Notes
Article history
The research reported in this issue of the journal was commissioned and funded by the HTA Programme on behalf of NICE as project number 07/35/01. The protocol was agreed in October 2007. The assessment report began editorial review in February 2008 and was accepted for publication in July 2008. The authors have been wholly responsible for all data collection, analysis and interpretation, and for writing up their work. The HTA editors and publisher have tried to ensure the accuracy of the authors’ report and would like to thank the referees for their constructive comments on the draft document. However, they do not accept liability for damages or losses arising from material published in this report.
Declared competing interests of authors
None
Permissions
Copyright statement
© 2009 Queen’s Printer and Controller of HMSO. This monograph may be freely reproduced for the purposes of private research and study and may be included in professional journals provided that suitable acknowledgement is made and the reproduction is not associated with any form of advertising. Applications for commercial reproduction should be addressed to: NCCHTA, Alpha House, Enterprise Road, Southampton Science Park, Chilworth, Southampton SO16 7NS, UK.
2009 Queen’s Printer and Controller of HMSO
Chapter 1 Background
Description of health problem
Influenza is a highly contagious, acute febrile respiratory infection caused by the influenza virus. Cases typically occur in a seasonal pattern, with localised epidemics during the winter months. Illness is generally self-limiting but bacterial complications may arise. Such complications can be life threatening in nature, particularly in the elderly and in individuals with co-morbidities. Worldwide pandemics of influenza may occur when a major new subtype arises, often originating from avian influenza. Circumstances of pandemic influenza and avian influenza are beyond the scope of this review.
Symptoms
Common symptoms of influenza include respiratory symptoms such as sneezing, runny nose, cough, sore throat and coryza, and systemic symptoms such as fever, malaise, myalgia, chills and headache. There may also be gastrointestinal (GI) symptoms such as nausea, vomiting and diarrhoea. The duration of the acute illness is usually around 3–4 days, but cough and malaise may persist for 1–2 weeks. It is also possible for individuals to be asymptomatic while infected with the influenza virus. 1,2
The symptoms of influenza can also arise from a number of other infectious diseases, known as influenza-like illnesses (ILIs). These can be caused by adenoviruses, rhinovirus, respiratory syncytial virus, parainfluenza virus and bacterial infections. Confirmation of influenza infection requires laboratory methods such as viral culture or serological examination of antibody titres.
Prognosis, complications and mortality
Influenza infection can cause unpleasant symptoms for 1–2 weeks but is usually self-limiting and does not generally require treatment in otherwise healthy adults. However, influenza can lead to complications, including secondary bacterial infection. Complications are more common in certain at-risk groups, including those aged over 65 years, infants beyond the age when maternally-derived antibodies provide protection (and those with congenital abnormalities) and individuals with co-morbidities such as chronic respiratory disease [including asthma and chronic obstructive pulmonary disease (COPD)], cardiovascular disease, chronic renal disease, diabetes mellitus or immunosuppression. 2
Complications of influenza are often respiratory; these include primary viral pneumonia, secondary bacterial pneumonia, bronchitis, bronchiolitis in children, exacerbations of asthma and chronic respiratory disease and otitis media. Additionally, influenza can cause a range of non-respiratory symptoms and complications, including febrile convulsions, toxic shock syndrome, Reye syndrome, encephalopathy, transverse myelitis, pericarditis and myocarditis. Some of these complications may require hospitalisation and can be life threatening, especially in the elderly or those with underlying disease. 1,2
The presence of complications increases the risk of mortality due to influenza. The mortality risk is highest in individuals who are elderly or have co-morbidities. Estimates of deaths each year in the UK that are thought to be caused by influenza range from 12,000 to 13,800. 3,4,5 The UK epidemic of 1989–90 was estimated to have caused in excess of 29,000 deaths. 1
The influenza virus
Influenza is an orthomyxovirus, comprising a lipid membrane surrounding a matrix protein shell and a core consisting of seven or eight ribonucleic acid (RNA)–nucleoprotein complexes. There are three serotypes of influenza virus – influenza A, B and C – which differ in their core proteins. Influenza A and B are responsible for nearly all influenza-associated clinical illnesses. The influenza virus contains two surface glycoproteins, which act as powerful antigens: haemagglutinin (H antigen) and neuraminidase (N antigen). Haemagglutinin facilitates the entry of the virus into cells of the respiratory epithelium, while neuraminidase facilitates the release of newly-produced viral particles (virions) from infected cells. An ion channel protein is also embedded in the lipid membrane; in influenza A this is the M2 protein and in influenza B it is the NB protein. The influenza virus infects epithelial cells of the upper and lower respiratory tracts, attaching to the cell membranes, invading the host cell and using the host cell machinery to reproduce. New viral particles are released by lysis (breaking open) of the host cells, which damages the epithelium and increases susceptibility to secondary bacterial infections. 6
Strains and subtypes
The World Health Organization (WHO) classification system for influenza is based on the antigenic type of the nucleoprotein core (A, B or C), the geographical location of first isolation, the strain serial number, the year of isolation and (for influenza A) the haemagglutinin (H) and neuraminidase (N) subtypes, with each item separated by a slash, e.g. A/Wuhan/359/95 (H3N2).
New strains and subtypes of influenza are produced as a result of ‘antigenic drift’ and ‘antigenic shift’. Antigenic drift arises from gene mutations causing changes in the amino acid sequence of haemagglutinin or neuraminidase, the main antigens associated with immunity, leading to changes in the antigenic nature of the virus, i.e. a new strain of influenza (within a subtype). Antigenic drift is associated with annual outbreaks, as the virus is able to infect individuals who had developed immunity to previous strains. Many individuals are likely to retain partial immunity, although infants have little or no immunity. Influenza A undergoes antigenic drift to a greater extent than influenza B.
Antigenic shift is said to occur when an entirely new subtype of influenza A is introduced into the population, causing disease and onward human-to-human transmission. Antigenic shift occurs when the H and/or N of the new subtype is introduced into humans from the avian reservoir of infection, primarily ducks that serve as a reservoir for 16 different subtypes of H and nine subtypes of N for the influenza A virus. Other animal reservoirs may also be implicated in antigenic shift. Antigenic shift occurred in 1918, when an H1N1 influenza A virus adapted to man. It occurred also in 1957 and 1968, when the genomes of the circulating human viruses were mixed with those of avian origin by genetic reassortment; this process of ‘gene shuffling’ occurs during dual infections with influenza A viruses of differing subtypes. Antigenic shift results in ‘pandemic influenza’ because populations across the world have little or no immunity to the new strains. Pandemics cause a very high morbidity and mortality burden;7 the 1918–19 pandemic is estimated to have caused up to 40 million deaths worldwide. Pandemics usually originate in Asia where chickens, ducks, pigs and humans live in very close proximity and where other social factors favour interspecies transmission of virus. However, as discussed above, pandemic influenza and avian influenza are not considered within this review.
Transmission
Influenza virus is passed easily from person to person and is spread by virus-laden respiratory secretions. Most infections appear to be transmitted by droplets that are expelled during coughing and sneezing rather than by aerosols. The incubation period is 1–3 days. People with influenza may begin shedding virus 1–2 days before symptoms appear. Nasal shedding peaks about 48 hours after onset of symptoms and adults usually remain infectious for up to 1 week (up to 2 weeks in children; viral shedding may also be prolonged in immunocompromised individuals). 2
Epidemiology
Seasonal outbreaks of infection with influenza occur most years during the winter months in the northern hemisphere. The UK influenza season may run from week 40 to week 25, but occurs typically between December and March. 8 Illnesses resembling influenza that occur in the summer are usually caused by other viruses. 8 Infections with influenza A account for approximately 80% of outbreaks, while influenza B accounts for approximately 20%. 9 Comparative studies indicate that A/H3N2 infections produce more severe illness than A/H1N1 infections and that influenza B is intermediate in severity. 2 Typically, there is an annual outbreak which appears abruptly, peaks within 2–3 weeks and lasts for around 5–7 weeks. Successive or overlapping waves of infection by different subtypes of influenza A or by influenza A and B may result in a more prolonged period of disease activity. 10
Influenza is a common condition that may affect all age groups. However, the risk of an individual contracting the disease depends on a number of factors, including the virulence of the circulating strain, the natural level of immunity (which depends on past exposure to influenza virus or vaccination, and the degree of cross-immunity to the circulating strain), health status, age (both those aged over 65 years and the very young are at increased risk) and living arrangements. Influenza outbreaks can occur within establishments where several people live or work in close proximity, e.g. residential homes, hospitals, schools and prisons. In addition, the virus is transmitted quite frequently between individuals who live in the same house. Many studies worldwide have shown that the highest attack rates occur in young children and that school-aged children play a central role in the dissemination of influenza in households and the community. 10
Incidence
Influenza activity during recent years is illustrated in Figures 1 and 2. The rate of general practitioner (GP) consultations for ILI is monitored in the UK, and thresholds for use in England are defined by the Health Protection Agency (HPA) as follows:8
-
Baseline rate: fewer than 30 new GP consultations per 100,000 population per week.
-
Normal seasonal activity: 30–200 new GP consultations per 100,000 population per week.
-
Epidemic activity: more than 200 new GP consultations per 100,000 population per week.
The thresholds for Wales are slightly different: the baseline rate is fewer than 25 new GP consultations per 100,000 population per week, normal seasonal activity relates to 25–100 new consultations and epidemic activity is defined as more than 400 new consultations per 100,000 population per week.
It should be noted that, since influenza activity varies from season to season, attack rates, complications and mortality rates would also be anticipated to vary.
Impact of influenza and significance for the National Health Service (NHS)
For most people, influenza causes illness lasting 1–2 weeks. A proportion of individuals may experience asymptomatic infection or mild illness. However, the disease can lead to complications and mortality, particularly in the elderly or those with certain co-morbidities.
In terms of resource implications, influenza causes an increase in GP consultations, medical treatment and hospitalisations, as well as increased absence from work. In primary care, adults aged 15–64 years account for most consultations for influenza-related illness. In a large UK study of subjects who had one or more diagnoses of influenza or ILI recorded within the General Practitioner Research Database (GPRD), 59.4% received prescription medications, the most frequently prescribed being antibiotics (45.2%) and antipyretics/analgesics (22.5%). 12 Patients with influenza were approximately six times more likely to use prescription medications than a matched control sample. 12 The incidence of consultations due to influenza across the study period was reported as being 14.5 per 1000 person-years. 12 Complications arising from influenza may require hospitalisation, particularly in elderly people with underlying cardiopulmonary disorders. 13
The prevention of influenza also has resource implications for the NHS. In the UK, groups recommended for influenza vaccination include people at risk of complications from influenza [those aged over 65 years; individuals with chronic respiratory disease, chronic cardiovascular disease, chronic renal disease, chronic liver disease, chronic neurological disease or diabetes mellitus; the immunosuppressed; individuals with human immunodeficiency virus (HIV) infection; and people in residential homes (elderly or other long-stay)], the carers of dependents whose welfare would be put at risk should their carer fall ill and health-care workers involved directly in patient care. Vaccination may also be considered for social care workers involved directly in care and household contacts of immunosuppressed individuals. 14 The requirement for influenza vaccination has also been extended to poultry workers, in order to reduce the risk of the development of a potentially serious new variant as a result of co-infection with avian and human influenza strains. 15 Therefore, the guidelines for vaccination cover both healthy individuals and people with underlying medical conditions. Prophylaxis with the antiviral drug oseltamivir is currently recommended by the National Institute for Health and Clinical Excellence (NICE) for at-risk persons who are not adequately protected by vaccination and have been exposed to influenza (and for at-risk persons living in residential homes who have been exposed to influenza, irrespective of vaccination status), provided that the individual can start taking oseltamivir within 48 hours of exposure to influenza. 16 These guidelines are described in more detail in Current usage in the NHS (later in this chapter).
Measurement of influenza activity in the community
Influenza has no pathognomonic features and can manifest itself, as can other respiratory viruses, in a range of ways, such as the common cold, bronchitis, bronchiolitis, exacerbations of asthma or COPD, pneumonia, croup and febrile convulsions. Therefore, the level of influenza activity in a community is quantified by a combination of two factors: (1) the number of cases of illness attributed to ILI (based on e.g. the number of clinic visits or absences from school/work) and (2) the laboratory-based identification of influenza virus in samples from individuals with ILI.
In 1947, WHO established a global influenza surveillance system (a network of laboratories) to monitor the emergence and spread of new strains of influenza. The information generated by this system aids the development of vaccines against currently-circulating influenza strains. Vaccination is an important aspect of influenza prophylaxis and the degree of match between vaccine and circulating strains within a particular season has considerable implications for the control of influenza activity. In the UK, the HPA monitors and records the incidence of seasonal influenza and uptake of seasonal influenza vaccine. The Centre for Infections conducts surveillance of influenza activity in the UK, carries out laboratory tests to identify which strains are in circulation and communicates this information to health professionals and the public. 8
Diagnosis
Influenza-like illness can be defined clinically according to symptoms; the exact definition varies, with different trials of influenza prevention using a range of indicators, often including raised temperature (usually ≥ 37.8°C) and/or symptoms such as cough, headache, sore throat or myalgia.
To determine whether an individual case of ILI is true influenza, presence of the influenza virus must be determined in a laboratory test. This may consist of isolation of influenza virus from a nose-and-throat swab or nasopharyngeal wash taken from the patient, by means of either viral culture or polymerase chain reaction (PCR). In addition, serum samples from the patient may be tested for the presence of influenza-specific antibodies using a haemagglutination inhibition assay (HAI); influenza infection is usually defined as a fourfold or higher increase in influenza-specific HAI titre between baseline and post-infection serum samples (known as seroconversion). Many influenza studies use both viral culture and HAI serum testing, while some also use PCR, and generally a positive result on one or more of the tests is taken to indicate influenza infection. However, laboratory confirmation of influenza would not routinely be carried out on people presenting to their GP with ILI. 1
Current service provision
Management of disease
The symptoms of influenza and other ILI are often self-limiting and require no medical intervention. Over-the-counter medications are available for symptomatic relief of influenza. The presence of secondary complications of influenza typically requires treatment including antibiotics, and may require hospitalisation.
NICE currently recommends zanamivir and oseltamivir for the treatment of at-risk adults who present with ILI and who can start therapy within 48 hours of the onset of their symptoms. 17 Oseltamivir is recommended for the treatment of children who present with ILI and who can start therapy within 48 hours of the onset of symptoms. 17 At-risk individuals are defined within the NICE guidance as those who:
-
have chronic respiratory disease (including asthma and COPD)
-
have significant cardiovascular disease (excluding people with hypertension only)
-
have chronic renal disease
-
are immunocompromised
-
have diabetes mellitus
-
are aged 65 years or older. 17
It should be noted that the current guidance for influenza vaccination differs to that outlined above in that, in addition to the at-risk groups defined above, vaccination is recommended for patients with chronic liver disease or chronic neurological disease and also for individuals who live within long-stay residential care facilities, carers, health-care workers and poultry workers. 15,18,19
Current service cost
There is very limited evidence concerning the total costs of treating influenza and ILI in the UK. The current value of the UK antiviral market for the prophylaxis and treatment of influenza has been estimated at approximately £800,000, of which around 89% is attributable to oseltamivir. 20 However, the true cost of managing influenza is likely to be considerably higher as a result of the additional costs of vaccination and the management of secondary complications arising from influenza infection.
Variation in services and/or uncertainty about best practice
There is currently relatively little antiviral usage in the UK, possibly as a result of lower levels of virus activity and/or consultation rates than in previous decades. In contrast, the use of oseltamivir in Japan has increased in recent years. 21
It should be noted that the market authorisations for the use of antiviral post-exposure prophylaxis stipulate that prophylaxis should be initiated within a specified period of exposure to an index case. This stipulation requires that patients present to their GP promptly, the timescale being affected by an individual’s propensity to seek medical care and issues relating to access to GP services.
There is variation in terms of the uptake of vaccination in indicated subgroups. Recent monitoring data from the HPA suggest that the uptake of influenza vaccination is around 79% in individuals aged over 65 years and around 42% in at-risk individuals aged under 65 years.
Relevant national guidelines
NICE has issued guidance relating to the use of amantadine and oseltamivir in prophylaxis16 and zanamivir, oseltamivir and amantadine in the treatment of influenza. 17 These recommendations are outlined in detail in Current usage in the NHS (see below).
In addition to national policy for influenza vaccination in at-risk groups, vaccination for people aged 65 years and above was promoted within the National Service Framework for Older People22 and for people with coronary heart disease in the National Service Framework for Coronary Heart Disease. 23
Description of technology under assessment
Summary of interventions
The clinical effectiveness and cost-effectiveness of amantadine, oseltamivir and zanamivir in the prophylaxis of influenza have been evaluated in this assessment. The following section summarises the product characteristics of each of these interventions using the Summary of Product Characteristics (SPC) for each drug24–29 (obtained from the electronic Medicine Compendium at www.medicines.org.uk) and information from the British National Formulary (BNF). 14
Amantadine (Lysovir®, Alliance Pharmaceuticals)
Description of intervention
Amantadine is a symmetrical C-10 primary amine with a cage-like structure, which is water soluble in hydrochloride salt form. 30 Amantadine hydrochloride exerts an antiviral effect on influenza type A by means of inhibition of the M2 ion channel, which results in the blocking of viral replication. 30 The antiviral activity of amantadine is restricted to influenza A. In addition, amantadine has weak dopamine agonist activity.
Licensed indications
Amantadine hydrochloride is indicated for:
-
the treatment of and prophylaxis against signs and symptoms caused by influenza A infection (as Lysovir, Alliance Pharmaceuticals)
-
the treatment of Parkinson’s disease (but not drug-induced extrapyramidal symptoms) (as Symmetrel®, Alliance Pharmaceuticals)
-
the treatment of herpes zoster (as Symmetrel).
Dosage and administration
Lysovir is available as reddish-brown, hard, gelatine capsules containing 100 mg amantadine hydrochloride, which are ingested orally. Symmetrel is available as 50 mg/5 ml syrup.
Adults and children over 10 years: 100 mg/day for as long as protection from influenza is required, usually for up to 6 weeks, or with influenza vaccination for 2–3 weeks after vaccination.
Treatment should be initiated within 48 hours of the onset of symptoms.
-
Adults: 100 mg/day for 4–5 days
-
Children aged 10–15 years: 100 mg/day for 4–5 days
-
Children under 10 years of age: dosage not established
-
Adults over 65 years of age: owing to the longer elimination half-life and reduced capacity for renal clearance of amantadine in elderly patients, a reduced dose of < 100 mg/day or 100 mg given at intervals of ≥ 1 day may be appropriate
-
Patients with renal impairment: dosage should be adjusted by reducing total daily dose or by increasing dosage interval in line with clearance of creatinine. Guidance is as follows:
Creatinine clearance (ml/minute) | Dose |
---|---|
< 15 | Lysovir contraindicated |
15–35 | 100 mg every 2–3 days |
> 35 | 100 mg/day |
Contraindications
Amantadine hydrochloride is contraindicated in patients who:
-
have epilepsy
-
have a history of gastric ulceration
-
have severe renal impairment
-
are pregnant, wish to become pregnant or are breastfeeding
-
have known hypersensitivity to amantadine or any excipients.
Cautions
Amantadine hydrochloride should be administered with caution to patients who:
-
have hepatic impairment
-
have renal impairment
-
have congestive heart disease (as the drug may cause exacerbation of oedema)
-
experience confusion or hallucinations
-
have underlying psychiatric disorders
-
are elderly
-
are receiving concomitant medications with potential to affect the central nervous system (CNS).
Abrupt withdrawal of amantadine therapy should be avoided in patients with Parkinson’s disease.
It should be noted that, while resistance to amantadine is well documented,30 it has been reported that levels of resistance among influenza isolates have risen dramatically on an international scale. 31 Development of resistance can occur relatively rapidly during treatment and can lead to the failure of prophylaxis, for example within the management of outbreaks of influenza in long-term care settings. 32
Adverse events
Adverse events associated with amantadine hydrochloride include anorexia, nausea, nervousness, insomnia, dizziness, inability to concentrate, convulsions, hallucinations, blurred vision, GI effects, livedo reticularis, peripheral oedema and skin rashes. It has been documented that adverse effects can occur frequently among recipients. 33 Central nervous system adverse events have been described as occurring most notably within the elderly population.
Oseltamivir (Tamiflu®, Roche)
Description of intervention
Oseltamivir is a neuraminidase inhibitor that exerts an antiviral effect on influenza A and B. 34 The drug inhibits viral release, preventing subsequent infection of adjacent cells. The SPC emphasises that oseltamivir is not a substitute for vaccination and that use should take into account official recommendations and variability of epidemiology and impact across patient populations and geographical locations.
Licensed indications
Oseltamivir is indicated for:
the post-exposure prophylaxis of influenza in patients aged 1 year and above who have had contact with a clinically diagnosed influenza index case when influenza is circulating in the community. The SPC states that the administration of oseltamivir should be decided on a case-by-case basis and that seasonal prophylaxis in subjects aged 1 year and above may be considered in exceptional circumstances (such as in the case of mismatch between vaccine and circulating strains of influenza or in the event of a pandemic).
the treatment of influenza in patients aged 1 year and above who present with influenza symptoms when influenza is circulating in the community. Treatment is effective when initiated within 48 hours of onset of the first symptoms.
Dosage and administration
Tamiflu is administered orally and is available as grey-yellow capsules containing 75 mg oseltamivir (as phosphate), 45 mg oseltamivir (as phosphate) or 30 mg oseltamivir (as phosphate), and as a powder (as phosphate) for reconstitution with water (12 mg/ml) as an oral suspension. The administration of 75 mg doses can be made up of one 75 mg capsule or one 30 mg capsule plus one 45 mg capsule or one 30 mg capsule plus one 45 mg dose of suspension. It should be noted that the BNF lists only the 75 mg dose of Tamiflu in capsule form. The administration of suspension is recommended in patients who are not able to swallow capsules. The SPC recommends that powder for oral suspension should be reconstituted by a pharmacist before it is dispensed to the patient.
Prophylaxis should be initiated as soon as possible within 48 hours of exposure to the index case.
Post-exposure prophylaxis
-
Adults and adolescents over 13 years: 75 mg for 10 days, for up to 6 weeks during an epidemic
-
Children aged 1–13 years: body weight under 15 kg, 30 mg once daily; body weight 15–23 kg, 45 mg once daily; body weight 23–40 kg, 60 mg once daily; body weight over 40kg, adult dose.
During a community outbreak of influenza, the recommended dose is 75 mg once daily for up to 6 weeks.
Dose adjustment is recommended for patients with severe renal impairment as follows:
Creatinine clearance (ml/minute) | Dose |
---|---|
> 30 | 75 mg once daily |
> 10 to ≤ 30 | 75 mg every second day |
or 30 mg suspension once daily | |
or 30 mg capsules once daily | |
≤ 10 | Not recommended |
Dialysis patients | Not recommended |
Treatment should be initiated as soon as possible within 48 hours of onset of symptoms.
-
Adults and adolescents over 13 years: 75 mg every 12 hours for 5 days
-
Children aged 1–13 years: body weight under 15 kg, 30 mg every 12 hours; body weight 15–23 kg, 45 mg every 12 hours; body weight 23–40 kg, 60 mg every 12 hours; body weight over 40 kg, adult dose.
Dose adjustment is recommended for patients with severe renal impairment as follows:
Creatinine clearance (ml/minute) | Dose |
---|---|
> 30 | 75 mg twice daily |
> 10 to ≤ 30 | 75 mg once daily |
or 30 mg suspension twice daily | |
or 30 mg capsule twice daily | |
≤ 10 | Not recommended |
Dialysis patients | Not recommended |
No adjustment of dose is required in the elderly, with the exception of patients with severe renal impairment. There is insufficient evidence to recommend dosage adjustment in children with renal impairment.
Contraindications
Oseltamivir is contraindicated in patients who have hypersensitivity to oseltamivir or any of its excipients.
Cautions
Oseltamivir should be administered with caution to patients who:
-
have renal impairment
-
are pregnant or breastfeeding
-
have conditions of such severity or instability that imminent hospitalisation may be required
-
are immunocompromised
-
have chronic cardiac and/or respiratory disease.
The dose should be reduced if creatinine clearance in patients is < 10–30 ml/minute and administration should be avoided if creatinine clearance is < 10 ml/minute.
Adverse events
Adverse events associated with oseltamivir include nausea, vomiting, abdominal pain, diarrhoea, dyspepsia, headache, fatigue, insomnia, dizziness, conjunctivitis, epistaxis, skin rashes, and – in very rare cases – hepatitis, Stevens–Johnson syndrome and toxic epidermal necrolysis. Neuropsychiatric disorders in children have also been reported.
Zanamivir [Relenza®, GlaxoSmithKline (GSK)]
Description of intervention
Zanamivir is a neuraminidase inhibitor that inhibits the replication of influenza A and B. 34 The SPC states that zanamivir is not a substitute for vaccination, as protection only lasts for as long as the drug is administered, and that the use of zanamivir should be decided on a case-by-case basis according to circumstances and the population in need of protection. The SPC recommends that the drug should be used only when reliable epidemiological data confirm the circulation of influenza in the community. Use of zanamivir should take into account official recommendations, epidemiological variation and varying impact of influenza across patient populations and geographical locations.
Licensed indications
Zanamivir is indicated for:
-
the post-exposure prophylaxis of influenza A and B in adults and children aged 5 years and above who have had contact with a clinically diagnosed case of influenza in a household. Relenza may be considered for use in seasonal prophylaxis in exceptional circumstances, for example when there is mismatch between circulating or vaccine strains or in the event of a pandemic.
-
the treatment of influenza A and B in adults and children aged 5 years and above who present with ILI when influenza is active in the community.
Dosage and administration
Relenza is available in the form of predispensed dry powder for inhalation in blisters containing 5 mg zanamivir per blister, delivered by means of oral inhalation using a Diskhaler® device. Each inhalation delivered (quantity released via mouthpiece of the Diskhaler) contains 4 mg zanamivir (the remainder appears to be lost in the inhalation process and is presumably retained within the Diskhaler apparatus).
Prophylaxis should be initiated as soon as possible and within 36 hours of exposure to an infected index case.
-
Adults and children aged 5 years and above: 10 mg once daily (i.e. two inhalations) for 10 days.
During an epidemic, prophylaxis may be administered.
-
Adults and children aged 12 years and above (as recommended in the BNF):18 10 mg once daily for up to 28 days.
Treatment should be initiated as soon as possible and within 48 hours of onset of symptoms in adults and within 36 hours of onset of symptoms in children.
-
Adults and children aged 5 years and above: 10 mg twice daily for 5 days.
No dose modification is required for individuals with renal or hepatic impairment or for elderly patients.
Contraindications
Zanamivir is contraindicated in patients who:
-
are pregnant or breastfeeding
-
are hypersensitive to any ingredient of the preparation.
Cautions
Zanamivir should be administered with caution to patients who:
-
have asthma and chronic pulmonary disease
-
have uncontrolled chronic illness
-
are immunocompromised
-
are pregnant.
According to the BNF, zanamivir should be used with caution in pregnancy and is contraindicated in breastfeeding women. However, according to the FDA, pregnancy and breastfeeding are cautions rather than contraindications. Other inhaled drugs, such as asthma medication, should be administered before zanamivir.
Adverse events
The following adverse events associated with zanamivir are described as occurring very rarely: bronchospasm, respiratory impairment, angioedema, urticaria and skin rashes.
Identification of important subgroups
A number of important subgroups should be considered in relation to the use of antivirals for influenza prophylaxis. Subgroups viewed to be at risk of developing influenza-associated complications were described earlier in this chapter (see Description of health problem). Within the guidance issued by NICE for the prophylaxis16 and treatment17 of influenza, populations viewed to be at risk include individuals who:
-
are aged 65 years or above
-
have chronic lung disease (including asthma and COPD)
-
have significant heart disease (excluding people with hypertension only)
-
have chronic renal disease
-
have diabetes mellitus
-
are immunocompromised.
Current usage in the NHS
Guidance was issued by NICE relating to the use of oseltamivir and amantadine in the prophylaxis of influenza16 and for the use of zanamivir, oseltamivir and amantadine for the treatment of influenza. 17 These guidance documents were issued in accordance with the expectation that vaccination would continue to be the mainstay of influenza prevention. Issued guidance relates solely to circumstances where it is known that influenza A or B is circulating in the community. To this end, NICE recommended that community-based virological systems should be used to monitor the circulation of influenza virus in the community. Guidance issued does not pertain to the circumstances of a pandemic or impending pandemic, or to the emergence of a widespread epidemic of a new influenza strain to which there is little or no community resistance.
At-risk groups were defined according to NICE guidance as described above.
NICE recommended that oseltamivir should be used in the prevention of influenza as follows:
-
for individuals who are aged 13 years and above
-
– and belong to an at-risk group
-
– and are not effectively protected by vaccination (e.g. individuals who have not received an influenza vaccination for that season, for whom vaccination may be contraindicated or has yet to take effect, or for whom vaccination has been undertaken but there is a mismatch between vaccine and circulating strains)
-
– and have been in close contact with an index case with ILI
-
– and can start taking oseltamivir within 48 hours of contact with the index case
-
-
for individuals who are aged 13 years and above
-
– and belong to an at-risk group (whether or not they have been vaccinated)
-
– and live in a residential care establishment where another individual has ILI (resident or staff member)
-
– and can start taking oseltamivir within 48 hours of contact with the index case.
-
For the purposes of the guidance, a residential care establishment was classed as a location where an at-risk person lived long term in order to receive continuing care alongside other individuals with care needs. Exposure to ILI was defined as having close contact with an individual who resides in the same home environment as a person who has been experiencing symptoms of ILI.
NICE did not recommend that oseltamivir should be used in post-exposure prophylaxis of influenza in healthy people aged under 65 years. The use of oseltamivir in seasonal prophylaxis was not recommended. The use of amantadine in post-exposure and seasonal influenza prophylaxis was not recommended.
It was recommended that amantadine should not be used in the treatment of influenza and that zanamivir or oseltamivir should not be used in the treatment of individuals who are healthy and are not at risk of developing complications from influenza.
The use of zanamivir and oseltamivir in line with their licensed indications was recommended for the treatment of:
-
adults (aged over 12 years) belonging to an at-risk group
-
– who present with ILI
-
– and can begin treatment within 48 hours of the onset of symptoms.
-
The use of oseltamivir in line with licensed indications was recommended for the treatment of:
-
children (aged over 1 year) belonging to an at-risk group
-
– who present with ILI
-
– and can begin treatment within 48 hours of the onset of symptoms.
-
It should be noted that, although the use of amantadine in the prophylaxis and treatment of influenza was not recommended by NICE, this drug is also licensed for the treatment of Parkinson’s disease and herpes zoster.
Anticipated costs associated with intervention
The costs associated with amantadine, oseltamivir and zanamivir are dependent on the setting for the prophylaxis, the mode of administration and the age of the patient (oseltamivir only). Acquisition costs for post-exposure prophylaxis and seasonal prophylaxis are presented in Tables 1 and 2 respectively. The capsule/tablet forms of prophylaxis are likely to be most relevant to adult populations as these allow for more precise measurements of dosage; for oseltamivir in children aged under 13 years, dosage is usually adjusted according to body weight. Prophylaxis is typically given to children under 13 years in suspension form based on body mass. The reader should note that while the BNF lists only 75 mg capsules and suspension, the SPC accessed via the electronic Medicine Compendium26,27 (www.medicines.org.uk) cites the additional availability of 30 mg and 45 mg capsules of oseltamivir. Amantadine, oseltamivir and zanamivir are self-administered and do not require administration by a health-care professional. It should be noted that diagnostic testing for influenza is not standard practice in the UK and is unlikely to represent a relevant cost associated with these products. The reader should also note that in November 2007 the manufacturer of zanamivir (GSK) applied to the Department of Health for a price modulation of two of their drugs, one of which was zanamivir. The current list price for zanamivir is £24.55 (five disks, four blisters per disk); the proposed price for zanamivir is £16.36 (Toni Maslen, Health Outcomes Programme Leader, GSK, 2007, personal communication). This price reduction was approved by the Department of Health with effect from 1 February 2008 but was not listed in the BNF14 at the time of submission of this report.
Regimen | Age (years) | Prophylaxis days/course | mg/dose | Doses/day | mg/course | Doses/pack | Packs required | Cost/pack | Cost/course |
---|---|---|---|---|---|---|---|---|---|
Amantadine (five-cap pack, Lysovir) | > 10 | 10 | 100 | 1 | 1000 | 5 | 2 | £2.40 | £4.80 |
Amantadine (14-cap pack, Lysovir) | > 10 | 10 | 100 | 1 | 1000 | 14 | 1 | £4.80 | £4.80 |
Amantadine (56-cap pack, Symmetrel) | > 10 | 10 | 100 | 1 | 1000 | 56 | 1 | £16.88 | £16.88 |
Amantadine (150 ml syrup, Symmetrel) | > 10 | 10 | 100 | 1 | 1000 | 15 | 1 | £5.55 | £5.55 |
Oseltamivir (cap) – adults | > 13 | 10 | 75 | 1 | 750 | 10 | 1 | £16.36 | £16.36 |
Oseltamivir (suspension) – adults | > 13 | 10 | 75 | 1 | 750 | 12 | 1 | £16.36 | £16.36 |
Oseltamivir (suspension) – children < 15 kg | < 14 | 10 | 30 | 1 | 300 | 30 | 1 | £16.36 | £16.36 |
Oseltamivir (suspension) – children 15–23 kg | < 14 | 10 | 45 | 1 | 450 | 20 | 1 | £16.36 | £16.36 |
Oseltamivir (suspension) – children 23–40 kg | < 14 | 10 | 60 | 1 | 600 | 15 | 1 | £16.36 | £16.36 |
Oseltamivir (suspension) – children > 40 kg | < 14 | 10 | 75 | 1 | 750 | 12 | 1 | £16.36 | £16.36 |
Zanamivir (powder) | > 5 | 10 | 10 | 1 | 100 | 10 | 1 | £24.55 | £24.55 |
Regimen | Age (years) | Prophylaxis days/course | mg/dose | Doses/day | mg/course | Doses/pack | Packs required | Cost/pack | Cost/course |
---|---|---|---|---|---|---|---|---|---|
Amantadine (five-cap pack, Lysovir) | > 10 | 42 | 100 | 1 | 4200 | 5 | 9 | £2.40 | £21.60 |
Amantadine (14-cap pack, Lysovir) | > 10 | 42 | 100 | 1 | 4200 | 14 | 3 | £4.80 | £14.40 |
Amantadine (56-cap pack, Symmetrel) | > 10 | 42 | 100 | 1 | 4200 | 56 | 1 | £16.88 | £16.88 |
Amantadine (150 ml syrup, Symmetrel) | > 10 | 42 | 100 | 1 | 4200 | 15 | 3 | £5.55 | £16.65 |
Amantadine following vaccination (five-cap pack, Lysovir) | > 10 | 21 | 100 | 1 | 2100 | 5 | 5 | £2.40 | £12.00 |
Amantadine following vaccination (14-cap pack, Lysovir) | > 10 | 21 | 100 | 1 | 2100 | 14 | 2 | £4.80 | £9.60 |
Amantadine following vaccination (56-cap pack, Symmetrel) | > 10 | 21 | 100 | 1 | 2100 | 56 | 1 | £16.88 | £16.88 |
Amantadine following vaccination (150 ml syrup, Symmetrel) | > 10 | 21 | 100 | 1 | 2100 | 15 | 2 | £5.55 | £11.10 |
Oseltamivir (cap) – adults | > 13 | 42 | 75 | 1 | 3150 | 10 | 5 | £16.36 | £81.80 |
Oseltamivir (suspension) – adults | > 13 | 42 | 75 | 1 | 3150 | 12 | 4 | £16.36 | £65.44 |
Oseltamivir (suspension) – children < 15 kg | < 14 | 42 | 30 | 1 | 1260 | 30 | 2 | £16.36 | £32.72 |
Oseltamivir (suspension) – children 15–23 kg | < 14 | 42 | 45 | 1 | 1890 | 20 | 3 | £16.36 | £49.08 |
Oseltamivir (suspension) – children 23–40 kg | < 14 | 42 | 60 | 1 | 2520 | 15 | 3 | £16.36 | £49.08 |
Oseltamivir (suspension) – children > 40 kg | < 14 | 42 | 75 | 1 | 3150 | 12 | 4 | £16.36 | £65.44 |
Zanamivir (powder) | > 5 | 28 | 10 | 1 | 280 | 10 | 3 | £24.55 | £73.65 |
Chapter 2 Definition of the decision problem
NICE has previously issued guidance on the use of amantadine and oseltamivir for the prevention of influenza. 16 When the original NICE guidance was issued, the licensed indications for zanamivir did not extend to its use as prophylaxis. Marketing authorisation has since been given for the use of zanamivir for the prophylaxis of influenza. This review presents an updated assessment of new and existing evidence for amantadine and oseltamivir, and an assessment of the clinical effectiveness and cost-effectiveness of zanamivir for the prophylaxis of influenza in England and Wales.
Decision problem
The decision problem has been defined as described below.
Interventions
Three prophylactic interventions are included in this assessment:
-
amantadine (Lysovir or Symmetrel, Alliance Pharmaceuticals)
-
oseltamivir (Tamiflu, Hoffman–La Roche Pharmaceuticals)
-
zanamivir (Relenza, GlaxoSmithKline Pharmaceuticals).
Relevant comparators
Amantadine, oseltamivir and zanamivir are compared with each other and with no prophylaxis (in which subjects received one of the following: placebo, no treatment or expectant treatment following onset of symptomatic influenza).
Populations and relevant subgroups
The interventions are evaluated in the post-exposure prophylaxis and seasonal prophylaxis settings. In the post-exposure setting, the assessment evaluates the clinical effectiveness and cost-effectiveness of the interventions in adults and children who have been exposed to a clinically-diagnosed case of influenza. In reality, effectiveness would be in terms of exposure to an index case with ILI, which may or may not subsequently be confirmed as influenza. Post-exposure prophylaxis was considered in the prevention of transmission of influenza from index cases to household contacts and in outbreak control within establishments where members of a community live or work in close proximity, for example within long-term care settings and boarding schools. In the seasonal setting, the assessment evaluates the clinical effectiveness and cost-effectiveness of the interventions in adults and children for whom seasonal prophylaxis would be appropriate in exceptional circumstances. In this case, exceptional circumstances relate to a high degree of mismatch between the circulating influenza virus and vaccine strains; as noted below, the effectiveness of influenza prophylaxis in pandemic situations is beyond the remit of this assessment.
The clinical effectiveness and cost-effectiveness of influenza prophylaxis for people who are at a higher risk of influenza infection or complications were considered. Where evidence was available, vaccination status was also taken into consideration.
Overall aims and objectives of assessment
The objectives of the assessment are:
-
to evaluate the clinical effectiveness of amantadine, oseltamivir, and zanamivir in the prophylaxis of influenza in terms of cases prevented, complications prevented, health-related quality of life (HRQoL), mortality, hospitalisations prevented, length of influenza illness and time to return to normal activities
-
to evaluate the incidence and impact of treatment-related adverse events
-
to estimate the incremental cost-effectiveness of amantadine, oseltamivir and zanamivir in comparison with each other and no prophylaxis
-
to identify gaps in the existing evidence base and those areas requiring further primary research
-
to estimate the annual cost to the NHS.
As outlined in Chapter 1 and noted above, the remit of this assessment does not include the circumstances of a pandemic, an impending pandemic or a widespread epidemic of a new strain of influenza to which there is little or no community resistance. The economic analysis considers a ‘typical’ influenza season as well as the potential impact of higher attack rates and vaccine mismatch. The interventions are appraised according to their licensed indications, with guidance to be issued in accordance with relevant marketing authorisations.
Chapter 3 Assessment of clinical effectiveness
Methods for reviewing effectiveness
A systematic review of the clinical effectiveness of amantadine, oseltamivir and zanamivir for influenza prophylaxis was undertaken according to the general principles recommended in the quality of reporting of meta-analyses (QUOROM) statement. 35 Methods for the review are detailed below.
Identification of studies
Systematic searches were undertaken to identify studies relating to the clinical effectiveness of amantadine, oseltamivir and zanamivir in the prevention of influenza A and B. The search strategy comprised the following main elements:
-
searching of electronic databases listed below
-
contact with experts in the field
-
handsearching of bibliographies of retrieved papers
-
scanning of electronic archives of key journals for relevant evidence published within the preceding 12 months (searched October 2007).
Sources searched
The electronic databases searched included MEDLINE; MEDLINE In-Process; EMBASE; Cochrane Database of Systematic Reviews, Cochrane Controlled Trials Register, Biosciences Information Service (BIOSIS), Cumulative Index to Nursing and Allied Health Literature (CINAHL), Database of Abstracts of Reviews of Effectiveness (DARE), NHS Economic Evaluation Database (EED) and Health Technology Assessment (HTA) databases; Office of Health Economics Health Economic Evaluations Database (OHE HEED), National Research Register (NNR); Science Citation Index (SCI); Current Controlled Trials (CCT); and ClinicalTrials.gov. Searches were undertaken in July 2007. Sponsor submissions to NICE were also handsearched.
Keyword strategies
The search strategies included subject headings and free text terms, combined using Boolean logic, to identify all published and unpublished data relating to the prevention of influenza A and B. The MEDLINE search strategy is presented in Appendix 1.
Search restrictions
Searches were restricted by publication type to controlled clinical trials, systematic reviews and economics or quality of life studies. Searches were not restricted by the date of publication or by language.
Inclusion and exclusion criteria
The following inclusion criteria were used to identify relevant studies for inclusion in the assessment.
Populations
The included populations comprised:
-
adults and children who have been exposed to a clinically-diagnosed case of influenza (which may or may not be true influenza)
-
adults and children for whom seasonal prophylaxis would be appropriate in exceptional circumstances, such as in the event of mismatch between the circulating influenza virus and vaccine strains; for the purposes of this assessment, we have considered healthy and at-risk children, adults and the elderly.
Interventions
Interventions comprised the following medications used for influenza prophylaxis administered in line with current UK marketing authorisations:
-
amantadine
-
oseltamivir
-
zanamivir.
Trials of these interventions in seasonal prophylaxis and post-exposure prophylaxis (both in prevention of the transmission of influenza within households and in outbreak control in settings where individuals live or work in close proximity) were included in the review. Trials in which interventions were used in prophylaxis against experimentally-induced influenza in line with licensed indications were also included. The results of these challenge studies should be interpreted with caution owing to their limited external validity. These studies are presented to provide a comprehensive review of the effectiveness of prophylaxis; they were not used to inform the health economic model.
Comparators
Interventions were compared with each other and no prophylaxis (in which subjects received one of the following: placebo, no treatment or expectant treatment following onset of symptomatic influenza).
Outcomes
Outcomes considered included:
-
cases prevented (measured in terms of symptomatic, laboratory-confirmed influenza or, in the absence of this outcome, clinical illness and/or infection)
-
complications prevented
-
adverse events
-
HRQoL
-
mortality
-
hospitalisations prevented
-
length of influenza illness
-
time to return to normal activities
-
cost and cost-effectiveness (see Chapter 4).
Study type
The study employed randomised controlled trials (RCTs). Had evidence not been available from RCTs, other study types would have been considered according to the hierarchy of evidence. Systematic reviews were not included in the analysis, but were handsearched to identify RCTs meeting the inclusion criteria of this review and retained for discussion.
The following exclusion criteria were used:
-
intervention medications not used in accordance with their licensed indications
-
studies published only in languages other than English.
Based on the above inclusion/exclusion criteria, study selection was undertaken by one reviewer, with involvement of a second reviewer when necessary to provide consensus on inclusion or exclusion of studies.
Data abstraction strategy
Data were extracted with no blinding to authors or journal, and were extracted by one reviewer using a standardised form. Any studies giving rise to uncertainty were reviewed independently by a second reviewer, and discrepancies, for example where studies were not clearly reported, were resolved by discussion. All data abstraction was checked and confirmed by a second reviewer.
Critical appraisal strategy
The quality of included RCTs was assessed using quality criteria based on those developed by the NHS Centre for Reviews and Dissemination;36 these are presented in Appendix 2. The purpose of such quality assessment was to provide a narrative account of trial quality for the reader. Quality assessment was confirmed by a second reviewer.
Methods of data synthesis
The outcomes defined above were presented within a narrative synthesis. Where quantitative synthesis was considered to be appropriate, statistical meta-analysis was undertaken using a random-effects model using RevMan software (version 4.2.10) in order to calculate pooled estimates for RRs for outcomes of interest. The presence of heterogeneity within the identified evidence and the lack of any direct comparative RCTs of antiviral prophylaxis were considered to preclude the use of sensitivity analyses and mixed-treatment comparisons.
Efficacy data are presented as RRs and protective efficacy (PE) (PE = 1–RR, expressed as a percentage). Where the RR or PE values were not described in the study publication, or where the value differed (usually by only a small margin) from that calculated from the formula below, the RR was calculated by the Assessment Group using the following formula (and marked with †):
where a = event present for treatment group, b = event present for control group, c = event absent for treatment group and d = event absent for control group.
Where publications have reported a 95% confidence interval (CI) around the RR or PE, these have been presented. Where no CI was published, it was calculated using the following formula (and marked with †):
Results
Quantity and quality of research available
As a result of the searches outlined above, a total of 1010 citations were identified, following removal of duplicates, and were screened for inclusion in the review of clinical effectiveness (Figure 3). Two hundred and eighty citations were rejected at the title stage, yielding 730 abstracts for screening, of which 551 were rejected on examination of the abstract. Of 179 full papers retrieved, 153 were excluded (of which 18 were not available for retrieval by information specialists or could not be read as they were not available in English). Of these, seven citations were excluded, since the full text was not available in English. 37–43 The articles that could not be obtained were unlikely to be relevant for inclusion, as they appeared to be conference abstracts and discussion papers. Papers that were excluded after close scrutiny are presented in Appendix 6, together with the justification for their exclusion. Twenty systematic reviews were identified; these were handsearched and retained for discussion. Twenty-six citations relating to 22 RCTs were included in the review of clinical effectiveness. One additional unpublished report was provided as evidence as part of the submissions by sponsors and is also presented. 44
Quantity of research available
A total of 26 published references presenting findings from 22 RCTs were considered relevant for inclusion in the review of clinical effectiveness of amantadine, oseltamivir and zanamivir for the prophylaxis of influenza. An additional unpublished report was identified in the sponsor submissions and included in the assessment, resulting in a total of 23 RCTs. 44 One included reference45 was a report of a pooled analysis of data relating to post-exposure prophylaxis of influenza using oseltamivir and zanamivir based on included trials. 46–49 No ongoing trials or trials due to report that met the inclusion criteria were identified in searches. All included articles are described below and grouped by intervention.
Amantadine
A total of eight full papers reporting eight RCTs that investigated the prophylactic use of amantadine against influenza were identified. Characteristics of these studies are presented in Table 3.
Study | Population characteristics | Trial design arms (no. of patients in each arm) | Preventative strategy | Prophylaxis duration | Source of funding |
---|---|---|---|---|---|
Aoki et al., 198658 | Healthy adults in a military setting (age not defined) | T1: amantadine 100 mg/day: 1980–1, n = 74; 1981–2 under 28 years, n = 21, over 29 years, n = 29; 1982–3, n = 46 | Seasonal | 39 days (1980–1), 32 days (1982–3) | National Health Research and Development Program of Canada and the Canadian Foundation for the Advancement of Clinical Pharmacology |
Six to eight individuals in each study year immunised against influenza in previous years | T2: placebo: 1980–1, n = 48; 1981–2 under 28 years, n = 16, over 29 years, n = 18; 1982–3, n = 33 | ||||
Pettersson et al., 198055 | Elderly subjects (mean ages T1 = 77.4 years, T2 = 79.0 years) living in a residential home, vaccination status unclear, but discussion states no adequate vaccine available | T1: amantadine 100 mg/day: randomised, n = 94; completing study, n = 89 | Seasonal | 9 weeks | Medica Ltd and Orion Diagnostica Ltd |
T2: placebo: randomised, n = 101; completing study, n = 99 | |||||
Reuman et al., 198957 | Healthy unvaccinated adults aged 18–55 years living in the community | T1: amantadine 100 mg/day, n = 159 | Seasonal | Presumed 6 weeks | E.I. DuPont de Nemours and Company, Inc. |
T2: placebo, n = 159 | |||||
Payler and Purdham, 198459 | Adolescent males aged 13–19 years in boarding school setting, 87% vaccinated | T1: amantadine 100 mg/day: randomised, n = 299; final analysis, n = 267 | Outbreak control | 14 days | Study medication supplied by Ciba–Geigy Pharmaceuticals |
T2: no specific treatment: randomised, n = 307; final analysis, n = 269 | |||||
Smorodintsev et al., 197060,61 | Male adults (recruitment pool aged 18–30 years), presumed healthy, in semi-isolated engineering school populations | T1: amantadine 100 mg/day (50.7% of 10,053): assigned to group, n = 5092; onset of influenza prior to dosing, n = 441; regularly or irregularly taking amantadine, n = 4559 | Outbreak control | Five of seven populations dosed for 30 days, two populations dosed for 12 days | Study medication supplied by E.I. DuPont de Nemours and Company, Inc. |
T2: placebo (31.6% of 10,053): assigned to group, n = 3175; onset of influenza prior to dosing, n = 307; receiving placebo, n = 2804 (3175 – 307 = 2868, 2804 included in analysis) | |||||
T3: internal control: individuals at same engineering schools as amantadine and placebo groups, but living at home rather than at school; received no prophylaxis (10.0% of 10,053), n = 1011 | |||||
T4: external control: individuals at an eighth engineering school; received no prophylaxis (7.7% of 10,053), assigned to group, n = 775 | |||||
Reuman et al., 198957 | Healthy unvaccinated adults aged 18–40 years | T1: amantadine 100 mg/day, n = 20 | Experimentally-induced influenza | 8 days (3 days pre-challenge and 5 days post-challenge) | E.I. DuPont de Nemours and Company, Inc. |
T2: placebo, n = 19 | |||||
Sears and Clements, 198763 | Healthy adults aged 18–40 years | T1: amantadine 100 mg/day, n = 22 | Experimentally-induced influenza | 8 days | E.I. DuPont de Nemours and Company, Inc. |
T2: placebo, n = 22 | |||||
Smorodintsev et al., 197062 | Healthy adults (age not defined) | T1: amantadine 100 mg/day, n = 19 | Experimentally-induced influenza | 12 days (24 hours before challenge and daily for 11 days) | Study medication supplied by E.I. DuPont de Nemours and Company, Inc. |
T2: placebo, n = 31 |
The original HTA review reported by Turner et al. 10 assessed the use of amantadine in influenza prophylaxis in children (aged under 18 years) and the elderly (aged over 65 years) only, as a Cochrane review of the use of amantadine in adults had recently been reported. 50 This Cochrane review has been subsequently updated33 and was handsearched to identify any additional citations for inclusion in the current review. Turner et al. 10 identified three trials of amantadine prophylaxis undertaken in children. 51–53 However, these studies and an additional trial54 are not included in this technology assessment report, as the dosage of amantadine is not established in children under 10 years of age according to licensed indications. Two prevention trials in the elderly were also included in the original assessment. 55,56 Of these studies, only the findings presented by Pettersson and colleagues55 are included in this update, while the trial reported by Leeming56 was excluded, as twice the currently licensed dose was administered to participants.
An additional seven trials of amantadine prophylaxis were identified by our searches. These included two studies that evaluated seasonal prophylaxis in healthy adults. 57,58 Further trials described amantadine prophylaxis in outbreak control in healthy adolescents in a boarding school59 and in adults in semi-isolated engineering school populations. 60,61 A further three reports of the prophylactic efficacy of amantadine against experimentally-induced influenza were identified. 57,62,63 One of these papers presented results from two separate trials examining the use of amantadine in seasonal prophylaxis and experimentally-induced influenza studies. 57
Four trials included in the Cochrane review of amantadine and rimantadine in influenza A in adults33 have been included in our assessment. 55,57,59,61 Justifications of study exclusions are reported in Appendix 6.
An abstract was available in English for a double-blind, placebo-controlled trial by Plesnik et al.,38 which suggested that amantadine at 100 mg/day reduced the incidence of serologically-confirmed infection and was well tolerated. However, as the full text was not available in English, this citation could not be included and is not presented in the review.
Oseltamivir
Nine studies (of which six were reported in full papers and a further three were abstracts) were identified that investigated the use of oseltamivir in prophylaxis against influenza in six original RCTs. Characteristics of these trials can be seen in Table 4.
Trial/data source | Population characteristics | Trial design arms (no. of patients in each arm) | Preventative strategy | Prophylaxis duration | Source of funding |
---|---|---|---|---|---|
WV15825 (Peters et al., 200164 and De Bock et al., 200065) | At-risk elderly subjects living in a residential home (mean age T1 = 81 years, T2 = 82 years), 98% with concomitant disease in each group | T1: oseltamivir 75 mg once daily, n = 276 | Seasonal | 6 weeks | Hoffman–La Roche |
T1: 80.4% vaccinated; T2: 80.1% vaccinated | T2: placebo, n = 272 | ||||
WV15673 (Hayden et al., 199966) | Healthy unvaccinated adults aged 18–65 years living in the community; conducted at study sites in Virginia, USA | T1: oseltamivir 75 mg once daily, n = 268 | Seasonal | 6 weeks | Hoffman–La Roche |
T2: placebo, n = 268 | |||||
WV15697 (Hayden et al., 199967) | Healthy unvaccinated adults aged 18–65 years living in the community; conducted at study sites in Texas and Kansas City, USA | T1: oseltamivir 75 mg once daily, n = 252 | Seasonal | 6 weeks | Hoffman–La Roche |
T2: placebo, n = 251 | |||||
WV15799 (Welliver et al., 200149) | Subjects of mixed age and health status living in households; adults and children aged 12 years and above (as contacts) | T1: oseltamivir 75 mg once daily, n = 493 | Post-exposure prophylaxis | 7 days | Hoffman–La Roche |
Contacts of all index cases: T1: 11.4% vaccinated; T2: 13.9% vaccinated | T2: placebo, n = 462 | ||||
Index cases did not receive treatment | |||||
WV16193 (Hayden et al., 200448,73,74) | Subjects of mixed age and health status; adults and children aged 1 year and above | Oseltamivir: post-exposure prophylaxis vs treatment on influenza onset (expectant treatment); index cases in both groups received treatment | Post-exposure prophylaxis | 10 days | Hoffman–La Roche |
Contacts: T1: 8% vaccinated; T2: 7% vaccinated | T1: oseltamivir prophylaxis 75 mg daily for 10 days, n = 400 | ||||
Index cases in both arms received treatment with oseltamivir 75 mg twice daily for 5 days | T2: oseltamivir treatment on influenza onset (expectant treatment) 75 mg twice daily for 5 days (less in children), n = 392 | ||||
Hayden et al., 200067 | Healthy adults aged 18–65 years | T1: oseltamivir 75 mg once daily, n = 19 | Experimentally-induced influenza | 7 days (1 day before challenge and 6 days after) | Hoffman–La Roche |
T2: placebo, n = 19 |
Four oseltamivir prevention trials were covered in the original HTA review;10 these were studies WV15825,64,65 WV15673,66 WV1569766 and WV15799. 49 Data for trials WV15673 and WV15697 were reported in the publication by Hayden et al. 66 both individually and combined across the two studies. All of these trials are included in the current assessment. An additional publication, by Hayden et al. ,47 examining the efficacy of oseltamivir in post-exposure prophylaxis within households present findings of an RCT published subsequent to the HTA review. 10 A further paper describes a trial of experimentally-induced influenza. 67 An additional publication45 describes a pooled analysis of data from oseltamivir post-exposure prophylaxis trials that are already included in the review. 46–49
An abstract in English was obtained for the trial by Kashiwagi et al. ,41 in which oseltamivir was administered to healthy adults at 75 mg once daily versus placebo for 6 weeks. This trial was previously reviewed by Jefferson et al. 68 However, the report was not available in full in English and was therefore excluded from this review.
Zanamivir
A total of 10 published reports of eight original RCTs were included in the assessment, of which eight were full papers and two were abstracts providing further reports of included studies. A further trial was identified within the sponsor submissions and is included, giving a total of nine RCTs. 44 These are presented in Table 5.
Trial | Population characteristics | Trial design arms (no. of patients in each arm) | Preventative strategy | Prophylaxis duration | Source of funding | Data source and additional information |
---|---|---|---|---|---|---|
NAIA3005 | Healthy adults aged 18 to 64 years from University communities | T1: zanamivir 10 mg once daily, n = 553 | Seasonal | 28 days | Glaxo Wellcome | Monto et al., 1999a70,71 |
T1: 14% vaccinated; T2: 14% vaccinated | T2: placebo, n = 554 | |||||
GlaxoSmithKline study 167/101 | Health-care workers aged 18 years and above | T1: zanamivir 10 mg once daily, n = 161 | Seasonal | 28 days | GlaxoSmithKline | Sponsor submission44 |
T2: placebo, n = 158 | ||||||
NAI30034 | At-risk adolescents and adults aged 12 years and above; high-risk defined as aged 65 years and above or chronic disorders of pulmonary or cardiovascular system or diabetes mellitus | T1: zanamivir 10 mg once daily: randomised, n = 1678; completed study, n = 1595 | Seasonal | 28 days | GlaxoSmithKline | LaForce et al., 200775 |
T1: 67% vaccinated; T2: 68% vaccinated | T2: placebo: randomised, n = 1685; completed study, n = 1594 | |||||
NAI30031 | Subjects of mixed age and health status; adults and children aged 5 years and above (as contacts) | T1: zanamivir 10 mg once daily, n = 661 | Post-exposure prophylaxis | 10 days | GlaxoSmithKline | Monto et al., 200247 |
Index cases: T1: 8% vaccinated; T2: 5% vaccinated | T2: placebo, n = 630 | |||||
Contacts: T1: 11% vaccinated; T2: 10% vaccinated | ||||||
Index cases did not receive treatment | ||||||
NAI30010 | Subjects of mixed age and health status; adults and children aged 5 years and above | T1: zanamivir inhaled 10 mg daily, n = 414 | Post-exposure prophylaxis | 10 days | Glaxo Wellcome | Hayden et al., 200046 |
Contacts: T1: 14% vaccinated; T2: 18% vaccinated | T2: placebo, n = 423 | |||||
Index cases were randomised to zanamivir twice daily or placebo | ||||||
NAIA2009/NAIB2009 | Subjects of mixed age and health status; unvaccinated adults and children aged 13–65 years (as contacts) | T1: zanamivir 10 mg inhaled daily, n = 144 | Post-exposure prophylaxis | 5 days | Presumed Glaxo Wellcome | Kaiser et al., 200072 |
Index cases did not receive treatment | T2: placebo, n = 144 | |||||
NAIA3004 | At-risk elderly subjects in long-term care (mean age T1 = 66.8 years, T2 = 67.2 years); 85% of subjects at randomisation at risk of complications or death due to influenza | T1: zanamivir once daily, n = 242 | Outbreak control | 14 days | Glaxo Wellcome | Ambrozaitis et al., 200576,77 |
T1: 9.6% vaccinated; T2: 8.8% vaccinated | T2: placebo, n = 252 | |||||
NAIA3003 | At-risk elderly subjects in long-term care (mean age T1 = 76.3 years, T2 = 74.8 years); 96% of subjects at randomisation at risk of complications or death due to influenza | T1: zanamivir 10 mg once daily, n = 12 for influenza B outbreak | Outbreak control | 14 days | Glaxo Wellcome | Gravenstein et al., 200578 |
T1: 99% vaccinated; T2: 92% vaccinated | T2: placebo n = 13, for influenza B outbreak |
Turner and colleagues10 evaluated five zanamivir prevention trials: studies NAIA2010,69 NAIA3005,70,71 NAIA30010,46 NAIA200972 and NAIB2009. 72 NAIA2009 and NAIB2009 were reported as a single trial in the published literature. All of these trials are included in the present assessment, with the exception of trial NAIA2010 reported by Schilling et al. ,69 in which the dose of zanamivir used was twice that of current licensed indications.
An additional six citations relating to zanamivir were identified by the systematic searches for inclusion in the clinical effectiveness review. Findings from a trial of zanamivir seasonal prophylaxis in at-risk adolescents and adults have been presented. 75 A report on the use of zanamivir in post-exposure prophylaxis within households has also been published,47 while two additional papers and one abstract provide reports of the use of zanamivir in outbreak control in at-risk elderly subjects within long-term care settings. 76–78 An additional paper describes a pooled analysis of data from zanamivir post-exposure prophylaxis trials that are already included in the review. 45
Quality of included research
The quality of the evidence included within the assessment was variable in terms of study design characteristics and clarity of reporting. Key study quality characteristics are summarised and presented in Appendices 3, 4 and 5.
Amantadine
The quality of the included eight RCTs relating to the prophylactic use of amantadine was relatively poor. No new amantadine prevention trials published since the original HTA assessment10 were identified. A considerable number of the older amantadine trials utilised a dose of 200 mg/day as opposed to the currently licensed adult dose of 100 mg/day18 and were therefore not considered to be suitable for inclusion in this review (see Inclusion and exclusion criteria, p. 17). Other amantadine prevention trials incorporated the use of doses in line with the current licence alongside inappropriate doses, but did not present data appropriate for inclusion separately and were therefore also excluded. Details of these studies can be found in Appendix 6.
Much of the amantadine prophylaxis evidence was not reported clearly, with a lack of detail on, for example, methods of randomisation of study subjects. 57,59–63 It was unclear in a number of trials whether allocation of treatment group was concealed. 55,57–59,62,63 One study publication failed to state clearly the number of participants randomised. 62 As only one report presented details of baseline characteristics of participants,55 it was generally not possible to assess whether baseline comparability between treatment groups had been achieved. It is therefore possible that potentially confounding variables may not have been adequately balanced among participants randomised to each trial arm. An additional four publications failed to state the eligibility criteria for participation in the trials. 55,60–62 A number of co-interventions were identified with the potential to affect outcomes, including vaccination,57–61 intake of medications that may affect study outcomes,64 and previous exposure to the experimental challenge strain. 62 The blinding of participants, those administering the intervention and outcome assessors was similarly difficult to judge and while many publications reported that a double-blind design was used, no further details were presented. Although all studies included at least 80% of randomised participants in the final analysis and only one study failed to report reasons for participants’ withdrawal,60,61 adherence to the intention-to-treat analysis was variable between studies.
Oseltamivir
The quality of the oseltamivir prophylaxis evidence presented was considerably more robust in terms of study design and reporting than that for amantadine. However, the randomisation methods used and concealment of allocation were unclear in the reporting of some studies. 48,49,73–74 All studies stated the number of participants randomised, and only one report failed to describe clearly the baseline characteristics and eligibility criteria,74 with all others judged to have achieved baseline comparability among subjects. A number of authors identified vaccination status,48,49,64,66 and recent use of antivirals48,64 or antibiotics48 as potentially confounding co-interventions. Clarity of reporting of blinding was variable among studies, and one study was described as being open-label in design. 48,73–78 All studies retained at least 80% of randomised subjects for use in the analysis and all, with the exception of two reports,64,67 presented reasons for withdrawal, but the analysis of only two studies could be considered to adhere to the intention-to-treat principle. 47,67,73-74
Zanamivir
The evidence base for the use of zanamivir in prophylaxis against influenza could also be considered to have a greater degree of internal validity than the trials of amantadine prophylaxis. However, there was a lack of detail on methods of randomisation46,47,72 and concealment of allocation. 46,72,76,77 All studies outlined the number of subjects who were randomised to each group and described baseline characteristics, with baseline comparability considered to have been reached to varying degrees in all trials. Baseline comparability was considered to be relatively weaker in one trial. 78 Vaccination status46,47,70,75,76,78 and recent use of antivirals46,76,78 were identified as co-interventions. More information was available on blinding procedures used in the zanamivir research (with additional information obtained from sponsor submissions) than for oseltamivir and amantadine prophylaxis trials, although there were some gaps in reporting in a number of studies. 46,47,70–72,75 However, all studies included more than 80% of randomised subjects in analyses, described reasons for withdrawal and utilised intention-to-treat analysis.
