Thursday, October 13, 2016

Zofran Injection, Flexi-Amp Injection





1. Name Of The Medicinal Product



Zofran Injection 2mg/ml. Zofran Flexi-amp Injection 2mg/ml.


2. Qualitative And Quantitative Composition



Zofran Injection 2mg/ml: 2ml glass ampoules each containing 4mg ondansetron (as hydrochloride dihydrate) in aqueous solution for intramuscular or intravenous administration. 4ml glass ampoules each containing 8mg ondansetron (as hydrochloride dihydrate) in aqueous solution for intravenous or intramuscular administration.



Zofran Flexi-amp injection 2mg/ml: 2ml plastic ampoules each containing 4 mg ondansetron (as hydrochloride dihydrate) in aqueous solution for intramuscular or intravenous administration. 4ml plastic ampoules each containing 8 mg ondansetron (as hydrochloride dihydrate) in aqueous solution for intravenous or intramuscular administration.



3. Pharmaceutical Form



Injection (aqueous solution).



4. Clinical Particulars



4.1 Therapeutic Indications



Adults:



Zofran is indicated for the management of nausea and vomiting induced by cytotoxic chemotherapy and radiotherapy. Zofran is indicated for the prevention and treatment of post-operative nausea and vomiting (PONV).



Paediatric Population:



Zofran is indicated for the management of chemotherapy-induced nausea and vomiting (CINV) in children aged



4.2 Posology And Method Of Administration



Chemotherapy and Radiotherapy:



Adults:



The emetogenic potential of cancer treatment varies according to the doses and combinations of chemotherapy and radiotherapy regimens used. The route of administration and dose of Zofran should be flexible in the range of 8-32mg a day and selected as shown below.



Emetogenic chemotherapy and radiotherapy: Zofran can be given either by rectal, oral (tablets or syrup), intravenous or intramuscular administration.



For most patients receiving emetogenic chemotherapy or radiotherapy, Zofran 8mg should be administered as a slow intravenous or intramuscular injection immediately before treatment, followed by 8mg orally twelve hourly.



To protect against delayed or prolonged emesis after the first 24 hours, oral or rectal treatment with Zofran should be continued for up to 5 days after a course of treatment.



Highly emetogenic chemotherapy: For patients receiving highly emetogenic chemotherapy, e.g. high-dose cisplatin, Zofran can be given either by oral, rectal, intravenous or intramuscular administration. Zofran has been shown to be equally effective in the following dose schedules over the first 24 hours of chemotherapy:



A single dose of 8mg by slow intravenous or intramuscular injection immediately before chemotherapy.



A dose of 8mg by slow intravenous or intramuscular injection immediately before chemotherapy, followed by two further intravenous or intramuscular doses of 8mg two to four hours apart, or by a constant infusion of 1mg/hour for up to 24 hours.



A single dose of 32mg diluted in 50-100ml of saline or other compatible infusion fluid (see Pharmaceutical Precautions) and infused over not less than 15 minutes immediately before chemotherapy.



The selection of dose regimen should be determined by the severity of the emetogenic challenge.



The efficacy of Zofran in highly emetogenic chemotherapy may be enhanced by the addition of a single intravenous dose of dexamethasone sodium phosphate, 20mg administered prior to chemotherapy.



To protect against delayed or prolonged emesis after the first 24 hours, oral or rectal treatment with Zofran should be continued for up to 5 days after a course of treatment.



Paediatric Population:



CINV in children aged



The dose for CINV can be calculated based on body surface area (BSA) or weight – see below. Weight-based dosing results in higher total daily doses compared to BSA-based dosing (sections 4.4.and 5.1).



Zofran injection should be diluted in 5% dextrose or 0.9% sodium chloride or other compatible infusion fluid (see section 6.6) and infused intravenously over not less than 15 minutes.



There are no data from controlled clinical trials on the use of Zofran in the prevention of delayed or prolonged CINV. There are no data from controlled clinical trials on the use of Zofran for radiotherapy-induced nausea and vomiting in children.



Dosing by BSA:



Zofran should be administered immediately before chemotherapy as a single intravenous dose of 5 mg/m2. The intravenous dose must not exceed 8 mg.



