antifungal agents Flashcards

1
Q

Candida species

A

C. albicans, C. glabrata, C. parapsilosis, C. tropicalis, C. krusei, C. guilliermondii, C. lusitaniae, C. auris* (multidrug-resistant)
Invasive candidiasis refers to severe forms of the disease (not uncomplicated candiduria and oropharyngeal or esophageal candidiasis)
Wide spectrum of invasive disease from catheter-associated infections to disseminated disease
Increased mortality if empiric antifungal therapy is delayed by 12 hours
Risk factors for invasive candidiasis: prolonged stay in ICU; central venous catheters; prolonged therapy with broad spectrum antibacterial agents; receipt of parenteral nutrition; recent surgery (especially abdominal); hemodialysis; diabetes mellitus

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

aspergillus

A

A mold that is ubiquitous in the environment
Primarily causes disease in immunocompromised hosts
Pulmonary system is most commonly affected (can occur anywhere though)
Definitive diagnosis requires a positive culture from a sterile site or histologic or radiologic evidence in a high-risk patient with negative cultures
Very difficult infection to treat

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

Cryptococcus neoformans

A

Encapsulated yeast that primarily affects the CNS and respiratory tract
More common in patients who are HIV-positive, who have received organ transplants, or high-dose corticosteroids
Cryptococcal meningitis – 30% mortality; residual neurologic deficits in 40%

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

Zygomycetes

A

Pathogens – Rhizopus, Absidia, Mucor
Most common infections: pulmonary system; paranasal sinuses with extension to the brain
Primary risk factors: diabetes mellitus; immunosuppression (with profound neutropenia); penetrating injuries from natural disasters (tornadoes, hurricanes, volcanic eruptions) or combat
Definitive diagnosis: tissue invasion on histopathologic exam with or without microbiologic evidence
Very poor prognosis

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

endemic (pathogenic) fungi

A

Pathogens – Histoplasma capsulatum, Blastomyces species, Coccidioides immitis
May cause disseminated disease via a primary pulmonary infection
May cause disease in normal host; higher risk in patients with suppressed cell-mediated immunity (HIV/AIDS, high-dose corticosteroids, TNF-α inhibitor therapy, transplant)
Geographic prevalence
-Histoplasma capsulatum – Midwestern states along Ohio and Mississippi river valleys; exposure to bat guano (cave exploration) or other large birds; demolition or construction
-Blastomyces species – Southeastern and Midwestern states along Ohio and Mississippi river valleys and Great Lakes region
-Coccidioides immitis/posadasii – cluster in southwestern United States (southern Arizona, southern California, southwest New Mexico, west Texas)

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

amphotericin B MOA

A

Binds to ergosterol and gets inserted into the fungal cytoplasmic membrane → disruption of the fungal cytoplasmic membrane → increased cell permeability → leakage of sodium/potassium/cellular constituents, loss of membrane potential, metabolic disruption → cell death
2. At low concentrations, K+ channel activity is increased
4
3. At higher concentrations, pores are formed in the membrane
4. Rapid onset of action
5. Resistance – decreased ergosterol biosynthesis, synthesis of alternative sterols
6. Pharmacodynamic parameter – Peak/MIC

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

amphotericin B SOA

A
Broad Spectrum
Candida species (not C. lusitaniae or C. auris)
Cryptococcus neoformans
Blastomyces dermatitidis
Histoplasma capsulatum
Coccidioides immitis
Aspergillus species** (reduced activity against A. terreus)
Mucor species
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

amphotericin B doexycholate PKs

A

Poorly absorbed after PO and IM administration – requires IV, intrathecal, intraventricular, topical, or bladder instillation
Rapidly and widely distributed into tissues – primarily deposits into reticuloendothelial tissues: liver, spleen, bone marrow
Poor penetration into CSF, even if meninges are inflamed (≈3% of serum)
Highly protein bound (> 90%) – mainly to β-lipoproteins
Not appreciably metabolized – ≈ 3% is excreted in the urine as active drug (most of the drug is degraded in situ)
Tri-exponential elimination – t½β 24-48 h; terminal elimination t½ 15 days (serum concentrations detected for at least 7 weeks after end of therapy)
Renal and hepatic impairment and hemodialysis do not affect drug clearance