Assessment of effectiveness
Critical review and synthesis of information
The outcomes considered in the clinical effectiveness review of the interventions used in influenza prophylaxis included cases prevented, complications prevented, adverse effects of treatment, HRQoL, mortality, hospitalisations prevented, length of influenza illness, time to return to normal activities, cost and cost-effectiveness. Not all of these outcomes were represented in the identified clinical effectiveness trials included in the review; none of the included studies reported outcomes relating to HRQoL or mortality. The primary outcome reported in the majority of included trials related to cases of influenza prevented as measured in terms of the incidence of symptomatic, laboratory-confirmed influenza (SLCI). Where SLCI data were not presented – typically in older trials of relatively lower quality – cases prevented by prophylaxis within trials were described in terms of clinical influenza, acute respiratory disease and/or infection. 58,60–63 The efficacy of prophylaxis in preventing cases of SLCI was most frequently reported as a protective efficacy statistic (1 – RR, expressed as a percentage). While a minority of papers presented some SLCI data by influenza type, the numbers of observed cases were too small to allow meaningful estimates of efficacy to be made by influenza type and therefore the total numbers of cases of SLCI are presented. These values are tabulated where appropriate within the data synthesis. In a small number of trials, this evidence was categorised by subgroup, in terms of age, risk (according to age and health status) and vaccination status, and is presented where available. The majority of trials also presented information on the occurrence of adverse events among participants, which is presented in text format, due to the large degree of variability in adverse events reported. A limited amount of information was reported relating to complications prevented, hospitalisations prevented, length of influenza illness and time to return to normal activities.
Amantadine
The included evidence focusing on amantadine prophylaxis against influenza was taken from relatively old trials of lower quality that were conducted across a broad range of population subgroups. There was considerable variability between trials in terms of vaccination levels, setting and duration of prophylaxis. Eight references reporting eight RCTs were identified. The Cochrane review investigating amantadine and rimantadine in influenza A incorporated the use of meta-analysis in their study. 33 However, the large degree of heterogeneity and variation in primary outcomes used in terms of cases prevented between the studies included in our review would suggest that the use of statistical meta-analysis would be inappropriate; as such, the results of these trials are presented in the form of a narrative synthesis.
Evidence for amantadine prophylaxis in children under 10 years is not presented in this systematic review; such data were excluded as amantadine dosage is not established in this age group according to licensed indications. The limited evidence that exists relating specifically to this younger age group was reported within the original HTA review. 10 No clinical trial evidence relating to the use of amantadine in the paediatric population has been published subsequently.
Two trials by Reuman et al. 57 and Aoki et al. 58 examining the use of amantadine in seasonal prophylaxis in healthy adults were identified and included in the systematic review of clinical effectiveness.
The RCT conducted by Reuman et al. 57 was undertaken in a healthy, unvaccinated adult population aged 18–55 years. Although this study also investigated the effects of amantadine at daily doses of 50 mg and 200 mg, only data relating to the use of the drug at the licensed dose of 100 mg per day are presented here. The reporting of the duration of the intervention is unclear within the reporting of this trial; it is assumed from the description of the trial methods to be over a period of 6 weeks. Subjects were excluded if chronic disease and abnormal clinical history and physical examination were evident prior to study entry. Clinical symptoms with influenza A infection were observed in 5 of 159 subjects in the placebo group (3.1%) and 2 of 159 subjects (1.3%) in the amantadine at 100 mg/day dosage group (RR 0.40,† 95% CI 0.08†–2.03†). The authors described a higher rate of adverse events in the treatment group receiving the higher dose of 200 mg/day but no differences between the arms receiving the licensed dose of 100 mg/day and placebo. Total adverse events were reported at a rate of 49/159 (31%) versus 47/159 (30%) in the placebo and amantadine arms respectively. Gastrointestinal adverse events occurred in 8% of subjects in each arm (12/159 for each arm). CNS-related adverse events were observed in 14% of amantadine-treated subjects (23/159) and 16% (25/159) of subjects in the placebo arm. One subject of the 159 in the placebo arm (0.6%) withdrew as a result of adverse events; no withdrawals were described in the amantadine 100 mg/day group. However, adherence to amantadine was relatively poor, with 49% of the amantadine-dosed participants and 58% of the placebo arm taking fewer than the total allotted tablets. This study suggests that the use of amantadine at the lower dose results in fewer adverse effects but that the low influenza attack rate does not allow meaningful conclusions to be drawn in relation to the efficacy of amantadine in preventing influenza illness and infection.
A study in which amantadine was administered to healthy military personnel for seasonal prophylaxis over two seasons for 32 days and 39 days respectively was reported by Aoki et al. 58 As discussed in Inclusion and exclusion criteria (p. 17), only data comparing effects in treatment arms receiving amantadine at a dose of 100 mg per day or placebo are presented in this review. Reasons for the unequal numbers in each treatment arm are unclear. Six to eight individuals per study season were described as being vaccinated in previous years (proportions not estimable). Primary outcomes that were reported related to the proportion of participants who developed acute respiratory tract infection, classification of disease and adverse effects. No differences in the incidence or classification of acute respiratory illness (ARI) were observed between the treatment arms. The trial findings were not reported clearly, in that one subject in the 1980–1 season and two subjects in the 1982–3 season are stated as developing acute influenza A, but no further detail was presented concerning the treatment arm in which these cases developed. However, the observed attack rates were so low that meaningful comparison of efficacy between arms is limited. In the 1980–1 season, withdrawals due to adverse effects were reported at a frequency of 1/49 (2.0%) in the placebo group and 1/75 (1.3%) in the amantadine 100 mg/day group. In 1982–3, these rates were described as 1/34 (2.9%) in the placebo group and 1/47 (2.1%) in the amantadine 100 mg/day group. No amantadine-related differences in adverse effects were observed between the placebo and amantadine 100 mg/day groups (no further data were presented).
A single trial by Pettersson et al. 55 in which amantadine was used for seasonal prophylaxis in elderly subjects was included in the systematic review. While the trial also investigated amantadine prophylaxis in different population groups and settings, the only data for amantadine administered in line with licensed indications and therefore suitable for inclusion related to residents of a home for the elderly who received amantadine at a dose of 100 mg/day versus placebo over a period of 9 weeks. The vaccination status of subjects was not clearly described in the trial publication, although it was stated in the discussion of the report that no adequate vaccine was available at the time of study; this suggests that the population could be considered to be unprotected by vaccination. Primary outcomes were reported in terms of the incidence of serologically-confirmed influenza infection, incidence of respiratory infections and adverse events. No data were reported for the incidence of serologically-confirmed influenza infection or incidence of respiratory infections in the elderly study population, as there was no evidence of an influenza epidemic in this group. Amantadine prophylaxis was described as being terminated in 5 of 94 (5.3%) and 2 of 101 (2.0%) subjects in the amantadine and placebo arms respectively. Although this evidence would suggest a potentially higher incidence of adverse events in the amantadine arm, a range of apparently non-drug-related reasons were cited for termination, including one fracture of caput femoris, two deaths attributable to carcinoma and myocardial infarction, no reason given (in one case) and compliance and practical issues (in a further two cases). One case of GI symptoms and one of chest pains were cited in the placebo arm.
No studies investigating the use of amantadine in the prevention of influenza in household contacts of influenza-infected index cases were identified for inclusion in the systematic review.
Two trials were identified in which amantadine was used for the control of influenza outbreaks. The trial reported by Payler and Purdham59 was undertaken in adolescent males in a boarding school, of whom 87% (525/606) were vaccinated for that season. Subjects were randomised to receive either amantadine 100 mg/day for 14 days or no treatment. In this study, the control arm was not placebo controlled. However, it is unlikely that a lack of blinding would have an impact on the reported incidence of SLCI, due to the nature of the manifested infectious illness and requirement of infection confirmed by laboratory tests. The incidence of clinical influenza was reported as being 7/267 (2.6%) in the amantadine arm versus 42/269 (15.6%) in the control group (p < 0.001, RR 0.17,† 95% CI 0.08†–0.37†). The incidence of clinical influenza that was laboratory confirmed was 3/267 (1.1%) in the subjects receiving amantadine compared with 29/269 (10.8%) in the control group (p < 0.001), resulting in a protective efficacy of 90% (95% CI 0.66†–0.97†). Of the three subjects developing symptomatic, laboratory-confirmed influenza in the amantadine arm, two were described as being vaccinated, while one subject was reported as unvaccinated. No information was given for the control arm. Urticaria was reported in 1 of 267 participants receiving amantadine (0.4%), while no adverse events were observed in the control group. The authors observed that eight of the nine subjects who developed laboratory-confirmed influenza A 3 days after the 14-day prophylactic period had ceased had received amantadine, highlighting that protection against influenza is not extended beyond the prophylactic period.
The second included RCT of amantadine in outbreak control was presented by Smorodintsev et al. 60,61 The composition of the study population was not clearly reported but appears to have consisted of healthy, unvaccinated adults based in semi-isolated engineering schools. Subjects in five of seven schools were dosed for 30 days, while subjects in two schools were dosed for 12 days. The reporting of the study was very unclear, with conflicting descriptions of the vaccination status of populations, varying from unvaccinated to partially vaccinated. Regardless of whether subjects received drug medication regularly or irregularly, clinical influenza occurred at rates of 214/4559 (4.7%) and 224/2804 (8.0%) in the amantadine and placebo groups respectively (RR 0.59,† 95% CI 0.49†–0.70†). Of 400 influenza cases that were selected at random, severity of symptoms in the amantadine group was reported as 56.0% mild and 9.0% severe; while symptoms were described as 38.0% mild and 19.0% severe in the placebo group (p < 0.01 for severe symptoms, p < 0.001 for mild symptoms), demonstrating milder disease in the amantadine-treated group. No further information was provided on the criteria for classing symptoms as mild or severe. Mean duration of overall illness was shorter in the amantadine group than in the placebo group (p < 0.05). A subset of non-ill subjects (n = 1825) were questioned about adverse effects, which occurred in 7.2% (94/1313) and 5.1% (26/512) of those questioned from the amantadine and placebo groups respectively, showing a non-significant 2.1% excess in the amantadine group. Statistically-significant (at 5%) excesses in dyspepsia (1.72%) and sleep disturbances (1.14%) were noted in the amantadine-dosed subjects. The applicability of this evidence is considerably hindered by poor reporting and lack of detail on population baseline characteristics. However, some limited evidence of the efficacy of amantadine in preventing and shortening the duration and severity of clinical influenza disease, and of a higher rate of adverse effects resulting from amantadine prophylaxis, were presented.
No studies investigating the use of amantadine in outbreak control in elderly populations were identified.
Three further trials of amantadine prophylaxis, in which subjects were challenged experimentally with influenza virus, were included in the systematic review. 57,62,63
Reuman et al. 57 undertook an RCT to determine the efficacy of amantadine in preventing experimentally-induced influenza A. Although the use of doses of amantadine at 50 mg/day and 200 mg/day were also investigated, only data relating to the use of amantadine at 100 mg/day and placebo are presented within this systematic review. Subjects were healthy, unvaccinated adults aged 18–40 years. Individuals who had a pre-study abnormal clinical history and physical examination or chronic disease were excluded from participation. Infection was noted in 18/19 (95%) placebo subjects and in 12/20 (60%) of amantadine-dosed subjects (p = 0.012). Symptomatic, laboratory-confirmed influenza was observed in a smaller proportion of subjects, i.e. 11/19 (58%) in the placebo arm and 3/20 (15%) in the amantadine arm (p = 0.0055), resulting in an RR of 0.26 (95% CI 0.09†–0.79†) and a protective efficacy of 74%. Amantadine at all doses was described as suppressing respiratory symptoms on days 2–6 following viral challenge and systematic symptoms on days 2 and 3 post challenge. Total length of illness was not reported. Total adverse events judged to be potentially drug related occurred in 50% of placebo subjects and 80% of subjects receiving amantadine at 100 mg/day (p = 0.27). These were stated as being mostly mild and transient and related to the GI and CNS systems. Three adverse events were rated as severe, comprising two cases of severe headache, of which one occurred in each treatment arm, and one case of dream abnormality in a subject receiving amantadine. No withdrawals were made in the placebo or amantadine at 100 mg/day arms.
Further evidence of the use of amantadine prophylaxis against experimentally-induced influenza A was published by Sears and Clements. 63 Healthy, unvaccinated adult subjects aged 18–40 years were randomised to receive either amantadine at 100 mg/day or placebo over a period of 8 days. Infection was serologically confirmed in 17/22 (77%) amantadine subjects and 20/22 (91%) subjects in the placebo group. Influenza illness was observed in 2/22 (9.1%) subjects receiving amantadine and 9/22 (40.9%) subjects receiving placebo, yielding a protective efficacy of 78% (p < 0.04) and an RR of 0.22† (95% CI 0.05†–0.91†). Severity of illness was also lower in the amantadine-dosed group. The authors stated that no adverse events were reported in the group who received amantadine.
Smorodintsev et al. 62 demonstrated the efficacy of amantadine at the lower dose of 100 mg/day versus the previously-used dose of 200 mg/day and placebo in the prevention of experimentally-induced influenza A in healthy medical student volunteers. Only data in which the licensed dose of 100 mg/day and placebo are compared are presented here. A protective efficacy of 42% against clinical influenza was reported for amantadine at 100 mg/day versus placebo (10/19 in the amantadine arm, 28/31 in the placebo arm; RR 0.58, 95% CI 0.37†–0.91†). This increased to a protective efficacy of 86% against serologically-confirmed influenza (1/19 in the amantadine arm, 12/31 in the placebo arm; RR 0.14,† 95% CI 0.02†–0.96†). No data were reported on adverse effects relating to a comparison of amantadine at the licensed dose with placebo; however, no drug-related side effects were reported overall.
Four trials presented evidence of varying levels of adherence to the study protocols. Payler and Purdham59 stated that only 2% of their subjects did not take amantadine, while 85% (number of subjects not reported) of participants in an additional trial of outbreak control were reported as taking amantadine without interruption over the study period, suggesting a relatively high level of adherence. 60 However, Reuman et al. 57 reported that approximately half of their study participants did not take all of the allotted study treatment (49% and 58% of amantadine and placebo groups respectively). The study by Aoki et al. 58 utilised laboratory testing of samples taken from tested study participants and demonstrated that 10% and 22% of subjects who had been randomised to receipt of amantadine and were tested in different study seasons showed no drug in samples. No amantadine was present in samples from placebo subjects.
No trials presented data on analysis of sensitivity of viral isolates to amantadine (see below).
As noted in the review by Jefferson et al. ,79 the evidence base relating to amantadine in prophylaxis against influenza was comparatively old and relatively poor in terms of study quality and reporting. The resulting data should therefore be interpreted with caution.
Owing to low attack rates, evidence of efficacy against SLCI in seasonal prophylaxis was limited. One study of amantadine used in outbreak control59 suggested high efficacy against SLCI in a boarding-school setting and demonstrated that protection against influenza is not conferred beyond the prophylactic period. Limited evidence for a lower incidence of clinical influenza and milder disease of shorter duration was presented. 60,61 Some evidence relating to the efficacy of amantadine in preventing experimentally-induced infection and SLCI was also identified. As such challenge studies are undertaken under experimental rather than clinical conditions, data drawn from these studies should be interpreted with caution with respect to external validity and applicability to clinical effectiveness, particularly with respect to the nature of challenge and the comparability of subjects in terms of pre-challenge antibody titres. However, as the evidence concerning amantadine prophylaxis against naturally-acquired influenza is sparse, it was considered useful to present the findings of the use of the drug in accordance with licensed indications against the development of experimentally-induced influenza in healthy adults, in order to supplement the evidence base presented here. Very limited interpretation can be made concerning the impact of vaccination status on the efficacy of amantadine prophylaxis, although the study reported by Payler and Purdham59 demonstrated that a small number of cases of SLCI developed in both vaccinated and unvaccinated subjects in the amantadine-treated arm.
Withdrawals due to adverse events and illness were similar in the amantadine and placebo groups, and adverse effects were similar in both groups, with the exception of the trial reported by Smorodintsev et al. 60,61 and the experimental challenge study by Reuman et al.,57 both of which demonstrated a higher incidence of adverse effects in the amantadine-treated subjects. Severe adverse effects also appeared to be higher in the amantadine-treated group. 57
None of the amantadine prophylaxis trials included in this review reported the assessment of sensitivity of influenza isolates to amantadine. However, as noted in Chapter 1, reports of the increasing emergence of amantadine-resistant influenza A strains31 present a significant challenge to the clinical effectiveness of amantadine in prophylaxis against influenza, and must be taken into account during the interpretation of the evidence presented in the clinical effectiveness review.
Oseltamivir
Nine references reporting six original RCTs of oseltamivir for the prophylaxis of influenza were identified.
No evidence that specifically relates to seasonal prophylaxis in children was identified.
Two RCTs investigating oseltamivir for seasonal prophylaxis were reported by Hayden et al. 66 (Table 6). The two trials were identically-designed multicentre studies undertaken in healthy, unvaccinated adults aged 18–65 years; the first trial was undertaken in Virginia (WV15673) and the second at sites in Texas and Kansas City (WV15697). Prophylaxis was administered for 6 weeks. Oseltamivir administered to subjects at a dose of 75 mg once daily conferred a protective efficacy against SLCI of 84% (95% CI 53–96) in trial WV15673 and a non-significant protective efficacy of 50% (95% CI –55 to 94) in trial WV15697. The authors reported a pooled estimate for protective efficacy against SLCI of 76% (95% CI 46–91; RR 0.24). When a meta-analysis of the data reported separately for each trial was undertaken by the Assessment Group, the RR of developing influenza for oseltamivir versus placebo was 0.27 (95% CI 0.09–0.83). Total withdrawals occurred in 21/519 (4%) of the placebo and 17/520 (3%) of the oseltamivir subjects. Withdrawals due to adverse effects or intercurrent illness occurred in 8/520 (1.5%) of the oseltamivir 75 mg/day group and in 10/519 (1.9%) of the placebo group. Upper GI adverse effects were greater in subjects receiving oseltamivir 75 mg/day (12.1%) than in those receiving placebo (7.1%) (difference 5.0%, 95% CI 1.4–8.6). Vomiting occurred in a higher proportion of subjects receiving the oseltamivir dose (2.5%) than in those receiving placebo (0.8%) (difference 1.7%, 95% CI 0.2–3.3).
Outcome | Trial | Total no. in placebo group | No. in placebo group with an event | Total no. in oseltamivir group | No. in oseltamivir group with an event | RR (95% CI) |
---|---|---|---|---|---|---|
SLCI | WV15673 | 268 | 19 | 268 | 3 | 0.16 (0.04–0.47) |
SLCI | WV15697 | 251 | 6 | 252 | 3 | 0.50 (0.06–1.55) |
Pooled (random effects) | 0.27 (0.09– 0.83) | |||||
(p = 0.21, I2 = 35.4%) |
Peters et al. 64 and De Bock et al. 65 presented the results from study WV15825, an RCT of oseltamivir in seasonal prophylaxis in a frail, elderly population residing within a residential care setting (Table 7). Prophylaxis with oseltamivir at 75 mg once daily for 6 weeks resulted in a 92% protective efficacy for SLCI (p = 0.002). When incidence in the vaccinated population only was analysed, a protective efficacy of 91% against SLCI (p = 0.003) was observed. For all individuals, receipt of oseltamivir resulted in an 86% relative reduction in secondary influenza complications [where complications included bronchitis (4/272), pneumonia (3/272) and sinusitis (1/272) in the placebo arm and bronchitis (1/276) in the oseltamivir group] (p = 0.037). In subjects with laboratory-confirmed influenza, the relative reduction in secondary complications was 78% (p = 1.14). Withdrawals due to adverse events or illness occurred at rates of 6.5% (18/276) and 4.0% (11/272) in the oseltamivir and placebo arms respectively. A similar proportion of subjects in each group experienced mild to moderate adverse events (around 60%); however, most of these were not considered by the study investigators to be drug related. Headaches occurred at a higher frequency in the oseltamivir group than in the placebo group (8.3% versus 5.5%) and GI adverse events were also more common among individuals receiving oseltamivir (14.9% versus 12.9%).
Outcome | Trial | Total no. in placebo group | No. in placebo group with an event | Total no. in oseltamivir group | No. in oseltamivir group with an event | RR (95% CI) |
---|---|---|---|---|---|---|
SLCI | WV15825 | 272 | 12 | 276 | 1 | 0.08 (0.01†–0.63†) |
SLCI (vaccinated subjects only) | WV15825 | 218 | 11 | 222 | 1 | 0.09 (0.01†–0.69†) |
Two RCTs, WV15799 reported by Welliver et al. 49 and WV16193 reported by Hayden et al. ,48,73 and Belshe et al.,74 investigating oseltamivir in the prevention of influenza in household contacts of index cases were identified (Table 8).
Outcome | Trial | Total no. in placebo group | No. in control group with an event | Total no. in oseltamivir group | No. in oseltamivir prophylaxis group with an event | RR (95% CI) |
---|---|---|---|---|---|---|
SLCI in contacts of all index cases | WV15799 | 462 | 34 | 493 | 4 | 0.11 (0.04–0.29) |
WV16193 | 392 | 40 | 400 | 11 | 0.27 (0.14–0.53) | |
Pooled (random effects) |
0.19 (0.08–0.45) (p = 0.15, I2 = 52.9%) |
|||||
SLCI in contacts of influenza-positive index cases | WV15799 | 206 | 26 | 209 | 3 | 0.11 (0.03, 0.33) |
WV16193 | 258 | 33 | 244 | 10 | 0.32 (0.16 to 0.65) | |
Pooled (random effects) |
0.21 (0.08–0.58) (p = 0.13, I2 = 56.3%) |
Welliver et al. 49 randomised household contacts of index cases to receive either 75 mg oseltamivir once daily or placebo for 7 days. Index cases did not receive antiviral treatment in either trial arm. Children under 12 years of age were excluded as contacts, but were eligible as index cases. A minor point is that subjects aged 12 years and above received the adult dose of 75 mg once daily, while dosing according to body weight is recommended in subjects aged less than 13 years. Although individuals with well-controlled co-morbidities were eligible for participation in the study, potential subjects with cancer, immunosuppression or chronic renal or liver disease were excluded. Prophylaxis resulted in a protective efficacy among individual contacts of all index cases of 89% (95% CI 71–96, p < 0.001). For individual contacts of influenza-positive index cases only, the protective efficacy was also 89% (95% CI 67–97, p < 0.001). Withdrawals due to adverse effects or illness occurred in 2/461 (0.4%) in the placebo arm and 5/494 (1.0%) oseltamivir subjects. Gastrointestinal adverse effects were reported in 7.2% of the placebo and 9.3% of the oseltamivir subjects, while nausea was evident in 2.6% and 5.5% of the placebo and oseltamivir subjects respectively. No abnormal results for safety or vital signs and no serious adverse events were observed.
A randomised, open-label trial (WV16193) in adults and children aged 1 year and above undertaken by Hayden et al. 48,73 and Belshe et al. 74 investigated the use of oseltamivir (75 mg once daily) in post-exposure prophylaxis in household contacts of index cases for 10 days versus expectant treatment, in which oseltamivir (75 mg twice daily) was administered for 5 days at the onset of influenza illness in contacts. In both trial arms, index cases received treatment. Post-exposure prophylaxis with oseltamivir for 10 days in individual household contacts resulted in a protective efficacy against SLCI of 73% (95% CI 47–86), including all households irrespective of whether the index case developed influenza. For individual contacts of influenza-positive index cases, the corresponding protective efficacy was lower, at 68% (95% CI 35–84). The proportion of contacts with laboratory-confirmed influenza with at least one secondary complication was broadly comparable between the post-exposure prophylaxis group and subjects receiving expectant treatment [7% (3/46) versus 5% (4/75)]; however, the more severe respiratory complications occurred in the expectant treatment arm only. The median time from start of treatment to alleviation of symptoms in contacts was also shorter in the post-exposure prophylaxis arm (n = 10) than in the expectant treatment arm (n = 33) [5.5 hours (0–87) versus 39.8 hours (0–627) (p = 0.103)]. Fewer contacts with laboratory-confirmed influenza in the post-exposure prophylaxis arm were bed bound compared with subjects in the expectant treatment group [7% (3/46) versus 28% (21/75)], demonstrating a milder form of disease. Withdrawals due to adverse events occurred at a rate of 1/410 (0.2%) in the post-exposure prophylaxis arm and 4/402 (1.0%) in the expectant treatment arm. Nausea was more common in subjects receiving oseltamivir for post-exposure once daily than treatment twice daily (8% versus 7%). However, vomiting was more frequent in the expectant treatment arm (10% versus 4.5%).
When the data for SLCI in the mixed adults and children populations from the Welliver et al. 49 and Hayden et al. 48 trials were pooled by meta-analysis using random effects, the resulting RR among household contacts of all index cases was 0.19 (95% CI 0.08–0.45), equating to a protective efficacy of 81%. For contacts of influenza-infected index cases only, the corresponding pooled RR was 0.21 (95% CI 0.08–0.58) and the resulting protective efficacy was 79%. A pooled RR for withdrawals generated by the Assessment Group yielded an RR of 0.85 (95% CI 0.09–7.72), favouring treatment.
An additional pooled analysis of data from the trials by Welliver et al. 49 and Hayden et al. 48 was reported by Halloran et al. ,45 who presented a pooled estimate of protective efficacy of oseltamivir post-exposure prophylaxis against illness of 81% (95% CI 35–94) and an 80% reduction in infectiousness (95% CI 48–72). The secondary analysis by Halloran et al. also assessed pathogenicity of influenza in the treatment and control arms of the household post-exposure prophylaxis trials. Pathogenicity was defined as the ability of the virus to cause disease in an infected person. It was calculated as the number of contacts with SLCI divided by the number of contacts with laboratory-confirmed influenza infections (symptomatic or asymptomatic). Pathogenicity was lower among subjects treated with oseltamivir than among control subjects. In the study by Welliver et al. ,49 reported pathogenicity in the control group was 34/60 (57%) and in the oseltamivir group it was 4/33 (12%); these data included contacts, regardless of whether the index case was influenza positive. In the study by Hayden et al. ,48 pathogenicity in the control group was 33/75 (44%) and in the oseltamivir group it was 10/46 (22%); note that for this study, data for contacts with an influenza-positive index case only were available for this analysis.
In the trials reported by Hayden et al. 48 and Welliver et al. 49 it was noted that, in some instances, the strain of influenza with which the contact cases were infected did not match that of the index case, thus indicating that illness was transmitted not from the index case but from a source external to the household setting.
Clinical outcomes from the trial by Hayden et al. 48 were also reported separately for paediatric household contacts aged 1–12 years (Table 9). It should be noted that this study allocated doses according to the child’s age banding, rather than body weight, as recommended by the BNF. 18 However, subsequent analysis has shown that the dosages used were broadly equivalent to those approved. 80 For individual contacts of all index cases, the protective efficacy against SLCI was 64% (RR 0.36, 95% CI 16–85). When contacts of influenza-infected index cases only were included in the analysis, the protective efficacy dropped to 55% (RR 0.45, 95% CI –13 to 82). Vomiting was more common in the expectant treatment group (20% versus 10%). No children withdrew as a result of adverse events.
Outcome | Trial | Total no. in placebo group | No. in control group with an event | Total no. in oseltamivir group | No. in oseltamivir prophylaxis group with an event | RR (95% CI) |
---|---|---|---|---|---|---|
SLCI in contacts of all index cases | WV16193 | 111 | 21 | 104 | 7 | 0.36 (0.15–0.84) |
SLCI in contacts of influenza-positive index cases | WV16193 | 74 | 18 | 55 | 6 | 0.45 (0.18–1.13) |
No studies describing the use of oseltamivir for control of influenza outbreaks were identified.
A single trial by Hayden et al. 67 of oseltamivir used in accordance with licensed indications in prophylaxis against experimentally-induced influenza B in healthy adults was identified. Influenza B infection was observed at rates of 17/19 (89%) in the oseltamivir group and 16/19 (84%) in the placebo group (RR 1.06,† 95% CI 0.83†–1.36†). Symptoms of upper respiratory tract illness were present in 2/19 (11%) oseltamivir subjects compared with 4/19 (21%) in the placebo arm (RR 0.50,† 95% CI 0.10†–2.41†), while fever was observed in 1/19 (5%) and 2/20 (10%) in the oseltamivir and placebo groups respectively (RR 0.53,† 95% CI 0.05†–5.34†). No serious adverse effects were reported. Adverse effects related to study treatment occurred in 1/19 (5.3%) subjects in each group. No treatment-related adverse effects were observed during the off-treatment follow-up period.
Adherence to the study regimens was reasonably high. In one study, 7% percent of placebo subjects and 11% of those in the oseltamivir arm were reported as taking less than 80% of study medication. 64 In another study, 53% of subjects in both oseltamivir and placebo arms took 100% of the prescribed doses, according to returned capsules. 66 In the study by Welliver et al. ,49 fewer than 1% of contacts in both placebo and oseltamivir arms did not take the allocated treatment.
A number of trials tested viral isolates for resistance to oseltamivir in vitro and found no evidence of reduced sensitivity (see below). 48,49,66,67
The trials included in this systematic review suggest that oseltamivir has a relatively high protective efficacy against SLCI in healthy adults. The protective efficacy against SLCI was notably high among the frail elderly living in residential care, among whom a clear reduction in influenza-associated complications was also observed. The efficacy against SLCI was broadly equivalent in vaccinated and unvaccinated individuals. The evidence for oseltamivir in post-exposure prophylaxis in the household setting has been reinforced by the publication of an additional trial48 since the original assessment. 10 Oseltamivir conveys a high protective efficacy against SLCI in household contacts and any resulting disease appears to be milder and of shorter duration. 48 As in the Cochrane review by Matheson et al. 81 of neuraminidase inhibitors in the prevention of influenza in children, only one RCT trial, WV16193,48 in which data relating specifically to children were presented, was identified. Prophylaxis in paediatric contacts was demonstrated to be reasonably effective. An experimental challenge study also demonstrated a lower incidence of illness in subjects receiving prophylaxis. 67
Withdrawals due to adverse events and illness were similar in both groups in all trials, bar one,64 which demonstrated a slightly higher incidence in frail, elderly subjects receiving oseltamivir. Two studies suggested that GI adverse effects were marginally higher among the oseltamivir-treated subjects. 49,66
No evidence of reduced sensitivity of viral isolates to oseltamivir was obtained. A number of publications have postulated that levels of resistance to neuraminidase inhibitors have been low. 82–84 However, additional reports from Japan85 and Europe86 (including the UK) have demonstrated the emergence of oseltamivir-resistant strains of influenza A. Recent surveillance data87 from within the UK have indicated that approximately 5% of influenza A (H1N1) isolates were oseltamivir resistant, but the HPA drew no conclusions with regard to the clinical significance of this finding, stating a requirement for the completion of further research before a judgement could be made. The clinical effectiveness evidence for the use of oseltamivir in prophylaxis against influenza should therefore be interpreted in light of the above reports of emerging resistance.
Zanamivir
Ten published articles presenting the results from eight RCTs were identified for inclusion in the systematic review of clinical effectiveness. An additional unpublished report was identified in the sponsor submissions and included in the assessment, resulting in a total of nine RCTs. The use of inhaled zanamivir only is considered within this assessment, hence trial arms in which doses of intranasal zanamivir were administered were excluded.
No data relating specifically to seasonal prophylaxis in children were identified.
Study NAIA3005 reported by Monto et al. 70,71 evaluated the use of zanamivir in seasonal prophylaxis in healthy adults (Table 10) aged 18–64 years and demonstrated a 68%† protective efficacy against SLCI (95% CI 37†–83†). When the unvaccinated subjects were analysed as a subgroup, the protective efficacy was 60% (95% CI 24–80). Potential symptoms relating to drug use were reported by 75% of subjects in both arms. Adverse effects considered by the authors to be potentially drug related were observed in 5% (27/554) of the placebo and 5% (30/553) of the zanamivir group, of which less than 1% in each arm was classed as severe. Total withdrawals occurred in 3% (17/554) and 2% (10/553) of the placebo and zanamivir arms respectively. Potentially drug-related withdrawals were made in 1.3% of the placebo and 0.7% of the zanamivir groups. A conference abstract71 presenting further information on the trial stated that significantly less time was lost from work in the zanamivir group (mean hours lost 1.4 hours versus 0.6 hours, p = 0.001). Total productive time lost was also less in the zanamivir group (1.8 hours versus 3.0 hours, p = 0.001). The authors stated that the trial was undertaken during a season in which the predominant circulating influenza A strain did not match the administered vaccine, demonstrating efficacy of prophylaxis during a circumstance of strain mismatch.
Outcome | Trial | Total no. in placebo group | No. in placebo group with an event | Total no. in zanamivir group | No. in zanamivir group with an event | RR (95% CI, p-value if available) |
---|---|---|---|---|---|---|
SLCI | NAIA3005 | 554 | 34 | 553 | 11 | 0.32 (0.17†–0.63†) |
SLCI in unvaccinated subjects only | NAIA3005 | No data | No data | No data | No data | 0.40 (0.20–0.76, p = 0.004) |
SLCI in unvaccinated subjects only | GSK study 167/101 | 156 | 6 | 160 | 3 | 0.49 (0.12†–1.92,† p = 0.3314) |
An unpublished report of a randomised, double-blind, placebo-controlled trial, presented as part of the sponsor submissions, described the use of zanamivir in seasonal prophylaxis in adult health-care workers (who were presumed to be healthy in the current assessment). 44 No statistical significance between treatment groups in the development of SLCI was observed (3/160 versus 6/156 in the zanamivir and placebo arms respectively in the non-vaccinated set, p = 0.3314). Adverse events occurred at similar rates in the zanamivir (67.7%) and placebo (62.2%) arms, of which 1.2% in the zanamivir subjects and 1.3% in the placebo subjects were considered to be drug related. One serious adverse event, which was not judged to be drug related, occurred in a zanamivir-treated subject.
Since the original HTA assessment was undertaken,10 a large-scale study of zanamivir seasonal prophylaxis in community-dwelling adolescents and adults aged 12 years and above at risk of complications of influenza has been published75 (Table 11). High risk was defined as being aged 65 years and above or having chronic pulmonary or cardiovascular disease or diabetes mellitus. For the intention-to-treat (ITT) population assessed for the development of SLCI during days 1–28 of prophylaxis, a protective efficacy of 83% was observed (RR 0.17, 95% CI 0.07–0.44, p < 0.001). For the per-protocol population, this value dropped to 75% (RR 0.25, 95% CI 0.09–0.70, p = 0.014). Protective efficacies against the development of SLCI during days 2–28 and 3–28 of the prophylactic period were 81% and 80% respectively. Data were also presented by high-risk condition, with RR values calculable for a number of subgroups: subjects with respiratory disease (RR 0.18, 95% CI 0.05†–0.61†), subjects with cardiovascular disease (no events in the zanamivir group) and subjects with diabetes (no events in the zanamivir group). When presented according to age, the incidence of SLCI was lower in the zanamivir group than the placebo group in subjects aged both below and above 50 years (50 years and above: zanamivir: 1/1276 (0.08%), placebo: 9/1270 (0.71%); below 50 years: zanamivir: 3/402 (0.75%), placebo: 14/415 (3%). 44 Relative risks were also calculable by vaccination status, with RRs of 0.17 (95% CI 0.02†–1.41†) and 0.17 (95% CI 0.05†–0.58†) of developing SLCI in vaccinated and unvaccinated subjects respectively. Confirmed influenza with complications was observed in 0.06% of zanamivir subjects and in 0.48% of those in the placebo arm, giving an RR of 0.12 (95% CI 0.02–0.73). Zanamivir was well tolerated, with no significant differences in total adverse effects between the two groups, with 51% in each group experiencing adverse effects (placebo: 851/1685, zanamivir: 850/1678). Potentially drug-related adverse events were observed in 9% and 10% of the placebo and zanamivir arms respectively. Drug-related serious adverse events occurred in 2/1685 of placebo subjects (0.12%, cardiac arrhythmia and dyspnoea/cough) and 1/1678 of zanamivir subjects(0.06%, acute resistant asthmatic bronchitis/acute rhinositis). In subjects with respiratory disease any adverse event was observed in 59% of each group (405/684 and 412/695 in the zanamivir and placebo arms respectively). 44 Subjects with cardiovascular disease for whom any adverse event was reported comprised 48% (159/331) and 49% (149/307) of the zanamivir and placebo arms respectively. 44 In diabetic subjects any adverse event was observed in 62% of the zanamivir group (223/359) and 52% of the placebo group (191/370). 44 There were 39 hospitalisations in the ITT population after the study commenced: 19 in the placebo group and 20 in the zanamivir group. 44 The mean length of stay across those subjects hospitalised was 3.8 days in placebo-treated subjects and 3.3 days in zanamivir-treated subjects,44 mean values 0.4 days and 0.3 days in the placebo and zanamivir groups respectively demonstrating no significant differences between arms. 44 Median time to alleviation of symptoms was shorter in the zanamivir group than in the placebo group (2.5 days versus 4.0 days).
Outcome | Trial | Total no. in placebo group | No. in placebo group with an event | Total no. in zanamivir group | No. in zanamivir group with an event | RR (95% CI, p-value if available) |
---|---|---|---|---|---|---|
SLCI in all cases | NAI30034 | 1685 | 23 | 1678 | 4 | 0.17 (0.07–0.44) |
SLCI in vaccinated subjects | NAI30034 | 1141 | 6 | 1116 | 1 | 0.17 (0.02†–1.41†) |
SLCI in unvaccinated subjects | NAI30034 | 544 | 17 | 562 | 3 | 0.17 (0.05†–0.58†) |
SLCI in subjects with respiratory disease | NAI30034 | 695 | 17 | 684 | 3 | 0.18 (0.05†–0.61†) |
SLCI in subjects with cardiovascular disease | NAI30034 | 307 | 1 | 331 | 0 | Not estimable |
SLCI in subjects with diabetes | NAI30034 | 370 | 3 | 359 | 0 | Not estimable |
Trial NAI3003475 also evaluated the efficacy of zanamivir in seasonal prophylaxis in subjects aged 65 years and above (Table 12). Of these, 13% had respiratory disease, 15% had cardiovascular disease, 9% had diabetes and 10% had two or three of the above risk factors. 44 SLCI was observed in 1/946 and 5/950 of the zanamivir and placebo group subjects respectively, resulting in an RR of 0.20 (0.02†–1.72†). The proportion experiencing any adverse events was 53% in each group (498/946 and 501/950 in the zanamivir and placebo arms respectively). 44
Outcome | Trial | Total no. in placebo group | No. in placebo group with an event | Total no. in zanamivir group | No. in zanamivir group with an event | RR (95% CI, p-value if available) |
---|---|---|---|---|---|---|
SLCI in subjects aged 65 and above | NAI30034 | 950 | 5 | 946 | 1 | 0.20 (0.02†–1.72†) |
A total of four trials of the use of zanamivir in post-exposure prophylaxis in households were included in the review. These were studies published by Hayden et al. 46 and Kaiser et al. 72 and a report by Monto and colleagues47 that was published subsequent to the cut-off date for inclusion of evidence in the original HTA review10 (Table 13).
Outcome | Trial | Total no. in placebo group | No. in placebo group with an event | Total no. in zanamivir group | No. in zanamivir group with an event | RR (95% CI, p-values if available) |
---|---|---|---|---|---|---|
SLCI in contacts of all index cases | NAI30010 | 423 | 40 | 414 | 7 | 0.18 (0.08†–0.39†) |
NAI30031 | 630 | 55 | 661 | 12 | 0.21 (0.11†–0.38†) | |
NAIA/B2009 | 144 | 9 | 144 | 3 | 0.33 (0.09†–1.21†) | |
Pooled (random effects) | 0.21 (0.13–0.33) | |||||
(p = 0.72, I2 = 0%) | ||||||
SLCI in contacts of influenza-positive index cases | NAI30010 | 215 | 33 | 195 | 6 | 0.20 (0.09†–0.47†) |
SLCI in contacts of influenza-positive index cases | NAI30031 | 398 | 51 | 368 | 9 | 0.19 (0.10†–0.38†) |
Pooled (random effects) | 0.19 (0.11–0.33) | |||||
(p = 0.93, I2 = 0%) |
Hayden et al. 46 presented evidence from trial NAI30010 that zanamivir, when administered to household contacts (aged 5 years and above) of index cases with ILI for 10 days, conveyed an RR of SLCI of 0.18 (95% CI 0.08†–0.39†). For individual contacts of influenza-positive index cases, the RR was 0.20 (95% CI 0.09†–0.47†). Total adverse events occurred in 50% of the placebo arm and 44% of subjects receiving zanamivir, of which 5% and 6% respectively were possibly drug related. Withdrawals for any reason were made in 5/423 (1.2%) and 3/414 (0.7%) of subjects in the placebo and zanamivir groups. One withdrawal due to adverse effects was made in the zanamivir group while none was made in the placebo group. Study medication was discontinued due to adverse events in 0.2% of the placebo group and 0.5% of the zanamivir arm. In contacts with laboratory-confirmed influenza, the median time to alleviation of symptoms without use of medication was 8.0 days in the placebo group and 5.5 days in the zanamivir group. The percentage of cases with complications requiring antibiotics was 8% in the placebo arm and 5% in the zanamivir arm. Index cases in households randomised to receive zanamivir also received zanamivir as treatment, while index cases in the placebo arm received placebo treatment.
Trials NAIA2009 and NAIB2009, performed by Kaiser et al. 72 and reported as a single trial in the literature, investigated the use of zanamivir for 5 days in household contacts of index cases with ILI. Index cases received no treatment. During the 5 days of prophylaxis, the RR for developing SLCI was 0.33 (95% CI 0.09†–1.21†) and during the 10 days after initiation of medication, the RR for SLCI was 0.36 (95% CI 0.12†–1.12†). Length of illness was shorter in the zanamivir group than in the placebo group (mean duration of significant influenza-like symptoms 0.2 days versus 0.6 days, p = 0.016). Potentially drug-related adverse effects occurred in 17% (25/144) of the placebo group and 19% (27/144) of the zanamivir group, and comprised primarily headaches, fatigue, nasal symptoms and throat discomfort.
In trial NAI30031, reported by Monto et al., which investigated the efficacy of zanamivir administered for 10 days as post-exposure prophylaxis in household contacts of index cases with ILI,46 protective efficacy for individual contacts was 79%† (95% CI 62†–89†; RR 0.21) in the ITT population (when calculated by the Assessment Group) and 81%† among individual contacts of influenza-positive index cases (95% CI 62†–90†; RR 0.19). Index cases did not receive treatment. For influenza A, the protective efficacy was 79% (95% CI 55–90; RR 0.21), and for influenza B, the reported protective efficacy was 87% (95% CI 64–95; RR 0.13). However, when calculated by the Assessment Group, the protective efficacy against influenza B was 79%† (95% CI 46†–92†; RR 0.21). The authors observed that, in some cases, there was a mismatch between the strains with which the contact cases and index cases were infected, demonstrating infection from an additional source of exposure. Significantly fewer households randomised to zanamivir prophylaxis reported a contact developing a complication of laboratory-confirmed influenza (2% versus 6%, p = 0.01). Adverse events (all of which were consistent with ILI) occurred in 52% of the placebo group and 42% of the zanamivir group. Adverse events considered by the investigators to be drug related were observed in 7% of placebo subjects and 6% of zanamivir subjects. Total withdrawals were made in 1.7% (11/630) of the placebo subjects and 0.9% (6/661) of the zanamivir subjects. No withdrawals were due to adverse events.
In contacts with SLCI from the zanamivir-treated group, the median time to alleviation of symptoms (5 days) was reduced by 1.5 days, from 6.5 days in the placebo group, demonstrating milder disease. 47 This is supported by evidence that households randomised to zanamivir and with at least one symptomatic ILI contact case spent less time confined to bed/incapacitated, with nearly a 1-day difference in the mean time confined to bed/incapacitated per household between treatment arms (1.8 days versus 2.6 days, p = 0.053).
Additional data relating to trial NAI30031 were identified from the sponsor submissions. 43 One contact case in the placebo group was hospitalised for more than 5 days. Two zanamivir-treated contact cases were also hospitalised. One contact case was hospitalised for less than 1 day and another for more than 5 days. The numbers were too low to make a meaningful comparison.
The need for non-prescription medications in households randomised to zanamivir was lower in subjects receiving zanamivir versus placebo (13% zanamivir versus 19% placebo, p = 0.076). The number of households requiring prescription medications was also lower (11% of zanamivir subjects versus 17% of placebo subjects, p = 0.100). Significantly fewer households receiving zanamivir required additional health-care contacts (20% of zanamivir subjects versus 32% of placebo subjects, p = 0.004). Among those households reporting at least one contact case with symptomatic ILI, the zanamivir group required a mean time off work/school of 10.9 hours per household compared with 15.1 hours for those in the placebo group (p = 0.693).
When data relating to SLCI were pooled by meta-analysis using a random-effects model, the combined protective efficacy was 79% [RR 0.21, 95% CI 0.13–0.33 (test of heterogeneity: p = 0.72, I2 = 0)]. The trial reported by Kaiser et al. 72 differed from the trials by Hayden et al. 46 and Monto et al. 47 in that all subjects were unvaccinated and prophylaxis was administered for 5 rather than 10 days. When data abstracted from the study reported by Kaiser et al. 72 were removed, the RR decreased to 0.20 (95% CI 0.12–0.32), corresponding to a slightly higher protective efficacy of 80% (test of heterogeneity: p = 0.77, I2 = 0).
When data for the incidence of SLCI in contacts of influenza-positive index cases from trials NAI3001046 and NAI3003147 were pooled, an RR of 0.19 (95% CI 0.11–0.33) was obtained (p = 0.93, I2 = 0%).
Halloran et al. 45 presented a pooled analysis of data from the trials by Hayden et al. 46 and Monto et al. ,47 proposing a prophylactic efficacy against illness of 75% (95% CI 54–86) and a reduction in infectiousness of 19% (95% CI –160 to 75). The secondary analysis by Halloran et al. also assessed pathogenicity of influenza in the treatment and control arms of the household post-exposure prophylaxis trials. Pathogenicity was defined as the ability of the virus to cause disease in an infected person and was calculated as the number of contacts with SLCI divided by the number of contacts with laboratory-confirmed influenza infections (symptomatic or asymptomatic). Pathogenicity was lower among subjects treated with zanamivir than among those in the placebo group. In the study reported by Hayden et al.,46 pathogenicity in the control group was reported as 40/66 (61%) while in the zanamivir group it was 7/26 (27%). In the study presented by Monto et al. 47 pathogenicity in the control group was 55/105 (52%) and in the zanamivir group this value was 12/48 (25%). Data from both of these studies included all contacts, whether or not the index case was influenza positive.
Two trials investigating zanamivir in preventing outbreaks of influenza in the elderly in long-term care settings were included. 76–78
Limited data relating to the prophylactic efficacy of zanamivir could be drawn from the trial by Gravenstein et al. 78 The study compared zanamivir with standard of care (rimantadine for influenza A and placebo for influenza B). As only 25 subjects were randomised during two outbreaks of influenza B and no subjects developed influenza, the data relating to influenza B were excluded from further analysis in the published report. Potentially drug-related adverse effects were reported in 38% of placebo subjects and 34% of zanamivir subjects. Withdrawals from the study due to adverse events occurred at rates of 0/13 in the placebo arm and 2/238 (0.8%) in the zanamivir arm. Early medication discontinuation due to adverse events was necessary in 0/13 of the placebo subjects and 11/238 (4.6%) of the zanamivir group.
The study by Ambrozaitis et al. 76,77 differed from that described above in that the elderly, at-risk subjects living in long-term care had a much lower proportion of vaccination (Table 14). During influenza A outbreaks, prophylaxis conferred a 32%† protective efficacy against SLCI as calculated by the Assessment Group (95% CI –27† to 67†). The authors noted that all cases of SLCI occurred in Lithuania (where none of subjects had been vaccinated). A higher protective efficacy of 70% (95% CI 13–89) was observed for laboratory-confirmed febrile illness. When subjects who became ill on days 1 or 2 were excluded, the protective efficacy against SLCI as calculated by the Assessment Group was 35%† (95% CI –40† to 70†). 44 No differences in SLCI were observed by age group. 44 Complications of SLCI during the first 28 days following prophylaxis initiation were observed at a lower rate in the zanamivir-treated subjects than in the placebo group, although this difference was not statistically significant (5% versus 6%, p = 0.653). Respiratory tract infections occurred in fewer subjects in the zanamivir arm (3% versus 6%), as did complications requiring antibiotics (2% versus 3%, p = 0.445). Withdrawals from the study due to adverse events were reported as 1/249 (0.4%)in the placebo arm and 2/240 (0.8%) in the zanamivir arm. Early discontinuation of medication due to adverse events occurred in 2/249 (0.8%) and 6/240 (2.5%) of the placebo and zanamivir subjects respectively. The following additional data were identified in the sponsor submissions. 44 Drug-related adverse effects were slightly higher in the zanamivir-treated arm [16/242 (7%)] than in the placebo arm [14/252 (6%)]. Serious adverse events occurred in 6/252 (2.4%) of placebo subjects and 6/242 (2.5%) of zanamivir subjects. There were no serious adverse events that were considered to be related to the study drug. Adverse events during prophylaxis in high-risk subjects were lower in the zanamivir group than in the placebo arm [64/202 (32%) versus 80/215 (37%)]. Subjects with high-risk respiratory conditions also experienced fewer adverse events when receiving zanamivir than did their placebo counterparts [(30/83 (36%) versus 32/80 (40%)].
Outcome | Trial | Total no. in placebo group | No. in placebo group with an event | Total no. in zanamivir group | No. in zanamivir group with an event | RR (95% CI) |
---|---|---|---|---|---|---|
SLCI | NAIA3004 | 249 | 23 | 240 | 15 | 0.68† (0.36†–1.27†) |
No trials in which zanamivir was used in accordance with licensed indications in prophylaxis against experimentally-induced influenza were identified.
Adherence in the zanamivir trials appeared to be high, suggesting the use of the Diskhaler for topical oral inhalation of drug to be acceptable to study participants. In one study, 95% and 97% of placebo and zanamivir-allocated participants took study doses over a 23–28 day period. 70 In another study, 90% of zanamivir subjects and 89% of placebo subjects took at least 24 doses for at least 24 days, with fewer than 1% requiring assistance in administering the drug. 75 In a further study, 97% of placebo group contacts and 99% of zanamivir group contacts took 8–10 doses (80–100%) of study medication. 47 Compliance in an additional study was high, with 98% of all participants taking 8–10 doses of the study drug. 46 In the studies by Ambrozaitis et al. 76,77 and Gravenstein et al. ,78 undertaken in the elderly, subjects who missed two or more consecutive days of medication were considered non-compliant. These proportions were very low, at 1% of total participants76 and 2% or less of total participants. 77
Several trials tested viral isolates for their susceptibility to zanamivir. 46,75,76,77 No evidence of resistance to zanamivir was observed, although rimantadine-resistant variants were reported by Gravenstein et al. 78
Convincing data were obtained for a relatively high protective efficacy of seasonal prophylaxis in healthy adults. The evidence base has been strengthened considerably by the publication of a large-scale trial specifically investigating the efficacy of zanamivir in seasonal prophylaxis in at-risk adolescents and adults, including the elderly. A very high protective efficacy was obtained; protective efficacy was also high when data were presented by age and risk subgroups. Post-exposure prophylaxis was also shown to be efficacious in preventing transmission of SLCI in households, with shorter and milder disease, fewer complications and a more rapid return to normal activities among subjects receiving the intervention. The evidence for outbreak control in the elderly in long-term care was more limited, but a relatively low protective efficacy against SLCI was demonstrated, with all cases occurring in unvaccinated subjects. Adverse events were similar in both treatment arms and across all studies.
Assessment of effectiveness
Discussion
The relative efficacies of amantadine, oseltamivir and zanamivir in preventing SLCI are summarised in Table 15. As in the previous HTA review,10,88 evidence for effectiveness of amantadine in prophylaxis was limited. However, amantadine was reported to be effective in preventing SLCI in healthy adolescents. The effectiveness of oseltamivir in prophylaxis against SLCI was demonstrated in a number of subgroups, particularly in seasonal prophylaxis in at-risk elderly subjects and in post-exposure prophylaxis in mixed households. Zanamivir was also shown to prevent influenza, most notably in seasonal prophylaxis among at-risk adults and adolescents, healthy and at-risk elderly individuals and in post-exposure prophylaxis in mixed households. Variation in the measurement and reporting of adverse events was observed among trials. However, no clear trends for the higher incidence of adverse events in treatment groups than in control groups (and vice versa) were observed for amantadine, oseltamivir or zanamivir or across interventions. Interventions appeared to be well tolerated, with few serious drug-related adverse events or drug-related withdrawals. Less evidence was available to demonstrate the effectiveness of the interventions in reducing the impact of influenza in terms of complications, hospitalisations, length of illness and time to return to normal activities. The identified studies suggested that oseltamivir and zanamivir may be effective in preventing influenza-associated complications. While there was no significant difference in numbers of subjects hospitalised between zanamivir and placebo groups, limited evidence was presented suggesting that individuals receiving zanamivir experienced a hospital stay of shorter duration. Limited evidence suggested that amantadine, oseltamivir and zanamivir were effective in shortening the length of influenza illness. The severity of symptoms was also reduced in amantadine-treated subjects. Additional evidence also suggested that fewer subjects receiving oseltamivir or zanamivir were incapacitated due to influenza illness, with a shorter time to return to normal activities. No evidence relating to HRQoL or mortality could be identified for inclusion in the clinical effectiveness review. As stated previously, the findings from the included trials in the clinical effectiveness review should be considered in conjunction with evidence for the development of antiviral resistance by influenza strains, particularly against amantadine, and of adverse events associated with amantadine, issues which may not be presented within the trials, but have the potential to have considerable impact on the use of the interventions in clinical practice.
Prophylactic strategy | Relative risk of developing SCLI (95% CI) | ||
---|---|---|---|
Amantadine | Oseltamivir | Zanamivir | |
Seasonal prophylaxis | |||
In healthy children | Dosage not established in children | NDA | NDA |
In at-risk children | Dosage not established in children | NDA | NDA |
In healthy adults | 0.40 (0.08–2.03)56 (from one trial) | 0.27 (0.09–0.83)66 (pooled estimate from two trials as reported by Assessment Group) | 0.32 (0.17–0.63)70 (from one trial) |
In at-risk adults and adolescents | NDA | NDA | 0.17 (0.07–0.44)75 (from one trial) |
In healthy elderly subjects | No data reported55 | NDA | 0.20 (0.02–1.72)75 (from one trial) |
In at-risk elderly subjects | No data reported55 | 0.08 (0.01–0.63)63 (98% subjects with concomitant disease; from one trial) | 0.20 (0.02–1.72)75 (from one trial) |
Post-exposure prophylaxis | |||
In mixed households | NDA | 0.19 (0.08–0.45)48,49 (from two trials) | 0.21 (0.13–0.33)46,47,72 (from three trials) |
In healthy children | Dosage not established in children | 0.36 (0.15–0.84)48 (from one trial) | NDA |
In at-risk children | Dosage not established in children | NDA (subjects with a number of chronic conditions excluded)47 | NDA |
In healthy adults and adolescents | 0.10 (0.03–0.34)59 (from one trial) | NDA | NDA |
In at-risk adults and adolescents | NDA | NDA | NDA |
In healthy elderly subjects | NDA | NDA | NDA |
In at-risk elderly subjects | NDA | NDA | 0.68 (0.36–1.27)76 (subjects 85% at risk of complications) |
Chapter 4 Assessment of cost-effectiveness
This chapter reports the methods and results of a systematic review of existing economic evaluations of influenza prophylaxis and the development of an independent health economic model to evaluate the cost-effectiveness of amantadine, oseltamivir and zanamivir for the seasonal prophylaxis and post-exposure prophylaxis of influenza. The systematic review of existing economic evaluations is presented below. The methods and results of the Assessment Group model are presented in Independent economic assessment (p. 61) and Cost-effectiveness results (p. 84) respectively.
Systematic review of existing cost-effectiveness evidence
Methods
The methods used to systematically search electronic databases to identify studies relating to the cost-effectiveness of amantadine, oseltamivir and zanamivir for the post-exposure prophylaxis and seasonal prophylaxis of influenza are described in Chapter 3 (see Methods for reviewing effectiveness, p. 17, and Appendix 1). Economic evaluations identified for inclusion in the review were also handsearched to identify other relevant cost-effectiveness studies of influenza prophylaxis that were not identified by the electronic searches. Alongside published economic evaluations, manufacturers’ submissions to NICE, where available, were also included in the review of economic evaluations. Appraisal of study quality was undertaken based on checklists for assessing quality in economic evaluations89 and mathematical models. 90
Results
Studies included in the review of cost-effectiveness
The systematic searches identified 580 citations of studies relating to the cost-effectiveness of amantadine, oseltamivir and zanamivir in the prevention of influenza. Titles and abstracts of each citation were screened for possible inclusion in the review. Of the initial 580 citations identified by the searches, full papers of 65 studies were retrieved for further detailed evaluation. Six of these studies met the inclusion criteria for the review described in Chapter 3 (see Inclusion and exclusion criteria, p. 17). In addition, one sponsor submission was received from Roche; this report included the details of a mathematical model to assess the cost-effectiveness of oseltamivir for the prophylaxis of influenza. Evidence concerning cost-effectiveness was not submitted by the manufacturers of zanamivir or amantadine. In total, seven economic evaluations were included in the systematic review. A summary of studies included or excluded from the review of cost-effectiveness is presented in Figure 4.
Table 16 details the characteristics of the seven studies included in the review of cost-effectiveness.
Roche submission, 200720 | Sander et al., 200691 | Risebroughet al., 200592 | Turner et al., 200310 | Scuffham and West, 200293 | Demicheli et al., 200094 | Patriarca et al., 198795 | |
---|---|---|---|---|---|---|---|
Type of economic analysis | Cost–utility analysis | Cost-effectiveness analysis and cost–utility analysis | Cost-effectiveness analysis | Cost-effectiveness analysis and cost–utility analysis | Cost-effectiveness analysis | Cost-effectiveness analysis | Cost-effectiveness analysis |
Health economic perspective | NHS | Health-care payer | Single government payer | NHS | Health-care financier | Ministry of Defence | Not reported (direct costs only included in evaluation) |
Health economic comparisons | (1) Oseltamivir; (2) Zanamivir; (3) Amantadine; (4) usual care | (1) Oseltamivir prophylaxis, no treatment; (2) no prophylaxis, no treatment; (3) no prophylaxis, oseltamivir treatment | (1) Oseltamivir prophylaxis; (2) amantadine prophylaxis; (3) no prophylaxis | (1) No intervention; (2) vaccination, no prophylaxis; (3) no vaccination, amantadine prophylaxis; (4) no vaccination, oseltamivir prophylaxis; (5) no vaccination, zanamivir prophylaxis; (6) vaccination plus amantadine prophylaxis; (7) vaccination plus oseltamivir prophylaxis; (8) vaccination plus zanamivir prophylaxis | (1) No intervention; (2) opportunistic vaccination; (3) comprehensive vaccination; (4) oseltamivir chemoprophylaxis; (5) rimantadine chemoprophylaxis; (6) oseltamivir treatment; (7) rimantadine treatment | (1) Vaccination; (2) amantadine prophylaxis; (3) NI | (1) No control; (2) vaccination, no chemoprophylaxis; (3) vaccination plus chemoprophylaxis; (4) outbreak control prophylaxis, no vaccination; (5) continuous chemoprophylaxis, no vaccination |
Type of prophylaxis | Seasonal prophylaxis (28–42 days depending on drug) and post-exposure prophylaxis (10 days) | Post-exposure prophylaxis; duration appears to be 7–10 days | Post-exposure prophylaxis; median duration of prophylaxis without ILI reported to be 12 days | Seasonal prophylaxis for 6 weeks (42 days) | Seasonal prophylaxis for 4 weeks (28 days) |
Seasonal prophylaxis for 62 days |
Post-exposure (outbreak) prophylaxis for 30 days; continuous seasonal prophylaxis for 3 months (≈ 91 days) |
Population characteristics | Healthy children (1–5 years); at-risk children (1–5 years); healthy children (1–15 years); at-risk children (1–15 years); healthy adults (> 15 years); at-risk adults (> 15 years) | Families with members ≥ 13 years | Elderly vaccinated patients in long-term care facility | Healthy adults, children, residential care elderly, high-risk adults | Elderly patients (age > 65 years in UK analysis) | British army effectives | Elderly nursing home residents |
Time horizon used in the analysis | Single influenza season; adjustments for loss in lifetime QALYs due to premature death | Single influenza season; adjustments for loss in lifetime QALYs due to premature death | 30 days (intended to reflect a single institutional outbreak) | Single influenza season; adjustments for loss in lifetime QALYs due to premature death | Typical (average) influenza season (including years of potential life lost due to premature death) | Single influenza season | Typical (average) influenza season (including years of potential life lost due to premature death) |
Health economic outcomes | Incremental cost per QALY gained (pairwise, i.e. oseltamivir versus comparator) | Incremental cost per ILI case avoided; incremental cost per QALY gained | Incremental costs (or savings) per ILI case avoided | Incremental cost per QALY gained; incremental cost per illness day avoided | Cost per life-year gained; cost per hospitalisation averted; cost per death averted; cost per morbidity day averted | Incremental cost per avoided case | Incremental cost per illness averted; incremental cost per hospitalisation averted; incremental cost per death averted |
Currency | Pounds sterling (£) | Pounds sterling (£) | Canadian dollars ($) | Pounds sterling (£) | Euro (€) | Pounds sterling (£) | US dollars ($) |
Modelling approach | Decision tree model | Decision tree model evaluated using Monte Carlo sampling | Decision tree model | Decision tree model | Decision tree model | Decision tree model | Decision tree model |
Potential conflicts of interest | Manufacturer of oseltamivir (Roche) | Study funded by Hoffman–La Roche | Study funded by Hoffman–La Roche | One author is an ad hocconsultant for Hoffman–La Roche and has received fees by other influenza prophylaxis sponsor companies |
Study funded by grants from Solvay, Aventis, Chiron, Berna and Medeva |
One author is an ad hocconsultant for Hoffman–La Roche | Not reported |
Review of existing economic evaluation studies
Roche submission to NICE
The Roche submission to NICE20 reports the use of a mathematical model to estimate the cost-effectiveness of oseltamivir for the seasonal prophylaxis and post-exposure prophylaxis of influenza. The cost-effectiveness model was submitted to NICE for scrutiny by the Assessment Group. The model presented within Roche’s submission is based on the simulating anti-influenza value and effectiveness (SAVE) model, and as such the structure and parameter set is similar to the model reported by Sander et al. 91 Twenty variations of the SAVE model were made available to the Assessment Group. The model compares oseltamivir prophylaxis with amantadine prophylaxis, zanamivir prophylaxis and no prophylaxis in the seasonal and post-exposure settings for four populations: otherwise healthy adults (including children > 12 years), at-risk adults (including children > 12 years), children aged 1–12 years and children aged 1–5 years. The analysis for children aged 1–5 years includes only usual care as a comparator for oseltamivir due to restrictions in the licensed indications of amantadine and zanamivir prophylaxis. It should also be noted that amantadine is licensed only in children aged 10 years or over; this prophylactic option is, however, included in the analysis for children aged 1–12 years. The base-case analysis was undertaken from the perspective of the NHS; secondary analysis was also reported from the societal perspective. The model is reported to use a lifetime horizon, whereby all important events occur within a 1-year time horizon with longer-term adjustments for quality-adjusted life-years (QALYs) lost as a result of premature death due to ILI. Cost-effectiveness is expressed in terms of the incremental cost per QALY gained, although this is based on pairwise comparisons of oseltamivir versus an alternative prophylactic option. In line with current recommendations from NICE,96 health outcomes were discounted at a rate of 3.5%; owing to the time frame used within the model, costs were not subjected to discounting.
