Medine.co.uk

Voriconazole 200 Mg Film-Coated Tablets

Informations for option: Voriconazole 200 Mg Film-Coated Tablets, show other option
Document: spc-doc_PL 04569-1388 change

SUMMARY OF PRODUCT CHARACTERISTICS

1 NAME OF THE MEDICINAL PRODUCT

Voriconazole 200 mg film-coated tablets

2    QUALITATIVE AND QUANTITATIVE COMPOSITION

Each tablet contains 200 mg voriconazole.

Excipient with known effect: each tablet contains 256.4 mg lactose monohydrate (tablet core and film-coating).

For the full list of excipients, see section 6.1.

3    PHARMACEUTICAL FORM

Voriconazole 200 mg film-coated tablets: white to off-white film-coated, capsule shaped, biconvex tablet debossed with “M164” on one side of the tablet and blank on the other side. Dimensions: 15.5 mm x 7.5 mm.

4    CLINICAL PARTICULARS

4.1    Therapeutic indications

Voriconazole is a broad spectrum, triazole antifungal agent and is indicated in adults and children aged 2 years and above as follows:

•    Treatment of invasive aspergillosis.

•    Treatment of candidaemia in non-neutropenic patients.

•    Treatment of fluconazole-resistant serious invasive Candida infections (including C. krusei).

   Treatment of serious fungal infections caused by Scedosporium spp. and Fusarium spp.

Voriconazole should be administered primarily to patients with progressive, possibly life-threatening infections.

4.2 Posology and method of administration

Posology

Electrolyte disturbances such as hypokalaemia, hypomagnesaemia and hypocalcaemia should be monitored and corrected, if necessary, prior to initiation and during voriconazole therapy (see section 4.4).

Adults and adolescents (12 to 14 years and >50 kg; 15 to 17 years regardless of body weight)

Therapy must be initiated with the specified loading dose regimen of Voriconazole to achieve plasma concentrations on Day 1 that are close to steady state. On the basis of the high oral bioavailability (96 %; see section 5.2), switching between intravenous and oral administration is appropriate when clinically indicated.

Detailed information on dosage recommendations is provided in the following table:

Oral

Patients 40 kg and above

Patients less than 40 kg*

Loading dose regimen

(first 24 hours)

400 mg every 12 hours

200 mg every 12 hours

Maintenance

dose

200 mg twice daily

100 mg twice daily

(after first 24 hours)

*This also applies to patients aged 15 years and older.

Dose adjustment

If patient response is inadequate, the maintenance dose may be increased to 300 mg twice daily for oral administration. For patients less than 40 kg the oral dose may be increased to 150 mg twice daily.

If patient is unable to tolerate treatment at a higher dose, reduce the oral dose by 50 mg steps to the 200 mg twice daily (or 100 mg twice daily for patients less than 40 kg) maintenance dose.

Phenytoin may be coadministered with voriconazole if the maintenance dose of voriconazole is increased from 200 mg to 400 mg orally, twice daily (100 mg to 200 mg orally, twice daily in patients less than 40 kg), see sections 4.4 and 4.5.

The combination of voriconazole with rifabutin should, if possible be avoided. However, if the combination is strictly needed, the maintenance dose of voriconazole may be increased from 200 mg to 350 mg orally, twice daily (100 mg to 200 mg orally, twice daily in patients less than 40 kg), see sections 4.4 and 4.5.

Efavirenz may be coadministered with voriconazole if the maintenance dose of voriconazole is increased to 400 mg every 12 hours and the efavirenz dose is reduced by 50 %, i.e. to 300 mg once daily. When treatment with voriconazole is stopped, the initial dosage of efavirenz should be restored (see sections 4.4 and 4.5).

Children (2 to <12 years) and young adolescents with low body weight (12 to 14 years and <50 kg).

Voriconazole should be dosed as children as these young adolescents may metabolize voriconazole more similarly to children than to adults.

The recommended dosing regimen is as follows:

Oral

Loading Dose Regimen (first 24 hours)

Not recommended

Maintenance Dose (after first 24 hours)

9 mg/kg twice daily (a maximum dose of 350 mg twice daily)

Note: Based on a population pharmacokinetic analysis in 112 immunocompromised paediatric patients aged 2 to <12 years and 26 immunocompromised adolescents aged 12 to <17 years.

It is recommended to initiate the therapy with intravenous regimen, and oral regimen should be considered only after there is a significant clinical improvement. It should be noted that an 8 mg/kg intravenous dose will provide voriconazole exposure approximately 2-fold higher than a 9 mg/kg oral dose.

These oral dose recommendations for children are based on studies in which voriconazole was administered as the powder for oral suspension. Bioequivalence between the powder for oral suspension and tablets has not been investigated in a paediatric population. Considering the assumed limited gastro-enteric transit time in paediatric patients, the absorption of tablets may be different in paediatric compared to adult patients. It is therefore recommended to use the oral suspension formulation in children aged 2 to <12.

Use in paediatric patients aged 2 to <12 years with hepatic or renal insufficiency has not been studied (see sections 4.8 and 5.2).

All other adolescents (12 to 14 years and >50 kg; 15 to 17 years regardless of body weight)

Voriconazole should be dosed as adults.

Dose adjustment

If patient response is inadequate, the dose may be increased by 1 mg/kg steps (or by 50 mg steps if the maximum oral dose of 350 mg was used initially). If patient is unable to tolerate treatment, reduce the dose by 1 mg/kg steps (or by 50 mg steps if the maximum oral dose of 350 mg was used initially).

Duration of treatment

Treatment duration should be as short as possible depending on the patient’s clinical and mycological response (see section 4.4).

For long term treatment greater than 6 months, a careful assessment of the benefit-risk balance should be considered (see sections 4.4 and 5.1).

Elderly

No dose adjustment is necessary for elderly patients (see section 5.2).

Renal impairment

The pharmacokinetics of orally administered voriconazole are not affected by renal impairment. Therefore, no adjustment is necessary for oral dosing for patients with mild to severe renal impairment (see section 5.2).

Voriconazole is haemodialysed with a clearance of 121 ml/min. A four hour haemodialysis session does not remove a sufficient amount of voriconazole to warrant dose adjustment.

Hepatic impairment

It is recommended that the standard loading dose regimens be used but that the maintenance dose be halved in patients with mild to moderate hepatic cirrhosis (Child-Pugh A and B) receiving voriconazole (see section 5.2).

Voriconazole has not been studied in patients with severe chronic hepatic cirrhosis (Child-Pugh C).

There is limited data on the safety of voriconazole in patients with abnormal Liver Function Tests (aspartate transaminase (AST), alanine transaminase (ALT), alkaline phosphatase (AP), or total bilirubin >5 times the upper limit of normal).

Voriconazole has been associated with elevations in liver function tests and clinical signs of liver damage, such as jaundice, and must only be used in patients with severe hepatic impairment if the benefit outweighs the potential risk. Patients with hepatic impairment must be carefully monitored for drug toxicity (see section 4.8).

Paediatric population

The safety and efficacy of voriconazole in children below 2 years has not been established. Currently available data are described in sections 4.8 and 5.1 but no recommendation on a posology can be made.

Method of administration

Voriconazole 50 mg and 200 mg film-coated tablets are to be taken at least one hour before, or one hour following a meal.

4.3 Contraindications

Hypersensitivity to the active substance or to any of the excipients (listed in section 6.1).

Co-administration with CYP3A4 substrates, terfenadine, astemizole, cisapride, pimozide or quinidine since increased plasma concentrations of these medicinal products can lead to QTc prolongation and rare occurrences of torsades de pointes (see section 4.5).

Co-administration with rifampicin, carbamazepine, mephobarbital and phenobarbital since these medicinal products are likely to decrease plasma voriconazole concentrations significantly (see section 4.5).

Co-administration with high dose ritonavir (400 mg and above twice daily) because ritonavir significantly decreases plasma voriconazole concentrations in healthy subjects at this dose. (see section 4.5, for lower doses see section 4.4).

