction. Example 4. – Preparation of compound (III)

 

Figure imgf000012_0001

26.11 g of the residue obtained in the Example 3 containing approximately 0.136 mol of the compound 2-chloromethyl-3-methoxy-4- chloropyridine (VI) were mixed with 370 ml of methylene chloride, to give a brown solution over which were added, at 20° – 25°C, 29.3 g (0.136 mol) of 5-difluoromethoxy-2-mercaptobenzimidazole (VII) and 17.10 ml (0.136 mol) of tetramethylguanidine (TMGH). The mixture was stirred at this temperature for 2 hours, after which 450 ml of water were added, with the pH being held to between 9.5 and 10. Thereafter the phases were decanted and the organic phase was washed 5×50 ml of a IN NaOH aqueous solution and, thereafter, with 2×50 ml of water. The organic phase was treated with 50 ml of water and an amount of HC1 30% sufficient to adjust the pH to between 5 and 6. Thereafter, the phases were decanted, and the organic phase was dried over anhydrous sodium sulfate, was filtered and washed, and evaporated at reduced pressure in a rotavap, to give a solid residue of melting point 64° – 73 °C that contains the compound (III). Thin layer chromatography on silica gel 60 F254, eluting with CHCl3/MeOH (15: 1), presented a main spot at Rf = 0.52. Yield 82%. The thus obtained compound 5-(difluoromethoxy)-2-[[(3-methoxy-4-chlorine-2 pyridinyl)methyl]mercapto]- lH-benzimidazole (III) was used as such in the following reaction Example 5. – Preparation of compound (IV)

 

Figure imgf000013_0001

25.8 g (0.0694 mol) of the compound (III) obtained in the Example 4 were mixed with 88 ml of methanol, to give a brown solution to which 3.7 ml of water, 0.99 g of ammonium molybdate and 0.78 g of sodium carbonate were added. The system was cooled to 0°C – 5°C, 3.4 ml (0.0756 mol) of 60% hydrogen peroxide were added, and the reaction mixture was held at 0°C – 5°C for 1 – 2 days, the end point of the reaction being checked by thin layer chromatography on silica gel 60 F254, eluting with CHCl3/MeOH (15: l).

During the reaction the presence of hydrogen peroxide in the reaction medium was controlled by testing with potassium iodide, water and starch. When effected on a sample containing hydrogen peroxide, it provides a brown-black colour. If the assay is negative before the chromatographic control indicates completion of the reaction, more hydrogen peroxide is added.

On completion of the reaction, 260 ml of water were added, the system was cooled to 0°C – 5°C again and the mixture was stirred for 2 hours at this temperature. The solid precipitate was filtered, washed with abundant water, and dried at a temperature below 60°C, to give 5-(difluoromethoxy)-2-[[(3- methoxy-4-chlorine-2-pyridinyl)methyl]sulfinyl]-lH-benzimidazole (IV), melting point 130° – 136°C, with an 83.5% yield. Thin layer chromatography on silica gel 60 F254, eluting with CHCl3/MeOH (15: 1), gave a main spot at Rf = 0.5.

Compound (IV) can be purified, if desired, by the following crystallization method:

5 g of crude product was suspended in 16 ml of acetone and was heated to boiling until a dark brown solution was obtained. Thereafter the thus obtained solution was allowed to cool down to room temperature and then was then chilled again to -20°C, at which temperature the mixture was held for 23 hours without stirring. Thereafter the solid was filtered and washed with 6×4 ml of acetone chilled to -20°C. Once dry, the resulting white solid weighed 2.73 g, had a point of melting of 142°C and gave a single spot in thin layer chromatography. The IR spectrum of the compound on KBr is given in Figure 1.

The acetonic solution comprising the mother liquors of filtration and the washes was concentrated to a volume of 20 ml and a further 5 g of crude compound were added. The above described crystallization process was repeated to obtain a further 4.11 g of purified product of characteristics similar to the previous one.

The acetonic solution from the previous crystallization was concentrated to a volume of 17 ml and a further 4 g of crude compound were added. The above described crystallization process was repeated to obtain a further 2.91 g of purified product of similar characteristics to the previous ones.

