Document

advertisement
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
Contents:No.
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
Subject
Contents
Concept of antibiotics and their classification
Antibiotics
Classification of antibiotics
Classes of antibiotics
1. Inhibitors of cell wall synthsis
Classification of inhibitors of cell walls synthesis
B-lactam antibiotics
Inhibitors of proteins synthesis:Aminoglycosides (AGs)
Tetracyclines
Chloramphenicol:
Macrolides:
Lincosamides :
Plasma membrane inhibitors
Nucleic acid inhibitors
Rifamycins
Quinolones
Ciprofloxacin, Norfloxacin, Ofloxacin,
Moxifloxacin, and trovafloxacin.
Metronidazole
Antimetabolites
Sulfonamides
Trimethoprim
Antifungal drugs
Antiviral drugs
Antiprotozoan drugs
Antihelminthic drugs
Pharmacology
Page No.
1
2
2
4
7
12
13
15
20
20
23
26
29
31
33
34
35
37
37
39
40
40
40
42
43
44
44
45
4
th
year
)1(
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)2(
Concept of antibiotics and their classification
Antibiotics:means "against life" defines are soluble compounds that are derived from certain M.O
that can inhibit the growth of bacteria or even destroy (kill) them and other M.O, such as
bacteria, fungi or protozoa, can differ from one another in the following ways:
a) Chemical properties.
b) Mechanism of action.
c) Pharmacokinetics.
d) Spectrum of activity.
e) Therapeutic uses.
f) Untoward effects.
The term originally referred to any agent with biological activity against living
organisms, however, "antibiotic" now is used to refer to substances with antibacterial,
antifungal, or anti-parasitical activity.
The first widely used antibiotic compounds used in modern medicine were produced
and isolated from living organisms, such as the penicillin class produced by fungi in the
genus penicillium or streptomycin from bacteria of the genus streptomyces. With
advances in organic chemistry many antibiotics are now also obtained by chemical
synthesis, such as the Sulfa drug.
Many antibiotics are relatively small molecules with a molecular weight less than
2000 Da.
Antibiotics are specific products of metabolism, or their modifications with high
physiological activity against individual groups of M.O (Viruses, Bacteria,
Streptomyces, Fungi, Algea, Protozoa) or against malignant tumours, that can
selectively slow down or completely inhibit their growth.
In contrast to some other metabolites, antibiotics are characterized by the following
two properties:
I: Unlike organic acids alcohols or the like compounds antibiotics are biologically active
against organisms sensitive to them. This means that an antibiotic substance has a high
physiological effect even in low concentrations. Penicillin for example, in the conc. Of
0.000001 gm/ml produces a pronounced bactericidal effect on bacteria sensitive to this
antibiotic.
II: The antibiotic action is selective, which means that each antibiotic is potent
biologically only against certain organisms, or groups of organisms, without affecting
appreciably other living organisms. Penicillin G for example inhibits the growth of only
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)3(
certain gram +ve bacteria (cocci, streptococci, etc.) and has no effect on G-ve bacteria,
fungi, or other organisms.
The antibiotics are not intermediate but final products of metabolism. They are
accumulated inside the cell or released by the cell in to the medium.
Antibiotics are formed in quantities smaller than organic acids alcohols, etc., but they
are physiologically most active products of metabolism.
Antibiotics were further developed in Britain following the re-discovery of penicillin in
1928 by Alexander Fleming. Fleming was studying Staphylocci at St. Mary's College in
London when he noticed a zone of inhibition surrounding a penicillium contaminant in
one of his cultures. The penicillium was initially identified as P. rubrum but was later
determined to be P. natatum
Definitions:
Antimicrobial, antimicrobic: any substances with sufficient antimicrobial activity that it
can be used in the treatment of infectious diseases.
Anti-infective agents: are chemical substances that kill or suppress the growth of M.O.
Bactericidal antibiotics: an antimicrobial that not only inhibits growth but is lethal to
bacteria.
Bacteriostatic antibiotic: an antimicrobial that inhibits growth but does not kill the
organisms.
Chemotherapeutics: a broad term that encompasses antibiotics, antimicrobials, and drugs
used in the treatment of cancer. In the context of infectious diseases, it implies the agent
is not an antibiotic.
Minimum inhibitory conc. (MIC): a laboratory term that defines the lowest conc.
(µg/ml) able to inhibit growth of the M.O.
Resistant: organism that are not inhibited by clinically achievable concentration of an
antimicrobial agent.
Sensitive: term applied to M.O indicating that they will be inhibited by conc of the
antimicrobic that can be achieved clinically.
Spectrum: an expression of the categories of M.O against which an antimicrobial is
typically active. A narrow spectrum agent has activity against only a few organisms. A
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)4(
broad spectrum agent has activity against organisms of diverse types (ex: G +ve and –ve
bacteria).
Susceptible: term applied to M.O indicating that they will be inhibited by conc of the
antimicrobic that can be achieved clinically.
Selective toxicity:
Clinically effective antimicrobial agent all exhibit selective toxicity toward the
parasite rather than the host (more damage to parasite than host), a characteristic that
differentiates them from the disinfectants in most cases, selective toxicity is explained
by action on microbial processes or structures that differ from those of mammalian cells,
for example, some agents act on bacterial cell wall synthesis, and other on functions of
the 70 S bacterial ribosome but not the 80 S eukaryotic ribosome, some antimicrobial
agents, such as penicillin, are essentially non toxic to the host, unless hypersensitivity
has developed.
Selective toxicity: is based on the ability of an antimicrobial agent to attack a target
present in bacteria but not humans.
Type of antibiotics:
1. Natural antibiotics : A number of natural products this substances secretions of fungi
or soil M.O. Soils are complex ecosystems, and it is not surprising that it is inhabitants
have evolved chemical defenses against each other. For example: Penicillin.
2. Semi-synthetic antibiotics: there are natural antibiotics (products) that have been
chemically modified in the laboratory (and pharmaceutical facility) to
a. Improve the efficacy of the natural product.
b. Reduce it is side effects.
c. Circumvent developing resistance by the targeted bacteria.
d. Expand the range of bacteria that can be treated with it.
3. Synthetic antibiotics: completely synthetic products, the sulfa drugs are examples.
Classification of antibiotics:
I: Classification of antibiotics by the mechanism of their biological action:
1. Antibiotics inhibiting synthesis of the cell wall
(ex: Penicillins, Bacitracin, Vancomycin, Cephalosporins, cycloserin, …etc)
2. Antibiotics causing membrane dysfunction
(ex: nystatin, trichomycin, endomycin, gramicidins,…etc)
3. Antibiotics inhibiting selectively the synthesis (metabolism) of nucleic acids
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)5(
a. Inhibiting synthesis of RNA
(ex: griseofulvin, canamycin, olivomycin, novobiocin, neomycin, actinomycin)
b. Inhibiting synthesis of DNA
(ex: ciprofloxacin, norfloxacin, sarcomycin, actidion, brunemycin)
4. Antibiotics inhibiting the synthesis of purins and pyrimidins
(ex: Sarcomycin, decoinin, asaserin)
5. Antibiotics inhibiting the synthesis of protein
(ex: gentamicin, tetracycline, tobramycin, erythromycin, chloramphenicol,…..etc)
6. Antibiotics inhibiting respiration
(ex: patulin, pyocynine, usnic acid,….etc)
7. Antibiotics inhibiting oxidative phosphorylation
(ex: colicins, oligomycin, gramicidins)
8. Antibiotics having antimetabolic properties these antibiotics acts as antimetabolites of
amino acids, vitamins, and nucleic acids.
(ex: trimethoprime, sulfamides, cotrimexazole)
II. Classification of antibiotics by the spectrum of their biological action:
1. Antibacterial antibiotics of narrow spectrum, active mostly against G+ve or G-ve
organisms. Ex: against G-ve bacteria: polymyxin , against G+ve bacteria: Penicillin
2. Broad spectrum antibacterial antibiotics, active mostly against G+ve and G –ve
bacteria ex: tetracycline, Aminoglcosides, chloramphenicol.