Coadministration of standard doses of voriconazole with efavirenz doses of 400 mg once daily or higher is contraindicated because efavirenz significantly decreases plasma voriconazole concentrations in healthy subjects at these doses. Voriconazole also significantly increases efavirenz plasma concentrations (see section 4.5, for lower doses see section 4.4).

Co-administration with ergot alkaloids (ergotamine, dihydroergotamine), which are CYP3A4 substrates, since increased plasma concentrations of these medicinal products can lead to ergotism (see section 4.5).

Co-administration with sirolimus, since voriconazole is likely to increase plasma concentrations of sirolimus significantly (see section 4.5).

Coadministration with St John's Wort (see section 4.5).

4.4 Special warnings and precautions for use

Hypersensitivity

Caution should be used in prescribing Voriconazole to patients with hypersensitivity to other azoles (see also section 4.8).

Cardiovascular

Voriconazole has been associated with QT interval prolongation. There have been rare cases of torsades de pointes in patients taking voriconazole who had risk factors, such as history of cardiotoxic chemotherapy, cardiomyopathy, hypokalaemia and concomitant medicinal products that may have been contributory. Voriconazole should be administered with caution to patients with potentially proarrhythmic conditions, such as:

•    Congenital or acquired QT-prolongation

•    Cardiomyopathy, in particular when heart failure is present

•    Sinus bradycardia

•    Existing symptomatic arrhythmias

•    Concomitant medicinal product that is known to prolong QT interval.

Electrolyte disturbances such as hypokalaemia, hypomagnesaemia and hypocalcaemia should be monitored and corrected, if necessary, prior to initiation and during voriconazole therapy (see section 4.2). A study has been conducted in healthy volunteers which examined the effect on QT interval of single doses of voriconazole up to 4 times the usual daily dose. No subject experienced an interval exceeding the potentially clinically relevant threshold of 500 msec (see section 5.1).

Hepatic toxicity

In clinical trials, there have been uncommon cases of serious hepatic reactions during treatment with voriconazole (including clinical hepatitis, cholestasis and fulminant hepatic failure, including fatalities). Instances of hepatic reactions were noted to occur primarily in patients with serious underlying medical conditions (predominantly haematological malignancy). Transient hepatic reactions, including hepatitis and jaundice, have occurred among patients with no other identifiable risk factors. Liver dysfunction has usually been reversible on discontinuation of therapy (see section 4.8).

Monitoring of hepatic function

Patients receiving voriconazole must be carefully monitored for hepatic toxicity. Clinical management should include laboratory evaluation of hepatic function (specifically AST and ALT) at the initiation of treatment with voriconazole and at least weekly for the first month of treatment. Treatment duration should be as short as possible ; however, if based on the benefit-risk assessment the treatment is continued (see section 4.2), monitoring frequency can be reduced to monthly if there are no changes in the Liver Function Tests.

If the liver function tests become markedly elevated, voriconazole should be discontinued, unless the medical judgment of the risk- benefit of the treatment for the patient justifies continued use.

Monitoring of hepatic function should be carried out in both children and adults.

Visual adverse reactions

There have been reports of prolonged visual adverse reactions, including blurred vision, optic neuritis and papilloedema (see section 4.8).

Renal adverse reactions

Acute renal failure has been observed in severely ill patients undergoing treatment with voriconazole. Patients being treated with voriconazole are likely to be treated concomitantly with nephrotoxic medicinal products and have concurrent conditions that may result in decreased renal function (see section 4.8).

Monitoring of renal function

Patients should be monitored for the development of abnormal renal function. This should include laboratory evaluation, particularly serum creatinine.

Monitoring of pancreatic function

Patients, especially children, with risk factors for acute pancreatitis (e.g. recent chemotherapy, haematopoietic stem cell transplantation (HSCT)), should be monitored closely during Voriconazole treatment. Monitoring of serum amylase or lipase may be considered in this clinical situation.

Dermatological adverse reactions

Patients have rarely developed exfoliative cutaneous reactions, such as Stevens-Johnson syndrome, during treatment with voriconazole. If patients develop a rash they should be monitored closely and Voriconazole discontinued if lesions progress.

In addition voriconazole has been associated with phototoxicity and pseudoporphyria. It is recommended that all patients, including children, avoid intense or prolonged exposure to direct sunlight during Voriconazole treatment and use measures such as protective clothing and sunscreen with high sun protection factor (SPF).

Long-term treatment

The following severe adverse events have been reported in relation with long-term voriconazole treatment, physicians should therefore consider the need to limit the exposure to voriconazole (see sections 4.2 and 5.1):

Squamous cell carcinoma of the skin has been reported in patients, some of whom have reported prior phototoxic reactions. If phototoxic reactions occur, multidisciplinary advice should be sought and the patient should be referred to a dermatologist. Voriconazole discontinuation should be considered. Dermatologic evaluation should be performed on a systematic and regular basis, whenever Voriconazole is continued despite the occurrence of phototoxicity-related lesions, to allow early detection and management of premalignant lesions. Voriconazole should be discontinued if premalignant skin lesions or squamous cell carcinoma are identified.

Non-infectious periostitis with elevated fluoride and alkaline phosphatase levels has been reported in transplant patients. If a patient develops skeletal pain and radiologic findings compatible with periostitis Voriconazole discontinuation should be considered after multidisciplinary advice.

Paediatric population

Safety and effectiveness in paediatric subjects below the age of two years has not been established (see sections 4.8 and 5.1). Voriconazole is indicated for paediatric patients aged two years or older. Hepatic function should be monitored in both children and adults. Oral bioavailability may be limited in paediatric patients aged 2 to <12 years with malabsorption and very low body weight for age. In that case, intravenous voriconazole administration is recommended.

Phenytoin (CYP2C9 substrate and potent CYP450 inducer)

Careful monitoring of phenytoin levels is recommended when phenytoin is coadministered with voriconazole. Concomitant use of voriconazole and phenytoin should be avoided unless the benefit outweighs the risk (see section 4.5).

Efavirenz (CYP450 inducer; CYP3A4 inhibitor and substrate)

When voriconazole is coadministered with efavirenz the dose of voriconazole should be increased to 400 mg every 12 hours and the dose of efavirenz should be decreased to 300 mg every 24 hours (see sections 4.2, 4.3 and 4.5).

Rifabutin (Potent CYP450 inducer)

Careful monitoring of full blood counts and adverse reactions to rifabutin (e.g. uveitis) is recommended when rifabutin is co-administered with voriconazole. Concomitant use of voriconazole and rifabutin should be avoided unless the benefit outweighs the risk (see section 4.5).

Ritonavir (potent CYP450 inducer; CYP3A4 inhibitor and substrate)

Coadministration of voriconazole and low dose ritonavir (100 mg twice daily) should be avoided unless an assessment of the benefit/risk to the patient justifies the use of voriconazole (see sections 4.3 and 4.5).

Everolimus (CYP3A4 substrate, P-gp substrate)

Co-administration of voriconazole with everolimus is not recommended because voriconazole is expected to significantly increase everolimus concentrations.

Currently there are insufficient data to allow dosing recommendations in this situation (see section 4.5).

Methadone (CYP3A4 substrate)

Frequent monitoring for adverse reactions and toxicity related to methadone, including QTc prolongation, is recommended when co-administered with voriconazole since methadone levels increased following co-administration of voriconazole. Dose reduction of methadone may be needed (see section 4.5).

Short acting opiates (CYP3A4 substrate)

Reduction in the dose of alfentanil, fentanyl and other short acting opiates similar in structure to alfentanil and metabolised by CYP3A4 (e.g. sufentanil) should be considered when co-administered with voriconazole (see section 4.5). As the half-life of alfentanil is prolonged in a 4-fold manner when alfentanil is co-administered with voriconazole and in an independent published study, concomitant use of voriconazole with fentanyl resulted in an increase in the mean AUC0.^> of fentanyl, frequent monitoring for opiate-associated adverse reactions (including a longer respiratory monitoring period) may be necessary.

Long acting opiates (CYP3A4 substrate)

Reduction in the dose of oxycodone and other long-acting opiates metabolized by CYP3A4 (e.g., hydrocodone) should be considered when coadministered with voriconazole. Frequent monitoring for opiate-associated adverse reactions may be necessary (see section 4.5).

