Cefaloridine

Jump to navigation Jump to search
Cefaloridine
Clinical data
AHFS/Drugs.comInternational Drug Names
ATC code
Pharmacokinetic data
ExcretionRenal
Identifiers
CAS Number
PubChem CID
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
E number{{#property:P628}}
ECHA InfoCard{{#property:P2566}}Lua error in Module:EditAtWikidata at line 36: attempt to index field 'wikibase' (a nil value).
Chemical and physical data
FormulaC19H17N3O4S2
Molar mass415.486 g/mol
3D model (JSmol)
  (verify)

WikiDoc Resources for Cefaloridine

Articles

Most recent articles on Cefaloridine

Most cited articles on Cefaloridine

Review articles on Cefaloridine

Articles on Cefaloridine in N Eng J Med, Lancet, BMJ

Media

Powerpoint slides on Cefaloridine

Images of Cefaloridine

Photos of Cefaloridine

Podcasts & MP3s on Cefaloridine

Videos on Cefaloridine

Evidence Based Medicine

Cochrane Collaboration on Cefaloridine

Bandolier on Cefaloridine

TRIP on Cefaloridine

Clinical Trials

Ongoing Trials on Cefaloridine at Clinical Trials.gov

Trial results on Cefaloridine

Clinical Trials on Cefaloridine at Google

Guidelines / Policies / Govt

US National Guidelines Clearinghouse on Cefaloridine

NICE Guidance on Cefaloridine

NHS PRODIGY Guidance

FDA on Cefaloridine

CDC on Cefaloridine

Books

Books on Cefaloridine

News

Cefaloridine in the news

Be alerted to news on Cefaloridine

News trends on Cefaloridine

Commentary

Blogs on Cefaloridine

Definitions

Definitions of Cefaloridine

Patient Resources / Community

Patient resources on Cefaloridine

Discussion groups on Cefaloridine

Patient Handouts on Cefaloridine

Directions to Hospitals Treating Cefaloridine

Risk calculators and risk factors for Cefaloridine

Healthcare Provider Resources

Symptoms of Cefaloridine

Causes & Risk Factors for Cefaloridine

Diagnostic studies for Cefaloridine

Treatment of Cefaloridine

Continuing Medical Education (CME)

CME Programs on Cefaloridine

International

Cefaloridine en Espanol

Cefaloridine en Francais

Business

Cefaloridine in the Marketplace

Patents on Cefaloridine

Experimental / Informatics

List of terms related to Cefaloridine

Editor-In-Chief: C. Michael Gibson, M.S., M.D. [1]

Overview

Cephaloridine (or cefaloridine) is a first generation semisynthetic derivative of cephalosporin C. It is unique among cephalosporins in that it exists as a zwitterion.

History

Since the discovery of cephalosporins P, N and C in 1948 there have been many studies describing the antibiotic action of cephalosporins and the possibility to synthesize derivatives. Hydrolysis of cephalosporin C, isolation of 7-aminocephalosporanic acid and the addition of side chains opened the possibility to produce various semi-synthetic cephalosporins. In 1962, cephalothin and cephaloridine were introduced.[1]

Cephaloridine was temporarily popular because it was better tolerated intramuscularly and attained in higher and more sustained levels in blood than cephalothin. However, it binds to proteins to a much lesser extent than cephalothin. Because it is also poorly absorbed after oral administration the use of this drug for humans declined rapidly, especially since the second generation of cephalosporins was introduced in the 1970s.[1] Today it is more commonly used in veterinary practice to treat mild to severe bacterial infections caused by penicillin resistant and penicillin sensitive Staphylococcus aureus, Escherichia coli, Streptococcus pyogenes, Streptococcus pneumoniae, Bacillus sutbtilis, Klebsiella, Clostridium diptheriae, Salmonella and Shigella.[2] Interest in studying cephalosporins was brought about by some unusual properties of cephaloridine. This antibiotic stands in sharp contrast to various other cephalosporins and to the structurally related penicillins in undergoing little or no net secretion by the mammalian kidney. Cephaloridine is, however, highly cytotoxic to the proximal renal tubule, the segment of the nephron responsible for the secretion of organic anions, including para-am-minohippurate (PAH), as well as the various penicillin and cephalosporin antibiotics. The cytotoxicity of cephaloridine is completely prevented by probenecid and several other inhibitors of organic anion transport, including the nearly nontoxic cephalothin.[3]