3. Antituberculosis antibiotics, active mostly against M. tuberculosis
Ex: Rifampin, streptomycin, cycloserin, canamycin
4. Antifungal antibiotics
(ex: Nystatin, griseofulvin, candicin, trichothecin, levorin)
5. Antitumour antibiotics
(ex: actinomycin C, mitomycin C, olivomycin, bruneomycin, rubomycins)
6. Antiamoebic antibiotics (ex: metronidazole, fumagillin)
7. Bacteriostatic antibiotics (ex: chloramphenicol, sulfonamides)
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)6(
8. Bactericidal antibiotics (ex: penicillin, Aminoglcosides, ciprofloxacin, cefotaxime)
III. Classification of antibiotics by the spectrum of their biological origin:
1. Antibiotics produced by M.O of the order Eubacterials.
a. Antbiotics formed by representatives of the genus Pseudomonas:
Pyocyanin
P. aeroginosa
Viscosin
P. viscose
b. Antbiotics formed by representatives of genera micrococcus, streptococcus,
diplococcus, chromobacterium, Escherichia, proteus
Nicin
S. lactis
Diplomycin
Diplococcus x-5
Prodigiosin
Chromobacterium prodigiosum (Serratia marcescens)
Coliformin E. coli
Protaptins
P. vulgaris
c. Antbiotics formed by genus Bacillus
Gramicidins
B. brevis
Subtilin
B. subtilis
Polymyxins
B. polymyxa
Colistatin
Bacillus unidentified sporous aerobe
2. Antibiotics formed by M.O belonging to the order streptomycetales:
Streptomycin
S. griseus
Tetracyclin
S. aureofaciencs, S. rimosus
Novobiocin
S. spheroids
Actinomycins
S. antibioticus
3. Antibiotics formed by imperfect fungi
Penicillin
P. chrysogenum
Griseofulvin
P. griseofulvum
Trichothecin
Trichothecium roseum
4. Antibiotics formed by fungi of the basidiomycete and ascomycete classes
Thermophillin
Basidiomycete Lenzites thermophila
Lenzitin
Lenzites septaria
Chetomin
Chaetomium cochloides (ascomycete)
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)7(
5. Antibiotics formed by Lichens, algae, and lower plants
Usnic (Usninic) acid
Lichen
Chlorellin
Chlorella vulgaris
6. Antibiotics formed by higher plants
Allicin
Allium sativum
Raphanin
Raphanus sativum
Phytoalexins Pisatin in peas (Pisum sativum)
Phaseolin in true beans (Phaseolus vulgaris)
7. Antibiotics of animal origin
Lysozyme, ecmolin, cruzin (Tripanosoma cruzi), Interferone
Classes of antibiotics
At the highest level, antibiotics can be classified as either bactericidal or bacteriostatic.
Bactericidal kill bacteria directly where bacteriostatic prevent them from dividing.
However, these classifications are based on laboratory behavior; in practice, both of these
are capable of ending a bacterial infection.
Generic Name
Amikacin
Gentamicin
Kanamycin
Neomycin
Netilmicin
Streptomycin
Tobramycin
Paromomycin
Antibiotics[6]
Brand Names
Common Uses
Aminoglycosides
Amikin
Infections caused by
Gram-negative
Garamycin
bacteria, such as
Kantrex
Escherichia coli and
Klebsiella
Netromycin
particularly
Pseudomonas
aeruginosa.
Nebcin
Effective against
Aerobic bacteria
(not
Humatin
obligate/facultative
anaerobes).
Possible Side Effects



Hearing loss
Vertigo
Kidney damage
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)8(
Ansamycins
Experimental, as
antitumor antibiotics
Geldanamycin
Herbimycin
Carbacephem
Loracarbef
Lorabid
Cefadroxil
Cefazolin
Cefalotin or Cefalothin
Carbapenems
Invanz
Bactericidal for both
Gram-positive and
Finibax
Gram-negative
Primaxin
organisms via
inhibition of cell
wall synthesis and
therefore useful for
empiric broadMerrem
spectrum
antibacterial
coverage. (Note
MRSA resistance to
this class.)
Cephalosporins (First generation)
Duricef
Ancef
Keflin
Cefalexin
Keflex
Ertapenem
Doripenem
Imipenem/Cilastatin
Meropenem
Cefaclor
Cefamandole
Cefoxitin
Cefprozil
Cefuroxime
Cefixime
Cefdinir
Cefditoren
Cefoperazone
Cefotaxime
Cefpodoxime
Cephalosporins (Second generation)
Ceclor
Mandole
Mefoxin
Cefzil
Ceftin, Zinnat
Cephalosporins (Third generation)
Suprax
Omnicef
Spectracef
Cefobid
Claforan














Gastrointestinal
upset and diarrhea
Nausea
Seizures
Headache
Rash and Allergic
reactions
Gastrointestinal
upset and diarrhea
Nausea (if alcohol
taken concurrently)
Allergic reactions
Gastrointestinal
upset and diarrhea
Nausea (if alcohol
taken concurrently)
Allergic reactions
Gastrointestinal
upset and diarrhea
Nausea (if alcohol
taken concurrently)
Allergic reactions
Dr. Zahra Muhsin Ali
Ceftazidime
Ceftibuten
Ceftizoxime
Ceftriaxone
Cefdinir
/ Lectures of antibiotics / Biology dep. /
4
th
year
)9(
Fortaz
Cedax
Rocephin
Cephalosporins (Fourth generation)

Cefepime

Maxipime

Gastrointestinal
upset and diarrhea
Nausea (if alcohol
taken concurrently)
Allergic reactions
Glycopeptides
Teicoplanin
Vancomycin
Vancocin
Macrolides
Azithromycin
Clarithromycin
Dirithromycin
Erythromycin
Roxithromycin
Troleandomycin
Telithromycin
Zithromax,
Sumamed,
Zitrocin
Biaxin
Ketek
Streptococcal
infections, syphilis,
respiratory
infections,
mycoplasmal
infections, Lyme
disease
Pneumonia


Nausea, vomiting,
and diarrhea
(especially at higher
doses)
Jaundice
Visual Disturbance, LIVER
TOXICITY. This
medication's approval in the
U.S. was controversial, and
one doctor went to jail in
followup attempts to
ascertain its safety because
she falsified the results of
her part of the testing
precisely because it seemed
to cause liver problems,
including liver failure, to a
greater extent than would be
expected of a common-use
antibiotic.[7]
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)10(
Antimetabolite,
Anticancer
Monobactams
Spectinomycin
Aztreonam
Penicillins
Amoxicillin
Ampicillin
Azlocillin
Carbenicillin
Cloxacillin
Dicloxacillin
Flucloxacillin
Mezlocillin
Meticillin
Nafcillin
Oxacillin
Penicillin
Piperacillin
Ticarcillin
Novamox

Wide range of
infections; penicillin
used for
streptococcal
infections, syphilis,
and Lyme disease
Polymyxin B
Enoxacin
Gatifloxacin
Levofloxacin
Lomefloxacin
Moxifloxacin
Norfloxacin
Ofloxacin
Trovafloxacin

Polypeptides
Eye, ear or bladder
infections; usually
applied directly to Kidney and nerve damage
the eye or inhaled (when given by injection)
into the lungs; rarely
given by injection
Quinolones
Bacitracin
Colistin
Ciprofloxacin

Gastrointestinal
upset and diarrhea
Allergy with serious
anaphylactic
reactions
Brain and kidney
damage (rare)
Ciproxin,
CiploxESTECINA Urinary tract
Tequin
Levaquin
Avelox
NOROXIN
Ocuflox
Trovan
infections, bacterial
prostatitis,
community-acquired
Nausea (rare), tendinosis
pneumonia,
(rare)
bacterial diarrhea,
mycoplasmal
infections,
gonorrhea
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)11(
Sulfonamides
Mafenide
Prontosil (archaic)
Sulfacetamide
Sulfamethizole
Sulfanilimide (archaic)
Sulfasalazine
Sulfisoxazole
Trimethoprim
TrimethoprimSulfamethoxazole (Cotrimoxazole) (TMPSMX)

Urinary tract
infections (except
sulfacetamide and
mafenide); mafenide
is used topically for
burns





Bactrim
Nausea, vomiting,
and diarrhea
Allergy (including
skin rashes)
Crystals in urine
Kidney failure
Decrease in white
blood cell count
Sensitivity to
sunlight
Tetracyclines

Demeclocycline
Doxycycline
Minocycline
Oxytetracycline
Vibramycin
Minocin
Terracin
Tetracycline
Sumycin
Arsphenamine
Salvarsan
Chloramphenicol
Chloromycetin
Clindamycin
Cleocin
Lincomycin
Ethambutol
Fosfomycin
Fusidic acid
Furazolidone
Fucidin
Syphilis, chlamydial
infections, Lyme
disease,
mycoplasmal
infections, acne
rickettsial infections
Others
Spirochaetal
infections (obsolete)
acne infections,
prophylaxis before
surgery
acne infections,
prophylaxis before
surgery
Antituberculosis



Gastrointestinal
upset
Sensitivity to
sunlight
Staining of teeth
(especially in
children)
Potential toxicity to
mother and fetus
during pregnancy
Dr. Zahra Muhsin Ali
Isoniazid
Linezolid
Metronidazole
Mupirocin
Nitrofurantoin
/ Lectures of antibiotics / Biology dep. /
4
th
year
Antituberculosis
Zyvox
Flagyl
Bactroban
Macrodantin,
Macrobid
Giardia
Platensimycin
Pyrazinamide
Quinupristin/Dalfopristin Syncercid
Antituberculosis
Rifampin or Rifampicin
Binds to the β
subunit of "RNA
polymerase" to
Reddish-orange sweat,
inhibit transcription
tears, and urine
of mostly "Grampositive" and
"mycobacteria"
Tinidazole
Generic Name
)12(
Brand Names
Common Uses
Possible Side Effects
1. Inhibitors of cell wall synthsis
Peptidoglycan a vital component of the bacterial cell wall (Fig. 1) is a compound
unique to bacteria and therefore provides an optimum target for selective toxicity.