Fluconazole (CYP2C9, CYP2C19 and CYP3A4 inhibitor)

Coadministration of oral voriconazole and oral fluconazole resulted in a significant increase in Cmax and AUCr of voriconazole in healthy subjects. The reduced dose and/or frequency of voriconazole and fluconazole that would eliminate this effect have not been established. Monitoring for voriconazole associated adverse reactions is recommended if voriconazole is used sequentially after fluconazole (see section 4.5).

Voriconazole 50 mg and 200 mg film-coated Tablets contain lactose. Patients with rare hereditary problems of galactose intolerance, the Lapp lactase deficiency or glucose-galactose malabsorption should not take this medicine.

4.5 Interaction with other medicinal products and other forms of interaction

Voriconazole is metabolised by, and inhibits the activity of, cytochrome P450 isoenzymes, CYP2C19, CYP2C9, and CYP3A4. Inhibitors or inducers of these isoenzymes may increase or decrease voriconazole plasma concentrations, respectively, and there is potential for voriconazole to increase the plasma concentrations of substances metabolised by these CYP450 isoenzymes.

Unless otherwise specified, drug interaction studies have been performed in healthy adult male subjects using multiple dosing to steady state with oral voriconazole at 200 mg twice daily (BID). These results are relevant to other populations and routes of administration.

Voriconazole should be administered with caution in patients with concomitant medication that is known to prolong QT interval. When there is also a potential for voriconazole to increase the plasma concentrations of substances metabolised by CYP3A4 isoenzymes (certain antihistamines, quinidine, cisapride, pimozide) coadministration is contraindicated (see below and section 4.3).

Interaction table

Interactions between voriconazole and other medicinal products are listed in the table below (once daily as “QD”, twice daily as “BID”, three times daily as “TID” and not determined as “ND”). The direction of the arrow for each pharmacokinetic parameter is based on the 90% confidence interval of the geometric mean ratio being within («■), below (j.) or above (|) the 80-125% range. The asterisk (*) indicates a two-way interaction. AUCx, AUCt and AUC0.^> represent area under the curve over a dosing interval, from time zero to the time with detectable measurement and from time zero to infinity, respectively.

The interactions in the table are presented in the following order: contraindications, those requiring dose adjustment and careful clinical and/or biological monitoring, and finally those that have no significant pharmacokinetic interaction but may be of clinical interest in this therapeutic field.

Medicinal product

[Mechanism of Interaction]

Interaction

Geometric mean changes (%)

Recommendations concerning coadministration

Astemizole, cisapride, pimozide, quinidine and terfenadine

[CYP3A4 substrates]

Although not studied, increased plasma concentrations of these medicinal products can lead to QTc prolongation and rare occurrences of torsades de pointes.

Contraindicated (see section 4.3)

Carbamazepine and long-acting barbiturates (e.g., phenobarbital, mephobarbital)

[potent CYP450 inducers]

Although not studied, carbamazepine and long-acting barbiturates are likely to significantly decrease plasma voriconazole concentrations.

Contraindicated (see section 4.3)

Efavirenz (a non-nucleoside reverse transcriptase inhibitor)

[CYP450 inducer; CYP3A4 inhibitor and substrate]

Efavirenz 400 mg QD, coadministered with voriconazole 200 mg BID)

Efavirenz Cmax | 38% Efavirenz AUCt | 44% Voriconazole Cmax l 61% Voriconazole AUCt 77%

Use of standard doses of voriconazole with efavirenz doses of 400 mg QD or higher is contraindicated (see section 4.3).

Efavirenz 300 mg QD, coadministered with voriconazole 400 mg BID

Compared to efavirenz 600 mg QD,

Efavirenz Cmax <->■ Efavirenz AUCt | 17%

Compared to voriconazole 200 mg BID,

Voriconazole Cmax | 23%

Voriconazole AUCt l 7%

Voriconazole may be co-administered with efavirenz if the voriconazole maintenance dose is increased to 400 mg BID and the efavirenz dose is decreased to 300 mg QD. When voriconazole treatment is stopped, the initial dose of efavirenz should be restored (see section 4.2 and 4.4).

Ergot alkaloids (e.g., ergotamine and dihydroergotamine)

[CYP3A4 substrates]

Although not studied, voriconazole is likely to increase the plasma concentrations of ergot alkaloids and lead to ergotism.

Contraindicated (see section 4.3)

Rifabutin

[potent CYP450 inducer]

300 mg QD

Voriconazole Cmax l 69% Voriconazole AUCt 78%

Concomitant use of voriconazole and rifabutin should be avoided unless the benefit outweighs the risk.

300 mg QD (co-administered with voriconazole 350 mg BID)*

Compared to voriconazole 200 mg BID,

Voriconazole Cmax l 4%

Voriconazole AUCt l 32%

The maintenance dose of voriconazole may be increased to 5 mg/kg intravenously BID or from 200 mg

to 350 mg orally BID

300 mg QD (co-administered with voriconazole 400 mg BID)*

Rifabutin Cmax | 195%

Rifabutin AUCt | 331%

Compared to voriconazole 200 mg BID,

(100 mg to 200 mg orally BID in patients less than 40 kg) (see section 4.2).

Voriconazole Cmax | 104%

Careful monitoring of full blood counts and

Voriconazole AUCt | 87%

adverse reactions to rifabutin (e.g., uveitis) is recommended when rifabutin is coadministered with voriconazole.

Rifampicin (600 mg QD)

Voriconazole Cmax j, 93%

Contraindicated (see

[potent CYP450 inducer]

Voriconazole AUCt j, 96%

section 4.3)

Ritonavir (protease inhibitor)

[potent CYP450 inducer; CYP3A4 inhibitor and substrate]

Ritonavir Cmax and AUCt <->■

Co-administration of

High dose (400 mg BID)

Voriconazole Cmax j, 66%

voriconazole and high doses of ritonavir

Voriconazole AUCt), 82%

(400 mg and above BID) is

contraindicated (see section 4.3).

Ritonavir Cmax j, 25%

Low dose (100 mg BID)*

Ritonavir AUCt j.13% Voriconazole Cmax j, 24%

Co-administration of voriconazole and low

Voriconazole AUCt j, 39%

dose ritonavir (100 mg BID) should be avoided, unless an assessment of the benefit/risk to the patient justifies the use of voriconazole.

St John's Wort

[CYP450 inducer; P-gp inducer]

In an independent published

Contraindicated (see

300 mg TID (co-administered

study,

section 4.3)

with voriconazole 400 mg single dose)

Voriconazole AUC0-co l 59%

Everolimus

[CYP3A4 substrate, P-gP substrate]


Although not studied, voriconazole is likely to significantly increase the plasma concentrations of everolimus.


Co-administration of voriconazole with everolimus is not recommended because voriconazole is expected to significantly increase everolimus concentrations (see section 4.4).


Fluconazole (200 mg QD)

[CYP2C9, CYP2C19 and CYP3A4 inhibitor]


Voriconazole Cmax | 57% Voriconazole AUCt | 79% Fluconazole Cmax ND Fluconazole AUCt ND


The reduced dose and/or frequency of voriconazole and fluconazole that would eliminate this effect have not been established. Monitoring for voriconazole-associated adverse reactions is recommended if voriconazole is used sequentially after fluconazole.


Phenytoin


[CYP2C9 substrate and potent CYP450 inducer]


300 mg QD


300 mg QD (co-administered with voriconazole 400 mg BID)*


Voriconazole Cmax l 49% Voriconazole AUCt 69%


Phenytoin Cmax | 67%

Phenytoin AUCt | 81%

Compared to voriconazole 200 mg BID,

Voriconazole Cmax | 34%

Voriconazole AUCt | 39%


Concomitant use of voriconazole and phenytoin should be avoided unless the benefit outweighs the risk. Careful monitoring of phenytoin plasma levels is recommended.


