MICRO PHARM Flashcards

1
Q

Ribavirin

A

Mechanism:

  • Inhibits synthesis of guanine nucleotides by competitively inhibiting inosine monophosphate dehydrogenase.
  • It is a nucleoside analog that can act on both RNA and DNA viruses. Can act on both RNA and DNA viruses but is mechanism of action in DNA viruses remains unclear.

Clinical Use:

  • Hepatitis C, RSV infection, and viral hemorrhagic fever (e.g., Lassa fever, Hantavirus infection).
  • For hepatitis C, it is always used in combination with other medications such as simeprevir or PEG-interferon-α.

Adverse Effects:
Nausea, irritability, hemolytic anemia, and severe teratogenicity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Carbapenems Clinical Use

A

IV imipenem (combined with cilastatin)
IV meropenem
IV ertapenem
IV doripenem
Imipenem is always given with cilastatin, which inhibits human dehydropeptidase I (a renal tubular enzyme that breaks down imipenem).
Meropenem is stable to dehydropeptidase I.

  • Last-resort drugs (used only in life-threatening infections or after other antibiotics have failed) because of the significant adverse effects
  • Broad-spectrum antibiotics with intrinsic beta-lactamase resistance
  • Gram-positive cocci (except for MRSA and Enterococcus faecalis, which are intrinsically resistant)
  • Gram-negative rods, including Pseudomonas aeruginosa (except ertapenem which has limited activity against Pseudomonas)
  • Anaerobes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Azithromycin Mechanism

A
Macrolide 
IV or oral form
Poor CNS penetration poor
Route of elimination → biliary
All macrolides (except azithromycin) have a short half-life.

Mechanism:
Bind to 23S ribosomal RNA molecule of the 50S subunit → blockage of translocation → inhibition of bacterial protein synthesis (bacteriostatic effect)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Tetracyclines

A

Oral or IV minocycline and tetracycline
Oral doxycycline and demeclocycline
CNS penetration → poor
Route of elimination: renal
Doxycycline → only gastrointestinal elimination (doxycycline is the only tetracycline that is not contraindicated in patients with renal failure)
Oral tetracyclines should not be taken with substances that contain large amounts of Ca2+, Mg2+, or Fe2+ (e.g., milk, antacids, iron supplements, respectively) because divalent cations inhibit the intestinal absorption of tetracyclines.

Mechanism:
Bind 30S subunit → aminoacyl-tRNA is blocked from binding to ribosome acceptor site → inhibition of bacterial protein synthesis (bacteriostatic effect)

Clinical Use:
Bacteria that lack a cell wall (e.g, Mycoplasma pneumoniae, Ureaplasma)
Intracellular bacteria, such as Rickettsia, Chlamydia, or Anaplasma (tetracyclines accumulate intracellularly and are, therefore, effective against intracellular pathogens)
Borrelia burgdorferi
Ehrlichia, Vibrio cholerae, Francisella tularensis
Cutibacterium acnes (topical tetracycline is used to treat acne)
Community-acquired MRSA (doxycycline)

Adverse Effects:
Hepatotoxicity
Deposition in bones and teeth → inhibition of bone growth (in children) and discoloration of teeth
Damage to mucous membranes (e.g., esophagitis, GI upset)
Photosensitivity (drug or metabolite in the skin absorbs UV radiation → photochemical reaction → formation of free radicals → damage to cellular components → inflammation (sunburn-like))
Demeclocycline can cause nephrogenic diabetes insipidus
Degraded tetracyclines are associated with Fanconi syndrome.
Rarely: pseudotumor cerebri

Contraindications:
Children < 8 years of age (except doxycycline)  
Pregnant women (except doxycycline)  
Patients with renal failure (except doxycycline)
Cautious use in patients with hepatic dysfunction 

Mechanisms of Resistance:
Plasmid-encoded transport pumps increase efflux out of the bacterial cell and decrease uptake of tetracyclines.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Macrolides

A

Oral or IV → erythromycin, azithromycin, clarithromycin
CNS penetration → poor
Route of elimination → biliary
All macrolides (except azithromycin) have a short half-life.

Mechanism:
Bind to 23S ribosomal RNA molecule of the 50S subunit → blockage of translocation → inhibition of bacterial protein synthesis (bacteriostatic effect)

Clinical Use:
Atypical pneumonia caused by Mycoplasma pneumonia, Legionella pneumophila, Chlamydophila pneumoniae
Bordetella pertussis
STIs caused by Chlamydia
Gram-positive cocci (especially for the treatment of streptococcal infection in patients who are allergic to penicillin)
Neisseria spp.
Second-line prophylaxis for N. meningitidis
Dual therapy with ceftriaxone for N. gonorrhoeae (azithromycin)
Mycobacterium avium (prophylaxis: azithromycin; treatment: azithromycin, clarithromycin)
H. pylori (clarithromycin is the part of triple therapy)
Ureaplasma urealyticum
Babesia spp. (azithromycin in combination with atovaquone)
Erythromycin is used off-label for the treatment of gastroparesis because it increases GI motility.

Adverse Effects:
Increased intestinal motility → GI upset
QT-interval prolongation, arrhythmia
Acute cholestatic hepatitis
Eosinophilia
Rash

Drug Interactions:
Erythromycin enhances the effect of oral anticoagulants (e.g., warfarin).
Erythromycin and clarithromycin increase theophylline serum concentrations due to CYP3A4 inhibition (cytochrome P450 inhibitors)

Contraindications:
Erythromycin estolate and clarithromycin are contraindicated in pregnant women (potentially hazardous to the fetus).
Azithromycin and clarithromycin are contraindicated in patients with hepatic failure (erythromycin should be used cautiously).
Cautious use of clarithromycin in patients with renal failure

Mechanisms of Resistance:
Methylation of the binding site of 23S rRNA prevents the macrolide from binding to rRNA.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Antipseudomonal penicillins

A

Piperacillin, ticarcillin, and carbenicillin.

Mechanism:
D-Ala-D-Ala structural analog.
Bind penicillin-binding proteins (transpeptidases). Block transpeptidase cross-linking of peptidoglycan in cell wall.
Activate autolytic enzymes.
Extended spectrum.
Penicillinase sensitive; use with β-lactamase inhibitors.

Clinical Use:
Pseudomonas spp. and gram ⊝ rods.

Adverse Reaction:
Hypersensitivity reactions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Fidaxomicin

A

Macrocyclic antibiotic (related to macrolides)

Mechanism:
Inhibits the sigma subunit of RNA polymerase, leading to protein synthesis impairment and cell death (bactericidal activity against C difficile)

Clinical Use:
First-line therapy for initial episodes of C. difficile infection (WBC < 15,000)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Acyclovir

A

Mechanism:
Guanosine analogs.
Monophosphorylated by HSV/VZV thymidine kinase and not phosphorylated in uninfected cells –> few adverse effects.
Triphosphate formed by cellular enzymes.
Preferentially inhibit viral DNA polymerase by chain termination.

Clinical Use:
HSV and VZV. Weak activity against EBV.
No activity against CMV.
Used for HSV- induced mucocutaneous and genital lesions as well as for encephalitis.
Prophylaxis in immunocompromised patients.
Only affects actively replicating viruses and has no effect on latent infections of HSV and VZV.
Valacyclovir, a prodrug of acyclovir, has better oral bioavailability.
For herpes zoster, use famciclovir.

Adverse Effects:
Obstructive crystalline nephropathy and acute renal failure if not adequately hydrated
Thrombotic thrombocytopenic purpura
Gastrointestinal symptoms (e.g., nausea)
↑ Transaminases

Mechanism of Resistance:
Mutated viral thymidine kinase.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Famciclovir

A

Mechanism:
Prodrug of penciclovir
Guanosine analog
Monophosphorylated by HSV/VZV thymidine kinase and not phosphorylated in uninfected cells –> few adverse effects.
Triphosphate formed by cellular enzymes.
Preferentially inhibit viral DNA polymerase by chain termination.

Clinical Use:
HSV and VZV. Weak activity against EBV.
No activity against CMV.
Used for HSV- induced mucocutaneous and genital lesions as well as for encephalitis.
Prophylaxis in immunocompromised patients.
No effect on latent forms of HSV and VZV.
For herpes zoster, use famciclovir.

Adverse Effects:
Obstructive crystalline nephropathy and acute renal failure if not adequately hydrated. 
Thrombotic thrombocytopenic purpura
Gastrointestinal symptoms (e.g., nausea)
↑ Transaminases

Mechanism of Resistance:
Mutated viral thymidine kinase.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Sofosbuvir

A

Mechanism:
NS5B inhibitor
Inhibition of NS5B (a RNA-dependent RNA polymerase) → chain termination and disruption of RNA synthesis → prevention of HCV replication

Clinical Use:
Used in combination with velpatasvir or ledipasvir for the treatment of chronic hepatitis C infection.

Adverse Effects:
Fatigue, headache, nausea.
Insomnia
Pruritus
Asthenia
It is contraindicated in severe renal insufficiency.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Glycylcyclines

A

Tigecycline is only agent in this class

Mechanism:
Tetracycline derivative.
Binds to 30S, inhibiting protein synthesis.
Generally bacteriostatic.
Biliary elimination
Should not be taken with milk, antacids, or iron supplements because divalent cations inhibit the absorption of glycylcyclines in the intestines.

Clinical use:
Gram-positive aerobes (not effective against S. viridans or Enterococci; limited efficacy against MSSA)
Gram ⊝
MRSA
VRE
Anaerobes (broad spectrum)
Partially effective against gram-negative aerobes (no effect against Proteus species)
Gram-intermediate bacteria: Borrelia, Mycoplasma, Rickettsia, Chlamydia
Infections requiring deep tissue penetration.

Adverse Effects:
GI upset (nausea, vomiting)
Hepatotoxicity
Deposition in bones and teeth 
Photosensitivity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Tigecycline

A

Glycylcycline class

Mechanism:
Tetracycline derivative.
Binds to 30S, inhibiting protein synthesis.
Generally bacteriostatic.
Biliary elimination
Should not be taken with milk, antacids, or iron supplements because divalent cations inhibit the absorption of glycylcyclines in the intestines.

Clinical use:
Gram-positive aerobes (not effective against S. viridans or Enterococci; limited efficacy against MSSA)
Gram ⊝
MRSA
VRE
Anaerobes (broad spectrum)
Partially effective against gram-negative aerobes (no effect against Proteus species)
Gram-intermediate bacteria: Borrelia, Mycoplasma, Rickettsia, Chlamydia
Infections requiring deep tissue penetration.

Adverse Effects:
GI upset (nausea, vomiting)
Hepatotoxicity
Deposition in bones and teeth 
Photosensitivity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Albendazole

A

Mechanism:
Inhibit microtubule polymerization resulting in decreased glucose uptake and glycogen synthesis, degeneration of mitochondria and endoplasmic reticulum and lysosomal release

Clinical Use:
Ascariasis
Enterobiasis
Trichuriasis
Strongyloidiasis
Hookworm infection
Cysticercosis
Trichinellosis
Toxocariasis
Adverse Effects:
Headache
GI upset (especially in short term use)
Increased liver enzymes
Rash, urticaria
Hair loss
Agranulocytosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Penicillin G

A

IV (crystalline) and IM (benzathine) form

Prototype β-lactam antibiotics.
Probenecid blocks the rebel tubular excretion (sometimes used to increase their serum levels or prolong their half life)
Often coadministered with aminoglycosides to aid their entry into the cell

Mechanism:
D-Ala-D-Ala structural analog. Bind penicillin-binding proteins (transpeptidases).
Block transpeptidase cross-linking of peptidoglycan in cell wall.
Activate autolytic enzymes.

Clinical Use:
Mostly used for gram ⊕ organisms (S pneumoniae, S pyogenes, Actinomyces, C. perfringens, S. agalactiae). Also used for gram ⊝ cocci (mainly N meningitidis) and spirochetes (mainly T pallidum).
Pasteurella
Rheumatic fever prophylaxis
Bactericidal for gram ⊕ cocci, gram ⊕ rods, gram ⊝ cocci, and spirochetes.
β-lactamase sensitive.

Adverse Effects:
Hypersensitivity reactions, direct Coombs ⊕ hemolytic anemia (binds to bacterial breakdown products that form haptens thus forming anti-IgG), drug-induced interstitial nephritis, cutaneous small vasculitis

Resistance:
β-lactamase cleaves the β-lactam ring.
Mutations in penicillin-binding proteins.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Penicillin V

A

Oral form

Prototype β-lactam antibiotics.
Probenecid blocks the rebel tubular excretion (sometimes used to increase their serum levels or prolong their half life)
Often coadministered with aminoglycosides to aid their entry into the cell

Mechanism:
D-Ala-D-Ala structural analog. Bind penicillin-binding proteins (transpeptidases).
Block transpeptidase cross-linking of peptidoglycan in cell wall.
Activate autolytic enzymes.

Clinical Use:
Mostly used for gram ⊕ organisms (S pneumoniae, S pyogenes, Actinomyces, C. perfringens, S. agalactiae). Also used for gram ⊝ cocci (mainly N meningitidis) and spirochetes (mainly T pallidum).
Pasteurella
Rheumatic fever prophylaxis
Bactericidal for gram ⊕ cocci, gram ⊕ rods, gram ⊝ cocci, and spirochetes.
β-lactamase sensitive.

Adverse Effects:
Hypersensitivity reactions, direct Coombs ⊕ hemolytic anemia (binds to bacterial breakdown products that form haptens thus forming anti-IgG), drug-induced interstitial nephritis, cutaneous small vasculitis

Resistance:
β-lactamase cleaves the β-lactam ring.
Mutations in penicillin-binding proteins.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Amoxicillin

A

Oral or IV
Combined with clavulanate

The molecular structure is similar to penicillin and therefore susceptible to degradation by β-lactamase (β-lactamase sensitive).
Oral bioavailability of amoxicillin is greater than that of ampicillin
D-Ala-D-Ala structural analog. Bind penicillin-binding proteins (transpeptidases).
Block transpeptidase cross-linking of peptidoglycan in cell wall.
Activate autolytic enzymes.

Clinical use:
Broader spectrum of activity than penicillin (extended-spectrum penicillin)
Gram-positive aerobes
Gram-negative rods (not effective against Enterobacter spp.)
Prophylaxis against encapsulated bacteria in asplenic patients and before dental procedures in patients with high risk for endocarditis
Otitis media and sinusitis caused by S. pneumonia, Moraxella and H influenzae

Most effective against:
H. pylori, H. influenzae, Enterococci, E. coli, Listeria monocytogenes, Proteus mirabilis, Salmonella, Shigella, Spirochetes (Lyme disease in pregnant and children)

Adverse effects:
Diarrhea
Pseudomembranous colitis 
Hypersensitivity reactions
Drug-induced rash (in patients with mononucleosis)  
Possibly acute interstitial nephritis

Mechanisms of resistance:
Cleavage of the β-lactam ring by penicillinases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Ampicillin

A

IV or IM form
With or without sulbactam

The molecular structure is similar to penicillin and therefore susceptible to degradation by β-lactamase (β-lactamase sensitive).
Oral bioavailability of ampicillin is lesser than that of amoxicillin
D-Ala-D-Ala structural analog. Bind penicillin-binding proteins (transpeptidases).
Block transpeptidase cross-linking of peptidoglycan in cell wall.
Activate autolytic enzymes.

Clinical use:
Broader spectrum of activity than penicillin (extended-spectrum penicillin)
Gram-positive aerobes
Gram-negative rods (not effective against Enterobacter spp.)
IV is good for aspiration pneumonia
Otitis media and sinusitis caused by S. pneumonia, Moraxella and H influenzae

Most effective against:
H. pylori, H. influenzae, Enterococci, E. coli, Listeria monocytogenes, Proteus mirabilis, Salmonella, Shigella, Spirochetes (Lyme disease in pregnant and children)

Adverse effects:
Diarrhea
Pseudomembranous colitis 
Hypersensitivity reactions
Drug-induced rash (in patients with mononucleosis)  
Possibly acute interstitial nephritis

Mechanisms of resistance:
Cleavage of the β-lactam ring by penicillinases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Dicloxacillin

A

Mechanism:
Same as penicillin.
D-Ala-D-Ala structural analog. Bind penicillin-binding proteins (transpeptidases). Block transpeptidase cross-linking of peptidoglycan in cell wall.
Activate autolytic enzymes.
Penicillinase resistant because bulky R group (e.g., isoxazolyl), which prevent bacterial β-lactamase from hydrolyzing the β-lactam ring

Clinical Use:
Narrow spectrum
Gram-positive aerobes, especially S. aureus (non-MRSA)
Penicillinase-resistant penicillins are not effective against Streptococcus viridans, Enterococci, or Listeria.
IV for systemic S aureus endocarditis and osteomelitis

Adverse Effect:
Interstitial nephritis (esp. associated with methicillin use)
Hypersensitivity reactions

Mechanism of Resistance:
Alteration of PBP binding site → reduced affinity → pathogen is not bound or inactivated by β-lactam (an altered PBP target site is one of the main virulence factors of MRSA)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Nafcillin

A

Mechanism:
Same as penicillin.
D-Ala-D-Ala structural analog. Bind penicillin-binding proteins (transpeptidases). Block transpeptidase cross-linking of peptidoglycan in cell wall.
Activate autolytic enzymes.
Penicillinase resistant because bulky R group (e.g., isoxazolyl), which prevent bacterial β-lactamase from hydrolyzing the β-lactam ring

Clinical Use:
Narrow spectrum
Gram-positive aerobes, especially S. aureus (non-MRSA)
Penicillinase-resistant penicillins are not effective against Streptococcus viridans, Enterococci, or Listeria.
IV for systemic S aureus endocarditis and osteomelitis

Adverse Effect:
Interstitial nephritis (esp. associated with methicillin use)
Hypersensitivity reactions

Mechanism of Resistance:
Alteration of PBP binding site → reduced affinity → pathogen is not bound or inactivated by β-lactam (an altered PBP target site is one of the main virulence factors of MRSA)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Oxacillin

A

Mechanism:
Same as penicillin.
D-Ala-D-Ala structural analog. Bind penicillin-binding proteins (transpeptidases). Block transpeptidase cross-linking of peptidoglycan in cell wall.
Activate autolytic enzymes.
Penicillinase resistant because bulky R group (e.g., isoxazolyl), which prevent bacterial β-lactamase from hydrolyzing the β-lactam ring

Clinical Use:
Narrow spectrum
Gram-positive aerobes, especially S. aureus (non-MRSA)
Penicillinase-resistant penicillins are not effective against Streptococcus viridans, Enterococci, or Listeria.
IV for systemic S aureus endocarditis and osteomelitis

Adverse Effect:
Interstitial nephritis (esp. associated with methicillin use)
Hypersensitivity reactions

Mechanism of Resistance:
Alteration of PBP binding site → reduced affinity → pathogen is not bound or inactivated by β-lactam (an altered PBP target site is one of the main virulence factors of MRSA)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Floxacillin

A

Mechanism:
Same as penicillin.
D-Ala-D-Ala structural analog. Bind penicillin-binding proteins (transpeptidases). Block transpeptidase cross-linking of peptidoglycan in cell wall.
Activate autolytic enzymes.
Penicillinase resistant because bulky R group (e.g., isoxazolyl), which prevent bacterial β-lactamase from hydrolyzing the β-lactam ring

Clinical Use:
Narrow spectrum
Gram-positive aerobes, especially S. aureus (non-MRSA)
Penicillinase-resistant penicillins are not effective against Streptococcus viridans, Enterococci, or Listeria.
IV for systemic S aureus endocarditis and osteomelitis

Adverse Effect:
Interstitial nephritis (esp. associated with methicillin use)
Hypersensitivity reactions

Mechanism of Resistance:
Alteration of PBP binding site → reduced affinity → pathogen is not bound or inactivated by β-lactam (an altered PBP target site is one of the main virulence factors of MRSA)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Methicillin

A

NO LONGER USED DUE TO HIGH ADVERSE EFFECTS

Mechanism:
Same as penicillin.
D-Ala-D-Ala structural analog. Bind penicillin-binding proteins (transpeptidases). Block transpeptidase cross-linking of peptidoglycan in cell wall.
Activate autolytic enzymes.
Penicillinase resistant because bulky R group (e.g., isoxazolyl), which prevent bacterial β-lactamase from hydrolyzing the β-lactam ring

Clinical Use:
Narrow spectrum
Gram-positive aerobes, especially S. aureus (non-MRSA)
Penicillinase-resistant penicillins are not effective against Streptococcus viridans, Enterococci, or Listeria.
IV for systemic S aureus endocarditis and osteomelitis

Adverse Effect:
Interstitial nephritis (esp. associated with methicillin use)
Hypersensitivity reactions

Mechanism of Resistance:
Alteration of PBP binding site → reduced affinity → pathogen is not bound or inactivated by β-lactam (an altered PBP target site is one of the main virulence factors of MRSA)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Mention Aminopenicillins (Penicillinase-Sensitive Penicillins)

A

Oral or IV: amoxicillin (combined with clavulanate )

IV or IM: ampicillin (with or without sulbactam)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Mention Antipseudomonal Penicillins

A

IV piperacillin (combined with tazobactam)
IV mezlocillin
IV ticarcillin
IV carbenicillin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Mention Penicillinase-Resistant Penicillins

A
Nafcillin
Dicloxacillin
Oxacillin
Floxacillin
Methicillin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

Piperacillin

A

Antipseudomonal penicillins
IV form
Combined with tazobactam

Mechanism:
D-Ala-D-Ala structural analog.
Bind penicillin-binding proteins (transpeptidases). Block transpeptidase cross-linking of peptidoglycan in cell wall.
Activate autolytic enzymes.
Extended spectrum.
Penicillinase sensitive; use with β-lactamase inhibitors.