The submission states that for oseltamivir versus amantadine and usual care, a cost-effectiveness analysis was undertaken. 20 The model assumes that oseltamivir and zanamivir are equivalent in terms of preventative efficacy and the submission reports a cost minimisation exercise for this comparison. However, the submission does not report the results of any head-to-head trials of zanamivir and oseltamivir prophylaxis (i.e. superiority, non-inferiority or equivalence trials) which provide any evidence to support the assumption of equivalence. Furthermore, the systematic review of clinical effectiveness presented in Chapter 3 did not identify any clinical evidence which could be considered to validate this assumption. Consequently, the use of a cost minimisation analysis for oseltamivir and zanamivir appears to be unjustified; even if equivalence trials were available, the comparative prophylactic effects would remain subject to uncertainty and should therefore be considered within the health economic analysis. Importantly, the Roche submission states that the preventative efficacy estimates have a considerable impact on the cost-effectiveness of oseltamivir prophylaxis. 20
Vaccination is not explicitly considered within the model, either as an option for influenza prevention or as a characteristic of the patient cohort. The studies used to estimate the preventative efficacy of zanamivir and oseltamivir included some patients who had been vaccinated and some patients who had not been vaccinated.
The model uses a deterministic decision tree approach which is reported to be appropriate as it captures a simple ILI pathway and events do not occur more than once. 20 The Roche submission argues that the results are conservative as the benefits of a contact case receiving prophylaxis and subsequently not infecting other individuals are not captured (herd immunity effects). The structural assumptions employed in the model are identical for seasonal and post-exposure prophylaxis settings. The model is reported to be based on ILI rather than true influenza alone, as it is intended to capture the impact of both true influenza and other ILI on costs and health outcomes.
The structures of the seasonal prophylaxis and post-exposure prophylaxis models are simple. For the post-exposure model, an individual who has been in contact with an ILI index case in a household may visit his or her GP to receive prophylaxis or may do nothing. For the seasonal prophylaxis model, the individual may or may not have been in contact with an index case when prophylaxis is initiated. The model assumes that one household member can obtain prescriptions for three contacts in the household. Contact cases may or may not go on to develop ILI. Individuals who develop ILI may be treated using oseltamivir (at-risk populations only) or usual care. Individuals who develop ILI may or may not develop complications. ILI complications are treated in an inpatient or outpatient setting depending on the severity of the complication. The model includes three complications: bronchitis, pneumonia and otitis media in children. Patients who develop ILI complications may survive or may die.
The model includes different attack rates for the seasonal prophylaxis models and for the post-exposure prophylaxis models; post-exposure attack rates are assumed to be higher than those for the seasonal prophylaxis models as contacts have by definition had previous exposure to an index case who may have influenza (Gavin Lewis, Head of Health Economics, Roche, personal communication). The submission states that the attack rates used in the post-exposure prophylaxis model are intended to represent the proportion of patients who, after being exposed to ILI, go on to develop ILI. 20 However, these are sourced from the oseltamivir post-exposure prophylaxis trial reported by Hayden et al. 48 and represent only laboratory-confirmed influenza, rather than all ILI. The attack rate for adults in the seasonal prophylaxis models was taken from Hayden et al. (assumed to be 4.8%). 66 The attack rate for children in the seasonal prophylaxis models was reported to be in the region of 10%,20 although the basis of this assumption is not reported in the submission. The methods used to derive upper and lower CIs around these attack rates are unclear from the submission.
The preventative efficacies of oseltamivir and zanamivir prophylaxis were sourced from a meta-analysis reported by Halloran et al. 45 The effectiveness of amantadine prophylaxis was derived from Monto et al. , although it should be noted that within this study patients received amantadine at a dose of 200 mg, which does not reflect its current licensed indications. 97 The model assumes that seasonal prophylaxis is effective across the whole influenza season; this is likely to be optimistic as patients may become susceptible to infection after they stop taking prophylaxis (see Chapter 3). Seasonal prophylaxis using zanamivir and oseltamivir are assumed to be equivalent to post-exposure prophylaxis using zanamivir and oseltamivir. The relative difference between amantadine as post-exposure prophylaxis and as seasonal prophylaxis was assumed to be the same as the relative difference for oseltamivir in each setting due to a lack of clinical trial evidence. The model does not include the possibility of resistance to amantadine, oseltamivir or zanamivir.
The probability of experiencing specific complications of ILI were sourced from a study reported by Meier et al. 12 It should be noted that these complication rates relate to ILI rather than true influenza alone (despite the claim that the model operates in terms of ILI, the Roche model actually appears to be based on true influenza attack rates). Complication rates due to influenza in children are assumed to be the same for both the 1–5 years age group and the 1–12 years age group. 20 The incidence of pneumonia and bronchitis was sourced from Meier et al. 12 However, the submission states that the incidence of otitis media is likely to be under-reported by Meier et al. Instead, the Roche model uses estimates sourced from oseltamivir clinical trial data;20 however, this estimate is only slightly higher than the estimate reported by Meier et al. (28% in Meier et al. versus 32.4% in the oseltamivir trials).
The probability of hospitalisation was taken from two US studies;98,99 these may not reflect UK practice. The model assumes that the probability of hospitalisation due to bronchitis is the same as that for other ILI. The probability of hospitalisation due to specific complications of ILI is assumed to be the same across the model populations. The model assumes the length of hospital stay to be 4 days for influenza and 7 days for pneumonia irrespective of patient population. The risk of death due to ILI is assumed to be the same as the risk of death due to ILI complications; this assumption is unlikely to be reasonable as ILI complications are known to increase the risk of death. It is likely that this assumption would overstate the benefits of avoiding a case of influenza.
The model includes HRQoL adjustments for individuals who develop influenza and complications of ILI. Utility estimates for patients experiencing an episode of influenza were derived from Likert valuations of patients with laboratory-confirmed influenza within the oseltamivir treatment trials. These rating scale data were converted to visual analogue scale (VAS) valuations and subsequently converted to time trade-off (TTO) utilities using a similar methodology to Turner et al. 10 Utility scores for patients with ILI, bronchitis and pneumonia were based on a Dutch person trade-off study reported by Stouthard et al. 100 Utility scores are applied for the duration of illness, based on clinical trial data (Gavin Lewis, Head of Health Economics, Roche, personal communication). In addition, the model includes the number of potential QALYs lost due to premature death resulting from ILI complications. Importantly, the model assumes that each potential year of life lost is valued at a state of perfect health; this assumption biases in favour of more effective prophylaxis options. The submission itself notes this assumption as a weakness of the model. 20
The model includes costs associated with drug acquisition, GP consultations, diagnostic tests, antibiotics and associated treatments, and hospitalisation for the treatment of ILI complications. Resource use estimates used in the model were derived from a variety of sources. Estimates of drug prescriptions, tests and investigations performed, primary and secondary care resource use for patients with influenza and certain complications were derived from the National Ambulatory Medical Care Survey (NAMCS);101 this is a US database, and may not reflect UK treatment patterns. Assumptions taken from this database were validated by Roche through a structured interview with one clinical expert. Sources for estimates of unit costs included the Personal Social Services Research Unit (PSSRU),102 the Monthly Index of Medical Specialties (MIMS) database,103 the MEDTAP database and the BNF. 14 Rates of antibiotic use were based on expert opinion.
Importantly, the model does not include the cost of drug wastage, and the cost of each prophylaxis course is calculated on the basis of the mean cost per tablet. The difference between the cost of oseltamivir with and without wastage is most pronounced in the seasonal prophylaxis indication for adults, these costs being £68.88 without wastage and £81.80 when wastage is included (see Modelling resource use and costs associated with influenza and other ILI, p. 76). Consequently, the acquisition cost of oseltamivir as seasonal prophylaxis is underestimated in the Roche submission. However, given the assumption of equivalence between oseltamivir and zanamivir, and the lower cost of a seasonal prophylaxis course using zanamivir, oseltamivir is actually dominated by zanamivir in this indication even when wastage is excluded.
The model assumes a single cost associated with hospitalisation due to ILI or ILI complications; this is quoted as £286 per day. This estimate is based on the cost of an inpatient day for mental health services; the justification for using this hospitalisation cost is unclear. 102 The model does not explicitly include the possibility of patients requiring intensive therapy unit (ITU) care or mechanical ventilation. A further potential problem with the SAVE model is that it assumes that all patients with ILI will incur GP consultation costs; this is not necessarily true as not all patients with ILI (whether influenza or not) will consult their GP. 104 Further, the model does not consider any costs associated with adverse events of prophylaxis or treatment using amantadine, oseltamivir or zanamivir.
The submission includes the details of one-way and probabilistic sensitivity analysis to explore uncertainty surrounding model parameters. The probabilistic sensitivity analysis was undertaken using @risk software alongside Microsoft excel.
It should be noted from the outset that the cost-effectiveness analysis presented within the Roche submission to NICE was not fully incremental; instead, 20 incremental cost-effectiveness ratios were presented for pairwise comparisons of oseltamivir versus amantadine, oseltamivir versus zanamivir and oseltamivir versus usual care for each population group across seasonal and post-exposure prophylaxis settings. The Assessment Group reanalysed the results presented within the Roche submission to generate fully incremental estimates of the cost-effectiveness of each prophylactic option compared with each other and usual care. The results of the reanalyses of the post-exposure models are presented in Tables 17–20.
Option | Costs | QALYs | Incremental cost | Incremental QALYs | ICER |
---|---|---|---|---|---|
Usual care | £44.54 | 109.619 | – | – | – |
Oseltamivir | £73.54 | 109.624 | £29.00 | 0.005 | £5800 |
Option | Costs | QALYs | Incremental cost | Incremental QALYs | ICER |
---|---|---|---|---|---|
Usual care | £44.84 | 108.678 | – | – | – |
Amantadine | £122.75 | 108.68 | Dominated by oseltamivir | ||
Oseltamivir | £84.74 | 108.683 | £39.90 | 0.005 | £7980 |
Zanamivir | £139.34 | 108.683 | _ | _ | Dominated by oseltamivir |
Option | Costs | QALYs | Incremental cost | Incremental QALYs | ICER |
---|---|---|---|---|---|
Usual care | £12.61 | 91.336 | – | – | – |
Amantadine | £89.65 | 91.337 | £77.04 | 0.001 | Extendedly dominated |
Oseltamivir | £92.84 | 91.339 | £3.19 | 0.002 | £26,743 |
Zanamivir | £126.35 | 91.339 | – | – | Dominated by oseltamivir |
Option | Costs | QALYs | Incremental cost | Incremental QALYs | ICER |
---|---|---|---|---|---|
Usual care | £13.30 | 85.119 | – | – | – |
Amantadine | £89.54 | 85.138 | £76.24 | 0.019 | Extendedly dominated |
Oseltamivir | £91.50 | 85.159 | £78.20 | 0.04 | £1955 |
Zanamivir | £123.60 | 85.159 | – | – | Dominated by oseltamivir |
The results suggest that the incremental cost-effectiveness of oseltamivir for post-exposure prophylaxis is consistently expected to be below £27,000 across all paediatric and adult populations. The finding that zanamivir is consistently dominated by oseltamivir is unsurprising, as the model assumes that oseltamivir and zanamivir have equivalent preventative efficacy and no differential impact on HRQoL due to adverse events, yet zanamivir is assumed to be more expensive than oseltamivir over the course of prophylaxis (the submission does not include the proposed price reduction for zanamivir). Uncertainty surrounding the relative efficacies of oseltamivir and zanamivir are not included in the model. The model suggests that amantadine is dominated or extendedly dominated by oseltamivir within each indication.
The results of the reanalyses of the seasonal prophylaxis models are presented in Tables 21–24.
Option | Costs | QALYs | Incremental cost | Incremental QALYs | ICER |
---|---|---|---|---|---|
Usual care | £28.58 | 109.623 | – | – | – |
Oseltamivir | £168.25 | 109.626 | £139.67 | 0.003 | £46,556.67 |
Option | Costs | QALYs | Incremental cost | Incremental QALYs | ICER |
---|---|---|---|---|---|
Usual care | £20.72 | 108.681 | – | – | – |
Amantadine | £95.48 | 108.683 | £74.76 | 0.002 | £37,380 |
Oseltamivir | £214.04 | 108.684 | £118.56 | 0.001 | £118,560 |
Zanamivir | £306.32 | 108.684 | – | – | Dominated by oseltamivir |
Option | Costs | QALYs |
Incremental cost |
Incremental QALYs |
ICER |
---|---|---|---|---|---|
Usual care | £8.18 | 91.337 | _ | – | – |
Amantadine | £87.22 | 91.338 | £79.04 | 0.001 | £79,040 |
Zanamivir | £302.07 | 91.339 | £214.85 | 0.001 | £214,850 |
Oseltamivir | £302.48 | 91.339 | _ | – | Dominated by zanamivir |
Option | Costs |
QALYs |
Incremental cost |
Incremental QALYs |
ICER |
---|---|---|---|---|---|
Usual care | £8.63 | 85.134 | – | _ | – |
Amantadine | £86.93 | 85.146 | £78.30 | 0.012 | £6525.00 |
Zanamivir | £300.78 | 85.16 | £213.85 | 0.014 | £15,275.00 |
Oseltamivir | £301.21 | 85.16 | – | – | Dominated by zanamivir |
The reanalysis of the seasonal prophylaxis models presented in Tables 21–24 suggests that the incremental cost-effectiveness of oseltamivir is expected to be around £46,000 per QALY gained for children aged 1–5 compared with best supportive care, and around £116,000 per QALY gained for children aged 1–12 compared with amantadine. As noted above, amantadine is licensed only in children aged over 10 years, hence this comparison can be considered valid only for children aged 11 or 12 years. Oseltamivir is expected to be dominated by zanamivir for otherwise healthy and at-risk individuals aged over 12 years. The Roche models suggest that prophylaxis using amantadine or zanamivir is likely to have a cost-effectiveness ratio below £20,000 per QALY gained in the at-risk population aged 12 years or older.
The Roche submission reported the results of several one-way sensitivity analyses as well as probabilistic sensitivity analysis for each of the pairwise cost-effectiveness comparisons. The one-way sensitivity analysis was undertaken to explore the impact of changing assumptions regarding attack rates, GP visits to receive prophylaxis, health utilities for ILI, bronchitis and pneumonia, preventative efficacy rates and the number of years of life lost. Both the seasonal prophylaxis and post-exposure prophylaxis models were reported to be highly sensitive to changes in assumptions regarding attack rates and the number of GP visits required per household.
In a similar manner to the deterministic health economic analysis, the results of the probabilistic sensitivity analysis were reported using cost-effectiveness planes and cost-effectiveness acceptability curves (CEACs) only for pairwise comparisons of oseltamivir versus amantadine and oseltamivir versus usual care. This is inappropriate as all options should be compared incrementally. A fully incremental reanalysis of uncertainty was not possible due to the structural limitations of the model (the model was capable of comparing only two prophylaxis options simultaneously). In addition, the submission states that pairwise comparisons were not undertaken for oseltamivir versus zanamivir due to the assumption of equivalence between these products; this is inappropriate as there is clearly uncertainty surrounding the relative efficacies of these drugs. Consequently, the correct interpretation of the probabilistic sensitivity analysis is problematic.
Tables 25 and 26 show the probabilities that oseltamivir has a cost-effectiveness ratio that is better than £20,000 and £30,000 per QALY gained compared with the next best comparator identified in the incremental reanalysis of the deterministic cost-effectiveness analysis submitted by Roche. These tables have been constructed by the Assessment Group from the simulation outputs used to generate the CEACs within the Roche submission.
Population | Comparison (non-dominated) |
Probability cost-effective at £20,000 per QALY gained |
Probability cost-effective at £30,000 per QALY gained |
---|---|---|---|
Children aged 1–5 years | Usual care | 0.91 | 0.97 |
Children aged 1–12 years | Usual care | 0.94 | 0.99 |
Otherwise healthy individuals aged > 12 years | Usual care | 0.18 | 0.65 |
At-risk individuals aged > 12 years | Usual care | 1.00 | 1.00 |
Population | Comparison (non-dominated) |
Probability cost-effective at £20,000 per QALY gained |
Probability cost-effective at £30,000 per QALY gained |
---|---|---|---|
Children aged 1–5 years | Usual care | 0.07 | 0.2 |
Children aged 1–12 years | Amantadine | 0.01 | 0.04 |
Otherwise healthy individuals aged > 12 years | Dominated by zanamivir in the deterministic analysis | NA | NA |
At-risk individuals aged > 12 years | Dominated by zanamivir in the deterministic analysis | NA | NA |
Tables 25 and 26 suggest that the probability that post-exposure prophylaxis using oseltamivir is optimal at thresholds of £20,000 is in excess of 0.90 in the paediatric and at-risk populations (i.e. there is a high probability that oseltamivir produces more net benefit than its relevant comparators at a threshold of £20,000 per QALY). The probability that oseltamivir post-exposure prophylaxis has a cost per QALY ratio below £20,000 is around 0.18 for healthy adults; the probability that oseltamivir post-exposure prophylaxis has a cost per QALY ratio below £30,000 is around 0.65 in the healthy adult group. In the seasonal prophylaxis setting, oseltamivir is unlikely to be cost-effective at £30,000 per QALY gained in children aged 1–5 and 1–12 years. Within its adult indications, oseltamivir was dominated by zanamivir within the deterministic analysis; given the assumption of equivalent efficacy between oseltamivir and zanamivir, one would expect zanamivir to be optimal irrespective of the assumed willingness-to-pay threshold.
Sander et al. – Post-exposure influenza prophylaxis with oseltamivir: cost-effectiveness and cost–utility in families in the UK
Sander et al. 91 present the methods and results of a cost-effectiveness and cost–utility analysis of oseltamivir as post-exposure prophylaxis from the perspective of the NHS (health-care payer perspective). The model simulates the experience of 100,000 hypothetical family members aged ≥ 13 who receive oseltamivir prophylaxis or no prophylaxis (with or without treatment for symptomatic ILI). The cost-effectiveness and cost–utility of oseltamivir prophylaxis is estimated by means of comparison with two alternatives: (1) no prophylaxis and no treatment and (2) no prophylaxis followed by treatment of ILI using oseltamivir. The model does not include options for sequential prophylaxis and treatment using antivirals, nor does it include other licensed prophylactic options such as amantadine or zanamivir. The health economic outcomes used within the analysis were the incremental cost per ILI case avoided and the incremental cost per QALY gained. The analysis uses a time horizon of a single influenza season; the cost–utility analysis also includes adjustments for QALYs lost as a result of premature death due to secondary complications of influenza.
The model uses a decision tree modelling approach, evaluated using Monte Carlo simulation methods to evaluate first-order uncertainty surrounding costs and health outcomes for each option. The decision tree model includes chance nodes describing the uncertainty surrounding the probability of ILI infection, the treatment of ILI (oseltamivir or no antiviral treatment), the onset of complications due to ILI or influenza and subsequent outpatient treatment, inpatient treatment and eventual death. The model does not include the impact of herd immunity upon clinical effectiveness or cost-effectiveness outcomes. The model includes two types of influenza-related complications: pneumonia and bronchitis. These are reported to have been included in the model because of their high incidence within the model population and their definite association with influenza, and because oseltamivir reduces the risk of these complications and other hospitalisation. 91 The model assumes that patients cannot develop more than one complication attributable to ILI.
The base-case ILI attack rate in contact cases was assumed to be 8%, based on clinical trials of oseltamivir prophylaxis within households. 48,49 The GP diagnostic certainty rate (i.e. sensitivity) was assumed to be 70%; however, a reference is not provided for the source of this assumption. The rate of true influenza infection in index cases was taken from clinical trials of oseltamivir as prophylaxis. 48,49 The model assumes that oseltamivir reduces the number of cases when used prophylactically and the duration of disease when used as treatment. The model also assumes that while prophylaxis may reduce the probability of experiencing ILI, and hence the probability of secondary complications, it does not affect the clinical course of complications once they manifest. The probability of avoiding clinically-proven influenza using post-exposure prophylaxis with oseltamivir was assumed to be 89%, based on a clinical trial reported by Welliver et al. 49 This estimate of efficacy is noticeably higher than the efficacy rates demonstrated in the trial reported by Hayden et al. 48 (62%), which are not used in the base-case health economic analysis.
The model includes HRQoL impacts associated with the incidence of ILI, bronchitis, pneumonia and QALY losses due to premature death. The approach to valuing the number of QALYs lost because of premature death from secondary influenza complications is similar to that reported by Turner et al. ,10 but certain underlying assumptions differ between the models. Patient HRQoL was measured within the clinical trials used to inform the health economic model using Likert visual analogue scales for health, sleep and usual activities (based on studies WV15670, WV15671, WV 15730 and M76001). Visual analogue scale scores were transformed into TTO index utilities using an algorithm based on econometric work undertaken by researchers at the University of York. 104 Time with complications was multiplied by their respective utility scores to estimate QALY losses. Life-years lost due to premature death were calculated using UK life tables, based on an assumed age at death. The analysis assumes that premature death due to complications was associated with a loss of 34.24 life-years, each of which is valued at a state equivalent to perfect health (one life-year lost is assumed to equal one QALY lost). As noted above, this assumption is also applied in the Roche submission to NICE. 20 This assumption is highly optimistic, and favours the oseltamivir prophylaxis option as this has the greatest efficacy in terms of avoiding influenza and related complications. The impact of this assumption on the cost-effectiveness of oseltamivir prophylaxis is not addressed within the sensitivity analysis. The majority of events occurred within 1 year and were not subjected to discounting, which is appropriate. The loss of QALYs due to premature death was discounted at a rate of 1.5% per year.
The cost impact of oseltamivir-related adverse events is not included in the model; the authors state that the adverse events observed in clinical trials of oseltamivir were ‘generally mild, self-limiting and did not result in health-care service utilisation’. 91 The impact of adverse events of treatment using oseltamivir, however, is included in the QALY estimate, which serves to reduce the number of QALYs gained for the oseltamivir treatment group. Resource use data relating to the prevention and treatment of influenza was derived from the NAMCS. 101 This resource use relates to estimates for drug prescriptions, diagnostic tests and investigations for ILI, bronchitis and pneumonia, and primary and secondary care admissions for patients with influenza and selected complications. Other resource use items included the cost of oseltamivir, GP visits, specialist visits, antibacterials for the treatment of ILI-related complications, bronchitis, pneumonia, over-the-counter medications and hospitalisation. The use of these resource use data may be problematic, as US treatment patterns for ILI and secondary complications may not reflect those in the UK.
A number of sensitivity analyses were undertaken alongside the underlying probabilistic analysis. These included varying the ILI attack rate for contact cases, varying assumptions regarding health-care resource utilisation and assumptions regarding the diagnostic accuracy of GPs in identifying influenza, as well as undertaking the analysis from the societal perspective. The sensitivity analysis also considers the impact of a lower efficacy rate of 60%, which reflects the results of the oseltamivir post-exposure prophylaxis clinical trial reported by Hayden et al. 48 The simulation model uses Monte Carlo sampling to handle both first- and second-order uncertainty surrounding costs and health outcomes.
Under the base-case assumptions, the model estimates the incremental cost-effectiveness of oseltamivir post-exposure prophylaxis versus no prophylaxis to be £467 per ILI case avoided, while the incremental cost–utility is estimated to be £29,938 per QALY gained. The incremental cost-effectiveness and cost–utility of oseltamivir prophylaxis versus no prophylaxis followed by oseltamivir treatment were estimated to be £451 per ILI case avoided and £52,202 per QALY gained. The results of the uncertainty analysis suggested that reduced prophylactic effectiveness for oseltamivir results in considerably less favourable estimates of cost-effectiveness and cost–utility. Assumptions concerning higher attack rates and reduced GP utilisation resulted in marked improvements in the cost-effectiveness and cost–utility of oseltamivir. When the economic analysis was undertaken from the societal perspective, oseltamivir was reported to dominate the no prophylaxis options. The probabilistic sensitivity analysis suggests that the probability that post-exposure prophylaxis using oseltamivir has a cost-effectiveness of better than £30,000 is 50% compared with no prophylaxis and 10% compared with oseltamivir treatment.
Risebrough et al. – Economic evaluation of oseltamivir phosphate for post-exposure prophylaxis of influenza in long-term care facilities
Risebrough et al. 92 report the methods and results of a decision-analytic model to evaluate the cost-effectiveness of post-exposure prophylaxis versus no prophylaxis in long-term care facilities. The model includes three treatment options: post-exposure prophylaxis using oseltamivir, post-exposure prophylaxis using amantadine and no prophylaxis. The analysis was undertaken from the perspective of the single government payer in Canada. Zanamivir was excluded from the analysis because of difficulties in drug administration experienced by elderly patients. The primary health economic outcome for the analysis was reported to be the incremental cost per ILI case avoided compared with usual care (no prophylaxis); however, the model results are presented only in terms of costs and consequences which are not synthesised to produce incremental cost-effectiveness ratios. All patients are assumed to have received prior vaccination for influenza. The model uses a time horizon of 30 days, which is intended to represent the approximate duration of one institutional outbreak.
The model uses a decision tree structure to evaluate the costs and health outcomes associated with each of the three options. The first chance node relates to whether an outbreak occurs within the given care facility. Following an outbreak, patients in the prophylaxis arms begin post-exposure prophylaxis for 12 days using either amantadine or oseltamivir. For patients receiving amantadine, the model includes the possibility of developing amantadine resistance, while adverse events may be experienced by individuals receiving either prophylactic option. The model then includes the possibility that the individual develops ILI from which they may experience a complication, recover without complication, or die. If the ILI case is complicated, the patient may be treated in the care facility or, alternatively, may be transferred to hospital. The model does not include the expected effects of herd immunity. The model does not differentiate between specific complications experienced by individuals developing ILI. The incidence of ILI complications has an impact only on the cost side of the model; the impact of ILI and prophylaxis on HRQoL is not included in the economic analysis.
The authors assume an ILI attack rate in vaccinated residents of 17%. This estimate was reported to have been derived from a number of case–control studies and RCTs. The precise statistical methods used to derive this baseline attack rate (e.g. statistical meta-analysis) is unclear. The model does not include the possibility of patients receiving antiviral treatment following the onset of ILI. At the time of the analysis, the authors reported that there were no RCTs evaluating oseltamivir or amantadine as post-exposure prophylaxis in the nursing home setting. 91 Therefore, the authors assumed that post-exposure prophylaxis using oseltamivir would be at least as effective as seasonal prophylaxis using oseltamivir, and that amantadine would be at least as effective as rimantadine. Relative risk reductions in ILI incidence of 60% and 63% were assumed for amantadine and oseltamivir respectively. The authors assumed that prophylaxis using either amantadine or oseltamivir would result in a 50% relative reduction in antibiotic use, serious complications and death; no evidence is provided to support the validity of this assumption. The model includes the possibility of patients withdrawing from therapy as a result of the incidence of adverse events.
The model includes acquisition costs for amantadine and oseltamivir, serum creatinine tests and oral antibiotics, as well as the cost of hospitalisation for the management of influenza or other respiratory infections and the cost of hospitalisation due to adverse events. A cost is included for death resulting from ILI in an acute hospital. Dose adjustments are included in the cost of amantadine. Acquisition costs for amantadine were taken from the Ontario Drug Benefit Formulary, while the cost of oseltamivir was based on the manufacturer’s wholesale price. Serum creatinine test costs were taken from the Ministry of Health Schedule of Benefits. 106 The costs of hospitalisation due to adverse events were based on authors assumptions. The cost of transfer to an acute care facility for treatment of influenza complications was based on the average of all hospitalisations for influenza or other respiratory procedures per case mix group, derived from the Ontario Case Costing Initiative. 106 Higher costs were assigned to those complications that have potentially life-threatening complexity; the same cost was assumed irrespective of the patient’s outcome. Neither costs nor health outcomes were adjusted for time preferences.
The authors undertook one-way sensitivity analysis and best/worst-case scenario analysis, varying cost and event probability parameter values to identify the key determinants of cost-effectiveness. The sensitivity analysis explored the impact of changing assumptions concerning the relative efficacy of amantadine and oseltamivir versus placebo, the cost of serum creatinine testing, the incidence of adverse events, the attack rate for ILI, the outbreak rate and the rate of amantadine resistance. The sensitivity analysis also explored the impact of including the cost of nurse or pharmacist time to review the patient chart and to calculate the creatinine clearance. Finally, the cost-effectiveness of rimantadine was also explored in the sensitivity analysis. Probabilistic sensitivity analysis was not undertaken within this study.
In the base-case analysis, the study suggests that post-exposure prophylaxis using oseltamivir or amantadine is expected to reduce the incidence of ILI cases, hospitalisation and death compared with no prophylaxis. Both options are also expected to produce cost-savings as compared against no prophylaxis. When compared in terms of the incremental cost per ILI case avoided, oseltamivir is expected to dominate both amantadine and no prophylaxis. The sensitivity analysis suggests that the analysis is sensitive to the amantadine dose calculation. The use of alternative assumptions concerning the attack rate for ILI, the outbreak rate and the rate of amantadine resistance did not affect the base-case conclusions. The sensitivity analysis also suggested that if rimantadine were available in Canada, at 32% of the cost of oseltamivir, it would be the least expensive option; however, the authors suggest that oseltamivir would remain the most effective option. The worst-case scenario for amantadine resulted in improvements in ILI cases avoided, albeit at a greater cost than no prophylaxis. In the worst-case scenario, oseltamivir remained more effective and less costly compared with the amantadine and no prophylaxis options.
Turner et al. – Systematic review and economic decision modelling for the prevention of influenza A and B
Turner et al. 10 report the methods and results of a mathematical decision model to evaluate the cost-effectiveness of amantadine, zanamivir and oseltamivir in the prevention and treatment of influenza A and B. This study formed the assessment report used to inform the 2003 NICE appraisal of oseltamivir and amantadine for the prevention of influenza. 16 The analysis was undertaken from the perspective of the NHS, although reduced time from work is considered within the sensitivity analysis. The model includes eight preventative options: (1) no prophylaxis, (2) vaccination, (3) amantadine prophylaxis, (4) zanamivir prophylaxis, (5) oseltamivir prophylaxis, (6) vaccination plus amantadine prophylaxis, (7) vaccination plus zanamivir prophylaxis and (8) vaccination plus oseltamivir prophylaxis. All antiviral strategies relate to seasonal prophylaxis over a period of 6 weeks (42 days). Post-exposure prophylaxis using amantadine, oseltamivir and zanamivir are not included in the economic model; the model has since been adapted to examine the cost-effectiveness of post-exposure prophylaxis; however, the results of this work have not been released into the public domain. 107 The assessment report also evaluated the cost-effectiveness of treatment options for influenza A and B; however, these options are considered separately from the antiviral prophylaxis options. Cost-effectiveness is expressed in terms of the incremental cost per QALY gained and the incremental cost per influenza illness day avoided. The model uses a time horizon of a single influenza season, and includes QALY losses resulting from premature death due to influenza. The model estimates the cost-effectiveness of prophylaxis in four discrete subgroups: healthy adults, high-risk adults, children and residential care elderly.
The model uses a decision tree approach to evaluate the costs and health outcomes for each prophylactic option. Chance nodes are used to describe the probability of a patient developing influenza (dependent on the prophylaxis option), and QALY losses and costs are assigned to each branch. Costs and benefits for patients with influenza are modified for strategies including vaccination, on the basis that vaccination may reduce the severity of secondary complications. The model includes two complications: pneumonia and otitis media (the latter is included only in the paediatric model).
The model operates on the basis of true influenza rather than ILI. As treatments for influenza are evaluated separately from prophylaxis and vaccination options, the exclusion of ILI may be reasonable because costs and benefits in patients with ILI which is not true influenza are not expected to differ between prophylaxis options (and would therefore cancel each other out in the cost-effectiveness calculations). Baseline attack rates for true influenza were estimated using random-effects meta-analyses of placebo arm outcomes from relevant trials included in the systematic review. The preventative efficacy of each prophylaxis option was estimated by calculating the odds ratio of developing influenza, adjusted for the probability of compliance. The protective benefit of the prophylaxis options was assumed to apply only to the period over which patients are taking prophylaxis. The benefit of prophylaxis in vaccinated patients was assumed to be cumulative, such that the relative benefit of prophylaxis was applied to the baseline influenza attack rate excluding the expected number of cases protected by prior vaccination. The probability that an individual presents to the GP with influenza was based on a UK study of excess ILI consultations over a 10-year period reported by the Royal College of General Practitioners (RCGP)5 and the baseline influenza attack rate derived from the meta-analysis. 10 The probability of presentation was estimated by dividing the number of excess ARI consultations by the expected number of individuals who are expected to develop influenza in each population group. As the number of patients who present with true influenza is unknown, the numerator for this calculation was based on excess ARI consultations, assuming that all excess consultations are due to influenza. This approach, therefore, implies that the rate of non-influenza ILI consultations is constant over the year, and is likely to represent the maximum theoretical impact of influenza over a season. 5
The model includes HRQoL impacts associated with the incidence of influenza, adverse events resulting from the use of amantadine, the incidence of pneumonia and otitis media, and a QALY loss resulting from premature death due to complications. QALY losses due to influenza were derived from VAS scores collected in trials of oseltamivir for the treatment of influenza (studies WV15670, WV15671, WV15730, WV15819, WV15876, WV15978, WV15812 and WV15872). QALYs were derived by recalibrating Likert score data to VAS scores which were then converted into TTO scores. 104 QALY losses due to premature death were estimated on the basis of mean age of death due to influenza within the model subgroup, remaining life expectancy, age-specific utility scores and the discount rate. QALYs lost due to premature death were discounted at a rate of 1.5% in the base-case analysis in line with recommendations from NICE at the time of the assessment. The valuation of serious adverse events due to amantadine was based on an assumed EuroQol-5D (EQ-5D) profile. Adverse events resulting from the use of oseltamivir and zanamivir were assumed to have no impact on HRQoL. The valuation of secondary complications of influenza (pneumonia and otitis media) was based on WHO disability weights for lower respiratory conditions. 109
The model includes the costs associated with GP visits, prophylaxis and vaccination acquisition and inpatient hospital stays. The cost of a GP consultation in the surgery or at home was derived from the PSSRU; this cost was weighted by the frequency of home and surgery visits to generate a mean cost per visit for the elderly population and for the healthy adult population. The mean cost of a GP visit for the paediatric model was assumed to be the same as for the healthy adult model. The cost of antiviral prophylaxis was based on a 6-week course, assuming 50% of the recommended dose. Each drug cost was inflated to account for container fees and pharmacy prescribing fees, although these cost adjustments do not form part of NICE’s methods guidance. 96 The cost of vaccination was taken from payments to GPs for vaccination and included an administration cost. Hospitalisation costs were based on Health-care Resource Groups (HRGs); the HRGs assumed for hospitalisation differed according to the population under consideration. Owing to the short time horizon for the analysis, costs were not subjected to discounting.
Simple uncertainty analysis was undertaken using one-way and two-way sensitivity analyses surrounding the base-case model specification. This included varying assumptions in relation to influenza attack rates, the probability of death and the value of QALY losses due to premature death resulting from influenza complications. Joint uncertainty in model parameters was evaluated using probabilistic sensitivity analysis; parameter uncertainty was propagated through the model using Monte Carlo sampling techniques. However, results are presented as CIs surrounding the cost-effectiveness ratio; CEACs for prophylaxis are not presented in the report.
In the base-case analysis, amantadine, oseltamivir and zanamivir were dominated by vaccination. The combined option of amantadine plus vaccination yielded an incremental cost per QALY gained of £28,920 compared with vaccination alone in the residential care population. The incremental cost-effectiveness ratio (ICER) of amantadine for all other populations was considerably higher, ranging from £124,854 to £909,210. When adverse events were excluded from the model, the results of the probabilistic sensitivity analysis suggested that the probability that amantadine resulted in an incremental cost per QALY gained below £30,000 was around 45% for the elderly residential care population. However, this is a conservative assumption which favours amantadine. For the other populations, the probability that amantadine has an incremental cost per QALY gained below £30,000 was less than 1%. For the combined option of oseltamivir plus vaccination, the incremental cost per QALY gained for the residential population was £64,841 compared with vaccination alone. For all of the remaining populations, the ICERs were markedly less favourable, ranging from £251,004 to £1,693,168. The probabilistic sensitivity analysis suggested that the probability that oseltamivir has an incremental cost per QALY gained that is below £30,000 was 3% or less for all populations. Zanamivir was also dominated by vaccination. For the combined option of zanamivir plus vaccination, the incremental cost per QALY gained for the residential population was £84,682 compared with vaccination alone. The incremental cost per QALY gained ranged from £324,414 to £2,188,039 for the remaining populations. The uncertainty analysis suggested that the probability that zanamivir has an ICER that is below £30,000 per QALY gained was less than 1%.
Scuffham and West – Economic evaluation of strategies for the control and management of influenza in Europe
Scuffham and West93 report the use of a decision model to estimate the ICER of six influenza control strategies compared with no intervention in elderly populations in England, France and Germany. The options included in the model are opportunistic vaccination, comprehensive vaccination, chemoprophylaxis using oseltamivir, chemoprophylaxis using rimantadine, treatment using oseltamivir and treatment using rimantadine. The costs and health effects of zanamivir and amantadine were not included in the model. The analysis was undertaken from the perspective of the health-care financier for each country. The analysis reports marginal health economic outcomes in terms of the cost per hospitalisation averted, cost per death averted, cost per life-year gained and cost per morbidity day averted. The time horizon used within the model was a typical (average) influenza season.
The modelling approach adopted by the authors was not explicitly stated; however, the text indicates that a decision tree modelling methodology was employed. The model estimates the proportion of patients who develop clinical symptoms of ILI, a percentage of whom will visit their GP for treatment and may receive symptomatic treatment or antibiotics for complications of ILI. The model includes the possibility that patients who develop complications may require hospitalisation and the possibility that complications may lead to premature death. The model does not include any herd immunity effects associated with vaccination or prophylaxis.
The model includes the cost of hospitalisation due to complications including influenza and pneumonia, other ARI and congestive heart failure. The model does not include any valuation of the impact of influenza complications upon HRQoL, hence complications appear to be included in the model only in terms of costs avoided. The number of premature deaths due to influenza by age group was taken from a study by Fleming et al. 5 Based on UK hospitalisation data, the authors estimated the years of potential life lost for the healthy 80-year-old population to be 7 years; owing to the likely presence of co-morbidities, the authors assumed that premature death due to influenza would result in a mean loss of 3.5 potential years of life. The authors did not discount costs as almost all relevant events occur within a single influenza season. The potential life-years lost due to premature death resulting from secondary influenza complications was discounted at a rate of 1.5%.
The authors assumed an attack rate for ILI of 10%. This estimate was sourced from excess GP consultation rates, current rates of vaccination and expert opinion. Excess GP consultation rates were taken from a study based on national data collected by the Weekly Returns Service (WRS) of the RCGP and from national data for hospital admissions and deaths. 110 These are modelled independently of ILI attack rates. The probability of after-hours GP consultations was derived from expert opinion, while the percentage of GP home visits was taken from the UK population-based study of incidence, risk factors, complications and drug treatment of influenza reported by Meier et al. 12 The efficacy of chemoprophylaxis was taken from a review reported by Demicheli et al. 94 Based on this review, the authors assumed that neuraminidase inhibitors (NIs), specifically oseltamivir, reduce the incidence of influenza by 55%, while ion-channel inhibitors, specifically rimantadine, reduce the incidence of influenza by 35%. The authors assumed that when taken as prophylaxis, these therapies result in the same proportional reductions as vaccination in terms of GP consultation, hospitalisation and death. The model does not appear to include parameters describing the probability that a patient with ILI has true influenza. However, the estimates of the clinical efficacy of prophylaxis relate specifically to laboratory-confirmed influenza, not ILI. This appears to represent an inconsistency in the parameterisation of the model.
The model includes a number of different resource use items including GP consultations, after-hours visits and home visits, antibiotics, hospitalisations due to influenza and pneumonia, other respiratory illness and congestive heart failure, vaccination acquisition and administration costs, and antiviral prophylaxis and treatment. Unit costs were derived from the PSSRU,111 national sources of hospitalisation data,112 Department of Health publications on prescription costs113 and national tariff estimates. 114 The authors assumed that prophylaxis and treatment did not result in any adverse events. Non-compliance with prophylaxis was included in the model at a weekly rate of 5%.
The authors report the results of a large number of simple sensitivity analyses relevant to each option for the prevention and/or treatment of influenza. This included varying assumptions concerning the years of potential life lost resulting from premature death due to influenza complications, the discount rate for health outcomes, ILI attack rates, excess GP consultations, the number of excess hospital admissions for influenza complications and the number of premature deaths due to ILI complications. Specifically with regard to the prophylaxis options, the sensitivity analysis included varying assumptions regarding GP consultations to receive chemoprophylaxis, compliance rates, the dosage of oseltamivir, the percentage of prophylaxis used during the 4-week peak of the influenza season and drug price. Despite the extensive use of simple sensitivity analysis, the authors did not undertake probabilistic sensitivity analysis, and the impact of joint uncertainty in model parameters is not captured within the analysis.
Under the base-case assumptions, the authors report the marginal cost per life-year gained for oseltamivir to be €197,919 compared with no intervention. The cost per hospitalisation averted for oseltamivir is reported to be €114,774, while the cost per death averted is reported to be €657,544. The cost per morbidity day averted, excluding and including deaths, is reported to be €1198 and €373 respectively. The results of the sensitivity analysis are reported only in terms of the benefit : cost ratio (ratio of the strategy costs minus the costs of hospitalisation averted) and the cost per morbidity day averted. The findings of the sensitivity analysis based on the latter outcome measure are particularly difficult to interpret in a policy context. The analysis is reported to be most sensitive to changes in the timing of the programme, the price and dose of the prophylactic, and the assumed loss in potential life-years due to premature death.
Demicheli et al. – Prevention and early treatment of influenza in healthy adults
Demicheli and colleagues93 report the use of a model to estimate the cost-effectiveness and cost–utility of influenza prevention in healthy adults from the perspective of the Ministry of Defence (MOD). The health economic analysis was undertaken alongside three ongoing Cochrane reviews; the results of these reviews led to marked changes in the scope of the proposed economic analysis and the final economic models presented in the paper. 94 The authors state that potential preventative options to be evaluated within the final model were vaccination, oral amantadine, oral rimantadine and oral oseltamivir. However, costs and health outcomes are presented for three preventative options: vaccination, amantadine prophylaxis and a third option denoted ‘NI prophylaxis’. Although the authors justify the exclusion of zanamivir from the analysis because of trials apparently including only laboratory-confirmed outcomes, the exclusion of rimantadine is not justified within the paper, and the NI option is not directly specified as representing oseltamivir. The primary health economic outcome for the analysis was the incremental cost per avoided case. The time horizon used within the analysis was not explicitly reported; however, the analysis appears to relate to a single influenza season (i.e. a 1-year time horizon).
The authors adopted a decision tree approach to evaluate the differences in benefits and costs of the alternative options for the prevention of influenza. The authors report that they simplified an initially complicated decision tree model structure to include only the possibility of developing influenza and the possibility of experiencing adverse events due to prophylaxis. The model does not include the costs and health impacts of complications due to influenza or ILI and, as a consequence, the model does not include the possibility of death. It is reasonable to argue that the specification of this model is poor, as the results of the analysis ignore key costs and benefits associated with influenza prevention.
The model appears to operate in terms of true influenza cases rather than ILI cases, although this is not entirely clear. Influenza attack rates were derived from influenza sickness rates for 1997 obtained from the Defence Analytical Services Agency (DASA). The model assumes an incidence rate for influenza of 5.7 per 1000; while this value appears very low, incidence rates of up to 400 per 1000 were explored within the sensitivity analysis. The model does not include the possibility of a patient with symptomatic influenza presenting to a health-care professional for treatment. The effectiveness of the amantadine, NIs and vaccination were obtained from three Cochrane reviews of the clinical effectiveness of vaccination and prevention of influenza.
The model includes acquisition costs associated with influenza prevention, which were derived from the Defence Medical Supply Agency and authors’ assumptions. 94 No other cost components appear to be included in the results of the model. The impact of administration costs on overall cost-effectiveness is explored within the sensitivity analysis. A formal price year is not reported. The authors do not mention the use of discounting, which appears to be appropriate given the restrictive scope of the model (i.e. the exclusion of complications and death).
The authors undertook simple sensitivity analysis exploring the impact of improved/worsened preventative efficacy of vaccination and prophylaxis, improved adverse event profiles for vaccination and antiviral prophylactics, duration of prophylaxis and the inclusion of administration costs for prevention. Probabilistic sensitivity analysis was not undertaken in this study.
Costs and health outcomes are not reported in a disaggregated form, and it is difficult to establish whether the results are true incremental comparisons between the options, or whether they are compared marginally against a policy of no prevention. The text appears to indicate the latter to be the case. Under the base-case assumptions, the marginal cost per case avoided for vaccination, amantadine and NI (presumably oseltamivir) are reported to be £2807, £9458, and £88,193 respectively. The uncertainty analysis suggests that under most conditions vaccination is likely to be the most cost-effective option. The key determinant of cost-effectiveness appears to be the influenza incidence rate, for which higher rates are expected to result in more favourable cost-effectiveness ratios for vaccination and prophylaxis. The robustness and reliability of the results of this analysis are severely restricted by the limited scope of the model and the limited reporting of the economic evaluation.
Patriarca et al. – Prevention and control of type A influenza infections in nursing homes
This study95 reports the methods and results of a model of the cost-effectiveness of options for the prevention of influenza A in the elderly nursing home population. The model includes four options for the prevention of influenza A: vaccination without chemoprophylaxis, vaccination with amantadine post-exposure prophylaxis following an outbreak of influenza (30 days’ duration), amantadine post-exposure prophylaxis following an outbreak of influenza (30 days’ duration) with no prior vaccination, and amantadine as seasonal prophylaxis (3 months’ duration) with no prior vaccination. All options are compared with a strategy of no control. Cost-effectiveness is expressed in terms of the incremental cost per illness averted, the incremental cost per hospitalisation averted and the incremental cost per death averted. The perspective of the analysis is not explicitly reported; however, the authors state that only direct costs were included in the analysis. The time horizon for the analysis is unclear; however, the authors state that they did not include future medical costs associated with deaths averted.
The authors used a decision tree model to evaluate the incremental costs and health outcomes for each preventative option. Chance nodes are used to describe the probability that an individual is immune or susceptible to influenza A, the probability of community exposure, the efficacy of vaccination, the possibility of a nursing home outbreak and the possibility that an individual will or will not become ill. Patients who become ill experience one of four possible outcomes: infection and survive, infection and die, hospitalisation and survive or hospitalisation and die. The model is reported to include the impact of herd immunity although the precise methods for including this factor are unclear. Respiratory complications only are included in the model.
The incidence of disease during the course of an outbreak was based on the experience of 41 separate vaccine efficacy studies conducted in nursing homes during the period 1972–85. The probability of an outbreak was estimated according to the results of a case–control study;115 this probability was adjusted for the vaccination and chemoprophylaxis options to account for herd immunity effects. The model assumes an overall attack rate of 43% during influenza outbreaks and 16% at other times. The model does not include the possibility of antiviral treatment for patients who develop ILI. The authors assumed that 80% of residents who completed the course of chemoprophylaxis would be fully protected. The probability of recovery/death with or without hospitalisation following influenza infection for patients receiving amantadine prophylaxis was assumed to be the same as for vaccination. More favourable outcomes were assumed for patients who received both vaccination and prophylaxis, although this was reported to be based on only limited clinical evidence. The impact of adverse events is not included in the effectiveness aspect of the model.
The model includes costs associated with vaccination, acquisition costs for amantadine prophylaxis and costs of diagnostic tests, treatments, ambulance and hospitalisation for influenza infections and associated complications. Administrative costs were excluded from the analysis for the chemoprophylaxis options, but were included for vaccination. The authors state that adverse events associated with amantadine are not associated with excess medical care costs; however, the authors did include the costs of treating fractures and soft-tissue injuries resulting from dizziness or postural hypotension for patients receiving amantadine. Costs of influenza infections and associated complications were sourced from 1986 prospective payment schedules for appropriate diagnosis-related groups and other sources. Physician charges were based on Medicare Part B payments. The authors do not make any reference to the use of discounting within the analysis.
One-way and multiway sensitivity analyses were undertaken surrounding the efficacy of influenza vaccination, the efficacy of chemoprophylaxis, and assumptions concerning risk reductions in hospitalisation and death for patients receiving prophylaxis. The authors also undertook a threshold analysis to determine how much amantadine and vaccination would have to cost before these options would no longer result in savings in direct medical costs. Finally, the authors explored the impact on cost-effectiveness of changing the exposure rate to influenza viruses. Probabilistic sensitivity analysis was not undertaken.
The option of outbreak prophylaxis was excluded from the analysis as it was the least effective and most expensive programme. 95 Marginal cost-effectiveness ratios are presented for vaccination plus chemoprophylaxis versus vaccination alone, continuous chemoprophylaxis versus vaccination alone, and continuous chemoprophylaxis versus vaccination plus chemoprophylaxis. The combination of vaccination and chemoprophylaxis during an outbreak was reported to result in demonstrable improvements in outcome at a modest increase in cost. However, the cost-effectiveness calculations include only the program costs, and do not account for expected cost savings in medical care costs. This omission biases against more effective prevention options. The authors report that changing assumptions regarding efficacy and the risk of hospitalisation and death exerted only minor or negligible effects on the clinical and economic outputs of the model. The authors report that varying exposure to influenza led to a proportionate reduction in the number of cases and a subsequent reduction in the cost-effectiveness of each programme. Increasing the level of coverage of vaccination and chemoprophylaxis led to a progressive decline in morbidity and increases in cost-effectiveness.
Summary of existing economic evaluations of amantadine, oseltamivir and zanamivir for the prophylaxis of influenza
The economic models included in this systematic review cover a broad range of prophylaxis options and settings including seasonal, post-exposure and outbreak control prophylaxis using amantadine, oseltamivir and zanamivir. The relevant populations examined within the economic analyses include children, elderly, at-risk adults and healthy adults with or without prior vaccination. However, the majority of studies included in the review do not include all relevant prophylaxis options for the prevention of influenza (i.e. amantadine, oseltamivir and zanamivir). The Roche submission20 and the study reported by Turner et al. 10 adopted the broadest scope in terms of prophylaxis options and populations. Included studies consistently adopted a short time horizon (a typical influenza season); however, most also accounted for long-term survival or quality-adjusted survival losses resulting from death due to secondary complications of influenza. Only three studies10,20,91 presented health economic results in terms of the incremental cost per QALY gained.
The majority of the models included in the review appear to operate on the basis of ILI rather than true influenza alone. However, one study93 appears inappropriately to apply relative reductions of true laboratory-confirmed influenza to the baseline ILI attack rate. The models include a range of secondary complications affecting costs and consequences; these include pneumonia, bronchitis, other ARI and congestive heart failure in adult populations and otitis media in children. One study did not specify which complications were included in the economic model,92 yet costs and consequences of managing these complications were included in the economic analysis. One study did not include the costs and health consequences resulting from secondary complications, nor did it include the possibility of premature death due to influenza. 94
The review highlights a paucity of good quality evidence relating to many aspects of the decision problem. In particular, many of the models are underpinned by assumptions concerning fundamental parameters such as the underlying ILI or influenza attack rate, the probability that an individual with influenza presents to his or her GP and assumptions regarding the treatment of secondary influenza-related complications, each of which has the propensity to considerably influence the resulting cost-effectiveness estimates. A key problem concerns the absence of robust estimates of the effectiveness of prophylaxis in the specific population under consideration, and the need to make assumptions of equivalence for prophylaxis across different population subgroups. In instances where the impact of influenza on HRQoL has been incorporated into the analysis, this has been drawn consistently from Likert scale data, from clinical trials of oseltamivir which are then mapped onto health utilities or from indirect utility estimates. None of these data are ideal. The limitations of the existing economic models included in the review studies are summarised in Box 1.
Independent economic assessment
Cost-effectiveness modelling methods
This section details the methods employed in the development of the independent Assessment Group model to assess the cost-effectiveness and cost–utility of influenza prophylaxis using amantadine, oseltamivir and zanamivir. The model structure and many of the parameter values draw upon the modelling work undertaken by Turner et al. 10 in the previous assessment of oseltamivir, amantadine and zanamivir for the seasonal prophylaxis of influenza. Key differences between these models include the incorporation of NICE guidance on the use of NIs for the treatment of symptomatic influenza-like illness,116 the inclusion of post-exposure prophylaxis options, an updated systematic review of the effectiveness of influenza prophylaxis (see Chapter 3) and updated estimates of cost and health outcomes associated with influenza and other ILI-related complications.
-
Failure to include all relevant prophylaxis options in the evaluation
-
Failure to model secondary complications and death
-
Failure to account for the impact of disease and prevention on health-related quality of life
-
Use of unrealistically favourable assumptions regarding the value of avoiding death due to secondary complications (i.e. one life-year lost is equal to one QALY lost)
-
Application of laboratory-confirmed influenza preventative efficacy estimates to reduction in ILI baseline attack rate
-
Failure to incorporate all relevant cost components into cost-effectiveness estimates
-
Use of US resource use data which may not reflect UK treatment patterns for the management of secondary complications of influenza
-
Failure to undertake incremental cost-effectiveness analysis (including uncertainty analysis)
-
Failure to account for joint uncertainty in model parameters using probabilistic sensitivity analysis.
Model scope
Interventions and comparators
The model evaluates the incremental costs and health outcomes of seasonal prophylaxis and post-exposure prophylaxis of influenza using amantadine, oseltamivir and zanamivir in comparison with each other and no prophylaxis.
Model population
Cost-effectiveness estimates for influenza prophylaxis using oseltamivir, amantadine and zanamivir are presented for six discrete subgroups: children aged 1–14 years (with at-risk medical condition or otherwise healthy), adults aged 15–64 years (with at-risk medical condition or otherwise healthy) and elderly adults aged over 65 years (with at-risk medical condition or otherwise healthy). In addition, the analysis considers the impact of prophylaxis for individuals who have been vaccinated against influenza and for individuals who have not been previously vaccinated. Although the model structure is identical for all subgroups, the analyses differ in terms of influenza attack rates, prophylaxis dose, prophylactic efficacy and prognosis following influenza onset.
Health economic outcomes
The primary health economic outcome used in the economic model is the incremental cost per QALY gained. This is calculated for all non-dominated prophylactic options compared with the next most effective option. Options that are dominated (simple or extended) are ruled out of the analysis.
Time horizon and time preferences
The model assumes that all events of interest occur within a single influenza season; hence, the time horizon is effectively 1 year in duration. As such, costs and health outcomes arising within this period are not subjected to discounting. However, as secondary complications of influenza and other ILI may result in premature death, the model also accounts for potential years of life lost beyond this time horizon; these are adjusted to account for the expected level of quality of life. Quality adjusted life-years lost because of premature death resulting from the incidence of influenza-related complications are discounted at a rate of 3.5%, in line with current recommendations from NICE. 96 A summary of the scope of the economic comparisons is presented in Table 27 (note that the duration of prophylaxis is assumed to be in line with licensed indications).
Prevention strategy | Duration of prophylaxis (seasonal) | Duration of prophylaxis (post-exposure) | Dosage per day |
---|---|---|---|
Amantadine | 42 days (21 days for patients who have previously been vaccinated) | 10 days | 100 mg |
Oseltamivira | 42 days | 10 days | 75 mg |
Zanamivir | 28 days | 10 days | 10mg |
Model structure
The model uses a decision-analytic (decision tree) approach to estimate the incremental costs and health outcomes associated with each influenza prophylaxis strategy compared with each other and no prophylaxis. The model operates on the basis of ILI which includes true influenza as well as other illnesses that are clinically similar to influenza, e.g. respiratory syncytial virus (RSV). The costs and health outcomes of other ILI are included in the model as these are often indistinguishable from true influenza and may result in additional health-care management costs as well as QALY losses. Furthermore, much of the literature relating to the consequences of influenza infection is actually based on the broader group of ILI including influenza. 12 Costs and health outcomes are estimated for three groups of patients: (1) individuals who develop true influenza; (2) individuals who develop other ILI which is not influenza; and (3) individuals who do not develop influenza or ILI. The prophylactic options evaluated within the model are effective only against the influenza virus, thus effective protection against influenza is assumed to reduce the probability of developing true influenza but will have no impact on other ILI.
A simplified description of the model structure is presented in Figure 5. Patients may receive seasonal or post-exposure prophylaxis using amantadine, oseltamivir or zanamivir, or no prophylaxis. The probability that a contact case will develop influenza is dependent on the influenza attack rate, the prophylactic efficacy of the strategy under consideration over the period in which the patient is taking prophylaxis, the probability that the influenza is influenza A (amantadine only), the degree of resistance to the prophylactic drug (amantadine only), and whether the patient has been previously vaccinated. In terms of post-exposure prophylaxis, the model assumes that the patients are prescribed prophylaxis within 48 hours of exposure to an infected index case, in line with licensed indications. Patients receiving vaccination and/or prophylaxis (amantadine only) may experience adverse events which may detract from the their HRQoL and may incur additional medical treatment costs. If patients do not develop ILI, no further costs or health outcomes are considered for these patients in the model. If a patient does develop influenza or other ILI, he or she may seek medical treatment in either primary care (i.e. GP consultation) or secondary care [i.e. presenting at an accident and emergency (A&E) department]. If the patient presents with symptomatic ILI, he or she may be considered appropriate for treatment using oseltamivir or zanamivir (if the patient presents within 48 hours of developing ILI symptoms and is considered to be at risk of developing secondary complications of influenza). 116
A proportion of patients who develop ILI are expected to develop secondary complications, including respiratory complications such as bronchitis, pneumonia or otitis media, or an exacerbation of an existing underlying condition (including cardiac, renal and CNS complications). 12 If a patient develops an ILI complication, he or she is assumed to seek medical attention for treatment. The model assumes that antibiotics may be prescribed for the treatment of uncomplicated ILI cases as well as for the treatment of ILI-related complications. 12 A proportion of patients who develop complications of ILI are assumed to require hospitalisation. The model assumes that a proportion of complications will result in premature death.
The decision model includes the administration and acquisition costs of influenza vaccination and prophylaxis, the costs of treatment of symptomatic ILI using NIs in at-risk groups, the costs of consultation in primary and secondary care, the costs of managing secondary complications of influenza and ILI and the costs of hospitalisation for individuals with severe complications of ILI. Quality-adjusted life-year losses are included for individuals who develop uncomplicated ILI, adverse events of prophylaxis (amantadine only), complications of ILI and premature death due to ILI complications.
Key model assumptions
-
Other ILI which is not influenza may also result in complications (including RSV and Mycoplasma pneumoniae). It should be noted that the complications arising from influenza may differ in reality from those for other ILI such as RSV (this is a limitation in the use of the data from Meier et al. ;12 see Parameters relating to the onset of influenza and other ILI, below). However, since the costs and effects associated with other ILI are the same for each prophylaxis group, these do not affect the resulting estimates of cost-effectiveness.
-
Prophylaxis using amantadine, oseltamivir and zanamivir are effective only against true influenza.
-
Antiviral prophylaxis is effective in preventing influenza only for the period over which the patient is taking the drug. For seasonal prophylaxis, the model assumes that a patient may be protected over a proportion of the whole influenza season. However, it should be noted that monitoring of influenza activity takes place at a national level and the duration for which activity exceeds the national threshold may not reflect influenza activity at the local level. The importance of this assumption is tested in the sensitivity analysis (see One-way/multiway sensitivity analysis and scenario analysis, p. 90). For the sake of simplicity, the model assumes that the risk of infection is constant for the period when influenza is circulating; this is in line with the previous models, reviewed in Systematic review of existing cost-effectiveness evidence (p. 43).
-
The joint benefit of vaccination followed by prophylaxis is assumed to be cumulative (the effectiveness of prophylaxis is applied to any remaining influenza cases which are not effectively protected by vaccination).
-
The model assumes that amantadine, oseltamivir and zanamivir would be used as prophylaxis when influenza is known to be circulating in the community (the threshold is currently set at 30 new ILI GP consultations per 100,000 population). 8
-
The model assumes that the prescription of seasonal prophylaxis and post-exposure prophylaxis of influenza requires a consultation with a GP. The possibility of multiple courses of antiviral prophylaxis being prescribed to an index case on behalf of other household contacts is explored in the sensitivity analysis. The model assumes that prophylaxis is not given at the same time as influenza vaccination, hence a second visit is required.
-
If an individual develops a secondary complication of ILI (whether or not this is due to influenza), the course of the complication is unaffected by the prior use of prophylaxis. Treatment of symptomatic influenza using oseltamivir or zanamivir is assumed to reduce the incidence of complications in at-risk patients. If a patient has already developed a complication while receiving prophylaxis, it is unlikely that antiviral treatment will provide any additional benefit. Given the simple structure of the model, the analysis assumes that patients who receive antiviral prophylaxis and subsequent treatment for symptomatic ILI develop complications after being prescribed treatment. This assumption is likely to favour prophylaxis as it increases the costs of treating symptomatic influenza. Assumptions surrounding the use of antiviral treatment following prophylaxis are explored in the sensitivity analysis.
-
Patients who experience adverse events due to prophylaxis are likely to consult their GP for advice.
-
Adverse events due to oseltamivir and zanamivir are mild, self-limiting and have no impact on a patient’s HRQoL. Adverse events due to amantadine prophylaxis may be more severe and may result in a reduction in the patient’s quality of life.
-
Antiviral treatment of symptomatic influenza and ILI using zanamivir and oseltamivir is given in line with current NICE recommendations. 116 The choice of NI for the treatment of symptomatic ILI is assumed to be independent of the prophylactic strategy under consideration. Antiviral treatment is assumed to incur an additional cost in patients who have previously received prophylaxis. For example, if a patient is prescribed oseltamivir prophylaxis, subsequently develops symptomatic ILI and is given oseltamivir treatment, a separate prescription of the drug is required.
-
All patients who develop complications due to influenza and other ILI present to a health-care professional for treatment.
-
Patients who develop either uncomplicated or complicated ILI may be prescribed antibiotics.
-
Patients who stop taking prophylaxis are assumed to do so at the beginning of the course and hence do not gain any additional protection over patients who do not receive prophylaxis (the impact of assumptions regarding withdrawal rates are explored in the sensitivity analysis – see One-way/multiway sensitivity analysis and scenario analysis, p. 90).
-
The costs of diagnostic tests (blood tests, sputum tests, chest X-ray) for patients presenting with respiratory complications are assumed to be included in the unit costs of GP consultation and A&E consultation.
-
Owing to limitations in the evidence base, the model assumes that only complicated ILI cases may result in hospitalisation and death. These assumptions are explored in the sensitivity analysis (see One-way/multiway sensitivity analysis and scenario analysis, p. 90).
-
The model includes only those health benefits accrued by patients receiving influenza prophylaxis; potential benefits accrued through decreased transmission of influenza as a result of the use of prophylaxis are not considered in the health economic model.