Phenytoin may be coadministered with voriconazole if the maintenance dose of voriconazole is increased to 5 mg/kg IV BID or from 200 mg to 400 mg oral BID, (100 mg to 200 mg oral BID in patients less than 40


kg) (see section 4.2).


Anticoagulants


Warfarin (30 mg single dose, co- administered with 300 mg BID voriconazole)

[CYP2C9 substrate]


Other oral coumarins (e.g.,

phenprocoumon,

acenocoumarol)

[CYP2C9 and CYP3A4 substrates]


Benzodiazepines (e.g., midazolam, triazolam, alprazolam)

[CYP3A4 substrates]


Immunosuppressants


[CYP3A4 substrates]


Sirolimus (2 mg single dose)


Ciclosporin (In stable renal transplant recipients receiving chronic ciclosporin therapy)


Tacrolimus (0.1 mg/kg single dose)


Maximum increase in prothrombin time was approximately 2-fold


Although not studied, voriconazole may increase the plasma concentrations of coumarins that may cause an increase in prothrombin time.


Although not studied clinically, voriconazole is likely to increase the plasma concentrations of benzodiazepines that are metabolised by CYP3A4 and lead to a prolonged sedative effect.


In an independent published study, Sirolimus Cmax | 6.6-fold

Sirolimus AUC0_®| 11-fold


Ciclosporin Cmax | 13% Ciclosporin AUCr | 70%


Tacrolimus Cmax | 117% Tacrolimus AUCt | 221%


Close monitoring of prothrombin time or other suitable anticoagulation tests is recommended, and the dose of anticoagulants should be adjusted accordingly.


Dose reduction of benzodiazepines should be considered.


Co-administration of voriconazole and sirolimus is contraindicated (see section 4.3).


When initiating voriconazole in patients already on ciclosporin it is recommended that the ciclosporin dose be halved and ciclosporin level carefully monitored. Increased ciclosporin levels have been associated with nephrotoxicity. When voriconazole is discontinued, ciclosporin levels must be carefully monitored and the


dose increased as necessary.

When initiating voriconazole in patients already on tacrolimus, it is recommended that the tacrolimus dose be reduced to a third of the original dose and tacrolimus level carefully monitored. Increased tacrolimus levels have been associated with nephrotoxicity. When voriconazole is discontinued, tacrolimus levels must be carefully monitored and the dose increased as necessary.

Long Acting Opiates

[CYP3A4 substrates]

Oxycodone (10 mg single dose)

In an independent published study,

Oxycodone Cmax| 1.7-fold

Oxycodone AUC0^> T 3.6-fold

Dose reduction in oxycodone and other long-acting opiates metabolized by CYP3A4 (e.g., hydrocodone) should be considered. Frequent monitoring for opiate-associated adverse reactions may be necessary.

Methadone (32-100 mg QD)

[CYP3A4 substrate]

R-methadone (active) Cmax | 31%

R-methadone (active)

AUCt T 47%

S-methadone Cmax | 65%

S-methadone AUCr | 103%

Frequent monitoring for adverse reactions and toxicity related to methadone, including QT prolongation, is recommended. Dose reduction of methadone may be needed.

Non-Steroidal AntiInflammatory Drugs (NSAIDs)

[CYP2C9 substrates]

Frequent monitoring

Ibuprofen (400 mg single dose)

Diclofenac (50 mg single dose)

S-Ibuprofen Cmax | 20% S-Ibuprofen AUC0^> t 100%

Diclofenac Cmax | 114% Diclofenac AUC0^> t 78%

for adverse reactions and toxicity related to NSAIDs is recommended. Dose reduction of NSAIDs may be needed.

Omeprazole (40 mg QD)*

[CYP2C19 inhibitor; CYP2C19 and CYP3A4 substrate]

Omeprazole Cmax| 116% Omeprazole AUCt | 280% Voriconazole Cmax | 15% Voriconazole AUCt | 41%

Other proton pump inhibitors that are CYP2C19 substrates may also be inhibited by voriconazole and may result in increased plasma concentrations of these medicinal products.

No dose adjustment of voriconazole is recommended.

When initiating voriconazole in patients already receiving omeprazole doses of 40 mg or above, it is

recommended that the omeprazole dose be halved.

Oral Contraceptives*

[CYP3A4 substrate; CYP2C19 inhibitor]

Norethisterone/ethinylestradiol (1 mg/0.035 mg QD)

Ethinylestradiol Cmax | 36%

Ethinylestradiol AUCt | 61%

Norethisterone Cmax | 15% Norethisterone AUCt | 53% Voriconazole Cmax | 14% Voriconazole AUCt | 46%

Monitoring for adverse reactions related to oral contraceptives, in addition to those for voriconazole, is recommended.

Short Acting Opiates

[CYP3A4 substrates]

Alfentanil (20 gg/kg single dose, with concomitant naloxone)

Fentanyl (5 gg/kg single dose)

In an independent published study,

Alfentanil AUC0^> t 6-fold

In an independent published study,

Fentanyl AUC0^> t 134-fold

Dose reduction of alfentanil, fentanyl and other short acting opiates similar in structure to alfentanil and metabolised by CYP3A4 (e.g., sufentanil) should be considered. Extended and frequent monitoring for respiratory depression and other opiate-associated adverse reactions is recommended.

Statins (e.g., lovastatin)

Although not studied clinically, voriconazole is

Dose reduction of statins should be

[CYP3A4 substrates]

likely to increase the plasma concentrations of statins that are metabolised by CYP3A4 and could lead to rhabdomyolysis.

considered.

Sulfonylureas (e.g., tolbutamide, glipizide, glyburide)

[CYP2C9 substrates]

Although not studied, voriconazole is likely to increase the plasma concentrations of sulfonylureas and cause hypoglycaemia.

Careful monitoring of blood glucose is recommended. Dose reduction of sulfonylureas should be considered.

Vinca Alkaloids (e.g., vincristine and vinblastine)

[CYP3A4 substrates]

Although not studied, voriconazole is likely to increase the plasma concentrations of vinca alkaloids and lead to neurotoxicity.

Dose reduction of vinca alkaloids should be considered.

Other HIV Protease Inhibitors (e.g., saquinavir, amprenavir and nelfinavir)

[CYP3A4 substrates and inhibitors]

Not studied clinically. In vitro studies show that voriconazole may inhibit the metabolism of HIV protease inhibitors and the metabolism of voriconazole may also be inhibited by HIV protease inhibitors.

Careful monitoring for any occurrence of drug toxicity and/or lack of efficacy, and dose adjustment may be needed.

Other Non-Nucleoside Reverse Transcriptase Inhibitors (NNRTIs) (e.g., delavirdine, nevirapine)

[CYP3A4 substrates, inhibitors or CYP450 inducers]

Not studied clinically. In vitro studies show that the metabolism of voriconazole may be inhibited by NNRTIs and voriconazole may inhibit the metabolism of NNRTIs.

The findings of the effect of efavirenz on voriconazole suggest that the metabolism of voriconazole may be induced by a NNRTI.

Careful monitoring for any occurrence of drug toxicity and/or lack of efficacy, and dose adjustment may be needed.

Cimetidine (400 mg BID)

[non-specific CYP450 inhibitor and increases gastric pH]

Voriconazole Cmax | 18% Voriconazole AUCt | 23%

No dose adjustment

Digoxin (0.25 mg QD)

[P-gp substrate]

Digoxin Cmax ^ Digoxin AUCt

No dose adjustment

Indinavir (800 mg TID)

[CYP3A4 inhibitor and

Indinavir Cmax <->■ Indinavir AUCt <->■

No dose adjustment

substrate]

Voriconazole Cmax <->■ Voriconazole AUCt <->■

Macrolide antibiotics

Erythromycin (1 g BID)

[CYP3A4 inhibitor]

Azithromycin (500 mg QD)

Voriconazole Cmax and AUCt <->■

Voriconazole Cmax and AUCt <->■

The effect of voriconazole on either erythromycin or azithromycin is unknown.