Oral dosing can commence twelve hours later and may be continued for up to 5 days (Table 1).



The total daily dose must not exceed adult dose of 32 mg.



Table 1: BSA-based dosing for Chemotherapy - Children aged













BSA




Day 1 (a,b)




Days 2-6(b)




< 0.6 m2




5 mg/m2 i.v. plus 2 mg syrup after 12 hrs




2 mg syrup every 12 hrs




0.6 m2




5 mg/m2 i.v. plus 4 mg syrup or tablet after 12 hrs




4 mg syrup or tablet every 12 hrs



a The intravenous dose must not exceed 8mg.



b The total daily dose must not exceed adult dose of 32 mg



Dosing by bodyweight:



Weight-based dosing results in higher total daily doses compared to BSA-based dosing (sections 4.4. and 5.1).



Zofran should be administered immediately before chemotherapy as a single intravenous dose of 0.15 mg/kg. The intravenous dose must not exceed 8 mg.



Two further intravenous doses may be given in 4-hourly intervals. The total daily dose must not exceed adult dose of 32 mg.



Oral dosing can commence twelve hours later and may be continued for up to 5 days (Table 2).



Table 2: Weight-based dosing for Chemotherapy - Children aged













Weight




Day 1 (a,b)




Days 2-6(b)







Up to 3 doses of 0.15 mg/kg every 4 hrs




2 mg syrup every 12 hrs




> 10 kg




Up to 3 doses of 0.15 mg/kg every 4 hrs




4 mg syrup or tablet every 12 hrs



a The intravenous dose must not exceed 8mg.



b The total daily dose must not exceed adult dose of 32 mg.



Elderly:



Zofran is well tolerated by patients over 65 years and no alteration of dosage, dosing frequency or route of administration are required.



Patients with Renal Impairment:



No alteration of daily dosage or frequency of dosing, or route of administration are required.



Patients with Hepatic Impairment:



Clearance of Zofran is significantly reduced and serum half-life significantly prolonged in subjects with moderate or severe impairment of hepatic function. In such patients a total daily dose of 8mg should not be exceeded and therefore parenteral or oral administration is recommended.



Post-Operative Nausea and Vomiting (PONV):



Adults:



For the prevention of PONV Zofran can be administered orally or by intravenous or intramuscular injection.



Zofran may be administered as a single dose of 4mg given by intramuscular or slow intravenous injection at induction of anaesthesia.



For treatment of established PONV a single dose of 4mg given by intramuscular or slow intravenous injection is recommended.



Paediatric population



PONV in children aged



For prevention of PONV in paediatric patients having surgery performed under general anaesthesia, a single dose of ondansetron may be administered by slow intravenous injection (not less than 30 seconds) at a dose of 0.1mg/kg up to a maximum of 4mg either prior to, at or after induction of anaesthesia.



For the treatment of PONV after surgery in paediatric patients having surgery performed under general anaesthesia, a single dose of Zofran may be administered by slow intravenous injection (not less than 30 seconds) at a dose of 0.1mg/kg up to a maximum of 4mg.



There are no data on the use of Zofran in the treatment of PONV in children below 2 years of age.



Elderly:



There is limited experience in the use of Zofran in the prevention and treatment of PONV in the elderly, however Zofran is well tolerated in patients over 65 years receiving chemotherapy.



Patients with Renal Impairment:



No alteration of daily dosage or frequency of dosing, or route of administration are required.



Patients with Hepatic Impairment:



Clearance of Zofran is significantly reduced and serum half life significantly prolonged in subjects with moderate or severe impairment of hepatic function. In such patients a total daily dose of 8mg should not be exceeded and therefore parenteral or oral administration is recommended.



Patients with poor Sparteine/Debrisoquine Metabolism:



The elimination half-life of ondansetron is not altered in subjects classified as poor metabolisers of sparteine and debrisoquine. Consequently in such patients repeat dosing will give drug exposure levels no different from those of the general population. No alteration of daily dosage or frequency of dosing are required.



4.3 Contraindications



Hypersensitivity to any component of the preparation.



4.4 Special Warnings And Precautions For Use



Hypersensitivity reactions have been reported in patients who have exhibited hypersensitivity to other selective 5HT3 receptor antagonists.