The acetonic solution from the previous crystallization was concentrated to a volume of 15 ml and a further 4 g of crude compound were added. The above described crystallization process was repeated to obtain a further 3.3 g of purified product of similar characteristics to the previous ones.

The acetonic solution from the previous crystallization was concentrated to a volume of 16 ml and a further 4.36 g of crude compound were added. The above described crystallization process was repeated to obtain a further 3.62 g of purified product of similar characteristics to the previous ones.

Finally, the acetonic solution from the previous crystallization was concentrated to a volume of 10 – 12 ml and held at -20°C for two days without stirring. Thereafter, the solid was filtered and washed with 5×3 ml of acetone chilled to -20°C. Once dry, the solid weighed 1.26 g and had similar characteristics to the previous ones.

The total yield of all the crystallizations was 80%.

Example 6. – Preparation of pantoprazole

 

Figure imgf000015_0001

12.95 g (0.0334 mol) of compound (IV) purified by crystallization of Example 5 were mixed with 38 ml of N,N-dimethylacetamide and thereafter 7.03 g (0.1003 mol) of potassium methoxide were added, while holding the temperature to between 20°C and 30°C, whereby a dark brown mixture was obtained. The system was held at approximately 25°C for about 23 hours, after which, once the reaction was complete, the pH was adjusted to 7 with the addition of 3.82 ml of acetic acid. The N,N-dimethylacetamide was removed at reduced pressure at an internal temperature of not more than 75°C. 65 ml of water and 50 ml of methylene chloride were added over the thus obtained residue, followed by decantation of the phases. Once the phases were decanted, the aqueous phase was extracted a with further 3×25 ml of methylene chloride, the organic phases were pooled and the resulting solution dried over anhydrous sodium sulfate, was filtered and washed, and evaporated at reduced pressure in a rotavap, to give a crude residue over which 55 ml of water were added, to give a suspension (if the product does not solidify at this point the water is decanted and a further 55 ml of water are added to remove remains of N,N-dimethylacetamide that hinder the solidification of the product). The solid was filtered and, after drying, 11.61 g of crude pantoprazole of reddish brown colour were obtained (Yield 90%). The thus obtained crude product was decoloured by dissolving the crude product in 150 ml of methanol, whereby a dark brown solution was obtained. 7.5 g of active carbon were added, while maintaining stirring for 45 minutes at 25°C – 30°C, after which the carbon was filtered out and the filter was washed. The methanol was then removed in the rotavap at reduced pressure, a temperature below 40°C. 10.33 g of a solid residue were obtained and were mixed with 14.9 ml of methylethylketone, and the suspension was heated to 45°C for about 10 minutes, after which it was cooled, first to room temperature and then to -20°C. This temperature was held over night and thereafter the solid was filtered, washed with 6×5 ml of methylethylketone chilled to -20°C. Once dry, 7.75 g of a white solid, melting point 140°C – 141 °C, were obtained. Thin layer chromatography on silica gel F254, eluting with CHCl3/MeOH (15: 1), gave a single spot at Rf =

0.41 and a IR spectrum corresponding identically with that of pantoprazole.

The ketonic solution comprising the mother liquors of filtration and the washes, was concentrated to 9.7 ml, was heated to 40°C, was held at this temperature for about five minutes and was then cooled, first to room temperature and then to -20°C, this temperature being held for 4 hours. At the end of this time, the solid was filtered and was washed with 4×2 ml of methylethylketone chilled to -20°C. Once dry, 0.42 g of a white solid of similar characteristics to the previous one was obtained.

The ketone solution from the previous treatment was concentrated to 3.1 ml, was heated to 40°C, was held to this temperature for about five minutes and then was cooled, first to room temperature and then to -20°C, this temperature being held for 4 hours. At the end of this time, the solid was filtered and was washed with 5×3 ml of methylethylketone chilled to – 20°C. Once dry, 0.41 g of a white-beige solid of similar characteristics to the previous one was obtained. The total yield, including purifications, was 67%.

If a whiter solid is desired, one or several washes can be carried with isopropyl acetate as follows: 6.6 g of pantoprazole from the methylethylketone treatment were suspended in 50 ml of isopropyl acetate. The system (white suspension) was stirred for about 30 minutes at 25°C, was then cooled to 0°C – 5°C, was stirred for about 15 minutes at this temperature and the solid was then filtered, was washed with 3×15 ml of isopropyl acetate. Once dry, 6.26 g of a pure white solid were obtained.