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

Amphotericin B lipid-associated formulations PKs

A

All 3 formulations have different PK patterns
Achieve tissue concentrations from 90% lower to 500% higher than deoxycholate formulation
80-90% reduction in concentrations in the kidneys

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

amphotericin B clinical uses

A
Disseminated candidiasis
Cryptococcosis
Aspergillosis
Histoplasmosis
Blastomycosis
Coccidioidomycosis
Mucormycosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

amphotericin B doexycholate dosing and administration

A

Test dose of 0.1 mg/kg or 1 mg over 20 to 30 minutes (do not pre-medicate)
Increase total daily dose to 0.3-1.0 mg/kg/day (usual 0.4-0.6 mg/kg/day; up to 1.5 mg/kg for aspergillosis and mucor)
Generally infused over 4-6 hours
-May be infused over 1 hour in patients who tolerate slower infusions (fever more common early in therapy compared to 4-h infusion)
-Rapid infusions in patients with severely compromised renal function may develop acute hyperkalemia and ventricular fibrillation
-Data suggest significantly fewer adverse events if administered as continuous infusion over 24 hours (conflicts with PD parameter)
Intrathecal or intraventricular administration – 0.1 mg initially; increase gradually to a maximum of 0.5 mg every 48-72 hours

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

L-AmB dosing and admin

A

1.5-6 mg/kg daily, infused over 2 hours (if using an in-line filter, mean pore diameter must be ≥ 1 micron)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

ABLC dosing and admin

A

5 mg/kg daily, infused at 2.5 mg/kg/hr

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

ABCD dosing and admin

A

3-4 mg/kg daily, infused at 1 mg/kg/hour (administer test dose; infusion may be shortened to 2 hours if no reactions)

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

ampB deoxycholate adverse reactions

A

Infusion-related:
-Headache, fever, chills, arthralgias, myalgias, nausea, vomiting, tachypnea, hypotension
—Pretreat with acetaminophen or aspirin, antihistamines, meperidine, phenothiazines
—Hydrocortisone 25-50 mg may be added to the infusion container
—Tolerance develops over time
-Thrombophlebitis
—Slow infusion (4-6 hrs); rotate infusion sites; in-line filters (>0.22 micron)
—Heparin 500-1,000 units may be added to infusion bag
Non-infusion-related:
-Nephrotoxicity – dose-dependent increase in serum creatinine and BUN
—Mechanism – direct vasoconstriction of afferent renal arterioles resulting in cortical ischemia and decrease in GFR
—Permanent loss of renal function related to total dose
—Management and prevention of nephrotoxicity - Sodium repletion – appears to be useful in patients who are sodium depleted; 0.5-1 L normal saline over 30 minutes before AMB and 0.5-1 L normal saline after completion of infusion; Hydration and adjustment of daily dose
-Hypokalemia – supplementation of 100 mEq/day or more may be required
-Hypomagnesemia
-Bicarbonate wasting
-Anemia – normochromic, normocytic (drug-induced decrease in erythropoietin production); reversible
Intrathecal administration – peripheral nerve pain, headache, vomiting, paresthesias, paraplegia, seizures, difficulty voiding, impaired vision

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

ampB lipid-associated formulation adverse reactions

A

Less nephrotoxicity & infusion-related toxicities with lipid-associated formulations (except ABCD had similar infusion-related toxicities [fever, chills] compared to deoxycholate)
High-dose liposomal amphotericiln (7.5 mg/kg/d) associated with high nephrotoxicity rates
L-AmB – manage infusion-related reactions with diphenhydramine
Most studies show similar clinical outcomes to deoxycholate

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

ampB DIs

A

Nephrotoxic agents – potentiation of nephrotoxicity
Digoxin and skeletal muscle relaxants – potentiation secondary to hypokalemia
Flucytosine – increase therapeutic effect; potential for increased toxicity