-
A proportion of influenza cases are assumed to be resistant to amantadine. Although there is some evidence of resistance for the NIs, these rates are low and are excluded from the base-case analysis. The impact of resistance to oseltamivir is considered within the sensitivity analysis (see One-way/multiway sensitivity analysis and scenario analysis, p. 90).
Model parameters
Lists of all model parameters for the seasonal prophylaxis and post-exposure prophylaxis models by subgroup are presented in Appendix 7.
Event probabilities
Baseline influenza attack rate
The baseline influenza attack rate describes the probability that an individual will develop influenza over the influenza season. The model assumes that the probability of developing influenza differs among children, adults and elderly individuals. Different attack rates are also assumed between the seasonal and post-exposure prophylaxis models, as probability of influenza infection is likely to be higher in an individual who has been in frequent close contact with an index case with symptomatic ILI in the household. In terms of seasonal prophylaxis, the clinical trials included in this review do not represent a good basis for estimating the probability of developing influenza as they include different levels of exposure to influenza vaccination across each subgroup; one would expect that this would result in lower attack rates than in the unvaccinated population. For the seasonal prophylaxis model, influenza attack rates were derived from a large meta-analysis of placebo arm groups of clinical trials of influenza vaccination versus no influenza vaccination reported by Turner et al. 10 The model uses the actual patient numbers presented in the summary of each meta-analysis to estimate the mean and distribution of the attack rate. Beta distributions were used to describe the uncertainty surrounding these parameters.
This source does not, however, provide a useful basis for estimating attack rates for the post-exposure prophylaxis models, as individuals eligible for post-exposure prophylaxis have, by definition, been exposed to an index case with symptomatic influenza or ILI. Consequently, one would expect the attack rate for these individuals to be higher than the attack rate in an individual who has not been exposed to an index case. Attack rates for the post-exposure prophylaxis options were sourced from the trials of post-exposure prophylaxis included in the systematic review (see Chapter 3). For the paediatric subgroup the attack rate was taken directly from the subgroup analysis reported by Hayden et al. ,48 as this was the only study which presented a subgroup analysis for the paediatric population. For the working-age adult and elderly populations, the attack rate was taken from a pooled analysis of placebo group attack rates reported in five trials of post-exposure prophylaxis. 46–49,72 It should be noted that patient-level data were not available, hence these attack rates relate to populations that are mixed in terms of subject age. Beta distributions were used to characterise the uncertainty surrounding these attack rates. The attack rates are presented in Table 28.
Type of prophylaxis | Age group | Number of patients with influenza | Number of patients at risk | Attack rate |
---|---|---|---|---|
Seasonal | Children (0–15 years) | 256 | 1469 | 0.174 |
Seasonal | Adults (16–64 years) | 104 | 1670 | 0.062 |
Seasonal | Elderly (65+ years) | 57 | 1098 | 0.052 |
Post-exposure | Children (0–15 years) | 21 | 111 | 0.189 |
Post-exposure | Adults (16–64 years) | 18 | 2051 | 0.088 |
Post-exposure | Elderly (65+ years) | 18 | 2051 | 0.088 |
Probability that an ILI is influenza
The probability of developing ILI during the influenza season was not available from the literature. Instead, the model uses data provided by the RCGP concerning the probability that a case of ILI is true influenza. Within the health economic model, this probability is divided by the true influenza attack rate to provide an estimate of the broader ILI attack rate in each subgroup (accounting for true influenza and other ILIs). Data relating to the probability that ILI is influenza was based on an analysis of swabs taken from individuals with symptomatic ILI collected during routine surveillance over the influenza seasons 2003–4 to 2006–7 (Dr Alex Elliott, RCGP, personal communication). These data relate to those weeks when influenza was known to be circulating in the community, as defined by the 30/100,000 ILI GP consultation threshold;8 they are shown in Table 29.
Season | Week | ILI consultation rate | Number of swabs | Number influenza A | Number influenza B | Total |
---|---|---|---|---|---|---|
2003–4 | 44 | 36.42 | 7 | 1 | – | 1 |
45 | 47.24 | 73 | 35 | – | 35 | |
46 | 61.79 | 120 | 60 | – | 60 | |
47 | 54.69 | 58 | 36 | – | 36 | |
48 | 52.79 | 43 | 20 | – | 20 | |
49 | 57.86 | 78 | 31 | – | 31 | |
50 | 36.96 | 53 | 18 | – | 18 | |
51 | 41.20 | 25 | 9 | – | 9 | |
52 | 33.03 | 23 | 4 | – | 4 | |
2004–5 | 1 | 38.91 | 15 | 5 | – | 5 |
2 | 34.89 | 27 | 13 | – | 13 | |
3 | 33.26 | 16 | 4 | – | 4 | |
4 | 30.45 | 29 | 14 | 1 | 15 | |
5 | 34.26 | 27 | 15 | 2 | 17 | |
6 | 32.28 | 31 | 14 | 1 | 15 | |
2005–6 | 5 | 36.90 | 81 | 10 | 42 | 52 |
6 | 41.60 | 89 | 8 | 43 | 51 | |
7 | 42.21 | 63 | 10 | 19 | 29 | |
2006–7 | 6 | 37.64 | 120 | 69 | 1 | 70 |
7 | 43.85 | 153 | 82 | – | 82 | |
8 | 38.17 | 125 | 55 | – | 55 | |
All weeks/years | 1256 | 513 | 109 | 622 |
Table 29 suggests that one would expect fewer influenza cases among the ILI cases when the consultation rate falls to baseline levels. The model assumes that the probability that ILI is true influenza is 0.50 across all subgroups (622/1256). Uncertainty surrounding this parameter was modelled using a beta distribution.
Probability that influenza is influenza A
The probability that a case of influenza is influenza A is based on virological surveillance data provided by the HPA (Dr Piers Mook, HPA, personal communication). These data relate to 12 influenza seasons from 1995–6 to 2006–7; they are shown in Table 30.
Season | Influenza A positive | Influenza B positive | Total number of swabs |
---|---|---|---|
2006–7 | 168 | 2 | 170 |
2005–6 | 28 | 85 | 113 |
2004–5 | 76 | 29 | 105 |
2003–4 | 124 | 0 | 124 |
2002–3 | 20 | 20 | 40 |
2001–2 | 39 | 1 | 40 |
2000–1 | 35 | 93 | 128 |
1999–2000 | 77 | 0 | 77 |
1998–9 | 49 | 17 | 66 |
1997–8 | 58 | 1 | 59 |
1996–7 | 74 | 69 | 143 |
1995–6 | 75 | 0 | 75 |
The probability that influenza A is the dominant influenza strain during a given influenza season was calculated from the data shown in Table 30; this gives a probability of 0.75 (influenza B is assumed to be dominant during the 2002–3 season). The probability that a case of influenza is influenza A was then modelled separately for those years where influenza A is dominant and those where influenza B is dominant. For years in which influenza A is dominant, the probability that an influenza case is influenza A was estimated to be 0.86 (740/859). For years in which influenza B is dominant, the probability that an influenza case is influenza A was estimated to be 0.30 (83/281). The overall mean probability that a case of influenza is influenza A is estimated to be 0.72. Beta distributions were used to characterise the uncertainty surrounding the probability that influenza A is dominant and the probability that an influenza case is influenza A given the dominant influenza strain during a given influenza season. These data are used to modify the effectiveness of amantadine which is effective only against influenza A.
Duration of the influenza season
The model assumes that individuals who are effectively protected against influenza by vaccination are protected over the entire influenza season. Individuals receiving influenza prophylaxis are assumed to be protected over the period for which they are taking the drug. Assuming the antivirals are prescribed when influenza is known to be circulating, the preventative efficacies of the antivirals were adjusted according to the proportion of the influenza season for which the individual is taking the drug. Data relating to the duration of the influenza season (when the number of new GP ILI consultations is in excess of 30 per 100,000 population at the current threshold or 50 per 100,000 population at the previous threshold)8 for influenza seasons 1987–8 to 2006–7 were made available to the assessment team by the RCGP (Dr Alex Elliot, RCGP, personal communication). These data are shown in Table 31.
Winter | Epidemic weeks | Consultation rate threshold used to estimate duration |
---|---|---|
1987–8 | 21 | 50 per 100,000 population |
1988–9 | 10 | 50 per 100,000 population |
1989–90 | 9 | 50 per 100,000 population |
1990–1 | 11 | 50 per 100,000 population |
1991–2 | 10 | 50 per 100,000 population |
1992–3 | 10 | 50 per 100,000 population |
1993–4 | 11 | 50 per 100,000 population |
1994–5 | 12 | 50 per 100,000 population |
1995–6 | 11 | 50 per 100,000 population |
1996–7 | 13 | 50 per 100,000 population |
1997–8 | 7 | 50 per 100,000 population |
1998–9 | 8 | 50 per 100,000 population |
1999–2000 | 7 | 50 per 100,000 population |
2000–1 | 11 | 30 per 100,000 population |
2001–2 | 7 | 30 per 100,000 population |
2002–3 | 1 | 30 per 100,000 population |
2003–4 | 9 | 30 per 100,000 population |
2004–5 | 6 | 30 per 100,000 population |
2005–6 | 3 | 30 per 100,000 population |
2006–7 | 3 | 30 per 100,000 population |
Based on the previously higher influenza threshold of 50 per 100,000 population, the mean duration of the influenza season was estimated to be 10.77 weeks. Using the current threshold of 30 new GP consultations per 100,000 population, the mean duration was estimated to be 5.71 weeks. Data relating to the current threshold are assumed in the base-case analysis; the impact of assuming the previous higher threshold is considered within the sensitivity analyses (see One-way/multiway sensitivity analysis and scenario analysis, p. 90). Uncertainty surrounding the duration of the influenza season was modelled using a gamma distribution; a standard error of 7 days was assumed within the analysis.
Modelling the preventative efficacy of vaccination
The preventative efficacy of influenza vaccination for children, adults and the elderly was derived from meta-analyses of RCTs presented within three recent Cochrane reviews of influenza vaccination. 117–119 The model assumes that influenza vaccination and prophylaxis are effective against true influenza but not ILI and that inactive parenteral vaccines represent the mainstay of vaccination use in England and Wales.
The Cochrane reviews report the RR of experiencing influenza for vaccination versus placebo to be 0.36 (95% CI 0.28–0.48) in healthy children, 0.35 (95% CI 0.25–0.49) in healthy adults and 0.42 (95% CI 0.27–0.66) in elderly populations. These preventative efficacy rates are assumed to be the same for otherwise healthy and at-risk groups within each age band. The propagation of these RRs leads to a proportionate reduction in the probability of experiencing secondary ILI complications and death within vaccinated patients. It should be noted that the health economic analysis reported by Turner et al. 10 assumed that influenza vaccination also had an impact in terms of reducing the probability of pneumonia, hospitalisation and mortality in adult and elderly patient groups, based on a meta-analysis of influenza vaccination in the elderly reported by Gross et al. 120 However, the odds ratios for these end points appear to relate to pneumonias, hospitalisations and deaths in the intention-to-treat populations within trials of vaccination versus no vaccination; hence, the inclusion of these effects is likely to result in double counting and an overestimate of the benefits of vaccination. Additional benefits of vaccination in terms of reducing pneumonias, hospitalisations and mortality are thus not included in the Assessment Group model presented here. It should also be noted that vaccination status is incorporated as a characteristic of the subgroups included in the assessment; vaccination is not considered as an option for this assessment.
The benefit of prior influenza vaccination is applied in the model to vaccinated subgroups by reducing the probability of developing influenza without prophylaxis. This is calculated as the probability of developing ILI minus the probability that ILI is influenza multiplied by 1 minus the RR of influenza for vaccination. The preventative efficacy of prophylaxis is then applied to any remaining cases of influenza which are not effectively protected by vaccination (the probability of developing other ILI is unaffected by vaccination). This approach appears to be the most reasonable, given the inconsistent availability of separate efficacy estimates for amantadine, oseltamivir and zanamivir prophylaxis in vaccinated and unvaccinated subgroups.
Modelling the preventative efficacy of antiviral prophylaxis
Estimates of the preventative efficacy of amantadine, oseltamivir and zanamivir in reducing SLCI were derived from evidence included in the systematic review of clinical effectiveness presented in Chapter 3. The model assumes that amantadine, oseltamivir and zanamivir as seasonal prophylaxis are effective only for the period in which the patient is taking the drug. In the absence of evidence concerning the relationship between the point at which patients withdraw from prophylaxis and the protective benefits of prophylaxis in these patients, the model assumes that patients who withdraw from prophylaxis do so at the beginning of the prophylaxis course and receive no protective benefit over individuals who do not receive prophylaxis. This assumption is in line with the previous modelling work reported by Turner et al. 10
Preventative efficacy of prophylaxis using amantadine
The systematic review of clinical effectiveness presented in Chapter 3 highlighted a paucity of evidence relating to the efficacy of amantadine in both the seasonal and post-exposure prophylaxis settings. Two studies were available relating to the seasonal prophylaxis of influenza using amantadine;57,58 data relating to the relative protective benefit of amantadine compared with placebo was available only from the study reported by Reuman et al. 57 This study included healthy adults who had not been vaccinated; a mean RR of 0.40 (95% CI 0.08–2.03) was estimated from the event data reported within the clinical trial publication. Owing to the absence of additional or alternative studies, this parameter estimate was applied to all subgroups in the seasonal prophylaxis model, hence the model assumes that the preventative efficacy of amantadine is independent of age and risk status. It should be noted that the systematic searches did not identify any direct evidence of the benefit of amantadine in the paediatric population in line with licensed indications (see Chapter 3); therefore we have extrapolated efficacy estimates from the adult population.
The systematic review did not identify any clinical trials of the effectiveness of amantadine in the post-exposure prophylaxis setting within households (see Chapter 3). However, one study was identified which examined the efficacy of amantadine in outbreak control in healthy adolescents in a boarding school over a period of 14 days. 59 The majority of subjects recruited within this study had been previously vaccinated for influenza. Prior vaccination does not necessarily confound the analysis of the efficacy of prophylaxis; however, it is likely that the presence of effective vaccination would reduce the statistical power of the trial comparison (as a result of lower attack rates in both prophylaxis and placebo groups). Efficacy estimates within the outbreak control setting were assumed to be similar to those for amantadine when used as post-exposure prophylaxis, as the duration of prophylaxis is similar (assuming post-exposure prophylaxis using amantadine would be taken for a duration of 10 days). Based on the event data reported in the clinical trial publication, the RR of amantadine versus placebo was estimated to be 0.10 (95% CI 0.03–0.34). Owing to a lack of any alternative evidence, this RR was applied to all subgroups in the model.
The model assumes that a proportion of patients develop amantadine-resistant disease; these patients are assumed to derive no prophylactic benefit from amantadine. Surveillance data (provided as academic-in-confidence) were provided by the HPA regarding the proportion of H1N1 and H3N2 isolates that were resistant to amantadine during the years 2004–7. Resistance may occur in either strain; recent data suggest that amantadine resistance is considerably higher in the H3N2 strain. Based on the data for the 2006–7 influenza season, the model assumes that 37% of influenza A cases are resistant to amantadine. This proportion is a crude estimate based on the experience over a single influenza season and may vary considerably as resistance levels and the ratio of H3N2 and H1N1 strains vary from year to year.
Preventative efficacy of prophylaxis using oseltamivir
The systematic review of clinical effectiveness presented in Chapter 3 identified a more substantial evidence base relating to the effectiveness of oseltamivir in the prophylaxis of influenza. Two studies of seasonal prophylaxis using oseltamivir were identified;64,66 one study66 recruited healthy adults (unvaccinated), while the other trial recruited at-risk elderly subjects in a residential home (> 80% of subjects vaccinated in intervention and control groups). 64 The study reported by Hayden et al. 66 was applied to the otherwise healthy and at-risk paediatric and working-age adult populations, while preventative efficacy estimates from the study reported by Peters et al. 64 were applied to the otherwise healthy and at-risk elderly populations. Based on event data reported by Hayden et al. ,66 the RR of developing influenza was estimated to be 0.24 (95% CI 0.10–0.58). Analysis of event data reported by Peters et al. 64 suggested an RR of developing influenza of 0.08 (95% CI 0.01–0.63). It is unclear whether the difference between efficacy rates from these two trials is a result of differences in terms of study population, underlying risk or another unknown source of heterogeneity.
Two studies were identified which evaluated the preventative efficacy of oseltamivir in the post-exposure prophylaxis of influenza. 48,49 The preventative efficacy of oseltamivir for the healthy adult group was based on a random-effects meta-analysis of these two studies; the mean RR used in the model was estimated to be 0.19 (95% CI 0.08–0.45). Importantly, within the two trials included in the meta-analysis one trial included paediatric and adult subjects48 while the other included only adult subjects. 49 Owing to a paucity of alternative evidence, this RR was applied to all otherwise healthy and at-risk adult populations. In the paediatric population, the RR of developing influenza following oseltamivir post-exposure prophylaxis was modelled on the subgroup analysis reported by Hayden et al. ;48 the mean RR of developing influenza for children was 0.36 (95% CI 0.16–0.80). This RR was applied to both the otherwise healthy and at-risk paediatric subgroups.
Preventative efficacy of prophylaxis using zanamivir
The systematic review identified two clinical trials relating to the benefit of zanamivir for the seasonal prophylaxis of influenza. 70,75 The study reported by Monto et al. 70 recruited healthy adults, the majority of whom were unvaccinated. The study reported by La Force et al. 74 recruited at-risk adults; subjects recruited into this study had a higher level of vaccination. Based on the event data reported in the clinical trial paper, the RR of developing influenza in healthy adults was estimated to be 0.32 (95% CI 0.17–0.63). 70 This estimate was applied to the otherwise healthy and at-risk children subgroups as well as to the healthy adult subgroup. Similarly, the RR of developing influenza in at-risk adults was estimated to be 0.17 (95% CI 0.06–0.50); this RR was applied to the at-risk adult working age subgroup. 75 The RR for the elderly populations was based on a subgroup analysis reported by LaForce et al. ;75 this RR was estimated to be 0.20 (95% CI 0.02–1.72).
The review identified three trials which reported the clinical efficacy of zanamivir versus placebo for the post-exposure prophylaxis of influenza in adults72 and children and adults. 46,47 The RR of developing influenza in all subgroups receiving zanamivir was estimated using a random-effects meta-analysis of these three trials; the RR was estimated to be 0.21 (95% CI 0.13–0.33). One study did evaluate zanamivir as outbreak control in largely at-risk elderly subjects;76 the model does not use efficacy data from this study because of differences in the duration of prophylaxis. The use of the meta-analysis estimate for zanamivir in post-exposure prophylaxis in households represents a bias in favour of zanamivir in this subgroup.
Relative risks and 95% CIs (shown in parentheses) used in the model are summarised in Table 32. The footnotes detail whether each RR is based on trial evidence relating exclusively to the model subgroup, trial evidence that includes the subgroup or trial evidence relating to other subgroups.
Intervention | Healthy children | At-risk children | Healthy adults | At-risk adults | Healthy elderly | At-risk elderly |
---|---|---|---|---|---|---|
Vaccination | 0.36 (0.28–0.48) | 0.36c (0.28–0.48) | 0.35 (0.25–0.49) | 0.35c (0.25–0.49) | 0.42 (0.27–0.66) | 0.42c (0.27–0.66) |
Amantadine (seasonal) | 0.40c (0.08–2.03) | 0.40c (0.08–2.03) | 0.40a (0.08–2.03) | 0.40a (0.08–2.03) | 0.40c (0.08–2.03) | 0.40c (0.08–2.03) |
Amantadine (post-exposure) | 0.10b (0.03–0.34) | 0.10b (0.03–0.34) | 0.10b (0.03–0.34) | 0.10b (0.03–0.34) | 0.10c (0.03–0.34) | 0.10c (0.03–0.34) |
Oseltamivir (seasonal) | 0.24c (0.10–0.58) | 0.24c (0.10–0.58) | 0.24a (0.10–0.58) | 0.24a (0.10–0.58) | 0.08b (0.01–0.63) | 0.08b (0.01–0.63) |
Oseltamivir (post-exposure) | 0.36a (0.16–0.80) | 0.36a (0.16–0.80) | 0.19b (0.08–0.45) | 0.19b (0.08–0.45) | 0.19b (0.08–0.45) | 0.19b (0.08–0.45) |
Zanamivir (seasonal) | 0.32c (0.17–0.63) | 0.32c (0.17–0.63) | 0.32a (0.17–0.63) | 0.17b (0.06–0.50) | 0.20b (0.02–1.72) | 0.20b (0.02–1.72) |
Zanamivir (post-exposure) | 0.21b (0.13–0.33) | 0.21b (0.13–0.33) | 0.21b (0.13–0.33) | 0.21b (0.13–0.33) | 0.21b/c (0.13–0.33) | 0.21b/c (0.13–0.33) |
It should be noted that the evidence surrounding the effectiveness of amantadine, oseltamivir and zanamivir within specific subgroups is not ideal, and decisions regarding the appropriate inclusion of specific preventative efficacy estimates are not straightforward. For the most part, preventative efficacy is assumed to be the same across a number of age and risk subgroups (even those where there is no trial evidence relating to the subgroup under consideration, e.g. amantadine post-exposure prophylaxis in the elderly). In other instances, where multiple sources exist, there are known heterogeneities between study populations (age, risk status, level of prior vaccination), methods of end-point measurement and duration of prophylaxis. It is unclear whether differences observed in these preventative efficacy estimates are a result of one or a combination of these known heterogeneities or some other underlying differences between the studies. The uncertainty surrounding all RRs of developing influenza for vaccination and prophylaxis was modelled using lognormal distributions; estimates of preventative efficacy were sampled from a normal distribution characterised by the logmean RR and the standard error of the log of the RR. The reader should be aware that there is likely to be a greater level of uncertainty surrounding these effectiveness estimates than the uncertainty reflected in data from the studies included in the systematic review.
Adverse events due to influenza vaccination and prophylaxis
The model includes the possibility of experiencing adverse events for patients receiving vaccination and/or amantadine prophylaxis. The probability of experiencing adverse events due to vaccination was based on data reported by Turner et al. ,76 sourced from an observational study of a 2-day work absence per 100 healthy adults as a result of influenza vaccination. 121 Although larger surveillance data sources are available [e.g. the vaccine adverse event reporting system (VAERS)], these tend to be insensitive in the identification of minor adverse events. Adverse events due to vaccination are assumed to be self-limiting, to require no treatment and to have no impact on HRQoL. However, the model does assume that patients experiencing adverse events due to vaccination will consult their GP for advice.
Evidence concerning the incidence of adverse events due to influenza prophylaxis is equivocal. In some instances, higher adverse event rates were reported in the placebo groups of the trials than the intervention groups, while in other instances, rates were higher in the intervention groups (see Chapter 3). In most cases, it is unclear whether adverse events are related to the prophylaxis or the clinical condition. This is further complicated by the poor reporting of the severity of adverse events within the clinical trials. The evidence does not allow for a robust comparison of adverse event rates between amantadine, oseltamivir and zanamivir. In the absence of more robust estimates from the trials included in the systematic review (see Chapter 3), assumptions regarding the probability of adverse events for amantadine, oseltamivir and zanamivir were drawn from the previous modelling work reported by Turner et al. 10 In line with Turner et al. ,10 the model assumes that the adverse events associated with the NIs are self-limiting, incur no treatment cost and have no impact on HRQoL. There is evidence, however, that amantadine can result in severe neuropsychiatric adverse events including behavioural changes, delirium, hallucinations, agitations and seizures. 10,14 In an attempt to capture these health effects, a utility decrement of 0.20 is assumed per day of adverse events for a mean duration of 5 days, based on the analysis reported by Turner et al. 10 The model assumes that the probability of experiencing adverse events due to amantadine is 5%. The QALY loss associated with amantadine adverse events was characterised using a beta distribution, while the duration of adverse events was modelled using a gamma distribution.
Withdrawal rates for influenza prophylaxis
In the absence of better quality evidence identified from the clinical trials included in the systematic review (see Chapter 3), withdrawal rates from prophylaxis were based on those reported within the previous modelling study reported by Turner et al. 10 The probability of withdrawal for amantadine was assumed to be 5.7% in children and healthy adults, and 14.7% in at-risk adults and elderly individuals. The probability of withdrawal was assumed to be 2% for oseltamivir and 1.3% for zanamivir across all model subgroups. 10 Uncertainty surrounding withdrawal rates was modelled using beta distributions.
Parameters relating to the onset of influenza and other influenza-like illnesses
Probability of an individual with ILI presenting symptomatically
There is considerable uncertainty surrounding the probability that an individual with ILI will consult a health-care professional in either primary or secondary care. The model reported by Turner et al. 10 used evidence from a study of the excess GP consultations reported by Fleming. 112 The use of these data implies the assumption that all excess GP consultations over the influenza season compared with the baseline rate are due to influenza. The validity of this assumption is questionable,122 as other ILIs such as RSV are often more prevalent during the influenza season, thus accounting for an unknown proportion of excess cases between the influenza season and baseline periods. Instead, the ILI consultation rate was based on a European ILI surveillance study reported by van Noort et al. 104 This study used an internet-based approach to monitoring ILI symptoms and consultations in the general population in the Netherlands, Belgium and Portugal. The study reported highly variable consultation rates for individuals with ILI ranging from 25% to 67%. The model assumes that the true probability that an individual with symptomatic ILI will present is likely to be at the lower end of this range. The model assumes a central estimate of 0.25; uncertainty surrounding this parameter value was modelled using a beta distribution assuming a subjectively large standard error (alpha = 5, beta = 15). The probability of presentation with ILI is assumed to be the same for all subgroups included in the model. The impact of this assumption is explored in the sensitivity analysis (see One-way/multiway sensitivity analysis and scenario analysis, p. 90).
Probability of an individual presenting within 48 hours of symptomatic onset of ILI
Treatment using oseltamivir is currently recommended only for those individuals who are considered to be at high risk of developing complications who present within 48 hours of symptomatic onset. 116 The probability of an individual presenting with ILI within 48 hours of onset was derived from a study reported by Ross et al. 123 The model assumes that half of those presenting on day 2 would be within the 48-hour cut-off; this assumption is in line with the previous model reported by Turner et al. 10 In this study, the probability of presentation within 48 hours was reported to be 52%, 16% and 11% in the paediatric, working-age adult and elderly populations respectively. These probabilities are assumed to be the same in otherwise healthy and at-risk populations. The uncertainty surrounding these parameters was modelled using beta distributions based on the empirical data reported by Ross et al. 123
Probability that an individual presenting within 48 hours is prescribed an NI for the treatment of ILI
In line with current recommendations from NICE concerning the use of NIs for the treatment of influenza and other ILI, the model assumes that oseltamivir and zanamivir are prescribed only for patients who are at risk of secondary complications of ILI (including elderly patients over 65 years of age). For the paediatric population who are eligible for treatment, the model assumes that patients are treated using oseltamivir. For at-risk adult populations, the model assumes that 89% of patients receive oseltamivir, based on data reported within the submission to NICE by Roche. 20 The remaining 11% of patients are assumed to receive treatment using zanamivir.
Probability of developing complications due to influenza and other ILI
The incidence of complications associated with influenza and ILI is not reported in detail within clinical trials of influenza prophylaxis (see Chapter 3). Instead, the probability of developing a complication of influenza or other ILI was taken directly from a large UK-based observational study reported by Meier et al. 12 This study collected and analysed data concerning the incidence, risk factors, clinical complications and drug utilisation associated with influenza and ILI using data collected in the GPRD in the period 1991–6. A total of 141,293 patients in the database were reported to have one or more diagnoses of influenza or ILI. Data concerning the incidence of specific complications, including exacerbations of underlying diseases and death due to influenza, were reported by age group (1–14 years, 15–49 years, 50–64 years, and > 65 years) and by presence of pre-existing chronic diseases. The rates of specific complications reported by Meier et al. 12 are shown in Table 33.
Type of complication | 1–14 years | 15–49 years | 50–64 years | > 65 years | ||||
---|---|---|---|---|---|---|---|---|
Healthy | Predisposed | Healthy | Predisposed | Healthy | Predisposed | Healthy | Predisposed | |
Respiratory tract | 1697 | 520 | 4530 | 1337 | 1106 | 604 | 819 | 754 |
Bronchitis | 113 | 21 | 748 | 203 | 309 | 167 | 273 | 256 |
Pneumonia | 29 | 9 | 185 | 35 | 52 | 27 | 106 | 97 |
URTI | 1470 | 302 | 3502 | 684 | 722 | 300 | 457 | 346 |
Cardiac | 0 | 0 | 4 | 9 | 7 | 20 | 9 | 59 |
CNS | 17 | 0 | 85 | 10 | 16 | 5 | 21 | 23 |
Renal | 2 | 0 | 5 | 3 | 4 | 2 | 5 | 12 |
Other | 701 | 156 | 646 | 143 | 141 | 49 | 195 | 171 |
Otitis media | 684 | 153 | 454 | 94 | 46 | 16 | 21 | 11 |
GI bleeding | 17 | 2 | 171 | 44 | 81 | 22 | 67 | 49 |
Death | 0 | 1 | 21 | 5 | 12 | 11 | 110 | 114 |
Total patients | 2311 | 650 | 5185 | 1472 | 1252 | 670 | 981 | 936 |
Total complications | 2417 | 676 | 5270 | 1502 | 1274 | 680 | 1049 | 1019 |
Complications per patient | 1.05 | 1.04 | 1.02 | 1.02 | 1.02 | 1.01 | 1.07 | 1.09 |
Number in group | 17,201 | 3695 | 69,231 | 12,195 | 16,017 | 5402 | 10,145 | 7407 |
Total cases in age group | 20,896 | 81,426 | 21,419 | 17,552 |
Data concerning complication rates for the predisposed group were assumed to represent the at-risk populations within the model. Complication rates among the 15–49 year age group and the 50–64 year age group were combined to represent the working-age adult model populations. Uncertainty surrounding the probability of experiencing a complication of influenza within each population group was modelled using beta distributions, while the multinomial probabilities of experiencing specific complications were modelled using Dirichlet distributions with minimally informative priors based on the methods reported by Briggs et al. 124 The model assumes that the risk of developing complications is the same for influenza and other ILI.
It should be noted that the use of this study is flawed in that many of the ILIs reported by Meier et al. will be caused by viruses other than influenza. This problem is compounded further as the study reported ILI complications over the whole year rather than the influenza season, hence the proportion of episodes caused by other ILIs is likely to be higher than that for the period when influenza is known to be circulating. A limitation of these data, and their use in the model, is that the rates of complications resulting from other ILIs which are not influenza may not reflect complication rates due to influenza infection.
Effectiveness of NIs for the treatment of symptomatic influenza
The efficacy and safety of oseltamivir and zanamivir for the treatment of influenza and other ILI is beyond the scope of this assessment and is scheduled for reappraisal in 2008. However, both zanamivir and oseltamivir are currently recommended for treatment of symptomatic influenza and ILI in at-risk individuals. 116 Evidence concerning the safety and efficacy of the NIs for the treatment of ILI was derived from the earlier HTA report by Turner et al. 10 The model assumes that oseltamivir and zanamivir reduce the probability of experiencing complications due to influenza and lead to a modest reduction in the impact of influenza on quality of life compared with best symptomatic care alone. The model assumes an odds ratio for all complications for zanamivir versus no treatment of 0.49 (95% CI 0.23–1.04) in all at-risk populations, while the odds ratio for complications for oseltamivir versus no treatment is assumed to be 0.65 (95% CI 0.43, 0.97) in the at-risk paediatric population and 0.40 (95% CI 0.16–0.93) in at-risk adult and elderly populations. 10 The model assumes that the NIs are not effective in reducing complications due to other ILIs which are not influenza. The odds ratios derived from Turner et al. 10 relate to reductions in complications requiring antibiotics; owing to the high rates of antibiotic use for the treatment of ILI-related complications,12 and the absence of alternative evidence, the model assumes that these efficacy rates are applied to all ILI-related complications. It is possible that this may overstate the benefit of zanamivir and oseltamivir in terms of reducing complications due to influenza and other ILI. A summary of treatment efficacy values assumed within the model is shown in Table 34.
Odds ratios (and 95% CIs) for reduction in complications | ||
---|---|---|
Population | Odds ratio for zanamivir | Odds ratio for oseltamivir |
Healthy children | 0.70 (0.52–0.96) | 0.65 (0.43–0.97) |
At-risk children | 0.49 (0.23–1.04) | 0.65 (0.43–0.97) |
Healthy adults | 0.70 (0.52–0.96) | 0.40 (0.16–0.93) |
At-risk adults | 0.49 (0.23–1.04) | 0.40 (0.16–0.93) |
Healthy elderly | 0.70 (0.52–0.96) | 0.40 (0.16–0.93) |
At-risk elderly | 0.49 (0.23–1.04) | 0.40 (0.16–0.93) |
As noted in Model structure (above), the model assumes that the use of neuraminidase inhibitors for the treatment of symptomatic influenza is independent of the prophylactic strategy and requires a further prescription (any remaining NI prophylaxis at the point of infection cannot be used as treatment at a higher dose). The impact of this assumption is explored in the sensitivity analysis by excluding the possibility of NI treatment for patients who develop symptomatic ILI.
Probability of receiving antibiotics
The model assumes that antibiotics may be prescribed for both patients who present with uncomplicated ILI and those who present with complicated ILI. The probability that an individual with or without complications is prescribed antibiotics was derived from the study reported by Meier et al. 12 The probability that a patient with uncomplicated influenza or ILI receives antibiotics was estimated to be 0.28, 0.42 and 0.55 in the paediatric, adult and elderly populations respectively. The probability that a patient with complicated influenza or ILI receives antibiotics was estimated to be 0.71, 0.80 and 0.74 in the paediatric, adult and elderly populations respectively. Owing to a lack of evidence to the contrary, these values are assumed to be the same for both the otherwise healthy and the at-risk populations. Uncertainty surrounding these probabilities was modelled using beta distributions based on the empirical data reported by Meier et al. ,12 as shown in Table 35.
Patients without complications | Patients with complications | |||
---|---|---|---|---|
Age group | Number receiving antibiotics | Number in group | Number receiving antibiotics | Number in group |
1–14 years | 2183 | 3093 | 4997 | 17,910 |
15–64 years | 6983 | 8726 | 39,622 | 94,338 |
> 65 years | 1527 | 2068 | 8544 | 15,620 |
Probability of hospitalisation due to ILI-related complications
The model assumes that patients who experience ILI-related complications may require hospitalisation. As noted above, the clinical trials of influenza prophylaxis do not consistently report the incidence of complications and as such do not provide any information regarding the probability that an individual requires hospitalisation. Furthermore, data relating to hospitalisation rates were not available from the study by Meier et al. 12 Instead, the probability of hospitalisation was derived from hospitalisation rates for lower RTIs reported within a meta-analysis of 10 trials of oseltamivir for the treatment of symptomatic influenza reported by Kaiser et al. 99 The probability of hospitalisation for individuals with influenza-related complications was estimated from the placebo arm data across the 10 included studies; this probability was estimated to be 0.11 (5/46) in the otherwise healthy children and working-age adult subgroups and 0.16 (15/95) in the at-risk subgroups (including otherwise healthy elderly). 99 The data presented in the study publication did not allow for these estimates to be subdivided further according to age group; this is unfortunate as age is likely to affect the risk of hospitalisation. Uncertainty surrounding the probability of hospitalisation was modelled using beta distributions based on the empirical data reported by Kaiser et al. 99
A proportion of patients who are hospitalised may require ITU care with or without mechanical ventilation. The previous model reported by Turner et al. 10 assumed that 4.9% (22/453) of patients undergo mechanical ventilation. No alternative evidence could be identified, hence the model uses these same parameter values. A beta distribution was used to characterise the uncertainty surrounding this parameter.
Probability of death due to ILI-related complications
The probability of death due to ILI-related complications was taken from the population-based study reported by Meier et al. 12 The probability of death due to influenza complications was observed to be very low in the paediatric and adult populations (< 1%); this probability was observed to be considerably higher in the elderly patients represented within the database (10–11%). The risk of death due to complications of ILI was observed to be slightly elevated in the predisposed populations compared with the otherwise healthy patients. As complications may be a result of true influenza or other ILI, the model assumes that the probability of death is the same for those patients who develop complications due to influenza and for those patients who develop complications due to other ILI. Uncertainty surrounding this parameter was modelled using beta distributions based on the empirical data reported by Meier et al. ,12 as shown in Table 36.
Population | Number of deaths | Number of complications | Probability of death |
---|---|---|---|
Healthy children | 0 | 2417 | 0.00 |
At-risk children | 1 | 676 | 0.00 |
Healthy adults | 33 | 6544 | 0.005 |
At-risk adults | 16 | 2182 | 0.007 |
Healthy elderly | 110 | 1049 | 0.1049 |
At-risk elderly | 114 | 1019 | 0.112 |
Modelling resource use and costs associated with influenza and other ILI
The model includes the acquisition and administration costs for vaccination, antiviral prophylaxis and treatment, costs associated with the management of adverse events, consultation costs, antibiotics, and hospitalisation costs for managing severe ILI-related complications. As the time horizon for the model is effectively 1 year in duration, costs were not subjected to discounting.
Costs of prophylaxis and treatment using amantadine, oseltamivir and zanamivir
Prophylaxis and treatment were costed according to BNF list prices at the time of the assessment. The number of doses of prophylaxis required using amantadine, oseltamivir and zanamivir was calculated based on the dosages and durations in line with licensed indications (see Table 27). The model assumes that seasonal prophylaxis using amantadine is given for a period of 6 weeks (42 days) for patients who have not been previously vaccinated, and 3 weeks (21 days) for patients who have been previously vaccinated. The model assumes that seasonal prophylaxis using oseltamivir is given for a period of 6 weeks (42 days). Seasonal prophylaxis using zanamivir is assumed to be given for a period of 4 weeks (28 days). The model assumes that post-exposure prophylaxis using amantadine, oseltamivir and zanamivir is given for a period of 10 days. The duration of treatment of symptomatic ILI using oseltamivir and zanamivir is assumed to be 5 days. In line with licensed indications, the daily dosage of amantadine prophylaxis and zanamivir prophylaxis is assumed to be 100 mg and 10 mg respectively for all populations. The cost of prophylaxis and treatment using oseltamivir for children assumes a mean body mass of between 23 kg and 40 kg. Unit costs for amantadine, oseltamivir and zanamivir were taken from the BNF No. 54. 14 Amantadine is available in both capsule and syrup form, and oseltamivir is available as capsules and as a suspension for reconstitution with water. The model assumes that prophylaxis for adults is administered using capsules rather than syrup or suspension, as this allows for more reliable dosing (Dr Andrew Ross, RCGP, personal communication). The cost of each prophylaxis course and treatment course includes the possibility of wastage. Where multiple products were available, the least expensive is assumed. The costs of prophylaxis used in the model are presented in Table 37.
Seasonal prophylaxis | Post-exposure prophylaxis | Treatment | ||||
---|---|---|---|---|---|---|
Drug | Adults | Children | Adults | Children | Adults | Children |
Amantadine prophylaxis (unvaccinated) | £14.40 | £14.40 | £4.80 | £4.80 | NA | NA |
Amantadine prophylaxis (previously vaccinated) | £9.60 | £9.60 | £4.80 | £4.80 | NA | NA |
Oseltamivir prophylaxis | £81.80 | £73.65 | £16.36 | £16.36 | £16.36 | £16.36 |
Zanamivir prophylaxis | £73.65 | £73.65 | £24.55 | £24.55 | £24.55 | £24.55 |
In the base-case analysis, the model assumes that each prescription of prophylaxis requires a GP consultation. The model assumes also that the administration of vaccination and the prescription of antiviral prophylaxis require separate consultations. The impact of prescribing multiple courses of prophylaxis for contact cases is explored in the sensitivity analysis (see One-way/multiway sensitivity analysis and scenario analysis, p. 90).
Cost of vaccination
The cost of influenza vaccination was estimated from list prices derived from BNF 54. 14 Current unit costs for influenza vaccine products range from £4.98 to £6.59, including both proprietary and non-proprietary products (Table 38). Recommended influenza vaccines vary between influenza seasons; the mean vaccine price was assumed within the model (£5.63). The model assumes that influenza vaccination is administered by a GP; the cost of vaccination is assumed to include the cost of a GP consultation based on costs reported by Curtis and Netten. 102 A GP visit is assumed to cost £25. As these costs are common to all patients receiving vaccination, these parameters have no impact on the incremental cost-effectiveness of influenza prophylaxis.
Product | Type of vaccine | Unit cost |
---|---|---|
Inactivated influenza vaccinea | Suspension of formaldehyde-inactivated influenza virus (split virion) | £6.29 |
Inactivated influenza vaccinea | Suspension of propiolactone-inactivated influenza virus (surface antigen) | £3.98 |
Agrippal® | Suspension of formaldehyde-inactivated influenza virus (surface antigen) | £5.03 |
Begrivac® | Suspension of formaldehyde-inactivated influenza virus (split virion) | £5.03 |
Enzira® | Suspension of inactivated influenza virus (split virion) | £6.59 |
Fluarix® | Suspension of formaldehyde-inactivated influenza virus (split virion) | £4.49 |
Imuvac® | Suspension of formaldehyde-inactivated influenza virus (surface antigen) | £6.59 |
Influvac subunit® | Suspension of formaldehyde-inactivated influenza virus (surface antigen) | £5.22 |
Mastaflu® | Suspension of formaldehyde-inactivated influenza virus (surface antigen) | £6.50 |
Viroflu® | Suspension of inactivated influenza virus (surface antigen, virosome) | £6.59 |
Cost of ILI presentation
The model assumes that patients present with symptomatic ILI either to their GP (in the surgery or at home) or at an A&E department. The probability that a patient with influenza or other ILI requires a home visit was derived from the study reported by Ross et al. 123 Counts of patients with ILI who had home visits were reported in aggregate form for patients aged under 75 and those aged over 75. Further data regarding the proportion of consultations which took place at home within each age group were provided by the lead author of this study (Dr Andrew Ross, RCGP, personal communication). The proportion of home visits was low in the paediatric and adult populations (5% and 8% respectively); the proportion was considerably higher in the elderly population (38%). Beta distributions were used to characterise the uncertainty surrounding this parameter based on the empirical data provided by Dr Ross of the RCGP. The proportion of all ILI presentations that take place in A&E departments was based on clinical opinion (Professor Robert Read, University of Sheffield, personal communication); the model assumes that 3% of patients present to A&E (range 1–5%). A beta distribution was used to capture the uncertainty surrounding this quantity. This parameter was assumed to be the same for otherwise healthy and at-risk paediatric, adult and elderly populations.
Unit costs for GP surgery consultations and home visits were derived from the PSSRU102 while the cost of an A&E consultation was derived from the NHS reference costs. 125 The model assumes that a GP surgery consultation costs £25,102 a home visit costs £69102 and an A&E attendance costs £95.56 (first attendance data code 180F). 125 The unit costs for A&E attendances are assumed to include the costs of diagnostic tests (e.g. blood and sputum tests, lung function tests, etc.). Based on the information reported above, the model assumes a mean cost of presentation with symptomatic ILI of £29.52 for children, £30.73 for working-age adults and £43.20 for elderly individuals.
Cost of antibiotics for the treatment of ILI-related complications
The model assumes that antibiotics are prescribed for individuals presenting with uncomplicated ILI as well as those presenting with influenza and ILI-related complications. The precise antibiotic prescribed depends on the type of complication; for simplicity, the model assumes that the preferred antibiotic for the treatment of symptomatic ILI and related complications is co-amoxiclav. In its non-proprietary tablet form, the unit cost for co-amoxiclav is £6.80 for a 21-tablet course. 14
Cost of managing adverse events resulting from vaccination and prophylaxis
The model assumes that adverse events resulting from vaccination and prophylaxis (amantadine only) incur additional costs due to additional GP attendances. As noted above, the cost of a GP attendance was assumed to be £25. 102 It should be noted that not all patients who experience adverse events will consult their GP, hence it is possible that the costs of managing adverse events is overestimated in the model, although the impact of this bias on cost-effectiveness outcomes is minor. The model assumes that adverse events due to oseltamivir and zanamivir are mild, self-limiting and incur no additional medical costs.
Cost of hospitalisation due to serious complications of influenza and other ILI
The cost of hospitalisation for serious complications was taken from the NHS reference costs 2005–2006. 125 The unit cost for lobar, atypical or viral pneumonia (D14) without complications was assumed; this was divided by the mean length of stay to derive an estimate of the daily cost of hospitalisation. The standard error for this parameter was estimated by dividing the interquartile range by 1.349 and dividing this by the square root of the number of submissions. This cost was then multiplied by the assumed duration of inpatient stay within each population group reported by Turner et al. 10
Mean lengths of hospitalisation stay due to ILI-related complications were taken from Turner et al. ;10 these are assumed to differ substantially between the paediatric, adult and elderly population subgroups. Turner et al. 10 did not include any uncertainty surrounding these estimates, hence the degree of uncertainty surrounding these mean values has been subjectively modelled using gamma distributions. These data are shown in Table 39.
Population | Mean length of stay (days) | Assumed standard error |
---|---|---|
Healthy children | 2.3 | 3 |
At-risk children | 2.3 | 3 |
Healthy adults | 11.9 | 3 |
At-risk adults | 11.9 | 3 |
Healthy elderly | 15 | 3 |
At-risk elderly | 15 | 3 |
A proportion of patients with particularly severe complications may require ITU care and mechanical ventilation; Turner et al. 10 note that the proportion of cases requiring mechanical ventilation is not known. The model uses the same value reported by Turner et al. 10 (probability of ITU care = 0.05). The typical duration of intensive care required for severely complicated cases was derived from a descriptive study of pneumonia management in the US reported by Oliveira et al. 126 Oliveira et al. report a mean duration of intensive care unit (ICU) stay of 28 days ± 26 days (10 patients). It should be noted, however, that this study may not reflect UK treatment patterns. Uncertainty surrounding this parameter was modelled using a lognormal distribution. The cost per intensive care day was taken from the NHS reference costs 2005–2006. 125 The cost per critical care day was assumed to be £1345.39 (Critical care level 2 code TCCS CC1L2).
Modelling the impact of influenza and ILI on health-related quality of life
The model estimates the number of QALYs lost due to adverse events of prophylaxis (amantadine only), influenza and ILI episodes, complications resulting from influenza and other ILI, and premature death as a result of secondary complications of ILI. In contrast to conventional methods for deriving the number of QALYs gained by the typical patient receiving a given health intervention, the model operates in terms of the number of QALYs lost over the influenza season including an estimate of the impact of premature death due to ILI complications. The difference in QALYs lost between one prophylactic option and its best comparator gives an estimate of the number of QALYs saved, ceteris paribus. It should be noted from the outset that the clinical trials of influenza prophylaxis did not include direct evaluation of the impact of the prophylaxis or disease on health utility using a preference-based valuation method. This problem is compounded by the paucity of reliable health utility estimates indirectly available within the literature. As such, the estimates of HRQoL employed within the model should be treated with caution.
QALYs lost due to influenza and ILI episodes
Previous evaluations of influenza and its prevention and treatment have used health utility scores derived using the EQ-5D127 or the Health Utilities Index, mark III (HUI3)128 based on general population valuations or retrospective valuations from individuals with a history of virologically-confirmed influenza. These studies were based on small numbers of subjects (n < 25). The study reported by Griffin127 reported an extreme value for the utility associated with influenza infection which is valued as a state worse than death (utility = –0.066). 127 It is likely that the impact of influenza on quality of life will be greatest when the illness is at its peak, and that it will have a lesser impact in the first and last days of illness.
The methodology reported by Turner et al. 10 was used to generate QALY loss estimates for cases of influenza and other ILI (see Review of exisiting economic evaluation studies, p. 43). The expected QALY loss due to an episode of influenza was estimated using data collected in five clinical trials of oseltamivir for the treatment of influenza in healthy adults and at-risk and elderly populations. Within these studies, a 10-point Likert scale was completed daily for up to 21 days by patients receiving oseltamivir treatment and patients receiving placebo. The scale employed was similar to a VAS, using a lower anchor which had a score of 0 describing ‘worst possible health’ and an upper anchor which had a score of 10 describing ‘normal health for someone your age’. As the upper anchor on the rating scale did not describe a notional state of ‘best possible health’, Turner et al. 10 recalibrated the upper anchor to represent mean utility scores for each age group using data from the Measurement and Valuation of Health (MVH) study. 105 The VAS equivalent data were then converted into TTO utility scores based on a VAS–TTO transformation algorithm reported by the MVH group. 105 Turner et al. 10 assumed that missing values resulted from the respondent having returned to normal health; missing values were therefore imputed as ‘normal health’ utility scores. The number of QALYs gained over the 21-day period was estimated for the healthy adult and at-risk and elderly populations for oseltamivir and placebo. The number of QALYs lost due to an influenza episode was calculated as the expected QALYs gained in the non-influenza population over 21 days minus the QALYs lost due to influenza over 21 days. For example, assuming a baseline utility of 0.90 without influenza, and a mean 21-day QALY loss of 0.041 with influenza, the number of QALYs lost due to influenza is calculated as (0.90 × 21) – (0.041 × 365)/365.
As equivalent data were not available from the zanamivir trials, the model assumes that the impact of zanamivir treatment on HRQoL is equivalent to that for oseltamivir. Data were not available for the paediatric population; therefore, the model assumes the same QALY loss as in the healthy adult population. The model also assumes that the QALY loss for an uncomplicated influenza episode is the same as that for an uncomplicated ILI episode. Mean QALY gains over 21 days used in the model are presented in Table 40. In their earlier report, Turner et al. 10 modelled the uncertainty in the data, but did not account for additional uncertainty resulting from the process of mapping from Likert data collected in the trials to a VAS and subsequently to TTO utilities. In order to better reflect this uncertainty, the model uses the mean QALY scores and an assumed level of additional uncertainty (subjectively assigned). These parameters were modelled using beta distributions.
Population | Oseltamivir mean QALY | Placebo mean QALY |
---|---|---|
Healthy children | 0.042 | 0.041 |
At-risk children | 0.030 | 0.028 |
Healthy adults | 0.042 | 0.041 |
At-risk adults | 0.030 | 0.028 |
Healthy elderly | 0.030 | 0.028 |
At-risk elderly | 0.030 | 0.028 |
QALYs lost due to adverse events due to prophylaxis
The model assumes that adverse events due to amantadine impact upon a patient’s health-related quality of life. The model assumes a utility decrement of 0.20 for a mean duration of 5 days based on the previous work reported by Turner et al. 10 Uncertainty surrounding the disutility of adverse events was modelled using a beta distribution, whilst uncertainty surrounding the duration of adverse events was modelled using a gamma distribution, assuming a standard error of 1 day.
QALYs lost due to ILI-related complications
In principle, the Likert scale data collected within the oseltamivir trials should have included quality of life valuations for individuals who experienced serious complications of influenza (or at least those occurring within the 21-day evaluation period). However, it should be noted that beyond the first 7 days, the number of respondents in the treatment and placebo groups declined considerably. The model assumes that serious complications such as respiratory illness and the exacerbation of underlying health problems are not captured within these valuations, and that such complications result in a further reduction in a patient’s HRQoL.
Systematic searches were undertaken to identify studies reporting preference-based valuations of the impact of influenza, ILI and related complications on HRQoL (see Appendix 1). The searches did not identify any published studies that reported preference-based valuations of the impact of the range of ILI complications associated with influenza and ILI (bronchitis, pneumonia, otitis media and exacerbation of an underlying condition, e.g. asthma). Instead, health utility decrements for secondary complications were derived from a modelling study of vaccination against a variety of diseases. 129 Within this study, committee HUI (mark II) scores were derived for a number of health states associated with influenza and ILI (Table 41). These utility estimates represent the consensus of the committee who undertook the valuation exercise and as such do not include any estimates of uncertainty. Wide standard errors were assumed within the probabilistic sensitivity analysis based on lognormal distributions.
Parameter | Committee HUI values | Mean decrement from baseline | Assumed lower 95% CI | Assumed upper 95% CI |
---|---|---|---|---|
Baseline utility score | 0.90 |
– |
– | – |
Utility – moderate to severe respiratory illness | 0.75 | 0.15 | 0.05 | 0.25 |
Utility – exacerbation of cardiac/asthma complication | 0.53 | 0.37 | 0.27 | 0.47 |
Utility – other complications | 0.53 | 0.37 | 0.27 | 0.47 |
The duration over which these utility decrements are applied was based on clinical trial data presented within the Roche submission,20 sourced from clinical trials of oseltamivir. The duration of each illness was derived simply by calculating the number of days between the onset of the complication and its resolution (Gavin Lewis, Roche, personal communication). The submission contained data relating to the duration of pneumonia, bronchitis and otitis media in children, healthy adults and at-risk groups. The mean duration of disutility for any respiratory complication was estimated by weighting the durations observed in the clinical trials by the ratio of pneumonia : bronchitis in each age group, as reported by Meier et al. 12 In the absence of any alternative evidence, the duration of other respiratory complications was assumed to follow this same pattern. The uncertainty analysis assumes a large standard error of 3 days for each subgroup; uncertainty surrounding these quantities was modelled using gamma distributions. Owing to a lack of alternative evidence, the duration of other non-respiratory complications is assumed to be the same as that for respiratory complications. Table 42 shows the assumed durations for these reductions in HRQoL.
Population | Respiratory and other complications | Otitis media | ||
---|---|---|---|---|
Mean duration (days) | Assumed standard error | Mean duration (days) | Assumed standard error | |
Healthy children | 7.89 | 3.00 | 9.36 | 3.00 |
At-risk children | 8.07 | 3.00 | 9.36 | 3.00 |
Healthy adults | 9.23 | 3.00 | 9.36 | 3.00 |
At-risk adults | 10.65 | 3.00 | 9.36 | 3.00 |
Healthy elderly | 10.88 | 3.00 | 9.36 | 3.00 |
At-risk elderly | 10.87 | 3.00 | 9.36 | 3.00 |
QALYs lost due to premature death resulting from ILI complications
The expected number of QALYs lost due to premature death resulting from secondary complications of ILI was also based on the methods reported by Turner et al. 10 Crude estimates of the mean age of death due to influenza for the paediatric, adult and elderly populations were derived from data reported by the Office for National Statistics (ONS; DH2). 130 Interim life tables for England and Wales were then used to calculate the expected number of life-years lost due to premature death for each age group based on the mean age of death. Life-years lost were weighted by general population utility scores derived from Kind et al. 131 to generate estimates of the number of QALYs lost within each age group. Expected QALYs lost were discounted at a rate of 3.5%. It should be noted that while the risk of death due to ILI complications is higher in the at-risk groups, the estimate of the number of QALYs lost is assumed to be the same for the healthy and at-risk populations; this assumption may be biased in favour of prophylaxis within the at-risk population subgroups. Table 43 shows the modelled estimates of the expected discounted QALYs for each population group. 96
Population subgroup | Expected QALYs (discounted at 3.5%) |
---|---|
Children | 24.74 |
Adults | 13.37 |
Elderly | 2.95 |
Calculation of cost-effectiveness
The central estimates of cost-effectiveness are based on the expected costs and QALYs lost for each option, as calculated from the results of the stochastic model. This approach is intended to capture any non-linearities in the model parameter distributions. The calculation of cost-effectiveness is fully incremental, whereby each prophylactic strategy is compared against its next best comparator. Prophylactic strategies which are dominated (simple or extended) are ruled out of the analysis.
Uncertainty analysis
One-way sensitivity analysis and scenario analysis
Simple one-way sensitivity analysis and scenario analysis were undertaken to examine the impact of changing model assumptions on the incremental cost-effectiveness of alternative prophylaxis options (the results of these analyses are presented in One-way/multiway sensitivity analysis and scenario analysis, p. 90). Details of these sensitivity analyses are detailed below.
Sensitivity analysis 1: proposed price reduction for zanamivir
In November 2007, the manufacturer of zanamivir (GSK) applied to the Department of Health for a price modulation of two of their drugs, one of which was zanamivir. The current list price for zanamivir is £24.55 (five disks, four blisters per disk); the new proposed price for zanamivir is £16.36 (Toni Maslen, Health Outcomes Programme Leader, GSK, personal communication). This proposed price reduction for zanamivir was approved by the Department of Health with effect from 1 February 2008 but was not listed in the BNF (No. 54)14 at the time of submission. This scenario analysis presents the central estimates of cost-effectiveness of influenza prophylaxis including this proposed price reduction for zanamivir. All other parameter values and assumptions in this analysis are the same as those in the base-case analysis presented in Central estimates of cost-effectiveness (see below). The reader should note that where zanamivir remains dominated by another prophylaxis option despite the price change, the slight differences in the cost-effectiveness of the remaining prophylactic options from the base case results are due to sampling errors in the stochastic model.
Sensitivity analysis 2: deterministic estimates of cost-effectiveness
The base-case health economic analysis is based upon the expected (mean) costs and health outcomes for each prophylactic option, drawn from the stochastic model. The second scenario presents the cost-effectiveness results based on the deterministic model.
Sensitivity analysis 3: cost-effectiveness of oseltamivir given in suspension form
The base-case analysis assumes that seasonal prophylaxis using oseltamivir is prescribed in capsule form to all adult populations, as this is likely to ensure more accurate dosing. However, in principle, oseltamivir given as suspension may allow for less wastage than in capsule form, thus leading to a reduction in the cost of the drug. A 56-cap pack of oseltamivir provides 10×75 mg tablets providing 750 mg of the drug (10 doses) while a 75 ml bottle (60 mg/5 ml) of oseltamivir in suspension form provides a total of 900 mg of the drug (12 doses of 75 mg). While both products cost £16.36 per unit, the use of suspension could, in principle, offer savings over oseltamivir capsules.
Sensitivity analysis 4: multiple prescriptions
The base-case model assumes that each prescription of prophylaxis requires a GP consultation; for vaccinated patients, the model assumes that prophylaxis can be given during the same consultation as the influenza vaccine. The Roche model assumed that four prescriptions of prophylaxis could be obtained per GP attendance. This scenario analysis assumes that four prescriptions may be obtained per individual, resulting in a reduction in the cost of GP attendances for unvaccinated patients. 20
Sensitivity analyses 5 and 6: reduced vaccine efficacy
Sensitivity analysis was undertaken assuming a lower efficacy rate for vaccination to capture the potential impact of a mismatch between vaccine and circulating strains of influenza. Scenario 5 assumes an RR for vaccination of 0.50, while scenario 6 assumes an RR of 0.75.
Sensitivity analysis 7: protection over entire influenza season
The base-case analysis assumes that patients receiving seasonal prophylaxis are at risk of infection when they stop taking the drug. This scenario assumes that the patient is protected over the entire influenza season.
Sensitivity analysis 8: no antiviral treatment for symptomatic influenza
This sensitivity analysis assumes that patients who develop symptomatic ILI do not receive antiviral treatment using oseltamivir or zanamivir.
Sensitivity analysis 9: equivalent efficacy for oseltamivir and zanamivir prophylaxis
There is uncertainty surrounding the relative efficacy of oseltamivir and zanamivir for the prophylaxis of influenza. The Roche model assumed that oseltamivir and zanamivir had equivalent efficacy. This scenario assumes that oseltamivir and zanamivir are equivalent, and uses the most favourable efficacy estimate for NIs within the model subgroup under evaluation.
Sensitivity analysis 10: no adverse events
There is uncertainty regarding the cost and health impact of adverse events associated with influenza prophylaxis. The base-case model assumes that individuals receiving prophylaxis may experience adverse events that may lead to additional medical care costs and a further loss of quality of life for amantadine. This scenario explores the impact of assuming no costs or health impacts associated with adverse events.
Sensitivity analysis 11: no withdrawals from prophylaxis
The model assumes that a proportion of patients withdraw from prophylaxis, and that patients who withdraw gain no protective benefit against influenza. This scenario assumes a withdrawal probability of 0.
Sensitivity analyses 12–16: resistance against oseltamivir
The base-case model assumes that resistance to oseltamivir is 0. These scenarios explore the impact of oseltamivir resistance on resulting cost-effectiveness estimates. Levels of resistance against amantadine are assumed to be the same as the base-case value for each scenario.
Sensitivity analysis 17: lower attack rates
Previous models of influenza prophylaxis have reported that cost-effectiveness estimates are highly sensitive to the true influenza attack rate. This scenario assumes that the attack rate is half that of the base case in each model subgroup.
Sensitivity analysis 18: higher attack rates
This scenario assumes that the attack rate is double that of the base case in each model subgroup.
Sensitivity analysis 19: use of a higher threshold for influenza activity
The base-case analysis assumes that seasonal prophylaxis will be used when the GP consultation rate for ILI is in excess of 30 per 100,000 population. 8 This scenario analysis examines the potential impact of using the previous influenza threshold of 50 consultations per 100,000 population on the cost-effectiveness of prophylaxis. This analysis draws on parameter values reported by Turner et al. 10 which was undertaken when the previous influenza threshold was implemented.
Sensitivity analysis 20: lower GP consultation rate
The base-case model assumes that the probability that an individual with symptomatic ILI consults a health-care professional is 0.25; however, this is based on a single survey and is associated with considerable uncertainty. This sensitivity analysis assumes that the probability that an individual with symptomatic ILI consults their GP is half the base-case value.
Sensitivity analysis 21: higher GP consultation rate
This sensitivity analysis assumes that the probability that an individual with symptomatic ILI consults their GP is double the base-case value.
Sensitivity analysis 22: alternative mapping function for influenza QALY loss
The base-case model uses rating scale data from clinical trials, mapped to a VAS, and subsequently mapped to TTO to generate QALY losses for the period in which an individual has influenza. This sensitivity analysis uses an alternative mapping function, converting VAS data into EQ-5D utilities.
Sensitivity analysis 23: lower QALY losses for at-risk groups
The base-case model assumes that the likely reduction in expected QALYs lost due to premature death as a result of influenza complications is the same in healthy and at-risk populations. This analysis assumes that the expected QALY loss in the at-risk group is half the value used in the base case.
Sensitivity analysis 24: complication utility decrements halved
The evidence concerning the impact of ILI complications on health outcomes is scarce and subject to considerable uncertainty. This analysis assumes a 50% reduction in utility decrements associated with ILI complications.
Sensitivity analysis 25: impact of assumptions regarding hospitalisation in uncomplicated cases
The base-case model assumes that uncomplicated ILI cases do not result in hospitalisation or death. Scenario 25 assumes that 10% of uncomplicated cases result in hospitalisation.
Sensitivity analysis 26: undiscounted cost-effectiveness estimates
Within the base-case model analysis, health outcomes were discounted at a rate of 3.5%. This analysis presents cost-effectiveness estimates without discounting.
Probabilistic sensitivity analysis
Comprehensive probabilistic sensitivity analysis was undertaken to explore the joint uncertainty in model parameters on the cost-effectiveness of each prophylaxis option. Uncertainty in model parameters was propagated through the model using Monte Carlo sampling techniques (5000 samples) to generate information on the probability that each prophylactic option is optimal (i.e. that it produces the greatest amount of net benefit). The results of the probabilistic sensitivity analysis are presented as incremental cost-effectiveness acceptability curves [see Probabilistic sensitivity analysis results (p.102), Appendix 10 and Appendix 11].
Model validation
The validity of the model was tested extensively. The model structure was reviewed throughout the model development process; the validity of key model assumptions was reviewed by clinical experts and compared with assumptions used in previous health economic models of influenza prophylaxis. At the end of the model development process, the logical consistency of the model structure and the handling of model parameters were checked by the lead modeller and also by a second modeller who was not involved in the assessment. In addition, every model parameter and its distributional characteristics were checked against the source data that were used to inform it. Finally, the expectation of probabilistic samples of each model parameter was checked against its parameter mean to identify any programming errors and any areas of non-linearity introduced through the model structure.