Clinical Use:
Pseudomonas spp. and gram ⊝ rods.
Also effective against anaerobes (e.g., Bacteroides fragilis)
Gram-positive aerobes (not effective against S. viridans)

Adverse Reaction:
Hypersensitivity reactions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Mezlocillin

A

Antipseudomonal penicillins
IV form

Mechanism:
D-Ala-D-Ala structural analog.
Bind penicillin-binding proteins (transpeptidases). Block transpeptidase cross-linking of peptidoglycan in cell wall.
Activate autolytic enzymes.
Extended spectrum.
Penicillinase sensitive; use with β-lactamase inhibitors.

Clinical Use:
Pseudomonas spp. and gram ⊝ rods.
Also effective against anaerobes (e.g., Bacteroides fragilis)
Gram-positive aerobes (not effective against S. viridans)

Adverse Reaction:
Hypersensitivity reactions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Ticarcillin

A

Antipseudomonal penicillins
IV form

Mechanism:
D-Ala-D-Ala structural analog.
Bind penicillin-binding proteins (transpeptidases). Block transpeptidase cross-linking of peptidoglycan in cell wall.
Activate autolytic enzymes.
Extended spectrum.
Penicillinase sensitive; use with β-lactamase inhibitors.

Clinical Use:
Pseudomonas spp. and gram ⊝ rods.
Also effective against anaerobes (e.g., Bacteroides fragilis)
Gram-positive aerobes (not effective against S. viridans)

Adverse Reaction:
Hypersensitivity reactions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Carbenicillin

A

Antipseudomonal penicillins
IV form

Mechanism:
D-Ala-D-Ala structural analog.
Bind penicillin-binding proteins (transpeptidases). Block transpeptidase cross-linking of peptidoglycan in cell wall.
Activate autolytic enzymes.
Extended spectrum.
Penicillinase sensitive; use with β-lactamase inhibitors.

Clinical Use:
Pseudomonas spp. and gram ⊝ rods.
Also effective against anaerobes (e.g., Bacteroides fragilis)
Gram-positive aerobes (not effective against S. viridans)

Adverse Reaction:
Hypersensitivity reactions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

Mention Streptogramins

A

Quinupristin

Dalfopristin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

Quinupristin

A

Streptogramin

Synergistic effect with Dalfopristin
Route of elimination → biliary and renal

Mechanism:
Inhibits the late phase of protein synthesis
Binds to 50S subunit → elongation of polypeptide is prevented → release of incomplete chains
Bacteriostatic when used separately, but bactericidal when used in combination with Dalfopristin

Clinical use:
Skin and skin structure infection with:
-S. aureus (methicillin-susceptible and resistant)
-S. pyogenes
-Vancomycin-resistant E. faecium (VRE)
-Not effective against E. faecalis
Adverse effects:
Inhibits CYP3A4 
Nausea, vomiting
Diarrhea
Headache
Arthralgia, myalgia
Thrombophlebitis
Rash, pruritus
Pseudomembranous colitis

Mechanisms of resistance:
Modification of bacterial ribosome binding site
Enzyme-mediated methylation (cross resistance with clindamycin and macrolides)
Efflux pumps

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

Dalfopristin

A

Streptogramin

Synergistic effect with Quinupristin
Route of elimination → biliary and renal

Mechanism:
Inhibits the early phase of protein synthesis
Binds to 23S portion of 50S subunit → change of conformation → enhanced binding of quinupristin
Inhibits peptidyl transferase
Bacteriostatic when used separately, but bactericidal when used in combination with Quinupristin

Clinical use:
Skin and skin structure infection with:
-S. aureus (methicillin-susceptible and resistant)
-S. pyogenes
-Vancomycin-resistant E. faecium (VRE)
-Not effective against E. faecalis
Adverse effects:
Inhibits CYP3A4 
Nausea, vomiting
Diarrhea
Headache
Arthralgia, myalgia
Thrombophlebitis
Rash, pruritus
Pseudomembranous colitis

Mechanisms of resistance:
Modification of bacterial ribosome binding site
Enzyme-mediated methylation (crossresistance with clindamycin and macrolides)
Efflux pumps

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

Mention 1st generation cephalosporins

A

Oral → cephalexin

IV, IM → cefazolin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Mention 2nd generation cephalosporins

A
Cefaclor
Cefuroxime
Cefoxitin
Cefotetan
Cefprozil
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Mention 3rd generation cephalosporins

A
Cefixime
Cefpodoxime
Cefotaxime
Ceftazidime
Ceftriaxone
Cefdinir
Cefoperazone
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

Mention 4th generation cephalosporins

A

IV → cefepime

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Mention 5th generation cephalosporins

A

IV → ceftaroline

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

Cephalexin

A

1st generation cephalosporin

Mechanism:
β-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases.
Bactericidal.

Clinical Use:
Gram ⊕ cocci, Proteus mirabilis, E coli, Klebsiella pneumoniae.
1st generation—PEcK.

Organisms typically not covered by 1st–4th generation cephalosporins are LAME: Listeria, Atypicals (Chlamydia, Mycoplasma), MRSA, and Enterococci.

Adverse Effects:
Hypersensitivity reactions, autoimmune hemolytic anemia, disulfiram-like reaction, vitamin K deficiency which increase the risk of bleeding. Low rate of cross- reactivity even in penicillin-allergic patients.
Increase nephrotoxicity of aminoglycosides.
Neurotoxicity (can lower seizure threshold)

Mechanism of resistance:
Inactivated by cephalosporinases (a type of β-lactamase).
Structural change in penicillin-binding proteins (transpeptidases).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Cefazolin

A

1st generation cephalosporin

Mechanism:
β-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases.
Bactericidal.

Clinical Use:
Gram ⊕ cocci, Proteus mirabilis, E coli, Klebsiella pneumoniae.
Perioperative wound infection prophylaxis (covers S. aureus)
1st generation—PEcK.

Organisms typically not covered by 1st–4th generation cephalosporins are LAME: Listeria, Atypicals (Chlamydia, Mycoplasma), MRSA, and Enterococci.

Adverse Effects:
Hypersensitivity reactions, autoimmune hemolytic anemia, disulfiram-like reaction, vitamin K deficiency which increase the risk of bleeding. Low rate of cross- reactivity even in penicillin-allergic patients.
Increase nephrotoxicity of aminoglycosides.
Neurotoxicity (can lower seizure threshold)

Mechanism of resistance:
Inactivated by cephalosporinases (a type of β-lactamase).
Structural change in penicillin-binding proteins (transpeptidases).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

Cefaclor

A

2nd generation cephalosporin
Oral form

Mechanism:
β-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases.
Bactericidal.

Clinical Use:
Gram ⊕ cocci, Proteus mirabilis, E coli, Klebsiella pneumoniae, H. influenzae
Klebsiella aerogenes (previously known as Enterobacter aerogenes), Neisseria, Serratia marcescens
2nd generation—HENS PEcK.

Organisms typically not covered by 1st–4th generation cephalosporins are LAME: Listeria, Atypicals (Chlamydia, Mycoplasma), MRSA, and Enterococci.

Adverse Effects:
Hypersensitivity reactions, autoimmune hemolytic anemia, disulfiram-like reaction, vitamin K deficiency which increase the risk of bleeding. Low rate of cross- reactivity even in penicillin-allergic patients.
Increase nephrotoxicity of aminoglycosides.
Neurotoxicity (can lower seizure threshold)

Mechanism of resistance:
Inactivated by cephalosporinases (a type of β-lactamase).
Structural change in penicillin-binding proteins (transpeptidases).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

Cefuroxime

A

2nd generation cephalosporin
Oral form

Mechanism:
β-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases.
Bactericidal.

Clinical Use:
Gram ⊕ cocci, Proteus mirabilis, E coli, Klebsiella pneumoniae, H. influenzae
Klebsiella aerogenes (previously known as Enterobacter aerogenes), Neisseria, Serratia marcescens
2nd generation—HENS PEcK.

Organisms typically not covered by 1st–4th generation cephalosporins are LAME: Listeria, Atypicals (Chlamydia, Mycoplasma), MRSA, and Enterococci.

Adverse Effects:
Hypersensitivity reactions, autoimmune hemolytic anemia, disulfiram-like reaction, vitamin K deficiency which increase the risk of bleeding. Low rate of cross- reactivity even in penicillin-allergic patients.
Increase nephrotoxicity of aminoglycosides.
Neurotoxicity (can lower seizure threshold)

Mechanism of resistance:
Inactivated by cephalosporinases (a type of β-lactamase).
Structural change in penicillin-binding proteins (transpeptidases).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

Cefoxitin

A

2nd generation cephalosporin
IV form

Mechanism:
β-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases.
Bactericidal.

Clinical Use:
Gram ⊕ cocci, Proteus mirabilis, E coli, Klebsiella pneumoniae, H. influenzae
Klebsiella aerogenes (previously known as Enterobacter aerogenes), Neisseria, Serratia marcescens
2nd generation—HENS PEcK.

Organisms typically not covered by 1st–4th generation cephalosporins are LAME: Listeria, Atypicals (Chlamydia, Mycoplasma), MRSA, and Enterococci.

Adverse Effects:
Hypersensitivity reactions, autoimmune hemolytic anemia, disulfiram-like reaction, vitamin K deficiency which increase the risk of bleeding. Low rate of cross- reactivity even in penicillin-allergic patients.
Increase nephrotoxicity of aminoglycosides.
Neurotoxicity (can lower seizure threshold)

Mechanism of resistance:
Inactivated by cephalosporinases (a type of β-lactamase).
Structural change in penicillin-binding proteins (transpeptidases).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

Cefotetan

A

2nd generation cephalosporin
IV form

Mechanism:
β-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases.
Bactericidal.

Clinical Use:
Gram ⊕ cocci, Proteus mirabilis, E coli, Klebsiella pneumoniae, H. influenzae
Klebsiella aerogenes (previously known as Enterobacter aerogenes), Neisseria, Serratia marcescens
2nd generation—HENS PEcK.

Organisms typically not covered by 1st–4th generation cephalosporins are LAME: Listeria, Atypicals (Chlamydia, Mycoplasma), MRSA, and Enterococci.

Adverse Effects:
Hypersensitivity reactions, autoimmune hemolytic anemia, disulfiram-like reaction, vitamin K deficiency which increase the risk of bleeding. Low rate of cross- reactivity even in penicillin-allergic patients.
Increase nephrotoxicity of aminoglycosides.
Neurotoxicity (can lower seizure threshold)

Mechanism of resistance:
Inactivated by cephalosporinases (a type of β-lactamase).
Structural change in penicillin-binding proteins (transpeptidases).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

Cefprozil

A

2nd generation cephalosporin

Mechanism:
β-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases.
Bactericidal.

Clinical Use:
Gram ⊕ cocci, Proteus mirabilis, E coli, Klebsiella pneumoniae, H. influenzae
Klebsiella aerogenes (previously known as Enterobacter aerogenes), Neisseria, Serratia marcescens
2nd generation—HENS PEcK.

Organisms typically not covered by 1st–4th generation cephalosporins are LAME: Listeria, Atypicals (Chlamydia, Mycoplasma), MRSA, and Enterococci.

Adverse Effects:
Hypersensitivity reactions, autoimmune hemolytic anemia, disulfiram-like reaction, vitamin K deficiency which increase the risk of bleeding. Low rate of cross- reactivity even in penicillin-allergic patients.
Increase nephrotoxicity of aminoglycosides.
Neurotoxicity (can lower seizure threshold)

Mechanism of resistance:
Inactivated by cephalosporinases (a type of β-lactamase).
Structural change in penicillin-binding proteins (transpeptidases).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

Cefixime

A

3rd generation cephalosporin
Oral form

Mechanism:
β-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases.
Bactericidal.

Clinical Use:
Least active among all cephalosporins against Gram ⊕ cocci
Serious gram ⊝ infections resistant to other β-lactams.
Perioperative wound infection prophylaxis (covers S. aureus)

Organisms typically not covered by 1st–4th generation cephalosporins are LAME: Listeria, Atypicals (Chlamydia, Mycoplasma), MRSA, and Enterococci.

Adverse Effects:
Hypersensitivity reactions, autoimmune hemolytic anemia, disulfiram-like reaction, vitamin K deficiency which increase the risk of bleeding. Low rate of cross- reactivity even in penicillin-allergic patients.
Increase nephrotoxicity of aminoglycosides.
Neurotoxicity (can lower seizure threshold)

Mechanism of resistance:
Inactivated by cephalosporinases (a type of β-lactamase).
Structural change in penicillin-binding proteins (transpeptidases).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

Cefpodoxime

A

3rd generation cephalosporin
Oral form

Mechanism:
β-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases.
Bactericidal.

Clinical Use:
Least active among all cephalosporins against Gram ⊕ cocci
Serious gram ⊝ infections resistant to other β-lactams.
Perioperative wound infection prophylaxis (covers S. aureus)

Organisms typically not covered by 1st–4th generation cephalosporins are LAME: Listeria, Atypicals (Chlamydia, Mycoplasma), MRSA, and Enterococci.

Adverse Effects:
Hypersensitivity reactions, autoimmune hemolytic anemia, disulfiram-like reaction, vitamin K deficiency which increase the risk of bleeding. Low rate of cross- reactivity even in penicillin-allergic patients.
Increase nephrotoxicity of aminoglycosides.
Neurotoxicity (can lower seizure threshold)

Mechanism of resistance:
Inactivated by cephalosporinases (a type of β-lactamase).
Structural change in penicillin-binding proteins (transpeptidases).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

Cefotaxime

A

3rd generation cephalosporin
IV form

Mechanism:
β-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases.
Bactericidal.

Clinical Use:
Least active among all cephalosporins against Gram ⊕ cocci
Serious gram ⊝ infections resistant to other β-lactams.
Perioperative wound infection prophylaxis (covers S. aureus)

Organisms typically not covered by 1st–4th generation cephalosporins are LAME: Listeria, Atypicals (Chlamydia, Mycoplasma), MRSA, and Enterococci.

Adverse Effects:
Hypersensitivity reactions, autoimmune hemolytic anemia, disulfiram-like reaction, vitamin K deficiency which increase the risk of bleeding. Low rate of cross- reactivity even in penicillin-allergic patients.
Increase nephrotoxicity of aminoglycosides.
Neurotoxicity (can lower seizure threshold)

Mechanism of resistance:
Inactivated by cephalosporinases (a type of β-lactamase).
Structural change in penicillin-binding proteins (transpeptidases).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

Ceftazidime

A

3rd generation cephalosporin
IV form

Mechanism:
β-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases.
Bactericidal.

Clinical Use:
Least active among all cephalosporins against Gram ⊕ cocci
Serious gram ⊝ infections resistant to other β-lactams.
Perioperative wound infection prophylaxis (covers S. aureus)
Pseudomonas

Organisms typically not covered by 1st–4th generation cephalosporins are LAME: Listeria, Atypicals (Chlamydia, Mycoplasma), MRSA, and Enterococci.

Adverse Effects:
Hypersensitivity reactions, autoimmune hemolytic anemia, disulfiram-like reaction, vitamin K deficiency which increase the risk of bleeding. Low rate of cross- reactivity even in penicillin-allergic patients.
Increase nephrotoxicity of aminoglycosides.
Neurotoxicity (can lower seizure threshold)

Mechanism of resistance:
Inactivated by cephalosporinases (a type of β-lactamase).
Structural change in penicillin-binding proteins (transpeptidases).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

Ceftriaxone

A

3rd generation cephalosporin
IM and IV form
Excreted in bile

Mechanism:
β-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases.
Bactericidal.

Clinical Use:
Serious gram ⊝ infections resistant to other β-lactams.
Can cross blood-brain barrier.
Perioperative wound infection prophylaxis (covers S. aureus)
Ceftriaxone—meningitis, gonorrhea, disseminated Lyme disease.

Organisms typically not covered by 1st–4th generation cephalosporins are LAME: Listeria, Atypicals (Chlamydia, Mycoplasma), MRSA, and Enterococci.

Adverse Effects:
Hypersensitivity reactions, autoimmune hemolytic anemia, disulfiram-like reaction, vitamin K deficiency which increase the risk of bleeding. Low rate of cross- reactivity even in penicillin-allergic patients.
Increase nephrotoxicity of aminoglycosides.
Neurotoxicity (can lower seizure threshold)

Mechanism of resistance:
Inactivated by cephalosporinases (a type of β-lactamase).
Structural change in penicillin-binding proteins (transpeptidases).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

Cefdinir

A

3rd generation cephalosporin

Mechanism:
β-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases.
Bactericidal.

Clinical Use:
Least active among all cephalosporins against Gram ⊕ cocci
Serious gram ⊝ infections resistant to other β-lactams.
Perioperative wound infection prophylaxis (covers S. aureus)

Organisms typically not covered by 1st–4th generation cephalosporins are LAME: Listeria, Atypicals (Chlamydia, Mycoplasma), MRSA, and Enterococci.

Adverse Effects:
Hypersensitivity reactions, autoimmune hemolytic anemia, disulfiram-like reaction, vitamin K deficiency which increase the risk of bleeding. Low rate of cross- reactivity even in penicillin-allergic patients.
Increase nephrotoxicity of aminoglycosides.
Neurotoxicity (can lower seizure threshold)

Mechanism of resistance:
Inactivated by cephalosporinases (a type of β-lactamase).
Structural change in penicillin-binding proteins (transpeptidases).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

Cefoperazone

A

3rd generation cephalosporin

Mechanism:
β-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases.
Bactericidal.

Clinical Use:
Least active among all cephalosporins against Gram ⊕ cocci
Serious gram ⊝ infections resistant to other β-lactams.
Pseudomonas

Organisms typically not covered by 1st–4th generation cephalosporins are LAME: Listeria, Atypicals (Chlamydia, Mycoplasma), MRSA, and Enterococci.

Adverse Effects:
Hypersensitivity reactions, autoimmune hemolytic anemia, disulfiram-like reaction, vitamin K deficiency which increase the risk of bleeding. Low rate of cross- reactivity even in penicillin-allergic patients.
Increase nephrotoxicity of aminoglycosides.
Neurotoxicity (can lower seizure threshold)

Mechanism of resistance:
Inactivated by cephalosporinases (a type of β-lactamase).
Structural change in penicillin-binding proteins (transpeptidases).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

Cefepime

A

4th generation cephalosporin
IV form

Mechanism:
β-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases.
Bactericidal.

Clinical Use:
Gram ⊝ organisms, with increase activity against Pseudomonas and gram ⊕ organisms.
Penetrate CNS (so can be used in meningitis)
Broad spectrum
Used for severe life-threatening infections (including nosocomial)

Adverse Effects:
Hypersensitivity reactions, autoimmune hemolytic anemia, disulfiram-like reaction, vitamin K deficiency which increase the risk of bleeding. Low rate of cross- reactivity even in penicillin-allergic patients.
Increase nephrotoxicity of aminoglycosides.
Neurotoxicity (can lower seizure threshold)

Mechanism of resistance:
Inactivated by cephalosporinases (a type of β-lactamase).
Structural change in penicillin-binding proteins (transpeptidases).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

Ceftaroline

A

5th generation cephalosporin
IV form

Mechanism:
β-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases.
Bactericidal.

Clinical Use:
Broad gram ⊕ and gram ⊝ organism coverage
Unlike 1st–4th generation cephalosporins, ceftaroline covers Listeria, MRSA, and Enterococcus faecalis— does not cover Pseudomonas and atypicals (Chlamydia, Mycoplasma, Legionella)

Adverse Effects:
Hypersensitivity reactions, autoimmune hemolytic anemia, disulfiram-like reaction, vitamin K deficiency which increase the risk of bleeding. Low rate of cross- reactivity even in penicillin-allergic patients.
Increase nephrotoxicity of aminoglycosides.
Neurotoxicity (can lower seizure threshold)

Mechanism of resistance:
Inactivated by cephalosporinases (a type of β-lactamase).
Structural change in penicillin-binding proteins (transpeptidases).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

β-lactamase Inhibitors

A

Clavulanate (combined with amoxicillin)
Avibactam (combined with ceftazidime)
Sulbactam (combined with ampicillin)
Tazobactam (combined with piperacillin)

Prevent the destruction of β-lactam antibiotics by β-lactamases and increase the spectrum of the antibiotic activity.
Can be coadministered with β-lactamase-sensitive penicillins in order to treat β-lactamase-producing organisms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

Aztreonam

A

Monobactam
IV form

Less susceptible to β-lactamases.
Prevents peptidoglycan cross-linking by binding to penicillin- binding protein 3.
Synergistic with aminoglycosides.
No cross-allergenicity with penicillins.

Clinical Use:
Effective against Gram ⊝ rods only, including nosocomial Pseudomonas, H. influenzae, and N. meningitidis
No activity against gram ⊕ rods or anaerobes
Alternative for penicillin-allergic patients (no cross-sensitivity with penicillins) and those with renal insufficiency who cannot tolerate aminoglycosides
Broad-spectrum coverage in combination with vancomycin or clindamycin

Adverse Effects:
Usually nontoxic; occasional GI upset.
Injection reactions
Rash

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

Vancomycin

A

Glycopeptide
Oral or IV form

Mechanism:
Bind terminal D-ala-D-ala of cell-wall precursor peptides → inhibition of cell wall synthesis (peptidoglycan formation) → bacterial death (bactericidal effect against most gram-positive bacteria)
Bacteriostatic against C. difficile
CNS penetration → only when there is increased permeability of the meningeal vessels (i.e., with meningeal inflammation)
Route of elimination → renal (via glomerular filtration)

Clinical Use:
Especially effective against multidrug-resistant organisms
Effective against a wide range of gram-positive bacteria only
MRSA
S. epidermidis
Enterococci (if not vancomycin resistant enterococci)
C. difficile (causing pseudomembranous colitis) → administered orally
Penetrate bone so its useful treatment for MRSA osteomyelitis
First line empiric treatment of endocarditis.

