Novel strategies in antibacterial drug developmentref
:
antivirulence-directed therapy
target virulent bacteria
requires discovery of a lethal target present only in
virulent bacteria
vaccination directed against virulence determinants has
also been proposed
this approach would prevent acquisition of antibiotic
resistance genes by non-pathogenic bacteria
genomics : new targets for inhibiting bacterial growth and
variability : block vital metabolic pathway, efflux pump
inhibitors, quorum-sensing signalling systems, hoped that
mutations in the targeted gene that may occur through drug
pressure will not be compatible with viability. Identify target
gene => clone gene, express, purify and crystallize =>
identify active site and screen molecule against site => test
in whole bacteria. 200-400 bacterial genes are essential genes
and are a priori candidates for killing sites by
inhibitors. Comparative bacterial genomics allows further
prioritization of ORFs that are conserved in pathogens but
absent in higher eukaryotes, and for which a function can often
be predicted by bioinformatics. There are several cases in which
additional knowledge is likely to increase markedly the
probability of finding new macromolecular targets. An
under-explored target is the transglycosylase step of
peptidoglycan assembly in the cell wall, in which the
chemo-enzymatic synthesis of the complex lipoglycopeptide
substrate, lipid II, now enables modern configurations of
high-throughput assays. In Gram-positive bacteria, it has been
known for some time that cell-surface proteins can covalently
attach to peptidoglycan. Bacterial genomic analyses have
identified multiple sortases in different pathogens. Deciphering
the selectivity of sortases for groups of proteins that end up
at the cell surface might be a prelude to the screening or
design of inhibitors. Sortases should be readily accessible to
ligands, and therefore good drug targets, but it remains to be
proven how responsive they will be for bacteriostatic or
bacteriocidal action. A final example is the non-classical
pathway of isoprenoid synthesis. In contrast to the classical
mevalonate pathway found in eukaryotes, many bacteria, including
clinically important pathogens, have recently been found to
contain the genes that encode a separate pathway that has
deoxyxylulose-5-phosphate as an intermediate rather than
mevalonate. The 7 enzymes of the deoxyxylulose-5-phosphate
pathway are not present in humans; these enzymes are essential
for lipid I and II biosynthesis as key intermediates in
peptidoglycan assembly.
tageting non-multiplying bacteria (antibiotic kills the
multiplying bacteria) : combination strategies. When antibiotic
dose decreases below a threshold, metabolism in non-multiplying
bacteria starts and some of them begin to multiply, becoming
susceptible to the next dose of antibiotic
overcoming resistance : strategies to combat efflux
pumps-mediated drug resistance
apramycin, which mimics a short section of RNA,
interrupts the plasmid's replication by sticking to RNA
strands that carry its genetic information. This 'blocks' the
strands, at which point the host bacterium categorizes the
plasmid as a foreign body and ejects it. Apramycin is not
likely to be used in clinical trials, because it is quite
toxic. But now that the chemists have proved how it works,
they are looking for other molecules that could do the same
job without causing problems for the patient.
Lancet or phenol coefficient : a measure of the bactericidal
activity of a chemical compound in relation to phenol. The test is standardized
(Rideal-Walker method, U. S. Department of Agriculture method). The
coefficient is calculated by dividing the concentration of the test
compound at which it kills the test organism in 10 minutes, but not
in 5 minutes, by the concentration of phenol that kills the organism
under the same conditions. It can be determined in the absence of
organic matter, or in the presence of a standard amount of added
organic matter.
Woods-Fildes theory : the theory that the antibacterial
activity of at least some chemotherapeutic drugs (especially the
sulfonamides) is a consequence of a competitive inhibition of
essential metabolic reactions of the microorganism.
isoniazid / isoniazide
(INH) / isonicotinic acid hydrazide(INAH®,
Nydrazid®,
Rifamate®,
...) is a pro-drug, which, after activation by the
mycobacterial katG-encoded catalase peroxidase,
reacts nonenzymatically with NAD+ and NADP+
to generate several isonicotinoyl adducts of these pyridine
nucleotides :
the acyclic 4S isomer of isoniazid-NAD, targets
the inhA-encoded enoyl-ACP reductase, an enzyme
essential for mycolic acid biosynthesis in Mycobacterium
tuberculosis. It inhibits pyridoxal phosphorylation
to vitamin B6 (Mycobacteria cells
contain low levels of such a kinase) : so
pyridoxine-dependent reactions are inhibited (e.g.
enoyl-ACP reductase of fatty acid synthase (FAS) II, which
converts D2-unsaturated
to
saturated fatty acids on the pathway to mycolic acid
biosynthesis). It should be co-administered with vitamin B6 /
pyridoxine
to minimize adverse side effects from pyridoxal kinase
inhibition.
the acyclic 4R isomer of isoniazid-NADP inhibits
the M. tuberculosisdihydrofolate reductase
(DHFR), an enzyme essential for nucleic acid
synthesis. This biologically relevant form of the
isoniazid adduct is a subnanomolar bisubstrate inhibitor
of M. tuberculosis DHFR. Expression of M.
tuberculosis DHFR in Mycobacterium smegmatis
mc2155 protects cells against growth inhibition by
isoniazid by sequestering the drugref.
ethionamide
(Trecator-SC®) inhibits the acetylation of
isoniazid in vitro.
prothionamide
pyrazinamide (PZA)
(pyrazine analog of nicotinamide) inhibits mycobacterial
fatty acid synthase (FAS) I, involved in mycolic acid
biosynthesis.
D-ethambutol
hydrochloride (Myambutol®).
It
blocks
arabinosyl transferases involved in synthesis of lipoarabinomannan
(LAM) and
mycolic acids. It is the only chemotherapeutical effective on
Mycobacterium
marinum.
Side effects : visual
disturbances
daptomycin (Cidecin®,
Cubicin®)
: rapidly bactericidal, i.v. only, may cause myalgia,
efficacy for bacteremia/endocarditis not demonstrated
riminophenazines :
B663 / clofazimine /
clofazamine (Lamprene®) inhibits Na+/K+
ATPase and acts as an immunosuppressor
B669
b-lactams have been
introduced in 1940 :
penicillins :
tetrahydrothiazole-containing (penem
group) compounds that irreversibly inhibit the
transpeptidase activity of penicillin-binding protein (PBP)
3. 1 international unit of penicillin : the specific
penicillin activity contained in 0.6 mg
of the international standard sodium salt of penicillin II
or G.
natural
penicillins(Gram +ve Bacteria and Neisseria gonorrhoeae).
Resistance to penicillin today occurs in as many as 80% of
all strains of Staphylococcus aureus :
surprisingly, Streptococcus pyogenes has never
fully developed resistance to penicillin !
semisynthetic penicillins (Gram -ve rods). A mold produces
the main part of the molecule (6-aminopenicillanic acid)
which can be modified chemically by the addition of side
shains. Resistant to b-lactamase.
dihydropenicillin F
a-aminopenicillins / 2nd
generation
amoxicillin (Amoxil®,
Hiconcil®; Pulsys® is a
once-daily formulation; Flumox® in
combination with flucloxacillin)
ampicillin
(Omnipen®, Polycillin®,
Pentrexyl®, Servicillin®, ...)
is effective orally (prodrugs : bacampicillin
(Penglobe®, Spectrobid®),
etacillin, pivampicillin (Pondocillin®),
talampicillin and metampicillin)
Side effects : type I, type II, type III and type
IV
hypersensitivities. Before the initiation of cephalosporin
therapy, skin testing should be performed for patients with a
suspected history of IgE-mediated reactions to penicillinsref.
The rate of cross-reactivity of > 10% between penicillins
and cephalosporins has not been
well established. 3 retrospective clinical studies reported
rates of reaction to carbapenems
of 9 to 11% among inpatients with a reported penicillin
allergyref1,
ref2,
ref3.
An earlier study reported that of 20 subjects with a positive
penicillin skin test, 10 reacted to an imipenem reagentref.
oxacephems (oxacephem group)
: effective against Gram +ve and Gram -ve
Bacteria, penicillinase-resistant.
latamoxef
moxalactam (Moxam®)
cephalosporins (Gram +ve and Gram -ve Bacteria) (from 7-aminocephalosporanic
acid).
Cephem group. They tend to be
resistant to b-lactamases/penicillinases
from S. aureus. They bind PBP3.
ceftobiprole
medocaril / BAL 5788 / RO 65-5788 / JNJ 30982081
is the water-soluble prodrug of the pyrrolidinone
cephalosporin, ceftobiprole / BAL 9141 / RO 63-9141,
which has activity against P.aeruginosaref
carbapenems (Gram +ve cocci and Gram -ve
rods, P.aeruginosa, anaerobes, Enterococci)
(the N atom in the penem group of penicillin is substitued
by a C atom => carbopenem group).
They arise from modifications of the chemically
unstable thienamycin.
Parenteral administration.
group 1 : no activity against nonfermenting bacteria
tebipenem / ME121 (Japan)
panipenem + betamipron (PAPM/BP)
(Carbenin® + betamipron) : to inhibit
panipenem uptake into the renal tubule and prevent
nephrotoxicity
group 2 : activity against nonfermentating bacteria
imipenem
(IPM)
/ MK 0787 / N-formimidoiyl
thienamycin + cilastatin
(IPM/CS) (an inhibitor of renal dihydropeptidase
I (DHP-I)-mediated hydrolysis in proximal renal
tubule) (Imipem®, Primaxin®, Tienem®).
As it is a zwitterion it can penetrates the outer
membrane in Gram -ve Bacteria.
The trans-hydroxyethil side chain differs from
the cis-aminoacyl side chain in
penicillins and so this molecule is not a
substrate for b-lactamase.
It binds PBP a/Ib.
biapenem (BIPM)
(Japan) is more stable than imipenem, meropenem and
panipenem to hydrolysis by human renal DHP-I, and
therefore does not require the coadministration of a
DHP-I inhibitor.
group 3 : tomopenem
ertapenem (Invanz®) is
a once-daily parenteral group 1 carbapenem antibiotic used
in the treatment of complicated intraabdominal infectionref1,
ref2,
ref3.
Several characteristics of ertapenem make its use
attractive as a potential preoperative antimicrobial agent
in elective colorectal surgery, since it is characterized
by rapid intravenous administration, appropriate coverage
against potential pathogens, a long half-life (so it does
not require a second administration during most
surgeries), and a safety profile similar to that of other
commonly used antibioticsref1,
ref2,
ref3,
ref4
faropenem daloxate
(Farom®), a new oral penem
RO4908463 / CS-023
olivanic acids / olivanates
MM4550
MM13902
MM17880
MM22380
MM22381
MM22382
MM22383
In recent years, the number of class D beta-lactamases with
carbapenem-hydrolysing properties has increased substantially.
Based on amino acid sequence identities, these class D or
OXA-type carbapenemases are divided into eight distantly
related groups, and they are only remotely related to other
class D beta-lactamases. A putative ancestor to one of the
plasmid-encoded OXA-type carbapenemases has been found.
OXA-type carbapenemases are not integrated into integrons as
gene cassettes like many class D oxacillinases, but most of
the OXA-type carbapenemases are instead encoded by chromosomal
genes. Some of these OXA-type carbapenemases are widely
dispersed in Pseudomonas aeruginosa and especially in
Acinetobacter baumannii. Although most of the OXA-type
carbapenemases show only weak carbapenemase activity,
carbapenem resistance may result from a combined action an
OXA-type carbapenemase and a secondary resistance mechanism
such as porin deficiencies or overexpressed efflux pumps. This
article reviews the phylogeny and the genetic environments of
the encoding genes and kinetic properties of the OXA-type
carbapenemasesref
b-lactamase /
penicillinase competitive irreversible inhibitors
(they are effective only if coupled to a penicillin whose
pharmacokinetics is identical) :
glycopeptides
(heptapeptide-attached sugars) have been introduced in 1958 (Gram +ve Bacteria)
vancomycin group
vancomycin
hydrochloride (Lyphocin®,
Vancocin®,
Vancoled®)
inhibits both transglycosylation and transpeptidation
reactions during peptidoglycan assembly: it makes 5
hydrogen bonds to the -D-Ala-D-Ala dipeptide terminus of each
uncrosslinked peptidoglycan side chain, inhibiting both
the transglycosilase and transpeptidase activity of PBPs.
Nowadays vancomycin-resistant
Enterococci (VRE)
and vancomycin-resistant
Staphylococcus aureus (VRSA)
(MBC > 32 mg/mL) appeared
thanks to a plasmid-coded enzyme that creates -D-Ala-D-lactate
termini, resulting in the loss of 1 hydrogen bond that
significantly reduces the binding constant ! The VanA
strains are resistant to both vancomycin and teicoplanin, while the VanB
strains are resistant to vancomycin only.
Side effects : nephrotoxicity,
histamine-mediate "red-man
syndrome"
(anaphylactoid reaction). Cross-resistance to glycopeptides
can arise from use of avoparcin as growth promoter
in animal feeding.
oritavancin / LY333328
(Nuvocid®) is a semisynthetic lipoglycopeptide
analogue of vancomycin that contains the heptapeptide core
common to all glycopeptides. It differs from vancomycin by
the presence of a hydrophobic N-4-(4-chlorophenyl)benzyl
(also referred to as 4'-chlorobiphenylmethyl) substituent
on the disaccharide sugar, the addition of a
4-epi-vancosamine monosaccharide to the amino acid residue
in ring 6, and the replacement of the vancosamine moiety
by 4-epi-vancosamine. One mechanism of action of
oritavancin is inhibition of transglycosylation (important
in peptidoglycan synthesis) by binding to
D-alanyl-D-alanine stem termini in Gram-positive bacteria.
The inhibition of peptidoglycan synthesis via inhibition
of transglycosylation is common to all glycopeptides
(vancomycin) and lipoglycopeptides. Secondary binding of
oritavancin to the pentaglycyl (Asp/Asn) bridging segment
in peptidoglycan also occurs, which distinguishes it from
vancomycin and contributes to oritavancin’s activity
versus vancomycin-resistant organisms. The presence of the
hydrophobic 4'-chlorobiphenylmethyl group allows for
interaction and disruption of the cell membrane, resulting
in depolarization, permeabilization, and
concentration-dependent, rapid cell death. This mechanism
is shared with telavancin but not vancomycin and results
in activity against daptomycin-nonsusceptible organisms.
In conclusion, oritavancin’s mechanism of action involves
at least 3 known mechanisms: inhibition of
transglycosylation, inhibition of transpeptidation, and
cell membrane interaction/disruption. Oritavancin’s
multiple mechanisms of action confer activity against
vancomycin-susceptible and -resistant organisms, as well
as rapid, concentration-dependent killing versus actively
growing, stationary phase, and biofilm-producing
Gram-positive bacteria. Its concentration-dependent
activity and prolonged half-life allow for single-dose
treatment
teicoplanin group
: N-methylLeu in 1 and Lys in 3 are replaced by
substituted phenylGlys
teicoplanin
(Targocid®, Targosid®) (a
lipoglycopeptide less toxic than vancomycin)
telavancin
ristocetin
(it also binds to vWF (which in fact is a.k.a. ristocetin
cofactor) causing human platelets aggregation)
dalbavancin (Pfizer)
: a new milestone of outpatient antimicrobial therapy
(OPAT) ? One-shot schedule (1-week half-life; 60%
accumulates in bone)
eremomycin
BRL 47761
ramoplanin (a
glycolipodepsipeptide consisting of 17 amino acids cyclized
to a macrolactone) inhibits transpeptidase activity of PBPs
bacitracin zinc (AK-Tracin®,
Baci-IM®, Baciferm®; Polysporin®
in combination with polymixin B;
Bimixin® in combination with neomycin;
Neosporin®
in combination with both neomycin and
polymixin B)
(Gram +ve Bacteria) needs
a divalent cation (Mg2+, Zn2+, Co2+,
Ni2+ or Cu2+) to bind the
undecaprenol-P-P and preventing it from being dephosphorylated
by a membrane pyrophosphatase. It has a high kidney toxicity
which precludes its systemic use. It is present in many
topical antibiotic preparations, and since it is not absorbed
by the gut, it is given to "sterilize" the bowel prior to
surgery.
D-cycloserine
(Seromycin®)
(Gram -ve and Gram +ve Bacteria) (from Streptomyces
orchidaceous) enters bacterial cells by means of an
active transport system for Gly and can reach a relatively
high intracellular concentration inhibiting Ala racemase and D-Ala-D-Ala
synthetase. Side effects : as it
is fairly toxic (NMDA-R partial agonist) it
has limited use as a secondary drug for TBC.
spirocyclohexene
griseofulvin
inhibits microtubule formation in mitotic spindle. It may act
as a photosensitizer.
antibacterial interfering with cell membrane integrity : these
antibiotics disorganize the structure or inhibit the function of
bacterial membranes. However, due to the similarities in
phospholipids in eubacterial and eukaryotic membranes, this
action is rarely specific enough to permit these compounds to be
used systemically.
cyclic polypeptides
polymyxin. Effective
mainly against Gram -ve Bacteria
: usage is usually limited to topical usage and is
occasionally given for UTIs caused by Pseudomonas
strains that are gentamicin-, carbenicillin- and
tobramycin-resistant. The balance between effectiveness and
damage to the kidney and other organs is dangerously close,
and the drug should only be given under close supervision in
the hospital.
polymixin B sulfate
(a mixture of polymixin B1 and B2)
(Aerospin®; Polysporin®
in combination with bacitracin;
Neosporin®
in combination with bacitracin
and neomycin; Neosporin®
in combination with gramicidin and neomycin;
Polytrim® in combination with trimethoprim) binds to the
lipid-A portion (the hyppo) of LPSref
polymixin E /
colistin (Acetylcolistin®, Colisticina®,
Colistin-600®, Colimicin®,
Coly-Mycin-S®, Multimycine®) (bactericidal) from Aerobacillus
colistinus
polyene : inhibits sterols present in cell wall-less Bacteria, but
they are mainly used as antifungal agents.
platensimycin, a
previously unknown class of antibiotics produced by Streptomyces
platensis. Platensimycin demonstrates strong,
broad-spectrum Gram-positive antibacterial activity by
selectively inhibiting cellular lipid biosynthesis. This
anti-bacterial effect is exerted through the selective
targeting of b-ketoacyl-(acyl-carrier-protein
(ACP)) synthase I/II (FabF/B) in the synthetic pathway
of fatty acids. Direct binding assays show that platensimycin
interacts specifically with the acyl-enzyme intermediate of
the target protein, and X-ray crystallographic studies reveal
that a specific conformational change that occurs on acylation
must take place before the inhibitor can bind. Treatment with
platensimycin eradicates Staphylococcus aureus
infection in mice. Because of its unique mode of action,
platensimycin shows no cross-resistance to other key
antibiotic-resistant strains tested, including MRSA, VISA and
VRE. Platensimycin is the most potent inhibitor reported for
the FabF/B condensing enzymes, and is the only inhibitor of
these targets that shows broad-spectrum activity, in vivo
efficacy and no observed toxicityref.
antibacterial interfering with
protein synthesis
impairing EF-G
steroid antibacterials
fusidic acid (against Gram
+ve Bacteria; bactericidal
in vitro at high concentrations) (from Fusidium
coccineum) inhibits EF-G
oxazolidinones
(initiation inhibitors) (introduced in 2000) inhibit
the bacterial pre-translational initiation complex formation
=> bacteriostatic
linezolid (Zyvox®)
i.v. and p.o. Side effects :
thrombocytopenia, hyperlactatemia, metabolic acidosis, and
peripheral neuropathy are adverse effects related to the
drug's capacity for interference with mitochondrial
functionref;
efficacy for bacteremia/endocarditis not demonstrated
AZD2563
PNU288234
ranbezolid
DA7687
tedizolid (Sivextro®)
eperezolid
posizolid
radezolid
sutezolid
peptidyl transferase
phenicols / phenyl
propanoids have been introduced in 1949: effective
against Gram +ve and Gram -ve cocchi and Salmonella
typhi
chloramphenicol
(currently it is produced entirely by chemical
synthesis) (AK-Chlor®, Chloromycetin®,
Chloroptic®, Ocu-Chlor®; Colbiocin®
in combination with rolitetracycline
and sodium colistimetate) inhibits the bacterial enzyme
peptidyl transferase in the 50S subunit
Side effects : since
mitochondria probably originated from prokaryotic cells
and have 70S ribosomes, they are subject to inhibition.
The eukaryotic cells most likely to be inhibited by
chloramphenicol are those undergoing rapid multiplication,
thereby rapidly multiplying mitochondria.
such cells include the blood forming cells of the
bone marrow, the inhibition of which could present as
acquired
aplastic
anemia in
1:50,000. Bone marrow toxicity was recognized to be
associated with chloramphenicol in 2 ways:
a dose-dependent reversible marrow depression
that disappears when the drug is stopped
an idiosyncratic reaction that causes
irreversible marrow failure (albeit quite rarely)
that is not dose-dependent and may occur at quite
low drug levels. It should be noted that cases of
the idiosyncratic reaction have been described
following the use of chloramphenicol eye drops
in newborns it may cause gray syndrome (a potentially
fatal condition seen in neonates, particularly
premature infants, characterized by an ashen gray
cyanosis, listlessness, weakness, and gray
syndrome, a potentially fatal condition
seen in neonates, particularly premature infants, due
to a reaction to chloramphenicol, characterized by an
ashen gray cyanosis, listlessness, weakness, and systemic
arterial hypotension) due to
inadequate glucuronidation with drug accumulation.
Chloramphenicol was a commonly used antibacterial agent in
the 1950s and, like other antimicrobials in use today, was
commonly used in situations where no specific antibiotic
is required (such as in a viral respiratory infection).
Now it is seldom used in human medicine except in
life-threatening situations (e.g. typhoid fever). R
plasmid-encoded resistance gene = chloramphenicol
acetyltransferase(CAT). The only
antimicrobial whose enteric absorption (palmitate) is
better than parenteral absorption (succinate)
In Jan 2002, chloramphenicol was detected in animal feed
in Europe. This contamination was traced to fish/seafood
products coming from the Far East. A commentary from the
Chinese Ministry of Agriculture on the "Draft Report
for the Residue Control in Live Animals and Animal
Products by EC Inspection Mission to China"ref
mentions that chloramphenicol was discontinued from the
Chinese Veterinary Pharmarcopoeia in 2000. An
investigation into the contamination of shrimp revealed
that 'the prawn-peeling workers had not worn protective
gloves in the past, causing an itchy symptom on
their hands, so some of the workers used chloromycetin
(chloramphenicol) to treat their hands in order to avoid
the itching, and, as a result, the prawns were polluted".
The 2002 FDA report regarding chloramphenicol testing in
imported seafoodref
contains this discussion: "Until recently, the sensitivity
of the methodology to detect chloramphenicol in shrimp
could find the drug down to 5 parts per billion (ppb).
Recently, Canada, and the European Union (EU), have
refined their methods to detect even lower levels, and,
have taken action on food products from China and Viet Nam
found to be contaminated by chloramphenicol. The FDA has
modified its methodology to confirm chloramphenicol levels
in shrimp and crayfish to 1ppb and is further modifying
the methods to detect 0.3 ppb, which will place the U.S.
methodology in line with Canada and the EU. The new
methodology for testing for chloramphenicol to the level
of 1 ppb will be used to test imported shrimp and crayfish
that are suspected to contain chloramphenicol. FDA will
detain, and refuse entry to, any product it identifies,
and confirms, as containing chloramphenicol". On 5-6 Jun
2002, a senior delegation of Chinese officials met with
the FDA to discuss the issue of chloramphenicol residues
in shrimp and crayfish. The delegation informed the FDA
that, on 5 Mar 2002, China banned the use
of chloramphenicol in animals and animal feeds. They also
informed the FDA that they are initiating testing of
shrimp, crayfish, and other animal-derived foods intended
for export, to ensure the absence of chloramphenicol and
other drug residues. The FDA and China exchanged
information on testing methodologies. The FDA informed the
Chinese officials that the Agency would take enforcement
action against products found to be in violation.
tiamphenicol
azidamfenicol
florfenicol
(Aquaflor® and Nuflor®)
pleuromutilin
(Econor®) : a natural antibiotic diterpene
that binds to the ribosomal peptidyl transferase
centre (PTC) of the 50S ribosomal subunit with its
tricyclic mutilin core positioned in a tight pocket at the
A-tRNA binding siteref
and interacts with domain V of 23S RNAref.
It is effective against resistant mycoplasma infection in
immunocompromised patientsref
azamulin
[14-O-(5-(2-amino-1,3,4-triazolyl)thioacetyl)-dihydromutilin]
is an azole derivativeref
retapamulin
tiamulin / 81.723 HFU
is used in the control and treatment of veterinary
gram-positive and gram-negative pathogens, with a
particular emphasis on infections in swineref.
It has exceptional activity (MIC < 1 µg/ml) against
anaerobic bacterial species, intestinal spirochetes, and
Mycoplasma sppref
macrolides have been
introduced in 1952 (against most Bacteria
; bactericidal for a few Gram +ve Bacteria) :
macrocylic (12÷22 carbon atoms) lactones linked through
glycoside bonds with amino sugars. Binding to the rRNA 23S
(P8 protein ?) in the 50S ribosomal subunit inhibits
elongation of the protein by peptidyl transferase or
prevents translocation of the ribosome or both.
erythromycin
base (E-Mycin®,
Ilotycin®,
...),
estolate
(Ilosone®),
gluceptate
(Ilotycin
Gluceptate®), ethylsuccinate (E.E.S.®,
...) + sulfisoxazole or lactobionate (Erythrocyn
Lactobionate-I.V.®) ; Benzamycin®,
Persa-Gel®, ... in combination with benzoyl
peroxide. From Streptomyceserythreus : not effective
against Enterobacteriaceae. Bactericidal in vitro
at high concentrations.
spiramycin (Rovamycine®,
Ulcar®) for treatment of toxoplasmosis and
cryptosporidiosis
Cross-resistance to macrolides can arise from use of tylosin
and virginiamycin as additives in animal feeding.
third-generation macrolides / 3-ketolides (a
3-keto group replacesL-cladinose
group of macrolides)
telithromycin
(Ketek®) is 99% effective in vitro
against Streptococcus
pneumoniae. i.v. only, may cause nausea
and vomiting, and acute
hepatitisref.
It inhibits secretion of IL-1b
and TNF-aref
fidaxomicin (previously
referred to as OPT-80), a macrocyclic antibiotic, is
more active in vitro than vancomycin, by a factor of
approximately 8, against clinical isolates of C.
difficile, including NAP1/BI/027 strains
clindamycin
(Cleocin®, Dalacin C®) pediatric
(Cleocin Pediatric®),
topical (Cleocin T®) or topical phosphate
(Cleocin Phosphate®, Actiza®) is
7-chloro-7-deoxylincomycin, effective against Gram +ve Bacteria and Gram -ve Neisseria, Hemophilus
influenzae,Bacteroides
spp.). Velac® in combination
with tretinoin.
Cross-resistance to lincosamides can arise from use of tylosin
and virginiamycin as additives in animal feeding.
streptogramins
(introduced in 1962) bind 50S impairing both early peptide
chain elongation and late peptide chain extrusion => bacteriostatic. They are a mix of
:
A (macrolide)
dalfopristin
B (cyclic peptide)
quinupristin
pristinamycin / RP
59500 = quinupristin + dalfopristin (Q/D)
(Synercid®) : they are semisynthetic
derivatives of pristinamycins.
i.v. only, not active against E.faecalis,
efficacy for bacteremia/endocarditis not demonstrated
Cross-resistance to streptogramins can arise from use of tylosin
and virginiamycin as additives in animal feeding.
viomycin (Viocin®)
: a basic polypeptide antibiotic administered
intramuscularly (along with other drugs) in the treatment of
Mycobacterium
tuberculosis.
May cause ototoxicity
nocathiacins are
cyclic thiazolyl peptides with inhibitory activity against
gram-positive bacteria
BMS-249524 (nocathiacin I), identified from
screening a library of compounds against a multiply
antibiotic-resistant Enterococcus faecium strain,
was used as a lead chemotype to obtain additional
structurally related compounds
2 more water-soluble derivatives, BMS-411886
and BMS-461996ref
The 23S rRNA A2058G alteration mediates macrolide,
lincosamide, and streptogramin B resistance in the
bacterial domain and determines the selectivity of macrolide
antibiotics for eubacterial ribosomes, as opposed to eukaryotic
ribosomes. However, this mutation is associated with a disparate
resistance phenotype: it confers high-level resistance to
ketolides in mycobacteria but only marginally affects ketolide
susceptibility in streptococci. Mutational alteration of the
polymorphic 2057-2611 base pair from A-U to G-C in isogenic
mutants of Mycobacterium
smegmatis significantly affects susceptibility to
ketolides but does not influence susceptibility to other
macrolide antibiotics. In addition, the 2057-2611 polymorphism
determines the fitness cost of the 23S rRNA A2058G resistance
mutation. Polymorphic nucleotides mediate the disparate
phenotype of genotypically identical resistance mutations and
provide an explanation for the large species differences in the
epidemiology of defined drug resistance mutationsref.
tetracyclines (bacteriostatic) : products of the
aromatic poliketide biosynthetic pathway in Streptomyces,
act by binding the 30S subunit and blocking the
binding of aminoacyl tRNA to the A site on the ribosome.
As most Bacteria possess an active transport
system for tetracycline that allows intracellular
accumulation of the antibiotic at concentrations 50 times
as great as that in the medium, a blood level which is
harmless to animal tissues can halt protein synthesis in
invading Bacteria.
doxycycline / doxycyclin
(Bassado®, Doxymycin®, Doryx®,
Vibra®, Vibra-Tabs™, Vibramycin®,;
low-dose oral formulation for dermatology : Oracea®)
does not cause the photosensitizing reaction that may
occur with conventional tetracycline. Divalent cations,
gluten products, and calcium do not materially interfere
with its absorption. Since doxycycline is a highly
lipid-soluble antimicrobial, a loading dose is necessary
for optimal results. Therefore, for moderate or severe
infections, the usual dose, given intravenously or
orally, should be doubled for 72 hours and then reduced
to the usual dose (ie, 200 mg every 12 hours reduced to
100 mg every 12 hours). The entire dose (intravenous or
oral) may be given once daily. Doxycycline should be
taken with food and should not be given to pregnant
patients or young children
lymecycline
(Tetralysal®)
methacycline
(Rondomycin®)
minocycline
(Minocin®, ...) is 10-folds more
lipid-soluble than conventional tetracycline whereas
doxycycline is only 5-folds more lipid-soluble. The
clinical importance of this characteristic is that
minocycline has particularly good tissue penetration and
excellent CNS penetration. It is effective also against
MRSA. Food and divalent cations interfere minimally or
not at all with oral absorption, and photosensitizing
reactions are rare. Because of its high
lipid-solubility, the drug is maintained in high
concentrations in middle ear fluid; thus, it has been
implicated in vestibular side effects. It should not be
given to pregnant patients or young children.
Minocycline-induced hyperpigmentation can be severely
disfiguring and is more likely to occur in certain
populations of patients (e.g., those with pemphigus,
pemphigoid, or atopic dermatitis). It is important to
recognize this condition early and offer an alternative
treatment, since symptoms can take months to years to
resolve once the drug is withdrawn. There are 4 types of
minocycline-induced cutaneous hyperpigmentationref:
type I occurs on the face within inflammatory
tissue
type II occurs on the arms and legs in a
circumscribed distribution
type III appears diffusely muddy-brown on
sun-exposed skin
Some newly discovered members of the tetracycline family
(e.g. chelocardin) have been shown to act by
inserting into the bacterial membrane, not by inhibiting
protein synthesis.
Side effects : contraindicated
during
pregnancy
and before age 8; renal toxicity (75%),
hepatotoxicity, neurotoxicity, systemic
arterial hypertension,
hirsutism,
gingival hyperplasia, GI toxicity, Steven-Johnsons's
syndrome
glycylcyclines are
tetracycline derivatives
tigecycline / GAR936
: not transported by the tetracycline efflux pumps (Staphylococcus aureus,
including MSSA, GISA, MRSA, Streptococcus
pneumoniae, includnig PRSP, VRE, Acinetobacter,
Bacteroides fragilis, Acinetobacter, Enterobacter;
inactive against Proteus spp. and
P.aeruginosa). Serum levels sometimes lower than
the MIC, but high tissue distribution far above the MIC,
low inhibitory quotient in the blood but a high
inhibitory quotient in tissue. Nausea and vomiting is
the most commonly reported adverse eventref.
Appropriate targets :
peritonitis, but in combination therapy if suspicion
of Pseudomonas involvement (e.g. postoperative
peritonitis, peritonitis treated with antibiotics before
surgery)
nosocomial pneumonia (post-operative pneumonia,
early-onset VAP, VAP due to Enterobacter, Acinetobacter)
severe community-acquired pneumonia
complicated skin and skin tissue infections
(postoperative cellulitis)
infections with resistant Gram-positive pathogens
(VRE, MRSA, GISA, VRSA)
infections with resistant Gram-negative pathogens
(Enterobacter, Acinetobacter)
catheter-related bacteremia ?
endocarditis ?
aminoglycosides
(initiation inhibitors) have been introduced in 1950
(against Gram -ve Bacteria
and just a little Gram +ve Bacteria).
They
bind
to the S12 (a.k.a. P10) protein in the 30S subunit of the
bacterial ribosome blocking the binding of initiator N-fMet-
tRNAMet to the ribosome and preventing the normal
dissociation of ribosomes into their subunits, leaving them
mainly in their 70S form, impairing polysomes formation. It
stabilizes aa-tRNA on the ribosome both with a cognate and
with a near-cognate codon in the A site by altering the
rates of GTP hydrolysis by elongation factor Tu (EF-Tu),
resulting in almost identical rates of GTP hydrolysis and
virtually complete loss of selectivity. The difference in
spelling (-micin or -mycin) reflects the
isolation from Streptomyces spp. or Micromonospora
spp., respectively. They can be classified according to
their aminocyclitol :
only topical PO use before GI tract surgery (no if
hemorrhages are suspected !) : very toxic when
administered IV !
kanamycin sulfate (Kantrex®)
: restricted spectrum, but if given IV it is
effective even against Staphyloccous
aureus
amikacin / BBK8
(Amikin®)
arbekacin (Habekacin®)
bekanamycin
dibekacin
tobramycin
(Aktob®, Tobral®, Tobrex®,
Nebcin®; ZyLet® in
combination with loteprednol)
intermediate spectrum
neomycin sulfate
(Ribostamycin®, Mycifradin®,
Neo-Fradin®; Neosporin
G.U. Irrigant®
in combination with polymixin
B; Neosporin® in
combination with bacitracin
and polymixin
B; Neosporin® in
combination with gramicidin and polymixin B) or undecylenate
ribostamycin
framicetin (topical use)
paramomycin
(Humatin®) : effective even against Staphyloccous aureus
gentamicin
sulfate (Gentalyn®, Garamycin®,
Genoptic®, Gent-AK®, Gentacidin®,
...) : broad spectrum. Gentamicin
surgical implant is a biodegradable leave-behind
implant impregnated with gentamicin, and is
indicated as an adjunct to systemic antibiotic
therapy for the treatment and prevention of
post-surgical acquired infection in both hard and
soft tissues. The product was developed using
Innocoll's proprietary collagen-based technology,
CollaRx®, and
has been approved in 49 countries. It is marketed
under the following different trade names: Collatamp® G, Collatamp® EG, Sulmycin® Implant, Garamycin® Schwamm, Duracol®, Duracoll®, Gentacol®, Gentacoll®, Garacol®, Garacoll® and Cronocol® in Europe, Central and South America,
Middle East, Africa and Asia
netilmicin
(Netromycin®) : semisynthetic
sisomicin /
thiostrepton (Siomycin®) derived
from Micromonospora inyoensis, closely
related to the C1a component of the
gentamicin complex; it is bactericidal for many
gram-negative and some gram-positive organisms,
having a range of activity similar to that of
gentamicin.
sisomicin sulfate : the sulfate salt of
sisomicin, used in the treatment of infections
caused by susceptible gram-negative organisms;
administered intravenously or intramuscularly.
Resistance genes : aminoglycoside
acetylases (AAC), adenylylases (AAD), and phosphorylases
(APH).
Side effects :
reversible acute
renal
failure
lasting up to 20 days after suspension (evaluate
[Ala-aminopeptidase]urine]
damage to the vestibulocochleary (auditory) nerves
=> permanent deafness
and/or temporaneous dizziness lasting 2-6 months.
gentamicin-induced hearing loss averages 8% for a short
course of therapyref
but may be higher in developing countries, where
aminoglycosides are frequently the only affordable
antibiotics and are sold over the counter. No therapy
presently exists to prevent ototoxicity. Animal models
suggest that ototoxicity is caused by reactive oxygen
species and is attenuated by antioxidantsref.
Salicylate is a clinically promising antidoteref
that can be administered as aspirin. 195 patients received
80 to 160 mg of gentamicin twice daily by intravenous
infusion (generally for 5 to 7 days) and were randomly
assigned to receive 14 days of supplementation either with
3 g of aspirin per day, divided into 3 doses (89
patients), or with placebo. The incidence of hearing loss
in the placebo group was within the anticipated range
(13%) but was significantly lower in the aspirin group
(3%). The efficacy of the gentamicin therapy was not
affected by the administration of aspirin. However,
gastric symptoms were more common in the aspirin groupref.
hexamethylenamine / hexamine / methenamine
mandelate and hippurate (Urex®, Hiprex®,
Mandelamine®) : the compound decomposes in water at
acidic pH according to the following reaction :
quinolones have been
introduced in 1962 (bactericidal against
Gram -ve Bacteria) at low
doses inhibit the religation of the doubly cleaved DNA whose
5' ends are tethered on 2 Tyr residues in type II bacterial
topoisomerases [both DNA gyrase (1 Tyr per GyrA
subunit in the active (GyrA)2(GyrB)2
tetramer) and DNA topoisomerase IV], while at high doses (e.g.
those present in urine) inhibit N-methyltransferase
1st generation quinolones : urinary
antiseptic, uneffective against Pseudomonas aeruginosa
2nd generation quinolones : urinary
antiseptic, effective even against Pseudomonas
aeruginosa
cinoxacin (Cinobac®,
Cinoxacin®)
flumequine
novobiocin (Albamycin®)
binds the B subunit of DNA gyrase inhibiting ATP
hydrolysis
pipemidic acid
(Dolcol®, Pipram®)
rosoxacin (Eradacil®)
3rd generation / 6-fluoroquinolones (FQ) :
systemic distribution (expecially used against bacterial
respiratory and UTIs, prostatitis, osteomyelitis,
and Salmonella
typhi).
Fluoroquinolones,
broad-spectrum antibiotics that are widely perceived to have
favorable adverse-effect profiles, have become the most
prescribed antibiotics in the USAref.
From 1995 to 2002, the number of fluoroquinolone
prescriptions in the USA increased by a factor > 3,
reaching about 22 million prescriptions per yearref.
The available fluoroquinolones have well-established
differences in antimicrobial activity, but their disparate
adverse-effect profiles are increasingly being recognized.
Serious adverse events have led to the withdrawal or
restriction of several fluoroquinolones in recent yearsref
ciprofloxacin
(Ciloxan®, Cipro®, Ciproxin®)
: PO, i.v.. In European ICUs it is effective against 89%
of MSSA, 7% of MRSA, 92% of E.coli, 78% of Enterobacter
spp., and 73% of P.aeruginosaref.
Ciprofloxacin is absorbed from the gastro-intestinal
tract. Oral bioavailability is approximately 70% and a
peak plasma concentration is achieved 0,5 to 2 hours
after oral dosing. Absorption may be delayed by the
presence of food, but is not substantially affected
overall. The plasma half-life is about 3,5 to 4,5 hours
and there is evidence of moderate accumulation.
Half-life may be prolonged in severe renal failure and
to some extent in the elderly. Plasma protein binding
ranges from 20 to 40%. Ciprofloxacin is widely
distributed in the body and tissue penetration is
extensive. It appears in the cerebrospinal fluid, but
the concentrations are only about 10% of those in the
plasma when the meninges are not inflamed. Ciprofloxacin
crosses the placenta, and is distributed into the breast
milk. High concentrations are achieved in the bile.
Ciprofloxacin is eliminated principally by urinary
excretion, but non-renal clearance may account for about
a third of elimination and includes hepatic metabolism,
biliary excretion, and possibly transluminal secretion
across the intestinal mucosa. Urinary excretion is by
active tubular secretion as well as glomerular
filtration and is virtually complete within 24 hours.
About 40 to 50% of an oral dose is excreted unchanged in
the urine and about 15% as metabolites Faecal excretion
over 5 days has accounted for 20 to 35% of an oral dose.
At least 4 active metabolites have been identified. Oxociprofloxacin
appears to be the major urinary metabolite, and sulphociprofloxacin
is the primary faecal metabolite. Only small amounts of
ciprofloxacin are removed by haemodialysis or peritoneal
dialysis. Polymorphism of the mexR gene which is
involved in the resistance to drugs like ciprofloxacin.
Mutations in mexR result in increased resistance to
multiple antibiotics due to overexpression of this
efflux system. The MexR product contains 147 amino acids
with a molecular mass of 16,964 Da.
ofloxacin (Floxin®,
Ocuflox®, Tarivid®)
long-acting (1 / die) and broad spectrum
fleroxacin (Megalone®)
lomefloxacin
(Maxaquin®)
pefloxacin (Peflocin®)
temafloxacin
(Omniflox®), recalled because of hemolysis,
renal failure, and hypoglycemiaref
(Rubinstein E. History of quinolones and their side
effects. Chemotherapy 2001;47:Suppl 3:3-8, 44)
uvofloxacin
clinafloxacin
difloxacin (Dicural®)
enrofloxacin for
veterinary use
garenofloxacin
gatifloxacin (Tequin®)
: limited data suggest that as compared with other
currently available fluoroquinolones, gatifloxacin
(Tequin, Bristol-Myers Squibb) may be uniquely associated
with increased risks of both hypoglycemia and
hyperglycemiaref1,
ref2,
ref3,
ref4,
ref5,
ref6,
ref7,
ref8,
ref9,
ref10,
ref11,
ref12,
ref13,
ref14,
ref15
(Tailor SA, Simor AE, Cornish W, Phillips E, Knowles S,
Rachlis A. Analysis of spontaneous report of hypoglycemia
and hyperglycemia associated with marketed systemic
fluoroquinolones made to the Canadian Adverse Drug
Reaction Monitoring Program. Can J Hosp Pharm
2004;57:12-17; Frothingham R. Gatifloxacin associated with
a 56-fold higher rate of glucose homeostasis abnormalities
than comparator quinolones in FDA Spontaneous Reporting
Database. In: Program and abstracts of the 44th
Interscience Conference on Antimicrobial Agents and
Chemotherapy, Washington, D.C., October 30–November 2,
2004. Washington, D.C.: American Society for Microbiology,
2004:19; Létourneau G, Morrison H, McMorran M.
Gatifloxacin: hypoglycemia and hyperglycemia. Can Adverse
React Newsl 2003;13(3):1-2). Although the mechanism of
these apparently competing adverse effects is not fully
understood, studies in animals suggest that although
gatifloxacin can promote insulin release and hypoglycemia
by blocking the ATP-sensitive potassium channels of
pancreatic islet cells, it can also trigger the
vacuolation of pancreatic b
cells, leading to reduced insulin levels and hyperglycemiaref1,
ref2,
ref3,
ref4.
Evidence that gatifloxacin causes dysglycemic effects in
humans consists of data from case reports, small studies
in healthy volunteers or hospital inpatients, and one
small post-marketing studyref1,
ref2,
ref3,
ref4,
ref5
(Grasela DM, Lacreta F, Kollia G, Randall D, Stoltz R,
Berger S. Lack of effect of multiple-dose gatifloxacin
(GAT) on oral glucose tolerance (OGTT), glucose and
insulin homeostasis, and glyburide pharmacokinetics (PK)
in patients with type II non-insulin-dependent diabetes
mellitus (NIDDM). In: Program and abstracts of the 39th
Interscience Conference on Antimicrobial Agents and
Chemotherapy, San Francisco, September 26–29, 1999.
Washington, D.C.: American Society for Microbiology,
1999:11; Schwarzbard L, Lodise TP, Lomaestro BM, Smith R.
Comparison of glucose intolerance (GI) between
gatifloxacin (G) and levofloxacin (L) in elderly,
hospitalized patients. In: Program and abstracts of the
45th Interscience Conference on Antimicrobial Agents and
Chemotherapy, Washington, D.C., December 16–19, 2005.
Washington, D.C.: American Society for Microbiology,
2005:463). These studies yielded conflicting conclusions
regarding the effects of gatifloxacin on blood glucose
levels, but some reports strongly suggest the existence of
a causal relationref.
For example, one recent report described two patients in
whom profound hyperglycemia (glucose, 942 to 1456 mg/dl)
developed shortly after gatifloxacin therapy but who had
no subsequent evidence of diabetesref.
The use of gatifloxacin among outpatients is associated
with an increased risk of in-hospital treatment for both
hypoglycemia and hyperglycemia : so it should not be used
in diabetics and other high risk patients)ref
gemifloxacin
mesylate (Factive®)
grepafloxacin
(Raxar®) : recalled because of QT-interval
prolongationref1,
ref2
(Rubinstein E. History of quinolones and their side
effects. Chemotherapy 2001;47:Suppl 3:3-8, 44)
levofloxacin
(Levaquin®, Quixin®, Tavanic® in
Europe, Cravit® in Asia) (i.v.)
marbofloxacin for
veterinary use
moxifloxacin (Avelox®,
Avalox®) (i.v.) has recently shown promise in a
murine model of Mycobacterium
tuberculosisref
sparfloxacin (Spara®,
Zagam®) : recalled because of QT-interval
prolongationref1,
ref2
(Rubinstein E. History of quinolones and their side
effects. Chemotherapy 2001;47:Suppl 3:3-8, 44)
tosufloxacin
trovafloxacin
(Trovan®) : recalled because of hepatotoxicityref1,
ref2.
ABT-492
Side effects :
fluoride causes cartilage alterations (use is forbidden
during pregnancy and in children) => bilateral acute or
chronic Achilles
calcaneal
tendinitis
=> tendon rupture, more common in patients over 60
years of age. The latency period between the start of
treatment and the appearance of the first symptoms range
from 1 to 510 days with a median of 6 days. Most patients
recover within 2 months after cessation of therapy, but
26% have not yet recovered at followup.
relapse in patients who suffered juvenile epilepsy
photosensitivity (use sunglasses)
neurologic impairment
incidence of convulsions and anxiety due to action as
GABAA
antagonists
nausea and vomiting
Simultaneous administration of antacids
containing magnesium or aluminium and
ciprofloxacin or other quinolones decreases the gastrointestinal
absorption of those antibacterial agents due to drug-cation
chelationref.
rifampin (Rifadin®,
Rimactane®) is a semisynthetic derivative of rifamycin
B active against Gram +ve Bacteria,
Mycobacterium
tuberculosis
and some Gram -ve Bacteria.
Rifampicin acts quite specifically against the b subunit of the bacterial RNA
polymerase and apparently blocks the entry of the first
nucleotide which is necessary to activate the polymerase.
It is effective orally and crosses the blood-brain
barrier, so it is useful for treatment of bacterial
meningitis.
CBR703 series compounds inhibit known catalytic
activities of RNAP (nucleotide addition, pyrophosphorolysis,
and Gre-stimulated transcript cleavage) but not translocation
of RNA or DNA when translocation is uncoupled from catalysis.
While screening for a new broad-spectrum antibiotic, J&J
researchers discovered that a class of compounds called diarylquinolines
worked against Mycobacteria smegmatis : chemical
tinkering led them to the even more potent R207910. It potently
inhibits both drug-sensitive and drug-resistant Mycobacterium
tuberculosis in vitro (MIC 0.06 µg/ml). In mice, R207910
exceeds the bactericidal activities of isoniazid and rifampin by
at least 1 log. As expected, resistance to R207910 developed
when given to mice as a monotherapy, but the J&J team's
experiments with mice have convinced leading TB researchers that
swapping the drug for 1 of the 3 in the most popular triple-drug
combination now used would delay development of resistant
strains. Substitution of drugs included in the WHO's first-line
tuberculosis treatment regimen (rifampin, isoniazid and
pyrazinamide) with R207910 accelerates bactericidal activity,
leading to complete culture conversion after 2 months of
treatment in some combinations, about half the time it takes
using the standard treatmen. A single dose of R207910 inhibits
mycobacterial growth for 1 week. Plasma levels associated with
efficacy in mice are well tolerated in healthy human volunteersref.
Human versions of the synthase are not affected by the drug and
initial studies in humans show that injections are safe, at
least for short periods of time. Trials in people who are
actively sick are now under way, although it will be many years
before the drug makes it to the market. One of the drugs in the
standard tuberculosis cocktail does not mix well with many HIV
drugs, but a regimen with compound J would be fine. Another
potential bonus is that the compound may be useful against
latent tuberculosis, as it kills even when the bacteria are not
actively reproducing. Although the outbreaks of
multidrug-resistant tuberculosis (MDR-TB) that afflicted New
York City in the 1990s were relatively minor when compared to
the burden of global tuberculosis [HN1]ref1,
ref2,
they served to raise public and political awareness. The result
was that for the first time TB control was included on the
agenda of the G8 economic summit meetings. The world's leaders
lent their support to that of nongovernmental organizations,
such as the Global Alliance for TB Drug Development (GATDD) and
the WHO, by encouraging industry and academia to engage in the
development of new drugs to treat this chronic respiratory
disease. This was a crucial event given that TB claims up to 2
million lives annually worldwide, blights myriad communities
principally in developing countries, and that no new TB drugs
have been discovered in the past 40 yearsref.
The current treatment for TB recommended by WHO--known as
directly observed therapy short-course (DOTS)--requires patients
to adhere to a 3- or 4-drug regimen comprising isoniazid,
rifampin, pyrazinamide, and/or ethambutol for a minimum of 6
months. Many patients fail to complete therapy because of drug
side effects and the complicated drug regimen, resulting in
relapse--often in the form of MDR-TB, which is even more
difficult to treat. An ideal new TB drug should be highly
active, so that treatment duration can be reduced to <3
months; it should kill the persistent bacilli that might
otherwise reactivate later in life; and it must show activity
against MDR-TB strains. Optimally, a new therapeutic agent would
be specific for Mycobacterium tuberculosis and also
compatible with existing TB drugs, because combination therapy
will remain mandatory to combat this major killerref.
Its lead compound was identified by adopting a medium-throughput
screening approach using live mycobacteria rather than the more
popular target-based, high-throughput screening that uses
robotics to screen millions of compounds for inhibitors of
critical functions such as key enzyme activities. This proved a
very astute decision because it avoided problems with drug
permeability (which always affect the target-based screens at a
later stage) by identifying active compounds that freely entered
the mycobacteria. After optimization by synthetic chemistry, the
investigators were left with 20 interesting drug candidates; of
these, R207910 showed the best activity profile. R207910 is
bactericidal and exquisitely active against a broad range of
mycobacteria [HN11], displaying little or no activity against
the other microorganisms tested. Crucially, R207910 is active
against both the drug-sensitive and drug-resistant forms of M.
tuberculosis. This organic compound of 555.51 daltons,
which contains both planar hydrophobic moieties and
hydrogen-bonding acceptor and donor groups, displays perfect
drug-like features that satisfy most of Lipinski's rules for
good drug candidatesref.
Pharmacokinetic and pharmacodynamic studies in different animal
models have confirmed the excellent drug-like properties of
diarylquinolines. To identify the target of R207910, mutants of
M. tuberculosis were isolated and the related
faster-growing organism M. smegmatis that were resistant
to R207910, and characterized by whole-genome sequencing. 2
different missense mutations in the atpE gene, which encodes the
C subunit of ATP synthase, the enzyme that uses the
transmembrane proton-motive force to generate ATP for the cellref.
Model of the mycobacterial ATP synthase showing the position of
mutations that confer resistance to the diarylquinoline drug
R207910ref.
ATP synthase has 2 major structural domains, F0 and F1,
that act as a biological rotary motorref.
F1 is composed of 9 subunits (a3,
b3, g,
d, e) and
is located in the cytoplasm, where it generates ATP. F0
spans the cytoplasmic membrane and contains 13 to 15 subunits
(a, b2, c9-12) arranged as a symmetrical
disc. The F0 and F1 domains are linked by
subunits g, e,
d, and b2. Rotation of the
transmembrane disc and the central stalk is driven by the
proton-motive force. The c subunit is an a-helical
hairpin structure with a short connecting loop. Notably, the
A63P mutation is very near E60, the glutamic acid residue whose
carboxyl group is protonated during proton translocation. The
proton-motive force fuels the rotation of the transmembrane disk
and the central stalk, which in turn modulates the nucleotide
affinity in the catalytic b subunit,
leading to the production of ATP. The c subunit has a hairpin
structure with 2 a helices and a
short connecting loop. The 2 mutations affect the
membrane-spanning a helices of the
ATP synthase c subunit and may restrict binding of R207910 to
the enzyme. Although biochemical confirmation is now required,
it is possible that the drug impedes assembly of the mobile disk
or interferes with its rotational properties, leading to
inadequate synthesis of ATP. A puzzling feature of R207910 is
its exceptional specificity for mycobacteria. ATP synthase is a
ubiquitous enzyme found in most living organisms, including
humans. There is very limited sequence similarity between the
mycobacterial and human AtpE proteins, which bodes well for the
safety of the compound, as borne out by the phase I study in
human volunteers. The mycobacteria-specific activity of R207910ref
may also be the consequence of limited sequence similarity among
bacterial AtpE proteins. However, those antitubercular agents
that show highly restricted activity (such as isoniazid,
ethionamide, and pyrazinamide) are all prodrugs requiring
activation by a mycobacterial enzymeref.
Although its chemical structure gives no clues to potential
activation sites, R207910 may also prove to be a prodrug. Mouse
studies already show that this compound can greatly shorten the
duration of therapy, both alone and in association with current
antitubercular agents. Furthermore, the equally remarkable
activity of R207910 against M. ulcerans--the
agent of an emerging human disease called Buruli ulcerref,
for which surgery is the only cure--also raises expectations for
a safer treatment for this disfiguring afflictionref.
sulfonamides (bacteriostatic). They were
introduced as chemotherapeutic agents by Domagk, Mietsch and
Klarer in 1935, who showed that one of these compounds (the
substituted sulfanilamide ...
... derived from the intracellular degradation of the
bacterial stain 4-sulfonamide-2',4'-diaminobenzol or red
Prontosil ...
...) had the effect of curing mice with infections
caused by b-hemolytic Streptococcus
spp.. Chemical modifications of the compound sulfanilamide
gave compounds with even higher and broader antibacterial
activity. Bacteria which are almost always sensitive
to the sulfonamides include Gram +ve Streptococci
(b-hemolytic Streptococci and
Streptococcus pneumoniae (even if some Streptococci
can assume folate from the environment, bypassing the
synthesis blockage)) and Gram -ve Bacteria
(E. coli, Neisseria meningitidis). They cross
the blood-brain barrier.
rapidly absorbed and rapidly eliminated sulfonamides
sulfisoxazole diolamine (Gantrisin®,
...; Pediazole® in combination with
erythromycin ethylsuccinate for otitis media; ? in
combination with phenazopyridine
for UTIs)
sulfacetamide (AK-Sulf®, Bleph-10®,
Cetamide®, Isopto Cetamide®,
Klaron®, Sebizon®, Sodium Sulamyd®,
Sulf-10®, Sultrim®... ; Sulfacet-R®
in combination with sulfur)
silver sulfadiazine (SSD) (SSD®,
Silvadene®, ...)is the most commonly used
topical antibacterial agent for the treatment of burn
wounds. It has many clinical advantages, including a
broad spectrum of antimicrobial activity, low toxicity,
and minimal pain on application. The current formulation
of silver sulfadiazine contains a lipid soluble carrier,
polypropylene glycol, that has certain disadvantages,
including pseudo-eschar formation and the need for twice
daily application. A new formulation of silver
sulfadiazine in a water soluble gel, poloxamer 188, can
be applied once a day and its water solubility allow
easy application and removal. Silver compounds are used
widely as effective antimicrobial agents to combat
pathogens (bacteria, viruses and eukaryotic
microorganisms) in the clinic and for public health
hygiene. Silver cations (Ag+) are
microcidal at low concentrations and used to treat
burns, wounds and ulcers. Ag is used to coat catheters
to retard microbial biofilm development. Ag is used in
hygiene products including face creams, "alternative
medicine" health supplements, supermarket products for
washing vegetables, and water filtration cartridges. Ag
is generally without adverse effects for humans, and argyria
is
rare and mostly of cosmetic concern.Resistance to silver
compounds as determined by bacterial plasmids and genes
has been defined by molecular genetics. Silver
resistance conferred by the Salmonella plasmid
pMGH100 involves nine genes in three transcription
units. A sensor/responder (SilRS) two-component
transcriptional regulatory system governs synthesis of a
periplasmic Ag(I)-binding protein (SilE) and two efflux
pumps (a P-type ATPase (SilP) plus a 3-protein
chemiosmotic RND Ag(I)/H+ exchange system (SilCBA)). The
same genes were identified on five of 19 additional IncH
incompatibility class plasmids but thus far not on other
plasmids. Of 70 random enteric isolates from a local
hospital, isolates from catheters and other Ag-exposed
sites, and total genomes of enteric bacteria, 10 have
recognizable sil genes. The centrally located 6 genes
are found and functional in the chromosome of Escherichia
coliK-12,
and
also occur on the genome of E. coli O157:H7.
The use of molecular epidemiological tools will
establish the range and diversity of such resistance
systems in clinical and non-clinical sources
sulfadoxine
(Fansidar® in combination with pyrimethamine)
sulfamazole
sulfamazone
sulfamethizole (Thiosulfil Forte®)
sulfametopirazine
sulfametoxypiridazine
sulfametrol
succinylsulfathiazole
Side effects : renal and hepatic
toxicity (they may cause kernicterus in newborns by
displacing bilirubin from plasma proteins in the face of
increased bilirubin production from fetal erythrocyte
turnover, decreased bilirubin conjugation, acidosis, and
decreased blood-brain barrier), agranulocytosis,
thrombocytopenia, acquired
aplastic
anemia,
hemolytic anemia in G6PD- patients, GI toxicity, Steven-Johnsons's
syndrome
=> Lyell's
syndrome.
p-aminosalicylic
acid (PAS) competes with p-aminobenzoic acid
(PABA)
pyrimethamine /
2,4-diamino-5-(p-chlorophenyl)-6-ethyl-
pyrimidine (Fansidar® in combination with
sulfadoxine; Maloprim®,
Deltaprim® in combination with dapsone)
tetroxyprim
bacterial
DHF reductase inhibitors :
trimethoprim (TMP)
(Trimplex®, Proloprim®, ...) (bacteriostatic) : it is often
administered together with bacteriostatic
sulfamethoxazole
as co-trimoxazole /
cotrimoxazole / TMP/SMX (Bactrim®, Cotrim®,
Cofatrim®, Primsol®, Septra®,
...), becoming bactericidal
against Gram -ve Bacteria;
Polytrim® in combination with polymyxin B
icloprim
albomycin is an analog of
ferrichrome, made by some Fungi.
ClpP (a core unit of a major bacterial protease
complex) inhibitors : acyldepsipeptides have
antibacterial activity against Gram-positive bacteria in
vitro and in several rodent models of bacterial
infection. The acyldepsipeptides are active against isolates
that are resistant to antibiotics in clinical application. Clp
is usually tightly regulated and strictly requires a member of
the family of Clp-ATPases and often further accessory proteins
for proteolytic activation. Binding of acyldepsipeptides to
ClpP eliminates these safeguards. The
acyldepsipeptide-activated ClpP core is capable of proteolytic
degradation in the absence of the regulatory Clp-ATPases. Such
uncontrolled proteolysis leads to inhibition of bacterial cell
division and eventually cell deathref
diphenyl urea
caragenin
LN-I-284
PZ-601
ME-1036
D-159a
igromycin A
cethromycin
BAL 3076 (monobactam + b-lactamase
inhibitor)
friulimicin
SASP
CBR-2092 : rifamicin SV + fluoroquinolone
Adverse events :
direct toxicity
cardiac effects - prolongation of the QTc
intervalref
renal toxicityref1,
ref2.
Very ill patients may have impaired absorption, altered
clearance due to renal dysfunction (increased risk of adverse
events, increased antibiotic doses are often required in burn
patients)ref
superinfection
antibiotics can change the normal microbial floraref,
allowing overgrowth of opportunistic infections (e.g. Candida
spp. in the gastrointestinal tract)ref
Leading parenteral antibiotic products in the global hospital market
for prophylaxis and therapy in USA and 5 European countries
Current status of antibacterial resistance in the ICU (NNIS, 2002 vs
1997-2001)ref
:
The quantity of antimicrobials used in food animals in Denmark has
declined 54% from peak use between 1994 and 2001. Before the
programme began, most pigs and broiler chickens in Denmark were
given antimicrobials, such as avilamycin, avoparcin,
tylosin, streptogramins and virginiamycin,
for most of their lives. After withdrawal, average use declined to
0.4 days in broiler chickens (with life span of around 42 days) and
7.9 days in pigs (with life span of around 170 days). Pork
production
in Denmark has continued to increase, and effects on poultry
production were small. Ending the use of the above antimicrobials
has greatly reduced the reservoir of resistant Enterococcus faecium in the food animal
reservoir, thus reducing the reservoir of resistance genes. For
example, resistance to avilamycin, avoparcin, and streptogramins in
Enterococcus faecium isolates from broiler chickens declined
from 60-80% before withdrawal of antimicrobials to only 5-35% after.
The carboxylic
ionophoresref
are open-chained oxygenated heterocyclic rings with a single
terminal carboxyl group of moderate molecular weight (200-2000).
It is a drug for use in animals only, and there is no comparable
drug or drug category for humans. Ionophores form lipid-soluble
complexes with polar cations (K+, Na+, Ca2+
and Mg2+), have a diverse antibacterial spectra and are
produced by fungi, predominantly Streptomyces sp. These
compounds are used as anticoccidial and growth promotant feed
additives. Clinical pathology of ionophore-induced toxicityref1,
ref2,
ref3,
ref4,
ref5
includes elevated enzyme levels of muscle origin such as aspartate
transaminase (AST) and creatine phosphokinase (CPK). Other serum
enzymes that may be elevated are lactic dehydrogenase (LDH) and
alkaline phosphatase. Additonally, there may be elevations in
blood urea nitrogen and bilirubin. Serum calcium and potassium
levels may fall to life-threatening levels in horses.
Hemoconcentration may also occur. The ionophores are generally
safe at prescribed levels in intended species. Certain management
situations increase the possibility of toxicoses including
overdose, mixing errors, premix consumption, and misuse in
non-target species such as horses, adult turkeys, and dogs.
Concurrent administration of other drugs, including
chloramphenicol, erythromycin, sulfonamides and cardiac
glycosides, can potentate ionophore toxicosis.
calcium ionophores clearly have
effects in human cells. Binding of calcium can produce the signs
and symptoms of hypocalcemia and can upregulate or
downregulate cellular functions based on the role of calcium in
such processesref1,
ref2.
lasalocid / X-537A is a
divalent ionophore used in broiler chickens (Avatec®)
for the prevention of coccidiosis caused by Eimeria
sp. and in cattle (Bovatec®) for improved feed
efficiency and increased rate of weight gain. It gets into
laying hens and consequently eggs through batches of chicken
feed which have been contaminated by accident : 12% of UK eggs
tested contained residues. It is unclear if the levels in the
eggs would cause clinical or subclinical effects in humans.
However, rules of the European Union have banned lasalocid
from being added to feed that is given to laying hens.
Although this problem may have existed before, the maximum
level of lasalocid reported in eggs has risen significantly
over the past year from 620 mg/kg
in 2002 to 3450 in 2003. 2 of the 250 samples contained levels
above 3000 mg/kg. People with
cardiac arrhythmia, children, and adults who eat 3 or 4 eggs a
day are most at risk from the drug (babies under 6 months
should not be fed eggs because of the potential risk of
allergic reactions), which has been linked to increased heart
and breathing rates, paralysis and even sudden adult death
syndrome in poultry. Consumers should be aware of the
potential danger and limit their daily egg intake. Eat organic
if you can and certainly give organic eggs to children, and 2
eggs in one day would be the maximum you should have
Spray for poultry processing companies to apply to contaminated
surface of raw poultry :
10% trisodium phosphate (TSP)
0.1% acidified sodium chlorite (ASC)
0.1 and 0.5% cetylpyridinium chloride (CPC) (Cecure®),
used
since
the 1930s in mouthwashes and throat lozenges, is effective
against Campylobacter
jejuniref,
Salmonella spp., Escherichia coli,
and Listeria spp.. It has no taste or
smell, does not change the color of meat, and leaves a residue
on foods only if they contain a lot of surface fat
1% Tween 80
water (50°C at application)
Scientists of the Kosan Bioscience
have overcome an important hurdle in the race to develop new antibiotics:
they have made bacteria efficiently churn out chemicals that could
prove to be useful drugs. Over the past few decades, bacteria have
evolved to resist our antibiotics. As a result, hospitals have seen
a dramatic rise in drug-resistant infections, many of which are
fatal. To come up with new antibiotics, scientists often work with
the natural chemical defences of fungi and bacteria, altering these
natural antibiotics to make new ones. Researchers have also
attempted to genetically engineer bacteria to pump out new chemicals
directly. Although chemicals produced this way have not yet been
used to fight human disease, the approach has produced some
promising compounds, including :
lantibiotics are
ribosomally-synthesised antimicrobial peptides produced by
Gram-positive bacteria that are characterised by the presence of
lanthionine and/or methyllanthionine residues.
Other unusual post-translationally modified amino acids, most
frequently dehydroalanine and dehydrobutyrine,
can also be present. While it has been frequently suggested that
these peptides have the potential to be utilised in a wide range
of medical applications, to date no actual therapeutic
applications have been convincingly described. More recently,
however, they have been the focus of much attention as a
consequence of improved biotechnological capabilities, an
improved understanding of lantibiotic biosynthesis and mode of
action, and their high specific activity against multi-drug
resistant bacteria. Rational mutagenesis strategies
('intelligenetics') have been implemented to alter individual
residues with a view to ultimately widening the active pH range,
improve stability, and enhance binding to cell wall targets with
the ultimate aim of optimising their antimicrobial activity. It
is hoped that as a consequence of this improved knowledge the
most suitable application of individual lantibiotics will become
apparent. It should also prove possible, in the near future, to
generate tailor-made lantibiotics and utilise biosynthetic
enzymes to incorporate modified amino acids into non-lantibiotic
peptides. In the shorter term, the extensive characterisation of
lantibiotics will be instrumental in reassuring drug industry
regulators of their safety and facilitate the widespread
application of these novel antimicrobial agents in medicineref.
The antimicrobial action of bacteriocins from Gram-positive
bacteria is based on interaction with the cytoplasmic membrane
of sensitive bacteriaref
:
nisin : a cationic, polycyclic
bacteriocin of 34 residues, including several unusual dehydro
residues and thioether-bridged lanthionines. Models based on
studies with artificial membrane systems propose that it forms
wedge-like poration complexes in the membrane by electrostatic
interaction between the positively charged C terminus of the
peptide and anionic membrane phospholipids. Nisin can also
permeabilise membranes via a targeted mechanism by using lipid
II, the bactoprenol-bound precursor of the bacterial cell
wall, as a docking molecule. Another consequence of binding
with lipid II is the inhibition of peptidoglycan biosynthesis.
mersacidine and actagardine also form a
complex with lipid II, but binding only blocks the
incorporation of lipid II into peptidoglycan, resulting in
slow cell lysis rather than pore formation.
plantaricin C and pediocin PD-1 share a
conserved sequence motif, which is most probably involved in
the binding of these bacteriocins to lipid II, with
mersacidine and actagardine. Although pediocin PD-1 and
plantaricin C may inhibit peptidoglycan biosynthesis, pore
formation is rather due to electrostatic interaction between
the positively charged unbridged N-terminus and
anionic phospholipids in the cytoplasmic membrane of sensitive
cells.
epilancin 15X, a novel
lantibiotic from a clinical strain of Staphylococcus
epidermidisref
But scientists have stumbled in trying to get bacteria to spit out
chemicals belonging to a class known as polyketides.
Polyketides are molecules that contain large rings of carbon and
oxygen atoms, and include the well-known antibiotic erythromycin.
DNA sequences that produce bits of the proteins that make
polyketides were taken from several different bacteria, mixed and
matched nside Escherichia coli bacteria. This much has been
done before, but the hard part was getting the resulting bits of
protein to combine into a functioning polyketide-making machine.
Polyketide proteins are very large, so it is particularly difficult
to get the components to attach together. To achieve this, special
sequences were added to the ends of their genetic fragments that in
turn made the protein fragments 'sticky'. This meant the protein
bits joined up "like Lego building blocks", resulting in new
proteins conformations and new polyketidesref. The team
has yet to test whether these polyketides have antibiotic powers.
But they anticipate that at least some will be useful. A few natural
polyketides attack cancer cells, so some of the bioengineered ones
might have anti-cancer properties too.
In Africa's ongoing struggle against tuberculosis, a group of
scientists and industry representatives are now exploring a plan to
introduce copper pipes, doorknobs and work
surfaces to the country's waterways and clinics. The metal's known
antibiotic activity could provide a simple way to help fight the
deadly infection. Past research has shown that copper has strong
anti-bacterial properties against worrisome pathogens such as the
superbug MRSA. Whereas all cells need a bit of copper to grow, an
excess can overwhelm a cell's mechanism to bind to the metal,
effectively killing it from over-exertion. MRSA was unable to
survive on copper alloy surfaces for > 90 minutes (Noyce J. O.,
Michels H.& Keevil C. W. . J. Hosp. Infect., 63. 289 - 297
(2006)). Recent research from a team in South Africa shows that
copper also wipes out the bacterium responsible for tuberculosis
(TB) — one of Africa's biggest killers. Laboratory tests showed that
after 48 hours of exposure, pure copper and 5 of its alloys could
inhibit growth in TB bacteria, including strains resistant to usual
drugs, with no signs of regrowth over the study period of 15 days.
TB is an extremely resilient bug. It often grows back on, say, a
stainless-steel surface that has been cleaned with disinfectant,
after just a couple of days. So the copper results are promising.
Such results have spurred ideas to make hospital surfaces from
copper alloys, to help keep background levels of infectious bacteria
down. The simple change has resulted in a consistent and significant
reduction in surface contamination by Staphylococcus aureus,
Escherichia coli and Pseudomonas aeruginosa. Whether
it is the best way to keep bugs in check, however, is still up for
debate. It can take up to 6 hours for copper to kill bacteria, so
there remains a window of opportunity when infection can occur. And
TB is transmitted by the inhalation of airborne bugs, so
self-sanitizing surfaces might not have as big an impact as hoped
with that disease. But reducing the chance of infection in a country
such as South Africa, where an estimated 5.3 million people live
with HIV and are particularly vulnerable to opportunistic
infections, may be of great benefit. Full water treatment systems
are obviously better than copper alone, as they are designed to
address a number of potential problems with the water supply. But
it's good to have a back-up system. Rural water-treatment systems
are not always correctly monitored. Here copper would be a good
additional barrier. Representatives from the International Copper Development Association,
along with African industry participants, engineers and faculty from
the Stellenbosch University, gathered at a workshop in Johannesburg
immediately after the IFIC conference to plan how best to use copper
to fight disease in Africa. They are still at the planning stage,
but at least one trial will begin before the end of 2006 at the
Kayamandi township clinic outside Stellenbosch. Copper is not the
answer for everything, but what we are looking at here is copper's
role in making a significant contribution. In-vitroantibiotic-induced endotoxin release may
depend on antibiotic class, presence of serum, type of organism,
site of antibiotic action and Gram-stain. Endotoxin release may be
different in late or early lysis, proportional to the number of
killed pathogens. Morphology of bacteria may have an impact on
endotoxin release and phagocytosis. Antibiotic-treated animals may
show higher endotoxin levels with a higher survival rate than
untreated animals. Plasma endotoxin may increase despite decreasing
bacteremia. There may be a similar killing rate by different
antibiotics but a difference in endotoxin release. Intestinal
endotoxin does not necessarily correlate to the level of
gram-negative bacteria. However, the alteration of the gut content
by pretreatment may be associated with reduced endotoxemia and
increased survival. Antibiotic-induced endotoxin release may be
different depending on the type of infection, the location of
infection, the virulence of strains, Gram-stain, mode of application
and dosage of antibiotic. Different antibiotics may induce the
release of different forms of endotoxin which may be lethal for
sensitized animals. The combination of antibiotics with inhibitors
of endotoxin or the pro-inflammatory response may be responsible for
increased survival by decrease of endotoxin release. The clinical
significance of antibiotic-induced endotoxin release is documented
only in a few clinical disorders, e.g., meningitis, urosepsis. The
difference in endotoxin release by PBP 2-specific antibiotics, e.g.,
imipenem, and PBP 3-specific antibiotics, e.g., ceftazidime, may not
be visible in each study. Patients with increased MOF scores may
profit from treatment with antibiotics known to decrease endotoxin.
In conclusion, the clinical significance of antibiotic-induced
endotoxin release remains to be clarified. Type of pathogen and its
virulence may be more important than recently suggested.
gram-positive pathogens were just recently recognized as an
important factor for the development of the host response. In case
of FUO in ICU patients either failure of treatment, e.g., failure of
source control in IAI, or a side effect of antibiotic treatment,
e.g., endotoxin release, should be considered as a cause of the
feverref.