Cost-effectiveness results
This section presents the results of the cost-effectiveness analysis of amantadine, oseltamivir and zanamivir for the prevention of influenza. The central estimates of cost-effectiveness for each of the six model subgroups with and without previous influenza vaccination are presented below. As noted in Calculation of cost-effectiveness (see above), all central estimates of cost-effectiveness are based on expected costs and health outcomes generated by the stochastic model. The next section, One-way/multiway sensitivity analysis and scenario analysis (see p. 90), presents the results of the simple sensitivity analysis and scenario analysis to identify key determinants of the cost-effectiveness of amantadine, oseltamivir and zanamivir for the prevention of influenza. The subsequent section, Probabilistic sensitivity analysis results, presents the results of the probabilistic sensitivity analysis using CEACs.
Central estimates of cost-effectiveness
Seasonal prophylaxis model results
Tables 44–49 present the central estimates of cost-effectiveness for seasonal prophylaxis using amantadine, oseltamivir and zanamivir for the six model subgroups. The reader should note that these central estimates are based on the BNF prices of amantadine, oseltamivir and zanamivir at the time of the assessment.
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £17.72 | 0.0043 | – | – | – |
Amantadine | £56.23 | 0.0040 | – | – | Extendedly dominated |
Zanamivir | £112.15 | 0.0033 | – | – | Dominated |
Oseltamivir | £85.51 | 0.0028 | £67.79 | 0.0015 | £44,007 |
Previously vaccinated individuals | |||||
Amantadine | £78.64 | 0.0030 | – | – | Dominated |
No prophylaxis | £43.23 | 0.0030 | – | – | Dominates |
Zanamivir | £140.36 | 0.0026 | – | – | Dominated |
Oseltamivir | £115.05 | 0.0024 | £71.81 | 0.0006 | £129,357 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £29.89 | 0.0109 | – | – | – |
Amantadine | £66.92 | 0.0097 | – | – | Extendedly dominated |
Zanamivir | £121.56 | 0.0083 | – | – | Dominated |
Oseltamivir | £93.57 | 0.0071 | £63.68 | 0.0038 | £16,630 |
Previously vaccinated individuals | |||||
No prophylaxis | £51.71 | 0.0075 | – | – | – |
Amantadine | £86.84 | 0.0073 | – | – | Extendedly dominated |
Zanamivir | £147.86 | 0.0065 | – | – | Dominated |
Oseltamivir | £122.06 | 0.0061 | £70.34 | 0.0014 | £51,069 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £6.63 | 0.0020 | – | – | – |
Amantadine | £46.49 | 0.0019 | – | – | Extendedly dominated |
Zanamivir | £103.70 | 0.0015 | – | – | Extendedly dominated |
Oseltamivir | £111.09 | 0.0013 | £104.45 | 0.0007 | £147,505 |
Previously vaccinated individuals | |||||
Amantadine | £71.34 | 0.0014 | – | – | Dominated |
No prophylaxis | £35.64 | 0.0014 | – | – | Dominates |
Zanamivir | £133.74 | 0.0012 | – | – | Extendedly dominated |
Oseltamivir | £141.6 | 0.0011 | £105.9 | 0.0002 | £427,184 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £13.57 | 0.0046 | – | – | – |
Amantadine | £52.74 | 0.0042 | – | – | Extendedly dominated |
Zanamivir | £108.33 | 0.0033 | – | – | Extendedly dominated |
Oseltamivir | £115.63 | 0.0030 | £102.06 | 0.0016 | £63,552 |
Previously vaccinated individuals | |||||
Amantadine | £75.94 | 0.0032 | – | – | Dominated |
No prophylaxis | £40.39 | 0.0031 | – | – | Dominates |
Zanamivir | £137.67 | 0.0027 | – | – | Extendedly dominated |
Oseltamivir | £145.53 | 0.0025 | £105.14 | 0.0006 | £186,651 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £10.43 | 0.0048 | – | – | – |
Amantadine | £49.93 | 0.0044 | – | – | Extendedly dominated |
Zanamivir | £106.16 | 0.0035 | – | – | Extendedly dominated |
Oseltamivir | £112.80 | 0.0028 | £102.38 | 0.0021 | £49,742 |
Previously vaccinated individuals | |||||
Amantadine | £74.16 | 0.0035 | – | – | Dominated |
No prophylaxis | £38.59 | 0.0035 | – | – | Dominates |
Zanamivir | £136.02 | 0.0029 | – | – | Extendedly dominated |
Oseltamivir | £143.54 | 0.0026 | £104.95 | 0.0009 | £121,728 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £13.45 | 0.0062 | – | – | – |
Amantadine | £52.63 | 0.0057 | – | – | Extendedly dominated |
Zanamivir | £108.39 | 0.0045 | – | – | Extendedly dominated |
Oseltamivir | £114.54 | 0.0036 | £101.09 | 0.0027 | £38,098 |
Previously vaccinated individuals | |||||
No prophylaxis | £40.75 | 0.0045 | – | – | – |
Amantadine | £76.25 | 0.0044 | – | – | Extendedly dominated |
Zanamivir | £137.84 | 0.0037 | – | – | Extendedly dominated |
Oseltamivir | £145.15 | 0.0033 | £104.40 | 0.0011 | £93,763 |
Group 1: healthy children
The model results presented in Table 44 suggest that the most effective seasonal prophylaxis option for healthy children is oseltamivir, irrespective of vaccination status. Oseltamivir is expected to produce a small improvement in terms of QALY losses avoided compared with the other prophylactic strategies; however, this is not the most expensive prophylactic option. Zanamivir is less effective and more expensive than oseltamivir, irrespective of vaccination status, hence it is ruled out by simple dominance and is not included in this analysis. For healthy children who have not been previously vaccinated against influenza, amantadine is expected to be ruled out by extended dominance, as oseltamivir has a more favourable incremental cost-effectiveness ratio. For healthy children who have been previously vaccinated, amantadine is expected to be dominated by no prophylaxis. For unvaccinated children, the incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be around £44,000 per QALY gained. For healthy children who have received prior vaccination, the incremental cost-effectiveness of oseltamivir compared with no prophylaxis is estimated to be approximately £129,000 per QALY gained.
Group 2: at-risk children
The model results presented in Table 45 suggest that the most effective seasonal prophylaxis option for at-risk children is oseltamivir irrespective of whether or not they have been previously vaccinated. Again, zanamivir is expected to be less effective and more expensive than oseltamivir, hence it is ruled out of the analysis by simple dominance. Amantadine is expected to be ruled out of the analysis by extended dominance (again oseltamivir has a more favourable cost-effectiveness ratio). The incremental cost-effectiveness of oseltamivir compared with no prophylaxis is estimated to be approximately £17,000 per QALY gained in unvaccinated at-risk children and £51,000 per QALY gained in at-risk children who have previously been vaccinated against influenza.
Group 3: healthy adults
The results presented in Table 46 suggest that oseltamivir is expected to be the most effective option for seasonal prophylaxis of influenza in healthy adults. This analysis suggests that zanamivir is expected to be slightly less expensive than oseltamivir, but is ruled out by extended dominance. For unvaccinated healthy adults amantadine is ruled out of the analysis by extended dominance, while for vaccinated healthy adults amantadine is expected to be dominated by no prophylaxis. The incremental cost-effectiveness of oseltamivir compared with no prophylaxis is estimated to be approximately £148,000 per QALY gained in unvaccinated healthy adults and £427,000 per QALY gained in healthy adults who have previously been vaccinated.
Group 4: at-risk adults
Table 47 suggests that oseltamivir is expected to be the most effective option for seasonal prophylaxis in at-risk adults. As with the healthy adult model, zanamivir is expected to be ruled out by extended dominance as oseltamivir has a lower incremental cost-effectiveness ratio. For unvaccinated at-risk adults, amantadine is expected to be ruled out by extended dominance, while for vaccinated individuals, amantadine is expected to be less effective and more expensive than a policy of no prophylaxis. The incremental cost-effectiveness of oseltamivir compared with no prophylaxis is estimated to be approximately £64,000 per QALY gained in unvaccinated individuals and £187,000 per QALY gained in at-risk adults who have previously been vaccinated against influenza.
Group 5: healthy elderly
The cost-effectiveness results presented in Table 48 suggest that oseltamivir is expected to be the most effective seasonal prophylaxis option for elderly adults who are otherwise healthy. As with the working-age adult models, zanamivir is expected to be ruled out by extended dominance. Amantadine is expected to be ruled out by extended dominance for unvaccinated individuals, and is dominated by no prophylaxis in vaccinated populations. The incremental cost-effectiveness of oseltamivir compared with no prophylaxis is estimated to be around £50,000 per QALY gained in unvaccinated healthy elderly adults and around £122,000 per QALY gained in healthy elderly adults who have previously been vaccinated.
Group 6: at-risk elderly
The results presented in Table 49 suggest that oseltamivir is expected to be the most effective seasonal prophylaxis option for at-risk elderly adults. Zanamivir and amantadine are both ruled out of the analysis by extended dominance. The incremental cost-effectiveness of oseltamivir compared with amantadine is estimated to be around £38,000 per QALY gained in unvaccinated at-risk elderly individuals and £94,000 per QALY gained in at-risk elderly adults who have previously been vaccinated.
Post-exposure prophylaxis model results
Tables 50–55 present the central estimates of cost-effectiveness for post-exposure prophylaxis of influenza using amantadine, oseltamivir and zanamivir for the six model subgroups.
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £18.96 | 0.0047 | – | – | – |
Amantadine | £46.40 | 0.0039 | – | – | Extendedly dominated |
Oseltamivir | £54.35 | 0.0032 | – | – | Extendedly dominated |
Zanamivir | £61.18 | 0.0029 | £42.22 | 0.0018 | £23,225 |
Previously vaccinated individuals | |||||
No prophylaxis | £44.09 | 0.0032 | – | – | – |
Amantadine | £73.84 | 0.0030 | – | – | Extendedly dominated |
Oseltamivir | £83.30 | 0.0027 | – | – | Extendedly dominated |
Zanamivir | £91.00 | 0.0026 | £46.91 | 0.0007 | £71,648 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £32.56 | 0.0118 | – | – | – |
Amantadine | £57.55 | 0.0097 | – | – | Extendedly dominated |
Oseltamivir | £63.97 | 0.0082 | – | – | Extendedly dominated |
Zanamivir | £69.76 | 0.0073 | £37.20 | 0.0045 | £8233 |
Previously vaccinated individuals | |||||
No prophylaxis | £53.57 | 0.0081 | – | – | – |
Amantadine | £82.44 | 0.0074 | – | – | Extendedly dominated |
Oseltamivir | £91.35 | 0.0068 | – | – | Extendedly dominated |
Zanamivir | £98.67 | 0.0065 | £45.10 | 0.0016 | £27,684 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £9.17 | 0.0028 | – | – | – |
Amantadine | £38.48 | 0.0024 | – | – | Extendedly dominated |
Zanamivir | £55.19 | 0.0017 | – | – | Dominated |
Oseltamivir | £46.94 | 0.0017 | £37.77 | 0.0011 | £34,181 |
Previously vaccinated individuals | |||||
No prophylaxis | £37.36 | 0.0019 | – | – | – |
Amantadine | £67.80 | 0.0019 | – | – | Extendedly dominated |
Zanamivir | £85.67 | 0.0015 | – | – | Dominated |
Oseltamivir | £77.46 | 0.0015 | £40.10 | 0.0004 | £103,706 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £19.34 | 0.0064 | – | – | – |
Amantadine | £47.10 | 0.0055 | – | – | Extendedly dominated |
Zanamivir | £61.49 | 0.0040 | – | – | Dominated |
Oseltamivir | £53.18 | 0.0039 | £33.85 | 0.0025 | £13,459 |
Previously vaccinated individuals | |||||
No prophylaxis | £44.32 | 0.0044 | – | – | – |
Amantadine | £74.21 | 0.0041 | – | – | Extendedly dominated |
Zanamivir | £91.27 | 0.0035 | – | – | Dominated |
Oseltamivir | £83.04 | 0.0035 | £38.73 | 0.0009 | £43,970 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £17.75 | 0.0082 | – | – | – |
Amantadine | £45.76 | 0.0069 | – | – | Extendedly dominated |
Zanamivir | £60.50 | 0.0051 | – | – | Dominated |
Oseltamivir | £52.17 | 0.0050 | £34.42 | 0.0032 | £10,716 |
Previously vaccinated individuals | |||||
No prophylaxis | £43.82 | 0.0059 | – | – | – |
Amantadine | £73.59 | 0.0054 | – | – | Extendedly dominated |
Zanamivir | £90.52 | 0.0045 | – | – | Dominated |
Oseltamivir | £82.27 | 0.0045 | £38.45 | 0.0014 | £28,473 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £22.88 | 0.0106 | – | – | – |
Amantadine | £50.05 | 0.0089 | – | – | Extendedly dominated |
Zanamivir | £63.68 | 0.0065 | – | – | Dominated |
Oseltamivir | £55.33 | 0.0065 | £32.45 | 0.0041 | £7866 |
Previously vaccinated individuals | |||||
No prophylaxis | £47.50 | 0.0076 | – | – | – |
Amantadine | £76.92 | 0.0070 | – | – | Extendedly dominated |
Zanamivir | £93.37 | 0.0059 | – | – | Dominated |
Oseltamivir | £85.11 | 0.0058 | £37.60 | 0.0017 | £21,608 |
Group 1: healthy children
The model results presented in Table 50 suggest that zanamivir is expected to be the most effective option for the post-exposure prophylaxis of influenza in otherwise healthy children. In this instance, oseltamivir and amantadine are ruled out of the analysis by extended dominance. The incremental cost-effectiveness of zanamivir versus no prophylaxis is estimated to be £23,000 per QALY gained for unvaccinated healthy children and around £72,000 in vaccinated healthy children.
The reader should note that oseltamivir is the only licensed prophylactic in children under the age of 5 years; the incremental cost-effectiveness ratio for oseltamivir versus no prophylaxis is expected to be around £24,000 per QALY gained and £74,000 per QALY gained in unvaccinated and vaccinated groups respectively.
Group 2: at-risk children
The cost-effectiveness results presented in Table 51 suggest that zanamivir is expected to be the most effective option for the post-exposure prophylaxis of influenza in at-risk children. Oseltamivir and amantadine are expected to be ruled out of the analysis by extended dominance for unvaccinated and vaccinated subgroups. The incremental cost-effectiveness of zanamivir versus no prophylaxis is estimated to be around £8000 per QALY gained in unvaccinated at-risk children and approximately £28,000 per QALY gained in vaccinated at-risk children.
For at-risk children under the age of 5 years, the incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be around £9000 per QALY gained for unvaccinated at-risk children and around £29,000 per QALY gained for vaccinated at-risk children.
Group 3: healthy adults
The cost-effectiveness estimates presented in Table 52 suggest that oseltamivir is expected to be the most effective option for the post-exposure prophylaxis of influenza in healthy adults. Within this subgroup, zanamivir is expected to be dominated by oseltamivir irrespective of vaccination status. Amantadine is expected to be ruled out by extended dominance. The incremental cost-effectiveness of oseltamivir versus no prophylaxis is estimated to be £34,000 per QALY gained for vaccinated healthy adults and around £104,000 per QALY gained for unvaccinated healthy adults.
Group 4: at-risk adults
Table 53 suggests that oseltamivir is expected to be the most effective option for the post-exposure prophylaxis of influenza in at-risk adults. Again, zanamivir is expected to be dominated by oseltamivir irrespective of vaccination status. Amantadine is again expected to be ruled out by extended dominance. The incremental cost-effectiveness of oseltamivir versus no prophylaxis is estimated to be around £13,000 per QALY gained for unvaccinated at-risk adults and £44,000 per QALY gained for previously vaccinated at-risk adults.
Group 5: healthy elderly
Table 54 suggests that oseltamivir is expected to be the most effective option for the post-exposure prophylaxis of influenza in otherwise healthy elderly adults. Zanamivir is again expected to be dominated by oseltamivir and is hence ruled out of the analysis. Amantadine is expected to be ruled out of the analysis by extended dominance. The incremental cost-effectiveness of oseltamivir versus no prophylaxis is estimated to be around £11,000 per QALY gained for unvaccinated healthy elderly individuals and around £28,000 per QALY gained for at-risk elderly who have previously been vaccinated against influenza.
Group 6: at-risk elderly
The model results presented in Table 55 suggest that oseltamivir is expected to be the most effective option for the post-exposure prophylaxis of influenza in at-risk elderly individuals. Zanamivir is expected to be dominated by oseltamivir and is ruled out of the analysis. Amantadine is expected to be ruled out by extended dominance. The incremental cost-effectiveness of oseltamivir versus no prophylaxis is estimated to be around £8000 per QALY for vaccinated at-risk elderly individuals and around £22,000 per QALY gained for at-risk elderly individuals who have previously been vaccinated.
One-way/multiway sensitivity analysis and scenario analysis
This section presents one-way and multiway sensitivity analysis to explore the impact of changing parameter assumptions on the incremental cost-effectiveness of amantadine, oseltamivir and zanamivir for the prevention of influenza. Descriptions of these scenarios are presented in Uncertainty analysis (p. 82).
Sensitivity analysis – cost-effectiveness results including proposed reduction in the price of zanamivir
Tables 56–67 present the results of the model incorporating the proposed price reduction for zanamivir. The reader should note that as these results are based on the stochastic model, they are subject to a small degree of Monte Carlo sampling error.
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £17.71 | 0.0043 | – | – | – |
Amantadine | £56.20 | 0.0040 | – | – | Extendedly dominated |
Zanamivir | £87.59 | 0.0033 | – | – | Dominated |
Oseltamivir | £85.49 | 0.0028 | £67.78 | 0.0015 | £43,870 |
Previously vaccinated individuals | |||||
Amantadine | £78.64 | 0.0030 | – | – | Dominated |
No prophylaxis | £43.22 | 0.0030 | – | – | Dominates |
Zanamivir | £115.80 | 0.0026 | – | – | Dominated |
Oseltamivir | £115.05 | 0.0024 | £71.82 | 0.0006 | £129,888 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £29.62 | 0.0109 | – | – | – |
Amantadine | £66.68 | 0.0097 | – | – | Extendedly dominated |
Zanamivir | £96.83 | 0.0084 | – | – | Dominated |
Oseltamivir | £93.38 | 0.0071 | £63.76 | 0.0038 | £16,598 |
Previously vaccinated individuals | |||||
No prophylaxis | £51.53 | 0.0075 | – | – | – |
Amantadine | £86.66 | 0.0074 | – | – | Extendedly dominated |
Zanamivir | £123.14 | 0.0066 | – | – | Dominated |
Oseltamivir | £121.90 | 0.0061 | £70.37 | 0.0014 | £50,902 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £6.57 | 0.0020 | – | – | – |
Amantadine | £46.40 | 0.0019 | – | – | Extendedly dominated |
Zanamivir | £79.09 | 0.0015 | – | – | Extendedly dominated |
Oseltamivir | £111.04 | 0.0013 | £104.46 | 0.0007 | £147,083 |
Previously vaccinated individuals | |||||
Amantadine | £71.26 | 0.0014 | – | – | Dominated |
No prophylaxis | £35.58 | 0.0014 | – | – | Dominates |
Zanamivir | £109.11 | 0.0012 | – | – | Extendedly dominated |
Oseltamivir | £141.56 | 0.0011 | £105.98 | 0.0002 | £427,802 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £13.70 | 0.0046 | – | – | – |
Amantadine | £52.83 | 0.0042 | – | – | Extendedly dominated |
Zanamivir | £83.85 | 0.0033 | £70.15 | 0.0013 | £53,159 |
Oseltamivir | £115.69 | 0.0030 | £31.84 | 0.0003 | £108,379 |
Previously vaccinated individuals | |||||
Amantadine | £76.03 | 0.0031 | – | – | Dominated |
No prophylaxis | £40.47 | 0.0031 | – | – | Dominates |
Zanamivir | £113.17 | 0.0026 | £72.70 | 0.0005 | £157,216 |
Oseltamivir | £145.58 | 0.0025 | £32.41 | 0.0001 | £313,592 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £10.48 | 0.0048 | – | – | – |
Amantadine | £49.98 | 0.0044 | – | – | Extendedly dominated |
Zanamivir | £81.63 | 0.0035 | – | – | Extendedly dominated |
Oseltamivir | £112.82 | 0.0028 | £102.34 | 0.0021 | £49,590 |
Previously vaccinated individuals | |||||
Amantadine | £74.21 | 0.0035 | – | – | Dominated |
No prophylaxis | £38.63 | 0.0035 | – | – | Dominates |
Zanamivir | £111.48 | 0.0029 | – | – | Extendedly dominated |
Oseltamivir | £143.55 | 0.0026 | £104.92 | 0.0009 | £120,292 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £13.46 | 0.0062 | – | – | – |
Amantadine | £52.64 | 0.0057 | – | – | Extendedly dominated |
Zanamivir | £83.81 | 0.0045 | – | – | Extendedly dominated |
Oseltamivir | £114.53 | 0.0036 | £101.07 | 0.0027 | £37,968 |
Previously vaccinated individuals | |||||
No prophylaxis | £40.75 | 0.0045 | – | – | – |
Amantadine | £76.26 | 0.0044 | – | – | Extendedly dominated |
Zanamivir | £113.26 | 0.0037 | – | – | Extendedly dominated |
Oseltamivir | £145.15 | 0.0033 | £104.40 | 0.0011 | £93,581 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £18.92 | 0.0047 | – | – | – |
Amantadine | £46.38 | 0.0040 | – | – | Extendedly dominated |
Oseltamivir | £54.34 | 0.0032 | – | – | Dominated |
Zanamivir | £52.98 | 0.0029 | £34.06 | 0.0018 | £18,717 |
Previously vaccinated individuals | |||||
No prophylaxis | £44.04 | 0.0032 | – | – | – |
Amantadine | £73.81 | 0.0030 | – | – | Extendedly dominated |
Oseltamivir | £83.28 | 0.0027 | – | – | Dominated |
Zanamivir | £82.79 | 0.0026 | £38.75 | 0.0007 | £59,412 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £32.38 | 0.0119 | – | – | – |
Amantadine | £57.38 | 0.0098 | – | – | Extendedly dominated |
Oseltamivir | £63.82 | 0.0082 | – | – | Dominated |
Zanamivir | £61.45 | 0.0073 | £29.07 | 0.0045 | £6390 |
Previously vaccinated individuals | |||||
No prophylaxis | £53.42 | 0.0081 | – | – | – |
Amantadine | £82.29 | 0.0075 | – | – | Extendedly dominated |
Oseltamivir | £91.22 | 0.0068 | – | – | Dominated |
Zanamivir | £90.37 | 0.0065 | £36.96 | 0.0016 | £22,663 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £9.23 | 0.0028 | – | – | – |
Amantadine | £38.54 | 0.0024 | – | – | Extendedly dominated |
Zanamivir | £47.03 | 0.0017 | – | – | Dominated |
Oseltamivir | £46.96 | 0.0017 | £37.73 | 0.0011 | £34,099 |
Previously vaccinated individuals | |||||
No prophylaxis | £37.40 | 0.0019 | – | – | – |
Amantadine | £67.84 | 0.0019 | – | – | Extendedly dominated |
Zanamivir | £77.51 | 0.0015 | – | – | Dominated |
Oseltamivir | £77.49 | 0.0015 | £40.09 | 0.0004 | £103,573 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £19.18 | 0.0064 | – | – | – |
Amantadine | £46.94 | 0.0055 | – | – | Extendedly dominated |
Zanamivir | £53.20 | 0.0040 | – | – | Dominated |
Oseltamivir | £53.09 | 0.0039 | £33.92 | 0.0025 | £13,539 |
Previously vaccinated individuals | |||||
No prophylaxis | £44.20 | 0.0044 | – | – | – |
Amantadine | £74.10 | 0.0041 | – | – | Extendedly dominated |
Zanamivir | £82.99 | 0.0035 | – | – | Dominated |
Oseltamivir | £82.96 | 0.0035 | £38.75 | 0.0009 | £44,163 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £17.70 | 0.0082 | – | – | – |
Amantadine | £45.74 | 0.0069 | – | – | Extendedly dominated |
Zanamivir | £52.28 | 0.0051 | – | – | Dominated |
Oseltamivir | £52.14 | 0.0050 | £34.44 | 0.0032 | £10,734 |
Previously vaccinated individuals | |||||
No prophylaxis | £43.78 | 0.0059 | – | – | – |
Amantadine | £73.56 | 0.0054 | – | – | Extendedly dominated |
Zanamivir | £82.30 | 0.0045 | – | – | Dominated |
Oseltamivir | £82.24 | 0.0045 | £38.46 | 0.0013 | £28,608 |
Option | Costs | QALYs lost | Incremental cost | Incremental QALYs | Incremental cost per QALY gained |
---|---|---|---|---|---|
Unvaccinated individuals | |||||
No prophylaxis | £22.75 | 0.0106 | – | – | – |
Amantadine | £49.95 | 0.0089 | – | – | Extendedly dominated |
Zanamivir | £55.39 | 0.0065 | – | – | Dominated |
Oseltamivir | £55.24 | 0.0064 | £32.49 | 0.0041 | £7892 |
Previously vaccinated individuals | |||||
No prophylaxis | £47.41 | 0.0075 | – | – | – |
Amantadine | £76.84 | 0.0069 | – | – | Extendedly dominated |
Zanamivir | £85.10 | 0.0058 | – | – | Dominated |
Oseltamivir | £85.04 | 0.0058 | £37.63 | 0.0017 | £21,749 |
Post-exposure prophylaxis
Results for post-exposure prophylaxis are presented in Tables 62–67.
Table 68 summarises the ICERs presented in the base-case analysis and those including the proposed reduction in the price of zanamivir.
Population | Base case (incremental cost per QALY gained) | Price reduction for zanamivir (incremental cost per QALY gained) | ||||
---|---|---|---|---|---|---|
Amantadine | Zanamivir | Oseltamivir | Amantadine | Zanamivir | Oseltamivir | |
Seasonal prophylaxis | ||||||
Healthy children | ||||||
Unvaccinated | Ext dom | Dom | £44,007 | Ext dom | Dom | £43,870 |
Vaccinated | Dom | Dom | £129,357 | Dom | Dom | £129,888 |
At-risk children | ||||||
Unvaccinated | Ext dom | Dom | £16,630 | Ext dom | Dom | £16,598 |
Vaccinated | Ext dom | Dom | £51,069 | Ext dom | Dom | £50,902 |
Healthy adults | ||||||
Unvaccinated | Ext dom | Ext dom | £147,505 | Ext dom | Ext dom | £147,083 |
Vaccinated | Dom | Ext dom | £427,184 | Dom | Ext dom | £427,802 |
At-risk adults | ||||||
Unvaccinated | Ext dom | Ext dom | £63,552 | Ext dom | £53,159 | £108,379 |
Vaccinated | Dom | Ext dom | £186,651 | Dom | £157,216 | £313,592 |
Healthy elderly | ||||||
Unvaccinated | Ext dom | Ext dom | £49,742 | Ext dom | Ext dom | £49,590 |
Vaccinated | Dom | Ext dom | £121,728 | Dom | Ext dom | £120,292 |
At-risk elderly | ||||||
Unvaccinated | Ext dom | Ext dom | £38,098 | Ext dom | Ext dom | £37,968 |
Vaccinated | Ext dom | Ext dom | £93,763 | Ext dom | Ext dom | £93,581 |
Post-exposure prophylaxis | ||||||
Healthy children | ||||||
Unvaccinated | Ext dom | £23,225 | Ext dom | Ext dom | £18,717 | Dom |
Vaccinated | Ext dom | £71,648 | Ext dom | Ext dom | £59,412 | Dom |
At-risk children | ||||||
Unvaccinated | Ext dom | £8233 | Ext dom | Ext dom | £6390 | Dom |
Vaccinated | Ext dom | £27,684 | Ext dom | Ext dom | £22,663 | Dom |
Healthy adults | ||||||
Unvaccinated | Ext dom | Dom | £34,181 | Ext dom | Dom | £34,099 |
Vaccinated | Ext dom | Dom | £103,706 | Ext dom | Dom | £103,573 |
At-risk adults | ||||||
Unvaccinated | Ext dom | Dom | £13,459 | Ext dom | Dom | £13,539 |
Vaccinated | Ext dom | Dom | £43,970 | Ext dom | Dom | £44,163 |
Healthy elderly | ||||||
Unvaccinated | Ext dom | Dom | £10,716 | Ext dom | Dom | £10,734 |
Vaccinated | Ext dom | Dom | £28,473 | Ext dom | Dom | £28,608 |
At-risk elderly | ||||||
Unvaccinated | Ext dom | Dom | £7866 | Ext dom | Dom | £7892 |
Vaccinated | Ext dom | Dom | £21,608 | Ext dom | Dom | £21,749 |
The summary of cost-effectiveness results presented in Table 68 shows that the proposed price reduction has no impact on the majority of economic comparisons presented in the base-case analysis. In terms of seasonal prophylaxis, the cost-effectiveness of zanamivir is no longer ruled out by extended dominance in at-risk adults; however, the incremental cost-effectiveness ratio for zanamivir versus no prophylaxis remains in excess of £50,000 per QALY gained for these comparisons. In terms of the post-exposure prophylaxis of influenza, the price reduction has no impact on the adult and elderly subgroup analyses, as zanamivir consistently remains dominated by oseltamivir. The proposed price reduction is, however, expected to lead to an improvement in the cost-effectiveness of zanamivir for otherwise healthy and at-risk children. For unvaccinated healthy children, the reduction in the price of zanamivir is expected to result in a reduction in the cost-effectiveness of zanamivir versus no prophylaxis from £23,000 per QALY gained to £19,000 per QALY gained. The cost-effectiveness of zanamivir in vaccinated, otherwise healthy children is expected to be in excess of £59,000 per QALY gained. For unvaccinated at-risk children, the lower price for zanamivir is expected to lead to an improvement in the cost-effectiveness of zanamivir versus no prophylaxis from £8000 per QALY gained to £6000 per QALY gained. For vaccinated at-risk children, the cost-effectiveness of zanamivir is improved from £28,000 per QALY gained to £23,000 per QALY gained.
One-way sensitivity analysis and scenario analysis results
Healthy children
The results of the simple sensitivity analysis for the healthy children subgroup are presented in Table 69.
Scenario | Unvaccinated | Vaccinated | ||||
---|---|---|---|---|---|---|
Amantadine | Zanamivir | Oseltamivir | Amantadine | Zanamivir | Oseltamivir | |
Seasonal prophylaxis | ||||||
Base case (stochastic model) | Ext dom | Dom | £44,007 | Dom | Dom | £129,357 |
1. Price reduction zanamivir | Ext dom | Dom | £43,870 | Dom | Dom | £129,888 |
2. Base-case deterministic model | Ext dom | Dom | £42,244 | Dom | Dom | £124,523 |
3. Oseltamivir as suspension | Ext dom | Dom | £42,244 | Dom | Dom | £124,523 |
4. Multiple prescriptions | Ext dom | Dom | £42,244 | Dom | Dom | £124,523 |
5. Vaccine efficacy = 50% | Ext dom | Dom | £42,244 | Dom | Dom | £88,526 |
6. Vaccine efficacy = 25% | Ext dom | Dom | £42,244 | Ext dom | Dom | £57,672 |
7. 100% protection over influenza season | Ext dom | Dom | £42,244 | Ext dom | Dom | £124,523 |
8. No antiviral treatment | Ext dom | Dom | £42,244 | Dom | Dom | £124,523 |
9. Best-case efficacy for NIs | Ext dom | Dom | £42,244 | Dom | Dom | £124,523 |
10. No adverse events | Ext dom | Dom | £42,244 | Ext dom | Dom | £124,523 |
11. No withdrawals | Ext dom | Dom | £41,319 | Dom | Dom | £121,952 |
12. 10% resistance for oseltamivir | Ext dom | Dom | £47,387 | Dom | Dom | £138,807 |
13. 20% resistance for oseltamivir | Ext dom | Dom | £53,815 | Dom | Dom | £156,663 |
14. 30% resistance for oseltamivir | Ext dom | Dom | £62,079 | Dom | Dom | £179,620 |
15. 40% resistance for oseltamivir | Ext dom | £573,163 | £73,099 | Dom | £1,599,296 | £210,229 |
16. 50% resistance for oseltamivir | Ext dom | £117,218 | £88,526 | Dom | £332,782 | £253,083 |
17. Attack rates halved | Ext dom | Dom | £88,526 | Dom | Dom | £253,083 |
18. Attack rates doubled | Ext dom | Dom | £19,104 | Ext dom | Dom | £60,243 |
19. Higher influenza threshold | Ext dom | Dom | £70,476 | Dom | Dom | £202,944 |
20. GP consultation rates halved | Ext dom | Dom | £42,560 | Dom | Dom | £124,839 |
21. GP consultation rates doubled | Ext dom | Dom | £41,613 | Dom | Dom | £123,891 |
22. VAS to EQ-5D mapping function | Ext dom | Dom | £38,980 | Dom | Dom | £114,902 |
23. QALY loss for at-risk halved | Ext dom | Dom | £42,244 | Dom | Dom | £124,523 |
24. Complication disutilities halved | Ext dom | Dom | £43,102 | Dom | Dom | £127,052 |
25. 10% uncomplicated hospitalised | Ext dom | Dom | £22,513 | Dom | Dom | £104,791 |
26. Undiscounted | Ext dom | Dom | £35,111 | Dom | Dom | £103,495 |
Post-exposure prophylaxis | ||||||
Base case (stochastic model) | Ext dom | £23,225 | Ext dom | Ext dom | £71,648 | Ext dom |
1. Price reduction zanamivir | Ext dom | £18,717 | Dom | Ext dom | £59,412 | Dom |
2. Base-case deterministic model | Ext dom | £23,217 | Ext dom | Ext dom | £71,668 | Ext dom |
3. Oseltamivir as suspension | Ext dom | £23,217 | Ext dom | Ext dom | £71,668 | Ext dom |
4. Multiple prescriptions | Ext dom | £19,634 | £11,322 | Ext dom | £61,717 | £38,627 |
5. Vaccine efficacy = 50% | Ext dom | £23,217 | Ext dom | Ext dom | £50,471 | Ext dom |
6. Vaccine efficacy = 25% | Ext dom | £23,217 | Ext dom | Ext dom | £32,302 | Ext dom |
7. 100% protection over influenza season | Ext dom | £23,217 | Ext dom | Ext dom | £71,668 | Ext dom |
8. No antiviral treatment | Ext dom | £23,217 | Ext dom | Ext dom | £71,668 | Ext dom |
9. Best-case efficacy for NIs | Ext dom | £630,864 | £18,875 | Ext dom | £1,759,576 | £59,607 |
10. No adverse events | Ext dom | £23,217 | Ext dom | Ext dom | £71,668 | Ext dom |
11. No withdrawals | Ext dom | £22,863 | Ext dom | Ext dom | £70,684 | Ext dom |
12. 10% resistance for oseltamivir | Ext dom | £23,217 | Ext dom | Ext dom | £71,668 | Ext dom |
13. 20% resistance for oseltamivir | Ext dom | £23,217 | Ext dom | Ext dom | £71,668 | Ext dom |
14. 30% resistance for oseltamivir | Ext dom | £23,217 | Ext dom | Ext dom | £71,668 | Ext dom |
15. 40% resistance for oseltamivir | Ext dom | £23,217 | Ext dom | Ext dom | £71,668 | Ext dom |
16. 50% resistance for oseltamivir | Ext dom | £23,217 | Dom | Ext dom | £71,668 | Ext dom |
17. Attack rates halved | Ext dom | £50,471 | Ext dom | Ext dom | £147,374 | Ext dom |
18. Attack rates doubled | Ext dom | £9590 | Ext dom | Ext dom | £33,816 | Ext dom |
19. Higher influenza threshold | Ext dom | £23,217 | Ext dom | Ext dom | £71,668 | Ext dom |
20. GP consultation rates halved | Ext dom | £23,533 | Ext dom | Ext dom | £71,984 | Ext dom |
21. GP consultation rates doubled | Ext dom | £22,585 | Ext dom | Ext dom | £71,037 | Ext dom |
22. VAS to EQ-5D mapping function | Ext dom | £21,423 | Ext dom | Ext dom | £66,131 | Ext dom |
23. QALY loss for at-risk halved | Ext dom | £23,217 | Ext dom | Ext dom | £71,668 | Ext dom |
24. Complication disutilities halved | Ext dom | £23,688 | Ext dom | Ext dom | £73,124 | Ext dom |
25. 10% uncomplicated hospitalised | Dom | £3485 | Dom | Ext dom | £51,937 | Ext dom |
26. Undiscounted | Ext dom | £19,296 | Ext dom | Ext dom | £59,566 | Ext dom |
The simple sensitivity analysis results presented in Table 69 suggest that the base-case seasonal prophylaxis cost-effectiveness estimates are sensitive to assumptions regarding influenza attack rates, the level of resistance against oseltamivir, vaccine efficacy, the threshold used to describe when influenza is circulating in the community (particularly the duration of the influenza season), the risk of hospitalisation in uncomplicated cases and the discount rate. Amantadine and zanamivir as seasonal prophylaxis remain dominated across almost all scenarios. The cost-effectiveness estimates for post-exposure prophylaxis are sensitive to the influenza attack rate, the use of multiple prescriptions of prophylaxis at a single GP visit, vaccine efficacy, assumptions regarding the relative effectiveness of oseltamivir and zanamivir and the risk of hospitalisation in uncomplicated cases. Amantadine and oseltamivir as post-exposure prophylaxis remain dominated or extendedly dominated by zanamivir in the majority of the scenarios presented for healthy children.
At-risk children
The results of the simple sensitivity analysis for the at-risk children subgroup are presented in Table 70.
Scenario | Unvaccinated | Vaccinated | ||||
---|---|---|---|---|---|---|
Amantadine | Zanamivir | Oseltamivir | Amantadine | Zanamivir | Oseltamivir | |
Seasonal prophylaxis | ||||||
Base case (stochastic model) | Ext dom | Dom | £16,630 | Ext dom | Dom | £51,069 |
1. Price reduction zanamivir | Ext dom | Dom | £16,598 | Ext dom | Dom | £50,902 |
2. Base-case deterministic model | Ext dom | Dom | £15,882 | Ext dom | Dom | £48,943 |
3. Oseltamivir as suspension | Ext dom | Dom | £15,882 | Ext dom | Dom | £48,943 |
4. Multiple prescriptions | Ext dom | Dom | £15,882 | Ext dom | Dom | £48,943 |
5. Vaccine efficacy = 50% | Ext dom | Dom | £15,882 | Ext dom | Dom | £34,479 |
6. Vaccine efficacy = 25% | Ext dom | Dom | £15,882 | Ext dom | Dom | £22,081 |
7. 100% protection over influenza season | Ext dom | Dom | £15,882 | Ext dom | Dom | £48,943 |
8. No antiviral treatment | Ext dom | Dom | £15,595 | Ext dom | Dom | £48,118 |
9. Best-case efficacy for NIs | Ext dom | Dom | £15,882 | Ext dom | Dom | £48,943 |
10. No adverse events | Ext dom | Dom | £15,882 | Ext dom | Dom | £48,943 |
11. No withdrawals | Ext dom | Dom | £15,510 | Ext dom | Dom | £47,910 |
12. 10% resistance for oseltamivir | Ext dom | Dom | £17,948 | Ext dom | Dom | £54,683 |
13. 20% resistance for oseltamivir | Ext dom | Dom | £20,531 | Ext dom | Dom | £61,857 |
14. 30% resistance for oseltamivir | Ext dom | Dom | £23,852 | Ext dom | Dom | £71,082 |
15. 40% resistance for oseltamivir | Ext dom | £229,215 | £28,280 | Ext dom | £641,536 | £83,381 |
16. 50% resistance for oseltamivir | £29,840 | £46,007 | £42,085 | Ext dom | £132,625 | £100,601 |
17. Attack rates halved | Ext dom | Dom | £34,479 | Dom | Dom | £100,601 |
18. Attack rates doubled | Ext dom | Dom | £6583 | Ext dom | Dom | £23,114 |
19. Higher influenza threshold | Ext dom | Dom | £27,226 | Ext dom | Dom | £80,454 |
20. GP consultation rates halved | Ext dom | Dom | £15,863 | Ext dom | Dom | £48,653 |
21. GP consultation rates doubled | Ext dom | Dom | £15,921 | Ext dom | Dom | £49,538 |
22. VAS to EQ-5D mapping function | Ext dom | Dom | £16,932 | Ext dom | Dom | £52,179 |
23. QALY loss for at-risk halved | Ext dom | Dom | £17,821 | Ext dom | Dom | £54,920 |
24. Complication disutilities halved | Ext dom | Dom | £16,041 | Ext dom | Dom | £49,435 |
25. 10% uncomplicated hospitalised | Ext dom | Dom | £8341 | Ext dom | Dom | £41,402 |
26. Undiscounted | Ext dom | Dom | £11,658 | Ext dom | Dom | £35,925 |
Post-exposure prophylaxis | ||||||
Base case (stochastic model) | Ext dom | £8233 | Ext dom | Ext dom | £27,684 | Ext dom |
1. Price reduction zanamivir | Ext dom | £6390 | Dom | Ext dom | £22,663 | Dom |
2. Base-case deterministic model | Ext dom | £8236 | Ext dom | Ext dom | £27,705 | Ext dom |
3. Oseltamivir as suspension | Ext dom | £8236 | Ext dom | Ext dom | £27,705 | Ext dom |
4. Multiple prescriptions | £2991 | £6797 | £4075 | Ext dom | £23,706 | £14,428 |
5. Vaccine efficacy = 50% | Ext dom | £8236 | Ext dom | Ext dom | £19,187 | Ext dom |
6. Vaccine efficacy = 25% | Ext dom | £8236 | Ext dom | Ext dom | £11,887 | Ext dom |
7. 100% protection over influenza season | Ext dom | £8236 | Ext dom | Ext dom | £27,705 | Ext dom |
8. No antiviral treatment | Ext dom | £8073 | Ext dom | Ext dom | £27,226 | Ext dom |
9. Best-case efficacy for NIs | Ext dom | £252,401 | £6491 | Ext dom | £705,940 | £22,858 |
10. No adverse events | Ext dom | £8236 | Ext dom | Ext dom | £27,705 | Ext dom |
11. No withdrawals | Ext dom | £8094 | Ext dom | Ext dom | £27,310 | Ext dom |
12. 10% resistance for oseltamivir | Ext dom | £8236 | Ext dom | Ext dom | £27,705 | Ext dom |
13. 20% resistance for oseltamivir | Ext dom | £8236 | Ext dom | Ext dom | £27,705 | Ext dom |
14. 30% resistance for oseltamivir | Ext dom | £8236 | Ext dom | Ext dom | £27,705 | Ext dom |
15. 40% resistance for oseltamivir | Ext dom | £8236 | Ext dom | Ext dom | £27,705 | Ext dom |
16. 50% resistance for oseltamivir | Ext dom | £8236 | Dom | Ext dom | £27,705 | Dom |
17. Attack rates halved | Ext dom | £19,187 | Ext dom | Ext dom | £58,125 | Ext dom |
18. Attack rates doubled | Ext dom | £2761 | Ext dom | Ext dom | £12,495 | Ext dom |
19. Higher influenza threshold | Ext dom | £8236 | Ext dom | Ext dom | £27,705 | Ext dom |
20. GP consultation rates halved | Ext dom | £8280 | Ext dom | Ext dom | £27,589 | Ext dom |
21. GP consultation rates doubled | Ext dom | £8147 | Ext dom | Ext dom | £27,943 | Ext dom |
22. VAS to EQ-5D mapping function | Ext dom | £8781 | Ext dom | Ext dom | £29,537 | Ext dom |
23. QALY loss for at-risk halved | Ext dom | £9242 | Ext dom | Ext dom | £31,088 | Ext dom |
24. Complication disutilities halved | Ext dom | £8319 | Ext dom | Ext dom | £27,983 | Ext dom |
25. 10% uncomplicated hospitalised | Dom | £696 | Dom | Ext dom | £20,165 | Ext dom |
26. Undiscounted | Ext dom | £6045 | Ext dom | Ext dom | £20,336 | Ext dom |
The simple sensitivity analysis results presented in Table 70 suggest that the base-case seasonal prophylaxis cost-effectiveness estimates for at-risk children are also sensitive to influenza attack rates, the level of resistance against oseltamivir, vaccine efficacy, the threshold used to describe when influenza is circulating in the community, the risk of hospitalisation in uncomplicated cases and the discount rate. Amantadine and zanamivir remain dominated by oseltamivir in almost every scenario in this subgroup. The cost-effectiveness estimates for post-exposure prophylaxis are also sensitive to the influenza attack rate, the use of multiple prescriptions of prophylaxis at a single GP visit, vaccine efficacy, assumptions regarding the relative effectiveness of oseltamivir and zanamivir and the risk of hospitalisation in uncomplicated cases. Amantadine and oseltamivir post-exposure prophylaxis are generally dominated or extendedly dominated by zanamivir within the at-risk children subgroup.
Healthy adults
The results of the simple sensitivity analysis for the healthy adult subgroup are presented in Table 71.
Scenario | Unvaccinated | Vaccinated | ||||
---|---|---|---|---|---|---|
Amantadine | Zanamivir | Oseltamivir | Amantadine | Zanamivir | Oseltamivir | |
Seasonal prophylaxis | ||||||
Base case (stochastic model) | Ext dom | Ext dom | £147,505 | Dom | Ext dom | £427,184 |
1. Price reduction zanamivir | Ext dom | Ext dom | £147,083 | Dom | Ext dom | £427,802 |
2. Base-case deterministic model | Ext dom | Ext dom | £141,659 | Dom | Ext dom | £410,832 |
3. Oseltamivir as suspension | Ext dom | Dom | £119,456 | Dom | Dom | £347,397 |
4. Multiple prescriptions | Ext dom | Ext dom | £141,659 | Dom | Ext dom | £410,832 |
5. Vaccine efficacy = 50% | Ext dom | Ext dom | £141,659 | Dom | Ext dom | £286,598 |
6. Vaccine efficacy = 25% | Ext dom | Ext dom | £141,659 | Dom | Ext dom | £189,972 |
7. 100% protection over influenza season | Ext dom | Ext dom | £141,659 | Dom | Ext dom | £410,832 |
8. No antiviral treatment | Ext dom | Ext dom | £141,659 | Dom | Ext dom | £410,832 |
9. Best-case efficacy for NIs | Ext dom | Ext dom | £141,659 | Dom | Ext dom | £410,832 |
10. No adverse events | Ext dom | Ext dom | £141,659 | Ext dom | Ext dom | £410,832 |
11. No withdrawals | Ext dom | Ext dom | £138,760 | Dom | Ext dom | £402,550 |
12. 10% resistance for oseltamivir | Ext dom | Ext dom | £157,763 | Dom | Ext dom | £456,845 |
13. 20% resistance for oseltamivir | Ext dom | Ext dom | £177,894 | Dom | Ext dom | £514,360 |
14. 30% resistance for oseltamivir | Ext dom | Ext dom | £203,776 | Dom | Ext dom | £588,309 |
15. 40% resistance for oseltamivir | Ext dom | £210,381 | Dom | Dom | £607,181 | Dom |
16. 50% resistance for oseltamivir | Ext dom | £210,381 | Dom | Dom | £607,181 | Dom |
17. Attack rates halved | Dom | Ext dom | £286,598 | Dom | Ext dom | £824,945 |
18. Attack rates doubled | Ext dom | Ext dom | £69,189 | Dom | Ext dom | £203,776 |
19. Higher influenza threshold | Ext dom | Ext dom | £230,072 | Dom | Ext dom | £663,441 |
20. GP consultation rates halved | Ext dom | Ext dom | £141,921 | Dom | Ext dom | £411,095 |
21. GP consultation rates doubled | Ext dom | Ext dom | £141,133 | Dom | Ext dom | £410,307 |
22. VAS to EQ-5D mapping function | Ext dom | Ext dom | £133,013 | Dom | Ext dom | £385,758 |
23. QALY loss for at-risk halved | Ext dom | Ext dom | £141,659 | Dom | Ext dom | £410,832 |
24. Complication disutilities halved | Ext dom | Ext dom | £143,075 | Dom | Ext dom | £414,940 |
25. 10% uncomplicated hospitalised | Ext dom | Dom | £110,466 | Dom | Ext dom | £379,639 |
26. Undiscounted | Ext dom | Ext dom | £119,801 | Dom | Ext dom | £347,440 |
Post-exposure prophylaxis | ||||||
Base case (stochastic model) | Ext dom | Dom | £34,181 | Ext dom | Dom | £103,706 |
1. Price reduction zanamivir | Ext dom | Dom | £34,099 | Ext dom | Dom | £103,573 |
2. Base-case deterministic model | Ext dom | Dom | £34,113 | Ext dom | Dom | £103,558 |
3. Oseltamivir as suspension | Ext dom | Dom | £34,113 | Ext dom | Dom | £103,558 |
4. Multiple prescriptions | Ext dom | Dom | £17,161 | Ext dom | Dom | £55,124 |
5. Vaccine efficacy = 50% | Ext dom | Dom | £34,113 | Ext dom | Dom | £71,507 |
6. Vaccine efficacy = 25% | Ext dom | Dom | £34,113 | Ext dom | Dom | £46,578 |
7. 100% protection over influenza season | Ext dom | Dom | £34,113 | Ext dom | Dom | £103,558 |
8. No antiviral treatment | Ext dom | Dom | £34,113 | Ext dom | Dom | £103,558 |
9. Best-case efficacy for NIs | Ext dom | £1,032,921 | £34,113 | Ext dom | £2,957,296 | £103,558 |
10. No adverse events | Ext dom | Dom | £34,113 | Ext dom | Dom | £103,558 |
11. No withdrawals | Ext dom | Dom | £33,365 | Ext dom | Dom | £101,422 |
12. 10% resistance for oseltamivir | Ext dom | £86,714 | £38,268 | Ext dom | £253,847 | £115,429 |
13. 20% resistance for oseltamivir | Ext dom | £42,326 | Ext dom | Ext dom | £127,024 | Ext dom |
14. 30% resistance for oseltamivir | Ext dom | £42,326 | Ext dom | Ext dom | £127,024 | Ext dom |
15. 40% resistance for oseltamivir | Ext dom | £42,326 | Ext dom | Ext dom | £127,024 | Ext dom |
16. 50% resistance for oseltamivir | Ext dom | £42,326 | Ext dom | Ext dom | £127,024 | Ext dom |
17. Attack rates halved | Ext dom | Dom | £71,507 | Dom | Dom | £210,397 |
18. Attack rates doubled | Ext dom | Dom | £15,416 | Ext dom | Dom | £50,139 |
19. Higher influenza threshold | Ext dom | Dom | £34,113 | Ext dom | Dom | £103,558 |
20. GP consultation rates halved | Ext dom | Dom | £34,376 | Ext dom | Dom | £103,821 |
21. GP consultation rates doubled | Ext dom | Dom | £33,588 | Ext dom | Dom | £103,033 |
22. VAS to EQ-5D mapping function | Ext dom | Dom | £32,031 | Ext dom | Dom | £97,238 |
23. QALY loss for at-risk halved | Ext dom | Dom | £34,113 | Ext dom | Dom | £103,558 |
24. Complication disutilities halved | Ext dom | Dom | £34,454 | Ext dom | Dom | £104,594 |
25. 10% uncomplicated hospitalised | Dom | Dom | £2920 | Ext dom | Dom | £72,366 |
26. Undiscounted | Ext dom | Dom | £28,849 | Ext dom | Dom | £87,579 |
The results presented in Table 71 suggest that the cost-effectiveness estimates for seasonal prophylaxis in healthy adults are sensitive to assumptions regarding influenza attack rates, the level of resistance against oseltamivir, vaccine efficacy, the threshold used to describe when influenza is circulating in the community, the risk of hospitalisation in uncomplicated cases and the discount rate. The post-exposure prophylaxis healthy adult model is sensitive to the influenza attack rate, the use of multiple prescriptions of prophylaxis at a single GP visit, vaccine efficacy and the risk of hospitalisation in uncomplicated cases.
At-risk adults
The results of the simple sensitivity analysis for the at-risk adult subgroup are presented in Table 72.
Scenario | Unvaccinated | Vaccinated | ||||
---|---|---|---|---|---|---|
Amantadine | Zanamivir | Oseltamivir | Amantadine | Zanamivir | Oseltamivir | |
Seasonal prophylaxis | ||||||
Base case (stochastic model) | Ext dom | Ext dom | £63,552 | Dom | Ext dom | £186,651 |
1. Price reduction zanamivir | Ext dom | £53,159 | £108,379 | Dom | £157,216 | £313,592 |
2. Base-case deterministic model | Ext dom | Ext dom | £60,742 | Dom | Ext dom | £179,061 |
3. Oseltamivir as suspension | Ext dom | Dom | £50,982 | Dom | Dom | £151,177 |
4. Multiple prescriptions | Ext dom | Ext dom | £60,742 | Dom | Ext dom | £179,061 |
5. Vaccine efficacy = 50% | Ext dom | Ext dom | £60,742 | Ext dom | Ext dom | £124,452 |
6. Vaccine efficacy = 25% | Ext dom | Ext dom | £60,742 | Ext dom | Ext dom | £81,979 |
7. 100% protection over influenza season | Ext dom | £50,868 | Dom | Ext dom | £150,850 | Dom |
8. No antiviral treatment | Ext dom | Ext dom | £60,133 | Dom | Ext dom | £177,346 |
9. Best-case efficacy for NIs | Ext dom | Ext dom | £55,732 | Dom | Ext dom | £164,748 |
10. No adverse events | Ext dom | Ext dom | £60,742 | Ext dom | Ext dom | £179,061 |
11. No withdrawals | Ext dom | Ext dom | £59,468 | Dom | Ext dom | £175,421 |
12. 10% resistance for oseltamivir | Ext dom | Ext dom | £67,821 | Dom | Ext dom | £199,287 |
13. 20% resistance for oseltamivir | Ext dom | £73,941 | £136,614 | Dom | £216,773 | £395,839 |
14. 30% resistance for oseltamivir | Ext dom | £73,941 | Dom | Dom | £216,773 | Dom |
15. 40% resistance for oseltamivir | Ext dom | £73,941 | Dom | Dom | £216,773 | Dom |
16. 50% resistance for oseltamivir | Ext dom | £73,941 | Dom | Dom | £216,773 | Dom |
17. Attack rates halved | Ext dom | Ext dom | £124,452 | Dom | Ext dom | £361,091 |
18. Attack rates doubled | Ext dom | Ext dom | £28,887 | Ext dom | Ext dom | £88,046 |
19. Higher influenza threshold | Ext dom | Ext dom | £99,605 | Dom | Ext dom | £290,100 |
20. GP consultation rates halved | Ext dom | Ext dom | £60,550 | Dom | Ext dom | £178,314 |
21. GP consultation rates doubled | Ext dom | Ext dom | £61,130 | Dom | Ext dom | £180,578 |
22. VAS to EQ-5D mapping function | Ext dom | Ext dom | £64,220 | Dom | Ext dom | £189,314 |
23. QALY loss for at-risk halved | Ext dom | Ext dom | £72,871 | Dom | Ext dom | £214,817 |
24. Complication disutilities halved | Ext dom | Ext dom | £61,222 | Dom | Ext dom | £180,476 |
25. 10% uncomplicated hospitalised | Ext dom | Ext dom | £47,704 | Dom | Ext dom | £166,024 |
26. Undiscounted | Ext dom | Ext dom | £51,290 | Dom | Ext dom | £151,197 |
Post-exposure prophylaxis | ||||||
Base case (stochastic model) | Ext dom | Dom | £13,459 | Ext dom | Dom | £43,970 |
1. Price reduction zanamivir | Ext dom | Dom | £13,539 | Ext dom | Dom | £44,163 |
2. Base-case deterministic model | Ext dom | Dom | £13,468 | Ext dom | Dom | £43,994 |
3. Oseltamivir as suspension | Ext dom | Dom | £13,468 | Ext dom | Dom | £43,994 |
4. Multiple prescriptions | Ext dom | Dom | £6017 | Ext dom | Dom | £22,704 |
5. Vaccine efficacy = 50% | Ext dom | Dom | £13,468 | Ext dom | Dom | £29,905 |
6. Vaccine efficacy = 25% | Ext dom | Dom | £13,468 | Ext dom | Dom | £18,947 |
7. 100% protection over influenza season | Ext dom | Dom | £13,468 | Ext dom | Dom | £43,994 |
8. No antiviral treatment | Ext dom | Dom | £13,301 | Ext dom | Dom | £43,542 |
9. Best-case efficacy for NIs | Ext dom | £452,511 | £13,468 | Ext dom | £1,298,402 | £43,994 |
10. No adverse events | Ext dom | Dom | £13,468 | Ext dom | Dom | £43,994 |
11. No withdrawals | Ext dom | Dom | £13,140 | Ext dom | Dom | £43,055 |
12. 10% resistance for oseltamivir | Ext dom | £36,590 | £15,295 | Ext dom | £110,056 | £49,212 |
13. 20% resistance for oseltamivir | Ext dom | £17,078 | Ext dom | Ext dom | £54,309 | Ext dom |
14. 30% resistance for oseltamivir | Ext dom | £17,078 | Ext dom | Ext dom | £54,309 | Ext dom |
15. 40% resistance for oseltamivir | Ext dom | £17,078 | Ext dom | Ext dom | £54,309 | Ext dom |
16. 50% resistance for oseltamivir | Ext dom | £17,078 | Ext dom | Ext dom | £54,309 | Ext dom |
17. Attack rates halved | Ext dom | Dom | £29,905 | Ext dom | Dom | £90,957 |
18. Attack rates doubled | Ext dom | Dom | £5250 | Ext dom | Dom | £20,513 |
19. Higher influenza threshold | Ext dom | Dom | £13,468 | Ext dom | Dom | £43,994 |
20. GP consultation rates halved | Ext dom | Dom | £13,499 | Ext dom | Dom | £43,881 |
21. GP consultation rates doubled | Ext dom | Dom | £13,406 | Ext dom | Dom | £44,223 |
22. VAS to EQ-5D mapping function | Ext dom | Dom | £14,239 | Ext dom | Dom | £46,513 |
23. QALY loss for at-risk halved | Ext dom | Dom | £16,158 | Ext dom | Dom | £52,779 |
24. Complication disutilities halved | Ext dom | Dom | £13,575 | Ext dom | Dom | £44,342 |
25. 10% uncomplicated hospitalised | Dom | Dom | £430 | Ext dom | Dom | £30,956 |
26. Undiscounted | Ext dom | Dom | £11,372 | Ext dom | Dom | £37,148 |
The results presented in Table 72 suggest that the cost-effectiveness estimates for seasonal prophylaxis in at-risk adults again are sensitive to assumptions regarding influenza attack rates, the level of resistance against oseltamivir, vaccine efficacy, the threshold used to describe when influenza is circulating in the community, the relative effectiveness of oseltamivir and zanamivir, the risk of hospitalisation in uncomplicated cases and the discount rate. The post-exposure prophylaxis healthy adult model is sensitive to the influenza attack rate, the use of multiple prescriptions of prophylaxis at a single GP visit, vaccine efficacy and the risk of hospitalisation in uncomplicated cases.
Healthy elderly
The results of the simple sensitivity analysis for the healthy elderly subgroup are presented in Table 73.
Scenario | Unvaccinated | Vaccinated | ||||
---|---|---|---|---|---|---|
Amantadine | Zanamivir | Oseltamivir | Amantadine | Zanamivir | Oseltamivir | |
Seasonal prophylaxis | ||||||
Base case (stochastic model) | Ext dom | Ext dom | £49,742 | Dom | Ext dom | £121,728 |
1. Price reduction zanamivir | Ext dom | Ext dom | £49,590 | Dom | Ext dom | £120,292 |
2. Base-case deterministic model | Ext dom | Ext dom | £47,609 | Dom | Ext dom | £116,346 |
3. Oseltamivir as suspension | Ext dom | Dom | £39,984 | Dom | Dom | £98,192 |
4. Multiple prescriptions | Ext dom | Ext dom | £47,609 | Dom | Ext dom | £116,346 |
5. Vaccine efficacy = 50% | Ext dom | Ext dom | £47,609 | Ext dom | Ext dom | £97,384 |
6. Vaccine efficacy = 25% | Ext dom | Ext dom | £47,609 | Ext dom | Ext dom | £64,201 |
7. 100% protection over influenza season | Ext dom | Ext dom | £47,609 | Ext dom | Ext dom | £116,346 |
8. No antiviral treatment | Ext dom | Ext dom | £47,055 | Dom | Ext dom | £115,020 |
9. Best-case efficacy for NIs | Ext dom | Ext dom | £47,609 | Dom | Ext dom | £116,346 |
10. No adverse events | Ext dom | Ext dom | £47,609 | Ext dom | Ext dom | £116,346 |
11. No withdrawals | Ext dom | Ext dom | £46,613 | Ext dom | Ext dom | £113,976 |
12. 10% resistance for oseltamivir | Ext dom | Ext dom | £53,140 | Dom | Ext dom | £129,515 |
13. 20% resistance for oseltamivir | Ext dom | Ext dom | £60,053 | Dom | Ext dom | £145,975 |
14. 30% resistance for oseltamivir | Ext dom | Ext dom | £68,941 | Dom | Ext dom | £167,138 |
15. 40% resistance for oseltamivir | Ext dom | £72,737 | Dom | Dom | £176,176 | Dom |
16. 50% resistance for oseltamivir | Ext dom | £72,737 | Dom | Dom | £176,176 | Dom |
17. Attack rates halved | Ext dom | Ext dom | £97,384 | Dom | Ext dom | £234,859 |
18. Attack rates doubled | Ext dom | Ext dom | £22,721 | Ext dom | Ext dom | £57,090 |
19. Higher influenza threshold | Ext dom | Ext dom | £77,972 | Dom | Ext dom | £188,639 |
20. GP consultation rates halved | Ext dom | Ext dom | £47,456 | Dom | Ext dom | £115,805 |
21. GP consultation rates doubled | Ext dom | Ext dom | £47,919 | Dom | Ext dom | £117,447 |
22. VAS to EQ-5D mapping function | Ext dom | Ext dom | £49,733 | Dom | Ext dom | £121,537 |
23. QALY loss for at-risk halved | Ext dom | Ext dom | £47,609 | Dom | Ext dom | £116,346 |
24. Complication disutilities halved | Ext dom | Ext dom | £47,856 | Dom | Ext dom | £116,950 |
25. 10% uncomplicated hospitalised | Ext dom | Dom | £35,219 | Dom | Ext dom | £103,957 |
26. Undiscounted | Ext dom | Ext dom | £44,358 | Dom | Ext dom | £108,402 |
Post-exposure prophylaxis | ||||||
Base case (stochastic model) | Ext dom | Dom | £10,716 | Ext dom | Dom | £28,473 |
1. Price reduction zanamivir | Ext dom | Dom | £10,734 | Ext dom | Dom | £28,608 |
2. Base-case deterministic model | Ext dom | Dom | £10,754 | Ext dom | Dom | £28,597 |
3. Oseltamivir as suspension | Ext dom | Dom | £10,754 | Ext dom | Dom | £28,597 |
4. Multiple prescriptions | Ext dom | Dom | £4897 | Ext dom | Dom | £14,651 |
5. Vaccine efficacy = 50% | Ext dom | Dom | £10,754 | Ext dom | Dom | £23,675 |
6. Vaccine efficacy = 25% | Ext dom | Dom | £10,754 | Ext dom | Dom | £15,061 |
7. 100% protection over influenza season | Ext dom | Dom | £10,754 | Ext dom | Dom | £28,597 |
8. No antiviral treatment | Ext dom | Dom | £10,615 | Ext dom | Dom | £28,257 |
9. Best-case efficacy for NIs | Ext dom | £355,876 | £10,754 | Ext dom | £850,316 | £28,597 |
10. No adverse events | Ext dom | Dom | £10,754 | Ext dom | Dom | £28,597 |
11. No withdrawals | Ext dom | Dom | £10,496 | Ext dom | Dom | £27,982 |
12. 10% resistance for oseltamivir | Ext dom | £28,930 | £12,190 | Ext dom | £71,872 | £32,015 |
13. 20% resistance for oseltamivir | Ext dom | £13,592 | Ext dom | Ext dom | £35,354 | Ext dom |
14. 30% resistance for oseltamivir | Ext dom | £13,592 | Ext dom | Ext dom | £35,354 | Ext dom |
15. 40% resistance for oseltamivir | Ext dom | £13,592 | Ext dom | Ext dom | £35,354 | Ext dom |
16. 50% resistance for oseltamivir | Ext dom | £13,592 | Ext dom | Ext dom | £35,354 | Ext dom |
17. Attack rates halved | Ext dom | Dom | £23,675 | Ext dom | Dom | £59,361 |
18. Attack rates doubled | Ext dom | Dom | £4294 | Ext dom | Dom | £13,215 |
19. Higher influenza threshold | Ext dom | Dom | £10,754 | Ext dom | Dom | £28,597 |
20. GP consultation rates halved | Ext dom | Dom | £10,810 | Ext dom | Dom | £28,552 |
21. GP consultation rates doubled | Ext dom | Dom | £10,641 | Ext dom | Dom | £28,689 |
22. VAS to EQ-5D mapping function | Ext dom | Dom | £11,234 | Ext dom | Dom | £29,873 |
23. QALY loss for at-risk halved | Ext dom | Dom | £10,754 | Ext dom | Dom | £28,597 |
24. Complication disutilities halved | Ext dom | Dom | £10,810 | Ext dom | Dom | £28,746 |
25. 10% uncomplicated hospitalised | Dom | Dom | Dom | Dom | Dom | £16,207 |
26. Undiscounted | Ext dom | Dom | £10,020 | Ext dom | Dom | £26,645 |
Table 73 suggests that the cost-effectiveness estimates are sensitive to assumptions regarding influenza attack rates, the level of resistance against oseltamivir, vaccine efficacy, the threshold used to describe when influenza is circulating in the community, the risk of hospitalisation in uncomplicated cases and the discount rate. The post-exposure prophylaxis healthy elderly model is sensitive to the influenza attack rate, the use of multiple prescriptions of prophylaxis at a single GP visit, vaccine efficacy and the risk of hospitalisation in uncomplicated cases.