Synthesis of peptidoglycan precursors starts in the cytoplasm, wall subunits are then
transported across the cytoplasmic membrane and finally inserted in to the growing
peptidoglycan molecule. Several different stages are therefore potential targets for
inhibition Fig. (2) The peptidoglycan (murein sac) component of the bacterial cell wall
gives it is shape and rigidity. This giant molecule is formed by weaving the linear
glycans N-acetylglucosamine and N-acetylmuramic acid into a basket-like structure.
Mature peptidoglycan is held together by cross-linking of short peptide side chains
hanging off the long glycan molecules. This cross linking process is the target of two of
the most important groups of antimicrobics, the B-lactams and the glycopeptides
(vancomycin and teicoplanin).
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)13(
Fig. (1) Structure of the Bacterial Cell Wall
Classification of inhibitors of cell walls synthesis
A) Inhibitor of cell wall synthesis
B-lactam antibiotics
other antibiotics (1. vancomycin,2. Bacitracin)
Dr. Zahra Muhsin Ali
Penicillin
/ Lectures of antibiotics / Biology dep. /
Cephalosporins
Carbapenems
a) Imipenem
b) Cilastatin
4
th
year
Monobactams
Aztreonam
B) B-lactamase inhibitors
1. Sulbactam
2. Tazobactam
3. Clavulanic acid
I: Classification of Penicillin
A) Natural penicillin
1. Benzylpenicillin PG
2. Phenoxymethylpenicillin PV
B) Semisynthetic penicillin
1. B-lactmase resistance
a. methicillin, b. oxacillin, c. cloxacillin, d. flucloxacillin
2. Extended spectrum
a. aminopenicillin (ampicillin, amoxicillin, tatampicillin, pivampicillin,
bacampicillin)
b. Amidiniopenicillin (pivamecillinam, mecillinam
c. carboxypenicillins (carbenicillin, ticacillin, piperacillin, mezlocillin, azlocillin)
II: Classification of Cephalosporins
1. 1st generation Cephalosporins including:
Cefadroxil, Cefzolin, Cefalexin, Cephalothin, Cephradine
2. 2nd generation Cephalosporins including:
Cefaclor, Cefamandole, Cefmetazole, Cefonicid, Cefotetan, Cefprozil, Cefuroxime
3. 3rd generation Cephalosporins including:
Cefdinir, Cefditoren, Cefoperazone, Cefpodoxime, Cefotaxime,
Ceftazidime,Ceftriaxone. Ceftizoxime
)14(
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)15(
4. 4th generation Cephalosporins including:
Cefepime, Cefpirome
* Cephalosporins have been classified as first, 2nd, 3rd, and 4th generation largely on this
basis of 1. Bacterial Susceptibility, 2. Resistance to B-lactamases
B-lactam antibiotics
Beta lactams contain a beta lactam ring and inhibit cell wall synthesis by binding to
penicillin binding proteins (PBP). Beta lactams comprise a very large family of different
groups of bactericidal compounds all containing the beta lactam ring. The different
groups with in the family are distinguished by the structure of the ring attached to the
beta-lactam ring in penicillins this is a five-membered ring in cephalosporins a sixmembered ring and by the side chains attached to these ring Fig. (3), PBP are membrane
proteins (ex: Carboxypeptidases, Transglycosylases, and Transpeptidases) capable of
binding to penicillin (hence the name PBP) and are responsible for the final stages of
cross linking of the bacterial cell wall structure, various bacteria differ in their number
and type of PBP as different B-lactams have variable affinity for different PBP bacteria
have
differing
B-lactam
susceptibility.
.
Fig. (4): The structure of B-lactam
Beta
lactam ring
Chemistry:
The structure of B-lactam consists of a thiazolidine ring (1) connected to a B-lactam ring
(2), which is attached to a side chain ®, the various penicillins differ in their side chain
structure Fig. (4) The basic structure of all B-lactams consists of a four membered Blactam ring, the type and structure of the side chain defines the class and
pharmacokinetics of the drugs.
* Mechanism of action
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)16(
Inhibition of one or more of these essential enzymes results in an accumulation of
precursor cell wall units leading to activation of the cells autolytic system and cell lysis
Fig (5).
B-lactam antimicrobics are usually highly bactericidal, but only to growing bacteria
synthesizing new cell wall. Killing involves attenuation and disruption of the developing
peptidoglycan "corset" liberation or activation of autolytic enzymes that further disrupt
weakened areas of the wall, and finally osmotic lysis from passage of water through the
cytoplasmic membrane to the hypertonic interior of the cell. As might be antipated, cell
wall-deficient organisms, such as Mycoplasma or L forms are unaffected , are not
susceptible to B-lactam antimicrobics.
The action of antimicrobics on peptidoglycan synthsis. The glucan backbone and the
amino acid side chains of peptidoglycan are shown. The transpeptidase enzyme
catalyzes the cross linking of the amino acid side chains. Penicillin and other B-lactames
bind to the transpeptidase, preventing it from carrying out its function. Vancomycin
binds directly to the amino acids, preventing the binding of transpeptidase.
The spectrum of activity and therapeutics uses of B-lactams varies widely
Different B-lactams have different clinical uses, but are not active against species that
lack a cell wall (ex: Mycoplasma) or those with very impenetrable walls such as
Mycobacteria, or intacellular pathogens such as Brucella, Legionella and Chlamydia.
1. Penicillin covers a narrow spectrum of bacteria where as the Carbapenems and some
penicillins (ex: Piperacillin/Tazobactam) have a very broad spectrum of coverage.
2. Penicillins cover most Streptococcal infections although some resistance has emerged
(ex: Penicillin resistant S. pneumoniae).
3. Meropenem, imipenem, Piperacillin, and Ceftazidime have good P. aeroginosae
coverage.
4. Cloxacillin, Cefazolin, Piperacillin/Tazobactam, and imipenem have good S. aureus
coverage.
5. The third generation Cephalosporins, Piperacillin, imipenem have good gram negative
coverage.
6. Some Enterococci are sensitive to amoxicillin, piperacillin, imipenem.
7. Aztreonam is active essentially only against aerobic gram –ve Bacilli.
8. Piperacillin use to P. aeroginosa infection and carbenicillin and ticarcillin active
against P. aeroginosa called it antipsudomonadal infections.
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)17(
9. Cloxacillin, oxacillin, methicillin is used to treatment of infection caused by
Staphylococci including skin and soft tissue infections, pneumonia, osteomylitis,
endocarditis, and septicemia
st
10. 1 generation Cephalosporins activity against Proteus mirabilis, E. coli, K.
pneumonia, Staphylococci and Streptococci
nd
11. 2
generation Cephalosporins activity against H. influnzae, some Enterobacter
aerogenes, some Neisseria spp., Klebsiella, E. coli, Proteus, Streptococc
i
rd
12. 3 generation Cephalosporins effective against Serratia marcescens, Meningitis, P.
aeroginosa
th
13. 4 generation Cephalosporins activity against Staphylococci and Streptococci and
Enterobacter, Enterobactereciae, E. coli, K. pneumonia, Proteus mirabilis, P.
aeroginosa, H. influenzae, Nesseria
14 Penicillin therapy is still the first choice for all types of
a) Gonococcal infections
b) Anthrax (B. anthrax)
c) Gas gangrene
d) Actinomycosis
e) Listeria infections
15. syphilis: PG is the most effective treatment for all stages of syphilis, since
Treponema pallidum, the M.O causing syphilis is very sensitive to PG.
Dverse effects (Side effect):
1. B-lactams are generally well tolerated and are considered relatively non toxic.
3. Allergic and hypersensitivity reactions are the most frequent adverse drug effect.
4. Uncommon side effects include drug induced fever, serum sickness, interstitial,
nephritis, hepatic toxicity, neutropenia and seizures.
5.Allergic manifestations including (a. Urticaria, angioedema, drug fever, Maculopapuar
rash, Serum sickness).
6. Adisulfira like effect including (Thrombocytopenia, neutropenia, interstitial nephritis)
Absorption, distribution, & excretion of B-lactam:
After intramuscular or intravenous administration, absorption of most penicillins is
rapid and complete. After oral administration, only 5-30% of the dose is absorbed,
depending on acid stability, binding to food, presence of buffers, etc. After absorption
penicillins are widely distributed in tissues and body fluids. Protein binding is 40-60%
for PG , ampicillin, and methecillin, 90% for nafcillin, and 95-98% for oxacillin and
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)18(
dicloxacillin. For most rapidly absorbed penicillins, a parenteral dose of 3-6 gm/24h
yields serum levels of approximately 1-6 µg/ml, in many tissues, penicillin
concentrations are similar to those in serum. Lower levels occur in joints, eye, and the
central nervous system. However in meningitis penetration is enhanced and levels of 0.2
µg/ml occur in the cerebrospinal fluid with a daily parenteral dose of 12 gm. Most of the
absorbed penicillin is rapidly excreted by the kidneys. The simultaneous administration
of probenecid or para-aminohippuric acid and procain are prolongs the duration of
penicillin in the body this state called depot preparation because probenecid and procain
are blocks the transport of penicillin in the proximal tubule (kidneys) this preparation of
penicillin provides an alternative means for maintaining high plasma levels of penicillin.