No dose adjustment

Mycophenolic acid (1 g single dose)

[UDP-glucuronyl transferase substrate]

Mycophenolic acid Cmax <->■ Mycophenolic acid AUCt <->■

No dose adjustment

Prednisolone (60 mg single dose)

[CYP3A4 substrate]

Prednisolone Cmax | 11% Prednisolone AUC0-co t 34%

No dose adjustment

Ranitidine (150 mg BID)

[increases gastric pH]

Voriconazole Cmax and AUCt <->■

No dose adjustment

4.6 Fertility, pregnancy and lactation

Pregnancy

There are no adequate data on the use of voriconazole in pregnant women available.

Studies in animals have shown reproductive toxicity (see section 5.3). The potential risk for humans is unknown.

Voriconazole must not be used during pregnancy unless the benefit to the mother clearly outweighs the potential risk to the foetus.

Women of child-bearing potential

Women of child-bearing potential must always use effective contraception during treatment.

Breastfeeding

The excretion of voriconazole into breast milk has not been investigated. Breastfeeding must be stopped on initiation of treatment with Voriconazole.

Fertility

In an animal study, no impairment of fertility was demonstrated in male and female rats (see section 5.3).

4.7 Effects on ability to drive and use machines

Voriconazole has a moderate influence on the ability to drive and use machines. It may cause transient and reversible changes to vision, including blurring, altered/enhanced visual perception and/or photophobia. Patients must avoid potentially hazardous tasks, such as driving or operating machinery while experiencing these symptoms.

4.8 Undesirable effects

a. Summary of the safety profile:

The safety profile of voriconazole is based on an integrated safety database of more than 2,000 subjects (1,655 patients in therapeutic trials). This represents a heterogeneous population, containing patients with haematological malignancy, HIV-infected patients with oesophageal candidiasis and refractory fungal infections, nonneutropenic patients with candidaemia or aspergillosis and healthy volunteers. Five hundred and sixty one (561) patients had a duration of voriconazole therapy of greater than 12 weeks, with 136 patients receiving voriconazole for over 6 months.

The most commonly reported adverse reactions were visual disturbances, pyrexia, rash, vomiting, nausea, diarrhoea, headache, peripheral oedema and abdominal pain.

The severity of the adverse reactions was generally mild to moderate. No clinically significant differences were seen when the safety data were analysed by age, race, or gender.

b. Tabulated list of adverse reactions:

In the table below, since the majority of the studies were of an open nature all causality adverse reactions, by system organ class and frequency are listed.

Frequency categories are expressed as: Very common (^ 1/10); Common (^ 1/100 to <1/10); Uncommon 1/1,000 to <1/100); Rare (^ 1/10,000 to <1/1,000); Very rare (<1/10,000); Not known (cannot be estimated from the available data)

Within each frequency grouping, undesirable effects are presented in order of decreasing seriousness.

Undesirable effects reported in subjects receiving voriconazole

System Organ Class

Adverse drug reactions

Infections and infestation

Common

Gastroenteritis, influenza-like illness

Rare

Pseudomembranous colitis

Blood and lymphatic system disorders

Common

Pancytopenia, bone marrow depression, leukopenia, thrombocytopenia, anaemia, purpura

Uncommon

Disseminated intravascular coagulation, agranulocytosis, lymphadenopathy, eosinophilia

Immune system disorders

Common

Sinusitis

Uncommon

Anaphylactoid reaction, hypersensitivity

Endocrine disorders

Uncommon

Adrenal insufficiency

Rare

Hyperthyroidism, hypothyroidism

Metabolism and nutrition disorders

Common

Hypoglycaemia, hypokalaemia

Uncommon

Hyponatraemia*

Psychiatric disorders

Common

Depression, hallucination, anxiety

Rare

Insomnia

Nervous system disorders

Very common

Headache

Common

Dizziness, confusional state, tremor, agitation, paraesthesia

Uncommon

Brain oedema, ataxia, vertigo, hypoaesthesia, hypertonia

Rare

Convulsion, encephalopathy, Guillain-Barre syndrome,

extrapyramidal symptoms, peripheral neuropathy

Eye disorders

Very common

Visual disturbances (including blurred vision (see section 4.4), chromotopsia and photophobia)

Uncommon

Papilloedema (see section 4.4), optic nerve disorder (including optic neuritis, see section 4.4), nystagmus, scleritis, blepharitis, diplopia

Rare

Optic atrophy, retinal haemorrhage, oculogyration, corneal opacity

Ear and labyrinth disorders

Rare

Hypoacusis, tinnitus

Cardiac disorders

Uncommon

Ventricular fibrillation, ventricular arrhythmia, syncope, supraventricular arrhythmia, supraventricular tachycardia, tachycardia, bradycardia, atrial arrhythmia

Rare

Torsades de pointes, ventricular tachycardia, atrioventricular complete block, bundle branch block, nodal rhythm

Vascular disorders

Common

Thrombophlebitis, hypotension, phlebitis

Rare

Lymphangitis

Respiratory, thoracic and mediastinal disorders

Common

Acute respiratory distress syndrome, pulmonary oedema, respiratory distress, chest pain

Gastrointestinal disorders

Very common

Abdominal pain, nausea, vomiting, diarrhoea

Uncommon

Pancreatitis, peritonitis, duodenitis, gingivitis, glossitis, swollen tongue, dyspepsia, constipation, dysgeusia

Hepatobiliary disorders

Common

Jaundice, cholestatic jaundice

Uncommon

Hepatic failure, hepatitis, hepatomegaly, cholecystitis, cholelithiasis

Rare

Hepatic coma

Skin and subcutaneous tissue disorders

Very common

Rash

Common

Exfoliative dermatitis, face oedema, phototoxic reaction, maculo-papular rash, macular rash, papular rash, cheilitis, pruritus, alopecia, erythema

Uncommon

Fixed drug eruption, Stevens-Johnson syndrome, angioneurotic oedema, allergic dermatitis, urticaria, drug hypersensitivity, psoriasis, eczema

Rare

Toxic epidermal necrolysis, erythema multiforme, discoid lupus erythematosis, pseudoporphyria

Not known

Squamous cell carcinoma

Musculoskeletal and connective tissue disorders

Common

Back pain

Uncommon

Arthritis

Not known

Periostitis

Renal and urinary disorders

Common

Renal failure acute, haematuria

Uncommon

Proteinuria, nephritis

Rare

Renal tubular necrosis

General disorders and administration site conditions

Very common

Pyrexia, oedema peripheral

Common

Injection site reaction / inflammation, chills, asthenia

Investigations

Common

Elevated liver function tests (including AST, ALT, alkaline phosphatase, gamma-glutamyl transpeptidase [GGT], lactate dehydrogenase [LDH], bilirubin), blood creatinine increased

Uncommon

Electrocardiogram QT corrected interval prolonged, blood urea increased, blood cholesterol increased

*Undesirable events identified during post-approval use

Visual disturbances

In clinical trials, voriconazole treatment-related visual disturbances were very common. In these studies, short-term as well as long-term treatment, approximately 30 % of subjects experienced altered/enhanced visual perception, blurred vision, colour vision change or photophobia. These visual disturbances were transient and fully reversible, with the majority spontaneously resolving within 60 minutes and no clinically significant long-term visual effects were observed. There was evidence of attenuation with repeated doses of voriconazole. The visual disturbances were generally mild, rarely resulted in discontinuation and were not associated with longterm sequelae. Visual disturbances may be associated with higher plasma concentrations and/or doses.

The mechanism of action is unknown, although the site of action is most likely to be within the retina. In a study in healthy volunteers investigating the impact of voriconazole on retinal function, voriconazole caused a decrease in the electroretinogram (ERG) waveform amplitude. The ERG measures electrical currents in the retina. The ERG changes did not progress over 29 days of treatment and were fully reversible on withdrawal of voriconazole.

There have been post-marketing reports of prolonged visual adverse events (see section 4.4).

Dermatological reactions

Dermatological reactions were common in patients treated with voriconazole in clinical trials, but these patients had serious underlying diseases and were receiving multiple concomitant medicinal products. The majority of rashes were of mild to moderate severity. Patients have rarely developed serious cutaneous reactions, including Stevens-Johnson syndrome, toxic epidermal necrolysis and erythema multiforme during treatment with voriconazole.