Adverse Effects:
Intravenous administration
Nephrotoxicity
Ototoxicity/vestibular toxicity
Thrombophlebitis
Red man syndrome (anaphylactoid reaction caused by rapid infusion of vancomycin; nonspecific mast cell degranulation → rapid release of histamine. Diffuse flushing of the skin, pruritus mainly of the upper body, muscle spasms and pain in the back and chest, possible hypotension and dyspnea. Can be prevented by slowing the rate of infusion and pretreating with antihistamines)
DRESS syndrome (drug reaction (anticonvulsant [hallopurinol] and sulfonamide) leading to eosinophilia and systemic symptoms)
Neutropenia
Dysgeusia and gastrointestinal side effects (e.g., nausea, vomiting, abdominal pain)
Oral administration → predominantly dysgeusia and gastrointestinal side effects

Mechanisms of Resistance:
Modification of amino acid D-Ala-D-Ala to D-Ala-D-Lac → occurs mainly in Enterococcus (e.g., E. faecium, less in E. faecalis)
Beta-lactamase resistant

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

Ricin

A

Castor oil toxin

Inhibits eukaryotic 60 subunit

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

Aminoglycosides

A

IV or IM gentamicin, amikacin, tobramycin, streptomycin
Oral neomycin, paromomycin
Poor CNS penetration
Route of elimination → renal (via glomerular filtration)

Mechanism:
Bind to 30S subunit of the bacterial ribosome → irreversible inhibition of initiation complex → inhibition of bacterial protein synthesis → cell death (bactericidal effect)
Misreading of mRNA
Blockage of translocation
Synergistic effect when combined with β-lactam antibiotics (β-lactams inhibit cell wall synthesis → facilitated entry of aminoglycoside drugs into the cytoplasm)

Clinical Use:
Severe gram-negative rod infections
Not effective against anaerobes (aminoglycosides require oxygen to be absorbed by cells)
Neomycin, which is not absorbed systemically, is administered orally to prepare the gut for bowel surgery.
Streptomycin is used as a second-line treatment for Mycobacterium tuberculosis and M. avium-intracellulare
Paromomycin is not significantly absorbed from the gastrointestinal tract, it is used as a luminal antibiotic in the treatment of hepatic encephalopathy and as a luminal amebicide (by eradicating intestinal cysts). First line treatment for amebiasis or giardiasis during pregnancy.
Gentamycin is the first line treatment of acute pyelonephritis in case of fluoroquinolone allergy or resistance

Adverse Effects:
Nephrotoxicity (proximal tubular injury and acute tubular necrosis visualized histologically as focal tubular epithelial necrosis, often with extensive granular casts that obstruct the tubular lumen and lead to rupture of the basement membrane)
Ototoxicity and vestibulotoxicity (risk of ototoxicity is higher when used concurrently with loop diuretics) resulting in tinnitus, ataxia, vertigo
Neuromuscular blockade (risk increases in the event of hypomagnesemia or myasthenia gravis, and in patients who use calcium channel blockers)
Teratogenicity

Contraindications:
Myasthenia gravis
Botulism
Pregnancy
Cautious use in patients with renal dysfunction

Mechanisms of resistance:
Inactivation via acetylation, phosphorylation, and/or adenylation by secreted bacterial transferase enzymes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

Paromomycin

A

Aminoglycoside
Oral form
Poor CNS penetration
Route of elimination → renal (via glomerular filtration)

Mechanism:
Bind to 30S subunit of the bacterial ribosome → irreversible inhibition of initiation complex → inhibition of bacterial protein synthesis → cell death (bactericidal effect)
Misreading of mRNA
Blockage of translocation
Synergistic effect when combined with β-lactam antibiotics (β-lactams inhibit cell wall synthesis → facilitated entry of aminoglycoside drugs into the cytoplasm)

Clinical Use:
Because it is not significantly absorbed from the gastrointestinal tract, it is used as a luminal antibiotic in the treatment of hepatic encephalopathy and as a luminal amebicide (by eradicating intestinal cysts).
First line treatment for amebiasis or giardiasis during pregnancy.

Adverse Effects:
Nephrotoxicity (proximal tubular injury and acute tubular necrosis visualized histologically as focal tubular epithelial necrosis, often with extensive granular casts that obstruct the tubular lumen and lead to rupture of the basement membrane)
Ototoxicity and vestibulotoxicity (risk of ototoxicity is higher when used concurrently with loop diuretics) resulting in tinnitus, ataxia, vertigo
Neuromuscular blockade (risk increases in the event of hypomagnesemia or myasthenia gravis, and in patients who use calcium channel blockers)
Teratogenicity

Contraindications:
Myasthenia gravis
Botulism
Pregnancy
Cautious use in patients with renal dysfunction

Mechanisms of resistance:
Inactivation via acetylation, phosphorylation, and/or adenylation by secreted bacterial transferase enzymes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

Gentamicin

A

Aminoglycoside
IV or IM form
Poor CNS penetration
Route of elimination → renal (via glomerular filtration)

Mechanism:
Bind to 30S subunit of the bacterial ribosome → irreversible inhibition of initiation complex → inhibition of bacterial protein synthesis → cell death (bactericidal effect)
Misreading of mRNA
Blockage of translocation
Synergistic effect when combined with β-lactam antibiotics (β-lactams inhibit cell wall synthesis → facilitated entry of aminoglycoside drugs into the cytoplasm)

Clinical Use:
Severe gram-negative rod infections
Not effective against anaerobes (aminoglycosides require oxygen to be absorbed by cells)
Gentamycin is the first line treatment of acute pyelonephritis in case of fluoroquinolone allergy or resistance

Adverse Effects:
Nephrotoxicity (proximal tubular injury and acute tubular necrosis visualized histologically as focal tubular epithelial necrosis, often with extensive granular casts that obstruct the tubular lumen and lead to rupture of the basement membrane)
Ototoxicity and vestibulotoxicity (risk of ototoxicity is higher when used concurrently with loop diuretics) resulting in tinnitus, ataxia, vertigo
Neuromuscular blockade (risk increases in the event of hypomagnesemia or myasthenia gravis, and in patients who use calcium channel blockers)
Teratogenicity

Contraindications:
Myasthenia gravis
Botulism
Pregnancy
Cautious use in patients with renal dysfunction

Mechanisms of resistance:
Inactivation via acetylation, phosphorylation, and/or adenylation by secreted bacterial transferase enzymes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

Amikacin

A

Aminoglycoside
IV or IM form
Poor CNS penetration
Route of elimination → renal (via glomerular filtration)

Mechanism:
Bind to 30S subunit of the bacterial ribosome → irreversible inhibition of initiation complex → inhibition of bacterial protein synthesis → cell death (bactericidal effect)
Misreading of mRNA
Blockage of translocation
Synergistic effect when combined with β-lactam antibiotics (β-lactams inhibit cell wall synthesis → facilitated entry of aminoglycoside drugs into the cytoplasm)

Clinical Use:
Severe gram-negative rod infections
Not effective against anaerobes (aminoglycosides require oxygen to be absorbed by cells)

Adverse Effects:
Nephrotoxicity (proximal tubular injury and acute tubular necrosis visualized histologically as focal tubular epithelial necrosis, often with extensive granular casts that obstruct the tubular lumen and lead to rupture of the basement membrane)
Ototoxicity and vestibulotoxicity (risk of ototoxicity is higher when used concurrently with loop diuretics) resulting in tinnitus, ataxia, vertigo
Neuromuscular blockade (risk increases in the event of hypomagnesemia or myasthenia gravis, and in patients who use calcium channel blockers)
Teratogenicity

Contraindications:
Myasthenia gravis
Botulism
Pregnancy
Cautious use in patients with renal dysfunction

Mechanisms of resistance:
Inactivation via acetylation, phosphorylation, and/or adenylation by secreted bacterial transferase enzymes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

Tobramycin

A

Aminoglycoside
IV or IM form
Poor CNS penetration
Route of elimination → renal (via glomerular filtration)

Mechanism:
Bind to 30S subunit of the bacterial ribosome → irreversible inhibition of initiation complex → inhibition of bacterial protein synthesis → cell death (bactericidal effect)
Misreading of mRNA
Blockage of translocation
Synergistic effect when combined with β-lactam antibiotics (β-lactams inhibit cell wall synthesis → facilitated entry of aminoglycoside drugs into the cytoplasm)

Clinical Use:
Severe gram-negative rod infections
Not effective against anaerobes (aminoglycosides require oxygen to be absorbed by cells)

Adverse Effects:
Nephrotoxicity (proximal tubular injury and acute tubular necrosis visualized histologically as focal tubular epithelial necrosis, often with extensive granular casts that obstruct the tubular lumen and lead to rupture of the basement membrane)
Ototoxicity and vestibulotoxicity (risk of ototoxicity is higher when used concurrently with loop diuretics) resulting in tinnitus, ataxia, vertigo
Neuromuscular blockade (risk increases in the event of hypomagnesemia or myasthenia gravis, and in patients who use calcium channel blockers)
Teratogenicity

Contraindications:
Myasthenia gravis
Botulism
Pregnancy
Cautious use in patients with renal dysfunction

Mechanisms of resistance:
Inactivation via acetylation, phosphorylation, and/or adenylation by secreted bacterial transferase enzymes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

Streptomycin

A

Aminoglycoside
IV or IM form
Poor CNS penetration
Route of elimination → renal (via glomerular filtration)

Mechanism:
Bind to 30S subunit of the bacterial ribosome → irreversible inhibition of initiation complex → inhibition of bacterial protein synthesis → cell death (bactericidal effect)
Misreading of mRNA
Blockage of translocation
Synergistic effect when combined with β-lactam antibiotics (β-lactams inhibit cell wall synthesis → facilitated entry of aminoglycoside drugs into the cytoplasm)

Clinical Use:
Severe gram-negative rod infections
Not effective against anaerobes (aminoglycosides require oxygen to be absorbed by cells)
Streptomycin is used as a second-line treatment for Mycobacterium tuberculosis and M. avium-intracellulare

Adverse Effects:
Nephrotoxicity (proximal tubular injury and acute tubular necrosis visualized histologically as focal tubular epithelial necrosis, often with extensive granular casts that obstruct the tubular lumen and lead to rupture of the basement membrane)
Ototoxicity and vestibulotoxicity (risk of ototoxicity is higher when used concurrently with loop diuretics) resulting in tinnitus, ataxia, vertigo
Neuromuscular blockade (risk increases in the event of hypomagnesemia or myasthenia gravis, and in patients who use calcium channel blockers)
Teratogenicity

Contraindications:
Myasthenia gravis
Botulism
Pregnancy
Cautious use in patients with renal dysfunction

Mechanisms of resistance:
Inactivation via acetylation, phosphorylation, and/or adenylation by secreted bacterial transferase enzymes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

Neomycin

A

Aminoglycoside
Oral form
Poor CNS penetration
Route of elimination → renal (via glomerular filtration)

Mechanism:
Bind to 30S subunit of the bacterial ribosome → irreversible inhibition of initiation complex → inhibition of bacterial protein synthesis → cell death (bactericidal effect)
Misreading of mRNA
Blockage of translocation
Synergistic effect when combined with β-lactam antibiotics (β-lactams inhibit cell wall synthesis → facilitated entry of aminoglycoside drugs into the cytoplasm)

Clinical Use:
Severe gram-negative rod infections
Not effective against anaerobes (aminoglycosides require oxygen to be absorbed by cells)
Neomycin, which is not absorbed systemically, is administered orally to prepare the gut for bowel surgery.

Adverse Effects:
Nephrotoxicity (proximal tubular injury and acute tubular necrosis visualized histologically as focal tubular epithelial necrosis, often with extensive granular casts that obstruct the tubular lumen and lead to rupture of the basement membrane)
Ototoxicity and vestibulotoxicity (risk of ototoxicity is higher when used concurrently with loop diuretics) resulting in tinnitus, ataxia, vertigo
Neuromuscular blockade (risk increases in the event of hypomagnesemia or myasthenia gravis, and in patients who use calcium channel blockers)
Teratogenicity

Contraindications:
Myasthenia gravis
Botulism
Pregnancy
Cautious use in patients with renal dysfunction

Mechanisms of resistance:
Inactivation via acetylation, phosphorylation, and/or adenylation by secreted bacterial transferase enzymes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

Minocycline

A
Tetracycline
IV and oral form
CNS penetration →  poor
Route of elimination → renal
Oral tetracyclines should not be taken with substances that contain large amounts of Ca2+, Mg2+, or Fe2+ (e.g., milk, antacids, iron supplements, respectively) because divalent cations inhibit the intestinal absorption of tetracyclines.

Mechanism:
Bind 30S subunit → aminoacyl-tRNA is blocked from binding to ribosome acceptor site → inhibition of bacterial protein synthesis (bacteriostatic effect)

Clinical Use:
Bacteria that lack a cell wall (e.g, Mycoplasma pneumoniae, Ureaplasma)
Intracellular bacteria, such as Rickettsia, Chlamydia, or Anaplasma (tetracyclines accumulate intracellularly and are, therefore, effective against intracellular pathogens)
Borrelia burgdorferi
Ehrlichia, Vibrio cholerae, Francisella tularensis

Adverse Effects:
Hepatotoxicity
Deposition in bones and teeth → inhibition of bone growth (in children) and discoloration of teeth
Damage to mucous membranes (e.g., esophagitis, GI upset)
DRESS syndrome
Photosensitivity: drug or metabolite in the skin absorbs UV radiation → photochemical reaction → formation of free radicals → damage to cellular components → inflammation (sunburn-like)
Degraded tetracyclines are associated with Fanconi syndrome.
Pseudotumor cerebri (rarely)

Contraindications:
Children < 8 years of age (except doxycycline)  
Pregnant women (except doxycycline)  
Patients with renal failure (except doxycycline)
Cautious use in patients with hepatic dysfunction 

Mechanisms of Resistance:
Plasmid-encoded transport pumps increase efflux out of the bacterial cell and decrease uptake of tetracyclines.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

Doxycycline

A

Tetracycline
Oral form
CNS penetration: poor
Gastrointestinal elimination (only tetracycline that is not contraindicated in patients with renal failure)
Oral tetracyclines should not be taken with substances that contain large amounts of Ca2+, Mg2+, or Fe2+ (e.g., milk, antacids, iron supplements, respectively) because divalent cations inhibit the intestinal absorption of tetracyclines.

Mechanism:
Bind 30S subunit → aminoacyl-tRNA is blocked from binding to ribosome acceptor site → inhibition of bacterial protein synthesis (bacteriostatic effect)

Clinical Use:
Bacteria that lack a cell wall (e.g, Mycoplasma pneumoniae, Ureaplasma)
Intracellular bacteria, such as Rickettsia, Chlamydia, or Anaplasma (tetracyclines accumulate intracellularly and are, therefore, effective against intracellular pathogens)
Borrelia burgdorferi
Ehrlichia, Vibrio cholerae, Francisella tularensis
Community-acquired MRSA (doxycycline)

Adverse Effects:
Hepatotoxicity
Deposition in bones and teeth → inhibition of bone growth (in children) and discoloration of teeth
Damage to mucous membranes (e.g., esophagitis, GI upset)
Photosensitivity: drug or metabolite in the skin absorbs UV radiation → photochemical reaction → formation of free radicals → damage to cellular components → inflammation (sunburn-like)
Degraded tetracyclines are associated with Fanconi syndrome.
Pseudotumor cerebri (rarely)

Contraindications:
Children < 8 years of age (except doxycycline)  
Pregnant women (except doxycycline)  
Patients with renal failure (except doxycycline)
Cautious use in patients with hepatic dysfunction 

Mechanisms of Resistance:
Plasmid-encoded transport pumps increase efflux out of the bacterial cell and decrease uptake of tetracyclines.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

Chloramphenicol

A

Mechanism:
Bind 50S subunit → blockage of peptidyltransferase → inhibition of bacterial protein synthesis (bacteriostatic effect)
CNS penetration → good
Route of elimination → renal elimination after hepatic metabolism

Clinical Use:
Meningitis (Haemophilus influenzae, Neisseria meningitidis, Streptococcus pneumoniae) and rickettsial diseases (eg, Rocky Mountain spotted fever [Rickettsia rickettsii]).
Limited use due to toxicity but often still used in developing countries because of low cost.

Adverse Effects:
Anemia (dose dependent), aplastic anemia (dose independent; due to bone marrow suppression), gray baby syndrome (in premature infants because they lack liver UDP-glucuronosyltransferase; hyponatremia, cyanosis, vomiting, flaccidity, hypothermia, and shock).
Potent inhibitory effect on cytochrome P450 isoforms CYP2C19 and CYP3A4

Contraindications:
Infancy
Pregnancy
Cautious use in patients with renal and/or hepatic dysfunction

Mechanism of resistance:
Plasmid-encoded acetyltransferase inactivates the drug.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

Clindamycin

A

Mechanism:
Binds to 50S subunit → blockage of peptide translocation (transpeptidation) → inhibition of peptide chain elongation → inhibition of bacterial protein synthesis (bacteriostatic effect)
CNS penetration → poor
Route of elimination → both renal and biliary
Actively transported in macrophages so can have penetration in abscesses

Clinical Use:
Anaerobic infections (eg, Bacteroides spp., Clostridium perfringens) in aspiration pneumonia, lung abscesses, and oral infections.
Also effective against invasive group A streptococcal infection (especially invasive infections)
Partially effective against gram-positive aerobes
Can be used in MRSA infections
Babesia (together with quinine)
Gardnerella vaginalis
Acne
Not effective against Enterococci

Treats anaerobic infections above the diaphragm vs metronidazole (anaerobic infections below diaphragm).

Adverse Effects:
Pseudomembranous colitis (C difficile overgrowth), fever, diarrhea, teratogenicity
Associated with esophagitis due to low pH and irritant effects on the distal esophagus

Contraindications:
In pregnant women during the 1st trimester and breastfeeding women, clindamycin should be used only if benefits outweigh the risks.

Cross-resistance with macrolides

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

Fosfomycin

A

Oral and IV form

Mechanism:
Inhibits enolpyruvate transferase (MurA) → no formation of N-acetylmuramic acid (a component of bacterial cell wall) → inhibition of cell wall synthesis → cell death (bactericidal effect)
Route of elimination → renal

Clinical use:
Women with uncomplicated urinary tract infections (e.g., cystitis due to E. coli or E. faecalis)

Adverse Effects:
Mild electrolyte imbalances (e.g., hypernatremia, hypokalemia)
Diarrhea

Contraindications:
Hypersensitivity

Mechanisms of resistance:
MurA mutations

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

Linezolid

A

Oxazolidinone
Oral form

Mechanism:
Binds to 23S subunit of 50S bacterial ribosome → inhibition of initiation complex formation → inhibition of bacterial protein synthesis (bacteriostatic effect)
Unlike other antibiotics that inhibit later stages of bacterial protein synthesis (e.g., elongation), linezolid inhibits protein synthesis at the very first step of translation.
Nonselective monoamine oxidase inhibition (MAOI)
Bactericidal against streptococci
CNS penetration → good
Route of elimination → both biliary and renal elimination after hepatic metabolism

Clinical Use:
Gram ⊕ species including MRSA and VRE.

Adverse Effects:
Bone marrow suppression (especially thrombocytopenia), peripheral neuropathy, serotonin syndrome (increase risk with SSRIs, MAOIs, SNRIs, bupropion), optic neuropathy (decrease visual acuity, decrease color perception, scotomas), GI upset.

Contraindications:
Concurrent use with MAOIs and selective serotonin reuptake inhibitors (SSRIs)

Mechanism of resistance:
Point mutation of 23S rRNA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

Erythromycin

A
Macrolide
Oral or IV form
CNS penetration → poor
Route of elimination → biliary
All macrolides (except azithromycin) have a short half-life.

Mechanism:
Bind to 23S ribosomal RNA molecule of the 50S subunit → blockage of translocation → inhibition of bacterial protein synthesis (bacteriostatic effect)

Clinical Use:
Atypical pneumonia caused by Mycoplasma pneumonia, Legionella pneumophila, Chlamydophila pneumoniae
Bordetella pertussis
STIs caused by Chlamydia
Gram-positive cocci (especially for the treatment of streptococcal infection in patients who are allergic to penicillin)
Neisseria spp.
Second-line prophylaxis for N. meningitidis
Ureaplasma urealyticum
Erythromycin is used off-label for the treatment of gastroparesis because it increases GI motility.

Adverse Effects:
Increased intestinal motility → GI upset
QT-interval prolongation, arrhythmia
Acute cholestatic hepatitis
Eosinophilia
Rash

Drug Interactions:
Erythromycin enhances the effect of oral anticoagulants (e.g., warfarin).
Erythromycin and clarithromycin increase theophylline serum concentrations due to CYP3A4 inhibition (cytochrome P450 inhibitors)

Contraindications:
Erythromycin estolate and clarithromycin are contraindicated in pregnant women (potentially hazardous to the fetus).

Mechanisms of Resistance:
Methylation of the binding site of 23S rRNA prevents the macrolide from binding to rRNA.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

Clarithromycin

A
Macrolide 
Oral or IV form
CNS penetration → poor
Route of elimination → biliary
All macrolides (except azithromycin) have a short half-life.

Mechanism:
Bind to 23S ribosomal RNA molecule of the 50S subunit → blockage of translocation → inhibition of bacterial protein synthesis (bacteriostatic effect)

Clinical Use:
Atypical pneumonia caused by Mycoplasma pneumonia, Legionella pneumophila, Chlamydophila pneumoniae
Bordetella pertussis
STIs caused by Chlamydia
Gram-positive cocci especially for the treatment of streptococcal infection in patients who are allergic to penicillin)
Neisseria spp.
Second-line prophylaxis for N. meningitidis
Mycobacterium avium (prophylaxis: azithromycin; treatment: azithromycin, clarithromycin)
H. pylori (clarithromycin is the part of triple therapy)
Ureaplasma urealyticum

Adverse Effects:
Increased intestinal motility → GI upset
QT-interval prolongation, arrhythmia
Acute cholestatic hepatitis
Eosinophilia
Rash

Drug Interactions:
Erythromycin and clarithromycin increase theophylline serum concentrations due to CYP3A4 inhibition (cytochrome P450 inhibitors)

Contraindications:
Erythromycin estolate and clarithromycin are contraindicated in pregnant women (potentially hazardous to the fetus) .
Azithromycin and clarithromycin are contraindicated in patients with hepatic failure (erythromycin should be used cautiously).
Cautious use of clarithromycin in patients with renal failure

Mechanisms of Resistance:
Methylation of the binding site of 23S rRNA prevents the macrolide from binding to rRNA.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

Polymyxin B

A

IV or IM form

Mechanism:
Cationic detergent (polypeptides) molecule that binds to phospholipids of the cytoplasmic membrane of gram-negative bacteria → increased membrane permeability → leakage of cell contents → cell death (bactericidal effect)
Binds to and inactivates endotoxins 
CNS penetration → poor
Route of elimination → mostly renal

Clinical Use:
Severe infections caused by multidrug-resistant gram-negative bacteria
Effective only against gram-negative bacteria including P. aeruginosa, K. pneumoniae, E. coli, Acinetobacter baumannii, and Enterobacteriaceae spp.
Proteus, Neisseria, and Serratia spp. are resistant to polymyxins.
Not effective against gram-positive bacteria (because gram-positive bacteria do not have an outer membrane)
Polymyxin B is a component of a triple antibiotic ointment (bacitracin, neomycin, and polymyxin B) used for superficial skin infections
Oral polymyxin B may be used to disinfect the bowel to prevent ICU infections (polymyxins have a poor oral absorption)

Adverse Effects:
Nephrotoxicity, neurotoxicity (e.g., paresthesias, weakness, speech disorders, neuromuscular blockage), respiratory failure.
Urticaria, eosinophilia, and/or anaphylactoid reactions

Contraindications:
Hypersensitivity to polymyxins
Cautious use in patients with renal dysfunction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

Polymyxin E (Colistin)

A

IV or IM form

Mechanism:
Cationic detergent (polypeptides) molecule that binds to phospholipids of the cytoplasmic membrane of gram-negative bacteria → increased membrane permeability → leakage of cell contents → cell death (bactericidal effect)
Binds to and inactivates endotoxins 
CNS penetration → poor
Route of elimination → mostly renal

Clinical Use:
Severe infections caused by multidrug-resistant gram-negative bacteria
Effective only against gram-negative bacteria including P. aeruginosa, K. pneumoniae, E. coli, Acinetobacter baumannii, and Enterobacteriaceae spp.
Proteus, Neisseria, and Serratia spp. are resistant to polymyxins.
Not effective against gram-positive bacteria (because gram-positive bacteria do not have an outer membrane)

Adverse Effects:
Nephrotoxicity, neurotoxicity (e.g., paresthesias, weakness, speech disorders, neuromuscular blockage), respiratory failure.
Urticaria, eosinophilia, and/or anaphylactoid reactions

Contraindications:
Hypersensitivity to polymyxins
Cautious use in patients with renal dysfunction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

Nitrofurantoin

A

Mechanism:
Reduced by bacterial nitroreductases to reactive metabolites → bind to bacterial ribosomes → impaired metabolism, impaired synthesis of protein, DNA, and RNA → cell death (bactericidal effect)
Route of elimination → primarily renal (rapidly excreted after absorption), small amounts in feces

Clinical Use:
Urinary tract pathogens
Gram-positive → Enterococci, Staphylococcus saprophyticus, group B streptococcus, Staphylococcus aureus, Staphylococcus epidermidis
Gram-negative → E. coli, Enterobacter spp., Shigella spp., Salmonella spp., Citrobacter spp, Neisseria spp, Bacteroides spp., Klebsiella spp.
Not effective against Pseudomonas and/or Proteus
Treatment and prophylaxis of acute uncomplicated UTIs (e.g., urethritis, cystitis)
Asymptomatic bacteriuria or symptomatic UTI in pregnant women
Should not be used in (suspected) pyelonephritis because nitrofurantoin does not achieve adequate concentration in renal tissue

Adverse Effects:
Pulmonary fibrosis
Hemolytic anemia in patients with G6PD deficiency
GI upset
Reversible peripheral neuropathy

Contraindications:
Children < 1 month of age
Breastfeeding women
Women at 38–42 weeks’ gestation or during delivery (because of the risk of hemolytic anemia in an infant due to the lack of erythrocyte glutathione peroxidase)
Hepatic dysfunction
Renal dysfunction with a creatinine clearance < 60 mL/min (in renal failure, urinary drug levels do not reach concentrations required to treat UTIs, and the retention of nitrofurantoin in systemic circulation increases the risk of adverse effects.)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

Sulfa Drugs

A

Antibiotics (sulfamethoxazole (SMX), sulfadiazine, sulfisoxazole)
Diuretics (thiazides, furosemide, acetazolamide)
Anti-inflammatory drugs (sulfasalazine, celecoxib)
Sulfonylureas
Probenecid

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

Trimethoprim (TMP)

A

Oral or IV cotrimoxazole (TMP/SMX)

Mechanism:
Inhibition of bacterial folic acid synthesis
Inhibit dihydrofolate reductase (DHFR) (DHFR uses NADPH to reduce dihydrofolic acid to tetrahydrofolic acid (THF); THF can be converted to methylene-THF; methylene-THF is an important cofactor for thymidylate synthetase, which catalyzes the conversion of deoxyuridine monophosphate (dUMP) to deoxythymidine monophosphate (dTMP))
Bacteriostatic but become bactericidal when combined with sulfonamides
The combination of TMP and SMX also prevents antibiotic resistance.
CNS penetration → good
Route of elimination → primarily renal (via tubular secretion) (only a small amount is excreted with bile)

Clinical Use: 
Common indications include UTIs and acute otitis media.
-TMP/SMX
Shigella
Salmonella
Empiric treatment for simple UTI
Prophylaxis and treatment of P. jirovecii
Prophylaxis of toxoplasmosis

Adverse Effects:
Megaloblastic anemia
Leukopenia, granulocytopenia (can be prevented with concomitant folinic acid administration)
In high doses → hyperkalemia, particularly in HIV-positive patients
Artificially increased creatinine (despite unchanged GFR) (similar to the actions of amiloride TMP blocks the epithelial Na+ channel in the DCT and collecting duct, this reduces transepithelial voltage and impairs Na/K exchange, leading to reduced potassium excretion and hyperkalemia. This effect is often magnified elderly population, renal failure and those given other K sparing diuretics, ACE inhibitors and ARBs).

Contraindications:
Children < 2 years of age
Pregnant women
Breastfeeding women
Cautious use in people with hepatic failure, renal failure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

Pyrimethamine

A

Oral sulfadiazine in combination with pyrimethamine

Mechanism:
Inhibition of bacterial folic acid synthesis
Inhibit dihydrofolate reductase (DHFR) (DHFR uses NADPH to reduce dihydrofolic acid to tetrahydrofolic acid (THF); THF can be converted to methylene-THF; methylene-THF is an important cofactor for thymidylate synthetase, which catalyzes the conversion of deoxyuridine monophosphate (dUMP) to deoxythymidine monophosphate (dTMP))
Bacteriostatic but become bactericidal when combined with sulfonamides
The combination of TMP and SMX also prevents antibiotic resistance.
CNS penetration → good
Route of elimination → primarily renal (via tubular secretion) (only a small amount is excreted with bile)

Clinical Use: 
Common indications include UTIs and acute otitis media.
-TMP/SMX
Shigella
Salmonella
Empiric treatment for simple UTI
Prophylaxis and treatment of P. jirovecii
Prophylaxis of toxoplasmosis

Adverse Effects:
Megaloblastic anemia
Leukopenia, granulocytopenia (can be prevented with concomitant folinic acid administration)
In high doses → hyperkalemia, particularly in HIV-positive patients
Artificially increased creatinine (despite unchanged GFR)

Contraindications:
Children < 2 years of age
Pregnant women
Breastfeeding women
Cautious use in people with hepatic failure, renal failure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

Sulfamethoxazole (SMX)

A

Sulfonamide
Oral or IV cotrimoxazole (TMP/SMX)

Mechanism:
Inhibition of bacterial folic acid synthesis
Inhibit dihydropteroate synthase.
Bacteriostatic but become bactericidal when combined with TMP
The combination of TMP and SMX also prevents antibiotic resistance.
CNS penetration → good
Route of elimination → primarily renal (via tubular secretion) (only a small amount is excreted with bile)
Metabolized in the liver via acetylation

Clinical Use: 
Common indications include UTIs and acute otitis media.
-TMP/SMX
Shigella
Salmonella
Empiric treatment for simple UTI
Prophylaxis and treatment of P. jirovecii
Prophylaxis of toxoplasmosis

Adverse Effects:
Fever
Photosensitivity
Sulfonamides hypersensitivity reactions (especially urticaria or hives) (sulfonamides are the second most common antibacterial to cause hypersensitivity reactions, after β-lactams)
Stevens-Johnson syndrome
Triggers hemolytic anemia in G6PD-deficient patients
Aplastic anemia, thrombocytopenia, and pancytopenia
Agranulocytosis
Hyperkalemia
GI upset
Nephrotoxicity (especially acute tubulointerstitial nephritis)
Kernicterus in infancy
Displacement of other drugs (e.g., warfarin) from albumin (SMX has a higher binding affinity to albumin, so it can displace drugs like warfarin from albumin, resulting in increased free plasma concentrations of the displaced drug)
Drug interactions due to CYP450 inhibition

Contraindications:
Children < 2 years of age
Pregnant women
Breastfeeding women
Cautious use in people with hepatic failure, renal failure

Mechanisms of Resistance:
Mutation in bacterial dihydropteroate synthase
Decreased uptake of sulfonamide
Increased para-aminobenzoate (PABA) synthesis (PABA is a substrate of dihydropteroate synthase and a precursor of folic acid)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

Sulfadiazine

A

Sulfonamide
Oral sulfadiazine in combination with pyrimethamine

Mechanism:
Inhibition of bacterial folic acid synthesis
Inhibit dihydropteroate synthase.
CNS penetration → good
Route of elimination → primarily renal (via tubular secretion) (only a small amount is excreted with bile)
Bacteriostatic but become bactericidal when combined with pyrimethamine (sequential block of folate synthesis)

Clinical use:
Common indications include UTIs and acute otitis media.
Shigella
Salmonella
Empiric treatment for simple UTI
Prophylaxis and treatment of P. jirovecii
Prophylaxis of toxoplasmosis

Adverse Effects:
Fever
Photosensitivity
Sulfonamides hypersensitivity reactions (especially urticaria or hives) (sulfonamides are the second most common antibacterial to cause hypersensitivity reactions, after β-lactams)
Stevens-Johnson syndrome
Triggers hemolytic anemia in G6PD-deficient patients
Aplastic anemia, thrombocytopenia, and pancytopenia
Agranulocytosis
Hyperkalemia
GI upset
Nephrotoxicity (especially acute tubulointerstitial nephritis)
Kernicterus in infancy
Displacement of other drugs (e.g., warfarin) from albumin (SMX has a higher binding affinity to albumin, so it can displace drugs like warfarin from albumin, resulting in increased free plasma concentrations of the displaced drug)
Drug interactions due to CYP450 inhibition

Contraindications:
Children < 2 years of age
Pregnant women
Breastfeeding women
Cautious use in people with hepatic failure, renal failure

Mechanisms of Resistance:
Mutation in bacterial dihydropteroate synthase
Decreased uptake of sulfonamide
Increased para-aminobenzoate (PABA) synthesis (PABA is a substrate of dihydropteroate synthase and a precursor of folic acid)

81
Q

Sulfisoxazole

A

Sulfonamide
Oral sulfisoxazole

Mechanism of action
Inhibition of bacterial folic acid synthesis
Inhibit dihydropteroate synthase.
CNS penetration → good
Route of elimination → primarily renal (via tubular secretion) (only a small amount is excreted with bile)

Clinical Use: 
Common indications include UTIs and acute otitis media.
Gram-positive bacteria and gram-negative bacteria
N. meningitidis
Chlamydia trachomatis
Nocardia asteroides
Toxoplasma gondii
Plasmodia spp.

Adverse Effects:
Fever
Photosensitivity
Sulfonamides hypersensitivity reactions (especially urticaria or hives) (sulfonamides are the second most common antibacterial to cause hypersensitivity reactions, after β-lactams)
Stevens-Johnson syndrome
Triggers hemolytic anemia in G6PD-deficient patients
Aplastic anemia, thrombocytopenia, and pancytopenia
Agranulocytosis
Hyperkalemia
GI upset
Nephrotoxicity (especially acute tubulointerstitial nephritis)
Kernicterus in infancy
Displacement of other drugs (e.g., warfarin) from albumin (SMX has a higher binding affinity to albumin, so it can displace drugs like warfarin from albumin, resulting in increased free plasma concentrations of the displaced drug)
Drug interactions due to CYP450 inhibition

Contraindications:
Children < 2 years of age
Pregnant women
Breastfeeding women
Cautious use in people with hepatic failure, renal failure

Mechanisms of Resistance:
Mutation in bacterial dihydropteroate synthase
Decreased uptake of sulfonamide
Increased para-aminobenzoate (PABA) synthesis (PABA is a substrate of dihydropteroate synthase and a precursor of folic acid)

82
Q

Daptomycin

A

Mechanism:
Incorporate K+ channels into the cell membrane of gram-positive bacteria → rapid membrane depolarization → loss of membrane potential → inhibition of synthesis of DNA, RNA, and proteins → cell death (bactericidal effect)
Route of elimination → renal

Clinical Use:
Gram-positive bacteria
S. aureus, especially MRSA
Mainly used in skin and skin structure infections, bacteremia, and endocarditis
Vancomycin-resistant Enterococci (VRE)
Not used in pneumonia (daptomycin is bound and inactivated by surfactant)

Adverse Effects:
Reversible myopathy
Muscle pain after 1 week starting therapy
Rhabdomyolysis (monitor creantinine phosphate (CPK))
Allergic pneumonitis

Contraindications:
Hypersensitivity

Mechanisms of Resistance:
Repulsion of daptomycin molecules due to the change in the bacterial surface charge

83
Q

Dapsone

A

Mechanism:
Competitive antagonist of para-aminobenzoic acid (PABA) for dihydropteroate synthetase → inhibition of dihydrofolic acid synthesis
Bacteriostatic and weakly bactericidal effect
Structurally different from sulfonamides but a similar mechanism of action
Route of elimination → mostly renal

Clinical Use:
M. leprae (lepromatous and tuberculoid leprosy)
P. jiroveci pneumonia (prophylaxis; treatment → used in combination with TMP as an alternative to TMP/SMX)
Dermatitis herpetiformis
Alternative to the combination of sulfadiazine and pyrimethamine for toxoplasmosis
In combination with pyrimethamine as an alternative for chloroquine-resistant malaria

Adverse Effects:
Methemoglobinemia
Triggers hemolytic anemia in patients with G6PD deficiency
Agranulocytosis
GI upset
Peripheral neuropathy

Contraindications:
G6PD deficiency
Cautious use in patients with renal and/or hepatic dysfunction

Mechanism of Resistance:
Mutations of folP1 gene coding for dihydropteroate synthase

84
Q

Mention 1st generation Fluoroquinolone

A

Nalidixic acid (oral)

85
Q

Fluoroquinolones

A

1st generation → nalidixic acid (oral)
2nd generation → norfloxacin, ciprofloxacin, ofloxacin (oral), enoxacin (oral or IV)
3rd generation → levofloxacin (oral or IV)
4th generation → moxifloxacin, gemifloxacin, gatifloxacin (oral)
Levofloxacin, moxifloxacin, and gemifloxacin are respiratory fluoroquinolones.

Mechanism:
Inhibition of prokaryotic topoisomerase II (DNA gyrase) and topoisomerase IV → DNA supercoiling → formation of double-stranded breaks → inhibition of DNA replication and transcription (bactericidal effect)
CNS penetration → good
Route of elimination → primarily renal (via glomerular filtration and tubular secretion)
Moxifloxacin undergoes biliary excretion.
Absorption is reduced when coadministered with polyvalent cations (e.g., magnesium, calcium, iron).

Clinical Use:
-Norfloxacin, ciprofloxacin, and ofloxacin
Gram-negative rods causing urinary and gastrointestinal infections
Some gram-positive pathogens
Genitourinary infections caused by Neisseria gonorrhoeae, Chlamydia trachomatis, and/or Ureaplasma urealyticum
Ciprofloxacin → Pseudomonas aeruginosa (e.g., malignant otitis externa)

-Levofloxacin, moxifloxacin, and gemifloxacin:
Atypical bacteria (e.g., Legionella spp., Mycoplasma spp., Chlamydophila pneumoniae)
Also effective against anaerobes
Gemifloxacin is highly potent against penicillin-resistant pneumococci.
Moxifloxacin → 2nd-line treatment of tuberculosis in patients who cannot tolerate antitubercular drugs and in multidrug resistant tuberculosis
Moxifloxacin poor antipseudomonal activity

Adverse Effects:
GI upset
Neurological symptoms (mild headache, dizziness, mood changes, peripheral neuropathy, can lower seizure threshold (increased risk in patients taking NSAIDs [NSAIDs increase the displacement of GABA from its receptor caused by quinolones] and those with a previous history of epilepsy))
Hyperglycemia/hypoglycemia (especially with gatifloxacin, which has been removed from the US market for this reason)
QT prolongation
Photosensitivity
Skin rash
Superinfection (most commonly with gram-positive pathogens)
Potentially life-threatening exacerbations in patients with myasthenia gravis
In children → potential damage to growing cartilage → reversible arthropathy
Muscle ache, leg cramps, tendinitis, tendon rupture, especially of the Achilles tendon (the risk of tendon rupture is higher for individuals over 60 years of age and for individuals on steroid therapy)
Increased risk for drug interactions as ciprofloxacin inhibits cytochrome P450

Contraindications:
Children and teenagers < 18 years of age (the only exception is ciprofloxacin which can be used in this cohort for complicated UTI including pyelonephritis and anthrax)
Patients > 60 years of age and those taking cortisol
Pregnant women
Breastfeeding women
Epilepsy, stroke, CNS lesions/inflammation
QT prolongation
Myasthenia gravis (potentially life-threatening exacerbations)
Cautious use in patients with renal failure, hepatic failure
Antacid use (aluminum- and magnesium-containing antacids decrease the absorption of fluoroquinolones)
Known aortic aneurysm or increased risk of aneurysms (e.g., Marfan syndrome, Ehlers-Danlos syndrome, advanced age, peripheral atherosclerotic disease, hypertension)

Mechanism of Resistance:
Chromosome-encoded mutation (altered amino acid composition) in DNA gyrase and topoisomerase IV enzymes
Altered cell wall permeability
Plasmid-encoded mutations in efflux pump proteins

86
Q

Mention 3rd generation Fluoroquinolone

A

Levofloxacin (oral or IV)

87
Q

Mention 4th generation Fluoroquinolone

A

Moxifloxacin
Gemifloxacin
Gatifloxacin

88
Q

Mention Respiratory Fluoroquinolones

A

Levofloxacin
Moxifloxacin
Gemifloxacin

89
Q

Nalidixic acid

A

1st generation fluoroquinolone

Mechanism:
Inhibition of prokaryotic topoisomerase II (DNA gyrase) and topoisomerase IV→ DNA supercoiling → formation of double-stranded breaks → inhibition of DNA replication and transcription (bactericidal effect)
CNS penetration → good
Route of elimination → primarily renal (via glomerular filtration and tubular secretion)
Absorption is reduced when coadministered with polyvalent cations (e.g., magnesium, calcium, iron).

Clinical Use:
-Norfloxacin, ciprofloxacin, and ofloxacin
Gram-negative rods causing urinary and gastrointestinal infections
Some gram-positive pathogens
Genitourinary infections caused by Neisseria gonorrhoeae, Chlamydia trachomatis, and/or Ureaplasma urealyticum
Ciprofloxacin → Pseudomonas aeruginosa (e.g., malignant otitis externa)

-Levofloxacin, moxifloxacin, and gemifloxacin:
Atypical bacteria (e.g., Legionella spp., Mycoplasma spp., Chlamydophila pneumoniae)
Also effective against anaerobes
Gemifloxacin is highly potent against penicillin-resistant pneumococci.
Moxifloxacin → 2nd-line treatment of tuberculosis in patients who cannot tolerate antitubercular drugs and in multidrug resistant tuberculosis

Adverse Effects:
GI upset
Neurological symptoms (mild headache, dizziness, mood changes, peripheral neuropathy, can lower seizure threshold (increased risk in patients taking NSAIDs [NSAIDs increase the displacement of GABA from its receptor caused by quinolones] and those with a previous history of epilepsy))
Hyperglycemia/hypoglycemia (especially with gatifloxacin, which has been removed from the US market for this reason)
QT prolongation
Photosensitivity
Skin rash
Superinfection (most commonly with gram-positive pathogens)
Potentially life-threatening exacerbations in patients with myasthenia gravis
In children → potential damage to growing cartilage → reversible arthropathy
Muscle ache, leg cramps, tendinitis, tendon rupture, especially of the Achilles tendon (the risk of tendon rupture is higher for individuals over 60 years of age and for individuals on steroid therapy)
Increased risk for drug interactions as ciprofloxacin inhibits cytochrome P450

Contraindications:
Children and teenagers < 18 years of age (the only exception is ciprofloxacin which can be used in this cohort for complicated UTI including pyelonephritis and anthrax)
Patients > 60 years of age and those taking cortisol
Pregnant women
Breastfeeding women
Epilepsy, stroke, CNS lesions/inflammation
QT prolongation
Myasthenia gravis (potentially life-threatening exacerbations)
Cautious use in patients with renal failure, hepatic failure
Antacid use (aluminum- and magnesium-containing antacids decrease the absorption of fluoroquinolones)
Known aortic aneurysm or increased risk of aneurysms (e.g., Marfan syndrome, Ehlers-Danlos syndrome, advanced age, peripheral atherosclerotic disease, hypertension)

Mechanism of Resistance:
Chromosome-encoded mutation in DNA gyrase and topoisomerase IV enzymes
Altered cell wall permeability
Plasmid-encoded mutations in efflux pump proteins

90
Q

Norfloxacin

A

2nd generation fluoroquinolone
Oral form
CNS penetration → good
Route of elimination → primarily renal (via glomerular filtration and tubular secretion)
Absorption is reduced when coadministered with polyvalent cations (e.g., magnesium, calcium, iron).