Respiratory events should be treated symptomatically and clinicians should pay particular attention to them as precursors of hypersensitivity reactions.



Very rarely and predominantly with intravenous Zofran, transient ECG changes including QT interval prolongation have been reported. Therefore, caution should be exercised in patients with cardiac rhythm or conduction disturbances, in patients treated with anti-arrhythmic agents or beta-adrenergic blocking agents and in patients with significant electrolyte disturbances.



As ondansetron is known to increase large bowel transit time, patients with signs of subacute intestinal obstruction should be monitored following administration.



In patients with adenotonsillar surgery prevention of nausea and vomiting with ondansetron may mask occult bleeding. Therefore, such patients should be followed carefully after ondansetron.



Paediatric Population:



Paediatric patients receiving ondansetron with hepatotoxic chemotherapeutic agents should be monitored closely for impaired hepatic function.



CINV



When calculating the dose on an mg/kg basis and administering three doses at 4-hourly intervals, the total daily dose will be higher than if one single dose of 5mg/m2 followed by an oral dose is given. The comparative efficacy of these two different dosing regimens has not been investigated in clinical trials. Cross-trial comparison indicates similar efficacy for both regimens (section 5.1).



4.5 Interaction With Other Medicinal Products And Other Forms Of Interaction



There is no evidence that ondansetron either induces or inhibits the metabolism of other drugs commonly co-administered with it. Specific studies have shown that there are no interactions when ondansetron is administered with alcohol, temazepan, furosemide, alfentanil, tramadol, morphine, lignocaine, thiopental, or propofol.



Ondansetron is metabolised by multiple hepatic cytochrome P-450 enzymes: CYP3A4, CYP2D6 and CYP1A2. Due to the multiplicity of metabolic enzymes capable of metabolising ondansetron, enzyme inhibition or reduced activity of one enzyme (e.g. CYP2D6 genetic deficiency) is normally compensated by other enzymes and should result in little or no significant change in overall ondansetron clearance or dose requirement.



Phenytoin, Carbamazepine and Rifampicin: In patients treated with potent inducers of CYP3A4 (i.e. phenytoin, carbamazepine, and rifampicin), the oral clearance of ondansetron was increased and ondansetron blood concentrations were decreased.



Tramadol: Data from small studies indicate that ondansetron may reduce the analgesic effect of tramadol.



Use of Zofran with QT prolonging drugs may result in additional QT prolongation. Concomitant use of Zofran with cardiotoxic drugs (e.g. anthracyclines) may increase the risk of arrhythmias (section 4.4).



4.6 Pregnancy And Lactation



The safety of ondansetron for use in human pregnancy has not been established. Evaluation of experimental animal studies does not indicate direct or indirect harmful effects with respect to the development of the embryo, or foetus, the course of gestation and peri- and post-natal development. However as animal studies are not always predictive of human response the use of ondansetron in pregnancy is not recommended.



Tests have shown that ondansetron passes into the milk of lactating animals. It is therefore recommended that mothers receiving Zofran should not breast-feed their babies.



4.7 Effects On Ability To Drive And Use Machines



Ondansetron has no or negligible influence on the ability to drive and use machines.



4.8 Undesirable Effects



Adverse events are listed below by system organ class and frequency. Frequencies are defined as: very common (



The following frequencies are estimated at the standard recommended doses of ondansetron according to indication and formulation.
















































Immune system disorders


 


Rare:




Immediate hypersensitivity reactions sometimes severe, including anaphylaxis.




Nervous system disorders


 


Very common:




Headache.




Uncommon:




Seizures, movement disorders including extrapyramidal reactions such as dystonic reactions, oculogyric crisis and dyskinesia(1).




Rare:




Dizziness during i.v. administration, which in most cases is prevented or resolved by lengthening the infusion period.




Eye disorders


 


Rare:




Transient visual disturbances (e.g. blurred vision) during i.v. administration.




Very rare:




Transient blindness predominantly during intravenous administration.(2)




Cardiac disorders


 


Uncommon:




Arrhythmias, chest pain with or without ST segment depression, bradycardia.