Antimicrobial activity:
All penicillins and cephalosporins have the same mode of action, B-lactams are
bactericidal, they bind to PBP and inhibit bacterial cell wall synthesis,they may also
activate endogenous autolytic enzymes resulting in bacterial cell lysis. PG and P V are
often measured in units (1million units=0.6gm), but the semisynthetic penicillins are
measured in grams. Whereas 0.002-1µg/ml of penicillin G is lethal for a majority of
susceptible G+ ve organisms, 10-100 times more is required to kill G-ve bacteria (except
neisseriae). The activity of penicillin also varies with their protein binding, which ranges
from 40% to more than 95% for different drugs.
Resistance to Beta lactams may involve one or more of the three possible mechanisms
1. Resistance by alteration in target site:
Some bacteria synthesize an additional PBP which has a much lower affinity for Blactams than normal PBP and is therefore able to continue cell wall synthesis when the
other PBP are inhibited, the gene which codes for the additional PBP is present on the
chromosome in all cells of a resistant population.
2. Resistance by alteration in access to the target site
This mechanism is found in gram –ve cells where B-lactams gain access to their PBP by
diffusion through protein channels (porins) in the outer membrane. Mutations in porin
genes result in a decrease in permeability of the outer membrane and hence resistance.
Strains resistant by this mechanism may exhibit cross resistance to unrelated antibiotics
that use the same porins.
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)19(
3. Reistance by production of B-lactamases
Bete-lactamases are enzymes that catalyze the hydrolysis of the B-lactam ring to yield
microbiologically inactive products. Genes encoding these enzymes are widespread in
the bacterial kingdom and are found on the chromosome and plasmids.
The B-lactamase of G+ve bacteria are released into the extracellular environment (Fig
5a) and resistance will only be manifest when a large population of cells is present. The
B-lactam of G-ve cells however remain within the periplasm (Fig. 5b).
Bacterial resistance production penicillinase and cephalosporins are a B-lactamase
which is produced by a number of bacteria. It can hydrolyze the B-lactam ring of
penicillin to form penicilloic acid and cephalosporinic acid a substance that has no
antibacterial activity.
Microorganisms that are capable of producing penicillinase and cephalosporins (Blactamase) including : S. aureus, Bacillus spp., Bacteroids spp., E. coli, proteus spp., P.
aeruginosa, Mycobacterium tuberculosis.
*Microorganisms can acquire the ability to produce penicillinase and cephalosporins
(B-lactamase) through
1. Plasmid acquisition via transduction in G+ve bacteria.
2. R factor acquisition during conjugation in G-ve bacteria.
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)20(
Inhibitors of proteins synthesis:1. Although protein synthesis proceeds in an essentially similar manner in prokaryotic
and eukaryotic cells its possible to exploit the differences (ex: 70 S vs. 80 S ribosome) to
achieve selective toxicity. The bacterial ribosome consists of a 70 S particle, that has 2
subunits: the 50 S (large) and the 30 S (Small): (Surprisingly: 50 S + 30 S=70 S).
There are 5important types of antibiotics that inhibit the function of bacterial ribosome
this group including:
1. Aminoglycosides (Including: Gentamicin, Amikacin, Streptomycin, Neomycin,
Tobramycin, Netilmicin, Spectinomicin, Framycetin, paramomycin, etc).
2. Chloramphenicol.
3. Tetracyclines (Including: Chlorotetracycline, Demethylchlorotetracycline,
Doxycyclin, Oxytetracycline).
4. Erythromycin.
5. Lincomycin.
1. Aminoglycosides (AGs)
Chemistry
The aminoglycosides are compounds containing characteristic amino sugars joined a
hexose nucleus in glycosidic linkage, all members of aminoglycosides group of
antibiotics have a six member aminocyclitol ring with attached amino sugars. The
individual agents differ in terms of the exact ring structure and the number and nature of
the amino sugar residues.
Mechanism of action:
Aminoglycosides acts by binding to specific proteins in the 30 S ribosomal subunit,
where they interfere with the binding of formylmethionyl-transfer RNA (fmet-tRNA) to
the ribosome (Fig.33.16), and therapy preventing the formation of initiation complexes
from which protein synthesis proceeds.
Aminoglycosides cause misreading of mRNA codons and tend to break a part
functional polysomes (Protein synthesis by multiple ribosomes tandem attached to a
single mRNA molecule) in to nonfunctional monosomes.
Pharmakinetics
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)21(
1. All of AGs are poorly absorbed after oral administration because of their polycatioic
structure.
2. All the AGs are more active in an alkaline environment.
3. Streptomycin, gentamicin, tobramycin, amikacin, kanamycin, and netilmicin are
absorbed rapidly after intramuscular or subcutaneous administration.
a. It is distributed in all extra cellular fluid.
b. It crosses the blood brain barrier only if the meninges are inflamed.
c. It is excreted by glomerular filtration.
4. Neomycin is also poorly absorbed following oral administration. It is most often
applied topically.
Preparations and administration:
1. Streptomycin
a. Intramuscular injection is the most common rate of administration.
b. To avoid the development of bacterial resistance, streptomycin therapy rarely is
extended beyond 10 days except in tuberculosis and subacute bacterial endocarditis.
2. Gentamycin
a. This drug can be administrated intramuscularly (Im.), intravenously (IV.), or as an
ointment or cream.
b. When renal function is impaired, peak and through serum conc. of gentamicin are
measured intermittently to allow optimal guidance for adjusting dosage and renal
function is monitored by means of the serum creatinine level.
3. Tobramycin, amikacin, and netilmicin can be given IV. or Im., kanamycin can be give
Im., IV., or orally.
4. Neomycin
a. This drug is available in the form of creams, ointments, and sprays, both alone and in
combination with polymyxin, bacitracin, other antibiotics, and corticosteroids.
b. It is also available for oral and parenteral administration, although it is rarely used
parenterally
Side effect of aminoglycosides
All of the aminoglycoseds are :
1. Ototoxicity and nephrotoxicity are the most serious side effects.
2. Dysfunction of the optic nerve can occur with streptomycin producing scotomas.
3. Neuromuscular junction blockade may when an AGs is given at high doses and in
combination with curariform drugs.
4. Hypersensitivity reactions can occur.
5. Super infection and intestinal malabsorption can occur following oral administration
of neomycin.
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)22(
6. Directly toxic to both components of the 8th cranial nerve leading to deafness, vertigo
and ataxia which may be irreversible. Avoid in patients with hearing loss.
7. Toxicity increases with age, pre-existing renal failure, volume depletion, duration of
therapy, high dose, and use in combination with certain nephrotoxic drugs.
8. AGs should generally not be used in pregnancy because of a risk of congenital
deafness, but can be given to breast feeding women because of minimal excretion in
breast milk or oral absorption by the infant in breast milk or oral absorption by infant.
Spectrum of activity
1. Gentamicin: against (proteus, pseudomonas, serratia, some strains S. aureus and etc.).
Gentamicin and tobramicin are major AGs they have an extended spectrum, which
includes staphylococci, enterobacteriacea, and of particular importance P. aeroginosa.
2. Amikacin and streptomycin are now primarily used in combination with other
antibiotics in the therapy of tuberculosis and other mycobacterium diseases.
Streptomycin is effective against the organisms that cause Yersinia pestis and tularemia
(Francisella tularensis).
Therapeutic use UTI and RTI, peritonitis, and bacterial meningitis.
3. Kanamycin has also been largely superseded by less toxic, more effective agents.
Against pseudomonas and most G+ve organism.
4. Netilmicin: active against bacteria that are resistant to gentamicin, P. aeroginosa,
Serratia, enterobacter and Klebsiella.
5. Neomycin is effective against many G-ve spp and is also effective G+ve (ex: S.
aureus).
Note:
Because of its serious effects when absorbed systemically is used most frequently in
dermatologic and ophthalmic ointments. In addition, neomycin can be used orally as a
bowel preparation for surgery or for the management of hepatic coma.
Resistance bacteria of AGs:
1. Resistance to AGs antibiotics may occur by alteration of the 30s ribosomal target
protein (ex: a single amino acid change in the p12 protein prevents streptomycin
binding). Resistance may be also a rise through alterations in cell wall permeability or in
the energy dependent transport a cross the cytoplasm membrane.
2. Production of Aminoglycoside modifying enzymes (Adenylases, acetylases,
phosphorylases) are the principal cause of resistance to AGs is most important
mechanism of acquired resistance. The genes for these enzymes are often plasmid
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)23(
mediated, located on transposons, and transferable from one bacterial species to another.
The enzymes alter the structure of the AGs molecules, thus inactivating the drug.
2. Tetracyclines:
1. The tetracyclines are a large family of antibiotics that were discovered as natural
products of Streptomyces bacteria beginning in the late 1940s, the group of tetracyclines
and the related compounds that are formed during their synthesis by only three
microorganisms (1. Str. aureofaciens , 2. Str. rimosus, 3. Nocardia sulphurea) several
semisynthetic preparations were obtained from the metabolites of these microorganisms.
They are used in medicine as well.
2. Common members of the tetracycline group
of antibiotics are:
• Oxytetracycline is a commonly used form of tetracycline that is also known
as Terramycin.
• Chlortetracyline is another commonly used form of tetracycline that is also
known as Aureomycin.