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

flucytosine MOAs

A

5-FC enters fungal cell → deaminated to 5-FU → 5-FU gets incorporated into fungal RNA → interference with protein synthesis
5-FC enters fungal cell → metabolized to 5-fluorodeoxyuridine monophosphate → inhibits thymidylate synthetase → interferes with DNA synthesis

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

flucytosine SOA

A

Cryptococcus neoformans**

Candida species

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

flucytosine PKs

A
Absorption – well absorbed orally (> 90%)
Distribution
-Penetrates into CSF ( ≈75% of serum)
-Negligible protein binding (2 to 4%)
Metabolism/Excretion
-Small amount converted to 5-FU
-85 to 95% excreted unchanged in urine
-Normal half-life – 3 to 5 hours; anephric half-life – 85 hours
-Removed by HD & PD
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

flucytosine clinical use

A

Combination therapy with amphotericin B for cryptococcal meningitis

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

flucytosine adverse reactions

A

Gastrointestinal (6%) – nausea, vomiting, diarrhea, abdominal pain, enterocolitis
Hematologic – Bone marrow suppression (more common with serum concentrations > 100 µg/ml)

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

flucytosine dosing/monitoring

A

Normal Renal Function – 100 to 150 mg/kg/DAY* PO in 4 divided doses (25 to 37.5 mg/kg/DOSE* Q6H) – available in 250 mg and 500 mg capsules
Renal Dysfunction - must be adjusted
Monitoring
-Baseline: CBC, platelets, Scr, BUN
-Reduce dose in patients with bone marrow or GI toxicity
-TDM – trough concentrations 20-40 µg/ml recommended to prevent rapid selection of resistance in yeast

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

ketoconazole MOA

A

Inhibits synthesis of ergosterol via inhibition of the fungal cytochrome P-450 dependent enzyme lanosterol 14-α-demethylase
Blocks formation of ergosterol → damage to fungal cell membrane → disruption of structural integrity → leakage of cytoplasm → inhibition of growth (fungistatic)

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

ketoconazole SOA

A

Candida albicans
Cryptococcus neoformans
Histoplasma capsulatum
Dermatophytes (tinea)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

ketoconazole PKs

A

Absorption
-Well absorbed orally (F=75%); peak concentration 1 to 2 hours after oral dosing
-Absorption is inversely related to gastric pH***
Distribution
-Widely distributed throughout body (CSF penetration is negligible)
-Protein binding 95 to 99%
Metabolism/Elimination
-Extensively metabolized in the liver
-Major excretory route is enterohepatic – 85 to 90% excreted in bile & feces
-Biphasic elimination – half-life 2 hours (during the first 8-12 hours); 9 hours thereafter
-Dosage adjustment not needed in renal failure; not removed by HD or PD

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

ketoconazole clinical uses

A

Should never be used orally for first-line therapy of any fungal infection due to risk of hepatotoxicity and drug interactions
Chronic mucocutaneous candidiasis
Histoplasmosis – in immunocompetent hosts

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

ketoconazole adverse reactions

A

Gastrointestinal (20-30%) – N/V/D; anorexia; abdominal pain
Hepatotoxicity**
Endocrine**
-Dose-dependent inhibition of adrenal steroid & testosterone synthesis → gynecomastia, decreased libido, oligospermia, loss of hair
-Menstrual irregularities

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

ketoconazole DIs

A

very potent inhibitor of CYP3A4
Drugs affecting gastric pH – increase pH, decrease bioavailability
Anticoagulants – prolonged PT
Rifampin – decrease ketoconazole concentrations
Cyclosporine, tacrolimus, sirolimus – increased concentrations
Phenytoin – decreased clearance, increased concentrations

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

Itraconazole MOA

A

Inhibits synthesis of ergosterol via inhibition of the fungal cytochrome P-450 dependent enzyme lanosterol 14-α-demethylase
Blocks formation of ergosterol → damage to fungal cell membrane → disruption of structural integrity → leakage of cytoplasm → inhibition of growth (fungistatic)