Vascular disorders


 


Common:




Sensation of warmth or flushing.




Uncommon:




Hypotension.




Respiratory, thoracic and mediastinal disorders


 


Uncommon:




Hiccups.




Gastrointestinal disorders


 


Common:




Constipation.




Hepatobiliary disorders


 


Uncommon:




Asymptomatic increases in liver function tests(3).




General disorders and administration site conditions


 


Common:




Local i.v. injection site reactions.



1. Observed without definitive evidence of persistent clinical sequelae.



2. The majority of the blindness cases reported resolved within 20 minutes. Most patients had received chemotherapeutic agents, which included cisplatin. Some cases of transient blindness were reported as cortical in origin.



3. These events were observed commonly in patients receiving chemotherapy with cisplatin.



Paediatric population



The adverse event profile in children and adolescents was comparable to that seen in adults.



4.9 Overdose



Little is known at present about overdosage with ondansetron, however, a limited number of patients received overdoses. Manifestations that have been reported include visual disturbances, severe constipation, hypotension and a vasovagal episode with transient second degree AV block. In all instances, the events resolved completely. There is no specific antidote for ondansetron, therefore in all cases of suspected overdose, symptomatic and supportive therapy should be given as appropriate. The use of ipecacuanha to treat overdose with ondansetron is not recommended, as patients are unlikely to respond due to the anti-emetic action of ondansetron itself.



5. Pharmacological Properties



5.1 Pharmacodynamic Properties



Ondansetron is a potent, highly selective 5HT3 receptor-antagonist. Its precise mode of action in the control of nausea and vomiting is not known. Chemotherapeutic agents and radiotherapy may cause release of 5HT in the small intestine initiating a vomiting reflex by activating vagal afferents via 5HT3 receptors. Ondansetron blocks the initiation of this reflex. Activation of vagal afferents may also cause a release of 5HT in the area postrema, located on the floor of the fourth ventricle, and this may also promote emesis through a central mechanism. Thus, the effect of ondansetron in the management of the nausea and vomiting induced by cytotoxic chemotherapy and radiotherapy is probably due to antagonism of 5HT3 receptors on neurons located both in the peripheral and central nervous system. The mechanisms of action in post-operative nausea and vomiting are not known but there may be common pathways with cytotoxic induced nausea and vomiting.



Ondansetron does not alter plasma prolactin concentrations.



The role of ondansetron in opiate-induced emesis is not yet established.



Paediatric population



CINV



The efficacy of ondansetron in the control of emesis and nausea induced by cancer chemotherapy was assessed in a double-blind randomised trial in 415 patients aged 1 to 18 years (S3AB3006). On the days of chemotherapy, patients received either ondansetron 5 mg/m2 intravenous + ondansetron 4 mg orally after 8-12 hrs or ondansetron 0.45 mg/kg intravenous + placebo orally after 8-12 hrs. Post-chemotherapy both groups received 4 mg ondansetron syrup twice daily for 3 days. Complete control of emesis on worst day of chemotherapy was 49% (5 mg/m2 intravenous + ondansetron 4 mg orally) and 41% (0.45 mg/kg intravenous + placebo orally). Post-chemotherapy both groups received 4 mg ondansetron syrup twice daily for 3 days. There was no difference in the overall incidence or nature of adverse events between the two treatment groups.



A double-blind randomised placebo-controlled trial (S3AB4003) in 438 patients aged 1 to 17 years demonstrated complete control of emesis on worst day of chemotherapy in:



• 73% of patients when ondansetron was administered intravenously at a dose of 5 mg/m2 intravenous together with 2-4 mg dexamethasone orally



• 71% of patients when ondansetron was administered as syrup at a dose of 8 mg + 2-4 mg dexamethasone orally on the days of chemotherapy.



Post-chemotherapy both groups received 4 mg ondansetron syrup twice daily for 2 days. There was no difference in the overall incidence or nature of adverse events between the two treatment groups.



The efficacy of ondansetron in 75 children aged 6 to 48 months was investigated in an open-label, non-comparative, single-arm study (S3A40320). All children received three 0.15 mg/kg doses of intravenous ondansetron, administered 30 minutes before the start of chemotherapy and then at four and eight hours after the first dose. Complete control of emesis was achieved in 56% of patients.