• Doxycycline is a semisynthetic form of tetracycline that has a longer-lasting
effect than the natural form of tetracycline.
• Minocyline is a semisynthetic form of tetracycline.
3. 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.
4. However, most bacteria possess an active transport system for tetracycline that will
allow intracellular accumulation of the antibiotic at concentrations 50 times as great as
that in the medium. This greatly enhances its antibacterial effectiveness and accounts for
its specificity of action, since an effective concentration cannot be accumulated in
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)24(
animal cells. Thus a blood level of tetracycline which is harmless to animal tissues can
halt protein synthesis in invading bacteria.
5.The tetracyclines have a remarkably low toxicity and minimal side effects when
taken by animals. The combination of their broad spectrum and low toxicity has led to
their overuse and misuse by the medical community and the wide-spread development
of resistance has reduced their effectiveness. Nonetheless, tetracyclines still have some
important uses, such as the use of doxycycline in the treatment of Lyme disease.
a. Chemistry:
Tetracyclins are four-ring molecules with five different sites for substitution, therapy
giving rise to a family of molecules with different substituents at different sites.
Members of the family differ more in their pharmacologic properties than in their
spectrum of activity.
b. Mechanism of action:
Tetracyclines are primarily bacteriostatic, inhibiting protein synthesis by binding to 30
S ribosomes at a point that blocks attachment of aminoacyl-tRNA to the acceptor site on
the mRNA ribosome complex. Unlike the aminoglycosides, their effect is reversible;
they are bacteriostatic rather than bactericidal.
c. Pharmakinetics of tetracyclines:
The tetracyclines are absorbed orally. The tetracyclines are chelated by divalent
cations, and their absorption and activity are reduced. Thus, they should not be taken
with dairy products or many antacid preparations. Tetracyclines are excreted in the bile
and urine in active form.
d. Side effects of tetracyclines:
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)25(
1. The tetracyclines have a strong affinity for developing bone and teeth, interference
with bone development and brown staining of teeth occurs in the fetus and children.
Systemic administration may cause liver damage. The drugs are deposited in the teeth
and bones because of their chelating properties and the formation of a tetracyclinecalcium orthophosphate complex. To which they give a yellowish color, and they are
avoided in children up to 8 years of ago.
2. Common complications of tetracyclines therapy are gastrointestinal disturbance due
to alteration of the normal flora, predisposing to superinfection with tetracyclineresistant organisms and vaginal or oral candidiasis (thrush) due to the opportunistic yeast
Candida albicans. Tetracyclines suppress diarrhea and encouraging overgrowth by
resistant and undesirable bacteria (ex: S. aureus) and fungi (ex: Candida).
3. Hypersensitivity reactions can occur.
4. When tetracyclines are administered orally, gastrointestinal irritation is common.
5. High doses of tetracyclines can produce hepatic dysfunction. This reaction is
exacerbated during pregnancy.
e. Preparations and administration:
Tetracyclines are available in formulations suitable for oral, IV, and Im administration,
they are also available for topical use, including ophthalmic solutions.
f. Spectrum of activity
1. Tetracycline is a broad-spectrum antibiotic that attacks both gram-positive
and gram-negative bacteria. Tetracyclines are able to combat pathogens that
invade cells because they can enter body tissues. They are used against urinary
tract infections, rickettsial infection, and chlamydial infections. Tetracyclines are also
used as the secondary treatment for gonorrhea and syphilis, Pseudomonas aeruginosa is
less sensitive but is generally susceptible to tetracycline concentrations that are
obtainable in the bladder.
2. Doxycycline is frequently used to treat chronic prostatitis, sinusitis, syphilis,
Chlamydia, pelvic inflammatory disease, acne and rosacea. In addition, it is used in the
treatment and prophylaxis of anthrax and in prophylaxis against malaria. It is also
effective against Yersinia pestis (the infectious agent of bubonic plague) and is
prescribed for the treatment of Lyme disease, ehrlichiosis and Rocky Mountain spotted
fever. Because doxycycline is one of the few medications that is effective in treating
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)26(
Rocky Mountain spotted fever (with the next best alternative being chloramphenicol), it
is indicated even for use in children for this illness.
3. Mycoplasmal infections.
Tetracyclines may be beneficial in the treatment of acne.
3. Chloramphenicol:
Ehrlich and his collaborators isolated from cultivated soil of Venezuela in 1947 the
strain of a Streptomyces species that produced an antibiotic with a broad antimicrobial
spectrum. The antibiotic was chloramphenicol, and the organism that produced it, Str.
venezuelae. Chloramphenicol was originally discovered and purified from the
fermentation of a Streptomyces species, but currently it is produced entirely by chemical
synthesis.
a. Chemistry
Chloramphenicol is a relatively simple molecule containing a nitrobenzene nucleus,
which is responsible for some of the toxic problems associated with the drug. Other
derivatives have been produced, but none is in widespread clinical use.
Fig. (1): Chemical structure of chloramphenicol
b. Mechanism of action of chloramphenicol:
Chloramphenicol has affinity for the large (50S) ribosomal subunit where it blocks the
action of peptidyl transferase, therapy preventing peptide bond synthesis (Fig. 33.16.).
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)27(
the drug has some inhibitory activity on human mitochondrial ribosomes (which are also
70 S) which may account for some of the dose-dependent toxicity to bone marrow.
c. Pharmakinetics of chloramphenicol:
1. Chloramphenicol is absorbed rapidly from the gastrointestinal tract.
2. Chloramphenicol is widely distributed in body fluids and reaches therapeutic levels
in CSF fluid, it is also present in bile, milk, and aqueous humor.
3. Chloramphenicol is metabolized in the liver by conjugation with glucuronic acid to
yield a microbiologically inactive form that is excreted by the kidneys.
4. It is metabolites are excreted in the urine.
d. Preparations and administration:
Chloramphenicol is well absorbed when given orally, but be given IV if the patient
cannot take drugs by mouth. Topical preparations (ophthalmic solutions and ointments)
are also available. It is well distributed in the body and penetrates host cells.
e. Side effects of chloramphenicol:
Nitrobenzene is a bone marrow suppressant, and the structurally similar
chloramphenicol molecule has similar effects. This toxicity takes two forms:
a. dose-dependent bone marrow suppression, which occurs if the drug is given for long
periods and is reversible when treatment is stopped.
b. an idiosyncratic reaction causing aplastic anemia, which is not dose dependent and
irreversible. It can occur after treatment has stopped, but is fortunately very rare,
occurring in about 1-30000 patients treated.
1. Hypersensitivity reactions can occur.
2. Chloramphenicol was once a highly prescribed antibiotic and a number of deaths
from anemia occurred before its use was curtailed.
3. Bone marrow depression resulting in panocytopenia.
4. Superinfections can occur, including oropharyngeal candidiasis and acute
staphylococcal enterocolitis.
5. Gray baby syndrome:
Chloramphenicol is also toxic to neonates, particularly premature babies whose liver
enzyme systems are incompletely developed. This can result in gray baby syndrome
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)28(
a. this condition is seen in neonates, especially premature infants, who have been given
relatively large doses of Chloramphenicol.
b. Cyanosis, respiratory irregularities, vasomotor collapse, abdominal distention, loose
green stools, and an ashen-gray color characterize this often fetal syndrome.
c. The condition develops because of the immature hepatic conjugating mechanism and
the inadequate mechanism for renal excretion in neonates.
5. Chloramphenicol
is an antibiotic of last resort because it inhibits formation of blood cells, causing
aplastic anemia.
6. The eucaryotic cells most likely to be inhibited by chloramphenicol are those
undergoing rapid multiplication, thereby rapidly synthesizing mitochondria.
Note:
Now it is seldom used in human medicine except in life-threatening situations (e.g.
typhoid fever).
f. Spectrum of activity:
Chloramphenicol is a broad-spectrum antibiotic that is small enough to effect
areas of the body that are too small for other antibiotics to enter. Is a protein synthesis
inhibitor that has a broad spectrum of activity but it exerts a bacteriostatic effect. It is
effective against intracellular parasites such as the rickettsiae.
The drug achieves satisfactory concentrations in the CSF, topical preparations are used
variety of bacterial species, both G-ve and G+ve, aerobes and anaerobes, including
intracellular organisms.
1. Chloramphenicol is the drug of choice for typhoid fever.
2. Chloramphenicol has been used in the treatment of bacterial meningitis caused by
(particularly H. influenzae) is effectively treated.
3. Rickettsial diseases and brucellosis can be treated with chloramphenicol; however,
tetracyclines are the preferred agents. Chloramphenicol use is now restricted to
treatment of rickettsial or ehrlichial infections in which tetracyclines cannot be used
because of hypersensitivity or pregnancy.
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)29(
In some developing countries Chloramphenicol use is more extensive because of its
low cost and proven efficacy in diseases such as typhoid fever and bacterial meningitis.
g. Resistance bacteria of Chloramphenicol
The most common mechanism of Chloramphenicol resistance involves the inactivation
of the drug by a plasmid mediated enzymatic mechanism which is easily transferred
with in G-ve bacterial populations. Chloramphenicol acetyl transferases produced by
resistant bacteria are intracellular, but capable of inactivating all Chloramphenicol in the
immediate environment of the cell. Acetylated Chloramphenicol fails to bind to the
ribosomal target.