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

itraconazole SOA

A
Aspergillus spp.**
Histoplasma capsulatum
Blastomyces dermatitidis
Candida species
Coccidioides immitis
Cryptococcus neoformans
Sporothrix schenckii
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

itraconazole PKs

A

Absorption
-Good absorption after oral administration (F ≈ 55%) – dependent on gastric acidity
-Capsules absorbed better when taken with meal or acidic cola beverage
-Oral solution better absorbed in fasting state
-Oral solution better absorbed than capsules – not interchangeable
Distribution
-Widely distributed throughout body tissues (poor CSF penetration)
-Highly protein bound (> 99%)
Metabolism/Elimination
-Metabolized predominantly by cytochrome P450 3A4 isoenzyme (inhibitor)
-Active metabolite – hydroxyitraconazole
-Long elimination half-life 20-36 hours
-Clearance decreases with higher doses due to saturable hepatic metabolism
-No dosage adjustment for renal dysfunction; not removed by HD or PD

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

Itraconazole clinical uses/indications

A

Histoplasmosis (first choice) – 200 mg PO daily (max dose 400 mg/day in 2 divided doses)
Aspergillosis (not first-line) – 200 to 400 mg PO daily
Blastomycosis – 200 mg PO daily (max dose 400 mg/day in 2 divided doses)
Life-threatening infections – administer oral loading dose of 200 mg three times daily for first 3 days of therapy, then 200-400 mg PO daily
Onychomycosis of toenails – 200 mg PO daily for 12 consecutive weeks
6. Onychomycosis of fingernails – 200 mg PO BID for 1 week; repeat 3 weeks later

34
Q

itraconazole therapeutic drug monitoring

A

Serum trough concentrations of 0.5-1 µg/ml associated with efficacy
Serum trough concentrations > 3 µg/ml associated with increased adverse events

35
Q

itraconazole adverse reactions

A

Hepatotoxicity
Congestive heart failure (boxed warning)*** – contraindicated in patients with ventricular dysfunction (CHF) or history of CHF → may cause negative inotropic effect; discontinue if signs/symptoms of CHF occur on therapy
Peripheral neuropathy
GI – nausea, abdominal discomfort
Rash
Contraindicated in pregnancy and nursing mothers

36
Q

itraconazole DIs

A

H2 antagonists, proton pump inhibitors, antacid – decrease itraconazole absorption
Drugs metabolized by CYP 3A4 – potent inhibitor
-Increased drug concentrations if administered with itraconazole: digoxin, quinidine, benzodiazepines, HMG-CoA reductase inhibitors (not pravastatin), rifabutin, cyclosporine, tacrolimus, sirolimus, protease inhibitors (ritonavir, indinavir, saquinavir)
-Decreased plasma concentrations of itraconazole: carbamazepine, phenytoin,
phenobarbital, rifampin, rifabutin, nevirapine
-Increased concentrations of itraconazole: clarithromycin, indinavir, ritonavir

37
Q

fluconazole MOA

A

Inhibits synthesis of ergosterol via inhibition of the fungal cytochrome P-450 dependent enzyme lanosterol 14-α-demethylase
Blocks formation of ergosterol → damage to fungal cell membrane → disruption of structural integrity → leakage of cytoplasm → inhibition of growth (fungistatic)

38
Q

fluconazole SOA

A

Candida species (less active vs. C. glabrata [up to 30% resistance]; not active vs. C. krusei)
C. albicans resistance reported (2.1-5.7%)
Cryptococcus neoformans
Histoplasma capsulatum
Blastomyces dermatitidis
Coccidioides immitis

39
Q

fluconazole PKs

A

Absorption
-Well absorbed orally (Tmax 1-2 hr); independent of gastric acidity; may be administered without regards to food
-Bioavailability > 90% & linear
Distribution
-Protein binding 11%
-Concentration in the CSF – 60 % of serum with uninflamed meninges; 80% of serum with inflamed meninges
Metabolism/Elimination
-Excreted unchanged in urine – 80% recovered unchanged in urine
-Elimination half-life ≈ 30 hours
-Dosage reduction required in renal insufficiency
-3 hour hemodialysis decreases serum concentrations by 50%