Structure & reactivity

Cephaloridine is a cephalosporin compound with pyridinium-1-ylmethyl and 2-thienylacetamido side groups. The molecular nucleus, of which all cephalosporins are derivatives, is A3-7-aminocephalosporanic acid. Conformations around the β-lactam rings are quite similar to the molecular nucleus of penicillin, while those at the carboxyl group exocyclic to the dihydrothiazine and thiazolidine rings respectively are different.[4]

Synthesis

Cephaloridine can be synthesised from Cephalotin and pyridine by deacetylation. This can be done by heating an aquous mixture of cephalotin, thiocyanate, pyridine and phosphoric acid for several hours. After cooling, diluting with water, and adjusting the pH with mineral acid, cephaloridine thiocyanate salt precipitates. This can be purified and converted to cephaloridine by pH adjustment or by interaction with ion-exchange resin.[5]

Clinical use of cephaloridine

Before the 1970s, cephaloridine was used to treat patients with urinary tract infections. Besides the drugs has been used successfully in the treatment of various lower respiratory tract infections. Cephaloridine was very effective to cure pneumococcal pneumonia. It has a high clinical and bacteriological rate of success in staphylococcal and streptococcal infections.[6]

Kinetics

Absorption

Cephaloridine is easy absorbed after intramuscular injection and poorly absorbed from the gastrointestinal tract.[7]

Distribution

The minor pathway of elimination is biliary excretion. When the blood serum concentration is 24 µg/ml, the corresponding biliary concentration is 10 µg/ml. In de spinal fluid the concentration of cephaloridine is 6-12% of the concentration in de blood and serum. Cephaloridine is distributed well into the liver, stomach wall, lung and spleen and is also found in fresh wounds one hour after injection. The concentration in the wound will decrease as the wound age increases. However, the drug is poorly penetrated into the cerebrospinal fluid and is found in a much smaller amount in the cerebral cortex.[7]

Pregnancy

When cephaloridine is administered to pregnant women, the drug crosses the placenta. Cephaloridine concentrations can be measured in the serum of the newborn up to 22 hours after labor, and can reach a level of 54% of the concentration in the maternal serum. When given an intramuscular dose of 1 g, a peak occurs in the cord blood after 4 hours. In amniotic fluid, the concentration takes about 3 hours to reach its antibacterial effect.[6]

Metabolism and Excretion

Urine specimens showed that no other microbiologically active metabolites were present except cephaloridine and that cephaloridine is excreted unchanged. Renal clearances were reported to be 146-280 ml/min, a plasma clearance of 167 ml/min/1,73m2 and a renal clearance of 125 ml/min/1,73m2. A serum half-life of 1,1-1,5 hour and a volume of distribution of 16 liters were reported.[7]

Pharmacokinetics

Pharmacokinetic analysis is not possible because appropriate data is not published. The physicochemical properties are almost the same as the other cephalosporins, therefore the pharmacokinetics are comparable.[7]

Adverse effects

Toxicity

Cephaloridine can cause kidney damage in humans, since it is actively taken up from the blood by the proximal tubular cells via an organic anion transporter (OAT) in the basolateral membrane. Organic anions are secreted through the proximal tubular cells via unidirectional transcellular transport. The organic anions are taken up from the blood into the cells across the basolateral membrane and extruded across the brush border membrane into the tubular fluid.[8] Cephaloridine is a substrate for OAT1 and thus can be transported into the proximal tubular cells, which form the renal cortex.[9] The drugs, however, cannot move readily across the luminal membrane since it is a zwitterion. The cationic group (pyridinium ring) of the compound probably inhibits the efflux through the membrane.[9][10] This results in an accumulation of cephaloridine in the renal cortex of the kidney, causing damage and necrosis of the S2 segment of the tubule.[8][9] However, there are no adverse effects on renal function if serum levels of cephaloridine are maintained between 20 and 80 μg/ml.[11]