Mechanism:
Inhibition of prokaryotic topoisomerase II (DNA gyrase) and topoisomerase IV→ DNA supercoiling → formation of double-stranded breaks → inhibition of DNA replication and transcription (bactericidal effect)

Clinical Use:
Gram-negative rods causing urinary and gastrointestinal infections
Some gram-positive pathogens
Genitourinary infections caused by Neisseria gonorrhoeae, Chlamydia trachomatis, and/or Ureaplasma urealyticum

Adverse Effects:
GI upset
Neurological symptoms (mild headache, dizziness, mood changes, peripheral neuropathy, can lower seizure threshold (increased risk in patients taking NSAIDs [NSAIDs increase the displacement of GABA from its receptor caused by quinolones] and those with a previous history of epilepsy))
Hyperglycemia/hypoglycemia (especially with gatifloxacin, which has been removed from the US market for this reason)
QT prolongation
Photosensitivity
Skin rash
Superinfection (most commonly with gram-positive pathogens)
Potentially life-threatening exacerbations in patients with myasthenia gravis
In children → potential damage to growing cartilage → reversible arthropathy
Muscle ache, leg cramps, tendinitis, tendon rupture, especially of the Achilles tendon (the risk of tendon rupture is higher for individuals over 60 years of age and for individuals on steroid therapy)

Contraindications:
Children and teenagers < 18 years of age (the only exception is ciprofloxacin which can be used in this cohort for complicated UTI including pyelonephritis and anthrax)
Patients > 60 years of age and those taking cortisol
Pregnant women
Breastfeeding women
Epilepsy, stroke, CNS lesions/inflammation
QT prolongation
Myasthenia gravis (potentially life-threatening exacerbations)
Cautious use in patients with renal failure, hepatic failure
Antacid use (aluminum- and magnesium-containing antacids decrease the absorption of fluoroquinolones)
Known aortic aneurysm or increased risk of aneurysms (e.g., Marfan syndrome, Ehlers-Danlos syndrome, advanced age, peripheral atherosclerotic disease, hypertension)

Mechanism of Resistance:
Chromosome-encoded mutation in DNA gyrase and topoisomerase IV enzymes
Altered cell wall permeability
Plasmid-encoded mutations in efflux pump proteins

91
Q

Ciprofloxacin

A

2nd generation fluoroquinolone
Oral form

Mechanism:
Inhibition of prokaryotic topoisomerase II (DNA gyrase) and topoisomerase IV→ DNA supercoiling → formation of double-stranded breaks → inhibition of DNA replication and transcription (bactericidal effect)
CNS penetration → good
Route of elimination → primarily renal (via glomerular filtration and tubular secretion)
Absorption is reduced when coadministered with polyvalent cations (e.g., magnesium, calcium, iron).

Clinical Use:
Gram-negative rods causing urinary and gastrointestinal infections
Some gram-positive pathogens
Genitourinary infections caused by Neisseria gonorrhoeae, Chlamydia trachomatis, and/or Ureaplasma urealyticum
Pseudomonas aeruginosa (e.g., malignant otitis externa)

Adverse Effects:
GI upset
Neurological symptoms (mild headache, dizziness, mood changes, peripheral neuropathy, can lower seizure threshold (increased risk in patients taking NSAIDs [NSAIDs increase the displacement of GABA from its receptor caused by quinolones] and those with a previous history of epilepsy))
Hyperglycemia/hypoglycemia (especially with gatifloxacin, which has been removed from the US market for this reason)
QT prolongation
Photosensitivity
Skin rash
Superinfection (most commonly with gram-positive pathogens)
Potentially life-threatening exacerbations in patients with myasthenia gravis
In children → potential damage to growing cartilage → reversible arthropathy
Muscle ache, leg cramps, tendinitis, tendon rupture, especially of the Achilles tendon (the risk of tendon rupture is higher for individuals over 60 years of age and for individuals on steroid therapy)
Increased risk for drug interactions as ciprofloxacin inhibits cytochrome P450

Contraindications:
Children and teenagers < 18 years of age (the only exception is ciprofloxacin which can be used in this cohort for complicated UTI including pyelonephritis and anthrax)
Patients > 60 years of age and those taking cortisol
Pregnant women
Breastfeeding women
Epilepsy, stroke, CNS lesions/inflammation
QT prolongation
Myasthenia gravis (potentially life-threatening exacerbations)
Cautious use in patients with renal failure, hepatic failure
Antacid use (aluminum- and magnesium-containing antacids decrease the absorption of fluoroquinolones)
Known aortic aneurysm or increased risk of aneurysms (e.g., Marfan syndrome, Ehlers-Danlos syndrome, advanced age, peripheral atherosclerotic disease, hypertension)

Mechanism of Resistance:
Chromosome-encoded mutation in DNA gyrase and topoisomerase IV enzymes
Altered cell wall permeability
Plasmid-encoded mutations in efflux pump proteins

92
Q

Ofloxacin

A

2nd generation fluoroquinolone
Oral form

Mechanism:
Inhibition of prokaryotic topoisomerase II (DNA gyrase) and topoisomerase IV→ DNA supercoiling → formation of double-stranded breaks → inhibition of DNA replication and transcription (bactericidal effect)
CNS penetration → good
Route of elimination → primarily renal (via glomerular filtration and tubular secretion)
Absorption is reduced when coadministered with polyvalent cations (e.g., magnesium, calcium, iron).

Clinical Use:
Gram-negative rods causing urinary and gastrointestinal infections
Some gram-positive pathogens
Genitourinary infections caused by Neisseria gonorrhoeae, Chlamydia trachomatis, and/or Ureaplasma urealyticum

Adverse Effects:
GI upset
Neurological symptoms (mild headache, dizziness, mood changes, peripheral neuropathy, can lower seizure threshold (increased risk in patients taking NSAIDs [NSAIDs increase the displacement of GABA from its receptor caused by quinolones] and those with a previous history of epilepsy))
Hyperglycemia/hypoglycemia (especially with gatifloxacin, which has been removed from the US market for this reason)
QT prolongation
Photosensitivity
Skin rash
Superinfection (most commonly with gram-positive pathogens)
Potentially life-threatening exacerbations in patients with myasthenia gravis
In children → potential damage to growing cartilage → reversible arthropathy
Muscle ache, leg cramps, tendinitis, tendon rupture, especially of the Achilles tendon (the risk of tendon rupture is higher for individuals over 60 years of age and for individuals on steroid therapy)
Increased risk for drug interactions as ciprofloxacin inhibits cytochrome P450

Contraindications:
Children and teenagers < 18 years of age (the only exception is ciprofloxacin which can be used in this cohort for complicated UTI including pyelonephritis and anthrax)
Patients > 60 years of age and those taking cortisol
Pregnant women
Breastfeeding women
Epilepsy, stroke, CNS lesions/inflammation
QT prolongation
Myasthenia gravis (potentially life-threatening exacerbations)
Cautious use in patients with renal failure, hepatic failure
Antacid use (aluminum- and magnesium-containing antacids decrease the absorption of fluoroquinolones)
Known aortic aneurysm or increased risk of aneurysms (e.g., Marfan syndrome, Ehlers-Danlos syndrome, advanced age, peripheral atherosclerotic disease, hypertension)

Mechanism of Resistance:
Chromosome-encoded mutation in DNA gyrase and topoisomerase IV enzymes
Altered cell wall permeability
Plasmid-encoded mutations in efflux pump proteins

93
Q

Levofloxacin

A

3rd generation respiratory fluoroquinolone
Oral or IV form

Mechanism:
Inhibition of prokaryotic topoisomerase II (DNA gyrase) and topoisomerase IV→ DNA supercoiling → formation of double-stranded breaks → inhibition of DNA replication and transcription (bactericidal effect)
CNS penetration → good
Route of elimination → primarily renal (via glomerular filtration and tubular secretion)
Absorption is reduced when coadministered with polyvalent cations (e.g., magnesium, calcium, iron).

Clinical Use:
Atypical bacteria (e.g., Legionella spp., Mycoplasma spp., Chlamydophila pneumoniae)
Also effective against anaerobes

Adverse Effects:
GI upset
Neurological symptoms (mild headache, dizziness, mood changes, peripheral neuropathy, can lower seizure threshold (increased risk in patients taking NSAIDs [NSAIDs increase the displacement of GABA from its receptor caused by quinolones] and those with a previous history of epilepsy))
Hyperglycemia/hypoglycemia (especially with gatifloxacin, which has been removed from the US market for this reason)
QT prolongation
Photosensitivity
Skin rash
Superinfection (most commonly with gram-positive pathogens)
Potentially life-threatening exacerbations in patients with myasthenia gravis
In children → potential damage to growing cartilage → reversible arthropathy
Muscle ache, leg cramps, tendinitis, tendon rupture, especially of the Achilles tendon (the risk of tendon rupture is higher for individuals over 60 years of age and for individuals on steroid therapy)
Increased risk for drug interactions as ciprofloxacin inhibits cytochrome P450

Contraindications:
Children and teenagers < 18 years of age (the only exception is ciprofloxacin which can be used in this cohort for complicated UTI including pyelonephritis and anthrax)
Patients > 60 years of age and those taking cortisol
Pregnant women
Breastfeeding women
Epilepsy, stroke, CNS lesions/inflammation
QT prolongation
Myasthenia gravis (potentially life-threatening exacerbations)
Cautious use in patients with renal failure, hepatic failure
Antacid use (aluminum- and magnesium-containing antacids decrease the absorption of fluoroquinolones)
Known aortic aneurysm or increased risk of aneurysms (e.g., Marfan syndrome, Ehlers-Danlos syndrome, advanced age, peripheral atherosclerotic disease, hypertension)

Mechanism of Resistance:
Chromosome-encoded mutation in DNA gyrase and topoisomerase IV enzymes
Altered cell wall permeability
Plasmid-encoded mutations in efflux pump proteins

94
Q

Moxifloxacin

A

4th generation respiratory fluoroquinolone
Oral or IV form

Mechanism:
Inhibition of prokaryotic topoisomerase II (DNA gyrase) and topoisomerase IV→ DNA supercoiling → formation of double-stranded breaks → inhibition of DNA replication and transcription (bactericidal effect)
CNS penetration → good
Undergoes biliary excretion.
Absorption is reduced when coadministered with polyvalent cations (e.g., magnesium, calcium, iron).

Clinical Use:
Atypical bacteria (e.g., Legionella spp., Mycoplasma spp., Chlamydophila pneumoniae)
Also effective against anaerobes
2nd-line treatment of tuberculosis in patients who cannot tolerate antitubercular drugs and in multidrug resistant tuberculosis

Adverse Effects:
GI upset
Neurological symptoms (mild headache, dizziness, mood changes, peripheral neuropathy, can lower seizure threshold (increased risk in patients taking NSAIDs [NSAIDs increase the displacement of GABA from its receptor caused by quinolones] and those with a previous history of epilepsy))
Hyperglycemia/hypoglycemia (especially with gatifloxacin, which has been removed from the US market for this reason)
QT prolongation
Photosensitivity
Skin rash
Superinfection (most commonly with gram-positive pathogens)
Potentially life-threatening exacerbations in patients with myasthenia gravis
In children → potential damage to growing cartilage → reversible arthropathy
Muscle ache, leg cramps, tendinitis, tendon rupture, especially of the Achilles tendon (the risk of tendon rupture is higher for individuals over 60 years of age and for individuals on steroid therapy)
Increased risk for drug interactions as ciprofloxacin inhibits cytochrome P450

Contraindications:
Children and teenagers < 18 years of age (the only exception is ciprofloxacin which can be used in this cohort for complicated UTI including pyelonephritis and anthrax)
Patients > 60 years of age and those taking cortisol
Pregnant women
Breastfeeding women
Epilepsy, stroke, CNS lesions/inflammation
QT prolongation
Myasthenia gravis (potentially life-threatening exacerbations)
Cautious use in patients with renal failure, hepatic failure
Antacid use (aluminum- and magnesium-containing antacids decrease the absorption of fluoroquinolones)
Known aortic aneurysm or increased risk of aneurysms (e.g., Marfan syndrome, Ehlers-Danlos syndrome, advanced age, peripheral atherosclerotic disease, hypertension)

Mechanism of Resistance:
Chromosome-encoded mutation in DNA gyrase and topoisomerase IV enzymes
Altered cell wall permeability
Plasmid-encoded mutations in efflux pump proteins

95
Q

Gemifloxacin

A

4th generation respiratory fluoroquinolone
Oral form

Mechanism:
Inhibition of prokaryotic topoisomerase II (DNA gyrase) and topoisomerase IV→ DNA supercoiling → formation of double-stranded breaks → inhibition of DNA replication and transcription (bactericidal effect)
CNS penetration → good
Route of elimination → primarily renal (via glomerular filtration and tubular secretion)
Absorption is reduced when coadministered with polyvalent cations (e.g., magnesium, calcium, iron).

Clinical Use:
Atypical bacteria (e.g., Legionella spp., Mycoplasma spp., Chlamydophila pneumoniae)
Also effective against anaerobes
Highly potent against penicillin-resistant pneumococci.

Adverse Effects:
GI upset
Neurological symptoms (mild headache, dizziness, mood changes, peripheral neuropathy, can lower seizure threshold (increased risk in patients taking NSAIDs [NSAIDs increase the displacement of GABA from its receptor caused by quinolones] and those with a previous history of epilepsy))
Hyperglycemia/hypoglycemia (especially with gatifloxacin, which has been removed from the US market for this reason)
QT prolongation
Photosensitivity
Skin rash
Superinfection (most commonly with gram-positive pathogens)
Potentially life-threatening exacerbations in patients with myasthenia gravis
In children → potential damage to growing cartilage → reversible arthropathy
Muscle ache, leg cramps, tendinitis, tendon rupture, especially of the Achilles tendon (the risk of tendon rupture is higher for individuals over 60 years of age and for individuals on steroid therapy)
Increased risk for drug interactions as ciprofloxacin inhibits cytochrome P450

Contraindications:
Children and teenagers < 18 years of age (the only exception is ciprofloxacin which can be used in this cohort for complicated UTI including pyelonephritis and anthrax)
Patients > 60 years of age and those taking cortisol
Pregnant women
Breastfeeding women
Epilepsy, stroke, CNS lesions/inflammation
QT prolongation
Myasthenia gravis (potentially life-threatening exacerbations)
Cautious use in patients with renal failure, hepatic failure
Antacid use (aluminum- and magnesium-containing antacids decrease the absorption of fluoroquinolones)
Known aortic aneurysm or increased risk of aneurysms (e.g., Marfan syndrome, Ehlers-Danlos syndrome, advanced age, peripheral atherosclerotic disease, hypertension)

Mechanism of Resistance:
Chromosome-encoded mutation in DNA gyrase and topoisomerase IV enzymes
Altered cell wall permeability
Plasmid-encoded mutations in efflux pump proteins

96
Q

Metronidazole

A

Oral or IV form

Mechanism:
Creates free radicals within the bacterial cell → DNA-strand breaks → cell death (bactericidal and antiprotozoal effect)
CNS penetration → good
Route of elimination → renal

Clinical Use:
Certain protozoa (e.g., Entamoeba histolytica, Giardia, Trichomonas)
Anaerobes (e.g., C. difficile, Bacteroides spp.)
Facultative anaerobes
Gardnerella vaginalis
Rosacea
Helicobacter pylori in place of amoxicillin (e.g., in case of penicillin allergy) as part of a triple therapy regimen
Not effective against aerobes (In anaerobes, metronidazole is reduced to its active metabolite. In aerobes, the presence of oxygen, which increases electron acceptance, prevents the reduction of metronidazole and its consequent formation into an active metabolite)

Adverse Effects:
Headache
Disulfiram-like reaction (nitroimidazoles inhibit acetaldehyde dehydrogenase → accumulation of acetaldehyde → immediate hangover-like symptoms after ethanol intake). Symptoms include flushing, tachycardia, and hypotension.
Metallic taste
Peripheral neuropathy (particularly with prolonged use)

97
Q

Tinidazole

A

Oral or IV form

Mechanism:
Creates free radicals within the bacterial cell → DNA-strand breaks → cell death (bactericidal and antiprotozoal effect)
CNS penetration → good
Route of elimination → renal

Clinical Use:
Certain protozoa (e.g., Entamoeba histolytica, Giardia, Trichomonas)
Anaerobes (e.g., C. difficile, Bacteroides spp.)
Facultative anaerobes
Gardnerella vaginalis
Rosacea
Helicobacter pylori in place of amoxicillin (e.g., in case of penicillin allergy) as part of a triple therapy regimen
Not effective against aerobes (In anaerobes, metronidazole is reduced to its active metabolite. In aerobes, the presence of oxygen, which increases electron acceptance, prevents the reduction of metronidazole and its consequent formation into an active metabolite)

Adverse Effects:
Headache
Disulfiram-like reaction (nitroimidazoles inhibit acetaldehyde dehydrogenase → accumulation of acetaldehyde → immediate hangover-like symptoms after ethanol intake). Symptoms include flushing, tachycardia, and hypotension.
Metallic taste
Peripheral neuropathy (particularly with prolonged use)

98
Q

Pentamidine

A

Mechanism:
Antimicrobial agent with a poorly understood mechanism

Clinical Use:
African trypanosomiasis
Babesiosis
Leishmaniasis
To prevent or treat Pneumocystis pneumonia (especially in patients unable to take TMP-SMX).

Adverse Effects:
Fatigue, dizziness, decreased appetite, and respiratory symptoms (e.g., cough, dyspnea, wheezing).

99
Q

Rifamycins

A

Oral or IV rifampin (rifampicin)
Oral rifabutin
Oral rifaximin
Oral rifapentine

Mechanism:
Inhibits bacterial DNA-dependent RNA-polymerase → prevention of transcription (mRNA synthesis) → inhibition of bacterial protein synthesis → cell death (bactericidal effect)
Route of elimination → biliary

Clinical Use:
Mycobacteria
Tuberculosis (together with isoniazid, pyrazinamide, and ethambutol)
Rifaximin is not absorbed systemically and is therefore used as a gut antibiotic.
Leprosy
-Long-term treatment of tuberculoid form → dapsone and rifampin (rifampin delays resistance to dapsone)
-Long-term treatment of lepromatous form → combination of dapsone, clofazimine, and rifampin
M. avium-intracellulare prophylaxis and treatment (rifabutin) (treatment of the most dangerous form, disseminated, consists of clarithromycin combined with ethambutol with a possible addition of rifabutin)
Meningococcal prophylaxis
Brucella
Chemoprophylaxis for people who had contact with children infected with H. influenzae type b
Rifaximin → second-line therapy in hepatic encephalopathy (can be added to nonabsorbable disaccharides (e.g., lactulose, lactitol) to decrease ammonia production by reducing the amount of urease-producing colonic bacteria)

Adverse Effects:
Harmless red-orange discoloration of body fluids (e.g., urine, tears)
Flu-like symptoms (fever, arthralgia; in severe cases hemolytic anemia, thrombocytopenia, and renal failure)
Minor hepatotoxicity (zone III)
Cytochrome P450 induction (CYP3A4, CYP2C9) (rifabutin is preferred for the treatment of mycobacterial infections in patients with HIV because it has a lower potential for CYP induction than rifampin; protease inhibitors and NNRTIs (e.g., efavirenz) are also cytochrome P450 substrates).
Intermittent therapy is associated with an increased risk of renal failure, so rifamycins should be administered daily (acute interstitial nephritis)

Contraindications:
Cautious use in patients with hepatic dysfunction

Mechanisms of Resistance:
RNA polymerase mutations decrease the affinity of rifamycins to RNA polymerase (mutations in the β subunit of the bacterial RNA polymerase confer near-complete resistance to the drug)
Resistance develops rapidly if used as monotherapy

100
Q

Rifampin (rifampicin)

A

Oral or IV form

Mechanism:
Inhibits bacterial DNA-dependent RNA-polymerase → prevention of transcription (mRNA synthesis) → inhibition of bacterial protein synthesis → cell death (bactericidal effect)
Route of elimination → biliary

Clinical Use:
Mycobacteria
Tuberculosis (together with isoniazid, pyrazinamide, and ethambutol)
Leprosy
-Long-term treatment of tuberculoid form → dapsone and rifampin (rifampin delays resistance to dapsone)
-Long-term treatment of lepromatous form → combination of dapsone, clofazimine, and rifampin
Meningococcal prophylaxis
Chemoprophylaxis for people who had contact with children infected with H. influenzae type b

Adverse Effects:
Harmless red-orange discoloration of body fluids (e.g., urine, tears)
Flu-like symptoms (fever, arthralgia; in severe cases hemolytic anemia, thrombocytopenia, and renal failure)
Minor hepatotoxicity
Cytochrome P450 induction (CYP3A4, CYP2C9) (rifabutin is preferred for the treatment of mycobacterial infections in patients with HIV because it has a lower potential for CYP induction than rifampin; protease inhibitors and NNRTIs (e.g., efavirenz) are also cytochrome P450 substrates).
Intermittent therapy is associated with an increased risk of renal failure, so rifamycins should be administered daily