4. Macrolides:
1. The investigators study intensely the antibacterial products of metabolism of various
streptomycetes a high molecular weight, Woodward (1957) gave the name of macrolides
to these compounds, also as the product of metabolism of Str. erythreus. Macrolide
antibiotics are characterized by the presence in their molecules of the macrocyclic
lactone ring connected with one or several carbohydrate residues, these are usually
amino sugars.
Chemistry:
Macrolides contain a lacton ring and one or more deoxy sugars.
Fig (1): The chemical structure of a macrolide antibiotic.
Azithromycin: Is a subclass of macrolide antibiotics. Azithromycin is one of the world's
best-selling antibiotics. It is s derived from erythromycin, but it differs chemically from
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)30(
erythromycin in that a methyl-substituted nitrogen atom is incorporated into the lactone
ring,
thus making the lactone ring 15-membered. Azithromycin is used to treat certain
bacterial infections, most often bacteria causing middle ear infections, tonsillitis, throat
infections, laryngitis, bronchitis, pneumonia and sinusitis. It is also effective against
certain sexually transmitted diseases, such as non-gonococcal urethritis and cervicitis.
The chemical structure of Azithromycin:
2. The most important members of the group are
erythromycin, clarithromycin, and azithromycin .
3. Mechanism of action of Macrolides:
Macrolides inhibit bacterial protein synthesis by binding to the 50S ribosomal subunit.
Binding inhibits elongation of the protein by peptidyl transferase or prevents
translocation of the ribosome or both. Macrolides are bacteriostatic for most bacteria but
are cidal for a few Gram-positive bacteria.
4. Pharmacokinetics:
a. All forms of erythromycin are absorbed following oral administration and diffuse in to
most body tissues and fluids except CSF fluid.
b. Macrolides are concentrated in the liver and is excreted primarily in bile and feces.
5. Preparations and administration:
a. Oral macrolides base is supplied as enteric coated tablets.
b. Macrolides ethylsuccinate, is available as granules or powder for oral suspension,
other available oral salts are stearate, and estolate.
c. Sterile macrolides gluceptate is available for parenteral administration.
d. Macrolides base is available in topical formulations.
6. Side effects of Macrolides:
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)31(
a. Cholestatic hepatitis has occurred in adults treated for a week or longer with the
estolate form. Hepatitis can also occur with the ethylsuccinate, and possibly with the
stearate. The hepatitis is uncommon and is reversible.
b. Epigastric distress can occur.
c. A high incidence of thrombophlebitis occurs when erythromycin is administered
intravenously even when the drug is dissolved in a large fluid volume.
d. Superinfection can occur.
e. Transient deafness has been reported, especially with higher doses.
f. Erythromycin causes nausea and vomiting after oral administration in a significant
number of patients. Jaundice is associated with some formulations of the drug.
7. Spectrum of activity:
a. This drug is effective against G+ve organism, including some strains of S. aureus that
are penicillin G resistance.
b. Neisseria species, some strains of H. influenzae, and Bordetella, Legionella,
Treponema, Mycoplasma species are sensitive to erythromycin.
c. In general, it is not very active against most G-ve bacilli.
d. Macrolide antibiotics can be administered
orally, making them the choice for treating children who have streptococcal and
staphylococcal infections. The most commonly used macrolide is Erythromycin,
which is used to treat legionellosis, mycoplasmal pneumonia, and streptococcal
and staphylococcal infections.
e. Clarithromycin is also active against mycobacterium. In addition, both azithromycin
and clarthromycin have demonstrated efficacy against Borrelia burgdorferi , the causal
agent of lyme disease and the protozoan parasite Toxoplasma gondii, which causes
toxoplasmosis.
5. Lincosamides :
1. The antibacterial products of metabolism of various streptomycetes, the product of
metabolism of Str. lincolnesis . including clindomycin and lincomycin.
Chemistry:
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)32(
Clindomycin is the 7-deoxy,7-chloro derivative of the parent drug, lincomycin, which it
has essentially replaced.
The chemical structure of Lincosamides
2.Pharmakinetics
a. Although it is well absorbed following oral administration, clindomycin most
often is administered parenterally.
b. The drug is widely distributed in body fluids and tissues but does not pass readily
into CSF fluid.
c. Most of the drug is metabolized to the inactive sulfoxide form, which then is
excreted in the bile and urine.
2. Mode of action: Lincosamides is chemically unrelated to the macrolides but has a
similar mode of action and spectrum, are a miscellaneous group of protein synthesis
inhibitors with activity similar to the macrolides.
3. Spectrum activity: Lincomycin has activity against Gram-positive bacteria and some
Gram-negative bacteria (Neisseria, H. influenzae). Clindamycin is usually used to treat
infections with anaerobic bacteria but can also be used to treat some protozoal diseases,
such as malaria. It is a common topical treatment for acne, and can be useful against
some methicillin-resistant Staphylococcus aureus (MRSA) infections. The most severe
common adverse effect of clindamycin is Clostridium difficile-associated diarrhea (the
most frequent cause of pseudomembranous colitis). Although this side-effect occurs
with almost all antibiotics, including beta-lactam antibiotics, it is classically linked to
clindamycin use.
4. Side effect: Pseudomembranous colitis can occur, resulting in diarrhea, abdominal
pain, fever, and mucus and blood in the stools. At one time, this condition was often
fatal, but now that the causative organism is known to be C. difficile, it can treated with
vancomycin.
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)33(
5.Resistance bacteria of Macrolides and clindamycin
a. Altered accumulation (Minimal outer membrane penetration, efflux pump)
b. Altered target (methylation of rRNA).
c. Enzymatic inactivation (phosphotransferase, esterase).
3. PLASMA MEMBRANE INHIBITORS
1. Plasma membrane inhibitors are antibiotics that interfere with the functionally
of the plasma membrane of a pathogen.
2. The commonly used plasma membrane
inhibitor antibiotic is Polymyxin B, which combats gram-negative bacteria such
as Pseudomonas. Today, when combined with neomycin, it is used in nonprescription
topical antibiotics for superficial infections.
a. The plasma membranes that encompass all kinds of living cells perform a variety of
vital functions. The structure of these membranes in bacterial cells differs from that in
mammalian cells and allows the application of some selectively toxic molecules, but
these are few in number compared with those acting at other target sites. The most
important are the polymyxins, which act on the membranes of G-ve bacteria, the
polyene antifungal agents (amphotericin B, nystatin) also act by inhibiting membrane
function.
b. Polymyxins:
The group of polymyxins includes antibiotics substances of the polypeptide nature
produced by various strains of B. polymyxa and B. circulans that differ in amino acid
composition , five various polymyxins were first differentiated: polymyxin A
(aerosporin), polymyxin B, C, D, E (Colistin), and M. Polymyxin B and E are the most
common members of the family still in clinical use.
The chemical structure of
polymyxin
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)34(
c. Polymyxins are bacteriocidal cyclic polypeptides that disrupt the structure of cell
membranes. The free amino groups of polymyxins act as cationic detergents, disrupting
the phospholipids structure of the cell membranes.
d. This antibiotics inhibiting membrane bound protein (enzyme) permeases which is
involved in transport processes (active and passive osmosis), in this state the antibiotics
is bacteriocidal, these biologically active compounds effect wall permeability in cells
sensitive to them. Cell permeability is known to depend in the first instance on the
cytoplasmic membrane.
8. Polymyxins act selectively on G –ve bacteria , Pseudomonas aeroginosa , fungicidal
effects, E. coli, K. pneumoniae, S. typhi, V. cholera, Brucella abortis.
9. Resistance due to chromosomally mediated alterations in membrane structure or
antibiotic uptake has been reported.
10. Polymyxins are primarily used topically but have also been for gut decontamination,
wounds irrigation and as a bladder washout.
11. In the past they have been used systemically, but due to poor distribution in tissues,
nephrotoxicity and neurotoxicity they have been superseded by less toxic agents.
4. NUCLEIC ACID INHIBITORS
Some antibiotics and chemotherapeutic agents affect the synthesis of DNA or RNA, or
can bind to DNA or RNA so that their messages cannot be read. Either case, of course,
can block the growth of cells. The majority of these drugs are unselective, however, and
affect animal cells and bacterial cells alike and therefore have no therapeutic application.
Two nucleic acid synthesis inhibitors which have selective activity against procaryotes
and some medical utility are the quinolones and rifamycins.
Nucleic acid inhibitors interfere with the formation of nucleic acids. The commonly
used nucleic acid inhibitor antibiotics follow.
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)35(
1. Rifamycins
1. Sensi et al., 1959 isolated from the culture of Str. mediterranei , an antibacterial
antibiotics which they named rifamycin (rifampin). It is a semisynthetic compound
derived from Amycolatopsis rifamycinica (formerly known as Amycolatopsis
mediterranei and Streptomyces mediterranei).
Chemistry:
Rifamycin belongs to the group of complex macrocyclic antibiotics, it is zwitterionic
and is soluble in water at low PH.