Another open-label, non-comparative, single-arm study (S3A239) investigated the efficacy of one intravenous dose of 0.15 mg/kg ondansetron followed by two oral ondansetron doses of 4 mg for children aged < 12 yrs and 8 mg for children aged



PONV



The efficacy of a single dose of ondansetron in the prevention of post-operative nausea and vomiting was investigated in a randomised, double-blind, placebo-controlled study in 670 children aged 1 to 24 months (post-conceptual age



Four double-blind, placebo-controlled studies have been performed in 1469 male and female patients (2 to 12 years of age) undergoing general anaesthesia. Patients were randomised to either single intravenous doses of ondansetron (0.1 mg/kg for paediatric patients weighing 40 kg or less, 4 mg for paediatric patients weighing more than 40 kg; number of patients = 735)) or placebo (number of patients = 734). Study drug was administered over at least 30 seconds, immediately prior to or following anaesthesia induction. Ondansetron was significantly more effective than placebo in preventing nausea and vomiting. The results of these studies are summarised in Table 3.



Table 3 Prevention and treatment of PONV in Paediatric Patients – Treatment response over 24 hours


































Study




Endpoint




Ondansetron %




Placebo %




p value




S3A380




CR




68




39







S3GT09




CR




61




35







S3A381




CR




53




17







S3GT11




no nausea




64




51




0.004




S3GT11




no emesis




60




47




0.004



CR = no emetic episodes, rescue or withdrawal



5.2 Pharmacokinetic Properties



Following oral administration, ondansetron is passively and completely absorbed from the gastrointestinal tract and undergoes first pass metabolism. Peak plasma concentrations of about 30ng/ml are attained approximately 1.5 hours after an 8mg dose. For doses above 8mg the increase in ondansetron systemic exposure with dose is greater than proportional; this may reflect some reduction in first pass metabolism at higher oral doses. Mean bioavailability in healthy male subjects, following the oral administration of a single 8 mg tablet, is approximately 55 to 60%. Bioavailability, following oral administration, is slightly enhanced by the presence of food but unaffected by antacids. Studies in healthy elderly volunteers have shown slight, but clinically insignificant, age-related increases in both oral bioavailability (65%) and half-life (5 hours) of ondansetron. Gender differences were shown in the disposition of ondansetron, with females having a greater rate and extent of absorption following an oral dose and reduced systemic clearance and volume of distribution (adjusted for weight).



The disposition of ondansetron following oral, intramuscular and intravenous dosing in adults is similar with a terminal half life of about 3 hours and steady state volume of distribution of about 140L. Equivalent systemic exposure is achieved after intramuscular and intravenous administration of ondansetron.



A 4mg intravenous infusion of ondansetron given over 5 minutes results in peak plasma concentrations of about 65ng/ml. Following intramuscular administration of ondansetron, peak plasma concentrations of about 25ng/ml are attained within 10 minutes of injection.



Following administration of ondansetron suppository, plasma ondansetron concentrations become detectable between 15 and 60 minutes after dosing. Concentrations rise in an essentially linear fashion, until peak concentrations of 20-30 ng/ml are attained, typically 6 hours after dosing. Plasma concentrations then fall, but at a slower rate than observed following oral dosing due to continued absorption of ondansetron. The absolute bioavailability of ondansetron from the suppository is approximately 60% and is not affected by gender. The half life of the elimination phase following suppository administration is determined by the rate of ondansetron absorption, not systemic clearance and is approximately 6 hours. Females show a small, clinically insignificant, increase in half-life in comparison with males.



Ondansetron is not highly protein bound (70-76%). Ondansetron is cleared from the systemic circulation predominantly by hepatic metabolism through multiple enzymatic pathways. Less than 5% of the absorbed dose is excreted unchanged in the urine. The absence of the enzyme CYP2D6 (the debrisoquine polymorphism) has no effect on ondansetron's pharmacokinetics. The pharmacokinetic properties of ondansetron are unchanged on repeat dosing.