Healthy elderly
Table 74 presents the results of the simple sensitivity analysis for the at-risk elderly subgroup.
Scenario | Unvaccinated | Vaccinated | ||||
---|---|---|---|---|---|---|
Amantadine | Zanamivir | Oseltamivir | Amantadine | Zanamivir | Oseltamivir | |
Seasonal prophylaxis | ||||||
Base case (stochastic model) | Ext dom | Ext dom | £38,098 | Ext dom | Ext dom | £93,763 |
1. Price reduction zanamivir | Ext dom | Ext dom | £37,968 | Ext dom | Ext dom | £93,581 |
2. Base-case deterministic model | Ext dom | Ext dom | £36,460 | Ext dom | Ext dom | £89,781 |
3. Oseltamivir as suspension | Ext dom | Dom | £30,545 | Ext dom | Dom | £75,699 |
4. Multiple prescriptions | Ext dom | Ext dom | £36,460 | Ext dom | Ext dom | £89,781 |
5. Vaccine efficacy = 50% | Ext dom | Ext dom | £36,460 | Ext dom | Ext dom | £75,072 |
6. Vaccine efficacy = 25% | Ext dom | Ext dom | £36,460 | Ext dom | Ext dom | £49,331 |
7. 100% protection over influenza season | Ext dom | Ext dom | £36,460 | Ext dom | Ext dom | £89,781 |
8. No antiviral treatment | Ext dom | Ext dom | £35,983 | Ext dom | Ext dom | £88,639 |
9. Best-case efficacy for NIs | Ext dom | Ext dom | £36,460 | Ext dom | Ext dom | £89,781 |
10. No adverse events | Ext dom | Ext dom | £36,460 | Ext dom | Ext dom | £89,781 |
11. No withdrawals | Ext dom | Ext dom | £35,688 | Ext dom | Ext dom | £87,942 |
12. 10% resistance for oseltamivir | Ext dom | Ext dom | £40,750 | Ext dom | Ext dom | £99,996 |
13. 20% resistance for oseltamivir | Ext dom | Ext dom | £46,113 | Ext dom | Ext dom | £112,764 |
14. 30% resistance for oseltamivir | Ext dom | Ext dom | £53,008 | Ext dom | Ext dom | £129,181 |
15. 40% resistance for oseltamivir | Ext dom | £55,953 | Dom | Ext dom | £136,192 | Dom |
16. 50% resistance for oseltamivir | Ext dom | £55,953 | Dom | Ext dom | £136,192 | Dom |
17. Attack rates halved | Ext dom | Ext dom | £75,072 | Dom | Ext dom | £181,714 |
18. Attack rates doubled | Ext dom | Ext dom | £17,154 | Ext dom | Ext dom | £43,815 |
19. Higher influenza threshold | Ext dom | Ext dom | £60,013 | Dom | Ext dom | £145,860 |
20. GP consultation rates halved | Ext dom | Ext dom | £36,317 | Ext dom | Ext dom | £89,304 |
21. GP consultation rates doubled | Ext dom | Ext dom | £36,751 | Ext dom | Ext dom | £90,754 |
22. VAS to EQ-5D mapping function | Ext dom | Ext dom | £37,709 | Ext dom | Ext dom | £92,858 |
23. QALY loss for at-risk halved | Ext dom | Ext dom | £57,467 | Dom | Ext dom | £141,511 |
24. Complication disutilities halved | Ext dom | Ext dom | £36,666 | Ext dom | Ext dom | £90,288 |
25. 10% uncomplicated hospitalised | Ext dom | Dom | £27,159 | Ext dom | Ext dom | £80,480 |
26. Undiscounted | Ext dom | Ext dom | £33,713 | Ext dom | Ext dom | £83,016 |
Post-exposure prophylaxis | ||||||
Base case (stochastic model) | Ext dom | Dom | £7866 | Ext dom | Dom | £21,608 |
1. Price reduction zanamivir | Ext dom | Dom | £7892 | Ext dom | Dom | £21,749 |
2. Base-case deterministic model | Ext dom | Dom | £7871 | Ext dom | Dom | £21,712 |
3. Oseltamivir as suspension | Ext dom | Dom | £7871 | Ext dom | Dom | £21,712 |
4. Multiple prescriptions | Ext dom | Dom | £3327 | Ext dom | Dom | £10,894 |
5. Vaccine efficacy = 50% | Ext dom | Dom | £7871 | Ext dom | Dom | £17,894 |
6. Vaccine efficacy = 25% | Ext dom | Dom | £7871 | Ext dom | Dom | £11,212 |
7. 100% protection over influenza season | Ext dom | Dom | £7871 | Ext dom | Dom | £21,712 |
8. No antiviral treatment | Ext dom | Dom | £7750 | Ext dom | Dom | £21,419 |
9. Best-case efficacy for NIs | Ext dom | £275,589 | £7871 | Ext dom | £659,137 | £21,712 |
10. No adverse events | Ext dom | Dom | £7871 | Ext dom | Dom | £21,712 |
11. No withdrawals | Ext dom | Dom | £7671 | Ext dom | Dom | £21,235 |
12. 10% resistance for oseltamivir | Ext dom | £21,970 | £8985 | Ext dom | £55,281 | £24,364 |
13. 20% resistance for oseltamivir | Ext dom | £10,072 | Ext dom | Ext dom | £26,954 | Ext dom |
14. 30% resistance for oseltamivir | Ext dom | £10,072 | Ext dom | Ext dom | £26,954 | Ext dom |
15. 40% resistance for oseltamivir | Ext dom | £10,072 | Ext dom | Ext dom | £26,954 | Ext dom |
16. 50% resistance for oseltamivir | Ext dom | £10,072 | Ext dom | Ext dom | £26,954 | Ext dom |
17. Attack rates halved | Ext dom | Dom | £17,894 | Ext dom | Dom | £45,576 |
18. Attack rates doubled | Ext dom | Dom | £2860 | Ext dom | Dom | £9780 |
19. Higher influenza threshold | Ext dom | Dom | £7871 | Ext dom | Dom | £21,712 |
20. GP consultation rates halved | Ext dom | Dom | £7908 | Ext dom | Dom | £21,662 |
21. GP consultation rates doubled | Ext dom | Dom | £7796 | Ext dom | Dom | £21,814 |
22. VAS to EQ-5D mapping function | Ext dom | Dom | £8141 | Ext dom | Dom | £22,456 |
23. QALY loss for at-risk halved | Ext dom | Dom | £12,406 | Ext dom | Dom | £34,222 |
24. Complication disutilities halved | Ext dom | Dom | £7915 | Ext dom | Dom | £21,835 |
25. 10% uncomplicated hospitalised | Dom | Dom | Dom | Dom | Dom | £12,411 |
26. Undiscounted | Ext dom | Dom | £7278 | Ext dom | Dom | £20,076 |
Table 74 suggests that the cost-effectiveness estimates are sensitive to assumptions regarding influenza attack rates, the level of resistance against oseltamivir, vaccine efficacy, the threshold used to describe when influenza is circulating in the community, the risk of hospitalisation in uncomplicated cases and the discount rate. The post-exposure prophylaxis at-risk elderly model is sensitive to the influenza attack rate, the use of multiple prescriptions of prophylaxis at a single GP visit, vaccine efficacy, and the risk of hospitalisation in uncomplicated cases.
Probabilistic sensitivity analysis results
Probabilistic sensitivity analysis was undertaken for the use of seasonal prophylaxis and post-exposure prophylaxis using amantadine, oseltamivir and zanamivir in each of the six subgroups, for vaccinated and unvaccinated patients. Cost-effectiveness acceptability curves for these 24 base case health economic comparisons are presented in Appendix 8. Probability sensitivity analysis was also undertaken for all health economic comparisons incorporating the proposed reduction in the price of zanamivir. Cost-effectiveness acceptability curves for these comparisons are presented in Appendix 9. For clarity of reporting, the results of the probabilistic sensitivity analysis are presented in tabular form in Tables 75 and 76. These tables show the probability that each prophylactic option produces the greatest incremental net benefit assuming cost-effectiveness thresholds of £20,000 per QALY gained and £30,000 per QALY gained.
Population | Probability optimal at £20,000/QALY | Probability optimal at £30,000/QALY | ||||||
---|---|---|---|---|---|---|---|---|
No prophylaxis | Amantadine | Oseltamivir | Zanamivir | No prophylaxis | Amantadine | Oseltamivir | Zanamivir | |
Seasonal prophylaxis | ||||||||
Healthy children | ||||||||
Unvaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 0.97 | 0.00 | 0.03 | 0.00 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
At-risk children | ||||||||
Unvaccinated | 0.27 | 0.03 | 0.70 | 0.00 | 0.02 | 0.03 | 0.94 | 0.01 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 0.97 | 0.00 | 0.03 | 0.00 |
Healthy adults | ||||||||
Unvaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
At-risk adults | ||||||||
Unvaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
Healthy elderly | ||||||||
Unvaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
At-risk elderly | ||||||||
Unvaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 0.89 | 0.02 | 0.08 | 0.01 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
Post-exposure prophylaxis | ||||||||
Healthy children | ||||||||
Unvaccinated | 0.63 | 0.00 | 0.22 | 0.15 | 0.15 | 0.00 | 0.40 | 0.45 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
At-risk children | ||||||||
Unvaccinated | 0.00 | 0.00 | 0.33 | 0.67 | 0.00 | 0.00 | 0.27 | 0.73 |
Vaccinated | 0.81 | 0.00 | 0.11 | 0.08 | 0.39 | 0.00 | 0.29 | 0.31 |
Healthy adults | ||||||||
Unvaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 0.81 | 0.00 | 0.19 | 0.00 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
At-risk adults | ||||||||
Unvaccinated | 0.02 | 0.00 | 0.89 | 0.10 | 0.00 | 0.00 | 0.84 | 0.16 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 0.96 | 0.00 | 0.04 | 0.00 |
Healthy elderly | ||||||||
Unvaccinated | 0.00 | 0.00 | 0.87 | 0.13 | 0.00 | 0.00 | 0.82 | 0.18 |
Vaccinated | 0.91 | 0.00 | 0.09 | 0.00 | 0.47 | 0.00 | 0.50 | 0.03 |
At-risk elderly | ||||||||
Unvaccinated | 0.00 | 0.00 | 0.83 | 0.17 | 0.00 | 0.00 | 0.77 | 0.23 |
Vaccinated | 0.64 | 0.00 | 0.35 | 0.01 | 0.15 | 0.00 | 0.78 | 0.07 |
Population | Probability optimal at £20,000/QALY | Probability optimal at £30,000/QALY | ||||||
---|---|---|---|---|---|---|---|---|
No prophylaxis | Amantadine | Oseltamivir | Zanamivir | No prophylaxis | Amantadine | Oseltamivir | Zanamivir | |
Seasonal prophylaxis | ||||||||
Healthy children | ||||||||
Unvaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 0.97 | 0.00 | 0.03 | 0.00 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
At-risk children | ||||||||
Unvaccinated | 0.25 | 0.03 | 0.70 | 0.03 | 0.01 | 0.02 | 0.91 | 0.05 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 0.97 | 0.00 | 0.03 | 0.00 |
Healthy adults | ||||||||
Unvaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
At-risk adults | ||||||||
Unvaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 0.99 | 0.00 | 0.00 | 0.01 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
Healthy elderly | ||||||||
Unvaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 0.97 | 0.00 | 0.00 | 0.03 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
At-risk elderly | ||||||||
Unvaccinated | 0.99 | 0.00 | 0.00 | 0.00 | 0.77 | 0.02 | 0.05 | 0.16 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
Post-exposure prophylaxis | ||||||||
Healthy children | ||||||||
Unvaccinated | 0.44 | 0.00 | 0.09 | 0.47 | 0.06 | 0.00 | 0.15 | 0.79 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 0.99 | 0.00 | 0.00 | 0.01 |
At-risk children | ||||||||
Unvaccinated | 0.00 | 0.00 | 0.15 | 0.85 | 0.00 | 0.00 | 0.15 | 0.85 |
Vaccinated | 0.70 | 0.00 | 0.04 | 0.26 | 0.24 | 0.00 | 0.12 | 0.65 |
Healthy adults | ||||||||
Unvaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 0.76 | 0.00 | 0.18 | 0.06 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 1.00 | 0.00 | 0.00 | 0.00 |
At-risk adults | ||||||||
Unvaccinated | 0.00 | 0.00 | 0.59 | 0.40 | 0.00 | 0.00 | 0.59 | 0.41 |
Vaccinated | 1.00 | 0.00 | 0.00 | 0.00 | 0.95 | 0.00 | 0.04 | 0.01 |
Healthy elderly | ||||||||
Unvaccinated | 0.00 | 0.00 | 0.62 | 0.38 | 0.00 | 0.00 | 0.62 | 0.38 |
Vaccinated | 0.90 | 0.00 | 0.07 | 0.03 | 0.42 | 0.00 | 0.38 | 0.20 |
At-risk elderly | ||||||||
Unvaccinated | 0.00 | 0.00 | 0.60 | 0.40 | 0.00 | 0.00 | 0.60 | 0.40 |
Vaccinated | 0.60 | 0.00 | 0.25 | 0.15 | 0.11 | 0.00 | 0.54 | 0.34 |
Uncertainty analysis results: base-case scenario
Table 75 presents the probability that each prophylactic option produces the greatest level of net benefit at thresholds of £20,000 per QALY gained and £30,000 per QALY gained for the base-case analysis. The option which is most likely to produce the greatest level of net benefit is highlighted in bold for each comparison.
Uncertainty analysis results: proposed price reduction for zanamivir
Table 76 presents the probability that each prophylactic option produces the greatest level of net benefit at thresholds of £20,000 per QALY gained and £30,000 per QALY gained, incorporating the proposed reduction in the price of zanamivir. The option which is most likely to produce the greatest level of net benefit is highlighted in bold for each comparison.
Budget impact analysis
This section presents estimates of the budget impact of a positive recommendation for each prophylactic option within each model subgroup in the light of current NICE recommendations. The analysis is based upon the expected cost of each prophylaxis strategy, including potential cost savings associated with the avoidance of influenza and other ILIs. Separate budget impact analyses are presented for seasonal prophylaxis and post-exposure prophylaxis. NICE currently recommends the use of oseltamivir as post-exposure prophylaxis in at-risk individuals aged over 13 years; this is taken to be the baseline cost, against which the incremental cost of each prophylactic option is compared.
The population of England and Wales is currently estimated to be around 53,728,600, based on data from the ONS. Of this figure, approximately 11,295,800 are aged under 16, 33,822,300 are working-age adults and 8,610,500 are elderly. The previous assessment by Turner et al. 10 suggested that approximately 12%, 25% and 42% of children, adults and elderly individuals respectively would be considered high risk. Recent evidence suggests that uptake of influenza vaccination is approximately 79% in individuals over the age of 65 years and around 42% in high-risk individuals who are under the age of 65. Data from the Department of Health suggest that the residential care home population in England and Wales is around 545,000 persons. These data were synthesised to crudely estimate the number of individuals who fall into each of the model subgroups (Table 77).
Population group | No. of individuals | Community dwelling | Residential care home |
---|---|---|---|
Healthy children | |||
Unvaccinated | 9,940,304 | 9,940,304 | 0 |
Vaccinated | 0 | 0 | 0 |
At-risk children | |||
Unvaccinated | 784,832 | 784,832 | 0 |
Vaccinated | 570,664 | 570,664 | 0 |
Healthy adults | |||
Unvaccinated | 25,366,725 | 25,366,725 | 0 |
Vaccinated | 0 | 0 | 0 |
At-risk adults | |||
Unvaccinated | 4,895,778 | 4,895,778 | 0 |
Vaccinated | 3,559,797 | 3,559,797 | 0 |
Healthy elderly | |||
Unvaccinated | 1,033,777 | 968,344 | 65,433 |
Vaccinated | 3,960,313 | 3,709,646 | 250,667 |
At-risk elderly | |||
Unvaccinated | 748,597 | 701,215 | 47,382 |
Vaccinated | 2,867,813 | 2,686,295 | 181,518 |
For the seasonal prophylaxis budget impact model, any individual within each subgroup could be potentially eligible to receive prophylaxis provided he or she is over the age specified within the licensed indications for each prophylaxis drug. The proportion of children who would be eligible for prophylaxis using amantadine, oseltamivir and zanamivir was estimated using data from the ONS. The estimated budget impact for seasonal prophylaxis options is presented in Table 78.
Population group | No. of individuals | Current policy | Expected cost per patient | Current cost | Additional budget impact over eligible population | |||||
---|---|---|---|---|---|---|---|---|---|---|
NP | Amantadine | Zanamivir | Oseltamivir | Amantadine | Zanamivir | Oseltamivir | ||||
Healthy children | ||||||||||
Unvaccinated | 10,240,912 | NP | £17.72 | £56.23 | £112.15 | £85.51 | £17.72 | £183,952,396 | £343,735,708 | £655,247,656 |
Vaccinated | – | NP | £43.23 | £78.64 | £140.36 | £115.05 | £43.23 | £0 | £0 | £0 |
At-risk children | ||||||||||
Unvaccinated | 808,567 | NP | £29.89 | £66.92 | £121.56 | £93.57 | £29.89 | £13,970,266 | £26,347,333 | £48,599,333 |
Vaccinated | 587,921 | NP | £51.71 | £86.84 | £147.86 | £122.06 | £51.71 | £9,632,657 | £20,090,863 | £39,033,738 |
Healthy adults | ||||||||||
Unvaccinated | 25,110,750 | NP | £6.63 | £46.49 | £103.70 | £111.09 | £6.63 | £1,000,937,591 | £2,437,345,426 | £2,622,899,442 |
Vaccinated | – | NP | £35.64 | £71.34 | £133.74 | £141.62 | £35.64 | £0 | £0 | £0 |
At-risk adults | ||||||||||
Unvaccinated | 4,846,375 | NP | £13.57 | £52.74 | £108.33 | £115.63 | £13.57 | £189,831,470 | £459,262,856 | £494,635,028 |
Vaccinated | 3,523,875 | NP | £40.39 | £75.94 | £137.67 | £145.53 | £40.39 | £125,278,544 | £342,831,299 | £370,497,449 |
Healthy elderly | ||||||||||
Unvaccinated | 1,033,813 | NP | £10.43 | £49.93 | £106.16 | £112.80 | £10.43 | £40,836,559 | £98,974,490 | £105,837,889 |
Vaccinated | 3,960,451 | NP | £38.59 | £74.16 | £136.02 | £143.54 | £38.59 | £140,896,254 | £385,872,027 | £415,636,663 |
At-risk elderly | ||||||||||
Unvaccinated | 748,623 | NP | £13.45 | £52.63 | £108.39 | £114.54 | £13.45 | £29,333,408 | £71,075,149 | £75,678,598 |
Vaccinated | 2,867,913 | NP | £40.75 | £76.25 | £137.84 | £145.15 | £40.75 | £101,827,060 | £278,453,842 | £299,415,220 |
For the post-exposure prophylaxis budget impact model, the population of interest relates to individuals who have come into contact with an index ILI case. The number of potentially eligible contact cases is crudely estimated by multiplying the number of individuals in each model subgroup by an estimated overall household ILI attack rate (the estimated household influenza attack rate multiplied by the probability that ILI is influenza). 20 The budget impact model assumes that if a household is infected, all contact cases will be eligible for prophylaxis if they present within 48 hours of contact with the index case. The model estimates the additional cost of each policy in the light of the existing NICE guidance (the ‘current policy cost’ column details the expected cost per patient of prophylaxis according to current NICE guidance). The budget impact for the residential care home population was based on an assumed ILI attack rate of 41%. 132 The estimated budget impact for post-exposure prophylaxis options is presented in Tables 79 and 80.
Population group | No. of individuals | Current policy | Expected cost per patient | Current cost | Additional budget impact over eligible population | |||||
---|---|---|---|---|---|---|---|---|---|---|
NP | Amantadine | Zanamivir | Oseltamivir | Amantadine | Zanamivir | Oseltamivir | ||||
Healthy children | ||||||||||
Unvaccinated | 2,067,940 | NP | £18.96 | £46.40 | £61.18 | £54.35 | £18.96 | £31,971,071 | £16,526,348 | £35,963,481 |
Vaccinated | – | NP | £44.09 | £73.84 | £91.00 | £83.30 | £44.09 | £0 | £0 | £0 |
At-risk children | ||||||||||
Unvaccinated | 163,273 | Ose/NP | £32.56 | £57.55 | £69.76 | £63.97 | £47.99 | –£254,503 | –£1,873,897 | £0 |
Vaccinated | 118,719 | Ose/NP | £53.57 | £82.44 | £98.67 | £91.35 | £72.13 | –£256,859 | –£1,966,380 | £0 |
Healthy adults | ||||||||||
Unvaccinated | 5,070,595 | NP | £9.17 | £38.48 | £55.19 | £46.94 | £9.17 | £23,136,834 | £36,323,410 | £29,814,690 |
Vaccinated | – | NP | £37.36 | £67.80 | £85.67 | £77.46 | £37.36 | £0 | £0 | £0 |
At-risk adults | ||||||||||
Unvaccinated | 978,625 | Ose | £19.34 | £47.10 | £61.49 | £53.18 | £24.61 | –£927,521 | £1,265,252 | £0 |
Vaccinated | 711,574 | Ose | £44.32 | £74.21 | £91.27 | £83.04 | £50.35 | –£978,796 | £911,673 | £0 |
Healthy elderly | ||||||||||
Unvaccinated | 195,544 | Ose | £17.75 | £45.76 | £60.50 | £52.17 | £21.63 | –£141,505 | £183,604 | £0 |
Vaccinated | 749,114 | Ose | £43.82 | £73.59 | £90.52 | £82.27 | £48.16 | –£733,300 | £696,777 | £0 |
At-risk elderly | ||||||||||
Unvaccinated | 141,601 | Ose | £22.88 | £50.05 | £63.68 | £55.33 | £26.54 | –£84,394 | £133,358 | £0 |
Vaccinated | 542,462 | Ose | £47.50 | £76.92 | £93.37 | £85.11 | £51.75 | –£500,882 | £505,397 | £0 |
Population group | No. of individuals | Current policy | Expected cost per patient | Current cost | Additional budget impact over eligible population | |||||
---|---|---|---|---|---|---|---|---|---|---|
NP | Amantadine | Zanamivir | Oseltamivir |
Amantadine |
Zanamivir | Oseltamivir | ||||
Healthy elderly | ||||||||||
Unvaccinated | 26,827 | Ose | £17.75 | £45.76 | £60.50 | £52.17 | £21.63 | –£19,413.59 | £25,189 | £0.00 |
Vaccinated | 102,774 | Ose | £43.82 | £73.59 | £90.52 | £82.27 | £48.16 | –£100,603.99 | £4,352,991 | £0.00 |
At-risk elderly | ||||||||||
Unvaccinated | 19,427 | Ose | £22.88 | £50.05 | £63.68 | £55.33 | £26.54 | –£11,578.37 | £721,445 | £0.00 |
Vaccinated | 74,422 | Ose | £47.50 | £76.92 | £93.37 | £85.11 | £51.75 | –£68,717.81 | £3,097,413 | £0.00 |
Chapter 5 Assessment of factors relevant to the NHS and other parties
Use of amantadine for Parkinson’s disease and herpes zoster virus
It should be borne in mind that, as amantadine is also licensed for the treatment of Parkinson’s disease and herpes zoster, individuals receiving the drug for these conditions may be protected against influenza A.
Herd immunity
The concept of herd immunity postulates that the higher the proportion of individuals in a population who are protected from an infection, the less likely it is that an outbreak of the same infection may become established in that community. With respect to influenza, it could be proposed that, where the number of individuals who are able to transmit the virus is reduced as a result of vaccination and/or influenza prophylaxis, unprotected individuals are less likely to become exposed to infection and are thus indirectly protected. Although this concept has not been modelled in this assessment, it should be considered that influenza prophylaxis in at-risk groups may result in herd immunity effects in the population with which they are in contact. Additional studies that examine the degree of viral shedding among subjects receiving prophylaxis versus placebo may provide further information with regard to this effect.
An additional issue relating to immunity against influenza was raised by study authors, who proposed that, while antivirals may be effective in preventing the development of SLCI, asymptomatic individuals may in fact have subclinical influenza infection, which may have the potential to confer immunity to the circulating strain on the exposed individual.
Additional support in using antivirals
In the clinical effectiveness review, a number of issues were identified relating to the external validity of a minority of the oseltamivir and zanamivir trials; these are discussed in Chapter 6. It was noted that in some studies, subjects who had lower levels of cognitive function and/or manual dexterity were excluded from participation. Therefore, it is possible that the reported levels of adherence and acceptability of the use of the Diskhaler device for delivery of zanamivir, and the ability of subjects to take oral antivirals independently, may not accurately reflect the scenario in the general population, and that older individuals or those with lower cognitive functioning and/or manual dexterity may require additional support from health- and social care professionals or carers in administration of antivirals.
Prescribing patterns for influenza prophylaxis
It was typically stipulated in the study inclusion criteria in the clinical trials of the use of oseltamivir and zanamivir in post-exposure prophylaxis that the administration of antivirals should be commenced within 48 hours of exposure to the ILI index case for oseltamivir and within 36 hours for zanamivir. In clinical practice, this requirement may be problematic, as it relies on both the identification of index cases and the initiation of prophylaxis in contact cases within the recommended cut-off period. In addition, initiation of post-exposure prophylaxis relies on the patient having access to GP services within the specified time period. The requirement for testing of creatinine clearance for dose adjustment for amantadine and oseltamivir also has the potential to affect the speed with which prophylaxis may be implemented.
A GP can usually prescribe medication only for individuals who present for consultation. The requirement for early identification of index cases and contact cases in post-exposure prophylaxis may lead to variations in prescribing practices, e.g. giving multiple prescriptions of prophylaxis to household contacts.
The future use of rapid diagnostic tests for influenza in clinical practice could be anticipated to facilitate the rapid identification of influenza-positive index cases and the circulation of influenza in the local community and, as such, has the potential to increase the clinical effectiveness of antivirals in prophylaxis.
Impact on primary care
Raised awareness of the availability of antiviral prophylaxis among the general population may lead to increased workloads for GPs and other primary health-care professionals. It should be noted that the economic analysis presented here makes very few assumptions about the way in which prophylaxis would be implemented or the infrastructure required to manage this. In certain patient groups, this may be a lesser issue (e.g. the use of post-exposure prophylaxis to manage opportunistically outbreaks in residential care homes) while for other settings the infrastructure may be of greater concern (e.g. introducing routine prophylaxis in schools).
Involvement of pharmacist in use of powder for oral suspension
As noted in Chapter 1, the summary of product characteristics for oseltamivir recommends that powder for oral suspension should be reconstituted by a pharmacist before being dispensed to the patient.
Chapter 6 Discussion
Statement of principal findings
Clinical effectiveness review
Twenty-six published references and one unpublished report relating to a total of 23 RCTs were included in the review of clinical effectiveness. The quality of the studies identified was variable and gaps in the evidence base limited the assessment of the clinical effectiveness of the interventions across population subgroups and settings. The evidence for amantadine prophylaxis across subgroups was very limited. However, evidence of the effectiveness of amantadine in preventing SLCI in outbreak control among adolescent subjects was identified. Oseltamivir was shown to be effective in preventing SLCI in a number of subgroups, particularly in seasonal prophylaxis in at-risk elderly subjects and in post-exposure prophylaxis in households of mixed composition. The effectiveness of zanamivir in preventing SLCI was also demonstrated, and was most convincing in trials of seasonal prophylaxis in at-risk adults and adolescents and in healthy and at-risk elderly subjects and in post-exposure prophylaxis in mixed households. Interventions appeared to be tolerated reasonably well by subjects, with a relatively low proportion of subjects experiencing drug-related adverse events and drug-related withdrawals. Very limited evidence was reported for the effectiveness of the interventions in preventing complications, hospitalisations and in minimising length of illness and time to return to normal activities. No data could be identified for HRQoL or mortality outcomes. Additional consideration should be paid to the issues of antiviral resistance and adverse events associated with amantadine during the interpretation of the findings of the review.
Cost-effectiveness review
Cost-effectiveness of amantadine, zanamivir and oseltamivir as seasonal prophylaxis
In healthy children
Amantadine and zanamivir as seasonal prophylaxis are expected to be dominated or extendedly dominated in the healthy children subgroup. The proposed reduction in the price of zanamivir does not affect this finding. The incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be greater than £44,000 per QALY gained. Assuming a willingness-to-pay threshold of £30,000 per QALY gained, the probability that no prophylaxis produces the greatest level of net benefit is expected to be around 0.97.
In at-risk children
Amantadine and zanamivir as seasonal prophylaxis are expected to be dominated or extendedly dominated in the at-risk children subgroup. Again, the proposed reduction in the price of zanamivir does not affect this finding. The incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be around £17,000 per QALY gained for at-risk children who have not been vaccinated. For at-risk children who have previously been vaccinated, the incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be in excess of £50,000 per QALY gained. The cost-effectiveness estimates for oseltamivir are based on efficacy data that have been drawn from a trial of seasonal prophylaxis in healthy adults. Assuming a willingness-to-pay threshold of £20,000 per QALY gained, the probability that oseltamivir is optimal in unvaccinated at-risk children is approximately 0.70 (this probability is also 0.70 when the proposed price reduction for zanamivir is included). Assuming a willingness-to-pay threshold of £30,000 per QALY gained, the probability that oseltamivir is optimal in unvaccinated at-risk children is around 0.94 (p = 0.91 when the proposed price reduction for zanamivir is included). For at-risk children who have previously been vaccinated, the probability that no prophylaxis is optimal at £30,000 per QALY gained is approximately 0.97 or higher.
In healthy adults
Amantadine and zanamivir as seasonal prophylaxis are expected to be dominated or extendedly dominated in the healthy adult subgroup. The incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be around £148,000 per QALY gained for healthy adults who have not been vaccinated and more than £427,000 per QALY gained for healthy adults who have been vaccinated. These estimates are based on a trial of oseltamivir as seasonal prophylaxis in healthy adults. Assuming a willingness-to-pay threshold of £30,000 per QALY gained, the probability that no prophylaxis is optimal is close to 1.0, irrespective of vaccination status.
In at-risk adults
Based on the current list price for zanamivir, the model suggests that both amantadine and zanamivir are ruled out of the analysis in at-risk adults. The incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be around £64,000 per QALY gained in unvaccinated at-risk adults and around £187,000 per QALY gained in previously vaccinated at-risk adults. These estimates are based on a trial of oseltamivir as seasonal prophylaxis in healthy adults. Assuming a willingness-to-pay threshold of £30,000 per QALY gained, the probability that no prophylaxis produces the greatest amount of net benefit is close to 1.0.
When the proposed price reduction for zanamivir is included in the analysis for at-risk adults, zanamivir is no longer dominated. The incremental cost-effectiveness of seasonal prophylaxis using zanamivir versus no prophylaxis is expected to be around £53,000 per QALY gained in unvaccinated at-risk adults and £157,000 per QALY gained in at-risk adults who have previously been vaccinated. The incremental cost-effectiveness of oseltamivir is expected to be around £108,000 per QALY gained in unvaccinated at-risk adults and around £314,000 per QALY gained in previously vaccinated at-risk adults. Assuming a willingness-to-pay threshold of £30,000 per QALY gained, the probability that no prophylaxis is optimal is around 0.99 for unvaccinated at-risk adults and close to 1.0 for previously vaccinated at-risk adults.
In healthy elderly individuals
In this subgroup, amantadine and zanamivir are expected to be dominated or extendedly dominated. The proposed reduction in the price of zanamivir does not affect this result. The incremental cost-effectiveness of oseltamivir versus no prophylaxis in healthy elderly individuals who have not been vaccinated is expected to be around £50,000 per QALY gained. For previously vaccinated healthy elderly individuals, the incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be greater than £120,000 per QALY gained. These estimates are based on a trial of oseltamivir as seasonal prophylaxis in elderly individuals. Assuming a willingness-to-pay threshold of £30,000 per QALY gained, the probability that no prophylaxis is expected to be optimal is close to 1.0 (this probability is around 0.97 and 1.0 when the proposed price reduction for zanamivir is included in the analysis for unvaccinated and vaccinated subgroups respectively).
In at-risk elderly individuals
In this subgroup, amantadine and zanamivir are expected to be extendedly dominated despite the proposed reduction in the price of zanamivir. The incremental cost-effectiveness of oseltamivir versus no prophylaxis in at-risk elderly individuals who have not been vaccinated is expected to be around £38,000 per QALY gained. For previously vaccinated at-risk elderly individuals, the incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be around £94,000 per QALY gained. These estimates are based on a trial of oseltamivir as seasonal prophylaxis in elderly subjects. Assuming a willingness-to-pay threshold of £30,000 per QALY gained, the probability that no prophylaxis is optimal is around 0.77 or higher.
The simple sensitivity analysis suggests that the cost-effectiveness of seasonal prophylaxis using amantadine, oseltamivir and zanamivir is sensitive to assumptions regarding the influenza attack rate, the level of resistance against oseltamivir, vaccine efficacy, the threshold used to describe when influenza is circulating in the community, the risk of hospitalisation in uncomplicated cases and the discount rate.
Cost-effectiveness of amantadine, zanamivir and oseltamivir as post-exposure prophylaxis
In healthy children
Amantadine and oseltamivir as post-exposure prophylaxis are expected to be dominated or extendedly dominated in the healthy children subgroup. For unvaccinated healthy children, the incremental cost-effectiveness of zanamivir post-exposure prophylaxis versus no prophylaxis is expected to be around £23,000 per QALY gained at the current list price, and around £19,000 per QALY gained when the proposed price reduction for zanamivir is included in the analysis. For vaccinated healthy children, the incremental cost-effectiveness of zanamivir is expected to be at least £59,000 per QALY gained; this estimate includes the proposed price reduction for zanamivir. These cost–utility estimates are based on effectiveness data derived from trials of post-exposure prophylaxis in households of mixed composition (children and adults). Based on the current list price for zanamivir, the probability that zanamivir is optimal in unvaccinated healthy children is expected to be 0.15 and 0.45 at willingness-to-pay thresholds of £20,000 and £30,000 per QALY gained respectively. When the proposed price reduction is included in the analysis, the probability that zanamivir is optimal in unvaccinated healthy children is expected to be 0.47 and 0.79 at willingness-to-pay thresholds of £20,000 and £30,000 per QALY gained respectively. For the vaccinated subgroup, the probability that no prophylaxis is optimal at a threshold of £30,000 per QALY gained is close to 1.0 (p = 0.99 when the proposed price reduction for zanamivir is included).
For children under the age of 5 years, oseltamivir is the only licensed antiviral prophylaxis option. The incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be around £24,000 per QALY gained and £74,000 per QALY gained in unvaccinated and vaccinated groups respectively.
In at-risk children
Amantadine and oseltamivir as post-exposure prophylaxis are expected to be dominated or extendedly dominated in the at-risk children subgroup. For unvaccinated at-risk children, the incremental cost-effectiveness of zanamivir post-exposure prophylaxis versus no prophylaxis is expected to be around £8000 per QALY gained at the current list price, and around £6000 per QALY gained when the proposed price reduction for zanamivir is included in the analysis. For vaccinated at-risk children, the incremental cost-effectiveness of zanamivir is expected to be around £28,000 per QALY gained at the current list price, and £23,000 per QALY gained when the proposed price reduction is included in the analysis. Again, these cost–utility estimates are based on effectiveness data derived from trials of post-exposure prophylaxis in households of mixed composition (children and adults). Based on its current list price, the probability that zanamivir is optimal in unvaccinated at-risk children is expected to be 0.67 and 0.73 at willingness-to-pay thresholds of £20,000 and £30,000 per QALY gained respectively. When the proposed price reduction is included in the analysis, the probability that zanamivir is optimal in unvaccinated at-risk children is expected to be 0.85 at willingness-to-pay thresholds of £20,000 and £30,000 per QALY gained. Based on the current list price for zanamivir, the probability that zanamivir is optimal in vaccinated at-risk children is expected to be 0.08 and 0.31 at willingness-to-pay thresholds of £20,000 and £30,000 per QALY gained respectively. When the proposed price reduction is included in the analysis, the probability that zanamivir is optimal in unvaccinated at-risk children is expected to be 0.26 and 0.65 at willingness-to-pay thresholds of £20,000 and £30,000 per QALY gained respectively.
For at-risk children under the age of 5 years, the incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be around £9000 per QALY gained for unvaccinated at-risk children and around £29,000 per QALY gained for vaccinated at-risk children.
In healthy adults
Amantadine and zanamivir prophylaxis are expected to be dominated or extendedly dominated in the healthy adult subgroup. The proposed price reduction for zanamivir does not affect this result. For unvaccinated healthy adults, the incremental cost-effectiveness of oseltamivir post-exposure prophylaxis versus no prophylaxis is expected to be around £34,000 per QALY gained. For previously vaccinated healthy adults, the incremental cost-effectiveness of oseltamivir is expected to be around £104,000 per QALY gained. These cost–utility estimates are based on effectiveness data derived from trials of post-exposure prophylaxis in households of mixed composition (children and adults). The probability that oseltamivir is optimal in unvaccinated otherwise healthy adults is expected to be around 0 and 0.19 at willingness-to-pay thresholds of £20,000 and £30,000 per QALY gained respectively. For healthy adults who have previously been vaccinated, the probability that oseltamivir is optimal is close to 0 at a willingness-to-pay threshold of £30,000 per QALY gained.
In at-risk adults
Amantadine and zanamivir prophylaxis are expected to be dominated or extendedly dominated in the at-risk adult subgroup. The proposed price reduction for zanamivir does not affect this result. For unvaccinated at-risk adults, the incremental cost-effectiveness of oseltamivir post-exposure prophylaxis versus no prophylaxis is expected to be around £13,000 per QALY gained. For previously vaccinated at-risk adults, the incremental cost-effectiveness of oseltamivir is expected to be around £44,000 per QALY gained. These cost–utility estimates are based on effectiveness data derived from trials of post-exposure prophylaxis in households of mixed composition (children and adults). Based on the current list price for zanamivir, the probability that oseltamivir is optimal in unvaccinated at-risk adults is 0.89 and 0.84 at willingness-to-pay thresholds of £20,000 and £30,000 per QALY gained respectively (p = 0.59 when the proposed price reduction for zanamivir is included in the analysis). For at-risk adults who have previously been vaccinated, the probability that no prophylaxis is optimal is around 0.96 at a willingness-to-pay threshold of £30,000 per QALY gained (p = 0.95 when the proposed price reduction for zanamivir is included).
In healthy elderly individuals
Amantadine and zanamivir prophylaxis are expected to be dominated or extendedly dominated in the healthy elderly subgroup. The proposed price reduction for zanamivir does not affect this result. For unvaccinated healthy elderly individuals, the incremental cost-effectiveness of oseltamivir post-exposure prophylaxis versus no prophylaxis is expected to be around £11,000 per QALY gained. For previously vaccinated healthy elderly individuals, the incremental cost-effectiveness of oseltamivir is expected to be around £28,000 per QALY gained. These cost–utility estimates are based on effectiveness data derived from trials of post-exposure prophylaxis in households of mixed composition (children and adults). Based on the current list price for zanamivir, the probability that oseltamivir is optimal in unvaccinated healthy elderly individuals is 0.87 and 0.82 at willingness-to-pay thresholds of £20,000 and £30,000 per QALY gained respectively (p = 0.62 when the proposed price reduction for zanamivir is included in the analysis). For healthy elderly individuals who have previously been vaccinated, the probability that oseltamivir is optimal is 0.09 and 0.50 at willingness-to-pay thresholds of £20,000 per QALY gained and £30,000 per QALY gained respectively (p = 0.07 and 0.38 when the proposed price reduction for zanamivir is included in the analysis).
In at-risk elderly individuals
Amantadine and zanamivir as post-exposure prophylaxis are expected to be dominated or extendedly dominated in the at-risk elderly subgroup. For unvaccinated at-risk elderly individuals, the incremental cost-effectiveness of oseltamivir post-exposure prophylaxis versus no prophylaxis is expected to be around £8000 per QALY gained. For vaccinated at-risk elderly individuals, the incremental cost-effectiveness of oseltamivir is expected to be around £22,000 per QALY gained. Again, these cost–utility estimates are based on effectiveness data derived from trials of post-exposure prophylaxis in households of mixed composition (children and adults). The probability that oseltamivir is optimal in unvaccinated at-risk elderly individuals is around 0.83 and 0.77 at willingness-to-pay thresholds of £20,000 and £30,000 per QALY gained (this probability is around 0.60 when the proposed price reduction for zanamivir is included in the analysis). For vaccinated at-risk elderly individuals, the probability that oseltamivir is optimal is 0.35 and 0.78 at willingness-to-pay thresholds of £20,000 per QALY gained and £30,000 per QALY gained respectively (p = 0.25 and 0.54 when the proposed price reduction for zanamivir is included in the analysis).
The simple sensitivity analysis suggests that the cost-effectiveness of post-exposure prophylaxis using amantadine, oseltamivir and zanamivir is sensitive to assumptions regarding the influenza attack rate, the level of resistance against oseltamivir, assumptions regarding the comparative efficacy of oseltamivir and zanamivir, the efficacy of influenza vaccination, multiple prescribing of prophylaxis to contact cases, the risk of hospitalisation in uncomplicated cases and the discount rate.
Strengths and limitations of the assessment
The methods used for reviewing the evidence for the clinical effectiveness of amantadine, oseltamivir and zanamivir in seasonal and post-exposure prophylaxis against influenza were comprehensive and systematic and we are confident that we identified all RCTs suitable for inclusion in the assessment. However, a limitation of the review was the necessity to exclude non-English studies, owing to time constraints. Where abstracts in English could be obtained for potentially relevant trials, the available data were discussed. An additional limitation was that a small number of full papers could not be retrieved by information specialists. However, as discussed earlier, it was considered unlikely that these articles were suitable for inclusion in the review.
The health economic model presented in Chapter 4 was developed following a detailed critical review of previous economic evaluations of influenza prophylaxis and clinical input. The review highlighted a number of concerns with previous health economic evaluations of amantadine, oseltamivir and zanamivir prophylaxis (see Chapter 4, Systematic review of existing cost-effectiveness data); the model presented here addresses each of these concerns. Despite this, the evidence base is subject to considerable uncertainty, and the evidence identified for the model is far from ideal, particularly in terms of the expected benefits of prophylaxis. The main limitation of the health economic model presented within this assessment is the use of a static rather than dynamic modelling approach. As such, the model captures only the benefits accrued by patients receiving prophylaxis, and does not include other potential indirect benefits accrued through decreased transmission of influenza through the use of prophylaxis. However, the use of a more sophisticated modelling approach would require additional assumptions and would not serve to reconcile the problems associated with an already limited evidence base (see below).
Uncertainties
Although a considerable amount of evidence was identified relating to the use of amantadine, oseltamivir and zanamivir in seasonal and post-exposure prophylaxis against influenza, the assessment of the clinical effectiveness of these interventions was limited by the variation in the quality of trials in terms of internal validity and clarity of reporting and by the heterogeneity between studies. The capacity of a number of trials to demonstrate efficacy against SLCI was hindered by low attack rates during the seasons under study. The quality of the study design and reporting of the amantadine prophylaxis trials was particularly poor and few data could be abstracted to inform the clinical effectiveness review. Further trials would be required to enable a meaningful evaluation of the effectiveness of this intervention. Stronger evidence was identified for the efficacy of both oseltamivir and zanamivir in preventing SLCI, with some limited data being available on the impact of the interventions on complications and hospitalisations, and on reducing length and severity of clinical disease across age groups, risk status groups and settings. However, significant gaps in knowledge still exist, which require further research. Further studies among those population groups considered to be at higher risk of influenza-associated complications are necessary to strengthen the evidence base for efficacy in the most clinically relevant subgroups. There is a particular requirement for further evidence relating to the clinical effectiveness of antivirals in post-exposure prophylaxis among elderly subjects, particularly in long-term care settings, as subjects over 65 years of age were not well represented in the post-exposure prophylaxis trials. Further research to investigate the use of zanamivir by patients with low cognitive function is warranted. Randomised controlled trials to investigate the use of oseltamivir in seasonal prophylaxis in healthy and at-risk children, at-risk adults and healthy elderly subjects, and the representation of a range of risk and age subgroups in post-exposure prophylaxis studies would be of value. Although the report by LaForce et al. 75 presented considerable evidence since the last HTA review10 concerning the protective efficacy of zanamivir in seasonal prophylaxis for at-risk adolescents and adults, further research is required on zanamivir in seasonal prophylaxis in healthy and at-risk children and healthy elderly subjects, and a more comprehensive representation of age and risk subgroups in studies of post-exposure prophylaxis in households is needed. Studies of influenza antiviral prophylaxis in which the effect of the confounding variable of vaccination is explored further are recommended. Research to assess the impact of seasonal prophylaxis in certain groups, such as children, on the transmission and circulation of influenza in the community would also be of value.
A number of head-to-head trials of antiviral interventions used in prophylaxis against influenza were identified and excluded in the clinical effectiveness review. Research was identified in which the efficacies of amantadine and rimantadine in prophylaxis against influenza were compared,133,134 while the evidence base for amantadine and rimantadine prophylaxis was reviewed in a recent Cochrane publication. 33 Additional data identified and excluded in this assessment examined the prophylactic efficacies of ribavirin versus amantadine134 and zanamivir versus rimantadine. 78 However, no relevant head-to-head RCTs in which amantadine, oseltamivir and/or zanamivir were directly compared could be identified. Such trials would be of significant value in determining the relative clinical effectiveness of these interventions in prophylaxis against influenza. The undertaking of a large-scale RCT of the efficacy of these interventions in seasonal and post-exposure prophylaxis with the incorporation of quality of life and resistance measurements would significantly expand the evidence base, although it is acknowledged that such a trial would require considerable resources.
The weaknesses in the clinical evidence base are directly relevant to the interpretation of the health economic model results. There is a marked paucity of robust evidence concerning the relative efficacy of alternative antiviral prophylactic drugs in specific subgroups. The non-exchangeability of studies of individual antivirals and the absence of head-to-head trials suggests that the use of more advanced Bayesian meta-analytic techniques (e.g. mixed treatment comparisons) would add little to the findings. As such, the economic analysis is pivoted on assumptions of equivalent efficacy of antivirals across numerous subgroups based on few trials (this is particularly the case for amantadine).
A number of attributes of the study designs of identified trials have implications for the interpretation of study findings. One issue relates to the variation in timing of prophylaxis within trials. Variation was evident in the timing of the onset of prophylaxis in experimental challenge studies, with subjects being dosed 1 day62,67 to 4 days63 before viral challenge. In the post-exposure prophylaxis studies based in households, prophylaxis in contact cases with oseltamivir began within 48 hours of the onset of symptoms in the index case;48,49 however, in the zanamivir trials prophylaxis was initiated within 36 hours of the onset of symptoms in the index case in two studies46,47 and where contacts had been exposed to an index case with ILI of no longer than 4 days’ duration. 72 Considerable variation was also present in the timing of the initiation of prophylaxis in trials of amantadine59–61 and zanamivir76,78 in outbreak control, where medications were administered upon levels of influenza activity reaching a level specific to that study. These variations in the onset of prophylaxis following exposure to influenza have the potential to impact on estimates of efficacy. Most studies of seasonal prophylaxis were initiated when influenza virus activity was detected locally or when virus was identified in the community and there was an increase in the observed cases of ILI. However, only two studies58,70 described the rationale for the length of prophylaxis administered, typically as a result of cessation of local activity. Therefore, the proportion of the influenza season across which subjects received prophylaxis varied from study to study. This variation in the period of prophylaxis is especially pertinent, as the risk of developing SLCI following antiviral prophylaxis is considered to be ongoing, with an apparent drop-off in efficacy on cessation of prophylaxis. Additional consideration should be afforded to the timing of the measurement of the primary outcome of SLCI in relation to the prophylactic period. In most cases, SLCI was reported across the whole prophylactic period. Some studies undertook additional analyses of data from days 2–4 of prophylaxis onwards, in order to exclude subjects who may have been infected with influenza virus prior to receipt of prophylaxis, but in whom clinical illness did not manifest until the early stages of the prophylaxis period. Only a small number of trials undertook follow-up measurement of SLCI beyond the period of prophylaxis, with obvious limitations for evaluation of any longer-term outcomes, such as the potential impact of subclinical infection on subjects. Variation was observed between the post-exposure prophylaxis trials undertaken in households in terms of whether index cases were treated with antivirals, which would be expected to have an impact on the transmission of virus to contacts.
An additional area of inconsistency between the different studies was the definition of clinical or symptomatic influenza, which was used to define SLCI. Around half the included studies defined symptomatic influenza as a raised temperature plus one or two additional symptoms, while other studies defined it as the presence of at least two of a list of symptoms which included raised temperature as one of the options. Also, of the 12 studies giving a specific value for a raised temperature, eight used ≥ 37.8°C, while three used ≥ 37.2°C and one used ≥ 37.3 °C. The study by Ambrozaitis et al. 76 defined SLCI as the presence of a new influenza-like sign or symptom, but also reported separately cases of ‘febrile SLCI’, which was defined as a new symptom plus a temperature of ≥ 37.8°C (and gave fewer cases than SLCI alone). Therefore, the number of cases of SLCI identified, and the protective efficacies reported by the different studies may vary depending on the definition of SLCI used.
The external validity of the RCTs must also be considered. A study by Diggory et al. 136 previously demonstrated that elderly individuals experienced difficulties in loading and priming the Diskhaler, by means of which zanamivir is administered by oral inhalation, and suggested that such practical difficulties posed a barrier to use among older patients. Conversely, the adherence data presented within the identified zanamivir trials would suggest that the use of the Diskhaler was acceptable to elderly study participants. 76,78 However, subjects who were unable to understand study personnel were excluded from trial participation by Ambrozaitis et al. 76 and Gravenstein et al.,78 while a requirement of participation in the trials by Monto et al. 47 and LaForce et al. 75 was that subjects should be able to use the Diskhaler adequately. It is therefore important to consider that individuals with low cognitive function or poor manual dexterity would not be represented in some of the study populations, and that such groups may experience difficulties in administering zanamivir independently in clinical practice. Similar external validity issues apply to the trials by Peters et al. 64 and Welliver et al. 49 in which individuals scoring below 7 on a mental status questionnaire were excluded from participation. Such patients may require support in taking oral antiviral prophylaxis.
It is important to highlight the emerging clinical evidence surrounding serious adverse events caused by NIs, in order to reflect the effects of these interventions on patients in clinical practice. Although a higher incidence of severe adverse events in oseltamivir and zanamivir was not apparent in the RCTs identified in this review, the occurrence of serious neuropsychiatric events among a minority of patients treated with NIs has been described;20,137 these circumstances should be monitored and taken into account during the interpretation of this evidence. Indeed, the assumptions made in the economic analysis reflect the current uncertainties regarding the incidence, duration and quality of life impact of adverse events caused by individual prophylactic drugs.
The emergence of variants of influenza that are resistant to amantadine, oseltamivir and/or zanamivir has significant potential to reduce the efficacy of these interventions in clinical practice. Although a number of identified trials tested viral isolates for resistance to oseltamivir and zanamivir in vitro and found no evidence of reduced sensitivity, as noted in Chapter 1 and Chapter 3, the emergence of strains of influenza resistant to amantadine, in particular, and also oseltamivir has been demonstrated and it is therefore important that, during interpretation of the clinical effectiveness evidence, such issues relating to antiviral resistance should be taken into account. Susceptibility should be continued to be monitored and testing of isolates should continue to be undertaken in future clinical trials. Variation in the levels of resistance to antivirals among influenza isolates was taken into account in the cost-effectiveness analysis. Although the base case assumes oseltamivir resistance to be 0 (as current levels of resistance to oseltamivir were considered sufficiently low to warrant exclusion from the base case), multiple sensitivity analyses were undertaken in order to assess the impact of variation in levels of resistance among influenza strains to the interventions under study. It should be noted that in the 2 weeks preceding completion of this assessment report, the HPA issued a press release stating that approximately 5% (8/162) of H1N1 influenza tested isolates were resistant to oseltamivir. However, further research and monitoring are required to fully assess the impact of this resistance. The sensitivity analysis undertaken using the economic model suggests that low levels of resistance do not have a marked impact on the cost-effectiveness of oseltamivir. However, increasing levels of resistance to oseltamivir do have the capacity to dramatically influence the conclusions of the economic analysis. It is therefore of key importance that the results of the economic analysis are interpreted in the light of current levels of influenza activity and resistance.
A further problem, noted in Chapter 4, is the complete absence of preference-based estimates of the impact of influenza and influenza prophylaxis on HRQoL. In addition, systematic searches were unable to identify robust estimates of the impact of influenza complications on quality of life. Consequently, the benefit side of the economic analysis is based entirely on an intermediate outcome measure (SLCI) and indirect estimates of its impact on health outcomes.
Chapter 7 Conclusions
The availability of clinical effectiveness data used to inform the cost-effectiveness modelling was limited for a number of population subgroups. This should be considered during the interpretation of the review findings.
Conclusions on the clinical effectiveness of influenza prophylaxis
Few data relating to the use of amantadine in prophylaxis could be identified and were taken from older trials of poorer quality. Oseltamivir and zanamivir were demonstrated to be effective in preventing SLCI in a number of subgroups. Interventions appeared to be well tolerated by subjects, with a relatively low incidence of few drug-related adverse events and drug-related withdrawals. Very limited evidence could be identified for the effectiveness of the interventions in preventing complications and hospitalisations and in minimising length of illness and time to return to normal activities. No data were identified relating to health-related quality of life or mortality outcomes. The increasing emergence of antiviral resistance among influenza isolates (particularly in the case of amantadine but also for oseltamivir) and the high frequency of adverse events associated with amantadine pose significant challenges to the use of the interventions in clinical practice and, whilst not directly reflected within the trials identified in the review, such issues must be considered during interpretation of the findings from the clinical effectiveness review.
Conclusions on the cost-effectiveness of influenza prophylaxis
Seasonal prophylaxis
In healthy children
Amantadine and zanamivir as seasonal prophylaxis are expected to be dominated or extendedly dominated in the healthy children subgroup. The proposed reduction in the price of zanamivir does not affect this finding. The incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be greater than £44,000 per QALY gained.
In at-risk children
Amantadine and zanamivir as seasonal prophylaxis are expected to be dominated or extendedly dominated in the at-risk children subgroup. Again, the proposed reduction in the price of zanamivir does not affect this finding. The incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be around £17,000 per QALY gained for at-risk children who have not been vaccinated. For at-risk children who have previously been vaccinated, the incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be in excess of £50,000 per QALY gained.
In healthy adults
Amantadine and zanamivir as seasonal prophylaxis are expected to be dominated or extendedly dominated in the healthy adult subgroup. The incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be around £148,000 per QALY gained for healthy adults who have not been vaccinated and greater than £427,000 per QALY gained for healthy adults who have been vaccinated.
In at-risk adults
Based on the current list price for zanamivir, the model suggests that both amantadine and zanamivir are ruled out of the analysis in at-risk adults. The incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be around £64,000 per QALY gained in unvaccinated at-risk adults and around £187,000 per QALY gained in previously vaccinated at-risk adults. When the proposed price reduction for zanamivir is included in the analysis for at-risk adults, zanamivir is no longer dominated. The incremental cost-effectiveness of seasonal prophylaxis using zanamivir versus no prophylaxis is expected to be around £53,000 per QALY gained in unvaccinated at-risk adults and £157,000 per QALY gained in at-risk adults who have previously been vaccinated. The incremental cost-effectiveness of oseltamivir is expected to be around £108,000 per QALY gained in unvaccinated at-risk adults and around £314,000 per QALY gained in previously vaccinated at-risk adults.
In healthy elderly individuals
In this subgroup, amantadine and zanamivir are expected to be dominated or extendedly dominated. The proposed reduction in the price of zanamivir does not affect this result. The incremental cost-effectiveness of oseltamivir versus no prophylaxis in healthy elderly individuals who have not been vaccinated is expected to be around £50,000 per QALY gained. For previously vaccinated healthy elderly individuals, the incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be greater than £120,000 per QALY gained.
In at-risk elderly individuals
In this subgroup, amantadine and zanamivir are expected to be extendedly dominated despite the proposed reduction in the price of zanamivir. The incremental cost-effectiveness of oseltamivir versus no prophylaxis in at-risk elderly individuals who have not been vaccinated is expected to be around £38,000 per QALY gained. For previously vaccinated at-risk elderly individuals, the incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be around £94,000 per QALY gained.
Post-exposure prophylaxis
In healthy children
Amantadine and oseltamivir as post-exposure prophylaxis are expected to be dominated or extendedly dominated in the healthy children subgroup. For unvaccinated healthy children, the incremental cost-effectiveness of zanamivir post-exposure prophylaxis versus no prophylaxis is expected to be around £23,000 per QALY gained at the current list price, and around £19,000 per QALY gained when the proposed price reduction for zanamivir is included in the analysis. For vaccinated healthy children, the incremental cost-effectiveness of zanamivir is expected to be at least £59,000 per QALY gained; this estimate includes the proposed price reduction for zanamivir.
For children under the age of 5 years, oseltamivir is the only licensed antiviral prophylaxis option. The incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be around £24,000 per QALY gained and £74,000 per QALY gained in unvaccinated and vaccinated groups respectively.
In at-risk children
Amantadine and oseltamivir as post-exposure prophylaxis are expected to be dominated or extendedly dominated in the at-risk children subgroup. For unvaccinated at-risk children, the incremental cost-effectiveness of zanamivir post-exposure prophylaxis versus no prophylaxis is expected to be around £8000 per QALY gained at the current list price, and around £6000 per QALY gained when the proposed price reduction for zanamivir is included in the analysis. For vaccinated at-risk children, the incremental cost-effectiveness of zanamivir is expected to be around £28,000 per QALY gained at the current list price, and £23,000 per QALY gained when the proposed price reduction is included in the analysis.
For at-risk children under the age of 5 years, the incremental cost-effectiveness of oseltamivir versus no prophylaxis is expected to be around £9000 per QALY gained for unvaccinated at-risk children and around £29,000 per QALY gained for vaccinated at-risk children.
In healthy adults
Amantadine and zanamivir prophylaxis are expected to be dominated or extendedly dominated in the healthy adult subgroup. The proposed price reduction for zanamivir does not affect this result. For unvaccinated healthy adults, the incremental cost-effectiveness of oseltamivir post-exposure prophylaxis versus no prophylaxis is expected to be around £34,000 per QALY gained. For previously vaccinated healthy adults, the incremental cost-effectiveness of oseltamivir is expected to be around £104,000 per QALY gained.
In at-risk adults
Amantadine and zanamivir prophylaxis are expected to be dominated or extendedly dominated in the at-risk adult subgroup. The proposed price reduction for zanamivir does not affect this result. For unvaccinated at-risk adults, the incremental cost-effectiveness of oseltamivir post-exposure prophylaxis versus no prophylaxis is expected to be around £13,000 per QALY gained. For previously vaccinated at-risk adults, the incremental cost-effectiveness of oseltamivir is expected to be around £44,000 per QALY gained.
In healthy elderly individuals
Amantadine and zanamivir prophylaxis are expected to be dominated or extendedly dominated in the healthy elderly subgroup. The proposed price reduction for zanamivir does not affect this result. For unvaccinated healthy elderly individuals, the incremental cost-effectiveness of oseltamivir post-exposure prophylaxis versus no prophylaxis is expected to be around £11,000 per QALY gained. For previously vaccinated healthy elderly individuals, the incremental cost-effectiveness of oseltamivir is expected to be around £28,000 per QALY gained.
In at-risk elderly individuals
Amantadine and zanamivir as post-exposure prophylaxis are expected to be dominated or extendedly dominated in the at-risk elderly subgroup. For unvaccinated at-risk elderly individuals, the incremental cost-effectiveness of oseltamivir post-exposure prophylaxis versus no prophylaxis is expected to be around £8000 per QALY gained. For vaccinated at-risk elderly individuals, the incremental cost-effectiveness of oseltamivir is expected to be around £22,000 per QALY gained.
Recommendations for research
It should be noted that increasing levels of resistance to antiviral prophylaxis have the capacity to dramatically influence the conclusions of the economic analysis. The results of the economic analysis should be interpreted in the light of current levels of influenza activity and resistance. The evidence base relating to the clinical effectiveness and cost-effectiveness of amantadine, oseltamivir and zanamivir in seasonal and post-exposure influenza prophylaxis would be reinforced by further research in the following areas:
-
Additional RCTs in subgroups for which data are currently lacking (as described in Chapter 6 and including assessments of oseltamivir in seasonal prophylaxis in children, at-risk adults and healthy elderly subjects; zanamivir in seasonal prophylaxis in children and healthy elderly subjects; and post-exposure prophylaxis trials of the interventions in elderly subjects and individuals with low cognitive function and/or manual dexterity)
-
RCTs in which the follow-up period extends beyond the duration of prophylaxis
-
head-to-head RCTs in which the clinical effectiveness of amantadine, oseltamivir and/or zanamivir in different subgroups is directly compared
-
quality of life studies to inform future economic decision modelling
-
further research concerning the incidence and management of complications caused by influenza.