40
Q

fluconazole clinical uses/dosing

A

Noninvasive candidiasis
-Oropharyngeal – 200 mg on day 1, then 100 mg daily for 2 weeks
-Esophageal – 200 mg on day 1, then 100 mg daily for minimum of 3 weeks (max 400 mg if poor response)
-Vaginal – 150 mg x 1 dose
Invasive candidiasis (e.g., candidemia) – 800 mg loading dose, then 400 mg daily
Prophylaxis in patients undergoing bone marrow transplantation – 400 mg daily
Candida urinary tract infection – 100-200 mg daily
Cryptococcal meningitis
-Alternative to amphotericin B ± flucytosine – 400 mg daily for 10-12 weeks after CSF negative
-Maintenance therapy – 200 mg daily (may be discontinued if CD4 count > 200 cells/mm3 for 6 months if no signs/symptoms of meningitis)

41
Q

fluconazole adverse reactions

A

Headache, nausea, anorexia
QT prolongation, torsades de pointes
Elevation of hepatic transaminases – may be seen with higher doses

42
Q

fluconazole drug interactions

A

Potent inhibitory of CYP2C9

Moderate inhibitor of CYP3A4

43
Q

voriconazole MOA

A

Inhibits synthesis of ergosterol via inhibition of the fungal cytochrome P-450 dependent enzyme lanosterol 14-α-demethylase
Blocks formation of ergosterol → damage to fungal cell membrane → disruption of structural integrity → leakage of cytoplasm → inhibition of growth (fungistatic)

44
Q

voriconazole SOA

A
Aspergillus species** (including amphotericin and itraconazole-resistant strains)
Scedosporium apiospermum
Candida species (C. albicans, C. tropicalis, C. glabrata, C. parapsilosis. C. krusei)
C. glabrata resistance (0.1-18.4%)
Histoplasma capsulatum
Blastomyces dermatitidis
Cryptococcus neoformans
Fusarium species
45
Q

voriconazole PKs

A

Absorption
-Oral bioavailability ≈ 96% (not affected by H2 antagonists, PPI, antacids)
-Tablets and oral suspension should be taken 1 hour before or after meals
Distribution
-Extensive tissue distribution (Vd 4.6 L/kg)
-Protein binding ≈ 58%
Metabolism
-Significantly metabolized by cytochrome P450 isoenzymes (2C19, 2C9, 3A4)*
-2C19 – genetic polymorphism (15-20% of Asian population, 3-5% of African American
and white populations expected to be poor metabolizers)
-Poor metabolizers – 4-fold higher AUC
Metabolism is saturable – pharmacokinetics are non-linear
* (2.5-fold increase in AUC with 300 mg PO q12h compared to 200 mg PO q12h; 2.3-fold increase in AUC with 4 mg/kg IV q12h compared to 3 mg/kg IV q12h)
Elimination
-less than 2% of dose excreted unchanged in urine
-Half-life 6 hours (dose-dependent)
-No difference in Cmax and AUC in mild to severe renal dysfunction
—NO dosage adjustment necessary for ORAL* dosing
—In patients with Clcr 30-50 ml/min, accumulation of the intravenous vehicle, sulfobutyl ether beta-cyclodextrin sodium (SBECD)
—AVOID IV VORICONAZOLE IF CLCR under 50 ML/MIN**

46
Q

voriconazole clinical uses/dosing

A

Oral and IV maintenance doses do not produce the same voriconazole exposures. A 200 mg oral dose provides an exposure similar to a 3 mg/kg IV dose; 300 mg oral dose provides an exposure similar to a 4 mg/kg IV dose.
If inadequate response, increase oral maintenance dose to 300 mg q12h
Adults who weigh less than 40 kg should receive half of the oral maintenance dose.
Oral maintenance dose should be decreased by 50% in mild-to-moderate hepatic cirrhosis