If a patient develops a rash they should be monitored closely and Voriconazole discontinued if lesions progress. Photosensitivity reactions have been reported, especially during long-term therapy (see section 4.4).

There have been reports of squamous cell carcinoma of the skin in patients treated with voriconazole for long periods of time; the mechanism has not been established (see section 4.4).

Liver function tests

The overall incidence of clinically significant transaminase abnormalities in the voriconazole clinical programme was 13.4 % (200/1493) of subjects treated with voriconazole. Liver function test abnormalities may be associated with higher plasma concentrations and/or doses. The majority of abnormal liver function tests either resolved during treatment without dose adjustment or following dose adjustment, including discontinuation of therapy.

Voriconazole has been infrequently associated with cases of serious hepatic toxicity in patients with other serious underlying conditions. This includes cases of jaundice, and rare cases of hepatitis and hepatic failure leading to death (see section 4.4).

d. Paediatric population

The safety of voriconazole was investigated in 285 paediatric patients aged 2 to <12 years who were treated with voriconazole in pharmacokinetic studies (127 paediatric patients) and in compassionate use programs (158 paediatric patients). The adverse reaction profile of these 285 paediatric patients was similar to that in adults. Postmarketing data suggest there might be a higher occurrence of skin reactions (especially erythema) in the paediatric population compared to adults. In the 22 patients less than 2 years old who received voriconazole in a compassionate use programme, the following adverse reactions (for which a relationship to voriconazole could not be excluded) were reported: photosensitivity reaction (1), arrhythmia (1), pancreatitis (1), blood bilirubin increased (1), hepatic enzymes increased (1), rash (1) and papilloedema (1). There have been post-marketing reports of pancreatitis in paediatric patients.

Reporting of suspected adverse reactions

Reporting suspected adverse reactions after authorisation of the medicinal product is important. It allows continued monitoring of the benefit/risk balance of the medicinal product. Healthcare professionals are asked to report any suspected adverse reactions via Yellow Card Scheme at: www.mhra.gov.uk/yellowcard.

4.9 Overdose

In clinical trials there were 3 cases of accidental overdose. All occurred in paediatric patients, who received up to five times the recommended intravenous dose of voriconazole. A single adverse reaction of photophobia of 10 minutes duration was reported.

There is no known antidote to voriconazole.

Voriconazole is haemodialysed with a clearance of 121 ml/min. In an overdose, haemodialysis may assist in the removal of voriconazole and SBECD from the body.

5 PHARMACOLOGICAL PROPERTIES

5.1 Pharmacodynamic properties

Pharmacotherapeutic group: Antimycotics for systemic use, triazole derivatives, ATC: J02AC03.

Mechanism of Action

Voriconazole is a triazole antifungal agent. The primary mode of action of voriconazole is the inhibition of fungal cytochrome P450-mediated 14 alpha-lanosterol demethylation, an essential step in fungal ergosterol biosynthesis. The accumulation of 14 alpha-methyl sterols correlates with the subsequent loss of ergosterol in the fungal cell membrane and may be responsible for the antifungal activity of voriconazole. Voriconazole has been shown to be more selective for fungal cytochrome P450 enzymes than for various mammalian cytochrome P-450 enzyme systems.

Pharmacokinetic/Pharmacodynamic Relationship

In 10 therapeutic studies, the median for the average and maximum plasma concentrations in individual subjects across the studies was 2425 ng/ml (inter-quartile range 1193 to 4380 ng/ml) and 3742 ng/ml (inter-quartile range 2027 to 6302 ng/ml), respectively. A positive association between mean, maximum or minimum plasma voriconazole concentration and efficacy in therapeutic studies was not found.

Pharmacokinetic-Pharmacodynamic analyses of clinical trial data identified positive associations between plasma voriconazole concentrations and both liver function test abnormalities and visual disturbances.

Clinical efficacy and safety

In vitro, voriconazole displays broad-spectrum antifungal activity with antifungal potency against Candida species (including fluconazole resistant C. krusei and resistant strains of C. glabrata and

C. albicans) and fungicidal activity against all Aspergillus species tested. In addition voriconazole shows in vitro fungicidal activity against emerging fungal pathogens, including those such as Scedosporium or Fusarium which have limited susceptibility to existing antifungal agents.

Clinical efficacy (defined as partial or complete response) has been demonstrated for

Aspergillus spp. including A. flavus, A. fumigatus, A. terreus, A. niger, A. nidulans, Candida spp., including C. albicans, C. glabrata, C. krusei, C. parapsilosis and C. tropicalis and limited numbers of C. dubliniensis, C. inconspicua, and C. guilliermondii, Scedosporium spp., including S. apiospermum, S. prolificans and Fusarium spp.

Other treated fungal infections (often with either partial or complete response) included isolated cases of Alternaria spp., Blastomyces dermatitidis, Blastoschizomyces capitatus, Cladosporium spp., Coccidioides immitis, Conidiobolus coronatus, Cryptococcus neoformans, Exserohilum rostratum, Exophiala spinifera, Fonsecaeapedrosoi, Madurella mycetomatis, Paecilomyces lilacinus, Penicillium spp. including P. marneffei, Phialophora richardsiae, Scopulariopsis brevicaulis and Trichosporon spp. including T. beigelii infections.

In vitro activity against clinical isolates has been observed for Acremonium spp., Alternaria spp., Bipolaris spp., Cladophialophora spp., andHistoplasma capsulatum, with most strains being inhibited by concentrations of voriconazole in the range 0.05 to 2 gg/ml.

In vitro activity against the following pathogens has been shown, but the clinical significance is unknown: Curvularia spp. and Sporothrix spp.

Breakpoints

Specimens for fungal culture and other relevant laboratory studies (serology, histopathology) should be obtained prior to therapy to isolate and identify causative organisms. Therapy may be instituted before the results of the cultures and other laboratory studies are known; however, once these results become available, antiinfective therapy should be adjusted accordingly.

The species most frequently involved in causing human infections include C. albicans, C. parapsilosis, C. tropicalis, C. glabrata and C. krusei, all of which usually exhibit MICs of less than 1 mg/L for voriconazole.

However, the in vitro activity of voriconazole against Candida species is not uniform. Specifically, for C. glabrata, the MICs of voriconazole for fluconazole-resistant isolates are proportionally higher than are those of fluconazole-susceptible isolates. Therefore, every attempt should be made to identify Candida to species level. If antifungal susceptibility testing is available, the MIC results may be interpreted using breakpoint criteria established by European Committee on Antimicrobial Susceptibility Testing (EUCAST).

EUCAST Breakpoints

Candida Species

MIC breakpoint (mg/L)

^ S (Susceptible)

>R (Resistant)

Candida albicans1

0.125

0.125

Candida tropicalis1

0.125

0.125

Candida parapsilosis1

0.125

0.125

Candida glabrata2

Insufficient evidence

Candida krusei3

Insufficient evidence

Other Candida spp.4

Insufficient evidence

1 Strains with MIC values above the Susceptible (S) breakpoint are rare, or not yet reported. The identification and antimicrobial susceptibility tests on any such isolate must be repeated and if the result is confirmed the isolate sent to a reference

laboratory.

2    In clinical studies, response to voriconazole in patients with C glabrata infections was 21% lower compared to C. albicans, C. parapsilosis and C. tropicalis. However, this reduced response was not correlated with elevated MICs.

3    In clinical studies, response to voriconazole in C. krusei infections was similar to C. albicans, C. parapsilosis and C. tropicalis. However, as there were only 9 cases available for EUCAST analysis, there is currently insufficient evidence to set clinical breakpoints for C. krusei.

4    EUCAST has not determined non-species related breakpoints for voriconazole.


Clinical Experience

Successful outcome in this section is defined as complete or partial response.

Aspergillus infections - efficacy in aspergillosis patients with poor prognosis Voriconazole has in vitro fungicidal activity against Aspergillus spp. The efficacy and survival benefit of voriconazole versus conventional amphotericin B in the primary treatment of acute invasive aspergillosis was demonstrated in an open, randomised, multicentre study in 277 immunocompromised patients treated for 12 weeks. Voriconazole was administered intravenously with a loading dose of 6 mg/kg every 12 hours for the first 24 hours followed by a maintenance dose of 4 mg/kg every 12 hours for a minimum of 7 days. Therapy could then be switched to the oral formulation at a dose of 200 mg every 12 hours. Median duration of IV voriconazole therapy was 10 days (range 2-85 days). After IV voriconazole therapy, the median duration of oral voriconazole therapy was 76 days (range 2-232 days).

A satisfactory global response (complete or partial resolution of all attributable symptoms signs, radiographic/bronchoscopic abnormalities present at baseline) was seen in 53 % of voriconazole-treated patients compared to 31 % of patients treated with comparator. The 84-day survival rate for voriconazole was statistically significantly higher than that for the comparator and a clinically and statistically significant benefit was shown in favour of voriconazole for both time to death and time to discontinuation due to toxicity.

This study confirmed findings from an earlier, prospectively designed study where there was a positive outcome in subjects with risk factors for a poor prognosis, including graft versus host disease, and, in particular, cerebral infections (normally associated with almost 100 % mortality).

The studies included cerebral, sinus, pulmonary and disseminated aspergillosis in patients with bone marrow and solid organ transplants, haematological malignancies, cancer and AIDS.

Candidaemia in non-neutropenic patients

The efficacy of voriconazole compared to the regimen of amphotericin B followed by fluconazole in the primary treatment of candidaemia was demonstrated in an open, comparative study. Three hundred and seventy non-neutropenic patients (above 12

years of age) with documented candidaemia were included in the study, of whom 248 were treated with voriconazole. 9 subjects in the voriconazole group and 5 in the amphotericin B followed by fluconazole group also had mycologically proven infection in deep tissue. Patients with renal failure were excluded from this study. The median treatment duration was 15 days in both treatment arms. In the primary analysis, successful response as assessed by a Data Review Committee (DRC) blinded to study medicinal product was defined as resolution/improvement in all clinical signs and symptoms of infection with eradication of Candida from blood and infected deep tissue sites 12 weeks after the end of therapy (EOT). Patients who did not have an assessment 12 weeks after EOT were counted as failures. In this analysis a successful response was seen in 41 % of patients in both treatment arms.

In a secondary analysis, which utilised DRC assessments at the latest evaluable time point (EOT, or 2, 6, or 12 weeks after EOT) voriconazole and the regimen of amphotericin B followed by fluconazole had successful response rates of 65 % and 71 %, respectively.

The Investigator’s assessment of successful outcome at each of these time points is shown in the following table.

Timepoint

Voriconazole (N=248)

Amphotericin B ffluconazole (N=122)

EOT

178 (72 %)

88 (72 %)

2 weeks after EOT

125 (50 %)

62 (51 %)

6 weeks after EOT

104 (42 %)

55 (45 %)

12 weeks after EOT

104 (42 %)

51 (42 %)

Serious refractory Candida infections

The study comprised 55 patients with serious refractory systemic Candida infections (including candidaemia, disseminated and other invasive candidiasis) where prior antifungal treatment, particularly with fluconazole, had been ineffective. Successful response was seen in 24 patients (15 complete, 9 partial responses). In fluconazole-resistant non albicans species, a successful outcome was seen in 3/3 C. krusei (complete responses) and 6/8 C. glabrata (5 complete, 1 partial response) infections. The clinical efficacy data were supported by limited susceptibility data.

Scedosporium and Fusarium infections

Voriconazole was shown to be effective against the following rare fungal pathogens:

Scedosporium spp.: Successful response to voriconazole therapy was seen in 16 (6 complete, 10 partial responses) of 28 patients with S. apiospermum and in 2 (both partial responses) of 7 patients with S. prolificans infection. In addition, a successful response was seen in 1 of 3 patients with infections caused by more than one organism including Scedosporium spp.

Fusarium spp.: Seven (3 complete, 4 partial responses) of 17 patients were successfully treated with voriconazole. Of these 7 patients, 3 had eye, 1 had sinus, and 3 had disseminated infection. Four additional patients with fusariosis had an infection caused by several organisms; two of them had a successful outcome.

The majority of patients receiving voriconazole treatment of the above mentioned rare infections were intolerant of, or refractory to, prior antifungal therapy.

Duration of treatment

In clinical trials, 561 patients received voriconazole therapy for greater than 12 weeks, with 136 patients receiving voriconazole for over 6 months.

Paediatric population

Sixty one paediatric patients aged 9 months up to 15 years who had definite or probable invasive fungal infections, were treated with voriconazole. This population included 34 patients 2 to <12 years old and 20 patients 12 to 15 years of age.

The majority (57/61) had failed previous antifungal therapies. Therapeutic studies included 5 patients aged 12 to 15 years, the remaining patients received voriconazole in the compassionate use programmes. Underlying diseases in these patients included haematological malignancies (27 patients) and chronic granulomatous disease (14 patients). The most commonly treated fungal infection was aspergillosis (43/61; 70 %).

Clinical Studies Examining QT Interval

A placebo-controlled, randomized, single-dose, crossover study to evaluate the effect on the QT interval of healthy volunteers was conducted with three oral doses of voriconazole and ketoconazole. The placebo-adjusted mean maximum increases in QTc from baseline after 800, 1200 and 1600 mg of voriconazole were 5.1, 4.8, and 8.2 msec, respectively and 7.0 msec for ketoconazole 800 mg. No subject in any group had an increase in QTc of >60 msec from baseline. No subject experienced an interval exceeding the potentially clinically relevant threshold of 500 msec.

5.2 Pharmacokinetic properties

General pharmacokinetic characteristics

The pharmacokinetics of voriconazole have been characterised in healthy subjects, special populations and patients. During oral administration of 200 mg or 300 mg twice daily for 14 days in patients at risk of aspergillosis (mainly patients with malignant neoplasms of lymphatic or haematopoietic tissue), the observed pharmacokinetic characteristics of rapid and consistent absorption, accumulation and non-linear pharmacokinetics were in agreement with those observed in healthy subjects.

The pharmacokinetics of voriconazole are non-linear due to saturation of its metabolism. Greater than proportional increase in exposure is observed with increasing dose. It is estimated that, on average, increasing the oral dose from 200 mg twice daily to 300 mg twice daily leads to a 2.5-fold increase in exposure (AUCr). The oral maintenance dose of 200 mg (or 100 mg for patients less than 40 kg) achieves a voriconazole exposure similar to 3 mg/kg IV. A 300 mg (or 150 mg for patients less than 40 kg) oral maintenance dose achieves an exposure similar to 4 mg/kg IV. When the recommended intravenous or oral loading dose regimens are administered, plasma concentrations close to steady state are achieved within the first 24 hours of dosing. Without the loading dose, accumulation occurs during twice daily multiple dosing with steady-state plasma voriconazole concentrations being achieved by Day 6 in the majority of subjects.

Absorption

Voriconazole is rapidly and almost completely absorbed following oral administration, with maximum plasma concentrations (Cmax) achieved 1-2 hours after dosing. The absolute bioavailability of voriconazole after oral administration is estimated to be 96 %.

Bioequivalence was established between the 200 mg tablets and the 40mg/ml oral suspension when administered as a 200 mg dose. When multiple doses of voriconazole are administered with high fat meals, Cmax and AUC are reduced by 34 % and 24 %, respectively. The absorption of voriconazole is not affected by changes in gastric pH.

Distribution

The volume of distribution at steady state for voriconazole is estimated to be 4.6 l/kg, suggesting extensive distribution into tissues. Plasma protein binding is estimated to be 58 %. Cerebrospinal fluid samples from eight patients in a compassionate programme showed detectable voriconazole concentrations in all patients.

Biotransformation

In vitro studies showed that voriconazole is metabolised by, the hepatic cytochrome P450 isoenzymes, CYP2C19, CYP2C9 and CYP3A4.

The inter-individual variability of voriconazole pharmacokinetics is high.