Special Patient Populations



Children and Adolescents (aged 1 month to 17 years)



In paediatric patients aged 1 to 4 months (n=19) undergoing surgery, weight normalised clearance was approximately 30% slower than in patients aged 5 to 24 months (n=22) but comparable to the patients aged 3 to 12 years. The half-life in the patient population aged 1 to 4 month was reported to average 6.7 hours compared to 2.9 hours for patients in the 5 to 24 month and 3 to 12 year age range. The differences in pharmacokinetic parameters in the 1 to 4 month patient population can be explained in part by the higher percentage of total body water in neonates and infants and a higher volume of distribution for water soluble drugs like ondansetron.



In paediatric patients aged 3 to 12 years undergoing elective surgery with general anaesthesia, the absolute values for both the clearance and volume of distribution of ondansetron were reduced in comparison to values with adult patients. Both parameters increased in a linear fashion with weight and by 12 years of age, the values were approaching those of young adults. When clearance and volume of distribution values were normalised by body weight, the values for these parameters were similar between the different age group populations. Use of weight-based dosing compensates for age-related changes and is effective in normalising systemic exposure in paediatric patients.



Population pharmacokinetic analysis was performed on 428 subjects (cancer patients, surgery patients and healthy volunteers) aged 1 month to 44 years following intravenous administration of ondansetron. Based on this analysis, systemic exposure (AUC) of ondansetron following oral or IV dosing in children and adolescents was comparable to adults, with the exception of infants aged 1 to 4 months. Volume was related to age and was lower in adults than in infants and children. Clearance was related to weight but not to age with the exception of infants aged 1 to 4 months. It is difficult to conclude whether there was an additional reduction in clearance related to age in infants 1 to 4 months or simply inherent variability due to the low number of subjects studied in this age group. Since patients less than 6 months of age will only receive a single dose in PONV a decreased clearance is not likely to be clinically relevant.



Renal Impairment



In patients with renal impairment (creatinine clearance 15-60 ml/min), both systemic clearance and volume of distribution are reduced following intravenous administration of ondansetron, resulting in a slight, but clinically insignificant, increase in elimination half-life (5.4h). A study in patients with severe renal impairment who required regular haemodialysis (studied between dialyses) showed ondansetron's pharmacokinetics to be essentially unchanged following intravenous administration.



Elderly



Specific studies in the elderly or patients with renal impairment have been limited to intravenous and oral administration. However, it is anticipated that the half-life of ondansetron after rectal administration in these populations will be similar to that seen in healthy volunteers, since the rate of elimination of ondansetron following rectal administration is not determined by systemic clearance.



Hepatic Impairment



Following oral, intravenous or intramuscular dosing in patients with severe hepatic impairment, ondansetron's systemic clearance is markedly reduced with prolonged elimination half-lives (15-32 h) and an oral bioavailability approaching 100% due to reduced pre-systemic metabolism. The pharmacokinetics of ondansetron following administration as a suppository have not been evaluated in patients with hepatic impairment.



5.3 Preclinical Safety Data



No additional data of relevance.



6. Pharmaceutical Particulars



6.1 List Of Excipients



Citric acid monohydrate, sodium citrate, sodium chloride, Water for Injections.



6.2 Incompatibilities



Zofran injection should not be administered in the same syringe or infusion as any other medication.



Ondansetron injection should only be mixed with those infusion solutions that are recommended.



6.3 Shelf Life



36 months (unopened). 24 hours (dilutions stored 2 - 8°C).



6.4 Special Precautions For Storage



Protect from light. Store below 30°C.



Dilutions of Zofran injection in compatible intravenous infusion fluids are stable under normal room lighting conditions or daylight for at least 24 hours, thus no protection from light is necessary while infusion takes place.



6.5 Nature And Contents Of Container



Zofran Injection: Type I clear glass snap-ring ampoules.



Zofran Flexi-amp injection: Polypropylene blow-fill-sealed ampoules with a twist-off top and overwrapped in a double foil blister.



5 ampoules are packed in a carton.



6.6 Special Precautions For Disposal And Other Handling



Zofran Injection and Zofran Flexi-amp injection should not be autoclaved.