47
Q

Voriconazole AEs

A
Visual disturbances** (20-30%) – blurred vision, color changes, photophobia, photopsia, hallucinations
Elevated liver function tests (13%; up to 33% with high dose) – aminotransferases, alkaline phosphatase, bilirubin
CNS – cognitive difficulties, memory loss with high dose
Peripheral neuropathy (9%)
Phototoxic skin reactions – may progress to squamous cell carcinoma (risk factors: prolonged treatment [> 6 months], strong sun exposure, ongoing immunosuppression or post-transplant phase, advanced age, light-colored skin)
Diffuse, painful periostitis secondary to excess fluoride (voriconazole contains fluorine) – associated with prolonged therapy; non-specific pain in hands, finger swelling, diffuse musculoskeletal pain
48
Q

Voriconazole therapeutic drug monitoring

A

Trough concentrations > 1 µg/ml associated with clinical response
Trough concentrations > 5.5 µg/ml associated with higher incidence of CNS adverse events

49
Q

Voriconazole drug interactions

A

Effect of other drugs on voriconazole – sig. decrease in voriconazole exposure
-Rifampin, rifabutin, carbamazepine (potent CYP 450 inducer) – co-administration contraindicated
Effect of voriconazole on other drugs – increased exposure due to inhibition of metabolism
-Sirolimus, terfenadine, astemizole, cisapride, pimozide, quinidine – contraindicated
-Cyclosporine, tacrolimus, warfarin, statins, benzodiazepines, calcium channel blockers, sulfonylureas, vinca alkaloids – careful monitoring and dosage adjustment required
-Protease inhibitors – ritonavir, saquiavir, amprenavir (no effect with indinavir)

50
Q

Posaconazole MOA

A

blocks synthesis of ergosterol – same as other triazole agents

51
Q

Posaconazole SOA

A
Candida species (less active against C. glabrata)
Aspergillus species**
Cryptococcus neoformans
Histoplasma capsulatum
Mucor species
Coccidioides species
52
Q

Posaconazole PKs

A

Absorption is affected by gastric pH (decreased absorption with PPIs)
Oral suspension: Tmax 3-4 hours
Delayed release tablets: Tmax 4-5 hours; absolute bioavailability 54%
Better absorbed when administered with food or acidic carbonated beverage and when administered in divided doses (200 mg q6h > 400 mg q12h > 800 mg q24h)
Protein binding 98%, primarily to albumin
≈ 17% metabolized by glucuronidation (not CYP450)
14% of administered dose recovered in urine; 71% in feces
Half-life 26-35 hours
IV formulation contains cyclodextrin – AVOID if CRCL less than 50 ml/min***

53
Q

posaconazole clinical uses

A

Prophylaxis of invasive Aspergillus and Candida infections in immunocompromised
patients (HSCT; hematologic malignancies with prolonged neutropenia)
Oropharyngeal candidiasis
Oropharyngeal candidiasis refractory to fluconazole and/or itraconazole
May be used as salvage therapy for treatment of aspergillosis or Mucor infections

54
Q

posaconazole therapeutic drug monitoring

A

Trough concentrations < 0.7 µg/ml associated with higher incidence of breakthrough fungal infections during prophylaxis
Trough concentrations > 1 µg/ml associated with treatment response for invasive aspergillosis
Currently no trough concentration associated with toxicity

55
Q

posaconazole DIs

A

strong inhibitor of CYP3A4
Cyclosporine – reduce dose to 3/4 of original dose
Tacrolimus – reduce dose to 1/3 of original dose
Sirolimus – contraindicated
Midazolam – dose reduction should be considered
Phenytoin – decreased Cmax and AUC of posaconazole
Rifabutin – decreased Cmax and AUC of posaconazole
PPIs and H2 antagonists – decreased Cmax and AUC of Posaconazole

56
Q

Posaconazole AEs

A

Nausea, vomiting, abdominal pain, diarrhea – 10-30%
Elevated AST, ALT, bilirubin, hypokalemia
Rash