In vivo studies indicated that CYP2C19 is significantly involved in the metabolism of voriconazole. This enzyme exhibits genetic polymorphism. For example, 15-20 % of Asian populations may be expected to be poor metabolisers. For Caucasians and Blacks the prevalence of poor metabolisers is 3-5 %. Studies conducted in Caucasian and Japanese healthy subjects have shown that poor metabolisers have, on average, 4fold higher voriconazole exposure (AUCT) than their homozygous extensive metaboliser counterparts.

Subjects who are heterozygous extensive metabolisers have on average 2-fold higher voriconazole exposure than their homozygous extensive metaboliser counterparts.

The major metabolite of voriconazole is the N-oxide, which accounts for 72 % of the circulating radiolabelled metabolites in plasma. This metabolite has minimal antifungal activity and does not contribute to the overall efficacy of voriconazole.

Elimination

Voriconazole is eliminated via hepatic metabolism with less than 2 % of the dose excreted unchanged in the urine.

After administration of a radiolabelled dose of voriconazole, approximately 80 % of the radioactivity is recovered in the urine after multiple intravenous dosing and 83 % in the urine after multiple oral dosing. The majority (>94 %) of the total radioactivity is excreted in the first 96 hours after both oral and intravenous dosing.

The terminal half-life of voriconazole depends on dose and is approximately 6 hours at 200 mg (orally). Because of non-linear pharmacokinetics, the terminal half-life is not useful in the prediction of the accumulation or elimination of voriconazole.

Pharmacokinetics in special patient groups Gender

In an oral multiple dose study, Cmax and AUCr for healthy young females were 83 % and 113 % higher, respectively, than in healthy young males (18-45 years). In the same study, no significant differences in Cmax and AUCr were observed between healthy elderly males and healthy elderly females (>65 years).

In the clinical programme, no dosage adjustment was made on the basis of gender. The safety profile and plasma concentrations observed in male and female patients were similar. Therefore, no dosage adjustment based on gender is necessary.

Elderly

In an oral multiple dose study Cmax and AUCr in healthy elderly males (>65 years) were 61 % and 86 % higher, respectively, than in healthy young males (18-45 years). No significant differences in Cmax and AUC were observed between healthy elderly females (> 65 years) and healthy young females (18- 45 years).

In the therapeutic studies no dosage adjustment was made on the basis of age. A relationship between plasma concentrations and age was observed. The safety profile of voriconazole in young and elderly patients was similar and, therefore, no dosage adjustment is necessary for the elderly (see section 4.2).

Paediatric

The recommended doses in children and adolescent patients are based on a population pharmacokinetic analysis of data obtained from 112 immunocompromised paediatric patients aged 2 to <12 years and 26 immunocompromised adolescent patients aged 12 to <17 years. Multiple intravenous doses of 3, 4, 6, 7 and 8 mg/kg twice daily and multiple oral doses (using the powder for oral suspension) of 4 mg/kg, 6 mg/kg, and 200 mg twice daily were evaluated in 3 paediatric pharmacokinetic studies. Intravenous loading doses of 6 mg/kg IV twice daily on day 1 followed by 4 mg/kg intravenous dose twice daily and 300 mg oral tablets twice daily were evaluated in one adolescent pharmacokinetic study. Larger inter-subject variability was observed in paediatric patients compared to adults. A comparison of the paediatric and adult population pharmacokinetic data indicated that the predicted total exposure (AUC) in children following administration of a 9 mg/kg IV loading dose was comparable to that in adults following a 6 mg/kg IV loading dose. The predicted total exposures in children following IV maintenance doses of 4 and 8 mg/kg twice daily were comparable to those in adults following 3 and 4 mg/kg IV twice daily, respectively. The predicted total exposure in children following an oral maintenance dose of 9 mg/kg (maximum of 350 mg) twice daily was comparable to that in adults following 200 mg oral twice daily. An 8 mg/kg intravenous dose will provide voriconazole exposure approximately 2-fold higher than a 9 mg/kg oral dose.

The higher intravenous maintenance dose in paediatric patients relative to adults reflects the higher elimination capacity in paediatric patients due to a greater liver mass to body mass ratio. Oral bioavailability may, however, be limited in paediatric patients with malabsorption and very low body weight for their age. In that case, intravenous voriconazole administration is recommended.

Voriconazole exposures in the majority of adolescent patients were comparable to those in adults receiving the same dosing regimens. However, lower voriconazole exposure was observed in some young adolescents with low body weight compared to adults. It is likely that these subjects may metabolize voriconazole more similarly to children than to adults. Based on the population pharmacokinetic analysis, 12- to 14-year-old adolescents weighing less than 50 kg should receive children’s doses (see section 4.2).

Renal impairment

In an oral single dose (200 mg) study in subjects with normal renal function and mild (creatinine clearance 41-60 ml/min) to severe (creatinine clearance <20 ml/min) renal impairment, the pharmacokinetics of voriconazole were not significantly affected by renal impairment. The plasma protein binding of voriconazole was similar in subjects with different degrees of renal impairment (see sections 4.2 and 4.4).

Hepatic impairment

After an oral single dose (200 mg), AUC was 233 % higher in subjects with mild to moderate hepatic cirrhosis (Child-Pugh A and B) compared with subjects with normal hepatic function. Protein binding of voriconazole was not affected by impaired hepatic function.

In an oral multiple dose study, AUC was similar in subjects with moderate hepatic cirrhosis (Child-Pugh B) given a maintenance dose of 100 mg twice daily and

subjects with normal hepatic function given 200 mg twice daily. No pharmacokinetic data are available for patients with severe hepatic cirrhosis (Child-Pugh C) (see sections 4.2 and 4.4).

5.3 Preclinical safety data

Repeated-dose toxicity studies with voriconazole indicated the liver to be the target organ. Hepatotoxicity occurred at plasma exposures similar to those obtained at therapeutic doses in humans, in common with other antifungal agents. In rats, mice and dogs, voriconazole also induced minimal adrenal changes. Conventional studies of safety pharmacology, genotoxicity or carcinogenic potential did not reveal a special hazard for humans.

In reproduction studies, voriconazole was shown to be teratogenic in rats and embryotoxic in rabbits at systemic exposures equal to those obtained in humans with therapeutic doses. In the pre and postnatal development study in rats at exposures lower than those obtained in humans with therapeutic doses, voriconazole prolonged the duration of gestation and labour and produced dystocia with consequent maternal mortality and reduced perinatal survival of pups. The effects on parturition are probably mediated by species-specific mechanisms, involving reduction of oestradiol levels, and are consistent with those observed with other azole antifungal agents. Voriconazole administration induced no impairment of male or female fertility in rats at exposures similar to those obtained in humans at therapeutic doses.

6    PHARMACEUTICAL PARTICULARS

6.1    List of excipients

Tablet core:

Lactose monohydrate Croscarmellose sodium Pregelatinised maize starch Povidone K29/32 Magnesium stearate

Film-coating:

Lactose monohydrate Hypromellose 15 mPas (E464) Titanium dioxide (E171) Triacetin

6.2 Incompatibilities

Not applicable.

6.3 Shelf life

2 years.

6.4 Special precautions for storage

This medicinal product does not require any special storage conditions.

6.5 Nature and contents of container

PVC-Aluminium foil blister packs of 14, 20, 28, 30, 90, 100, 14 x 1, 28 x 1, 30 x 1, 50 x 1, 56 x 1, 60 x 1, 100 x 1 film-coated tablets for Voriconazole 200 mg film-coated tablets.

Not all pack sizes may be marketed.

6.6 Special precautions for disposal

No special requirements for disposal.

Any unused medicinal product or waste material should be disposed of in accordance with local requirements.

7 MARKETING AUTHORISATION HOLDER

Generics [UK] Ltd trading as Mylan Potters Bar,

Hertfordshire,

EN6 1TL,

United Kingdom.

8    MARKETING AUTHORISATION NUMBER(S)

PL 04569/1388

9    DATE OF FIRST AUTHORISATION/RENEWAL OF THE AUTHORISATION

27/06/2014

10    DATE OF REVISION OF THE TEXT

27/06/2014