Contraindications:
Cautious use in patients with hepatic dysfunction

Mechanisms of Resistance:
RNA polymerase mutations decrease the affinity of rifamycins to RNA polymerase.
Resistance develops rapidly if used as monotherapy (mutations in the β subunit of the bacterial RNA polymerase confer near-complete resistance to the drug)

101
Q

Rifabutin

A

Oral form

Mechanism:
Inhibits bacterial DNA-dependent RNA-polymerase → prevention of transcription (mRNA synthesis) → inhibition of bacterial protein synthesis → cell death (bactericidal effect)
Route of elimination → biliary

Clinical Use:
Mycobacteria
Tuberculosis (together with isoniazid, pyrazinamide, and ethambutol)
M. avium-intracellulare prophylaxis and treatment (rifabutin) (treatment of the most dangerous form, disseminated, consists of clarithromycin combined with ethambutol with a possible addition of rifabutin)
Meningococcal prophylaxis
Chemoprophylaxis for people who had contact with children infected with H. influenzae type b

Adverse Effects:
Harmless red-orange discoloration of body fluids (e.g., urine, tears)
Flu-like symptoms (fever, arthralgia; in severe cases hemolytic anemia, thrombocytopenia, and renal failure)
Minor hepatotoxicity
Cytochrome P450 induction (CYP3A4, CYP2C9) (rifabutin is preferred for the treatment of mycobacterial infections in patients with HIV because it has a lower potential for CYP induction than rifampin; protease inhibitors and NNRTIs (e.g., efavirenz) are also cytochrome P450 substrates).
Intermittent therapy is associated with an increased risk of renal failure, so rifamycins should be administered daily

Contraindications:
Cautious use in patients with hepatic dysfunction

Mechanisms of Resistance:
RNA polymerase mutations decrease the affinity of rifamycins to RNA polymerase (mutations in the β subunit of the bacterial RNA polymerase confer near-complete resistance to the drug)
Resistance develops rapidly if used as monotherapy

102
Q

Rifaximin

A

Oral form

Mechanism:
Inhibits bacterial DNA-dependent RNA-polymerase → prevention of transcription (mRNA synthesis) → inhibition of bacterial protein synthesis → cell death (bactericidal effect)
Route of elimination → biliary

Clinical Use:
Not absorbed systemically and is therefore used as a gut antibiotic.
Second-line therapy in hepatic encephalopathy (can be added to nonabsorbable disaccharides (e.g., lactulose, lactitol) to decrease ammonia production by reducing the amount of urease-producing colonic bacteria)

Adverse Effects:
Harmless red-orange discoloration of body fluids (e.g., urine, tears)
Flu-like symptoms (fever, arthralgia; in severe cases hemolytic anemia, thrombocytopenia, and renal failure)
Minor hepatotoxicity
Cytochrome P450 induction (CYP3A4, CYP2C9) (rifabutin is preferred for the treatment of mycobacterial infections in patients with HIV because it has a lower potential for CYP induction than rifampin; protease inhibitors and NNRTIs (e.g., efavirenz) are also cytochrome P450 substrates).
Intermittent therapy is associated with an increased risk of renal failure, so rifamycins should be administered daily

Contraindications:
Cautious use in patients with hepatic dysfunction

Mechanisms of Resistance:
RNA polymerase mutations decrease the affinity of rifamycins to RNA polymerase.
Resistance develops rapidly if used as monotherapy (mutations in the β subunit of the bacterial RNA polymerase confer near-complete resistance to the drug)

103
Q

Rifapentine

A

Oral form

Mechanism:
Inhibits bacterial DNA-dependent RNA-polymerase → prevention of transcription (mRNA synthesis) → inhibition of bacterial protein synthesis → cell death (bactericidal effect)
Route of elimination → biliary

Clinical Use:
Mycobacteria
Meningococcal prophylaxis
Chemoprophylaxis for people who had contact with children infected with H. influenzae type b

Adverse Effects:
Harmless red-orange discoloration of body fluids (e.g., urine, tears)
Flu-like symptoms (fever, arthralgia; in severe cases hemolytic anemia, thrombocytopenia, and renal failure)
Minor hepatotoxicity
Cytochrome P450 induction (CYP3A4, CYP2C9) (rifabutin is preferred for the treatment of mycobacterial infections in patients with HIV because it has a lower potential for CYP induction than rifampin; protease inhibitors and NNRTIs (e.g., efavirenz) are also cytochrome P450 substrates).
Intermittent therapy is associated with an increased risk of renal failure, so rifamycins should be administered daily

Contraindications:
Cautious use in patients with hepatic dysfunction

Mechanisms of Resistance:
RNA polymerase mutations decrease the affinity of rifamycins to RNA polymerase.
Resistance develops rapidly if used as monotherapy (mutations in the β subunit of the bacterial RNA polymerase confer near-complete resistance to the drug)

104
Q

Isoniazid

A

Mechanism:
Prodrug and needs to be converted into its active metabolite by bacterial catalase-peroxidase (encoded by KatG).
Prevents cell wall synthesis by inhibiting the synthesis of mycolic acid
CNS penetration → variable
Metabolization → primarily hepatic
INH is converted into various metabolites (e.g., via acetylation), some of which are hepatotoxic (e.g., hydrazine, acetylhydrazine).
Main metabolic enzyme → N-acetyltransferase (NAT) → involved in metabolite formation and subsequent neutralization
The rate of NAT acetylation is genetically determined.
-Individuals with slow acetylation → higher half-life due to lack of hepatic NAT → increased risk of drug-induced toxicity
-Individuals with fast acetylation → lower half-life of active drug → administration of a higher dose is required for reaching the same blood concentration compared to individuals with slow acetylation
Inhibits cytochrome P450 isoforms (CYP1A2, CYP2A6, CYP2C19, and CYP3A4)
Route of elimination → renal elimination after hepatic metabolism (dose adjustment is not required in the case of renal failure)

Clinical Use:
Treatment of TB (together with rifampin, pyrazinamide, and ethambutol)
The only drug that can be used as monoprophylaxis against TB
First-line monotherapy for latent TB

Adverse Effects:
Hepatotoxicity
Anion gap metabolic acidosis
Drug-induced lupus erythematosus
High doses of INH can precipitate benzodiazepine-refractory seizures
Vitamin B6 deficiency (INH should be administered with pyridoxine to avoid vitamin B6 deficiency)
Peripheral neuropathy due to S-adenosylmethionine accumulation
Sideroblastic anemia (inhibits pyridoxine phosphokinase and decrease pyridoxine conversion to PLP, an ALA synthase cofactor), aplastic anemia, thrombocytopenia
Optic neuropathy (less common compared to treatment with ethambutol)
Pellagra

Contraindications:
Cautious use in patients with renal and/or hepatic dysfunction

Mechanisms of Resistance:
Mutations causing decreased KatG (coding for bacterial catalase-peroxidase) → decreased expression of catalase-peroxidase → less/no biologically active INH

105
Q

Pyrazinamide

A

Mechanism:
Not completely understood
Prodrug → converted into active form pyrazinoic acid
Most effective at acidic pH (e.g., in acidic phagolysosomes)
Bactericidal effect
CNS penetration → only when meninges are inflamed
Route of elimination → renal elimination after hepatic metabolism

Clinical Use:
M. tuberculosis

Adverse Effects:
Hyperuricemia (decrease uric acid clearance; can precipitate gout)
Hepatotoxicity

Contraindications:
Consider use in pregnant and breastfeeding women only if benefits outweigh the risks
Hepatic failure
Acute gout

Mechanisms of Resistance:
Mutations in RpsA gene coding for ribosomal protein S1 (the mechanism through which the mutation leads to the resistance to pyrazinamide is unknown)

106
Q

Ethambutol

A

Mechanism:
Inhibits arabinosyltransferase → ↓ carbohydrate polymerization → prevention of mycobacterial cell wall synthesis (bacteriostatic effect) (arabinosyltransferase catalyzes the production of arabinogalactan, a constituent of the cell wall of Mycobacteria)
CNS penetration → only when meninges are inflamed
Route of elimination → primarily renal

Clinical Use:
M. tuberculosis therapy (together with isoniazid, rifampin, and pyrazinamide)
M. avium-intracellulare treatment (together with azithromycin or clarithromycin; ciprofloxacin or rifabutin can be added)

Adverse Effects:
Optic neuropathy with decreased visual acuity and red-green color blindness which may result in irreversible blindness
Resistance develops rapidly if used as monotherapy

Contraindications:
Contraindicated in patients who are unable to report visual changes
Contraindicated in patients with optic neuritis

Mechanisms of Resistance:
Mutations of EmbCAB gene coding for arabinosyltransferase

107
Q

Antibiotics Contraindicated in Children

A

Aminoglycosides
Tetracyclines (< 8 years old (except doxycycline))
Tigecycline (< 8 years old)
Chloramphenicol
Fluoroquinolones
Sulfonamides, trimethoprim (TMP) and pyrimethamine (< 2 years old)
Nitrofurantoin (< 1 month old)
Ethambutol (young children who are unable to report visual changes)

108
Q

Antibiotics Contraindicated in Pregnant Women

A

Aminoglycosides
Tetracyclines
Tigecycline
Chloramphenicol
Fluoroquinolones
Sulfonamides with trimethoprim (TMP) or pyrimethamine
Macrolides (clarithromycin and erythromycin estolate) (the rate of hepatotoxicity is higher in pregnant women if using erythromycin estolate)

109
Q

Antibiotics Contraindicated in Individuals with Renal Failure

A
Fosfomycin
Aminoglycosides
Tetracyclines
Sulfonamides with trimethoprim (TMP) or pyrimethamine
Nitrofurantoin
110
Q

Antibiotics Contraindicated in Individuals with Hepatic Failure

A

Tetracyclines
Macrolides (azithromycin and clarithromycin)
Sulfonamides with trimethoprim (TMP) or pyrimethamine
Nitrofurantoin
Pyrazinamide

111
Q

Antibiotics Safe to Use in Individuals with Hepatic Failure

A
β-lactams
Vancomycin
Fosfomycin
Daptomycin
Polymyxin
Aminoglycosides
Clindamycin
Streptogramin (quinupristin, dalfopristin)
Linezolid
Ethambutol
112
Q

Antibiotics Safe to Use in Individuals with Renal Failure

A

β-lactams
Vancomycin
Daptomycin
Clindamycin
Streptogramin (quinupristin, dalfopristin)
Linezolid
Metronidazole
Rifamycins (should be administered daily because intermittent therapy is associated with an increased risk of renal failure)
Pyrazinamide
Ethambutol
Macrolides (erythromycin and azithromycin)

113
Q

Antibiotics Safe to Use in Pregnant Women

A

β-lactams
Fosfomycin
Daptomycin
Macrolides (azithromycin and erythromycin) (except erythromycin estolate)

114
Q

Amphotericin B

A

Routes of administration → intravenous, intrathecal, bladder irrigation

Mechanism:
Binds ergosterol (unique to fungi); forms membrane pores that allow leakage of electrolytes.
Binds cholesterol to a degree, which explains a larger number of its adverse effects

Clinical Use:
Severe systemic mycoses, such as:
-Cryptococcal disease (in combination with flucytosine for cryptococcal meningitis)
-Sporotrichosis
-Aspergillosis
-Blastomycosis
-Candidiasis
-Coccidioides (administered intrathecally for coccidioidal meningitis)
-Histoplasmosis
-Mucormycosis
-Fungal cystitis (administered via bladder irrigation)
-Fungal meningitis (intrathecal administration)

Adverse Effects:
Nephrotoxicity (lipid-based formulations of the drug and IV hydration reduce nephrotoxicity)
IV phlebitis (administration via a central line will reduce the risk of IV phlebitis)
Impaired renal tubule permeability → hypokalemia and hypomagnesemia (restoring K+ and Mg2+ while administering the drug can counter this effect)
Distal renal tubular acidosis (type 1)
Fever, chills (amphotericin B is sometimes referred to as “shake and bake”)
Anemia (suppression of renal EPO synthesis)
Arrhythmias
Hypotension
Headache

Contraindications:
Electrolyte disturbances (e.g., hypokalemia, hypomagnesemia)
Renal dysfunction
Pregnancy

115
Q

Nystatin

A
Mechanism:
Same as amphotericin B. 
Binds ergosterol (unique to fungi); forms membrane pores that allow leakage of electrolytes. 
Topical use only as too toxic for intravenous use
Clinical Use:
Topical
-Vaginal candidiasis
-Diaper rash
-NOT effective against dermatophytes
Oral (swish and swallow)
-Oropharyngeal candidiasis

Adverse effects
Gastrointestinal symptoms (e.g., diarrhea, nausea, pain)
Contact dermatitis
Stevens-Johnson syndrome

Contraindications:
Pregnancy

116
Q

Flucytosine

A

Route of administration → oral

Mechanism:
Converted to 5-fluorouracil by fungal cytosine deaminase, thereby inhibiting DNA and RNA synthesis

Clinical Use:
Combination with amphotericin B → systemic fungal infections (especially cryptococcal meningitis)
Monotherapy → non-life-threatening infections, such as genitourinary candida infections, that do not respond to other drugs

Adverse Effects:
Bone marrow suppression with pancytopenia
Hepatic injury (elevated liver enzymes, especially in patients with flucytosine blood levels > 100 mg/L)
Renal failure
Gastrointestinal upset

Contraindications:
Renal impairment
Pregnancy and breastfeeding

117
Q

Azoles

A

Group of antifungal agents with a broad spectrum of activity that can be divided into triazoles and imidazoles

Routes of administration:
Imidazoles → topical, oral
Triazoles → oral, IV

Mechanism:
Inhibition of 14-alpha demethylase, a fungal cytochrome P450 → ↓ fungal synthesis of ergosterol from lanosterol → ↓ levels of ergosterol → membrane instability → cellular death.
Ketoconazole → inhibition of 17α-hydroxylase/17,20-lyase
Inhibition of cytochrome P450 can influence serum concentrations of other medications
Cautious use in patients with renal and/or hepatic dysfunction
PPIs must be discontinued (azoles require a low pH to be absorbed)

Adverse Effects:
-Topical Use:
Local burning sensation
Pruritus
-Systemic Use:
Hepatotoxicity (inhibits cytochrome P450 → ↑ concentration of many drugs metabolized by CYP450 (e.g., warfarin, simvastatin, cyclosporine, theophylline))
(serum transaminases and alkaline phosphatase elevation (especially severe with ketoconazole))
Gynecomastia due to inhibition of testosterone synthesis (especially seen with ketoconazole)
Gastrointestinal upset
QT prolongation → torsade de pointes
Hypokalemia (especially ketoconazole and itraconazole)
Ketoconazole → adrenal cortex insufficiency (caused by inhibited cortisol synthesis (the effect can be used therapeutically to treat Cushing syndrome))
Voriconazole → dose-dependent, reversible visual disorders (e.g., photosensitivity)

Contraindications:
Cardiac arrhythmias
Severe cardiac insufficiency

118
Q

Clotrimazole

A

Imidazole derivative (topical, oral)

Mechanism:
Inhibition of 14-alpha demethylase, a fungal cytochrome P450 → ↓ fungal synthesis of ergosterol from lanosterol → ↓ levels of ergosterol → membrane instability → cellular death.
Cautious use in patients with renal and/or hepatic dysfunction
PPIs must be discontinued (azoles require a low pH to be absorbed)

Clinical Use:
Primarily topical fungal infections
Vaginal candidiasis
Tinea infections
Oropharyngeal candidiasis

Adverse Effects:
-Topical Use:
Local burning sensation
Pruritus
-Systemic Use:
Hepatotoxicity (inhibits cytochrome P450 → ↑ concentration of many drugs metabolized by CYP450 (e.g., warfarin, simvastatin, cyclosporine, theophylline))
(serum transaminases and alkaline phosphatase elevation (especially severe with ketoconazole))
Gynecomastia due to inhibition of testosterone synthesis (especially seen with ketoconazole)
Gastrointestinal upset
QT prolongation → torsade de pointes
Hypokalemia (especially ketoconazole and itraconazole)

119
Q

Miconazole

A

Imidazole derivative

Mechanism:
Inhibition of 14-alpha demethylase, a fungal cytochrome P450 → ↓ fungal synthesis of ergosterol from lanosterol → ↓ levels of ergosterol → membrane instability → cellular death.
Cautious use in patients with renal and/or hepatic dysfunction
PPIs must be discontinued (azoles require a low pH to be absorbed)

Clinical Use:
Primarily topical fungal infections
Vaginal candidiasis
Tinea infections
Oropharyngeal candidiasis

Adverse Effects:
-Topical Use:
Local burning sensation
Pruritus
-Systemic Use:
Hepatotoxicity (inhibits cytochrome P450 → ↑ concentration of many drugs metabolized by CYP450 (e.g., warfarin, simvastatin, cyclosporine, theophylline))
(serum transaminases and alkaline phosphatase elevation (especially severe with ketoconazole))
Gynecomastia due to inhibition of testosterone synthesis (especially seen with ketoconazole)
Gastrointestinal upset
QT prolongation → torsade de pointes
Hypokalemia (especially ketoconazole and itraconazole)

120
Q

Ketoconazole

A
Imidazoles derivative (topical, oral)
Triazoles → oral, IV

Mechanism:
Inhibition of 14-alpha demethylase, a fungal cytochrome P450 → ↓ fungal synthesis of ergosterol from lanosterol → ↓ levels of ergosterol → membrane instability → cellular death.
Ketoconazole → inhibition of 17α-hydroxylase/17,20-lyase
Inhibition of cytochrome P450 can influence serum concentrations of other medications
Cautious use in patients with renal and/or hepatic dysfunction
PPIs must be discontinued (azoles require a low pH to be absorbed)

Clinical Use:
Tinea versicolor
Prostate cancer
Psoriasis (applied topically to prevent fungal skin infections)
Hypercortisolism (e.g., Cushing syndrome) (because it inhibits cortisol synthesis)

Adverse Effects:
-Topical Use:
Local burning sensation
Pruritus
-Systemic Use:
Hepatotoxicity (inhibits cytochrome P450 → ↑ concentration of many drugs metabolized by CYP450 (e.g., warfarin, simvastatin, cyclosporine, theophylline)) (serum transaminases and alkaline phosphatase elevation (especially severe with ketoconazole))
Gynecomastia due to inhibition of testosterone synthesis (especially seen with ketoconazole)
Gastrointestinal upset
QT prolongation → torsade de pointes
Hypokalemia (especially ketoconazole and itraconazole)
Ketoconazole → adrenal cortex insufficiency (caused by inhibited cortisol synthesis (the effect can be used therapeutically to treat Cushing syndrome))

121
Q

Fluconazole

A

Triazoles derivative (oral, IV)

Mechanism:
Inhibition of 14-alpha demethylase, a fungal cytochrome P450 → ↓ fungal synthesis of ergosterol from lanosterol → ↓ levels of ergosterol → membrane instability → cellular death.
Cautious use in patients with renal and/or hepatic dysfunction
PPIs must be discontinued (azoles require a low pH to be absorbed)

Clinical Use:
Drug of choice maintenance therapy of cryptococcal meningitis in patients with AIDS (initially, cryptococcal meningitis is treated with amphotericin B PLUS either flucytosine or fluconazole)
Candidiasis (all forms)
Coccidioidomycosis
Histoplasmosis
Blastomycosis

Adverse Effects:
-Topical Use:
Local burning sensation
Pruritus
-Systemic Use:
Hepatotoxicity (inhibits cytochrome P450 → ↑ concentration of many drugs metabolized by CYP450 (e.g., warfarin, simvastatin, cyclosporine, theophylline))
(serum transaminases and alkaline phosphatase elevation (especially severe with ketoconazole))
Gynecomastia due to inhibition of testosterone synthesis (especially seen with ketoconazole)
Gastrointestinal upset
QT prolongation → torsade de pointes
Hypokalemia (especially ketoconazole and itraconazole)

122
Q

Voriconazole

A

Triazoles derivative (oral, IV)

Mechanism:
Inhibition of 14-alpha demethylase, a fungal cytochrome P450 → ↓ fungal synthesis of ergosterol from lanosterol → ↓ levels of ergosterol → membrane instability → cellular death.
Cautious use in patients with renal and/or hepatic dysfunction
PPIs must be discontinued (azoles require a low pH to be absorbed)

Clinical Use:
Drug of choice for invasive aspergillosis
Some forms of Candida

Adverse Effects:
-Topical Use:
Local burning sensation
Pruritus
-Systemic Use:
Hepatotoxicity (inhibits cytochrome P450 → ↑ concentration of many drugs metabolized by CYP450 (e.g., warfarin, simvastatin, cyclosporine, theophylline))
(serum transaminases and alkaline phosphatase elevation (especially severe with ketoconazole))
Gynecomastia due to inhibition of testosterone synthesis (especially seen with ketoconazole)
Gastrointestinal upset
QT prolongation → torsade de pointes
Hypokalemia (especially ketoconazole and itraconazole)
Voriconazole → dose-dependent, reversible visual disorders (e.g., photosensitivity)

123
Q

Itraconazole

A

Triazoles derivative (oral, IV)

Mechanism:
Inhibition of 14-alpha demethylase, a fungal cytochrome P450 → ↓ fungal synthesis of ergosterol from lanosterol → ↓ levels of ergosterol → membrane instability → cellular death.
Cautious use in patients with renal and/or hepatic dysfunction
PPIs must be discontinued (azoles require a low pH to be absorbed)

Clinical Use:
Histoplasmosis
Blastomycosis
Coccidioidomycosis
Sporotrichosis
Allergic bronchopulmonary aspergillosis, aspergilloma
Particularly effective in dermatophytosis (e.g., onychomycosis)
Oropharyngeal/esophageal candidiasis

Adverse Effects:
-Topical Use:
Local burning sensation
Pruritus
-Systemic Use:
Hepatotoxicity (inhibits cytochrome P450 → ↑ concentration of many drugs metabolized by CYP450 (e.g., warfarin, simvastatin, cyclosporine, theophylline))
(serum transaminases and alkaline phosphatase elevation (especially severe with ketoconazole))
Gynecomastia due to inhibition of testosterone synthesis (especially seen with ketoconazole)
Gastrointestinal upset
QT prolongation → torsade de pointes
Hypokalemia (especially ketoconazole and itraconazole)