Acknowledgements
We would like to thank Piers Mook, Joanna Ellis and Maria Zambon at the Health Protection Agency for providing advice and data for the model. We would also like to thank Alex Elliot, Andrew Ross and Douglas Fleming (Director) at the Royal College of General Practitioners for providing expert input and data for the modelling process. We would like to acknowledge Allan Wailoo, David Turner, John Brazier and Jim Chilcott for providing further methodological advice within the model development process. Myfanwy Lloyd-Jones provided methodological suggestions for the systematic review of clinical effectiveness. We would like to thank Hazel Pilgrim for her comments and validation of the model. Finally, we would like to thank Dr Rod Taylor (Peninsula Technology Assessment Group), Dr Martin Wiselka (University Hospitals of Leicester NHS Trust) and Dr Wei Shen Lim (Nottingham City Hospital) for commenting on this report.
Contribution of authors
Paul Tappenden (Senior Cost-effectiveness Modeller/Research Fellow) was the Assessment Group lead, undertook the cost-effectiveness review and developed the cost-effectiveness model. Rachel Jackson (Research Associate) and Katy Cooper (Research Associate) undertook the clinical effectiveness review. Emma Simpson (Research Fellow) was involved in the preparation of the scope and protocol and advised on the clinical effectiveness review. Angie Rees (Information Officer) performed the literature searches. Robert Read (Honorary Consultant Physician in Infectious Diseases) and Karl Nicholson (Consultant Physician and Professor of Infectious Diseases) provided clinical input to both the systematic review and health economic modelling throughout the assessment. Andrea Shippam helped in the retrieval of papers and in preparing and formatting the report. Jim Chilcott and Eva Kaltenthaler are guarantors.
About ScHARR
The School of Health and Related Research (ScHARR) is one of the four Schools that comprise the Faculty of Medicine at the University of Sheffield. ScHARR brings together a wide range of medical- and health-related disciplines including public health, general practice, mental health, epidemiology, health economics, management sciences, medical statistics, operational research and information science. It includes the Sheffield unit of the Trent Research and Development Support Unit (RDSU), which is funded by NIHR to facilitate high-quality health services research and capacity development.
The ScHARR Technology Assessment Group (ScHARR-TAG) synthesises research on the effectiveness and cost-effectiveness of health-care interventions for the NHS R&D Health Technology Assessment (HTA) Programme on behalf of a range of policy makers, including the National Institute for Health and Clinical Excellence (NICE). ScHARR-TAG is part of a wider collaboration of six units from other regions. The other units are: Southampton Health Technology Assessment Centre (SHTAC), University of Southampton; Aberdeen Health Technology Assessment Group (Aberdeen HTA Group), University of Aberdeen; Liverpool Reviews & Implementation Group (LRiG), University of Liverpool; Peninsular Technology Assessment Group (PenTAG), University of Exeter; NHS Centre for Reviews and Dissemination, University of York; and West Midlands Health Technology Assessment Collaboration (WMHTAC), University of Birmingham.
Disclaimers
The views expressed in this publication are those of the authors and not necessarily those of the HTA Programme or the Department of Health.
References
- Wiselka M. Influenza: diagnosis, management and prophylaxis. BMJ 1994;308:1341-5.
- Nicholson KG, Nicholson KG, Webster RG, Hay AJ. Textbook of influenza. Oxford: Blackwell Science; 1998.
- Tillett HE, Smith JWG, Gooch CD. Excess deaths attributable to influenza in England and Wales: age at death and certified cause. Int J Epidemiol 1983;12:344-52.
- Nicholson KG. Impact of influenza and respiratory syncytial virus on mortality in England and Wales from January 1975 to December 1990. Epidemiol Infect 1996;116:51-63.
- Fleming DM. The contribution of influenza to combined acute respiratory infections, hospital admissions and deaths in winter. Commun Dis Publ Health 2000;3:32-8.
- Ruigrok RWH, Nicholson KG, Webster RG, Hay AJ. Textbook of influenza. Oxford: Blackwell Science; 1998.
- Jefferson T, Demicheli V, Nicholson KG, Webster RG, Hay AJ. Textbook of influenza. Oxford: Blackwell Science; 1998.
- Health Protection Agency (HPA) 2007. www.hpa.org.uk/infections/topics_az/influenza/seasonal/default.htm.
- Douglas RG. Prophylaxis and treatment of influenza. New Engl J Med 1990;322:443-50.
- Turner D, Wailoo A, Nicholson K, Cooper N, Sutton, A, Abrams K. Systematic review and economic decision modelling for the prevention and treatment of influenza A and B. Health Technol Assess 2003;7:1-170.
- Health Protection Agency (HPA) Centre for Infections (CfI) . HPA National Influenza Season Summary 2006 2007 2007.
- Meier CR, Napalkov PN, Wegmuller Y, Jefferson T, Jick H. Population-based study on incidence, risk factors, clinical complications and drug utilisation associated with influenza in the United Kingdom. Eur J Clin Microbiol Infect Dis 2000;19:834-42.
- Nicholson KG, Wood JM, Zambon M. Influenza. Lancet 2003;362:1733-45.
- British Medical Association and Royal Pharmaceutical Society of Great Britain . British National Formulary 2007.
- Department of Health Immunisation programme for poultry workers . Guidance on the Implementation of a Seasonal Flu Vaccination Programme for Poultry Workers 2007.
- National Institute for Clinical Excellence . Guidance on the Use of Oseltamivir and Amantadine for the Prophylaxis of Influenza 2003.
- NHS and National Institute for Clinical Excellence . Guidance on the Use of Zanamivir, Oseltamivir and Amantadine for the Treatment of Influenza 2003.
- Joint Formulary Committee . British National Formulary 2007.
- Department of Health . The Influenza Immunisation Programme 2007 2008 2007.
- Roche . Tamiflu (oseltamivir) Influenza Prophylaxis: Achieving Clinical Excellence in Influenza Prophylaxis 2007:1-197.
- Monto AS, McKimm-Breschkin JL, Macken C, Hampson AW, Hay A, Klimov A, et al. Detection of influenza viruses resistant to neuraminidase inhibitors in global surveillance during the first 3 years of their use. Antimicrob Agents Chemother 2006;50:2395-402.
- Department of Health . National Service Framework for Older People 2001.
- Department of Health . National Service Framework for Coronary Heart Disease 2000.
- Alliance Pharmaceuticals . Lysovir 100 mg capsules. Summary of Product Characteristics 2005.
- Roche . Tamiflu 12 mg/ml powder for oral suspension. Summary of Product Characteristics 2007.
- Roche . Tamiflu 30 mg hard capsules. Summary of Product Characteristics 2007.
- Roche . Tamiflu 45 mg hard capsules. Summary of Product Characteristics 2007.
- Roche . Tamiflu 75 mg hard capsules. Summary of Product Characteristics 2007.
- GlaxoSmithKline . Relenza 5 mg/dose inhalation powder. Summary of Product Characteristics 2006.
- Aoki F, Nicholson KG, Webster RG, Hay AJ. Textbook of influenza. Oxford: Blackwell Science; 1998.
- Hayden FG. Antiviral resistance in influenza viruses – implications for management and pandemic response. New Engl J Med 2006;354:785-8.
- Schilling M, Gravenstein S, Drinka P, Cox N, Krause P, Povinelli L, et al. Emergence and transmission of amantadine-resistant influenza A in a nursing home. J Am Geriatr Soc 2004;52:2069-73.
- Jefferson T, Demicheli V, Di Pietrantonj C, Rivetti D. Amantadine and Rimantadine for Influenza A in Adults 2006.
- Penn C, Nicholson KG, Webster RG, Hay AJ. Textbook of influenza. Oxford: Blackwell Science; 1998.
- Moher D, Cook DJ, Eastwood S, Olkin I, Rennie D, Stroup DF. and for the QUOROM Group. Improving the quality of reports of meta-analyses of randomised controlled trials: the QUOROM statement. Lancet 1999;354:1896-900.
- NHS Centre for Reviews and Dissemination . Report 4: Undertaking Systematic Reviews on Effectiveness; CRDs Guidance for Those Carrying Out or Commissioning Reviews 2001.
- Hess P. Amantadine and rimantadine in the prophylaxis of influenza A. Med Hygiene 1982;40.
- Plesnik V, Heinz F, Bindas B, Cechova D, Eliasova J, Galetkova A, et al. Controlled study of influenza prophylaxis by VUFB amantadin. Cesk Epidemiol Mikrobiol Imunol 1977;26:216-26.
- Quilligan JJ, Hirayama M, Baernstein HD. The prevention of A2-influenza in children by chemoprophylaxis with amantadine hydrochloride. J Suisse Med 1966;96:1689-92.
- Smorodintsev AA, Romanov YA, Rumovski VI, Zlydniko DM. Effectiveness of hydrochloric amantadine (midantane) in prevention of artificially induced influenza infection. Vopr Virusol 1972:152-6.
- Kashiwagi S, Kudoh S, Watanabe A, Yoshimura I. Efficacy and safety of the selective oral neuraminidase inhibitor oseltamivir for prophylaxis against influenza – placebo-controlled double-blind multicenter phase III trial. Kansenshogaku Zasshi (Journal of the Japanese Association for Infectious Diseases) 2000;74:1062-76.
- Shinjoh M, Sato S, Sugaya N, Mitamura K, Takeuchi Y, Kosaki K, et al. Effect of post-exposure prophylaxis with oseltamivir for those in contacts with influenza patients in pediatric wards. Kansenshogaku Zasshi (Journal of the Japanese Association for Infectious Diseases) 2004;78:262-9.
- Diaz-Pedroche C, Lizasoain M, Lopez-Medrano F, Escalante F, Lumbreras C, Folgueira D, et al. Nosocomial outbreak of influenza in high-risk hematological patients. Efficacy of control measures and the use of zanamivir. Enferm Infecc Microbiol Clin 2006;24:10-3.
- GlaxoSmithKline Pharmaceuticals . Drugs for the Prophylaxis of Influenza – Review of Zanamivir 2007.
- Halloran M, Hayden FG, Yang Y, Longini IM, Monto AS. Antiviral effects on influenza viral transmission and pathogenicity: observations from household-based trials. Am J Epidemiol 2007;165:212-21.
- Hayden FG, Gubareva LV, Monto AS, Klein TC, Elliott MJ, Hammond JM, et al. Inhaled zanamivir for the prevention of influenza in families. New Engl J Med 2000;343:1282-9.
- Monto AS, Pichichero ME, Blanckenberg SJ, Ruuskanen O, Cooper C, Fleming DM, et al. Zanamivir prophylaxis: an effective strategy for the prevention of influenza types A and B within households. J Infect Dis 2002;186:1582-8.
- Hayden FG, Belshe R, Villanueva C, Lanno R, Hughes C, Small I, et al. Management of influenza in households: a prospective, randomized comparison of oseltamivir treatment with or without postexposure prophylaxis. J Infect Dis 2004;189:440-9.
- Welliver R, Monto AS, Carewicz O, Schatteman E, Hassman M, Hedrick J, et al. Effectiveness of oseltamivir in preventing influenza in household contacts: a randomized controlled trial. JAMA 2001;285:748-54.
- Jefferson TO, Demicheli V, Deeks JJ, Rivetti D. Amantadine and rimantadine for preventing and treating influenza A in adults. Cochrane Database of Systematic Reviews; 2000.
- Finklea JF, Hennessy AV, Davenport FM. A field trial of amantadine prophylaxis in naturally-occurring acute respiratory illness. Am J Epidemiol 1967;85:403-12.
- Quilligan JJ, Hirayama M, Baernstein HD. The suppression of A2 influenza in children by the chemoprophylactic use of amantadine. J Pediatr 1966;69:572-5.
- Leung P, McIntosh K, Chai H. Amantadine prophylaxis against influenza A/USSR in children with chronic asthma. J Allergy Clin Immunol 1979;63.
- Wright PF, Khaw KT, Oxman MN, Shwachman H. Evaluation of the safety of amantadine-HC1 and the role of respiratory viral infections in children with cystic fibrosis. J Infect Dis 1976;134:144-9.
- Pettersson RF, Hellström PE, Penttinen K, Pyhälä R, Tokola O, Vartio T, et al. Evaluation of amantadine in the prophylaxis of influenza A (H1N1) virus infection: a controlled field trial among young adults and high-risk patients. J Infect Dis 1980;142:377-83.
- Leeming JT. Amantidine hydrochloride and the elderly. BMJ 1969;1:313-14.
- Reuman PD, Bernstein DI, Keefer MC, Young EC, Sherwood JR, Schiff GM. Efficacy and safety of low dosage amantadine hydrochloride as prophylaxis for influenza A. Antiviral Res 1989;11:27-40.
- Aoki FY, Stiver HG, Sitar DS, Hammond GW, Milley EV, Vermeersch C, et al. Potential of influenza vaccine and amantadine to prevent influenza A illness in Canadian forces personnel 1980–1983. Mil Med 1986;151:459-65.
- Payler DK, Purdham PA. Influenza A prophylaxis with amantadine in a boarding school. Lancet 1984;1:502-4.
- Smorodintsev A, Karpuchin G, Zlydnikov D. The prospect of amantadine for prevention of influenza A in humans (effectiveness of amantadine during influenza A2/Hong Kong epidemics in January–February 1969 in Leningrad). Ann N Y Acad Sci 1970;173:44-73.
- Smorodintsev AA, Karapuhi GI, Zlydniko DM, Malyseva AM, Svecova EG, Burov SA, et al. Prophylactic effectiveness of amantadine hydrochloride in an epidemic of Hong-Kong influenza in Leningrad in 1969. Bull World Health Organ 1970;42:865-72.
- Smorodintsev AA, Zlydnikov DM, Kiseleva AM, Romanov JA, Kazantsev AP, Rumovsky VI. Evaluation of amantadine in artificially induced A2 and B influenza. JAMA 1970;213:1448-54.
- Sears SD, Clements ML. Protective efficacy of low-dose amantadine in adults challenged with wild-type influenza A virus. Antimicrob Agents Chemother 1987;31:1470-3.
- Peters PH, Gravenstein S, Norwood P, De Bock V, Van Couter A, Gibbens M, et al. Long-term use of oseltamivir for the prophylaxis of influenza in a vaccinated frail older population. J Am Geriatr Soc 2001;49:1025-31.
- De Bock V, Peters P, Von Planta T-A, Gibbens M, Ward P. Oral oseltamivir for prevention of influenza in the frail elderly. Clin Microbiol Infect 2000;6.
- Hayden FG, Atmar RL, Schilling M, Johnson C, Poretz D, Paar D, et al. Use of the selective oral neuraminidase inhibitor oseltamivir to prevent influenza. New Engl J Med 1999;341:1336-43.
- Hayden FG, Jennings L, Robson R, Schiff G, Jackson H, Rana B, et al. Oral oseltamivir in human experimental influenza B infection. Antiviral Ther 2000;5:205-13.
- Jefferson TO, Demicheli V, Di Pietrantonj C, Jones M, Rivetti D. Neuraminidase inhibitors for preventing and treating influenza in healthy adults. Cochrane Database of Systematic Reviews; 2006.
- Schilling M, Povinelli L, Krause P, Gravenstein M, Ambrozaitis A, Jones H, et al. Efficacy of zanamivir for chemoprophylaxis of nursing home influenza outbreaks. Vaccine 1998;16:1771-4.
- Monto AS, Robinson DP, Louise M, James H, Hinson M, Elliott MJ, et al. Zanamivir in the prevention of influenza among healthy adults: a randomized controlled trial. JAMA 1999;282:31-5.
- Monto AS, Robinson D, Griffin AD, Edmundson S. The effects of zanamivir on productivity in the prevention of influenza among healthy adults. J Antimicrob Chemother 1999;44.
- Kaiser L, Henry D, Flack NP, Keene O, Hayden FG. Short-term treatment with zanamivir to prevent influenza: results of a placebo-controlled study. Clin Infect Dis 2000;30:587-9.
- Hayden F, Belshe R, Villanueva C, Lanno R, Small I, Hughes C, et al. Oral oseltamivir prevents the spread of influenza between children in households. Abstracts of the Interscience Conference on Antimicrobial Agents and Chemotherapy 2002;42.
- Belshe R, Hayden F, Carewicz O, Lanno R, Martin C, Hughes C, et al. Effectiveness of oral oseltamivir in preventing spread of influenza-like illness in households with proven influenza. Abstracts of the Interscience Conference on Antimicrobial Agents and Chemotherapy 2001;41.
- LaForce C, Man CY, Henderson FW, McElhaney JE, Hampel FC, Bettis R, et al. Efficacy and safety of inhaled zanamivir in the prevention of influenza in community-dwelling, high-risk adult and adolescent subjects: a 28-day, multicenter, randomized, double-blind, placebo-controlled trial. Clin Ther 2007;29:1579-90.
- Ambrozaitis A, Gravenstein S, Van Essen G. A, Rubinstein E, Balciuniene L, Stikleryte A, et al. Inhaled zanamivir versus placebo for the prevention of influenza outbreaks in an unvaccinated long-term care population. J of the American Medical Directors Association 2005;6:367-74.
- Ambrozaitis, A, Van Essen G, Rubinstein E, Balciuniene L, Stikleryte A, Gravenstein S, et al. Inhaled zanamivir versus placebo for the prevention of influenza outbreaks in an unvaccinated long-term care population (NAIA3004). J Am Geriatr Soc 2001;49:S130-1.
- Gravenstein S, Drinka P, Osterweil D, Schilling M, Krause P, Elliott M, et al. Inhaled zanamivir versus rimantadine for the control of influenza in a highly vaccinated long-term care population. J Am Med Dir Assoc 2005;6:359-66.
- Jefferson T, Deeks JJ, Demicheli V, Rivetti D, Rudin M. Amantadine and rimantadine for preventing and treating influenza A in adults. Cochrane Database of Systematic Reviews; 2004.
- European Medicines Agency Scientific discussion . Tamiflu-H-402-II-20 2006.
- Matheson NJ, Harnden AR, Perera R, Sheikh A, Symmonds-Abrahams M. Neuraminidase inhibitors for preventing and treating influenza in children. Cochrane Database of Systematic Reviews; 2007.
- Aoki FY, Boivin G, Roberts N. Influenza virus susceptibility and resistance to oseltamivir. Antiviral Ther 2007;12:603-16.
- Hatakeyama S, Sugaya N, Ito M, Yamazaki M, Ichikawa M, Kimura K, et al. Emergence of influenza B viruses with reduced sensitivity to neuraminidase inhibitors. JAMA 2007;297:1435-42.
- Escuret V, Frobert E, Bouscambert-Duchamp M, Sabatier M, Grog I, Valette M, et al. Detection of human influenza A (H1N1) and B strains with reduced sensitivity to neuraminidase inhibitors. J Clin Virol 2007;41:25-8.
- Kiso M, Mitamura K, Sakai-Tagawa Y, Shiraishi K, Kawakami C, Kimura K, et al. Resistant influenza A viruses in children treated with oseltamivir: descriptive study. Lancet 2004;364:759-65.
- European Centre for Disease Prevention and Control . Emergence of Seasonal Influenza Viruses Type A H1N1 With Oseltamivir Resistance in Some European Countries at the Start of the 2007–8 Influenza Season 2008.
- Health Protection Agency . Oseltamivir Resistance in European Influenza Viruses – Update 2008.
- Cooper NJ, Sutton AJ, Abrams KR, Wailoo A, Turner DA, Nicholson KG. Effectiveness of neuraminidase inhibitors in treatment and prevention of influenza A and B: systematic review and meta-analyses of randomised controlled trials. BMJ 2003;326:1235-9.
- Drummond MF, Sculpher MJ, Torrance GW, O’Brien BJ, Stoddart GL. Methods for the economic evaluation of health care programmes. New York: Oxford University Press; 2007.
- Eddy DM. Assessing medical technology. Washington, DC: National Academy Press; 1985.
- Sander B, Hayden FG, Gyldmark M, Garrison LP. Post-exposure influenza prophylaxis with oseltamivir: cost effectiveness and cost utility in families in the UK. Pharmacoeconomics 2006;24:373-86.
- Risebrough NA, Bowles SK, Simor AE, McGeer A, Oh PI. Economic evaluation of oseltamivir phosphate for postexposure prophylaxis of influenza in long-term care facilities. J Am Geriatr Soc 2005;53:444-51.
- Scuffham P, West P. Economic evaluation of strategies for the control and management of influenza in Europe. Vaccine 2002;20:2562-78.
- Demicheli V, Jefferson T, Rivetti D, Deeks J. Prevention and early treatment of influenza in healthy adults. Vaccine 2000;18:957-1030.
- Patriarca PA, Arden NH, Koplan JP, Goodman RA. Prevention and control of type A influenza infections in nursing homes: benefits and costs of four approaches using vaccination and amantadine. Ann Intern Med 1987;107:732-40.
- National Institute for Health and Clinical Excellence . Guide to the Methods of Technology Appraisal 2004.
- Monto AS, Gunn RA, Bandyk MG, King CL. Prevention of Russian influenza by amantadine. JAMA 1979;241:1003-7.
- Cox NJ, Subbarao K. Global epidemiology of influenza: past and present. Annu Rev Med 2000;51:407-21.
- Kaiser L, Wat C, Mills T, Mahoney P, Ward P, Hayden F. Oseltamivir prevents complications from influenza-related lower respiratory tract infections. ACOG Clin Rev 2003;8.
- Stouthard M, Essink-Bot M, Bonsel G, . Disability weights for diseases in the Netherlands. Rotterdam, Netherlands: Department of Public Health, Erasmus University Rotterdam; 1997.
- Department of Health and Human Services . National Ambulatory Medical Care Survey (NACMS) 1997.
- Curtis L, Netten A. Unit costs of health and social care. Canterbury: Personal Social Services Research Unit (PSSRU); 2006.
- MIMS . The Prescribing Reference for General Practice 2007.
- van Noort SP. Gripenet: an internet-based system to monitor influenza-like illness uniformly across Europe. Euro Surveill 2007.
- MVH Group . The Measurement and Valuation of Health 1994.
- Ontario Ministry of Health and Long Term Care . Ontario Ministry of Health Schedule of Benefits and Fees 2007.
- Ontario Case Cost Initiative . Average Costs Per Case Mix Group (CMG) 2000–2001 2001.
- Wailoo A, Abrams K, Turner D, Sutton AJ, Cooper NJ, Nicholson KG. Review of influenza post-exposure prophylaxis. DSU report to the National Institute for Clinical Excellence; 2003.
- Murray CJ, Lopez AD. The global burden of disease: a comprehensive assessment of mortality and disabilities from diseases, injuries and risk factors in 1990 and projected to 2020. Cambridge, MA: Harvard School of Public Health; 1996.
- Fleming DM. The contribution of influenza to combined acute respiratory infections, hospital admissions and deaths in winter. Comm Dis Publ Health 2000;3:32-8.
- Netten A, Curtis L. Unit costs of health and social care. Canterbury: Personal Social Services Research Unit (PSSRU); 2000.
- CHKS Ltd . Hospitalisation Data for England and Wales 1995–1999 2000.
- Department of Health . Prescription Cost Analysis 2000.
- Department of Health . Drug Tariff National Health Service, England and Wales 1999.
- Patriarca PA, Weber JA, Parker RA, Orenstein WA, Hall WN, Kendal AP, et al. Risk factors for outbreaks of influenza in nursing homes. Am J Epidemiol 1986;124:114-9.
- National Institute for Clinical Excellence . Guidance on the Use of Zanamivir, Oseltamivir and Amantadine for the Treatment of Influenza 2003.
- Smith S, Demicheli V, Pietrantonj C, Harnden AR, Jefferson T, Matheson NJ, et al. Vaccines for preventing influenza in healthy children (review). The Cochrane Library; 2007.
- Jefferson TO, Rivetti D, Pietrantonj C, Rivetti A, Demicheli V. Vaccines for preventing influenza in healthy adults (review). The Cochrane Library; 2007.
- Rivetti D, Jefferson TO, Thomas R, Rudin M, Rivetti A, Pietrantonj C, et al. Vaccines for preventing influenza in the elderly (review). The Cochrane Library; 2007.
- Gross PA, Hermogenes AW, Sacks HS, Lau J, Levandowski RA. The efficacy of influenza vaccine in elderly persons: a meta-analysis and review of the literature. Ann Intern Med 1995;123:518-27.
- Nichol KL, Margolis KL, Lind A, Murdoch M, McFadden R, Hauge M, et al. Side effects associated with influenza vaccination in healthy working adults. A randomized, placebo-controlled trial. Ann Intern Med 1996;156:1546-50.
- Pitman R, Melegaro A, Gelb D, Siddiqui M, Gay N, Edmunds W. Assessing the burden of influenza and other respiratory infections in England and Wales. J Infect 2007;54:530-8.
- Ross AM, Kai J, Salter R, Ross J, Fleming DM. Presentation with influenza-like illness in general practice: implications for use of neuraminidase inhibitors. Commun Dis Publ Health 2000;3:256-60.
- Briggs AH, Ades AE, Price MJ. Probabilistic sensitivity analysis for decision trees with multiple branches: use of the Dirichlet distribution in a Bayesian framework. Med Decis Making 2003;23:341-50.
- Department of Health . NHS Reference Costs 2005–2006 2007.
- Oliveira EC, Marik PE, Colice G. Influenza pneumonia: a descriptive study. Chest 2001;119.
- Griffin ADP. Cost-effectiveness analysis of inhaled zanamivir in the treatment of influenza A and B in high-risk patients. Pharmacoeconomics 2001;19:293-301.
- Rothberg MBH. Management of influenza symptoms in healthy adults: cost-effectiveness of rapid testing and antiviral therapy. J Gen Intern Med 2003;18:808-15.
- Stratton KR, Durch JS, Lawrence RS. Vaccines for the 21st century: a tool for decisionmaking. Washington, DC: National Academy Press; 2007.
- Office for National Statistics . Mortality Statistics: Cause 2006.
- Kind P, Hardman G, Macran S. UK population norms for EQ-5D. Centre for Health Economics Discussion Paper Series; 1999.
- Gupta RK, Zhao H, Cooke M, Harling R, Regan M, Bailey L, et al. Public health responses to influenza in care homes: a questionnaire-based study of local health protection units. J Publ Health 2007;29:88-90.
- Quarles JM, Couch RB, Cate TR, Goswick CB. Comparison of amantadine and rimantadine for prevention of type A (Russian) influenza. Antiviral Res 1981;1:149-55.
- Dolin R, Reichman RC, Madore HP, Maynard R, Linton PN, Webber-Jones J. A controlled trial of amantadine and rimantadine in the prophylaxis of influenza A infection. New Engl J Med 1982;307:580-4.
- Cohen A, Togo Y, Khakoo R, Waldman R, Sigel M. Comparative clinical and laboratory evaluation of the prophylactic capacity of ribavirin, amantadine hydrochloride and placebo in induced human influenza type A. J Infect Dis 1976;133:A114-20.
- Diggory P, Fernandez C, Humphrey A, Jones V, Murphy M. Comparison of elderly people’s technique in using two dry powder inhalers to deliver zanamivir: randomised controlled trial. BMJ 2001;322:577-9.
- Pocock N. US FDA staff recommend additional neuropsychiatric warnings for oseltamivir and zanamivir. NHS National Electronic Library for Medicines; 2007.
Appendix 1 Literature search strategies
MEDLINE search strategy to identify clinical trials
1. Oseltamivir/526
2. (gs 4071 or gs 4104 or gs4104 or gs4071 or tamiflu).mp.
3. Amantadine/
4. amantadine.mp.
5. aman.mp.
6. amanta.mp.
7. amantadin.mp.
8. amantadina.mp.
9. amixx.mp.
10. cerebramed.mp.
11. endantadine.mp.
12. gen-amantadine.mp.
13. infecto-flu.mp.
14. infex.mp.
15. mantadix.mp.
16. midrantan.mp.
17. pms-amantadine.mp.
18. symadine.mp.
19. symmetrel.mp.
20. viregyt.mp.
21. wiregyt.mp.
22. tregor.mp.
23. oseltami.mp.
24. Zanamivir/
25. zanamivir.mp.
26. 2,3-didehydro-2,4-dideoxy-4-guanidino-n-acetyl-d-neuraminic acid.mp.
27. 2,3-didehydro-2,4-dideoxy-4-guanidinyl-n-acetylneuraminic acid.mp.
28. 4-guanidino-2,4-dideoxy-2,3-didehydro-n-acetylneuraminic acid.mp.
29. 4-guanidino-2-deoxy-2,3-didehydro-n-acetylneuraminic acid.mp.
30. 4-guanidino-neu5ac2en.mp.
31. 5-acetylamino-2,6-anhydro-4-guanidino-3,4,5-trideoxy-d-galacto-non-enoic acid.mp. -
32. (gg 167 or gg167).mp.
33. relenza.mp.
34. or/1–33
35. prophyla$.ti,ab.
36. prevent$.ti,ab.
37. 35 or 36
38. 37 and 34
39. randomized controlled trial.pt.
40. controlled clinical trial.pt.
41. randomized controlled trials/
42. random allocation/
43. double blind method/
44. single blind method/
45. or/39–44
46. clinical trial.pt.
47. exp clinical trials/
48. (clin$adj25 trial$).tw.
49. ((singl$or doubl$or trebl$or tripl$) adj25 (blind$or mask$)).tw.
50. placebos/
51. placebo$.tw.
52. random$.tw.
53. research design/
54. or/46–53
55. “comparative study”/
56. exp evaluation studies/
57. follow-up studies/
58. prospective studies/
59. (control$or prospectiv$or volunteer$).tw.
60. (control$or prospectiv$or volunteer$).tw.
61. or/55–60
62. 45 or 54 or 61
63. “animal”/
64. “human”/
65. 63 not 64
66. 62 not 65
67. 66 and 38
68. Influenza, Human/
69. 68 and 67
MEDLINE search strategy to identify utility estimates for influenza and related complications
1. Influenza/
2. (influenza or flu).tw.
3. 1 or 2
4. “Quality of Life”/
5. (quality of life or qol).ti,ab.
6. (quality adjusted life year or qaly).ti,ab.
7. utilit$.ti,ab.
8. Health Status Indicators/
9. disability adjusted life.tw.
10. daly$.tw.
11. (sf36 or sf 36 or short form 36 or shortform 36 or sf thirtysix or sf thirty six or shortform thirtysix or shortform thirty six or short form thirtysix or short form thirty six).tw.
12. (sf6 or sf 6 or short form 6 or shortform 6 or sf six or sfsix or shortform six or short form six).tw.
13. (sf12 or sf 12 or short form 12 or shortform 12 or sf twelve or sftwelve or shortform twelve or short form twelve).tw.
14. (sf16 or sf 16 or short form 16 or shortform 16 or sf sixteen or sfsixteen or shortform sixteen or short form sixteen).tw.
15. (sf20 or sf 20 or short form 20 or shortform 20 or sf twenty or sftwenty or shortform twenty or short form twenty).tw.
16. (euroqol or euro qol or eq5d or eq 5d).tw.
17. (hql or hqol or h qol or hrqol or hr qol).tw.
18. (hye or hyes).tw.
19. health$year$equivalent$.tw.
20. health utilit$.tw.
21. (hui or hui1 or hui2 or hui3).tw.
22. disutili$.tw.
23. rosser.tw.
24. quality of wellbeing.tw.
25. qwb.tw.
26. willingness to pay.tw.
27. standard gamble$.tw.
28. time trade off.tw.
29. time tradeoff.tw.
30. tto.tw.
31. exp models, economic/
32. economic model$.tw.
33. markov$.tw.
34. monte carlo.tw.
35. (decision$adj2 (tree$or analy$or model$)).tw.
36. letter.pt.
37. editorial.pt.
38. comment.pt.
39. or/36–38
40. or/4–35
41. (40 and 3) not 39
Appendix 2 Quality assessment
Quality assessment criteria for experimental studies
These quality assessment criteria were based on those proposed by the NHS Centre for Reviews and Dissemination. 35
Yes/No/Unclear/Not applicable | |
Was the method used to assign participants to the treatment groups really random? | |
What method of assignment was used? | |
Was the allocation of treatment concealed? | |
What method was used to conceal treatment allocation? | |
Was the number of participants who were randomised stated? | |
Were the eligibility criteria for study entry specified? | |
Were details of baseline comparability presented? | |
Was baseline comparability achieved? | |
Were the participants who received the intervention blinded to the treatment allocation? | |
Were the individuals who administered the intervention blinded to the treatment allocation? | |
Were the outcome assessors blinded to the treatment allocations? | |
Was the success of the blinding procedure assessed? | |
Were any co-interventions identified that may influence the outcomes for each group? | |
Was an intention-to-treat analysis included? | |
Were at least 80% of the participants originally included in the randomised process followed up in the final analysis? |
Appendix 3 Study quality characteristics for amantadine prophylaxis trials
Quality criterion | Reuman et al., 198957 (1)a | Reuman et al., 198957 (2)a | Aoki et al., 198658 | Pettersson et al., 198055 | Payler and Purdham, 198459 | Smorodintsev et al., 197060,61 | Sears and Clements, 198763 | Smorodintsev et al., 197062 |
---|---|---|---|---|---|---|---|---|
Was the method used to assign participants to the treatment groups really random? | Y | U | Y | Y | U | U | U | U |
Was the allocation of treatment concealed? | U | U | U | U | U | Y | U | U |
Was the number of participants who were randomised stated? | Y | Y | Y | Y | Y | Y | Y | U |
Were details of baseline comparability presented? | N | N | N | Y | N | N | N | N |
Was baseline comparability achieved? | U | U | U | N | U | U | U | U |
Were the eligibility criteria for study entry specified? | Y | Y | Y | N | Y | N | Y | N |
Were the outcome assessors blinded to allocation? |
U For self-notification of illness: Y |
U |
For measurement of amantadine concentrations in plasma and urine: Y For incidence of illness and adverse events: self-recorded: Y For serum HAI assessment: U For nurses classifying illness: U |
U For self-recorded symptoms and adverse effects: Y |
U | For adverse effect and morbidity assessment: Y | U | U |
Were the individuals who administered the intervention blinded to allocation? |
U Described as double blind but no further details |
U Described as double blind but no further details |
U Described as single blind but no further details |
U Described as double blind but no further details |
N | Y |
U Described as double blind but no further details |
U Described as double blind but no further details |
Were the participants receiving intervention blinded to allocation? | Y |
U Described as double blind but no further details |
Y | Y | N | Y |
U Described as double blind but no further details |
U Described as double blind but no further details |
Were the reasons for withdrawal stated? | Y | Y | Y | Y | Y | N | Y | NA |
Was intention-to-treat analysis included? | Y | Y | N |
Mixed For efficacy analysis: N For adverse event analysis: Y |
N | N | Y | Y |
Appendix 4 Study quality characteristics for oseltamivir prophylaxis trials
Peters et al., 200164 | Hayden et al., 199966 | Welliver et al., 200149 | Hayden et al., 200448,73,74 | Hayden et al., 200067 | |
---|---|---|---|---|---|
Was the method used to assign participants to the treatment groups really random? | Y | Y | U | U | U |
Was the allocation of treatment concealed? | Y | Y | U | U | U |
Was the number of participants who were randomised stated? | Y | Y | Y | Y | Y |
Were details of baseline comparability presented? | Y | Y | Y | Y | N |
Was baseline comparability achieved? | Y | Y | Y | Y | U |
Were the eligibility criteria for study entry specified? | Y | Y | Y | Y | U |
Were the outcome assessors blinded to allocation? |
U For self-recording of data: Y |
U For self-recording of data: Y |
U For self-recording of data: Y |
U For self-recording of data: Y |
U |
Were the individuals who administered the intervention blinded to allocation? |
U Described as double blind but no further details |
Y Double-blind labelling |
U Described as double blind but no further details |
N Open-label |
U Described as double blind but no further details |
Were the participants receiving intervention blinded to allocation? | Y |
Y Double-blind labelling |
U Described as double blind but no further details |
N Open-label |
U Described as double blind but no further details |
Were the reasons for withdrawal stated? | N | Y | Y | Y | N |
Was intention-to-treat analysis included? | U | N | N | Y | Y |
Appendix 5 Study quality characteristics for zanamivir prophylaxis trials
Quality criterion | Monto et al., 199972,73 | LaForce et al., 200775 | Monto et al., 200247 | Hayden et al., 200046 | Kaiser et al., 200072 | Ambrozaitis et al., 200576,77 | Gravenstein et al., 200578 | GSK study 167/10144 |
---|---|---|---|---|---|---|---|---|
Was the method used to assign participants to the treatment groups really random? | Y | Y | U | U | U | Y | Y | U |
Was the allocation of treatment concealed? | Y | Y44 | N44 | U | U | U | Y | U |
Was the number of participants who were randomised stated? | Y | Y | Y | Y | Y | Y | Y | Y |
Were details of baseline comparability presented? | Y | Y | Y | Y | Y | Y | Y | N |
Was baseline comparability achieved? | Y | Y | Y | Y | Y | Y | Y(albeit relatively weakly for age, sex, vaccination status, chronic cardiac condition and diabetes variables) | U |
Were the eligibility criteria for study entry specified? | Y | Y | Y | Y | Y | Y | Y | N |
Were the outcome assessors blinded to allocation? | U; for self-recording of data: Y | U; for self-recording of data: Y | U; for self-recording of data: Y | U; for self-recording of data: Y | U | Y | Y44 | U |
Were the individuals who administered the intervention blinded to allocation? | Y | Y44 | Y44 | U; described as double blind but no further details | U; described as double blind but no further details | Y | Y44 | U; described as double blind but no further details |
Were the participants receiving intervention blinded to allocation? | Y | Y | Y44 | U; described as double blind but no further details | U; described as double blind but no further details | Y | Y44 | U; described as double blind but no further details |
Were the reasons for withdrawal stated? | Y | Y | Y | Y | Y | Y | Y | N |
Was intention-to-treat analysis included? | Y | Y | Y | Y | Y | Y | Y | N |
Appendix 6 Studies excluded after close scrutiny with rationale
Study | Reason for exclusion |
---|---|
Aoki et al., 1985 | Not in line with licensed indications |
Bowles et al., 1999 | Not a randomised controlled trial |
Bowles et al., 2002 | Not a randomised controlled trial |
Bryson et al., 1980 | Not in line with licensed indications |
Bush et al., 2004 | Not a randomised controlled trial |
Calfee et al., 1999a | Not in line with licensed indications |
Calfee et al., 1999b | Not in line with licensed indications |
Callmander et al., 1968 | Not in line with licensed indications |
Cass et al., 2000 | Not in line with licensed indications |
Cohen et al., 1976 | Not in line with licensed indications |
Davies et al., 1988 | Not a randomised controlled trial |
Dawkins et al., 1968 | Analogue of amantadine hydrochloride. Not in line with licensed indications |
Degelau et al., 1990 | Not a randomised controlled trial |
Diaz-Pedroche et al., 2006 | Not available to read in English |
Dolin et al., 1982 | Not in line with licensed indications |
Drinka et al., 1998 | Comparison of short and long-term amantadine prophylaxis protocols |
Finklea et al., 1967 | Not in line with licensed indications – dosage not established in children |
Galbraith et al., 1969a | Data for subgroup in line with licensed indications not presented |
Galbraith et al., 1969b | Data for subgroup in line with licensed indications not presented |
Galbraith et al., 1971 | Data for subgroup in line with licensed indications not presented |
Hayden et al., 1981 | Not in line with licensed indications |
Hayden et al., 1996 | Not in line with licensed indications |
Hayden et al., 1999b | Not in line with licensed indications |
Hayden, 2001 | Abstract only. Insufficient data |
Hess, 1982 | Not available to read in English |
Hirji et al., 2001 | Not a randomised controlled trial |
Hirji et al., 2002 | Not a randomised controlled trial |
Jackson et al., 1963 | Not in line with licensed indications |
Kantor et al., 1980 | Not in line with licensed indications |
Kashiwagi et al., 2000 | Not available to read in English |
Lee et al., 2000 | Not a randomised controlled trial |
Leeming et al., 1969 | Not in line with licensed indications |
Leung et al., 1979 | Not in line with licensed indications |
Libow et al., 1996 | Not a randomised controlled trial |
Mate et al., 1970 | Not in line with licensed indications |
McLeod & Lau, 2001 | Not a randomised controlled trial |
Millet et al., 1982 | Not in line with licensed indications |
Monto et al., 1979 | Not in line with licensed indications |
Monto et al., 2004 | Not a randomised controlled trial |
Muldoon et al., 1976 | Not in line with licensed indications |
Nafta et al., 1970 | Not in line with licensed indications |
O’Donoghue et al., 1973 | Not in line with licensed indications |
Oker-Blom et al., 1970 | Not in line with licensed indications |
Peckinpaugh et al., 1970 | Not in line with licensed indications |
Peters et al., 1989 | Not a randomised controlled trial |
Plesnik et al., 1977 | Not available to read in English |
Quarles et al., 1981 | Not in line with licensed indications |
Quilligan et al., 1966a | Not in line with licensed indications – dosage not established in children |
Quilligan et al., 1966 | Not available to read in English |
Schapira et al., 1971 | Not in line with licensed indications |
Schilling et al., 1998 | Not in line with licensed indications |
Shinjoh et al., 2004 | Not available to read in English |
Smorodintsev et al., 1972 | Not available to read in English |
Somani et al., 1991 | Not a randomised controlled trial |
Stanley et al., 1965 | Not in line with licensed indications |
Togo et al., 1968 | Not in line with licensed indications |
Tyrrell et al., 1965 | Not in line with licensed indications |
Vogel, 2002 | Not a randomised controlled trial |
Walker et al., 1997 | Not in line with licensed indications |
Wendel et al., 1966 | Not in line with licensed indications |
Wright et al., 1974 | Not in line with licensed indications |
Wright et al., 1976 | Not in line with licensed indications – dosage not established in children |
Appendix 7 List of all model parameters
The following abbreviations are used in this appendix:
A&E, accident and emergency; CNS, central nervous system; GP, general practitioner; ICU, intensive care unit; ILI, influenza-like illness; ITU, intensive therapy unit; LOS, length of stay; NA, not applicable; QALY, quality-adjusted life-year; RR, relative risk; SE, standard error.
Distribution parameter key
Distribution type | Parameter 1 | Parameter 2 |
---|---|---|
Normal | Mean | SE |
Beta | Alpha | Alpha + beta |
Gamma | Alpha | Beta |
Lognormal | Ln mean | SE ln mean |
Dirichlet (multinomial) | Alpha | Beta |
List of model parameters – seasonal prophylaxis
No. | Parameter description | Distribution | Mean | Parameter 1 | Parameter 2 |
---|---|---|---|---|---|
Baseline event probabilities (disease) | |||||
1 | Baseline attack rate for influenza | Beta | 0.17 | 256 | 1469 |
2 | Probability ILI is influenza within epidemic period | Beta | 0.50 | 622 | 1256 |
3 | Probability influenza A strain is dominant | Beta | 0.75 | 9 | 12 |
4 | Probability influenza is influenza A in influenza A dominant seasons | Beta | 0.86 | 740 | 859 |
5 | Probability influenza is influenza A in influenza B dominant years | Beta | 0.30 | 83 | 281 |
6 | Probability influenza is influenza A | NA | 0.72 | 0.72 | – |
7 | Duration of influenza epidemic (days) | Gamma | 40 | 32.65 | 1.23 |
Effectiveness parameters (prevention) | |||||
8 | RR for influenza – vaccine | Lognormal | 0.36 | –1.02 | 0.14 |
9 | RR for influenza – amantadine prophylaxis | Lognormal | 0.40 | –0.92 | 0.83 |
10 | RR for influenza – oseltamivir prophylaxis | Lognormal | 0.24 | –1.43 | 0.45 |
11 | RR for influenza – zanamivir prophylaxis | Lognormal | 0.32 | –1.13 | 0.34 |
12 | Probability of amantadine resistance | Beta | 0.37 | 73.40 | 199 |
13 | Probability influenza case occurs within epidemic | Beta | 1 | 1 | 1125 |
14 | Probability influenza case avoidable – amantadine | NA | 1 | 1 | – |
15 | Probability influenza case avoidable – amantadine (vaccination) | NA | 0.53 | 0.53 | – |
16 | Probability influenza case avoidable – oseltamivir | NA | 1 | 1 | – |
17 | Percentage of influenza cases avoidable – zanamivir | NA | 0.70 | 0.70 | – |
Adverse events/withdrawals (prophylaxis) | |||||
18 | Probability adverse event – vaccination | Beta | 0.02 | 2 | 100 |
19 | Probability adverse event – amantadine prophylaxis | Beta | 0.05 | 10 | 200 |
20 | Probability withdrawal – amantadine prophylaxis | Beta | 0.06 | 13.56 | 237.89 |
21 | Probability withdrawal – oseltamivir prophylaxis | Beta | 0.02 | 1.72 | 86.11 |
22 | Probability withdrawal – zanamivir prophylaxis | Beta | 0.01 | 10.41 | 800.78 |
ILI event probabilities (treatment) | |||||
23 | Probability patient with ILI presents | Beta | 0.25 | 5 | 20 |
24 | Probability patient presents within 48 hours of ILI onset | Beta | 0.52 | 38 | 73 |
25 | Probability patient given antiviral Treatment | presents < 48 hours | NA | 0 | 0 | – |
26 | Probability patient receives oseltamivir | prescribed antiviral | NA | 1 | 1 | – |
27 | Probability patient receives zanamivir | prescribed antiviral | NA | 0 | 0 | – |
28 | Probability adverse events – oseltamivir treatment | Beta | 0.02 | 1.72 | 86.11 |
29 | Probability adverse events – zanamivir treatment | Beta | 0.01 | 10.41 | 800.78 |
30 | Probability complication – no treatment | Beta | 0.14 | 2417 | 17,201 |
31 | Odds ratio complication – oseltamivir treatment | Lognormal | 0.65 | 0 | 0 |
32 | Odds ratio complication – zanamivir treatment | Lognormal | 0.70 | –0.36 | 0.16 |
33 | Probability complication is respiratory | Dirichlet | 0.70 | 1698 | 2423 |
34 | Probability complication is cardiac | Dirichlet | 0 | 1 | 2423 |
35 | Probability complication is CNS | Dirichlet | 0.01 | 18 | 2423 |
36 | Probability complication is renal | Dirichlet | 0 | 3 | 2423 |
37 | Probability complication is otitis media | Dirichlet | 0.28 | 685 | 2423 |
38 | Probability complication is other | Dirichlet | 0.01 | 18 | 2423 |
39 | Probability respiratory complication is pneumonia | Beta | 0.02 | 29 | 1697 |
40 | Probability patient receives antibiotics | no complication | Beta | 0.28 | 4997 | 17,910 |
41 | Probability patient receives antibiotics | complication | Beta | 0.74 | 2183 | 2962 |
42 | Probability of influenza death | complication | Beta | 0 | 1 | 2311 |
Cost/resource parameters | |||||
43 | Cost of amantadine prophylaxis course (without vaccine) | NA | £14.40 | £14.40 | – |
44 | Cost of amantadine prophylaxis course (with vaccine) | NA | £9.60 | £9.60 | – |
45 | Cost of oseltamivir prophylaxis course | NA | £49.08 | £49.08 | – |
46 | Cost of zanamivir prophylaxis course | NA | £73.65 | £73.65 | – |
47 | Cost of oseltamivir treatment course | NA | £16.36 | £16.36 | – |
48 | Cost of zanamivir treatment course | NA | £24.55 | £24.55 | – |
49 | Days per course – amantadine prophylaxis | NA | £42 | £42 | – |
50 | Days per course – amantadine prophylaxis (prior vaccination) | NA | 21 | 21 | – |
51 | Days per course – oseltamivir prophylaxis | NA | 42 | 42 | – |
52 | Days per course – zanamivir prophylaxis | NA | 28 | 28 | – |
53 | Acquisition cost for vaccination | NA | £5.63 | £5.63 | – |
54 | Administration cost for vaccination | NA | £25 | £25 | – |
55 | Cost of attendance at GP surgery consultation | NA | £25 | £25 | – |
56 | Cost of attendance at GP home visit | NA | £69 | £69 | – |
57 | Cost of attendance at A&E | NA | £95.56 | £95.56 | – |
58 | Probability A&E attendance | patient presents (no complication) | Beta | 0.03 | 8.35 | 270.11 |
59 | Probability GP attendance | patient presents (no complication) | NA | 0.97 | 0.97 | – |
60 | Probability home GP visit | GP presentation (no complication) | Beta | 0.05 | 4 | 73 |
61 | Probability A&E attendance | patient presents (complication) | Beta | 0.03 | 8.35 | 270.11 |
62 | Probability GP attendance | patient presents (complication) | NA | 0.97 | 0.97 | – |
63 | Probability home GP visit | GP presentation (complication) | Beta | 0.05 | 4 | 73 |
64 | Cost of uncomplicated influenza presentation | NA | £29.52 | £29.52 | – |
65 | Cost of complicated influenza presentation | NA | £29.52 | £29.52 | – |
66 | Cost of antibiotics course | NA | £6.80 | £6.80 | – |
67 | Cost of anti-emetics course (metaclopramide 7-day course) | NA | £1.69 | £1.69 | – |
68 | Cost of managing adverse events – vaccination | NA | £25 | £25 | – |
69 | Cost of managing adverse events – amantadine prophylaxis | NA | £25 | £25 | – |
70 | Cost of inpatient episode | Gamma | £261.17 | £261.17 | 5.16 |
71 | Probability hospitalisation no treatment | complication | Beta | 0.11 | 5 | 46 |
72 | Probability ICU care | complication | Beta | 0.05 | 22 | 453 |
73 | Inpatient LOS (days) | Gamma | 2.30 | 1 | 4 |
74 | Cost of ITU day | Normal | £1345.39 | £1345.39 | £31.95 |
75 | ITU LOS (days) | Gamma | 28 | 11.60 | 2.41 |
76 | Expected cost of hospitalisation | NA | £2430.18 | £2430.18 | – |
HRQoL parameters | |||||
77 | 21-day QALYs for influenza case – no treatment | Beta | 0.04 | 4146 | 100,000 |
78 | 21-day QALYs for influenza case – oseltamivir treatment | Beta | 0.04 | 4247 | 100,000 |
79 | 21-day QALYs for influenza case – zanamivir treatment | Beta | 0.04 | 4247 | 100,000 |
80 | QALY loss for influenza case – no treatment | NA | 0.01 | 0.01 | – |
81 | QALY loss for influenza case – oseltamivir treatment | NA | 0.01 | 0.01 | – |
82 | QALY loss for influenza case – zanamivir treatment | NA | 0.01 | 0.01 | – |
83 | Utility decrement – adverse events | Beta | 0.20 | 200 | 1000 |
84 | Duration adverse events | Gamma | 0.01 | 25 | 0 |
85 | Utility decrement respiratory complication | Lognormal | 0.15 | –1.90 | 0.41 |
86 | Utility decrement cardiac complication | Lognormal | 0.37 | –0.99 | 0.14 |
87 | Utility decrement CNS complication | Lognormal | 0.37 | –0.99 | 0.14 |
88 | Utility decrement renal complication | Lognormal | 0.37 | –0.99 | 0.14 |
89 | Utility decrement otitis media complication | Lognormal | 0.15 | –1.90 | 0.41 |
90 | Utility decrement other complication | Lognormal | 0.37 | –0.99 | 0.14 |
91 | Duration respiratory complication (years) | Gamma | 0.02 | 6.92 | 1.14 |
92 | Duration cardiac complication (years) | Gamma | 0.02 | 6.92 | 1.14 |
93 | Duration CNS complication (years) | Gamma | 0.02 | 6.92 | 1.14 |
94 | Duration renal complication (years) | Gamma | 0.02 | 6.92 | 1.14 |
95 | Duration otitis media complication (years) | Gamma | 0.03 | 9.73 | 0.96 |
96 | Duration other complication (years) | Gamma | 0.02 | 6.92 | 1.14 |
97 | Utility general population 0–24 | Normal | 0.94 | 0.94 | 0.01 |
98 | Utility general population 25–34 | Normal | 0.93 | 0.93 | 0.01 |
99 | Utility general population 35–44 | Normal | 0.91 | 0.91 | 0.01 |
100 | Utility general population 45–54 | Normal | 0.85 | 0.85 | 0.01 |
101 | Utility general population 55–64 | Normal | 0.80 | 0.80 | 0.01 |
102 | Utility general population 65–74 | Normal | 0.78 | 0.78 | 0.01 |
103 | Utility general population > 75 | Normal | 0.73 | 0.73 | 0.02 |
104 | Percentage population female | NA | 0.51 | 0.51 | – |
105 | QALY loss for premature death | NA | 24.74 | 24.74 | – |
106 | Discount rate for QALYs | NA | 3.50% | 3.50% | – |
No. | Parameter description | Distribution | Mean | Parameter 1 | Parameter 2 |
---|---|---|---|---|---|
Baseline event probabilities (disease) | |||||
1 | Baseline attack rate for influenza | Beta | 0.17 | 256 | 1469 |
2 | Probability ILI is influenza within epidemic period | Beta | 0.50 | 622 | 1256 |
3 | Probability influenza A strain is dominant | Beta | 0.75 | 9 | 12 |
4 | Probability influenza is influenza A in influenza A dominant seasons | Beta | 0.86 | 740 | 859 |
5 | Probability influenza is influenza A in influenza B dominant years | Beta | 0.30 | 83 | 281 |
6 | Probability influenza is influenza A | NA | 0.72 | 0.72 | – |
7 | Duration of influenza epidemic (days) | Gamma | 40 | 32.65 | 1.23 |
Effectiveness parameters (prevention) | |||||
8 | RR for influenza – vaccine | Lognormal | 0.36 | –1.02 | 0.14 |
9 | RR for influenza – amantadine prophylaxis | Lognormal | 0.40 | –0.92 | 0.83 |
10 | RR for influenza – oseltamivir prophylaxis | Lognormal | 0.24 | –1.43 | 0.45 |
11 | RR for influenza – zanamivir prophylaxis | Lognormal | 0.32 | –1.13 | 0.34 |
12 | Probability of amantadine resistance | Beta | 0.37 | 73.40 | 199 |
13 | Probability influenza case occurs within epidemic | Beta | 1 | 1 | 1125 |
14 | Probability influenza case avoidable – amantadine | NA | 1 | 1 | – |
15 | Probability influenza case avoidable – amantadine (vaccination) | NA | 0.53 | 0.53 | – |
16 | Probability influenza case avoidable – oseltamivir | NA | 1 | 1 | – |
17 | Percentage of influenza cases avoidable – zanamivir | NA | 0.70 | 0.70 | – |
Adverse events/withdrawals (prophylaxis) | |||||
18 | Probability adverse event – vaccination | Beta | 0.02 | 2 | 100 |
19 | Probability adverse event – amantadine prophylaxis | Beta | 0.05 | 10 | 200 |
20 | Probability withdrawal – amantadine prophylaxis | Beta | 0.06 | 13.56 | 237.89 |
21 | Probability withdrawal – oseltamivir prophylaxis | Beta | 0.02 | 1.72 | 86.11 |
22 | Probability withdrawal – zanamivir prophylaxis | Beta | 0.01 | 10.41 | 800.78 |
ILI event probabilities (treatment) | |||||
23 | Probability patient with ILI presents | Beta | 0.25 | 5 | 20 |
24 | Probability patient presents within 48 hours of ILI onset | Beta | 0.52 | 38 | 73 |
25 | Probability patient given antiviral treatment | presents < 48 hours | NA | 1 | 1 | – |
26 | Probability patient receives oseltamivir | prescribed antiviral | NA | 1 | 1 | – |
27 | Probability patient receives zanamivir | prescribed antiviral | NA | 0 | 0 | – |
28 | Probability adverse events – oseltamivir treatment | Beta | 0.02 | 1.72 | 86.11 |
29 | Probability adverse events – zanamivir treatment | Beta | 0.01 | 10.41 | 800.78 |
30 | Probability complication – no treatment | Beta | 0.18 | 675 | 3695 |
31 | Odds ratio complication – oseltamivir treatment | Lognormal | 0.65 | 0 | 0 |
32 | Odds ratio complication – zanamivir treatment | Lognormal | 0.49 | –0.71 | 0.38 |
33 | Probability complication is respiratory | Dirichlet | 0.77 | 521 | 681 |
34 | Probability complication is cardiac | Dirichlet | 0 | 1 | 681 |
35 | Probability complication is CNS | Dirichlet | 0 | 1 | 681 |
36 | Probability complication is renal | Dirichlet | 0 | 1 | 681 |
37 | Probability complication is otitis media | Dirichlet | 0.23 | 154 | 681 |
38 | Probability complication is other | Dirichlet | 0 | 3 | 681 |
39 | Probability respiratory complication is pneumonia | Beta | 0.02 | 9 | 520 |
40 | Probability patient receives antibiotics | no complication | Beta | 0.28 | 4997 | 17,910 |
41 | Probability patient receives antibiotics | complication | Beta | 0.74 | 2183 | 2962 |
42 | Probability of influenza death | complication | Beta | 0 | 1 | 650 |
Cost/resource parameters | |||||
43 | Cost of amantadine prophylaxis course (without vaccine) | NA | £14.40 | £14.40 | – |
44 | Cost of amantadine prophylaxis course (with vaccine) | NA | £9.60 | £9.60 | – |
45 | Cost of oseltamivir prophylaxis course | NA | £49.08 | £49.08 | – |
46 | Cost of zanamivir prophylaxis course | NA | £73.65 | £73.65 | – |
47 | Cost of oseltamivir treatment course | NA | £16.36 | £16.36 | – |
48 | Cost of zanamivir treatment course | NA | £24.55 | £24.55 | – |
49 | Days per course – amantadine prophylaxis | NA | £42 | £42 | – |
50 | Days per course – amantadine prophylaxis (prior vaccination) | NA | 21 | 21 | – |
51 | Days per course – oseltamivir prophylaxis | NA | 42 | 42 | – |
52 | Days per course – zanamivir prophylaxis | NA | 28 | 28 | – |
53 | Acquisition cost for vaccination | NA | £5.63 | £5.63 | – |
54 | Administration cost for vaccination | NA | £25 | £25 | – |
55 | Cost of attendance at GP surgery consultation | NA | £25 | £25 | – |
56 | Cost of attendance at GP home visit | NA | £69 | £69 | – |
57 | Cost of attendance at A&E | NA | £95.56 | £95.56 | – |
58 | Probability A&E attendance | patient presents (no complication) | Beta | 0.03 | 8.35 | 270.11 |
59 | Probability GP attendance | patient presents (no complication) | NA | 0.97 | 0.97 | – |
60 | Probability home GP visit | GP presentation (no complication) | Beta | 0.05 | 4 | 73 |
61 | Probability A&E attendance | patient presents (complication) | Beta | 0.03 | 8.35 | 270.11 |
62 | Probability GP attendance | patient presents (complication) | NA | 0.97 | 0.97 | – |
63 | Probability home GP visit | GP presentation (complication) | Beta | 0.05 | 4 | 73 |
64 | Cost of uncomplicated influenza presentation | NA | £29.52 | £29.52 | – |
65 | Cost of complicated influenza presentation | NA | £29.52 | £29.52 | – |
66 | Cost of antibiotics course | NA | £6.80 | £6.80 | – |
67 | Cost of anti-emetics course (metaclopramide 7-day course) | NA | £1.69 | £1.69 | – |
68 | Cost of managing adverse events – vaccination | NA | £25 | £25 | – |
69 | Cost of managing adverse events – amantadine prophylaxis | NA | £25 | £25 | – |
70 | Cost of inpatient episode | Gamma | £261.17 | £261.17 | 5.16 |
71 | Probability hospitalisation no treatment | complication | Beta | 0.16 | 15 | 95 |
72 | Probability ICU care | complication | Beta | 0.05 | 22 | 453 |
73 | Inpatient LOS (days) | Gamma | 2.30 | 1 | 4 |
74 | Cost of ITU day | Normal | £1345.39 | £1345.39 | £31.95 |
75 | ITU LOS (days) | Gamma | 28 | 11.60 | 2.41 |
76 | Expected cost of hospitalisation | NA | £2430.18 | £2430.18 | – |
HRQoL parameters | |||||
77 | 21-day QALYs for influenza case – no treatment | Beta | 0.03 | 2820 | 100,000 |
78 | 21-day QALYs for influenza case – oseltamivir treatment | Beta | 0.03 | 2977 | 100,000 |