Fig: The chemical structure of rifampin
2. Mechanism of action: Rifampin inhibits RNA synthesis in bacteria and chlamydiae
by binding to DNA-dependent RNA polymerase, which prevents the initiation of RNA
synthesis. Selective toxicity is based on the far greater affinity for bacterial polymerases
than for the equivalent human enzymes.
3. Pharmacokinetics: Rifampin is well absorbed from the gastrointestinal tract, and it is
widely distributed in tissues and is excreted mainly through the liver. Rifamycins can
reach the cerebrospinal fluid and enter tissues and abscesses. The disadvantage of
rifamycins is that they cause urine, feces, saliva, sweat and tears to appear an orange-red
color. It is crosses the blood brain barrier and reaches high concentrations in saliva.
4. Preparations: Rifampin is available as capsules for oral use.
5. Side effects:
a. Urine, sweat, tears, and contact lenses may take on an orange color because of
rifampin administration, this effect is harmless.
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)36(
b. Light-chain proteinuria and impaired antibody response may occur.
c. Rifampin induces hepatic microsomal enzymes .
d. Rashes, gastrointestinal disturbances, and renal damage have been reported.
e. Jaundice and severe hepatic dysfunction are occasionally produced.
5. Spectrum activity:
Rifampicin (or rifampin) is a bactericidal antibiotic from the rifamycin group.
a. They are used to treat tuberculosis and leprosy. A commonly used rifamycin is
rifampin, which is used to attack Mycobacterium tuberculosis that causes tuberculosis
and leprosy. It has been found to have greater bactericidal effect against M. tuberculosis
than other anti-tuberculosis drugs, and it has largely replaced isoniazid as one of the
front-line drugs used to treat the disease, especially when isoniazid resistance is
indicated. It is effective orally and penetrates the cerebrospinal fluid so it is useful for
treatment of bacterial meningitis.
b. Rifampicin is typically used to treat Mycobacterium infections, including tuberculosis
and leprosy; and also has a role in the treatment of methicillin-resistant Staphylococcus
aureus (MRSA) in combination with fusidic acid. It is used in prophylactic therapy
against Neisseria meningitidis (meningococcal) infection. It is also used to treat
infection by Listeria monocytogenes, Neisseria gonorrhoeae, Haemophilus influenzae
and Legionella pneumophila.
6. Resistance due to chromosomally mediated alterations in target site (Mutant RNA
polymerase)
Three additional synthetic chemotherapeutic agents have been used in the treatment of
tuberculosis: isoniazid (INH), para-aminosalicylic acid (PAS), and ethambutol. The
usual strategy in the treatment of tuberculosis has been to administer a single antibiotic
(historically streptomycin, but now, most commonly, rifampicin is given) in conjunction
with INH and ethambutol. Since the tubercle bacillus rapidly develops resistance to the
antibiotic, ethambutol and INH are given to prevent outgrowth of a resistant strain. It
must also be pointed out that the tubercle bacillus rapidly develops resistance to
ethambutol and INH if either drug is used alone. Ethambutol inhibits incorporation of
mycolic acids into the mycobacterial cell wall. Isoniazid has been reported to inhibit
mycolic acid synthesis in mycobacteria and since it is an analog of pyridoxine (Vitamin
B6) it may inhibit pyridoxine-catalyzed reactions as well. Isoniazid is activated by a
mycobacterial peroxidase enzyme and destroys several targets in the cell. PAS is an antifolate, similar in activity to the sulfonamides. PAS was once a primary anti-tuberculosis
drug, but now it is a secondary agent, having been largely replaced by ethambutol.
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)37(
Isoniazid is also called isonicotinyl hydrazine or INH. Isoniazid is a first-line antituberculosis medication used in the prevention and treatment of tuberculosis. Isoniazid is
never used on its own to treat active tuberculosis because resistance quickly develops.
2. Quinolones
1. Quinolones are synthesis agents that interfere with replication of the bacterial
chromosome including Nalidixic acid.
2. Quinolones interfere with DNA gyrase (an enzyme), which is involved in DNA
replication.
3. Quinolones represent a large family of bacteriocidal synthetic agents which, in a
manner similar to the cephalosporins, can be generally grouped in categories or
"Generations" based on their spectrum of activity (Fig: 33:26). Nalidixic acid is the first
generation prototypes, but the addition of fluorine at position 6 of the main quinolone
ring (Fluoroquinolones) (Fig: 33:27) has improved antibacterial activity, leading to the
synthesis of many additional compounds, this compound including Ciprofloxacin,
Norfloxacin, Ofloxacin, Moxifloxacin, and trovafloxacin.
4. Mechanism of action
The antibacterial activity of Quinolones is due to their ability to inhibit the activity of
bacterial DNA gyrase enzyme and Topoisomerase II,IV (which is an enzyme necessary
to separate replicated DNA, and thereby inhibits cell division), which is essential for
DNA replication and allows supercoils to be relaxed and reformed. Binding of the drug
inhibits DNA gyrase activity. During replication of the bacterial chromosome, DNA
gyrase produces and removes supercoils in DNA ahead of the replication fork to
maintain the proper "Tension" required for efficient DNA duplication. Topoisomerase
IV similarly acts to remove supercoils and to separate newly formed DNA "Daughter"
strand after replication (Fig. 33.28). These enzymes thus act in concert to insure that the
DNA molecule has the proper conformation for efficient replication and packaging
within the cell. Quinolones are able to interfere with these essential enzymes in bacteria
while not affecting their counterparts in mammalian cells.
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)38(
5. Resistance to Quinolones is chromosomally mediated
a. Mutations which changes the target enzymes in a manner that affects Quinolones
binding (changing in DNA gyrase subunits structure resulting in lowered affinity for the
drug).
b. Changes in cell wall permeability, resulting in decreased uptake, or by efflux. These
mechanisms may also lead to cross-resistance to other unrelated agents affected by the
same process.
6. Quinolones are primarily administered orally since they are readily absorbed from the
gastrointestinal tract, achieving significant serum conc. and good distribution throughout
the body compartments. Excretion is mostly in the urine, though a small proportion is
excreted in the feces.
7. Spectrum activity:
Fluoroquinolones comprise a group of antibiotics that have a broad-spectrum, and are
able to enter cells to attack pathogens.
a. Nalidixic acid is only active against enterobacteria, and, although occasionally
employed in treatment of urinary tract infections, its use has largely been replaced by the
newer fluorinated compounds. The compound is unusual in that it is effective against
several types of Gram-negative bacteria such as E. coli, Enterobacter aerogenes, K.
pneumoniae and Proteus species which are common causes of UTIs. It is not usually
effective against Pseudomonas aeruginosa, and Gram-positive bacteria may be resistant.
b. Nalidixic acid is a bactericidal agent that binds to the Some quinolones penetrate
macrophages and neutrophils better than most antibiotics and are thus useful in
treatment of infections caused by intracellular parasites.
c. The fluoroquinolone, Cipro. (ciprofloxacin) was recently touted as the drug of choice
for treatment and prophylaxis of anthrax, which is caused by a Gram-positive bacillus,
Bacillus anthracis.
8. Side effects of Quinolones:
a. Gastrointestinal intolerance disturbances are the most common side effect of
Quinolones.
b. Neurotoxicity and Photosensitivity reaction are less common.
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)39(
c. The disadvantage of fluoroquinolones is that they may interfere with cartilage
development, making them unsafe for pregnant, not used to children because of possible
toxic effects on cartilage development.
women and children.
d. Confusion, dizziness, headache.
e. Rushes.
3. Metronidazole:
1. Metronidazole is a nitroimidazole with antiparasitic and antibacterial properties.
2. Mechanism of action:
After entry into the microbial cell the molecule is activated by reduction, and the
reduced intermediate products are responsible for antimicrobial activity, probably
through interaction with, and breakage of the cell's DNA. The reactive intermediates are
short-lived and decompose to none toxic inactive end products. Metronidazole is active
only against anaerobic organisms because only these can produce the low redox
potential necessary to reduce the parent drug.
3. Metronidazole is usually given orally or rectally. It is well absorbed and well
distributed in tissues and CSF. The drug is metabolized and most of the parent
compound and metabolites are excreted in the urine.
4. Metronidazole is also effective against other protozoan parasites such as
Giardia lamblia , Entamoeba coli and E. histolytica. It is an important agent for
treatment of infections caused by anaerobic bacteria.
5. Metronidazole resistance is relatively rare and appears to involve either an alteration
in uptake or a decrease in cellular reductase activity, therapy slowing the activation of
the intracellular drug.
6. The most serious side effects of Metronidazole involved the central nervous system
and include peripheral neuropathy. However, these are relatively uncommon and usually
seen only in patient on large doses or prolonged treatment.
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)40(
ANTIMETABOLITES
Antimetabolites are a group of drugs that interfere with metabolite, which is a
substance (such as an enzyme) that is necessary for cell metabolism. Common
antimetabolites follow.
1. Sulfonamides
-Sulfonamides or sulfa drugs are used to treat urinary tract infections and control
infections in burn patients. These were among the first synthetic antimicrobial
drugs used. Commonly used sulfonamides are:
• Silver sulfadiazine is used to control infections common in burn patients.
• TMP-SMZ is a combination of trimethoprim and sulfamethoxazole and is
used to combat intestinal tract and urinary tract gram-negative pathogens.