124
Q

Posaconazole

A

Triazoles derivative (oral, IV)

Mechanism:
Inhibition of 14-alpha demethylase, a fungal cytochrome P450 → ↓ fungal synthesis of ergosterol from lanosterol → ↓ levels of ergosterol → membrane instability → cellular death.
Cautious use in patients with renal and/or hepatic dysfunction
PPIs must be discontinued (azoles require a low pH to be absorbed)

Clinical Use:
Oropharyngeal candidiasis
Prophylactic treatment of invasive aspergillosis and Candida infections in immunocompromised patients

Adverse Effects:
Hepatotoxicity (inhibits cytochrome P450 → ↑ concentration of many drugs metabolized by CYP450 (e.g., warfarin, simvastatin, cyclosporine, theophylline)) (serum transaminases and alkaline phosphatase elevation (especially severe with ketoconazole))
Gynecomastia due to inhibition of testosterone synthesis (especially seen with ketoconazole)
Gastrointestinal upset
QT prolongation → torsade de pointes
Hypokalemia (especially ketoconazole and itraconazole)

125
Q

Isavuconazole

A

Triazoles derivative (oral, IV)

Mechanism:
Inhibition of 14-alpha demethylase, a fungal cytochrome P450 → ↓ fungal synthesis of ergosterol from lanosterol → ↓ levels of ergosterol → membrane instability → cellular death.
Cautious use in patients with renal and/or hepatic dysfunction
PPIs must be discontinued (azoles require a low pH to be absorbed)

Clinical Use:
Invasive aspergillosis
Invasive mucormycosis

Adverse Effects:
-Topical Use:
Local burning sensation
Pruritus
-Systemic Use:
Hepatotoxicity (inhibits cytochrome P450 → ↑ concentration of many drugs metabolized by CYP450 (e.g., warfarin, simvastatin, cyclosporine, theophylline)) (serum transaminases and alkaline phosphatase elevation (especially severe with ketoconazole))
Gynecomastia due to inhibition of testosterone synthesis (especially seen with ketoconazole)
Gastrointestinal upset
QT prolongation → torsade de pointes
Hypokalemia (especially ketoconazole and itraconazole)

126
Q

Terbinafine

A

Route of administration → oral

Mechanism:
Inhibition fungal squalene epoxidase → ↓ synthesis of ergosterol → accumulation of squalene → ↑ membrane permeability → cell death

Clinical Use:
Dermatophytosis (most commonly onychomycosis)
Tinea infections (e.g., tinea pedis)

Adverse Effects:
Hepatotoxicity 
Dysgeusia
Gastrointestinal upset
Headache
Exacerbation of autoimmune diseases (e.g., systemic lupus erythematosus)

Contraindications:
Impaired liver function
Systemic lupus erythematosus
Breastfeeding

127
Q

Echinocandins

A

Caspofungin
Anidulafungin
Micafungin

Route of administration → IV

Mechanism:
Inhibition of β-glucan synthesis → disruption of fungal cell wall synthesis → ↓ resistance against osmotic forces → cell death

Clinical Use:
Invasive aspergillosis (less toxic than amphotericin B)
Invasive candidiasis (particularly biofilm-embedded Candida)
Adverse Effects:
Flushing (due to release of histamine)
Hepatotoxicity (serum transaminases and alkaline phosphatase elevation)
Gastrointestinal upset
Hypotension
Phlebitis/pain at the injection site
Fever, shivering
128
Q

Ciclopirox

A

Route of administration → topical

Mechanism:
Not fully understood
Likely disruption of DNA, RNA, and protein synthesis

Clinical Use:
Tinea versicolor
Dermatophytosis (e.g., onychomycosis)
Seborrheic dermatitis of the scalp

Adverse Effects:
Facial edema
Ventricular tachycardia
Skin irritations (should not be applied to the eye or mucous membranes)
Pruritus
129
Q

Griseofulvin

A

Route of administration → oral

Mechanism:
Binds to keratin precursor cells → accumulation in keratin-rich tissues (e.g., nails, hair) → entry into the fungal cell → interference with microtubule function → disruption of fungal mitosis

Clinical Use:
Dermatophyte infections (e.g., tinea pedis)
Not effective against tinea versicolor

Adverse Effects:
Hepatotoxicity
Carcinogenicity
Teratogenicity
Enzyme induction → ↓ concentration of many drugs metabolized by cytochrome P450 (e.g., warfarin)
Confusion, headaches
Disulfiram-like reaction
Urticaria and severe skin reactions (e.g., Stevens-Johnson syndrome)

Contraindications:
Porphyria
Hepatic failure

130
Q

Eflornithine

A

Mechanism:
Inhibits ornithine carboxylase, which is required for cell differentiation and proliferation
CNS penetration

Clinical Use:
Trypanosoma brucei (not efficient in the treatment of East African sleeping sickness because T. b. rhodesiense has a much higher turnover rate of ornithine carboxylase than T.b. gambiense)
Adverse Effects:
Bone marrow toxicity
Seizures
Hearing loss
Alopecia, rash
Diarrhea
131
Q

Nifurtimox

A

Mechanism:
Not clearly understood
Most likely induces reactions that create oxidative stress
CNS penetration

Clinical Use:
Trypanosoma brucei
Second-line drug in the treatment of Chagas disease (American trypanosomiasis).

Adverse Effects:
Seizures
Cognitive impairment

132
Q

Melarsoprol

A

Mechanism:
A prodrug that contains arsenic
Taken up by amino-purine transporters (e.g., P2) of T. brucei → metabolized into an active melarsen oxide → lysis of T. brucei through an unknown mechanism
Only a small amount of melarsoprol (< 1%) enters the CSF.

Clinical Use:
Trypanosoma brucei

Adverse Effects:
Post-treatment reactive encephalopathy (PTRE) (clinical manifestations include headache, high fever, tremors, impaired speech, and seizures. In order to prevent PTRE, patients should receive glucocorticoids shortly before initiating therapy and until the end of treatment)
Jarisch-Herxheimer reaction
Nephrotoxicity
Peripheral neuropathy
Thrombophlebitis
133
Q

Suramin

A

Mechanism:
Not clearly understood
Most likely inhibits enzymes involved in DNA replication and protein synthesis
No CNS penetration

Clinical Use:
Onchocerciasis
Trypanosoma brucei

Adverse Effects:
Hypersensitivity (skin test with 100–200 mg of IV eflornithine must be performed before administering suramin)
Nephrotoxicity
Peripheral neuropathy
Hypogonadism among male patients
134
Q

Sodium stibogluconate

A

Mechanism:
Inhibits the production of purine triphosphate molecules (e.g. ATP), ultimately stalling the metabolism of Leishmania species.

Clinical Use:
All forms of Leishmaniasis.

135
Q

Permethrin

A

5% lotion

Mechanism: 
Topical application (additionally, ivermectin can be given orally) 
Blocks the deactivation of Na+ channels → uncontrollable depolarization of neuronal membranes → seizures and ultimately paralysis and death of arthropod
Should be applied to every part of the body and left on the skin for 8–12 hours before rinsing off; application may be repeated 1 week later.

Clinical Use:
Scabies (Sarcoptes scabiei)
Lice (Pediculosis capitis, pediculosis pubis)

Adverse Effects:
Pruritus
Erythema

136
Q

Dimeticone

A

Mechanism:
Local application in head lice infestation. Dosage is dependent on the hair length.
Penetrates into the respiratory orifice of lice and blocks breathing

Clinical Use:
Lice (Pediculosis capitis)

Adverse Effects:
Pruritus
Erythema

137
Q

Malathion

A

Mechanism:
Commonly used as an insecticide in agriculture and public recreation areas
Topical application (additionally, ivermectin can be given orally)
Irreversible acetylcholinesterase inhibitor

Clinical Use:
Scabies (Sarcoptes scabiei)
Lice (Pediculosis capitis, pediculosis pubis)

Adverse Effects:
Carcinogenic

138
Q

Bendazoles (e.g., albendazole, mebendazole)

A

Mechanism:
Inhibit microtubule polymerization resulting in:
-Decreased glucose uptake and glycogen synthesis
-Degeneration of mitochondria and endoplasmic reticulum
-Lysosomal release

Clinical Use:
Ascariasis
Enterobiasis
Trichuriasis
Strongyloidiasis
Hookworm infection
Cysticercosis
Trichinellosis
Toxocariasis
Adverse Effects:
Headache
GI upset (especially in short term use)
Increased liver enzymes
Rash, urticaria
Hair loss
Agranulocytosis
139
Q

Pyrantel pamoate

A

Mechanism:
Neuromuscular blocking agents that inhibit the release of acetylcholine and acetylcholinesterase

Clinical Use:
Ascariasis (in pregnant women)
Enterobiasis
Hookworm infection

Adverse Effects:
Headache
Fever
Dizziness
Nausea, vomiting
Diarrhea
Abdominal pain
Increased liver enzymes
140
Q

Praziquantel

A

Mechanism:
Causes vacuolization of the schistosome integument and increases cell membrane permeability to calcium resulting in paralysis, dislodgement, and death of parasite
A single dose is sufficient for most worms.

Clinical Use:
Taeniasis
Dyphllobothriasis
Schistosomiasis
Neurocysticercosis
Clonorchiasis
Paragonomiasis
Adverse Effects:
Nausea, vomiting
Abdominal pain
Increased liver enzymes
Rash, pruritis
Joint/muscle pain
Headache
Seizure
141
Q

Ivermectin

A

Mechanism:
Paralyzes parasite by:
-Enhancing γ-aminobutyric acid (GABA) mediated neurotransmission
-Binding to muscle glutamate-gated chloride ion channels → cell hyperpolarization

Clinical Use:
Onchocerciasis
Strongyloidiasis
Hookworm infection

Adverse Effects:
Fever
Dizziness
Tachycardia
Hypotension
Nausea, vomiting
Abdominal pain
Rash, pruritis
Lymphangitis
Joint/muscle pain
142
Q

Diethylcarbamazine

A

Mechanism:
Immobilizes microfilariae
Sensitizes the microfilariae to phagocytosis through immobilization
Alteration of surface structure and displacement from the attachment site

Clinical Use:
Loaiasis
Lymphatic filariasis

Adverse Effects:
Headache
Malaise, weakness
Anorexia, vomiting
Rash, facial swelling
Muscle and joint pain
Vision loss
Eosinophilia
Leukocytosis
143
Q

Chloroquine/hydroxychloroquine

A

Mechanism:
The precise mechanisms of action are unknown, but the following are the most widely accepted hypotheses:
-Antimalarial action → drug accumulation in the parasite’s food vacuole → inhibition of heme polymerization (and thus, detoxification) into hemozoin and formation of highly toxic heme-chloroquine complexes → lysis of membranes and death of the parasites
-Antirheumatoid action → interference with antigen processing in macrophages and other APCs → ↓ formation of peptide-MHC protein complexes → ↓ immune response against autoantigenic peptides

Clinical Use:
Treatment and prophylaxis of malaria due to Plasmodium malariae, P. ovale, or P. vivax (strains of P. falciparum are usually resistant)
Rheumatoid arthritis (basic therapy)
Systemic lupus erythematosus and discoid lupus erythematosus (without organ involvement) (hydroxychloroquine is used for therapy as well as prevention of further attacks in these autoimmune conditions)
Porphyria cutanea tarda (in low doses) (chloroquine binds to porphyrins which are then excreted by the kidneys)

Adverse Effects:
Gastrointestinal
Nausea with cramps (most common)
Anorexia
Vomiting
Visual disturbances:
-Irreversible bilateral retinopathy (key fundoscopic feature is bull's eye maculopathy and paracentral scotomata) (incidence of retinopathy from chloroquine and hydroxychloroquine (the less toxic metabolite of chloroquine) increases with both the dose and the duration of treatment)
-Blurred vision
-Photophobia
Pruritus (most commonly seen in dark-skinned individuals) (dark-skinned individuals have a higher amount of melanin. A number of studies have shown that chloroquine preferentially binds to melanocytes. This could be a reason why pruritus especially affects dark-skinned individuals)
Photosensitivity
Alopecia
Whitening of hair
Neurologic
Myasthenia-like muscle weakness
Sensorineural deafness
Tinnitus
Cranial nerve palsies
Torsades de pointes (due to possible QT prolongation), can cause sudden death
Thrombocytopenia, leukopenia
Worsening of preexisting conditions → epilepsy, psoriasis, porphyria and preexisting retinal damage

Mechanism of Resistance:
Attributed to the development of membrane efflux pumps, which decrease the intracellular concentration of the drug (occurs especially in P. falciparum)

144
Q

Amantadine

A

Mechanism:
M2 ion channel blocker (protons flow into the interior of the virus via the M2 ion channel, which leads to viral uncoating. Amantadine blocks these channels, inhibiting the release of the viral genome)
Weak NMDA receptor antagonist (increases dopamine release and blocks dopamine reuptake, reducing bradykinesia in patients with Parkinson disease.)

Clinical Use:
Influenza A (not recommended as first-line treatment, but may be given to patients with oseltamivir-resistant influenza A)
Parkinson disease

Adverse Effects:
Ataxia
Anxiety
Nausea
Vomiting
Livedo reticularis
Peripheral edema

Amantadine only

  • Orthostatic dysregulation
  • QT interval prolongation
145
Q

Rimantadine

A

Mechanism:
M2 ion channel blocker

Clinical Use:
Influenza A (not recommended as first-line treatment, but may be given to patients with oseltamivir-resistant influenza A)
Adverse Effects:
Ataxia
Anxiety
Nausea
Vomiting
Livedo reticularis
Peripheral edema
146
Q

Oseltamivir/Zanamivir

A

Mechanism:
Neuraminidase inhibitor: blockage of viral budding and prevention of viral dissemination into the bloodstream by inhibiting neuraminidase enzyme
Administration within 2 days of symptom onset is vital to reduce the duration of illness and alleviate symptoms.

Clinical Use:
Treatment of influenza A and B (reduces symptom duration if taken within 1–2 days of symptom onset)
Prophylaxis of influenza in adults and pediatric patients ≥ 5 years of age

Adverse Effects:
Upper respiratory tract infections
Nausea
Vomiting
Headache
147
Q

Peramivir

A

Mechanism:
Neuraminidase inhibitor: blockage of viral budding and prevention of viral dissemination into the bloodstream by inhibiting neuraminidase enzyme
Administration within 2 days of symptom onset is vital to reduce the duration of illness and alleviate symptoms.

Clinical Use:
Treatment of influenza A and B (reduces symptom duration if taken within 1–2 days of symptom onset)
Prophylaxis of influenza in adults and pediatric patients ≥ 5 years of age

Adverse Effects:
Upper respiratory tract infections
Nausea
Vomiting
Headache
148
Q

Valacyclovir

A

Valacyclovir (prodrug of acyclovir with greater oral bioavailability)

Mechanism:
Guanosine analogs.
Monophosphorylated by HSV/VZV thymidine kinase and not phosphorylated in uninfected cells –> few adverse effects.
Triphosphate formed by cellular enzymes.
Preferentially inhibit viral DNA polymerase by chain termination.

Clinical Use:
HSV and VZV. Weak activity against EBV.
No activity against CMV.
Used for HSV- induced mucocutaneous and genital lesions as well as for encephalitis.
Prophylaxis in immunocompromised patients.
Only affects actively replicating viruses and has no effect on latent infections of HSV and VZV.
For herpes zoster, use famciclovir.

Adverse Effects:
Obstructive crystalline nephropathy and acute renal failure if not adequately hydrated
Thrombotic thrombocytopenic purpura
Gastrointestinal symptoms (e.g., nausea)
↑ Transaminases

Mechanism of Resistance:
Mutated viral thymidine kinase.

149
Q

Penciclovir

A

Mechanism:
Guanosine analogs.
Monophosphorylated by HSV/VZV thymidine kinase and not phosphorylated in uninfected cells –> few adverse effects.
Triphosphate formed by cellular enzymes.
Preferentially inhibit viral DNA polymerase by chain termination.

Clinical Use:
HSV and VZV. Weak activity against EBV.
No activity against CMV.
Used for HSV- induced mucocutaneous and genital lesions as well as for encephalitis.
Prophylaxis in immunocompromised patients.
Only affects actively replicating viruses and has no effect on latent infections of HSV and VZV.
For herpes zoster, use famciclovir.

Adverse Effects:
Thrombotic thrombocytopenic purpura
Gastrointestinal symptoms (e.g., nausea)
↑ Transaminases

Mechanism of Resistance:
Mutated viral thymidine kinase.

150
Q

Famciclovir

A

Prodrug of penciclovir with greater oral bioavailability

Mechanism:
Guanosine analogs.
Monophosphorylated by HSV/VZV thymidine kinase and not phosphorylated in uninfected cells –> few adverse effects.
Triphosphate formed by cellular enzymes.
Preferentially inhibit viral DNA polymerase by chain termination.

Clinical Use:
HSV and VZV. Weak activity against EBV.
No activity against CMV.
Used for HSV- induced mucocutaneous and genital lesions as well as for encephalitis.
Prophylaxis in immunocompromised patients.
Only affects actively replicating viruses and has no effect on latent infections of HSV and VZV.
For herpes zoster, use famciclovir.

Adverse Effects:
Thrombotic thrombocytopenic purpura
Gastrointestinal symptoms (e.g., nausea)
↑ Transaminases

Mechanism of Resistance:
Mutated viral thymidine kinase.

151
Q

Ganciclovir Mechanism

A
  • Guanosine analog (nucleoside analog)
  • Phosphorylation to 5’ monophosphate by the CMV-coded UL97 kinase (serine/threonine kinase) → further phosphorylation to triphosphate by cellular kinases
    Incorportaion of the phosphorylated drug into the replicating viral DNA strand → inhibition of viral DNA polymerase → termination of viral DNA synthesis
  • Lower selectivity than acyclovir and penciclovir → inhibition of host cells’ DNA replication → higher toxicity
152
Q

Valganciclovir

A

Prodrug of ganciclovir with greater oral availability

Mechanism:
Guanosine analog (nucleoside analog)
Phosphorylation to 5' monophosphate by the CMV-coded UL97 kinase (serine/threonine kinase)  → further phosphorylation to triphosphate by cellular kinases
Incorportaion of the phosphorylated drug into the replicating viral DNA strand → inhibition of viral DNA polymerase → termination of viral DNA synthesis
Lower selectivity than acyclovir and penciclovir → inhibition of host cells' DNA replication → higher toxicity

Clinical Use:
Systemic treatment of choice for CMV retinitis in immunocompromised patients (e.g., patients with AIDS)
CMV prophylaxis in transplant recipients

Adverse Effects:
Myelotoxicity (pancytopenia (additive effect when administered with NRTIs))
Nephrotoxicity
Gastrointestinal symptoms (e.g., nausea)
CNS
Headache
Confusion
Paresthesias

Mechanism of Resistance:
Mutation of viral UL97 kinase

153
Q

Foscarnet (pyrophosphate analog)

A

Mechanism:
Inhibition of DNA/RNA polymerase and HIV reverse transcriptase → direct inhibition of viral DNA polymerases via attachment to the pyrophosphate-binding site of the enzyme
Does not require activation by viral kinase

Clinical Use:
CMV retinitis in immunocompromised patients when ganciclovir fails
Acyclovir-resistant HSV.

Adverse Effects:
Nephrotoxicity
Gastrointestinal symptoms (e.g., nausea)
Headache
Confusion
Seizures due to electrolyte abnormalities (e.g., hypocalcemia, hypokalemia)
Paresthesias
Leukopenia
Neutropenia

Mechanism of Resistance:
Mutation of viral DNA polymerase

154
Q

Cidofovir

A

Mechanism:
Viral DNA polymerase inhibitor with a long half-life
Direct inhibition of viral DNA polymerases
Does not require activation by viral kinase

Clinical Use:
CMV retinitis in immunocompromised individuals (e.g., patients with AIDS)
Acyclovir-resistant HSV

Adverse Effects:
Nephrotoxicity (reduced with administration of probenecid and IV fluids)

Mechanism of Resistance:
Mutation of viral DNA polymerase

155
Q

Fomivirsen

A

Mechanism:
Antisense antiviral that blocks translation of viral mRNA (possesses a base sequence that is complementary to CMV mRNA and therefore blocks protein synthesis)

Clinical Use:
Local treatment of CMV retinitis in patients with AIDS (intraocular injection)

Adverse Effects:
Eye infections
Increase in intraocular pressure
Macular edema

156
Q

Pegylated interferon-α and interferon-α

A
Mechanism:
Antiviral and immunomodulatory effect via intercellular and intracellular mechanisms (interferons bind to cellular receptors via protein kinase R (PKR))
Inhibition of viral protein synthesis
Promotion of breakdown of viral RNA
Increased expression of MHC class I molecules

Clinical Use:
Monotherapy in acute hepatitis C and chronic active hepatitis B
As a combination treatment in chronic hepatitis C (triple therapy with sofosbuvir, ribavirin, and interferon-α)

Adverse Effects:
Flu-like symptoms
Bone marrow suppression
CNS
Depressive mood
Seizures
Induction of autoantibodies
Myopathy
Contraindications:
Decompensated cirrhosis
Psychiatric conditions
Pregnancy
Autoimmune conditions
Leukopenia or thrombocytopenia
157
Q

Tenofovir

A
Mechanism:
Nucleotide analog (adenosine analog): phosphorylated to triphosphate in hepatic cells (activation) → binds to viral DNA polymerase → causes premature termination of DNA transcription

Clinical Use:
Chronic active hepatitis B
HIV

Adverse Effects:
Nephrotoxicity (e.g., Fanconi syndrome)
Headache and abdominal pain
Disease exacerbation is possible.