Metabolism

Cephaloridine is excreted in the urine without undergoing metabolism.[12] It inhibits organic ion transport in the kidney. This process is preceded by the lipid peroxidation. Thereafter, probably a combination of events, such as formation of a reactive intermediate, a free radical and stimulation of lipid peroxidation, lead to peroxidative damage to cell membranes and mitochondria. It is not yet clear whether metabolic activation by cytochromes P-450, chemical rearrangements, reductive activation or all these actions are involved.[9]

The hypotheses made about the mechanism of action causing the toxiciy of cephaloridine are:

  • Reactive metabolites are formed by cytochromes P-450 or emerge from destabilization of the β –lactam ring. Metabolic activation of the drugs might take place via cytochromes P-450, producing reactive metabolites. This hypothesis is based on the behaviour of some inhibitors of CYTP450, which decrease the toxicity, and some inducers of the monooxygenases which increase toxicity. It could also be possible that a reactive intermediate is formed due to the unstable β -lactam ring.[9] The pyridinium side-group of cephaloridine has unstable bonds to the core of the compound (in comparison with other cephalosporins). When this side-group leaves, the β –lactam ring is destabalized by intramolecular electron shifts.[13] Thus, the leaving group creates a reactive product.
  • Both lipid peroxidation and oxidative stress can cause membrane damage. Lipid peroxidation and oxidative stress take place as lipid peroxidation products, such as malondialdehyde, have been detected. Reduced glutathione (GSH) and NADPH are both depleted. Consequently, GSSG cannot be reduced to GSH. This leads to an increased toxicity since oxidative stress cannot be reduced. In addition, nephrotoxicity is augmented by deficiency of selenium or tocopherol. The pyridinium side-group interacts with the reduced NADP in a redox cycle. It has been suggested that superoxide anion radicals and hydroxyl radicals may be formed and that lipid peroxidation could be responsible for the toxicity of cephaloridine.[9][13]
  • Damage to the mitochondria and intracellular respiratory processes and reduced mitochondrial respiration can cause nephrotoxicity. The previously mentioned damages have been detected after exposure to cephalosporins.[9] β-lactam antibiotics injure mitochondria by an attack on the metabolic substrate carriers of the inner membrane.[13] Respiratory toxicity is caused by inactivation of mitochondrial anion substrate carriers.[8]

Symptoms of kidney damage caused by cephaloridine

Some symptoms caused by cephaloridine are: asymptomatic, enzymuria, proteinuria, tubular necrosis, increased urea level in blood, anemia, increased hydrogen ion level in blood, fatigue, increased blood pressure, increased blood electrolyte level, kidney dysfunction, kidney damage, impaired body water balance and impaired electrolyte balance.[14]

Complications caused by cephaloridine

Complications caused by the use of cephaloridine include seizures, coma, chronic kidney failure, acute kidney failure and death.[14]

Treatment of kidney damage caused by cephaloridine

The damage of the kidneys can be treated by removing the toxin from the body, monitoring and supporting kidney function (dialysis if necessary) and, in severe cases, kidney transplant. Supportive therapy in the acute phase can be done by fluid, electrolyte and hypertension management. Longer term management includes monitoring of renal function, close management of high blood pressure. Furthermore, dietary management may include protein and sodium management, adequate hydration and phosphate and potassium restriction. In case of chronic renal failure dietary management also includes erythropoietin agonists (since anaemia is associated with chronic renal failure), phosphate binders (in case of hyperphosphatemia), calcium supplements, Vitamin D supplements and sodium bicarbonate (to correct the acid-base disturbance).[14]