-Many of the synthetic chemotherapeutic agents (synthetic antibiotics) are competitive
inhibitors of essential metabolites or growth factors that are needed in bacterial
metabolism. Hence, these types of antimicrobial agents are sometimes referred to as
anti-metabolites or growth factor analogs, since they are designed to specifically
inhibit an essential metabolic pathway in the bacterial pathogen. At a chemical level,
competitive inhibitors are structurally similar to a bacterial growth factor or metabolite,
but they do not fulfill their metabolic function in the cell. Some are bacteriostatic and
some are bactericidal. Their selective toxicity is based on the premise that the bacterial
pathway does not occur in the host.
-Sulfonamides were introduced as chemotherapeutic agents by Domagk in 1935, who
showed that one of these compounds (prontosil) had the effect of curing mice with
infections caused by beta-hemolytic streptococci. Chemical modifications of the
compound sulfanilamide gave rise to compounds with even higher and broader
antibacterial activity. The resulting sulfonamides have broadly similar antibacterial
activity, but differ widely in their pharmacological actions. Bacteria which are almost
always sensitive to the sulfonamides include Streptococcus pneumoniae, beta-hemolytic
streptococci and E. coli. The sulfonamides have been extremely useful in the treatment
of uncomplicated UTI caused by E. coli, and in the treatment of meningococcal
meningitis (because they cross the blood-brain barrier).
The sulfonamides (e.g. Gantrisin and Trimethoprim) are inhibitors of the bacterial
enzymes required for the synthesis of tetrahydofolic acid (THF), the vitamin form of
folic acid essential for 1-carbon transfer reactions. Sulfonamides are structurally similar
to para aminobenzoic acid (PABA), the substrate for the first enzyme in the THF
pathway, and they competitively inhibit that step. Trimethoprim is structurally similar to
dihydrofolate (DHF) and competitively inhibits the second step in THF synthesis
mediated by the DHF reductase. Animal cells do not synthesize their own folic acid but
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)41(
obtain it in a preformed fashion as a vitamin. Since animals do not make folic acid, they
are not affected by these drugs, which achieve their selective toxicity for bacteria on this
basis.
The chemical structures of sulfanilamide and para-aminobenzoic acid (PABA). In
bacteria, sulfanilamide acts as a competitive inhibitor of the enzyme
dihydropteroate synthetase, DHPS, which catalyses the conversion of PABA to
dihydropteroate, a key step in folate synthesis. Folate is necessary for the cell to
synthesize nucleic acids (DNA and RNA), and in its absence, cells will be unable to
divide. Hence, sulfanilamide and other sulfonamides exhibit a bacteriostatic rather
than bactericidal effect.
- Resistance is widespread with plasmid mediated genes coding for an altered
dihydropteroate synthtase
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
ANTIFUNGAL DRUGS
Antifungal drugs inhibit the growth of fungi. The commonly used antifungal drugs
follow:
Polyenes
Polyenes are a group of antifungal antibiotics that combine with the ergosterol in the
plasma membrane of fungi. This causes the plasma membrane to become extremely
)42(
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
permeable, ultimately resulting in the death of the fungi. A commonly used polyene
is amphotericin B, which is used to treat histoplasmosis, coccidioidomycosis,
and blastomycosis. However, amphotericin B is toxic to kidneys. Enclosing
amphotericin
B in liposomes when administering the drug reduces its toxicity.
Imidazoles
Imidazoles interfere with fungal ergosterol synthesis. Commonly used imidazoles
are:
• Clotrimazole is used to treat cutaneous mycoses. Two example are athlete’s
foot and vaginal yeast infections.
• Miconazole is similar to clotrimazole. Both drugs are used topically and
sold without a prescription.
Triazoles
Triazoles are similar to imidazoles, but are less toxic than other antifungals.
Griseofulvin
Griseofulvin is an antibiotic used to treat dermatophytic fungal infections of the
hair and nails. This includes tinea capitis. Griseofulvin is taken orally and enters
the keratin of skin, hair, and nails. Its purpose is to interfere with fungal mitosis,
inhibiting reproduction.
Tolnaftate
Tolnaftate is a topical treatment for athlete’s foot. Its mechanism for action is
unknown.
ANTIVIRAL DRUGS
Antiviral drugs interfere with the replication of viruses. Commonly used antiviral
drugs follow.
Amantadine
Amantadine prevents a virus from entering the cell or from uncoating once the
virus enters the cell. It is used (though it has limited usefulness) to prevent
influenza A, but has no practical effect once the virus infects the cell.
Nucleoside analogs
Nucleoside analog antiviral drugs affect the synthesis of viral DNA or RNA.
Commonly used nucleoside analogs are:
• Acyclovir is used to combat viruses that cause herpes.
• Ribavirin is used to treat rotavirus-caused pneumonia in infants.
• Ganciclovir is used fight cytomegalovirus infections that are common in
transplant patients and patients who have AIDS.
• Trifluridine is used to treat the eye infection caused by acyclovir-resistant
herpes keratitis.
• Zidovudine (AZT) is used in HIV infection. This drug blocks the synthesis
of DNA from RNA by the enzyme reverse transcriptase.
)43(
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
ANTIPROTOZOAN DRUGS
Antiprotozoan drugs are used to combat parasitic infection, such as the disease
malaria. Commonly used antiprotozoan drugs follow.
Chloroquine
Chloroquine is a replacement for quinine, which was the traditional treatment
for malaria.
Mefloquine
Mefloquine is a secondary treatment for malaria when there is a resistance to
chloroquine.
Quinacrine
Quinacrine is used to treat giardiasis.
Diiodohydroxyquin
Diiodohydroxyquin is used to treat intestinal amoebic diseases. However,
diiodohydroxyquin can cause damage to the optic nerve if the dosage is not carefully
controlled.
Metronidazole
Metronidazole combats parasitic protozoa and obligate anaerobic bacteria. It is
used to treat vaginitis caused by Trichomonas vaginalis, giardiasis, and amoebic
dysentery.
Nifurtimox
Nifurtimox is used to combat Chagas’ disease. However, nifurtimox can cause
side effects, such as nausea and convulsions.
ANTIHELMINTHIC DRUGS
Antihelminthic drugs are used to combat helminths, which are parasitic flatworms.
Commonly used antihelminthic drugs follow.
Niclosamide
Niclosamide is used to treat tapeworm infections. This drug inhibits ATP production
in aerobic conditions.
Praziquantel
Praziquantel is also used to destroy tapeworm infections and is used to treat
schistosomiasis and other fluke-caused diseases. This drug alters the permeability
of the organism’s plasma membrane.
Mebendazole
Mebendazole is used to combat intestinal helminthic infections, such as ascariasis,
whipworms, and pinworms.
)44(
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)45(
Pharmacology:
Is study of the biochemical and physiologic a aspect of drug effects including
absorption, distribution, metabolism, toxicity, and specific mechanism of drug action.
The branches of Pharmacology including:
1. Pharmacognosy.
2. Pharmacodynamics.
3. Pharmacokinetics.
4. Therapeutics.
5. Toxicology.
6. Dosology.
7. Pharmacy.
Factors effecting drug absorption:
1. Dosage.
2. Route of administration including:
I. Alimentary routes including:
a. Orally.
b. Rectal.
c. Sublingual.
* Advantages of Alimentary administration:
1. An Alimentary route is generally the safest route of administration. The delivery of
the drug in to the circulation is slow after oral administration, so that rapid high blood
levels are avoided and untoward effects are less likely.
2. Alimentary routes allow for convenient dosage forms that do not require sterile
technique.
** Disadvantages of Alimentary administration:
1. The rate of absorption is variable. This becomes a problem if only a small range in
blood levels separates a drugs desired therapeutic effect from it is toxic effects.
2. Irritation of mucosal surfaces can occur.
3. The compliance of the patient is not ensured.
4. Oral administration of some drugs passages through firstly the liver and the
resulting initial hepatic metabolism is avoided by the sublingual route.
II. Parenteral routes
1. Intravenous (IV).
2. Intramuscular (IM).
3. Subcutaneous (SC).
4. Intrapertoneal (IP).
5. Epidural injection.
6. Transdermal.
Dr. Zahra Muhsin Ali
/ Lectures of antibiotics / Biology dep. /
4
th
year
)46(
* Advantages of Parenteral administration:
1. There is a rapid response, which may be required in an emergency.
2. The dose can often be more accurately delivered.
3. Parenteral administration provideds an alternative when the alimentary route is not
feasible (ex: When the patient is unconscious).
**Disadvantages of Parenteral administration:
1. The more rapid absorption can lead to increase in untoward effects.
2. Both a sterile formulation and the use of a septic technique are required.
3. Local irritation may occur at the site of injection.
III. Miscellaneous routes
1. Topical administration: is useful in the treatment of local conditions, there is
usually little systemic absorption.
2. In halation provides a rapid route of administration it is used for volatile
anesthetics and many bronchodilators.
Reference:
1. Antimicrobial chemotherapy edited by John M. B. Smith; John E. Payne and Thomas V. Berne.
(2000).
2. Antimicrobial Susceptibility testing edited by Richard Schwalbe and Avery C. Goodwin. (2007).
Download