57
Q

Isavuconazole MOA

A

Isavuconazonium sulfate is the prodrug of isavuconazole – metabolized by esterases in blood
Inhibits synthesis of ergosterol (key component of fungal cell membrane) by inhibiting lanosterol 14-alpha-demethylase (responsible for conversion of lanosterol to ergosterol) → depletion of ergosterol weakens cell membrane structure and function

58
Q

Isavuconazole SOA

A

Aspergillus specis*** (A. flavus, A, fumigatus, A. niger)
Mucor
Rhizopus

59
Q

Isavuconazole PKs

A

Linear pharmacokinetics up to 600 mg of isavuconazole
Well absorbed orally – absolute bioavailability 98%; Tmax 2-4 hours
IV and oral capsules are bioequivalent
May be administered orally with or without food
Very large volume of distribution ≈ 450 L
Highly protein bound (99% to albumin)
Plasma half-life 130 hours
Renal excretion is less than 1% – no dosage adjustment for renal impairment or ESRD
IV formulation does not contain cyclodextrin
Not removed by hemodialysis
No dosage adjustment in mild or moderate hepatic impairment

60
Q

Isavuconazole clinical uses and dosing

A

Invasive aspergillosis in patients ≥ 18 years of age
Invasive mucormycosis in patients ≥ 18 years of age
IV formulation must be administered through an in-line filter (pore size 0.2-1.2 microns) over at least 1 hour in 250 mL to reduce risk of infusion-related reactions (insoluble drug particulates may form after reconstitution)
Oral capsules should be swallowed whole; do not chew, crush, dissolve, or open the capsules

61
Q

Isavuconazole AEs

A

Most frequent: nausea (26%), vomiting (25%), diarrhea (22%), headache (17%)
Hepatic (16%) – increased LFTs (AST, ALT, alkaline phosphatase, total bilirubin); generally reversible and discontinuation of isavuconazole not usually required; severe hepatic events have been reported; evaluate LFTs at the start and during the course of therapy
Infusion-related reactions (hypotension, dyspnea, chills, dizziness, paresthesias, hypoesthesia) – discontinue infusion if occurs
Hypokalemia (14%)
Hypersensitivity and severe skin reactions (anaphylaxis, Stevens Johnson syndrome)

62
Q

Isavuconazole DIs

A

Isavuconazole is a substrate for CYP3A4, a moderate inhibitor of CYP3A4, CYP2C8, CYP2C9, CYP2C19, and CYP2D6, and a mild inhibitor of P-glycoprotein
CYP3A4 inhibitors/inducers
CYP3A4 substrates – isavuconazole increases concentrations of cyclosporine, tacrolimus, sirolimus, midazolam
P-glycoprotein substrates (digoxin)

63
Q

Isavuconazole CIs

A

Co-administration with strong CYP3A4 inhibitors (e.g., ketoconazole, high-dose ritonavir) → significant increase plasma isavuconazole concentrations
Co-administration with strong CYP3A4 inducers (rifampin, carbamazepine, St. John’s wort, long acting barbiturates) → significantly decrease plasma isavuconazole concentrations
Patients with familial short QT syndrome – isavuconazole shortens QT interval

64
Q

Echinocandin antifungal agents

A

caspofungin, micafungin, anidulafungin

65
Q

caspofungin MOA

A

Glucan synthesis inhibitor leading to noncompetitive inhibition 1,3-β-glucan, an integral polysaccharide component of fungal cell wall**
Together with chitin, glucan is responsible for the cell wall’s strength and shape – important in maintaining the osmotic integrity of the fungal cell wall***

66
Q

caspofungin SOA

A

Aspergillus species
Candida species, including azole-resistant strains (C. glabrata resistance up to 12%)
Less active vs. C. parapsilosis
Limited activity against Histoplasma capsulatum, Cryptococcus neoformans, Fusarium, and Mucor

67
Q

caspofungun PKs

A

Poor oral bioavailability – administered by IV infusion
Plasma concentration decline in polyphasic manner
-β-phase – half-life 9 to 11 hours
-γ-phase – half-life 40 to 50 hours
Highly protein bound to albumin (97%)
Slowly metabolized by hydrolysis and N-acetylation
≈ 1.4% excreted unchanged in urine
No dosage adjustment for renal insufficiency or mild hepatic impairment
Not removed by hemodialysis

68
Q

Caspofungin indications/dosing

A

Candidemia – 70 mg loading dose on day 1, then 50 mg q24h
Esophageal candidiasis – 50 mg q24h
Empiric therapy of presumed fungal infections in febrile neutropenia – 70 mg loading dose on day 1, then 50 mg q24h
Invasive aspergillosis in patients who are refractory to or intolerant to other therapies - 70 mg loading dose on day 1, then 50 mg q24h (may use 70 mg daily)
Reduce daily dose to 35 mg in severe hepatic dysfunction

69
Q

caspofungin DIs

A

Does not induce or inhibit CYP450 system; poor substrate for CYP450
Tacrolimus – decreases AUC, Cmax, and C12h of tacrolimus
Cyclosporine – increase caspofungin AUC by 35%
Inducers of drug clearance (efavirenz, nelfinavir, nevirapine, phenytoin, rifampin, carbamazepine) – may reduce caspofungin concentrations, may need to increase daily dose to 70 mg

70
Q

caspofungin adverse reactions

A

Histamine-mediated symptoms – rash, facial swelling, pruritus, flushing
Fever
Phlebitis at infusion site
Nausea, vomiting, headache
Laboratory – increased liver transaminases, decreased potassium, eosinophilia, increase in urine protein and RBC’s, decreased hemoglobin/hematocrit

71
Q

Micafungin MOA

A

inhibits synthesis of 1,3-β-glucan

72
Q

Micafungin SOA

A

Candida species, including azole-resistant strains (C. glabrata resistance up to 12%)
Less active vs. C. parapsilosis
Aspergillus species
Limited activity against Histoplasma capsulatum, Cryptococcus neoformans, Fusarium, and Mucor***

73
Q

Micafungin PKs

A

Not absorbed orally – give IV
Protein binding 99%
Half-life 14-17 hours
Metabolized by the liver – no dosage adjustment for renal dysfunction
Less than 15% eliminated renally as unchanged drug

74
Q

Micafungin clinical uses/dosing

A

Oropharyngeal and esophageal candidiasis – 50 to 100 mg daily
Candidemia – 100 mg daily
Aspergillosis – 150 mg daily
Prophylaxis of Candida infections in hematopoietic stem cell transplantation (HSCT) – 50 mg daily

75
Q

micafungin DIs

A

not metabolized via CYP450 pathways

76
Q

micafungin AEs

A
Hyperbilirubinemia
Nausea
Diarrhea
Eosinophilia
Rash, pruritus, urticarial – rare
77
Q

anidulafungin MOA

A

inhibits synthesis of 1,3-β-glucan

78
Q

anidulafungin SOA

A

Candida species, including azole-resistant strains (C. glabrata resistance up to 12%)
Less active vs. C. parapsilosis
Aspergillus species
Limited activity against Histoplasma capsulatum, Cryptococcus neoformans,
Fusarium, and Mucor**

79
Q

anidulafungin PKs

A

Not absorbed orally – must be administered IV
Protein binding – 84%
Long half-life: 26.5 hours in patients with fungal infections
Not metabolized or renally eliminated → undergoes slow chemical degradation
No dosage adjustments required for renal or hepatic dysfunction

80
Q

anidulafungin clinical uses/dosing

A

Candidemia and other Candida infections (peritonitis, intra-abdominal abscess) – 200 mg loading dose on day 1, then 100 mg daily for 14 days after last positive culture
Esophageal candidiasis – 200 mg loading dose on day 1, then 100 mg daily for at least 14 days (at least 7 days after resolution of symptoms) - Significantly higher relapse rate than fluconazole

81
Q

anidulafungin DIs

A

not a substrate, inducers, or inhibitor of CYP450

82
Q

anidulafungin AEs

A
overall, well-tolerated
Histamine-mediated symptoms – rash, urticaria, flushing, pruritus, hypotension
Diarrhea
Increased LFTs
Hypokalemia