 

 

 

Trade Names

Country Trade name Manufacturer
Germany Pantozol Nycomed
Rifun – “-
France Eupantol Altana
Inipomp Sanofi-Aventis
United Kingdom Protium ALTANA
Italy Pantekta Abbott
Pantopan Pharmacia
Pantork Altana
USA Protonix Wyeth
Ukraine Kontrolok Nycomed Oranienburg GmbH, Germany
Nolpaza Krka
Pultset Nobel Ilach Sanayi ve Ticaret AS, Turkey
Proksium JSC “Lubnyfarm”, Ukraine
various generic drugs

Formulations

  • ampoule 40 mg;
  • Tablets 40 mg

UV – spectrum

Conditions : Concentration – 1 mg / 100 ml
Solvent designation schedule
Methanol
Water
0.1 M HCl
0.1M NaOH
The absorption maximum 289 nm 291nm Observed
decay
295 nm
391 346 418
ε 16600 14700 17700

IR – spectrum

Wavelength (μm)
Wavenumber (cm -1 )

NMR Spectrum

 will be added

 

 

Links

  • EP 134 400 (Byk Gulden Lomberg; appl. 1.5.1984; CH-prior. 3.5.1983).
  • US 4,555,518 (Byk Gulden Lomberg; 26.11.1985; appl. 1.5.1984; CH-prior. 3.5.1983).
  • US 4,758,579 (Byk Gulden Lomberg; 19.7.1988; appl. 28.4.1987; CH-prior. 16.6.1984).
  • UV and IR Spectra. H.-W. Dibbern, RM Muller, E. Wirbitzki, 2002 ECV
  • NIST / EPA / NIH Mass Spectral Library 2008
  • Handbook of Organic Compounds. NIR, IR, Raman, and UV-Vis Spectra Featuring Polymers and Surfactants, Jr., Jerry Workman.Academic Press, 2000.
  • Handbook of ultraviolet and visible absorption spectra of organic compounds, K. Hirayama. Plenum Press Data Division, 1967.

References

  1.  Pali-Schöll I, Jensen-Jarolim E (April 2011). “Anti-acid medication as a risk factor for food allergy”. Allergy 66 (4): 469–77. doi:10.1111/j.1398-9995.2010.02511.xPMID 21121928.
  2.  [Dr. John Cooke, chair of Methodist Hospital’s cardiovascular services] [Houston Chronicle Health Zone dated Thursday, July 11, 2013 chron.com/refluxmeds] (Journal: Circulation)
  3. Jump up^ Meyer, U A (1996). “Metabolic interactions of the proton-pump inhibitors lansoprazole, omeprazole and pantoprazole with other drugs”. European journal of gastroenterology & hepatology8 (Suppl 1): S21–25. doi:10.1097/00042737-199610001-00005.
  4.  Steinijans, V. W.; Huber, R.; Hartmann, M.; Zech, K.; Bliesath, H.; Wurst, W.; Radtke, H. W. (1996). “Lack of pantoprazole drug interactions in man: An updated review”. International Journal of Clinical Pharmacology and Therapeutics 34 (6): 243–262. PMID 8793611.
  5.  Sachs G, Shin JM, Hunt R (December 2010). “Novel approaches to inhibition of gastric acid secretion”Curr Gastroenterol Rep 12 (6): 437–47. doi:10.1007/s11894-010-0149-5.PMC 2974194PMID 20924727.
  6.  Teva Announces Launch Of Generic Protonix Tablets
  7. Jump up^ Rubenstein, Sarah (29 January 2008). “Wyeth Plans Generic Protonix; Litigation With Teva to Continue”The Wall Street Journal. p. D9. Retrieved 25 October 2009.
  8. Jump up^ “Nycomed and Wyeth announce launch of an own generic version of PROTONIX – lawsuit to defend patent continues”. Retrieved 25 October 2009.[dead link]
  9. Jump up^ IntelliPharmaCeutics Press Release

External links

Share
Follow

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

Join other followers: