Вы находитесь на странице: 1из 9

Je

is
is
av
av
MM
ss
ev
ev
RR
Lec
Lec 2: General Principles
Principles of Antimicrobials by D.r Frederick C. Loyola
of Antimicrobials
ai
ai
June 23 , 2010
MM
June 23 , 2010
en
en
RR
I. Introduction (Gram staining)
oy
oy
II.
Ic
Chemotherapeutic Agents
Ic
F.
III.
F.
Factors in Antimicrobial Selection
ll
au A. Anrimicrobial Pharmacodynamics
au
P B. Kinetics of Bacterial Killing
P
vs C. Tissue Penetration
vs
Vi D. Bactrerial Resistance
Vi
ne E. Antibiotic Dosing
ne
le F. Other Considerations
le
Ar
Ar IV. Types of Antimicrobials
ňa
ňa A. Inhibitors of Cell Wall Synthesis
Ni
Ni 1. Penicillins
gg 2. Cephalosporins
an
an 3. Vancomycin
ad
ad 4. Others
DD B. Protein Synthesis Inhibitors
rr 1. Tetracyclines
he
he 2. Macrolides
ac
ac 3. Clindamycin
Te
Te 4. Clorampenicol
5. Aminoglycosides
ar
ar CHEMOTHERAPEUTIC AGENTS
be
be
GRAM STAINING Characteristics of Ideal Anti-Infective Agent
co
co
Ri
1. Selective toxicity, effective anti-infective
Ri
ie
ie
ck
ck 2. Capable of penetration in concentration that exceeds
Ni
Ni several folds the MIC of potential pathogen; high
ad
ad affinity for site of action
Gl
Gl 3. Resistant to inactivation by microbial enzymes, tissue
Je
Je fluid or products of inflammation and must not readily
stimulate microbial resistance
nz
nz
4. Orally active
Ay
Ay
hh
at 5. Long elimination half-life
at
KK 6. Devoid of adverse drug-drug interactions
oo 7. Absence of major rgan system toxic effects
Jh
Jh 8. Absence of developmental or behavioral toxic effects
ah
ah
en
en  Targets
 bacterial and fungal cell wall-synthesizing enzymes
Gi
Gi
do
(Beta lactams & Anti-fungal)
do
E
 bacterial ribosomes (Macrolides & Aminoglycosides)
E
lle
lle
ce
ce  enzymes required for nucleotide synthesis & DNA
Jo
Jo replication (Sulfas & Quinolones)
be
be  machinery of Viral replication (anti-viral drugs)
Jo
Jo
zz
Iv FACTORS IN ANTIBIOTIC SELECTION
Iv
kk
ar
ar SPECTRUM
MM BACTERIOSTATIC:
ll agents that interfere with growth or replication of the
he
he microorganism but do not kill it
ac
ac eg. chloramphenicol, the tetracyclines, erythromycin,
clindamycin, streptogramins, and linezolid
RR
””
“G
“G
nn BACTERICIDAL
hh antibiotic-mediated killing of the microorganism
Jo
Jo
nn Page 1 of 9
Ia
Ia
na
e.g., the aminoglycosides The bacteria has a natural resistance to the given
drug. Hence, it is ineffective.
A. ANTIMICROBIAL PHARMACODYNAMICS
 MIC - (minimum inhibitory concentration)
• Acquired
minimum concentration of antibiotic to Resistance is acquired when the bacteria has an
inhibit the growth of bacteria adapted exposure to the drug or to its environment
MIC50 (50%) or MIC90 (90%)
 PAE – (post antibiotic effect)
antibacterial activity persists beyond the
time during which measurable drug is present
 PALE – (post antibiotic leukocyte enhancing effect)

B. KINETICS OF BACTERIAL KILLING


 Concentration-dependent antibiotics
The higher the concentration, the greater the
bactericidal effect (e.g., Aminoglycosides and quinolones)
Prolonged PAE, significant PALE even at subminimal
inhibitory concentration ( phagocytosis is still there evn if level • Relative
of antibiotic has decreased beyond MIC With resistance but with increasing concentration the
Less frequent dosing is more effective drug it can effectively kill the organism
 Time-dependent antibiotics • Absolute
no much difference in the rate of killing with varying Absolute resistance with drug no matter what the
concentrations of the drugs. concentration
(e.g., Beta-lactams, Metronidazole, Vancomycin,
Macrolides,clindamycin, oxazolidinones) *Natural resistance can be relative and Acquired resistance
Bactericidal efficacy is dependent on the length of can be relative or absolute.
time the bacteria are exposed to free drug serum
concentrations of at least 4x the pathogen MIC E. ANTIBIOTIC DOSING
Goal: attain serum concentration of at least 4x MIC of  Renal Insufficiency
infecting organism at all times for at least 40-50% of the dosing  Loading and Maintenance dosing in renal
interval insufficiency
Most cost-effective manner of administration: by  Aminoglycoside dosing
continuous IV infusion or choose an antibiotic with long half  For drugs eliminated by the kidneys, the initial/loading dose
life. (if required) is unchanged, and the maintenance dose/dosing
interval are modified in proportion to the degree of renal
C. TISSUE PENETRATION insufficiency. If creatinine clearance is 40-60 ml/min, decrease
 Property of antibiotic the dose of renally eliminated antibiotic by 50% and maintain
the usual dosing interval.
e.g., lipid solubility, molecular size
 If crea clearance is 10-40 ml/min, decrease dose of renally
quaternary ions (with charge)- can’t enter the tissues
eliminated antibiotic by 50% and double the dosing interval.
 Tissue  Or alternatively, use antibiotic eliminated/ inactivated by the
e.g., adequacy of blood supply, presence of hepatic route in usual dose.
inflammation
 Hepatic Insufficiency
 Rarely a problem in acute infection due to increased  Decrease daily dose of hepatically eliminated antibiotic by
microvascular permeability from local release of chemical 50% in the presence of clinically severe liver disease.
inflammatory mediators.  Alternatively use antibiotic eliminated/inactivated by the
 Antibiotic tissue penetration is rarely a problem in acute renal route in usual dose.
infections due to increased microvascular permeability from
local release of chemical inflammatory substances or  Combined Renal & Hepatic Insufficiency
mediators.  There are no good dosing adjustment guidelines for
 Chronic infections and infections caused by intracellular patients with hepatorenal insufficiency. If renal insufficiency is
pathogens often rely on chemical properties of an antibiotic worse than hepatic insufficiency, antibiotic eliminated by the
(e.g., high lipid solubility, small molecular size) for adequate liver are often administered at half the daily dose. If hepatic
tissue penetration. insufficiency is more severe than the renal insufficiency, renally
eliminated antibiotics are usually administered and dosed in
D. BACTERIAL RESISTANCE proportion to renal function.
• Natural

Page 2 of 9
 Mode of Antibiotic Excretion/Excretory organ toxicity
 The mode of elimination/excretion does not necessarily
predispose to excretory organ toxicity. (e.g., nafcillin
(hepatically eliminated) is not hepatotoxic.

OTHER CONSIDERATIONS IN ANTIMICROBIAL THERAPY


 Bactericidal VS Bacteriostatic Therapy
 For most infections, bacteriostatic and bacreticidal
antibiotics inhibit/kill organisms at the same rate and should
not be a factor in antibiotic selection. However, bactericidal
antibiotics has an advantage in certain infections such as
endocarditis, meningitis, febrile neutropenia.

 Monotherapy VS Combination Therapy


 Monotherapy is preferred to combination therapy for most
infections:
Cost saving, less chances of medication error, fewer
missed doses/drug interactions The outer membrane or outer envelope is present in gram
 Combination is useful for drug synergy, extending spectrum negative but not gram positive organisms.
beyond what can be obtained with a single drug and  Relatively resistant to beta lactam antibiotics.
preventing drug resisance  Impaired penetration of the antibiotics to target PBPs
because of their impermeable outer cell wall membrane.
 Intravenous VS Oral switch therapy  Beta lactam can only enter via outer membrane protein
 Advantage of an early oral switch are: reduced cost, early channels (porins). Absence of the proper channel or down
hospital discharge, less need for home IV therapy, elimination regulation of its production can greatly impair drug entry into
of IV line infection the cell.

 Duration of therapy
 Most bacterial infections in normal hosts are treated with
antibiotics for 1-2 weeks. Maybe extended in: impaired
immunity (diabetes, SLE, Alcoholic liver disease, neutropenic,
diminished splenic function), Chronic bacterial infections
(endocarditis & osteomyelitis), chronic viral & fungal infections
and certain bacterial intracellular pathogens

ANTIMICROBIALS

A. Beta-Lactam Compounds & Other Inhibitors of Cell Wall


Synthesis
 Penicillins
 Cephalosporins & Cephamycins
 Other Beta Lactam Drugs:
 Monobactams
 Beta-Lactamase Inhibitors
 Carbapenems
 Other Inhibitors of Cell Wall Synthesis
 Vancomycin
 Teicoplanin
 Daptomycin
BETA LACTAMS:
 Fosfomycin
1. PENICILLINS
 Bacitracin
6-Aminopenicillanic Acid
 Cycloserine

Cell Wall Lysis:

Page 3 of 9
beta-lactamase producing staphylococci, T. pallidum,
Clostridium
 Pen V: oral form, indicated only for minor infections due to
poor bioavailabilty
 Benzathine/Procaine: IM yield low but prolonged drug
levels, for treatment of beta-hemolytic strep pharyngitis

CLINICAL USES OF PENICILLIN, RESISTANT TO STAPH


BETA-LACTAMASE
 thiozolidine ring is attached to the beta lactam ring that  The sole indication is infection by beta-lactamase –
carried the secondary amino group producing staphylococci (Methicillin, Nafcillin)
 For mild, local infections: Isoxazolyl (oxacillin) – 15-25
CLASSIFICATION OF PENICILLINS mg/kg/d
 Penicillins (e.g. Penicillin G)  For serious systemic staph infection: oxacillin or nafcillin 8-
VS gram-positive organism, gram-negative cocci & 12 g/d (50-100 mg/kg/d)
non-beta lactamase anaerobes
have little activity against gram negative rods and Clinical Uses of Extended-Spectrum Penicillins
they are susceptible to hydrolysis by beta-lactamases  Aminopenicillins
 Carboxypenicillins, Ureidopenicillins- good for
 Antistaphylococcal penicillins (e.g. Nafcillin) pseudomonas
VS staphylococci and streptococci  Greater activity against gram-negative bacteria
 Amoxicillin is better absorbed from the gut
 Extended-spectrum penicillins (e.g. ampicillin &  Ampicillin is effective for shigellosis but not for
antipseudomonal penicillins) salmonellosis
VS gram-negative organisms  For Pseudomonas: Carboxypenicillins + aminoglycosides
 Beta-lactamase inhibitors ( clavulanic acid, sulbactam,
LIMITATIONS
tazobactam): VS beta-lactamase-producing strains of Staph
 Instability at acidic pH aureus & other g- bacteria
 Susceptibility to destruction by beta-lactamase Minimal antibacterial activity but good bata-lactam
(penicillinase) inhibitors
 Relative inactivity against gram-negative bacilli SUICIDE INHIBITORS:
UNITS ( clavulanic acid, sulbactam, tazobactam)
 Pen G: 1600 units/mg (1M unit = 0.6 g) Preaparations:
MECHANISM OF ACTION 1. Amoxicliin + clavulanic acid (co-amoxiclav) in oral form
 Inhibit bacterial cell wall synthesis 2. Ampicillin + Sulbactam (Unasyn)
3. Piperacillin + Tazobactam (Tazosyn)
PHARMACOKINETICS OF PENICILLIN
 Oral absorption differs greatly for different penicillins, B-Lactamase
depending on acid stability and protein-binding (impaired
by food)
 Widely distributed in the body fluids and tissues
 Excreted into sputum and milk
 Poor penetration in the eye, prostrate and CNS except
with active inflammation of meninges if given in high
doses
 Rapidly excreted by the kidneys
 Normal half-life: approx. 30 mins
 Blood levels raised by giving probenecid
RESISTANCE
 Inactivation by B lactamase
 Modification of target PBPs
 Impaired penetration of drug to target PBPs
 Presence of an efflux pump
-Does not attain therapeutic concentration inside

CLINICAL USES OF PENICILLINS


 Pen G: drug of choice for infections caused by
streptococci, meningococci, enterococci, pneumococci, non-

Page 4 of 9
2. CEPHALOSPORINS  Ceftazidime & cefoperazone: VS P. aeruginosa
 Ceftizoxime & Moxalactam: VS B fragilis
 Cefixime & Ceftriaxone: first line drug for treatment of N.
gonorrhea
 Ceftriaxone & Cefotaxime: active against Meningitis
also with severe infections due to (PRSP*)

Fourth – Generation Cephalosporins


 Cefepime, Cefpirome
 More resistant to hydrolysis by beta-lactamase
 Active VS P aeruginosa, enterobacter, S aureus, and S
 7-aminocephalosporanic acid (betalactam ring fused with 6- pneumoniae (PRSP)
membered dihydrotiazine ring)  Half-life: 2 hours
 Adverse effects:
 Soluble in water Allergy
 Stable to pH and temperature changes Toxicity – Renal, Hypoprothrombinemia, severe
 Not active against enterococci and Listeria disulfiram-like reactions
monocytogenes Superinfection

First-Generation Cephalosporin None of the Cephalosporins is active against Enterococcus,


Listeria monocytogenes and MRSA
 Cefadroxil, cefazolin, cephalexin, cephalotin, cephapirin,
cephradine
ADVERSE EFFECTS & TOXICITIES
 very active against g+ cocci such as pneumococci,
streptocci and staph. They are not active against MRSA.  Sensitizing, may cause cross reactions in some with pen
allergy
 Tubular secretory blocking agents (probenecid) may
increase serum levels  Patients with anaphylactic reactions to pen should not
receive cephalosporins
 Cefazolin is almost the only first –generation parenteral
cephalosporin: drug of choice for surgical prophylaxis,  Cephalosporins with methylthiotetrazole group
alternative to an antistaphylococcal penicillin ( cefamandole, cefmetazole, cefotetan, cefoperazone)
 Rarely the drug of choice for any infection causes hypoprothrombinemia and bleeding---give Vit K.
also causes disulfiram-like effect

As we go up to the ladder, the gram- coverage becomes broader.


OTHER BETA-LACTAMS
But the coverage on g+ are sometimes compromised. But in 2nd
gen, generally speaking, the coverage against by 1st gen is  Monobactams (monocyclic B-lactam ring)
retained and with the addition of some other g- organisms. Aztreonam : VS gram-negative rods
 Beta-lactamase Inhibitors (suicidal inhibitors)
Second Generation Cephalosporins Clavulanic acid, sulbactam, tazobactam
 Cefaclor, cefamandole, cefonicid, cefuroxime(axetil), Treatment of mixed aerobic and anaerobic infections
cefprozil, loracarbef, ceforanide, cephamycins (cefoxitin), (intraabdominal infections)
cefmetazole, cefotetan)  Carbapenems
 Extended gram-negative coverage beta-lactam ring is attached to a five-membered ring,
 Dose: (oral) 10-15 mg/kg/d substitution of carbon for sulfur and unsaturation in the five
membered ring
 VS beta-lactamase producing H. influenza or Moraxella
Imipenem, meropenem, doripenem,ertapenem
catarrhalis
Choice for treatment of infection caused by
 Tx of sinusitis, otitis or lower RTI
enterobacter
 Cefoxitin, Cefotetan, Cefmetazole- peritonitis or
diverticulitis
3. VANCOMYCIN
 Cefuroxime: for community-acquired pneumonia
 For staphylococcal infection (MRSA)
-The only second-generation drug that crosses blood-
 Bactericidal
brain barrier.
 Primarily active against gram positive bacteria
 MOA: Inhibits cell wall synthesis by binding firmly to the D-
Third–Generation Cephalosporins
ala-D-Ala terminus of nascent peptidoglycan
 Cefoperazone, cefotaxime, ceftazidime, ceftriaxone,
 Poorly absorbed in GIT
cefixime, cefpodoxime proxetil, cefdinir, cefditoren pivoxil,
ceftibuten, ceftizoxime, moxalactam  Tx of enterocolitis due to Clostridium difficile
 Expanded gram-negative coverage  Indication: sepsis, endocarditis due to methicillin-resistant
 Some can cross blood brain barrier staphylococci, with gentamicin, as alternative regimen for
 Active VS citrobacter, serratia & providencia(*)

Page 5 of 9
treatment of enteroccocal endocarditis in patient with serious 1. The charged tRNA unit carrying amino acid 8 binds to the
penicillin allergy acceptor siteA on the 70s ribosome. The peptidyl tRNA at the
 Drug of choice for treatment of serious staphylococcal donor site, with amino acid 1 through 7,
infections resistant to usual antistaphylococcal antibiotics 2. Then binds the growing amino acid chain to amino acid 8
 Adverse Reaction: “Red man” or “red neck” syndrome (transpeptidation,).
3. The uncharged tRNA left at the donor site is released
3. OTHER INHIBITORS OF CELL WALL SYNTHESIS 4. The new 8-amino acid chain with its tRNA shifts to the
 Teicoplanin – glycopeptide peptidyl site (translocation). The antibiotic binding sites are
Long half-life: 45 – 70 hours shown schematically as triangles.
 Chloramphenicol (C) and Macrolides (M) bind to the 50s
 Daptomycin (mechanism unknown) subunit and block transpeptidation by inhibiting peptidyl
o Cyclic lipopeptide, same spectrum with vanco transferase(step 2) and translocation reaction and formation of
o More rapidly bactericidal in vitro initiation complex.
 The tetracyclines (T) bind to the 30s subunit and prevent
o Dose: 4 mg/kg for skin infection, 6 mg/kg for
binding of the incoming charged tRNA unit at the acceptor site
bacteremia and endocarditis
on the mRNA-ribosome complex(Step 1).
 Fosfomycin – stable salt of phosphonomycin
 Aminoglycosides bind to specific 30s subunit ribosomal
o inhibits enolpyruvate transferase
proteins.
o tx of uncomplicated LUTI  Protein synthesis is inhibited by aminoglycosides in at least
o Oral bioavailability: 40% 3 ways:
o Half – life: 4 hours 1. Interference w/ initiation complex of peptide
o Dose: single 3g for AUC formation,
o Safe for use in pregnancy 2. Misreading of mRNA which causes incorporation of
 Bacitracin - cyclic peptide incorrect amino acid into the peptide,
o interfere with dephosphorylation in cycling of the lipid 3. Breaking of polysomes into nonfunctional
carrier monosomes.
o no cross-resistance
o Useful for suppression of mixed bacterial flora in 1. TETRACYCLINES
surface lesions  Binds to 30s ribosomal subunit blocking attachment of
 Cycloserine aminoacyl tRNA to acceptor site
o Water-soluble, unstable at acid pH  Static
o Almost exclusively to treat tuberculosis resistant to  Resistance: decrease intracellular accumulation
first-line agents (impaired influx or increased efflux), ribosomal
o Inhibits alanine racemase (L-alanine to D-alanine) & protection to cause interference with binding, enzymatic
ligase enzyme inactivation
 Broad-spectrum Bacteriostatic
B. PROTEIN SYNTHESIS INHIBITORS  Chelate divalent metal ions/damage growing bones & teeth
 40-80% protein bound
Tetracyclines, Macrolides, Clindamycins, Chloramphenicol,  Cross the placenta and excreted in milk
Aminoglycosides & Streptogramins  Choice for patient with renal insufficiency: Doxycycline –
once daily/ Tigecycline as well not renally excreted
 Minocycline-maybe used for meningococcal carrier state
 Drug of choice for: Mycoplasma pneumoniae, chlamydiae,
rickettsiae, spirochetes, helicobacter pylori(+)
 Avoid in pregnant women and children below 8 years old,
to avoid deposition in growing bones and teeth
 Tigecycline
o First glycylcycline to reach the clinic,
o Newest among the group
o Effective even with tetracycline resistant strains
o Broad spectrum: even with MRSA, VISA,VRE, g-
enterics, anaerobes(B+/-), atypical agents
o Proteus and P aeroginosa- intrinsically resistant
o No dosage adjustment required for renally impaired
patients, elimination is primarily biliary
STEPS IN BACTERIAL PROTEIN SYNTHESIS AND
TARGETS OF SEVERAL ANTIBIOTICS ADVERSE REACTIONS
o GI effects

Page 6 of 9
o Bony structures and teeth- Chelates, binds enamel
dysplasia, bone deformity 2.C. MACROLIDES: Azithromycin
o Liver toxicity- with concomitant liver problem or with  15-atom lactone ring
pregnancy / can impair hepatic functions  Highly active against chlamydia
o Kidney toxicity- nitrogen retention, diuretics also  High tissue & phagocytic cells level – renal elimination
causes nitrogen retention, may give  Half-life is > 4 days
doxycycline or tigecycline  Once-daily dosing and short duration of treatment
o Local Tissue toxicity- Venous thrombosis to iv site  Given 1 hour before or after meals
o Photosensitization  Does not inactivate cytochrome P450 enzymes
o Vestibular reactions- Dizziness, vertigo, nausea with
high doses 2.D. MACROLIDES: Ketolides – (Telithromycin)
 Semisynthetic 14-membered-ring
2. MACROLIDES AND CLINDAMYCIN
 Oral bioavailability: 57%
 Bind to 50s subunit inhibiting translocation of peptidyl-
 Dose: 800 mg OD
tRNA from acceptor to donor site
 Treatment of respiratory tract infections
 Static / cidal in high doses
 Reversible inhibitor of the CYP3A4
 Resistance: reduced permeability or efflux pump; form
 May slightly prolong the QTc interval
methyltransferases that alters 50s drug binding (gram + cocci)
(MLS-type B resistance) & drug-inactivating enzymes (gram – Many macrolide resistant strains are susceptible to ketolides
because the structural modification of these compounds renders
rods)-esterases
them poor substrates for efflux pump mediated resistance.and
they bind with ribosomes of other bacteria with higher affinity
than other macrolides

3. CLINDAMYCIN
 Chlorine –substituted lincomycin
 VS strep, staph, pneumococci
 For severe anaerobic infection due to bacteroides & other
anaerobes
 Can be used for penetrating wounds of the abdomen (+
amino or cephalosporin)
 For prophylaxis of endocarditis with valvular heart disease
undergoing dental procedures rather than erythromycin
 The macrolides are a group of closely related compounds  10-20 mg/kg/d (0.15-0.3g every 6 hrs)
characterized by a macrocyclic lactone ring (usually containing  Causes drug induced colitis (C. difficile)
14 or 16 atoms) to which deoxy sugars are attached. The
prototype drug is erythromycin and the semi synthetic
compounds are clarithromycin and azithromycin. 4. CHLORAMPENICOL
Clindamycin is a chlorine-substituted derivative of lincomycin  Bind to 50s ribosomal subunit, inhibits peptidyltransferase
 Static
2.A. MACROLIDES: Erythromycin  Resistance: formation of acetlytransferases (transacetylase)

 Poorly soluble in water


 Unstable at acid pH
 VS gram –positive organisms
 Destroyed by stomach acid (needs enteric coating)
 Drug of choice: corynebacterial infection, respiratory,
neonatal, ocular or genital chlamydial infection, community-
acquired pneumonia, alternative for penicillin-allergic
individuals
 Prophylaxis against endocarditis during dental procedures
with valvular heart disease
 Soluble in alcohol, neutral, stable
2.B. MACROLIDES: Clarithromycin  Bacteriostatic, broad-spectrum
 More active against mycobacterium avium complex  Dose: 50-100 mg/kg/d
 250-500 mg BID  Completely and rapidly absorbed orally
 Longer half-life: 6 hours  Eye infections
 Lower frequency of GI intolerance
MECHANISM OF ACTION:
 Less frequent dosing

Page 7 of 9
Reversible binds to 50S ribosomes subunits of 5.E. Netilmicin
susceptible organisms and inhibits the peptidyl tranferase step  5-7 mg/kg/d
(transpeptidation) preventing aminoacids from being  Maybe active against genta and tobra resistant bacteria
transferred to growing peptide chains thus inhibiting protein
synthesis 5.F. Kanamycin & Neomycin
 Bowel preparation for elective surgery
ADVERSE REACTIONS  For topical and oral use only (too toxic for parenteral use)
 GI disturbances
 Bone marrow disturbances 5.G. Spectinomycin
Aplastic anemia  Aminocyclitol
 Toxicity with newborn infants  Alternative treatment for gonorrhea for penicillin-allergic
Gray baby syndrome: lack of effective glucoronic patients
acid conjugation  2 g IM (40 mg/kg)
 Interaction with other drugs
- Inhibits hepatic microsomal enzymes AMINOGLYCOSIDES
BIODISPOSITION AND TOXICITY
5. AMINOGLYCOSIDES  Polar compound, not absorbed orally or widely distributed
 Bind to 30s ribosomal subunit-  Renal clearance proportional to GFR--- decrease dose in
dec. initiation codon function- renal dysfunction
formation of initiation complex (static)  Short half-lives (2-3 hours) – conventionally given at 6-12
 Misreading of code- formation of inactive proteins (cidal) hours intervals, but once-daily dosing regimens now more
 Resistance: formation of drug-inactivating transferases common
 Adverse effects- nephrotoxicity: includes proteinuria,
 Bactericidal, accumulated intracellularly via O2 dependent hypokalemia, acidosis and ATN. Ototoxicity:from hair cell
uptake (anaerobes innately resistant) damage includes deafness and vestibular dysfunction, NM
 Water-soluble, stable, more active at alkali blockade, contact dermatitis (neomycin)
 Irreversible inhibitors of protein synthesis – binds to specific
30S subunit ribosomal proteins ONCE DAILY DOSING OF AMINOGLYCOSIDES
 Absorbed poorly in GI tract  Once-daily administration is clinically effective and causes
 Ototoxic and nephrotoxic less toxicity than conventional dosing regimens
 In high doses: curare-like effect with neuromuscular  Antibacterial effects depend mainly on peak blood level
blockade (rather than time) and continue with blood levels < MIC. A post
 VS gram-negative (often in combination), enteric bacteria, antibiotic effect (PAE)
synergistic with penicillins in enterococcus and P.  Toxicity depends both on blood level & the time such levels
aeroginosa are > than a specific threshold (ie, total dose)

5.A. Streptomycin
 Second line agent for tuberculosis tx NEWER AGENTS
 With penicillin: for enterococcal endocarditis
 With tetracycline: tularemia, plague, brucellosis 1. Streptogramins
 Most serious toxic effect: vestibular disturbance – vertigo  Quinupristin-dalfopristin
and loss of balance  Bactericidal (rapid) except with E faecium (slow)
 Active against g+cocci, MDR-strep, PRSP,MRSA
5.B. Gentamicin  Resistance : MLS-B type/inactivation of A and efflux
 Gram-positive and gram-negative  IV 7.5 mg/kg every 8-12 hrs
 For severe infections (sepsis and pneumonia)  Adverse effect: pain at infusion site, athralgia-myalgia
 5-6 mg/kg/d syndrome

5.C. Tobramycin 2. Oxazoladinones


 More active against pseudomonas++  Linezolid: bacteriostatic except for strep
 Also available in inhalational form (TOBI)  Inhibits protein synthesis by binding on 23 S
 (P. aeroginosa complicating cystic fibrosis) ribosomal RNA of the 50S subunit (unique-no cross resistance)
 High oral bioavailability:100%
5.D. Amikacin  Half-life: 4-6 hours
 For resistant organisms  For vancomycin-resistant E. faecium
 500 mg BID IV  Toxicity: thrombocytopenia and neutropenia

Page 8 of 9
MECHANISMS OF ACTIONS

Page 9 of 9

Вам также может понравиться