158
Q

Adefovir

A
Mechanism:
Nucleotide analog (adenosine analog): phosphorylated to triphosphate in hepatic cells (activation) → binds to viral DNA polymerase → causes premature termination of DNA transcription
159
Q

Entecavir

A

Mechanism:
Nucleoside analog: inhibition of reverse transcriptase

Clinical Use:
Frst-line drug in hepatitis B infection because of its potent antiviral activity and its low drug resistance rate
HIV

Adverse Effects:
Occasional
-Gastrointestinal symptoms
-Fever
-Headache
Rare
-Vasculitides
-Neuropathies
-Neutropenia
-Lactic acidosis
160
Q

Lamivudine

A

Mechanism:
Nucleoside analog: inhibition of reverse transcriptase

Clinical Use:
Chronic active hepatitis B (third-line treatment due to high rate of resistance).
HIV

Adverse Effects:
Adverse Effects:
Occasional
-Gastrointestinal symptoms
-Fever
-Headache
Rare
-Vasculitides
-Neuropathies
-Neutropenia
-Lactic acidosis
161
Q

Telbivudine

A

Mechanism:
Nucleoside analog: inhibition of reverse transcriptase

Clinical Use:
Chronic active hepatitis B
HIV

Adverse Effects:
Occasional
-Gastrointestinal symptoms
-Fever
-Headache
Rare
-Vasculitides
-Neuropathies
-Neutropenia
-Lactic acidosis
162
Q

Glecaprevir

A

Mechanism:
NS3/4A protease inhibitors
Inhibition of NS3/4A (an HCV serine protease required for viral replication) → ↓ viral replication
Available and used in combination with NS5A inhibitor pibrentasvir.

Clinical Use:
Chronic hepatitis C infection (all genotypes)

Adverse Effects:
Headache
Fatigue
Nausea

163
Q

Grazoprevir

A

Mechanism:
NS3/4A protease inhibitors
Inhibition of NS3/4A (an HCV serine protease required for viral replication) → ↓ viral replication
Available and used in combination with NS5A inhibitor elbasvir.

Clinical Use:
Chronic hepatitis C infection (genotypes 1a, 1b, and 4)

Adverse Effects:
Headache
Fatigue
Nausea

164
Q

Paritaprevir

A

Mechanism:
NS3/4A protease inhibitors
Inhibition of NS3/4A (an HCV serine protease required for viral replication) → ↓ viral replication
Only available in combination with ombitasvir, ritonavir, and dasabuvir.

Clinical Use:
Chronic hepatitis C infection (genotype 1 or 4)

Adverse Effects:
Headache
Fatigue
Nausea
Insomnia
Asthenia
Pruritus
Allergic skin reactions
165
Q

Simeprevir

A

Mechanism:
NS3/4A protease inhibitors
Inhibition of NS3/4A (an HCV serine protease required for viral replication) → ↓ viral replication

Clinical Use:
Chronic hepatitis C infection (as part of triple antiviral combination, in addition to peginterferon-α and ribavirin)

Adverse Effects:
Photosensitivity
Rash
Fatigue
Headache
Abdominal pain
Diarrhea
166
Q

Voxilaprevir

A

Mechanism:
NS3/4A protease inhibitors
Inhibition of NS3/4A (an HCV serine protease required for viral replication) → ↓ viral replication
Available and used in combination with sofosbuvir and velpatasvir in patients with initial treatment failure.

Clinical Use:
Chronic hepatitis C infection (all genotypes)

Adverse Effects:
Headache
Fatigue
Diarrhea
Nausea
167
Q

Daclatasvir

A

Mechanism:
Non-nucleoside NS5A polymerase inhibitors
Exact mechanism of action is unknown
Inhibition of the viral NS5A phosphoprotein, which is essential for replication → prevention of HCV RNA replication

Clinical Use:
Chronic hepatitis C infection

Adverse Effects:
Headache
Fatigue
Diarrhea
Nausea
168
Q

Elbasvir

A

Mechanism:
Non-nucleoside NS5A polymerase inhibitors
Exact mechanism of action is unknown
Inhibition of the viral NS5A phosphoprotein, which is essential for replication → prevention of HCV RNA replication

Clinical Use:
Chronic hepatitis C infection

Adverse Effects:
Headache
Fatigue
Diarrhea
Nausea
169
Q

Ledipasvir

A

Mechanism:
Non-nucleoside NS5A polymerase inhibitors
Exact mechanism of action is unknown
Inhibition of the viral NS5A phosphoprotein, which is essential for replication → prevention of HCV RNA replication

Clinical Use:
Chronic hepatitis C infection

Adverse Effects:
Headache
Fatigue
Diarrhea
Nausea
170
Q

Ombitasvir

A

Mechanism:
Non-nucleoside NS5A polymerase inhibitors
Exact mechanism of action is unknown
Inhibition of the viral NS5A phosphoprotein, which is essential for replication → prevention of HCV RNA replication

Clinical Use:
Chronic hepatitis C infection

Adverse Effects:
Headache
Diarrhea
Fatigue
Nausea
Insomnia
Asthenia
Pruritus
Skin reactions
171
Q

Pibrentasvir

A

Mechanism:
Non-nucleoside NS5A polymerase inhibitors
Exact mechanism of action is unknown
Inhibition of the viral NS5A phosphoprotein, which is essential for replication → prevention of HCV RNA replication

Clinical Use:
Chronic hepatitis C infection

Adverse Effects:
Headache
Fatigue
Nausea
Diarrhea
172
Q

Velpatasvir

A

Mechanism:
Non-nucleoside NS5A polymerase inhibitors
Exact mechanism of action is unknown
Inhibition of the viral NS5A phosphoprotein, which is essential for replication → prevention of HCV RNA replication

Clinical Use:
Chronic hepatitis C infection

Adverse Effects:
Headache
Diarrhea
Fatigue

173
Q

Dasabuvir

A

Mechanism:
NS5B inhibitors
Inhibition of NS5B (a RNA-dependent RNA polymerase) → chain termination and disruption of RNA synthesis → prevention of HCV replication

Clinical Use:
Chronic hepatitis C infection

Adverse Effects:
Fatigue
Headache
Insomnia
Pruritus
Asthenia
Nausea
174
Q

Baloxavir

A

Mechanism:
Inhibits the “cap snatching” endonuclease activity of the influenza virus RNA polymerase Žviral replication.

Clinical Use:
Treatment within 48 hours of symptom onset shortens duration of illness.

175
Q

Remdesivir

A

Mechanism:
Prodrug of an ATP analog.
The active metabolite inhibits viral RNA-dependent RNA polymerase and evades proofreading by viral exoribonuclease (ExoN)Ž –> decrease viral RNA production.

Clinical Use:
Recently approved for treatment of COVID-19 requiring hospitalization.

176
Q

Zidovudine (ZDV, formerly AZT)

A

Mechanism:
NRTIs act as nucleoside analogs → competitive blockage of nucleoside binding to reverse transcriptase → inhibition of formation of 3’ to 5’ phosphodiester linkages → termination of DNA chain → inhibition of RNA to DNA reverse transcription
Activation requires intracellular phosphorylation, thus their efficacy is reliant on kinase availability and activity, which varies depending on cell functionality and activation state.

Clinical Use:
HIV

Adverse Effects:
Bone marrow suppression → anemia (especially zidovudine), neutropenia
Mitochondrial toxicity (myopathy, peripheral neuropathy, hepatic steatosis, lactic acidosis)
HIV-associated lipodystrophy (Cushing-like syndrome): abnormal distribution of fat (loss of subcutaneous fatty tissue (lipoatrophy) in face and extremities; metabolic changes → impaired glucose tolerance, hyperlipoproteinemia (elevated triglycerides, elevated total cholesterol, lowered HDL); probable accumulation of fat in liver, muscles, abdomen, breasts, and neck (buffalo hump))

Resistance:
Caused by mutations in the gene that codes for reverse transcriptase (pol gene)

177
Q

Lamivudine (3TC)

A

Mechanism:
NRTIs act as nucleoside analogs → competitive blockage of nucleoside binding to reverse transcriptase → inhibition of formation of 3’ to 5’ phosphodiester linkages → termination of DNA chain → inhibition of RNA to DNA reverse transcription
Activation requires intracellular phosphorylation, thus their efficacy is reliant on kinase availability and activity, which varies depending on cell functionality and activation state.

Clinical Use:
HIV

Adverse Effects:
Bone marrow suppression → anemia (especially zidovudine), neutropenia
Mitochondrial toxicity (myopathy, peripheral neuropathy, hepatic steatosis, lactic acidosis)
HIV-associated lipodystrophy (Cushing-like syndrome): abnormal distribution of fat (loss of subcutaneous fatty tissue (lipoatrophy) in face and extremities; metabolic changes → impaired glucose tolerance, hyperlipoproteinemia (elevated triglycerides, elevated total cholesterol, lowered HDL); probable accumulation of fat in liver, muscles, abdomen, breasts, and neck (buffalo hump))

Resistance:
Caused by mutations in the gene that codes for reverse transcriptase (pol gene)

178
Q

Emtricitabine (FTC)

A

Mechanism:
NRTIs act as nucleoside analogs → competitive blockage of nucleoside binding to reverse transcriptase → inhibition of formation of 3’ to 5’ phosphodiester linkages → termination of DNA chain → inhibition of RNA to DNA reverse transcription
Activation requires intracellular phosphorylation, thus their efficacy is reliant on kinase availability and activity, which varies depending on cell functionality and activation state.

Clinical Use:
HIV

Adverse Effects:
Bone marrow suppression → anemia (especially zidovudine), neutropenia
Mitochondrial toxicity (myopathy, peripheral neuropathy, hepatic steatosis, lactic acidosis)
HIV-associated lipodystrophy (Cushing-like syndrome): abnormal distribution of fat (loss of subcutaneous fatty tissue (lipoatrophy) in face and extremities; metabolic changes → impaired glucose tolerance, hyperlipoproteinemia (elevated triglycerides, elevated total cholesterol, lowered HDL); probable accumulation of fat in liver, muscles, abdomen, breasts, and neck (buffalo hump))

Resistance:
Caused by mutations in the gene that codes for reverse transcriptase (pol gene)

179
Q

Abacavir (ABC)

A

Mechanism:
NRTIs act as nucleoside analogs → competitive blockage of nucleoside binding to reverse transcriptase → inhibition of formation of 3’ to 5’ phosphodiester linkages → termination of DNA chain → inhibition of RNA to DNA reverse transcription
Activation requires intracellular phosphorylation, thus their efficacy is reliant on kinase availability and activity, which varies depending on cell functionality and activation state.

Clinical Use:
HIV

Adverse Effects:
Bone marrow suppression → anemia (especially zidovudine), neutropenia
Mitochondrial toxicity (myopathy, peripheral neuropathy, hepatic steatosis, lactic acidosis)
Abacavir-related hypersensitivity syndrome (potentially life-threatening systemic reaction with fever and maculopapular rash). Abacavir should be avoided in HLA-B*57:01-positive patients
HIV-associated lipodystrophy (Cushing-like syndrome): abnormal distribution of fat (loss of subcutaneous fatty tissue (lipoatrophy) in face and extremities; metabolic changes → impaired glucose tolerance, hyperlipoproteinemia (elevated triglycerides, elevated total cholesterol, lowered HDL); probable accumulation of fat in liver, muscles, abdomen, breasts, and neck (buffalo hump))

Resistance:
Caused by mutations in the gene that codes for reverse transcriptase (pol gene)

180
Q

Stavudine (d4T)

A

Mechanism:
NRTIs act as nucleoside analogs → competitive blockage of nucleoside binding to reverse transcriptase → inhibition of formation of 3’ to 5’ phosphodiester linkages → termination of DNA chain → inhibition of RNA to DNA reverse transcription
Activation requires intracellular phosphorylation, thus their efficacy is reliant on kinase availability and activity, which varies depending on cell functionality and activation state.

Clinical Use:
HIV

Adverse Effects:
Bone marrow suppression → anemia (especially zidovudine), neutropenia
Mitochondrial toxicity (myopathy, peripheral neuropathy, hepatic steatosis, lactic acidosis)
Pancreatitis (didanosine/stavudine)
HIV-associated lipodystrophy (Cushing-like syndrome): abnormal distribution of fat (loss of subcutaneous fatty tissue (lipoatrophy) in face and extremities; metabolic changes → impaired glucose tolerance, hyperlipoproteinemia (elevated triglycerides, elevated total cholesterol, lowered HDL); probable accumulation of fat in liver, muscles, abdomen, breasts, and neck (buffalo hump))

Resistance:
Caused by mutations in the gene that codes for reverse transcriptase (pol gene)

181
Q

Didanosine (ddI)

A

Mechanism:
NRTIs act as nucleoside analogs → competitive blockage of nucleoside binding to reverse transcriptase → inhibition of formation of 3’ to 5’ phosphodiester linkages → termination of DNA chain → inhibition of RNA to DNA reverse transcription
Activation requires intracellular phosphorylation, thus their efficacy is reliant on kinase availability and activity, which varies depending on cell functionality and activation state.

Clinical Use:
HIV

Adverse Effects:
Bone marrow suppression → anemia (especially zidovudine), neutropenia
Mitochondrial toxicity (myopathy, peripheral neuropathy, hepatic steatosis, lactic acidosis)
Pancreatitis (didanosine/stavudine)
HIV-associated lipodystrophy (Cushing-like syndrome): abnormal distribution of fat (loss of subcutaneous fatty tissue (lipoatrophy) in face and extremities; metabolic changes → impaired glucose tolerance, hyperlipoproteinemia (elevated triglycerides, elevated total cholesterol, lowered HDL); probable accumulation of fat in liver, muscles, abdomen, breasts, and neck (buffalo hump))

Resistance:
Caused by mutations in the gene that codes for reverse transcriptase (pol gene)

182
Q

Tenofovir

A

Nucleotide analog, also called nucleotide reverse-transcriptase inhibitor (NtRTI)

Mechanism:
NRTIs act as nucleoside analogs → competitive blockage of nucleoside binding to reverse transcriptase → inhibition of formation of 3’ to 5’ phosphodiester linkages → termination of DNA chain → inhibition of RNA to DNA reverse transcription

Clinical Use:
HIV

Adverse Effects:
Bone marrow suppression → anemia (especially zidovudine), neutropenia
Mitochondrial toxicity (myopathy, peripheral neuropathy, hepatic steatosis, lactic acidosis)
HIV-associated lipodystrophy (Cushing-like syndrome): abnormal distribution of fat (loss of subcutaneous fatty tissue (lipoatrophy) in face and extremities; metabolic changes → impaired glucose tolerance, hyperlipoproteinemia (elevated triglycerides, elevated total cholesterol, lowered HDL); probable accumulation of fat in liver, muscles, abdomen, breasts, and neck (buffalo hump))
Tenofovir → nephrotoxicity (proximal tubular dysfunction, acute tubular necrosis, and subsequent acute kidney injury. Affected individuals show elevated creatinine, subnephrotic proteinuria, and glycosuria. Discontinuation lead to significant recovery and suggests reversibility of the toxic tenofovir effects.)

Resistance:
Caused by mutations in the gene that codes for reverse transcriptase (pol gene)

183
Q

Nevirapine

A

Mechanism:
Noncompetitive inhibitors of viral reverse transcriptase (NNRTIs) that bind to the reverse transcriptase at a different location than NRTIs
NNRTIs do not require intracellular phosphorylation for activation but are instead direct inhibitors.

Clinical Use:
HIV-1 (NOT HIV-2)

Adverse Effects:
Hepatotoxicity (nevirapine)
Hypersensitivity reactions: rash, Stevens-Johnson syndrome

184
Q

Efavirenz

A

Mechanism:
Noncompetitive inhibitors of viral reverse transcriptase (NNRTIs) that bind to the reverse transcriptase at a different location than NRTIs
NNRTIs do not require intracellular phosphorylation for activation but are instead direct inhibitors.

Clinical Use:
HIV-1 (NOT HIV-2)

Adverse Effects:
CNS toxicity and vivid or disturbing dreams (efavirenz)
Hypersensitivity reactions: rash, Stevens-Johnson syndrome

185
Q

Delavirdine

A

Mechanism:
Noncompetitive inhibitors of viral reverse transcriptase (NNRTIs) that bind to the reverse transcriptase at a different location than NRTIs
NNRTIs do not require intracellular phosphorylation for activation but are instead direct inhibitors.

Clinical Use:
HIV-1 (NOT HIV-2)

Adverse Effects:
Hypersensitivity reactions: rash, Stevens-Johnson syndrome

186
Q

HIV Protease Inhibitors

A

Indinavir, Ritonavir, Nelfinavir, Lopinavir, Atazanavir, Darunavir, Fosamprenavir, Saquinavir

Mechanism:
Inhibition of viral HIV-1 protease (encoded by pol gene) → inability to cleave viral mRNA into functional units → generation of impaired viral proteins → production of immature (noninfectious) virions

Clinical Use:
HIV

Adverse Effects:
GI upset (nausea, diarrhea)
Lipodystrophy and fat accumulation (are far more likely to cause fat accumulation than other ARTs)
Nephrolithiasis, crystal-induced nephropathy, and hematuria
Thrombocytopenia (indinavir)
Hyperglycemia (inhibition of insulin-dependent glucose transporters (GLUT 4) → peripheral insulin resistance → impaired glucose tolerance)

Ritonavir can be used to increase concentrations of other drugs since it inhibits cytochrome P450.

187
Q

Integrase Inhibitors

A

Raltegravir, Dolutegravir, Bictegravir, Elvitegravir

Mechanism:
Inhibition of the viral integrase → blockade of viral DNA integration into the host’s DNA → inhibition of viral replication

Clinical Use:
HIV

Adverse Effects:
↑ creatine kinase

188
Q

Enfuvirtide

A

Mechanism:
Inhibit binding or fusion of HIV virions with human cells.
Fusion inhibitor
Competitively binds to the viral protein gp41 and thereby prevents fusion with the cell

Clinical Use:
HIV-1 (NOT HIV-2)

Adverse Effects:
Skin irritation at the site of drug injection

189
Q

Maraviroc

A

Mechanism:
Blocks the CCR5 coreceptor on T cells and monocytes that is essential to cell infection for some HIV genotypes (R5 HIV-1) → inhibits gp120 interaction → prevents virus docking

Clinical Use:
HIV

Adverse Effects:
Hepatotoxicity, allergic reactions

190
Q

Cobicistat

A

Mechanism:
CYP3A4 inhibitor

Clinical Use:
Pharmacokinetic enhancement of antiretroviral therapy for HIV infection.
Decreases the breakdown of certain antiretroviral, including darunavir and atazanavir.

191
Q

Ganciclovir Clinical Use

A
  • Systemic treatment of choice for CMV retinitis in immunocompromised patients (e.g., patients with AIDS)
    CMV prophylaxis in transplant recipients
192
Q

Ganciclovir Adverse Effects

A
  • Myelotoxicity (pancytopenia (additive effect when administered with NRTIs))
  • Nephrotoxicity
  • Gastrointestinal symptoms (e.g., nausea)
  • Headache
  • Confusion
  • Paresthesias
193
Q

Ganciclovir Mechanism of Resistance

A

Mutation of viral UL97 kinase

194
Q

Azithromycin Clinical Use

A
Macrolide 
IV or oral form
Poor CNS penetration poor
Route of elimination → biliary
All macrolides (except azithromycin) have a short half-li fe.
  • Atypical pneumonia caused by Mycoplasma pneumonia, Legionella pneumophila, Chlamydophila pneumoniae
  • Bordetella pertussis
  • STIs caused by Chlamydia
  • Gram-positive cocci especially for the treatment of streptococcal infection in patients who are allergic to penicillin
  • Neisseria spp.
  • Second-line prophylaxis for N. meningitidis
  • Dual therapy with ceftriaxone for N. gonorrhoeae (azithromycin)
  • Mycobacterium avium (prophylaxis: azithromycin; treatment → azithromycin, clarithromycin)
  • Ureaplasma urealyticum
  • Babesia spp. (azithromycin in combination with atovaquone)
195
Q

Azithromycin Adverse Effects

A
Macrolide 
IV or oral form
Poor CNS penetration poor
Route of elimination: biliary
All macrolides (except azithromycin) have a short half-life.
  • Increased intestinal motility → GI upset
  • QT-interval prolongation, arrhythmia
  • Acute cholestatic hepatitis
  • Eosinophilia
  • Rash

Contraindications:
Azithromycin and clarithromycin are contraindicated in patients with hepatic failure (erythromycin should be used cautiously).

196
Q

Azithromycin Mechanism of Resistance

A
Macrolide 
IV or oral form
Poor CNS penetration poor
Route of elimination: biliary
All macrolides (except azithromycin) have a short half-life.

Methylation of the binding site of 23S rRNA prevents the macrolide from binding to rRNA.

197
Q

Carbapenems Adverse Effects

A

IV imipenem (combined with cilastatin)
IV meropenem
IV ertapenem
IV doripenem
Imipenem is always given with cilastatin, which inhibits human dehydropeptidase I (a renal tubular enzyme that breaks down imipenem).
Meropenem is stable to dehydropeptidase I.

  • Secondary fungal infections
  • CNS toxicity (can lower seizure threshold at high serum concentrations) (highest risk → imipenem; lowest risk → meropenem)
  • Gastrointestinal upset
  • Rash
  • Thrombophlebitis
198
Q

Carbapenems Mechanism of Resistance

A

IV imipenem (combined with cilastatin)
IV meropenem
IV ertapenem
IV doripenem
Imipenem is always given with cilastatin, which inhibits human dehydropeptidase I (a renal tubular enzyme that breaks down imipenem).
Meropenem is stable to dehydropeptidase I.

  • Inactivation by carbapenemase (type of β-lactamase produced by carbapenemase-producing Enterobacteriaceae (e.g., E. coli, K. pneumoniae, K. aerogenes))