Compatibility with intravenous fluids



Zofran injection should only be mixed with those infusion solutions which are recommended:



• Sodium Chloride Intravenous Infusion BP 0.9%w/v



• Glucose Intravenous Infusion BP 5%w/v



• Mannitol Intravenous Infusion BP 10%w/v



• Ringers Intravenous Infusion



• Potassium Chloride 0.3%w/v and Sodium Chloride 0.9%w/v Intravenous Infusion BP



• Potassium Chloride 0.3%w/v and Glucose 5%w/v Intravenous Infusion BP



In keeping with good pharmaceutical practice dilutions of Zofran injection in intravenous fluids should be prepared at the time of infusion or stored at 2-8oC for no more than 24 hours before the start of administration.



Compatibility studies have been undertaken in polyvinyl chloride infusion bags and polyvinyl chloride administration sets. It is considered that adequate stability would also be conferred by the use of polyethylene infusion bags or Type 1 glass bottles. Dilutions of Zofran in sodium chloride 0.9%w/v or in glucose 5%w/v have been demonstrated to be stable in polypropylene syringes. It is considered that Zofran injection diluted with other compatible infusion fluids would be stable in polypropylene syringes.



Compatibility with other drugs: Zofran may be administered by intravenous infusion at 1mg/hour, e.g. from an infusion bag or syringe pump. The following drugs may be administered via the Y-site of the Zofran giving set for ondansetron concentrations of 16 to 160 micrograms/ml (e.g. 8 mg/500 ml and 8 mg/50 ml respectively);



Cisplatin: Concentrations up to 0.48 mg/ml (e.g. 240 mg in 500 ml) administered over one to eight hours.



5-Fluorouracil: Concentrations up to 0.8 mg/ml (e.g. 2.4g in 3 litres or 400mg in 500ml) administered at a rate of at least 20 ml per hour (500 ml per 24 hours). Higher concentrations of 5-fluorouracil may cause precipitation of ondansetron. The 5-fluorouracil infusion may contain up to 0.045%w/v magnesium chloride in addition to other excipients shown to be compatible.



Carboplatin: Concentrations in the range 0.18 mg/ml to 9.9 mg/ml (e.g. 90 mg in 500 ml to 990 mg in 100 ml), administered over ten minutes to one hour.



Etoposide: Concentrations in the range 0.14 mg/ml to 0.25 mg/ml (e.g. 72 mg in 500 ml to 250 mg in 1 litre), administered over thirty minutes to one hour.



Ceftazidime: Doses in the range 250 mg to 2000 mg reconstituted with Water for Injections BP as recommended by the manufacturer (e.g. 2.5 ml for 250 mg and 10 ml for 2g ceftazidime) and given as an intravenous bolus injection over approximately five minutes.



Cyclophosphamide: Doses in the range 100 mg to 1g, reconstituted with Water for Injections BP, 5 ml per 100 mg cyclophosphamide, as recommended by the manufacturer and given as an intravenous bolus injection over approximately five minutes.



Doxorubicin: Doses in the range 10-100mg reconstituted with Water for Injections BP, 5 ml per 10 mg doxorubicin, as recommended by the manufacturer and given as an intravenous bolus injection over approximately 5 minutes.



Dexamethasone: Dexamethasone sodium phosphate 20mg may be administered as a slow intravenous injection over 2-5 minutes via the Y-site of an infusion set delivering 8 or 32mg of ondansetron diluted in 50-100ml of a compatible infusion fluid over approximately 15 minutes. Compatibility between dexamethasone sodium phosphate and ondansetron has been demonstrated supporting administration of these drugs through the same giving set resulting in concentrations in line of 32 microgram - 2.5mg/ml for dexamethasone sodium phosphate and 8 microgram - 1mg/ml for ondansetron.



Administrative Data


7. Marketing Authorisation Holder



Glaxo Operations UK Limited,



Greenford Road,



Greenford,



Middlesex, UB6 0HE



Trading as



GlaxoSmithKline UK



Stockley Park West



Uxbridge



Middlesex UB11 1BT



8. Marketing Authorisation Number(S)



PL00004/0375



9. Date Of First Authorisation/Renewal Of The Authorisation



23 October 2001



10. Date Of Revision Of The Text



10 October 2011



11. LEGAL STATUS


POM




No comments:

Post a Comment