References

  1. 1.0 1.1 Mason, I.S, Kietzmann, M., Cephalosporins-pharmalogical basis of clinical use in veterinary dermatology, Veterinary Dermatology 1999, 10, 187-192
  2. R.K. CHAUDHARY AND A.K. SRIVASTAVA, Disposition and dosage regimen of cephaloridine in calves, Vezerkny Research Communications, 13 (1989) 325-329. http://www.springerlink.com/content/l67g7035w9113317/fulltext.pdf
  3. Bruce M Tune and Doris Fravert, Mechanisms of cephalosporin nephrotoxicity: A comparison of cephaloridine and cephaloglycin, Kidney International (1980) 18, 591–600; doi:10.1038/ki.1980.177 http://www.nature.com/ki/journal/v18/n5/pdf/ki1980177a.pdf
  4. R. M. Sweet and L. F. Dahl, THE STRUCTURE OF CEPHALORIDINE HYDROCHLORIDE MONOHYDRATE, BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 1969 Vol. 34, No. 1
  5. Osol, A. and J.E. Hoover, et al. (eds.). Remington's Pharmaceutical Sciences. 15th ed. Easton, Pennsylvania: Mack Publishing Co., 1975., p. 1120
  6. 6.0 6.1 D.R. Owens, Advances in pharmacology and chemotherapy, Volume 13, Academic Press, Inc., p 83-170
  7. 7.0 7.1 7.2 7.3 Charles H. Nightingale, Douglas S. Greene, Richard Quintiliani, Pharmacokinetics and Clinical Use of Cephalosporin Antibiotics, Journal of Pharmaceutical Sciences, December 1975, Volume 64 number 12, page 1913-1914
  8. 8.0 8.1 8.2 Michio Takeda, Akihiro Tojo, Takashi Sekine, Makoto Hosoyamada, Yoshikatsu Kanai and Hitoshi Endou, Role of organic anion transporter 1 (OAT1) in cephaloridine (CER)-induced nephrotoxicity, Kidney International (1999), http://www.nature.com/ki/journal/v56/n6/full/4491152a.html
  9. 9.0 9.1 9.2 9.3 9.4 9.5 9.6 John A. Timbrell, Principles of Biochemical Toxicology, Informa Healthcare USA Inc., 2009, p 332-335
  10. Robert W. Schrier, Diseases Of The Kidney And The Urinary Tract, Lippincott Williams & Wilkins, 2007, p 1041 http://books.google.nl/books?id=ERqtOZMAiw0C&pg=PA1041&lpg=PA1041&dq=Cephaloridine+reactivity&source=bl&ots=tZiGHvQKvZ&sig=NBOP6rdDIOHJZ0bovIuCwZBkqVQ&hl=nl&ei=mGpeTcTeLs-UOsPGyekN&sa=X&oi=book_result&ct=result&resnum=1&ved=0CBYQ6AEwADgK#v=onepage&q=Cephaloridine%20reactivity&f=false
  11. J.F. Winchester, A.C. Kennedy, Absence of nephrotoxicity during chephaloridine therapy for urinary-tract infection, http://www.thelancet.com/journals/lancet/article/PIIS0140-6736(72)91908-3/abstract
  12. Marvin Turck, Cephalosporins and Related Antibiotics: An Overview, REVIEWS OF INFECTIOUS DISEASES • VOL. 4, SUPPLEMENT • SEPTEMBER-OCTOBER 1982, © 1982 by The University of Chicago. http://cid.oxfordjournals.org/content/4/Supplement_2/S281.full.pdf
  13. 13.0 13.1 13.2 Bruce M. Tune, Nephrotoxicity of beta-lactam antibiotics: mechanisms and strategies for prevention, Pediatr Nephrol (1997) 11: 768±772, http://www.springerlink.com/content/3q23m092mlpn7rv5/
  14. 14.0 14.1 14.2 http://www.wrongdiagnosis.com/k/kidney_damage_cephaloridine/intro.htm