79 | 21-day QALYs for influenza case – zanamivir treatment | Beta | 0.03 | 2977 | 100,000 |
80 | QALY loss for influenza case – no treatment | NA | 0.02 | 0.02 | – |
81 | QALY loss for influenza case – oseltamivir treatment | NA | 0.02 | 0.02 | – |
82 | QALY loss for influenza case – zanamivir treatment | NA | 0.02 | 0.02 | – |
83 | Utility decrement – adverse events | Beta | 0.20 | 200 | 1000 |
84 | Duration adverse events | Gamma | 0.01 | 25 | 0 |
85 | Utility decrement respiratory complication | Lognormal | 0.15 | –1.90 | 0.41 |
86 | Utility decrement cardiac complication | Lognormal | 0.37 | –0.99 | 0.14 |
87 | Utility decrement CNS complication | Lognormal | 0.37 | –0.99 | 0.14 |
88 | Utility decrement renal complication | Lognormal | 0.37 | –0.99 | 0.14 |
89 | Utility decrement otitis media complication | Lognormal | 0.15 | –1.90 | 0.41 |
90 | Utility decrement other complication | Lognormal | 0.37 | –0.99 | 0.14 |
91 | Duration respiratory complication (years) | Gamma | 0.02 | 7.24 | 1.11 |
92 | Duration cardiac complication (years) | Gamma | 0.02 | 7.24 | 1.11 |
93 | Duration CNS complication (years) | Gamma | 0.02 | 7.24 | 1.11 |
94 | Duration renal complication (years) | Gamma | 0.02 | 7.24 | 1.11 |
95 | Duration otitis media complication (years) | Gamma | 0.03 | 9.73 | 0.96 |
96 | Duration other complication (years) | Gamma | 0.02 | 7.24 | 1.11 |
97 | Utility general population 0–24 | Normal | 0.94 | 0.94 | 0.01 |
98 | Utility general population 25–34 | Normal | 0.93 | 0.93 | 0.01 |
99 | Utility general population 35–44 | Normal | 0.91 | 0.91 | 0.01 |
100 | Utility general population 45–54 | Normal | 0.85 | 0.85 | 0.01 |
101 | Utility general population 55–64 | Normal | 0.80 | 0.80 | 0.01 |
102 | Utility general population 65–74 | Normal | 0.78 | 0.78 | 0.01 |
103 | Utility general population > 75 | Normal | 0.73 | 0.73 | 0.02 |
104 | Percentage population female | NA | 0.51 | 0.51 | – |
105 | QALY loss for premature death | NA | 24.74 | 24.74 | – |
106 | Discount rate for QALYs | NA | 3.50% | 3.50% | – |
No. | Parameter description | Distribution | Mean | Parameter 1 | Parameter 2 |
---|---|---|---|---|---|
Baseline event probabilities (disease) | |||||
1 | Baseline attack rate for influenza | Beta | 0.06 | 104 | 1670 |
2 | Probability ILI is influenza within epidemic period | Beta | 0.50 | 622 | 1256 |
3 | Probability influenza A strain is dominant | Beta | 0.75 | 9 | 12 |
4 | Probability influenza is influenza A in influenza A dominant seasons | Beta | 0.86 | 740 | 859 |
5 | Probability influenza is influenza A in influenza B dominant years | Beta | 0.30 | 83 | 281 |
6 | Probability influenza is influenza A | NA | 0.72 | 0.72 | – |
7 | Duration of influenza epidemic (days) | Gamma | 40 | 32.65 | 1.23 |
Effectiveness parameters (prevention) | |||||
8 | RR for influenza – vaccine | Lognormal | 0.35 | –1.05 | 0.17 |
9 | RR for influenza – amantadine prophylaxis | Lognormal | 0.40 | –0.92 | 0.83 |
10 | RR for influenza – oseltamivir prophylaxis | Lognormal | 0.24 | –1.43 | 0.45 |
11 | RR for influenza – zanamivir prophylaxis | Lognormal | 0.32 | –1.13 | 0.34 |
12 | Probability of amantadine resistance | Beta | 0.37 | 73.40 | 199 |
13 | Probability influenza case occurs within epidemic | Beta | 1 | 1 | 1125 |
14 | Probability influenza case avoidable – amantadine | NA | 1 | 1 | – |
15 | Probability influenza case avoidable – amantadine (vaccination) | NA | 0.53 | 0.53 | – |
16 | Probability influenza case avoidable – oseltamivir | NA | 1 | 1 | – |
17 | Percentage of influenza cases avoidable – zanamivir | NA | 0.70 | 0.70 | – |
Adverse events/withdrawals (prophylaxis) | |||||
18 | Probability adverse event – vaccination | Beta | 0.02 | 2 | 100 |
19 | Probability adverse event – amantadine prophylaxis | Beta | 0.05 | 10 | 200 |
20 | Probability withdrawal – amantadine prophylaxis | Beta | 0.06 | 13.56 | 237.89 |
21 | Probability withdrawal – oseltamivir prophylaxis | Beta | 0.02 | 1.72 | 86.11 |
22 | Probability withdrawal – zanamivir prophylaxis | Beta | 0.01 | 10.41 | 800.78 |
ILI event probabilities (treatment) | |||||
23 | Probability patient with ILI presents | Beta | 0.25 | 5 | 20 |
24 | Probability patient presents within 48 hours of ILI onset | Beta | 0.16 | 104 | 668 |
25 | Probability patient given antiviral treatment | presents < 48 hours | NA | 0 | 0 | – |
26 | Probability patient receives oseltamivir | prescribed antiviral | NA | 0.89 | 0.89 | – |
27 | Probability patient receives zanamivir | prescribed antiviral | NA | 0.11 | 0.11 | – |
28 | Probability adverse events – oseltamivir treatment | Beta | 0.02 | 1.72 | 86.11 |
29 | Probability adverse events – zanamivir treatment | Beta | 0.01 | 10.41 | 800.78 |
30 | Probability complication – no treatment | Beta | 0.08 | 6509 | 85,248 |
31 | Odds ratio complication – oseltamivir treatment | Lognormal | 0.40 | –1 | 0 |
32 | Odds ratio complication – zanamivir treatment | Lognormal | 0.70 | –0.36 | 0.16 |
33 | Probability complication is respiratory | Dirichlet | 0.87 | 5637 | 6515 |
34 | Probability complication is cardiac | Dirichlet | 0 | 12 | 6515 |
35 | Probability complication is CNS | Dirichlet | 0.02 | 102 | 6515 |
36 | Probability complication is renal | Dirichlet | 0 | 10 | 6515 |
37 | Probability complication is otitis media | Dirichlet | 0.08 | 501 | 6515 |
38 | Probability complication is other | Dirichlet | 0.04 | 253 | 6515 |
39 | Probability respiratory complication is pneumonia | Beta | 0.04 | 237 | 5636 |
40 | Probability patient receives antibiotics | no complication | Beta | 0.42 | 19811 | 47,169 |
41 | Probability patient receives antibiotics | complication | Beta | 0.81 | 6983 | 8579 |
42 | Probability of influenza death | complication | Beta | 0.01 | 33 | 6437 |
Cost/resource parameters | |||||
43 | Cost of amantadine prophylaxis course (without vaccine) | NA | £14.40 | £14.40 | – |
44 | Cost of amantadine prophylaxis course (with vaccine) | NA | £9.60 | £9.60 | – |
45 | Cost of oseltamivir prophylaxis course | NA | £81.80 | £81.80 | – |
46 | Cost of zanamivir prophylaxis course | NA | £73.65 | £73.65 | – |
47 | Cost of oseltamivir treatment course | NA | £16.36 | £16.36 | – |
48 | Cost of zanamivir treatment course | NA | £24.55 | £24.55 | – |
49 | Days per course – amantadine prophylaxis | NA | £42 | £42 | – |
50 | Days per course – amantadine prophylaxis (prior vaccination) | NA | 21 | 21 | – |
51 | Days per course – oseltamivir prophylaxis | NA | 42 | 42 | – |
52 | Days per course – zanamivir prophylaxis | NA | 28 | 28 | – |
53 | Acquisition cost for vaccination | NA | £5.63 | £5.63 | – |
54 | Administration cost for vaccination | NA | £25 | £25 | – |
55 | Cost of attendance at GP surgery consultation | NA | £25 | £25 | – |
56 | Cost of attendance at GP home visit | NA | £69 | £69 | – |
57 | Cost of attendance at A&E | NA | £95.56 | £95.56 | – |
58 | Probability A&E attendance | patient presents (no complication) | Beta | 0.03 | 8.35 | 270.11 |
59 | Probability GP attendance | patient presents (no complication) | NA | 0.97 | 0.97 | – |
60 | Probability home GP visit | GP presentation (no complication) | Beta | 0.08 | 56 | 674 |
61 | Probability A&E attendance | patient presents (complication) | Beta | 0.03 | 8.35 | 270.11 |
62 | Probability GP attendance | patient presents (complication) | NA | 0.97 | 0.97 | – |
63 | Probability home GP visit | GP presentation (complication) | Beta | 0.08 | 56 | 674 |
64 | Cost of uncomplicated influenza presentation | NA | £30.73 | £30.73 | – |
65 | Cost of complicated influenza presentation | NA | £30.73 | £30.73 | – |
66 | Cost of antibiotics course | NA | £6.80 | £6.80 | – |
67 | Cost of anti-emetics course (metaclopramide 7-day course) | NA | £1.69 | £1.69 | – |
68 | Cost of managing adverse events – vaccination | NA | £25 | £25 | – |
69 | Cost of managing adverse events – amantadine prophylaxis | NA | £25 | £25 | – |
70 | Cost of inpatient episode | Gamma | £261.17 | £261.17 | 5.16 |
71 | Probability hospitalisation no treatment | complication | Beta | 0.11 | 5 | 46 |
72 | Probability ICU care | complication | Beta | 0.05 | 22 | 453 |
73 | Inpatient LOS (days) | Gamma | 11.90 | 16 | 1 |
74 | Cost of ITU day | Normal | £1345.39 | £1345.39 | £31.95 |
75 | ITU LOS (days) | Gamma | 28 | 11.60 | 2.41 |
76 | Expected cost of hospitalisation | NA | £4937.39 | £4937.39 | – |
HRQoL parameters | |||||
77 | 21-day QALYs for influenza case – no treatment | Beta | 0.04 | 4146 | 100,000 |
78 | 21-day QALYs for influenza case – oseltamivir treatment | Beta | 0.04 | 4247 | 100,000 |
79 | 21-day QALYs for influenza case – zanamivir treatment | Beta | 0.04 | 4247 | 100,000 |
80 | QALY loss for influenza case – no treatment | NA | 0.01 | 0.01 | – |
81 | QALY loss for influenza case – oseltamivir treatment | NA | 0.01 | 0.01 | – |
82 | QALY loss for influenza case – zanamivir treatment | NA | 0.01 | 0.01 | – |
83 | Utility decrement – adverse events | Beta | 0.20 | 200 | 1000 |
84 | Duration adverse events | Gamma | 0.01 | 25 | 0 |
85 | Utility decrement respiratory complication | Lognormal | 0.15 | –1.90 | 0.41 |
86 | Utility decrement cardiac complication | Lognormal | 0.37 | –0.99 | 0.14 |
87 | Utility decrement CNS complication | Lognormal | 0.37 | –0.99 | 0.14 |
88 | Utility decrement renal complication | Lognormal | 0.37 | –0.99 | 0.14 |
89 | Utility decrement otitis media complication | Lognormal | 0.15 | –1.90 | 0.41 |
90 | Utility decrement other complication | Lognormal | 0.37 | –0.99 | 0.14 |
91 | Duration respiratory complication (years) | Gamma | 0.03 | 9.46 | 0.98 |
92 | Duration cardiac complication (years) | Gamma | 0.03 | 9.46 | 0.98 |
93 | Duration CNS complication (years) | Gamma | 0.03 | 9.46 | 0.98 |
94 | Duration renal complication (years) | Gamma | 0.03 | 9.46 | 0.98 |
95 | Duration otitis media complication (years) | Gamma | 0.03 | 9.73 | 0.96 |
96 | Duration other complication (years) | Gamma | 0.03 | 9.46 | 0.98 |
97 | Utility general population 0–24 | Normal | 0.94 | 0.94 | 0.01 |
98 | Utility general population 25–34 | Normal | 0.93 | 0.93 | 0.01 |
99 | Utility general population 35–44 | Normal | 0.91 | 0.91 | 0.01 |
100 | Utility general population 45–54 | Normal | 0.85 | 0.85 | 0.01 |
101 | Utility general population 55–64 | Normal | 0.80 | 0.80 | 0.01 |
102 | Utility general population 65–74 | Normal | 0.78 | 0.78 | 0.01 |
103 | Utility general population > 75 | Normal | 0.73 | 0.73 | 0.02 |
104 | Percentage population female | NA | 0.51 | 0.51 | – |
105 | QALY loss for premature death | NA | 13.37 | 13.37 | – |
106 | Discount rate for QALYs | NA | 3.50% | 3.50% | – |
No. | Parameter description | Distribution | Mean | Parameter 1 | Parameter 2 |
---|---|---|---|---|---|
Baseline event probabilities (disease) | |||||
1 | Baseline attack rate for influenza | Beta | 0.06 | 104 | 1670 |
2 | Probability ILI is influenza within epidemic period | Beta | 0.50 | 622 | 1256 |
3 | Probability influenza A strain is dominant | Beta | 0.75 | 9 | 12 |
4 | Probability influenza is influenza A in influenza A dominant seasons | Beta | 0.86 | 740 | 859 |
5 | Probability influenza is influenza A in influenza B dominant years | Beta | 0.30 | 83 | 281 |
6 | Probability influenza is influenza A | NA | 0.72 | 0.72 | – |
7 | Duration of influenza epidemic (days) | Gamma | 40 | 32.65 | 1.23 |
Effectiveness parameters (prevention) | |||||
8 | RR for influenza – vaccine | Lognormal | 0.35 | –1.05 | 0.17 |
9 | RR for influenza – amantadine prophylaxis | Lognormal | 0.40 | –0.92 | 0.83 |
10 | RR for influenza – oseltamivir prophylaxis | Lognormal | 0.24 | –1.43 | 0.45 |
11 | RR for influenza – zanamivir prophylaxis | Lognormal | 0.17 | –1.75 | 0.54 |
12 | Probability of amantadine resistance | Beta | 0.37 | 73.40 | 199 |
13 | Probability influenza case occurs within epidemic | Beta | 1 | 1 | 1125 |
14 | Probability influenza case avoidable – amantadine | NA | 1 | 1 | – |
15 | Probability influenza case avoidable – amantadine (vaccination) | NA | 0.53 | 0.53 | – |
16 | Probability influenza case avoidable – oseltamivir | NA | 1 | 1 | – |
17 | Percentage of influenza cases avoidable – zanamivir | NA | 0.70 | 0.70 | – |
Adverse events/withdrawals (prophylaxis) | |||||
18 | Probability adverse event – vaccination | Beta | 0.02 | 2 | 100 |
19 | Probability adverse event – amantadine prophylaxis | Beta | 0.05 | 10 | 200 |
20 | Probability withdrawal – amantadine prophylaxis | Beta | 0.15 | 0.37 | 2.55 |
21 | Probability withdrawal – oseltamivir prophylaxis | Beta | 0.02 | 1.72 | 86.11 |
22 | Probability withdrawal – zanamivir prophylaxis | Beta | 0.01 | 10.41 | 800.78 |
ILI event probabilities (treatment) | |||||
23 | Probability patient with ILI presents | Beta | 0.25 | 5 | 20 |
24 | Probability patient presents within 48 hours of ILI onset | Beta | 0.16 | 104 | 668 |
25 | Probability patient given antiviral treatment | presents < 48 hours | NA | 1 | 1 | – |
26 | Probability patient receives oseltamivir | prescribed antiviral | NA | 0.89 | 0.89 | – |
27 | Probability patient receives zanamivir | prescribed antiviral | NA | 0.11 | 0.11 | – |
28 | Probability adverse events – oseltamivir treatment | Beta | 0.02 | 1.72 | 86.11 |
29 | Probability adverse events – zanamivir treatment | Beta | 0.01 | 10.41 | 800.78 |
30 | Probability complication – no treatment | Beta | 0.12 | 2166 | 17,597 |
31 | Odds ratio complication – oseltamivir treatment | Lognormal | 0.40 | –1 | 0 |
32 | Odds ratio complication – zanamivir treatment | Lognormal | 0.49 | –0.71 | 0.38 |
33 | Probability complication is respiratory | Dirichlet | 0.89 | 1942 | 2172 |
34 | Probability complication is cardiac | Dirichlet | 0.01 | 30 | 2172 |
35 | Probability complication is CNS | Dirichlet | 0.01 | 16 | 2172 |
36 | Probability complication is renal | Dirichlet | 0 | 6 | 2172 |
37 | Probability complication is otitis media | Dirichlet | 0.05 | 111 | 2172 |
38 | Probability complication is other | Dirichlet | 0.03 | 67 | 2172 |
39 | Probability respiratory complication is pneumonia | Beta | 0.03 | 62 | 1941 |
40 | Probability patient receives antibiotics | no complication | Beta | 0.42 | 19811 | 47,169 |
41 | Probability patient receives antibiotics | complication | Beta | 0.81 | 6983 | 8579 |
42 | Probability of influenza death | complication | Beta | 0.01 | 16 | 2142 |
Cost/resource parameters | |||||
43 | Cost of amantadine prophylaxis course (without vaccine) | NA | £14.40 | £14.40 | – |
44 | Cost of amantadine prophylaxis course (with vaccine) | NA | £9.60 | £9.60 | – |
45 | Cost of oseltamivir prophylaxis course | NA | £81.80 | £81.80 | – |
46 | Cost of zanamivir prophylaxis course | NA | £73.65 | £73.65 | – |
47 | Cost of oseltamivir treatment course | NA | £16.36 | £16.36 | – |
48 | Cost of zanamivir treatment course | NA | £24.55 | £24.55 | – |
49 | Days per course – amantadine prophylaxis | NA | £42 | £42 | – |
50 | Days per course – amantadine prophylaxis (prior vaccination) | NA | 21 | 21 | – |
51 | Days per course – oseltamivir prophylaxis | NA | 42 | 42 | – |
52 | Days per course – zanamivir prophylaxis | NA | 28 | 28 | – |
53 | Acquisition cost for vaccination | NA | £5.63 | £5.63 | – |
54 | Administration cost for vaccination | NA | £25 | £25 | – |
55 | Cost of attendance at GP surgery consultation | NA | £25 | £25 | – |
56 | Cost of attendance at GP home visit | NA | £69 | £69 | – |
57 | Cost of attendance at A&E | NA | £95.56 | £95.56 | – |
58 | Probability A&E attendance | patient presents (no complication) | Beta | 0.03 | 8.35 | 270.11 |
59 | Probability GP attendance | patient presents (no complication) | NA | 0.97 | 0.97 | – |
60 | Probability home GP visit | GP presentation (no complication) | Beta | 0.08 | 56 | 674 |
61 | Probability A&E attendance | patient presents (complication) | Beta | 0.03 | 8.35 | 270.11 |
62 | Probability GP attendance | patient presents (complication) | NA | 0.97 | 0.97 | – |
63 | Probability home GP visit | GP presentation (complication) | Beta | 0.08 | 56 | 674 |
64 | Cost of uncomplicated influenza presentation | NA | £30.73 | £30.73 | – |
65 | Cost of complicated influenza presentation | NA | £30.73 | £30.73 | – |
66 | Cost of antibiotics course | NA | £6.80 | £6.80 | – |
67 | Cost of anti-emetics course (metaclopramide 7-day course) | NA | £1.69 | £1.69 | – |
68 | Cost of managing adverse events – vaccination | NA | £25 | £25 | – |
69 | Cost of managing adverse events – amantadine prophylaxis | NA | £25 | £25 | – |
70 | Cost of inpatient episode | Gamma | £261.17 | £261.17 | 5.16 |
71 | Probability hospitalisation no treatment | complication | Beta | 0.16 | 15 | 95 |
72 | Probability ICU care | complication | Beta | 0.05 | 22 | 453 |
73 | Inpatient LOS (days) | Gamma | 11.90 | 16 | 1 |
74 | Cost of ITU day | Normal | £1345.39 | £1345.39 | £31.95 |
75 | ITU LOS (days) | Gamma | 28 | 11.60 | 2.41 |
76 | Expected cost of hospitalisation | NA | £4937.39 | £4937.39 | – |
HRQoL parameters | |||||
77 | 21-day QALYs for influenza case – no treatment | Beta | 0.03 | 2820 | 100,000 |
78 | 21-day QALYs for influenza case – oseltamivir treatment | Beta | 0.03 | 2977 | 100,000 |
79 | 21-day QALYs for influenza case – zanamivir treatment | Beta | 0.03 | 2977 | 100,000 |
80 | QALY loss for influenza case – no treatment | NA | 0.02 | 0.02 | – |
81 | QALY loss for influenza case – oseltamivir treatment | NA | 0.02 | 0.02 | – |
82 | QALY loss for influenza case – zanamivir treatment | NA | 0.02 | 0.02 | – |
83 | Utility decrement – adverse events | Beta | 0.20 | 200 | 1000 |
84 | Duration adverse events | Gamma | 0.01 | 25 | 0 |
85 | Utility decrement respiratory complication | Lognormal | 0.15 | –1.90 | 0.41 |
86 | Utility decrement cardiac complication | Lognormal | 0.37 | –0.99 | 0.14 |
87 | Utility decrement CNS complication | Lognormal | 0.37 | –0.99 | 0.14 |
88 | Utility decrement renal complication | Lognormal | 0.37 | –0.99 | 0.14 |
89 | Utility decrement otitis media complication | Lognormal | 0.15 | –1.90 | 0.41 |
90 | Utility decrement other complication | Lognormal | 0.37 | –0.99 | 0.14 |
91 | Duration respiratory complication (years) | Gamma | 0.03 | 12.60 | 0.85 |
92 | Duration cardiac complication (years) | Gamma | 0.03 | 12.60 | 0.85 |
93 | Duration CNS complication (years) | Gamma | 0.03 | 12.60 | 0.85 |
94 | Duration renal complication (years) | Gamma | 0.03 | 12.60 | 0.85 |
95 | Duration otitis media complication (years) | Gamma | 0.03 | 9.73 | 0.96 |
96 | Duration other complication (years) | Gamma | 0.03 | 12.60 | 0.85 |
97 | Utility general population 0–24 | Normal | 0.94 | 0.94 | 0.01 |
98 | Utility general population 25–34 | Normal | 0.93 | 0.93 | 0.01 |
99 | Utility general population 35–44 | Normal | 0.91 | 0.91 | 0.01 |
100 | Utility general population 45–54 | Normal | 0.85 | 0.85 | 0.01 |
101 | Utility general population 55–64 | Normal | 0.80 | 0.80 | 0.01 |
102 | Utility general population 65–74 | Normal | 0.78 | 0.78 | 0.01 |
103 | Utility general population > 75 | Normal | 0.73 | 0.73 | 0.02 |
104 | Percentage population female | NA | 0.51 | 0.51 | – |
105 | QALY loss for premature death | NA | 13.37 | 13.37 | – |
106 | Discount rate for QALYs | NA | 3.50% | 3.50% | – |
No. | Parameter description | Distribution | Mean | Parameter 1 | Parameter 2 |
---|---|---|---|---|---|
Baseline event probabilities (disease) | |||||
1 | Baseline attack rate for influenza | Beta | 0.05 | 57 | 1098 |
2 | Probability ILI is influenza within epidemic period | Beta | 0.50 | 622 | 1256 |
3 | Probability influenza A strain is dominant | Beta | 0.75 | 9 | 12 |
4 | Probability influenza is influenza A in influenza A dominant seasons | Beta | 0.86 | 740 | 859 |
5 | Probability influenza is influenza A in influenza B dominant years | Beta | 0.30 | 83 | 281 |
6 | Probability influenza is influenza A | NA | 0.72 | 0.72 | – |
7 | Duration of influenza epidemic (days) | Gamma | 40 | 32.65 | 1.23 |
Effectiveness parameters (prevention) | |||||
8 | RR for influenza – vaccine | Lognormal | 0.42 | –0.87 | 0.23 |
9 | RR for influenza – amantadine prophylaxis | Lognormal | 0.40 | –0.92 | 0.83 |
10 | RR for influenza – oseltamivir prophylaxis | Lognormal | 0.08 | –2.50 | 1.04 |
11 | RR for influenza – zanamivir prophylaxis | Lognormal | 0.20 | –1.61 | 1.09 |
12 | Probability of amantadine resistance | Beta | 0.37 | 73.40 | 199 |
13 | Probability influenza case occurs within epidemic | Beta | 1 | 1 | 1125 |
14 | Probability influenza case avoidable – amantadine | NA | 1 | 1 | – |
15 | Probability influenza case avoidable – amantadine (vaccination) | NA | 0.53 | 0.53 | – |
16 | Probability influenza case avoidable – oseltamivir | NA | 1 | 1 | – |
17 | Percentage of influenza cases avoidable – zanamivir | NA | 0.70 | 0.70 | – |
Adverse events/withdrawals (prophylaxis) | |||||
18 | Probability adverse event – vaccination | Beta | 0.02 | 2 | 100 |
19 | Probability adverse event – amantadine prophylaxis | Beta | 0.05 | 10 | 200 |
20 | Probability withdrawal – amantadine prophylaxis | Beta | 0.15 | 0.37 | 2.55 |
21 | Probability withdrawal – oseltamivir prophylaxis | Beta | 0.02 | 1.72 | 86.11 |
22 | Probability withdrawal – zanamivir prophylaxis | Beta | 0.01 | 10.41 | 800.78 |
ILI event probabilities (treatment) | |||||
23 | Probability patient with ILI presents | Beta | 0.25 | 5 | 20 |
24 | Probability patient presents within 48 hours of ILI onset | Beta | 0.11 | 18.5 | 164 |
25 | Probability patient given antiviral treatment | presents < 48 hours | NA | 1 | 1 | – |
26 | Probability patient receives oseltamivir | prescribed antiviral | NA | 0.89 | 0.89 | – |
27 | Probability patient receives zanamivir | prescribed antiviral | NA | 0.11 | 0.11 | – |
28 | Probability adverse events – oseltamivir treatment | Beta | 0.02 | 1.72 | 86.11 |
29 | Probability adverse events – zanamivir treatment | Beta | 0.01 | 10.41 | 800.78 |
30 | Probability complication – no treatment | Beta | 0.09 | 942 | 10,145 |
31 | Odds ratio complication – oseltamivir treatment | Lognormal | 0.40 | –1 | 0 |
32 | Odds ratio complication – zanamivir treatment | Lognormal | 0.70 | –0.36 | 0.16 |
33 | Probability complication is respiratory | Dirichlet | 0.86 | 820 | 948 |
34 | Probability complication is cardiac | Dirichlet | 0.01 | 10 | 948 |
35 | Probability complication is CNS | Dirichlet | 0.02 | 22 | 948 |
36 | Probability complication is renal | Dirichlet | 0.01 | 6 | 948 |
37 | Probability complication is otitis media | Dirichlet | 0.02 | 22 | 948 |
38 | Probability complication is other | Dirichlet | 0.07 | 68 | 948 |
39 | Probability respiratory complication is pneumonia | Beta | 0.13 | 106 | 819 |
40 | Probability patient receives antibiotics | no complication | Beta | 0.55 | 8544 | 15,620 |
41 | Probability patient receives antibiotics | complication | Beta | 0.80 | 1527 | 1916 |
42 | Probability of influenza death | complication | Beta | 0.11 | 110 | 981 |
Cost/resource parameters | |||||
43 | Cost of amantadine prophylaxis course (without vaccine) | NA | £14.40 | £14.40 | – |
44 | Cost of amantadine prophylaxis course (with vaccine) | NA | £9.60 | £9.60 | – |
45 | Cost of oseltamivir prophylaxis course | NA | £81.80 | £81.80 | – |
46 | Cost of zanamivir prophylaxis course | NA | £73.65 | £73.65 | – |
47 | Cost of oseltamivir treatment course | NA | £16.36 | £16.36 | – |
48 | Cost of zanamivir treatment course | NA | £24.55 | £24.55 | – |
49 | Days per course – amantadine prophylaxis | NA | £42 | £42 | – |
50 | Days per course – amantadine prophylaxis (prior vaccination) | NA | 21 | 21 | – |
51 | Days per course – oseltamivir prophylaxis | NA | 42 | 42 | – |
52 | Days per course – zanamivir prophylaxis | NA | 28 | 28 | – |
53 | Acquisition cost for vaccination | NA | £5.63 | £5.63 | – |
54 | Administration cost for vaccination | NA | £25 | £25 | – |
55 | Cost of attendance at GP surgery consultation | NA | £25 | £25 | – |
56 | Cost of attendance at GP home visit | NA | £69 | £69 | – |
57 | Cost of attendance at A&E | NA | £95.56 | £95.56 | – |
58 | Probability A&E attendance | patient presents (no complication) | Beta | 0.03 | 8.35 | 270.11 |
59 | Probability GP attendance | patient presents (no complication) | NA | 0.97 | 0.97 | – |
60 | Probability home GP visit | GP presentation (no complication) | Beta | 0.38 | 62 | 165 |
61 | Probability A&E attendance | patient presents (complication) | Beta | 0.03 | 8.35 | 270.11 |
62 | Probability GP attendance | patient presents (complication) | NA | 0.97 | 0.97 | – |
63 | Probability home GP visit | GP presentation (complication) | Beta | 0.38 | 62 | 165 |
64 | Cost of uncomplicated influenza presentation | NA | £43.20 | £43.20 | – |
65 | Cost of complicated influenza presentation | NA | £43.20 | £43.20 | – |
66 | Cost of antibiotics course | NA | £6.80 | £6.80 | – |
67 | Cost of anti-emetics course (metaclopramide 7-day course) | NA | £1.69 | £1.69 | – |
68 | Cost of managing adverse events – vaccination | NA | £25 | £25 | – |
69 | Cost of managing adverse events – amantadine prophylaxis | NA | £25 | £25 | – |
70 | Cost of inpatient episode | Gamma | £261.17 | £261.17 | 5.16 |
71 | Probability hospitalisation no treatment | complication | Beta | 0.16 | 15 | 95 |
72 | Probability ICU care | complication | Beta | 0.05 | 22 | 453 |
73 | Inpatient LOS (days) | Gamma | 15 | 25 | 1 |
74 | Cost of ITU day | Normal | £1345.39 | £1345.39 | £31.95 |
75 | ITU LOS (days) | Gamma | 28 | 11.60 | 2.41 |
76 | Expected cost of hospitalisation | NA | £5747.01 | £5747.01 | – |
HRQoL parameters | |||||
77 | 21-day QALYs for influenza case – no treatment | Beta | 0.03 | 2820 | 100,000 |
78 | 21-day QALYs for influenza case – oseltamivir treatment | Beta | 0.03 | 2977 | 100,000 |
79 | 21-day QALYs for influenza case – zanamivir treatment | Beta | 0.03 | 2977 | 100,000 |
80 | QALY loss for influenza case – no treatment | NA | 0.02 | 0.02 | – |
81 | QALY loss for influenza case – oseltamivir treatment | NA | 0.01 | 0.01 | – |
82 | QALY loss for influenza case – zanamivir treatment | NA | 0.01 | 0.01 | – |
83 | Utility decrement – adverse events | Beta | 0.20 | 200 | 1000 |
84 | Duration adverse events | Gamma | 0.01 | 25 | 0 |
85 | Utility decrement respiratory complication | Lognormal | 0.15 | –1.90 | 0.41 |
86 | Utility decrement cardiac complication | Lognormal | 0.37 | –0.99 | 0.14 |
87 | Utility decrement CNS complication | Lognormal | 0.37 | –0.99 | 0.14 |
88 | Utility decrement renal complication | Lognormal | 0.37 | –0.99 | 0.14 |
89 | Utility decrement otitis media complication | Lognormal | 0.15 | –1.90 | 0.41 |
90 | Utility decrement other complication | Lognormal | 0.37 | –0.99 | 0.14 |
91 | Duration respiratory complication (years) | Gamma | 0.03 | 13.15 | 0.83 |
92 | Duration cardiac complication (years) | Gamma | 0.03 | 13.15 | 0.83 |
93 | Duration CNS complication (years) | Gamma | 0.03 | 13.15 | 0.83 |
94 | Duration renal complication (years) | Gamma | 0.03 | 13.15 | 0.83 |
95 | Duration otitis media complication (years) | Gamma | 0.03 | 9.73 | 0.96 |
96 | Duration other complication (years) | Gamma | 0.03 | 13.15 | 0.83 |
97 | Utility general population 0–24 | Normal | 0.94 | 0.94 | 0.01 |
98 | Utility general population 25–34 | Normal | 0.93 | 0.93 | 0.01 |
99 | Utility general population 35–44 | Normal | 0.91 | 0.91 | 0.01 |
100 | Utility general population 45–54 | Normal | 0.85 | 0.85 | 0.01 |
101 | Utility general population 55–64 | Normal | 0.80 | 0.80 | 0.01 |
102 | Utility general population 65–74 | Normal | 0.78 | 0.78 | 0.01 |
103 | Utility general population > 75 | Normal | 0.73 | 0.73 | 0.02 |
104 | Percentage population female | NA | 0.51 | 0.51 | – |
105 | QALY loss for premature death | NA | 2.95 | 2.95 | – |
106 | Discount rate for QALYs | NA | 3.50% | 3.50% | – |
No. | Parameter description | Distribution | Mean | Parameter 1 | Parameter 2 |
---|---|---|---|---|---|
Baseline event probabilities (disease) | |||||
1 | Baseline attack rate for influenza | Beta | 0.05 | 57 | 1098 |
2 | Probability ILI is influenza within epidemic period | Beta | 0.50 | 622 | 1256 |
3 | Probability influenza A strain is dominant | Beta | 0.75 | 9 | 12 |
4 | Probability influenza is influenza A in influenza A dominant seasons | Beta | 0.86 | 740 | 859 |
5 | Probability influenza is influenza A in influenza B dominant years | Beta | 0.30 | 83 | 281 |
6 | Probability influenza is influenza A | NA | 0.72 | 0.72 | – |
7 | Duration of influenza epidemic (days) | Gamma | 40 | 32.65 | 1.23 |
Effectiveness parameters (prevention) | |||||
8 | RR for influenza – vaccine | Lognormal | 0.42 | –0.87 | 0.23 |
9 | RR for influenza – amantadine prophylaxis | Lognormal | 0.40 | –0.92 | 0.83 |
10 | RR for influenza – oseltamivir prophylaxis | Lognormal | 0.08 | –2.50 | 1.04 |
11 | RR for influenza – zanamivir prophylaxis | Lognormal | 0.20 | –1.61 | 1.09 |
12 | Probability of amantadine resistance | Beta | 0.37 | 73.40 | 199 |
13 | Probability influenza case occurs within epidemic | Beta | 1 | 1 | 1125 |
14 | Probability influenza case avoidable – amantadine | NA | 1 | 1 | – |
15 | Probability influenza case avoidable – amantadine (vaccination) | NA | 0.53 | 0.53 | – |
16 | Probability influenza case avoidable – oseltamivir | NA | 1 | 1 | – |
17 | Percentage of influenza cases avoidable – zanamivir | NA | 0.70 | 0.70 | – |
Adverse events/withdrawals (prophylaxis) | |||||
18 | Probability adverse event – vaccination | Beta | 0.02 | 2 | 100 |
19 | Probability adverse event – amantadine prophylaxis | Beta | 0.05 | 10 | 200 |
20 | Probability withdrawal – amantadine prophylaxis | Beta | 0.15 | 0.37 | 2.55 |
21 | Probability withdrawal – oseltamivir prophylaxis | Beta | 0.02 | 1.72 | 86.11 |
22 | Probability withdrawal – zanamivir prophylaxis | Beta | 0.01 | 10.41 | 800.78 |
ILI event probabilities (treatment) | |||||
23 | Probability patient with ILI presents | Beta | 0.25 | 5 | 20 |
24 | Probability patient presents within 48 hours of ILI onset | Beta | 0.11 | 18.5 | 164 |
25 | Probability patient given antiviral treatment | presents < 48 hours | NA | 1 | 1 | – |
26 | Probability patient receives oseltamivir | prescribed antiviral | NA | 0.89 | 0.89 | – |
27 | Probability patient receives zanamivir | prescribed antiviral | NA | 0.11 | 0.11 | – |
28 | Probability adverse events – oseltamivir treatment | Beta | 0.02 | 1.72 | 86.11 |
29 | Probability adverse events – zanamivir treatment | Beta | 0.01 | 10.41 | 800.78 |
30 | Probability complication – no treatment | Beta | 0.12 | 908 | 7407 |
31 | Odds ratio complication – oseltamivir treatment | Lognormal | 0.40 | –1 | 0 |
32 | Odds ratio complication – zanamivir treatment | Lognormal | 0.49 | –0.71 | 0.38 |
33 | Probability complication is respiratory | Dirichlet | 0.83 | 755 | 914 |
34 | Probability complication is cardiac | Dirichlet | 0.07 | 60 | 914 |
35 | Probability complication is CNS | Dirichlet | 0.03 | 24 | 914 |
36 | Probability complication is renal | Dirichlet | 0.01 | 13 | 914 |
37 | Probability complication is otitis media | Dirichlet | 0.01 | 12 | 914 |
38 | Probability complication is other | Dirichlet | 0.05 | 50 | 914 |
39 | Probability respiratory complication is pneumonia | Beta | 0.13 | 97 | 754 |
40 | Probability patient receives antibiotics | no complication | Beta | 0.55 | 8544 | 15,620 |
41 | Probability patient receives antibiotics | complication | Beta | 0.80 | 1527 | 1916 |
42 | Probability of influenza death | complication | Beta | 0.12 | 114 | 936 |
Cost/resource parameters | |||||
43 | Cost of amantadine prophylaxis course (without vaccine) | NA | £14.40 | £14.40 | – |
44 | Cost of amantadine prophylaxis course (with vaccine) | NA | £9.60 | £9.60 | – |
45 | Cost of oseltamivir prophylaxis course | NA | £81.80 | £81.80 | – |
46 | Cost of zanamivir prophylaxis course | NA | £73.65 | £73.65 | – |
47 | Cost of oseltamivir treatment course | NA | £16.36 | £16.36 | – |
48 | Cost of zanamivir treatment course | NA | £24.55 | £24.55 | – |
49 | Days per course – amantadine prophylaxis | NA | £42 | £42 | – |
50 | Days per course – amantadine prophylaxis (prior vaccination) | NA | 21 | 21 | – |
51 | Days per course – oseltamivir prophylaxis | NA | 42 | 42 | – |
52 | Days per course – zanamivir prophylaxis | NA | 28 | 28 | – |
53 | Acquisition cost for vaccination | NA | £5.63 | £5.63 | – |
54 | Administration cost for vaccination | NA | £25 | £25 | – |
55 | Cost of attendance at GP surgery consultation | NA | £25 | £25 | – |
56 | Cost of attendance at GP home visit | NA | £69 | £69 | – |
57 | Cost of attendance at A&E | NA | £95.56 | £95.56 | – |
58 | Probability A&E attendance | patient presents (no complication) | Beta | 0.03 | 8.35 | 270.11 |
59 | Probability GP attendance | patient presents (no complication) | NA | 0.97 | 0.97 | – |
60 | Probability home GP visit | GP presentation (no complication) | Beta | 0.38 | 62 | 165 |
61 | Probability A&E attendance | patient presents (complication) | Beta | 0.03 | 8.35 | 270.11 |
62 | Probability GP attendance | patient presents (complication) | NA | 0.97 | 0.97 | – |
63 | Probability home GP visit | GP presentation (complication) | Beta | 0.38 | 62 | 165 |
64 | Cost of uncomplicated influenza presentation | NA | £43.20 | £43.20 | – |
65 | Cost of complicated influenza presentation | NA | £43.20 | £43.20 | – |
66 | Cost of antibiotics course | NA | £6.80 | £6.80 | – |
67 | Cost of anti-emetics course (metaclopramide 7-day course) | NA | £1.69 | £1.69 | – |
68 | Cost of managing adverse events – vaccination | NA | £25 | £25 | – |
69 | Cost of managing adverse events – amantadine prophylaxis | NA | £25 | £25 | – |
70 | Cost of inpatient episode | Gamma | £261.17 | £261.17 | 5.16 |
71 | Probability hospitalisation no treatment | complication | Beta | 0.16 | 15 | 95 |
72 | Probability ICU care | complication | Beta | 0.05 | 22 | 453 |
73 | Inpatient LOS (days) | Gamma | 15 | 25 | 1 |
74 | Cost of ITU day | Normal | £1345.39 | £1345.39 | £31.95 |
75 | ITU LOS (days) | Gamma | 28 | 11.60 | 2.41 |
76 | Expected cost of hospitalisation | NA | £5747.01 | £5747.01 | – |
HRQoL parameters | |||||
77 | 21-day QALYs for influenza case – no treatment | Beta | 0.03 | 2820 | 100,000 |
78 | 21-day QALYs for influenza case – oseltamivir treatment | Beta | 0.03 | 2977 | 100,000 |
79 | 21-day QALYs for influenza case – zanamivir treatment | Beta | 0.03 | 2977 | 100,000 |
80 | QALY loss for influenza case – no treatment | NA | 0.02 | 0.02 | – |
81 | QALY loss for influenza case – oseltamivir treatment | NA | 0.01 | 0.01 | – |
82 | QALY loss for influenza case – zanamivir treatment | NA | 0.01 | 0.01 | – |
83 | Utility decrement – adverse events | Beta | 0.20 | 200 | 1000 |
84 | Duration adverse events | Gamma | 0.01 | 25 | 0 |
85 | Utility decrement respiratory complication | Lognormal | 0.15 | –1.90 | 0.41 |
86 | Utility decrement cardiac complication | Lognormal | 0.37 | –0.99 | 0.14 |
87 | Utility decrement CNS complication | Lognormal | 0.37 | –0.99 | 0.14 |
88 | Utility decrement renal complication | Lognormal | 0.37 | –0.99 | 0.14 |
89 | Utility decrement otitis media complication | Lognormal | 0.15 | –1.90 | 0.41 |
90 | Utility decrement other complication | Lognormal | 0.37 | –0.99 | 0.14 |
91 | Duration respiratory complication (years) | Gamma | 0.03 | 13.13 | 0.83 |
92 | Duration cardiac complication (years) | Gamma | 0.03 | 13.13 | 0.83 |
93 | Duration CNS complication (years) | Gamma | 0.03 | 13.13 | 0.83 |
94 | Duration renal complication (years) | Gamma | 0.03 | 13.13 | 0.83 |
95 | Duration otitis media complication (years) | Gamma | 0.03 | 9.73 | 0.96 |
96 | Duration other complication (years) | Gamma | 0.03 | 13.13 | 0.83 |
97 | Utility general population 0–24 | Normal | 0.94 | 0.94 | 0.01 |
98 | Utility general population 25–34 | Normal | 0.93 | 0.93 | 0.01 |
99 | Utility general population 35–44 | Normal | 0.91 | 0.91 | 0.01 |
100 | Utility general population 45–54 | Normal | 0.85 | 0.85 | 0.01 |
101 | Utility general population 55–64 | Normal | 0.80 | 0.80 | 0.01 |
102 | Utility general population 65–74 | Normal | 0.78 | 0.78 | 0.01 |
103 | Utility general population > 75 | Normal | 0.73 | 0.73 | 0.02 |
104 | Percentage population female | NA | 0.51 | 0.51 | – |
105 | QALY loss for premature death | NA | 2.95 | 2.95 | – |
106 | Discount rate for QALYs | NA | 3.50% | 3.50% | – |
List of model parameters – post-exposure prophylaxis
No. | Parameter description | Distribution | Mean | Parameter 1 | Parameter 2 |
---|---|---|---|---|---|
Baseline event probabilities (disease) | |||||
1 | Baseline attack rate for influenza | Beta | 0.19 | 21 | 111 |
2 | Probability ILI is influenza within epidemic period | Beta | 0.50 | 622 | 1256 |
3 | Probability influenza A strain is dominant | Beta | 0.75 | 9 | 12 |
4 | Probability influenza is influenza A in influenza A dominant seasons | Beta | 0.86 | 740 | 859 |
5 | Probability influenza is influenza A in influenza B dominant years | Beta | 0.30 | 83 | 281 |
6 | Probability influenza is influenza A | NA | 0.72 | 0.72 | – |
7 | Duration of influenza epidemic (days) | Gamma | 40 | 32.65 | 1.23 |
Effectiveness parameters (prevention) | |||||
8 | RR for influenza – vaccine | Lognormal | 0.36 | –1.02 | 0.14 |
9 | RR for influenza – amantadine prophylaxis | Lognormal | 0.10 | –2.26 | 0.60 |
10 | RR for influenza – oseltamivir prophylaxis | Lognormal | 0.36 | –1.03 | 0.41 |
11 | RR for influenza – zanamivir prophylaxis | Lognormal | 0.21 | –1.56 | 0.24 |
12 | Probability of amantadine resistance | Beta | 0.37 | 73.40 | 199 |
13 | Probability influenza case occurs within epidemic | Beta | 1 | 1 | 1125 |
14 | Probability influenza case avoidable – amantadine | NA | 1 | 1 | – |
15 | Probability influenza case avoidable – amantadine (vaccination) | NA | 1 | 1 | – |
16 | Probability influenza case avoidable – oseltamivir | NA | 1 | 1 | – |
17 | Percentage of influenza cases avoidable – zanamivir | NA | 1 | 1 | – |
Adverse events/withdrawals (prophylaxis) | |||||
18 | Probability adverse event – vaccination | Beta | 0.02 | 2 | 100 |
19 | Probability adverse event – amantadine prophylaxis | Beta | 0.05 | 10 | 200 |
20 | Probability withdrawal – amantadine prophylaxis | Beta | 0.06 | 13.56 | 237.89 |
21 | Probability withdrawal – oseltamivir prophylaxis | Beta | 0.02 | 1.72 | 86.11 |
22 | Probability withdrawal – zanamivir prophylaxis | Beta | 0.01 | 10.41 | 800.78 |
ILI event probabilities (treatment) | |||||
23 | Probability patient with ILI presents | Beta | 0.25 | 5 | 20 |
24 | Probability patient presents within 48 hours of ILI onset | Beta | 0.52 | 38 | 73 |
25 | Probability patient given antiviral treatment | presents < 48 hours | NA | 0 | 0 | – |
26 | Probability patient receives oseltamivir | prescribed antiviral | NA | 1 | 1 | – |
27 | Probability patient receives zanamivir | prescribed antiviral | NA | 0 | 0 | – |
28 | Probability adverse events – oseltamivir treatment | Beta | 0.02 | 1.72 | 86.11 |
29 | Probability adverse events – zanamivir treatment | Beta | 0.01 | 10.41 | 800.78 |
30 | Probability complication – no treatment | Beta | 0.14 | 2417 | 17,201 |
31 | Odds ratio complication – oseltamivir treatment | Lognormal | 0.65 | 0 | 0 |
32 | Odds ratio complication – zanamivir treatment | Lognormal | 0.70 | –0.36 | 0.16 |
33 | Probability complication is respiratory | Dirichlet | 0.70 | 1698 | 2423 |
34 | Probability complication is cardiac | Dirichlet | 0 | 1 | 2423 |
35 | Probability complication is CNS | Dirichlet | 0.01 | 18 | 2423 |
36 | Probability complication is renal | Dirichlet | 0 | 3 | 2423 |
37 | Probability complication is otitis media | Dirichlet | 0.28 | 685 | 2423 |
38 | Probability complication is other | Dirichlet | 0.01 | 18 | 2423 |
39 | Probability respiratory complication is pneumonia | Beta | 0.02 | 29 | 1697 |
40 | Probability patient receives antibiotics | no complication | Beta | 0.28 | 4997 | 17,910 |
41 | Probability patient receives antibiotics | complication | Beta | 0.74 | 2183 | 2962 |
42 | Probability of influenza death | complication | Beta | 0 | 1 | 2311 |
Cost/resource parameters | |||||
43 | Cost of amantadine prophylaxis course (without vaccine) | NA | £4.80 | £4.80 | – |
44 | Cost of amantadine prophylaxis course (with vaccine) | NA | £4.80 | £4.80 | – |
45 | Cost of oseltamivir prophylaxis course | NA | £16.36 | £16.36 | – |
46 | Cost of zanamivir prophylaxis course | NA | £24.55 | £24.55 | – |
47 | Cost of oseltamivir treatment course | NA | £16.36 | £16.36 | – |
48 | Cost of zanamivir treatment course | NA | £24.55 | £24.55 | – |
49 | Days per course – amantadine prophylaxis | NA | £10 | £10 | – |
50 | Days per course – amantadine prophylaxis (prior vaccination) | NA | 10 | 10 | – |
51 | Days per course – oseltamivir prophylaxis | NA | 10 | 10 | – |
52 | Days per course – zanamivir prophylaxis | NA | 10 | 10 | – |
53 | Acquisition cost for vaccination | NA | £5.63 | £5.63 | – |
54 | Administration cost for vaccination | NA | £25 | £25 | – |
55 | Cost of attendance at GP surgery consultation | NA | £25 | £25 | – |
56 | Cost of attendance at GP home visit | NA | £69 | £69 | – |
57 | Cost of attendance at A&E | NA | £95.56 | £95.56 | – |
58 | Probability A&E attendance | patient presents (no complication) | Beta | 0.03 | 8.35 | 270.11 |
59 | Probability GP attendance | patient presents (no complication) | NA | 0.97 | 0.97 | – |
60 | Probability home GP visit | GP presentation (no complication) | Beta | 0.05 | 4 | 73 |
61 | Probability A&E attendance | patient presents (complication) | Beta | 0.03 | 8.35 | 270.11 |
62 | Probability GP attendance | patient presents (complication) | NA | 0.97 | 0.97 | – |
63 | Probability home GP visit | GP presentation (complication) | Beta | 0.05 | 4 | 73 |
64 | Cost of uncomplicated influenza presentation | NA | £29.52 | £29.52 | – |
65 | Cost of complicated influenza presentation | NA | £29.52 | £29.52 | – |
66 | Cost of antibiotics course | NA | £6.80 | £6.80 | – |
67 | Cost of anti-emetics course (metaclopramide 7-day course) | NA | £1.69 | £1.69 | – |
68 | Cost of managing adverse events – vaccination | NA | £25 | £25 | – |
69 | Cost of managing adverse events – amantadine prophylaxis | NA | £25 | £25 | – |
70 | Cost of inpatient episode | Gamma | £261.17 | £261.17 | 5.16 |
71 | Probability hospitalisation no treatment | complication | Beta | 0.11 | 5 | 46 |
72 | Probability ICU care | complication | Beta | 0.05 | 22 | 453 |
73 | Inpatient LOS (days) | Gamma | 2.30 | 1 | 4 |
74 | Cost of ITU day | Normal | £1345.39 | £1345.39 | £31.95 |
75 | ITU LOS (days) | Gamma | 28 | 11.60 | 2.41 |
76 | Expected cost of hospitalisation | NA | £2430.18 | £2430.18 | – |
HRQoL parameters | |||||
77 | 21-day QALYs for influenza case – no treatment | Beta | 0.04 | 4146 | 100,000 |
78 | 21-day QALYs for influenza case – oseltamivir treatment | Beta | 0.04 | 4247 | 100,000 |
79 | 21-day QALYs for influenza case – zanamivir treatment | Beta | 0.04 | 4247 | 100,000 |
80 | QALY loss for influenza case – no treatment | NA | 0.01 | 0.01 | – |
81 | QALY loss for influenza case – oseltamivir treatment | NA | 0.01 | 0.01 | – |
82 | QALY loss for influenza case – zanamivir treatment | NA | 0.01 | 0.01 | – |
83 | Utility decrement – adverse events | Beta | 0.20 | 200 | 1000 |
84 | Duration adverse events | Gamma | 0.01 | 25 | 0 |
85 | Utility decrement respiratory complication | Lognormal | 0.15 | –1.90 | 0.41 |
86 | Utility decrement cardiac complication | Lognormal | 0.37 | –0.99 | 0.14 |
87 | Utility decrement CNS complication | Lognormal | 0.37 | –0.99 | 0.14 |
88 | Utility decrement renal complication | Lognormal | 0.37 | –0.99 | 0.14 |
89 | Utility decrement otitis media complication | Lognormal | 0.15 | –1.90 | 0.41 |
90 | Utility decrement other complication | Lognormal | 0.37 | –0.99 | 0.14 |
91 | Duration respiratory complication (years) | Gamma | 0.02 | 6.92 | 1.14 |
92 | Duration cardiac complication (years) | Gamma | 0.02 | 6.92 | 1.14 |
93 | Duration CNS complication (years) | Gamma | 0.02 | 6.92 | 1.14 |
94 | Duration renal complication (years) | Gamma | 0.02 | 6.92 | 1.14 |
95 | Duration otitis media complication (years) | Gamma | 0.03 | 9.73 | 0.96 |
96 | Duration other complication (years) | Gamma | 0.02 | 6.92 | 1.14 |
97 | Utility general population 0–24 | Normal | 0.94 | 0.94 | 0.01 |
98 | Utility general population 25–34 | Normal | 0.93 | 0.93 | 0.01 |
99 | Utility general population 35–44 | Normal | 0.91 | 0.91 | 0.01 |
100 | Utility general population 45–54 | Normal | 0.85 | 0.85 | 0.01 |
101 | Utility general population 55–64 | Normal | 0.80 | 0.80 | 0.01 |
102 | Utility general population 65–74 | Normal | 0.78 | 0.78 | 0.01 |
103 | Utility general population > 75 | Normal | 0.73 | 0.73 | 0.02 |
104 | Percentage population female | NA | 0.51 | 0.51 | – |
105 | QALY loss for premature death | NA | 24.74 | 24.74 | – |
106 | Discount rate for QALYs | NA | 3.50% | 3.50% | – |
No. | Parameter description | Distribution | Mean | Parameter 1 | Parameter 2 |
---|---|---|---|---|---|
Baseline event probabilities (disease) | |||||
1 | Baseline attack rate for influenza | Beta | 0.19 | 21 | 111 |
2 | Probability ILI is influenza within epidemic period | Beta | 0.50 | 622 | 1256 |
3 | Probability influenza A strain is dominant | Beta | 0.75 | 9 | 12 |
4 | Probability influenza is influenza A in influenza A dominant seasons | Beta | 0.86 | 740 | 859 |
5 | Probability influenza is influenza A in influenza B dominant years | Beta | 0.30 | 83 | 281 |
6 | Probability influenza is influenza A | NA | 0.72 | 0.72 | – |
7 | Duration of influenza epidemic (days) | Gamma | 40 | 32.65 | 1.23 |
Effectiveness parameters (prevention) | |||||
8 | RR for influenza – vaccine | Lognormal | 0.36 | –1.02 | 0.14 |
9 | RR for influenza – amantadine prophylaxis | Lognormal | 0.10 | –2.26 | 0.60 |
10 | RR for influenza – oseltamivir prophylaxis | Lognormal | 0.36 | –1.03 | 0.41 |
11 | RR for influenza – zanamivir prophylaxis | Lognormal | 0.21 | –1.56 | 0.24 |
12 | Probability of amantadine resistance | Beta | 0.37 | 73.40 | 199 |
13 | Probability influenza case occurs within epidemic | Beta | 1 | 1 | 1125 |
14 | Probability influenza case avoidable – amantadine | NA | 1 | 1 | – |
15 | Probability influenza case avoidable – amantadine (vaccination) | NA | 1 | 1 | – |
16 | Probability influenza case avoidable – oseltamivir | NA | 1 | 1 | – |
17 | Percentage of influenza cases avoidable – zanamivir | NA | 1 | 1 | – |
Adverse events/withdrawals (prophylaxis) | |||||
18 | Probability adverse event – vaccination | Beta | 0.02 | 2 | 100 |
19 | Probability adverse event – amantadine prophylaxis | Beta | 0.05 | 10 | 200 |
20 | Probability withdrawal – amantadine prophylaxis | Beta | 0.06 | 13.56 | 237.89 |
21 | Probability withdrawal – oseltamivir prophylaxis | Beta | 0.02 | 1.72 | 86.11 |
22 | Probability withdrawal – zanamivir prophylaxis | Beta | 0.01 | 10.41 | 800.78 |
ILI event probabilities (treatment) | |||||
23 | Probability patient with ILI presents | Beta | 0.25 | 5 | 20 |
24 | Probability patient presents within 48 hours of ILI onset | Beta | 0.52 | 38 | 73 |
25 | Probability patient given antiviral treatment | presents < 48 hours | NA | 1 | 1 | – |
26 | Probability patient receives oseltamivir | prescribed antiviral | NA | 1 | 1 | – |
27 | Probability patient receives zanamivir | prescribed antiviral | NA | 0 | 0 | – |
28 | Probability adverse events – oseltamivir treatment | Beta | 0.02 | 1.72 | 86.11 |
29 | Probability adverse events – zanamivir treatment | Beta | 0.01 | 10.41 | 800.78 |
30 | Probability complication – no treatment | Beta | 0.18 | 675 | 3695 |
31 | Odds ratio complication – oseltamivir treatment | Lognormal | 0.65 | 0 | 0 |
32 | Odds ratio complication – zanamivir treatment | Lognormal | 0.49 | –0.71 | 0.38 |
33 | Probability complication is respiratory | Dirichlet | 0.77 | 521 | 681 |
34 | Probability complication is cardiac | Dirichlet | 0 | 1 | 681 |
35 | Probability complication is CNS | Dirichlet | 0 | 1 | 681 |
36 | Probability complication is renal | Dirichlet | 0 | 1 | 681 |
37 | Probability complication is otitis media | Dirichlet | 0.23 | 154 | 681 |
38 | Probability complication is other | Dirichlet | 0 | 3 | 681 |
39 | Probability respiratory complication is pneumonia | Beta | 0.02 | 9 | 520 |
40 | Probability patient receives antibiotics | no complication | Beta | 0.28 | 4997 | 17,910 |
41 | Probability patient receives antibiotics | complication | Beta | 0.74 | 2183 | 2962 |
42 | Probability of influenza death | complication | Beta | 0 | 1 | 650 |
Cost/resource parameters | |||||
43 | Cost of amantadine prophylaxis course (without vaccine) | NA | £4.80 | £4.80 | – |
44 | Cost of amantadine prophylaxis course (with vaccine) | NA | £4.80 | £4.80 | – |
45 | Cost of oseltamivir prophylaxis course | NA | £16.36 | £16.36 | – |
46 | Cost of zanamivir prophylaxis course | NA | £24.55 | £24.55 | – |
47 | Cost of oseltamivir treatment course | NA | £16.36 | £16.36 | – |
48 | Cost of zanamivir treatment course | NA | £24.55 | £24.55 | – |
49 | Days per course – amantadine prophylaxis | NA | £10 | £10 | – |
50 | Days per course – amantadine prophylaxis (prior vaccination) | NA | 10 | 10 | – |
51 | Days per course – oseltamivir prophylaxis | NA | 10 | 10 | – |
52 | Days per course – zanamivir prophylaxis | NA | 10 | 10 | – |
53 | Acquisition cost for vaccination | NA | £5.63 | £5.63 | – |
54 | Administration cost for vaccination | NA | £25 | £25 | – |
55 | Cost of attendance at GP surgery consultation | NA | £25 | £25 | – |
56 | Cost of attendance at GP home visit | NA | £69 | £69 | – |
57 | Cost of attendance at A&E | NA | £95.56 | £95.56 | – |
58 | Probability A&E attendance | patient presents (no complication) | Beta | 0.03 | 8.35 | 270.11 |
59 | Probability GP attendance | patient presents (no complication) | NA | 0.97 | 0.97 | – |
60 | Probability home GP visit | GP presentation (no complication) | Beta | 0.05 | 4 | 73 |
61 | Probability A&E attendance | patient presents (complication) | Beta | 0.03 | 8.35 | 270.11 |
62 | Probability GP attendance | patient presents (complication) | NA | 0.97 | 0.97 | – |
63 | Probability home GP visit | GP presentation (complication) | Beta | 0.05 | 4 | 73 |
64 | Cost of uncomplicated influenza presentation | NA | £29.52 | £29.52 | – |
65 | Cost of complicated influenza presentation | NA | £29.52 | £29.52 | – |
66 | Cost of antibiotics course | NA | £6.80 | £6.80 | – |
67 | Cost of anti-emetics course (metaclopramide 7-day course) | NA | £1.69 | £1.69 | – |
68 | Cost of managing adverse events – vaccination | NA | £25 | £25 | – |
69 | Cost of managing adverse events – amantadine prophylaxis | NA | £25 | £25 | – |
70 | Cost of inpatient episode | Gamma | £261.17 | £261.17 | 5.16 |
71 | Probability hospitalisation no treatment | complication | Beta | 0.16 | 15 | 95 |
72 | Probability ICU care | complication | Beta | 0.05 | 22 | 453 |
73 | Inpatient LOS (days) | Gamma | 2.30 | 1 | 4 |
74 | Cost of ITU day | Normal | £1345.39 | £1345.39 | £31.95 |
75 | ITU LOS (days) | Gamma | 28 | 11.60 | 2.41 |
76 | Expected cost of hospitalisation | NA | £2430.18 | £2430.18 | – |
HRQoL parameters | |||||
77 | 21-day QALYs for influenza case – no treatment | Beta | 0.03 | 2820 | 100,000 |
78 | 21-day QALYs for influenza case – oseltamivir treatment | Beta | 0.03 | 2977 | 100,000 |
79 | 21-day QALYs for influenza case – zanamivir treatment | Beta | 0.03 | 2977 | 100,000 |
80 | QALY loss for influenza case – no treatment | NA | 0.02 | 0.02 | – |
81 | QALY loss for influenza case – oseltamivir treatment | NA | 0.02 | 0.02 | – |
82 | QALY loss for influenza case – zanamivir treatment | NA | 0.02 | 0.02 | – |
83 | Utility decrement – adverse events | Beta | 0.20 | 200 | 1000 |
84 | Duration adverse events | Gamma | 0.01 | 25 | 0 |
85 | Utility decrement respiratory complication | Lognormal | 0.15 | –1.90 | 0.41 |
86 | Utility decrement cardiac complication | Lognormal | 0.37 | –0.99 | 0.14 |
87 | Utility decrement CNS complication | Lognormal | 0.37 | –0.99 | 0.14 |
88 | Utility decrement renal complication | Lognormal | 0.37 | –0.99 | 0.14 |
89 | Utility decrement otitis media complication | Lognormal | 0.15 | –1.90 | 0.41 |
90 | Utility decrement other complication | Lognormal | 0.37 | –0.99 | 0.14 |
91 | Duration respiratory complication (years) | Gamma | 0.02 | 7.24 | 1.11 |
92 | Duration cardiac complication (years) | Gamma | 0.02 | 7.24 | 1.11 |
93 | Duration CNS complication (years) | Gamma | 0.02 | 7.24 | 1.11 |
94 | Duration renal complication (years) | Gamma | 0.02 | 7.24 | 1.11 |
95 | Duration otitis media complication (years) | Gamma | 0.03 | 9.73 | 0.96 |
96 | Duration other complication (years) | Gamma | 0.02 | 7.24 | 1.11 |
97 | Utility general population 0–24 | Normal | 0.94 | 0.94 | 0.01 |
98 | Utility general population 25–34 | Normal | 0.93 | 0.93 | 0.01 |
99 | Utility general population 35–44 | Normal | 0.91 | 0.91 | 0.01 |
100 | Utility general population 45–54 | Normal | 0.85 | 0.85 | 0.01 |
101 | Utility general population 55–64 | Normal | 0.80 | 0.80 | 0.01 |
102 | Utility general population 65–74 | Normal | 0.78 | 0.78 | 0.01 |
103 | Utility general population > 75 | Normal | 0.73 | 0.73 | 0.02 |
104 | Percentage population female | NA | 0.51 | 0.51 | – |
105 | QALY loss for premature death | NA | 24.74 | 24.74 | – |
106 | Discount rate for QALYs | NA | 3.50% | 3.50% | – |
No. | Parameter description | Distribution | Mean | Parameter 1 | Parameter 2 |
---|---|---|---|---|---|
Baseline event probabilities (disease) | |||||
1 | Baseline attack rate for influenza | Beta | 0.09 | 180 | 2051 |
2 | Probability ILI is influenza within epidemic period | Beta | 0.50 | 622 | 1256 |
3 | Probability influenza A strain is dominant | Beta | 0.75 | 9 | 12 |
4 | Probability influenza is influenza A in influenza A dominant seasons | Beta | 0.86 | 740 | 859 |
5 | Probability influenza is influenza A in influenza B dominant years | Beta | 0.30 | 83 | 281 |
6 | Probability influenza is influenza A | NA | 0.72 | 0.72 | – |
7 | Duration of influenza epidemic (days) | Gamma | 40 | 32.65 | 1.23 |
Effectiveness parameters (prevention) | |||||
8 | RR for influenza – vaccine | Lognormal | 0.35 | –1.05 | 0.17 |
9 | RR for influenza – amantadine prophylaxis | Lognormal | 0.10 | –2.26 | 0.60 |
10 | RR for influenza – oseltamivir prophylaxis | Lognormal | 0.19 | –1.66 | 0.44 |
11 | RR for influenza – zanamivir prophylaxis | Lognormal | 0.21 | –1.56 | 0.24 |
12 | Probability of amantadine resistance | Beta | 0.37 | 73.40 | 199 |
13 | Probability influenza case occurs within epidemic | Beta | 1 | 1 | 1125 |
14 | Probability influenza case avoidable – amantadine | NA | 1 | 1 | – |
15 | Probability influenza case avoidable – amantadine (vaccination) | NA | 1 | 1 | – |
16 | Probability influenza case avoidable – oseltamivir | NA | 1 | 1 | – |
17 | Percentage of influenza cases avoidable – zanamivir | NA | 1 | 1 | – |
Adverse events/withdrawals (prophylaxis) | |||||
18 | Probability adverse event – vaccination | Beta | 0.02 | 2 | 100 |
19 | Probability adverse event – amantadine prophylaxis | Beta | 0.05 | 10 | 200 |
20 | Probability withdrawal – amantadine prophylaxis | Beta | 0.06 | 13.56 | 237.89 |
21 | Probability withdrawal – oseltamivir prophylaxis | Beta | 0.02 | 1.72 | 86.11 |
22 | Probability withdrawal – zanamivir prophylaxis | Beta | 0.01 | 10.41 | 800.78 |
ILI event probabilities (treatment) | |||||
23 | Probability patient with ILI presents | Beta | 0.25 | 5 | 20 |
24 | Probability patient presents within 48 hours of ILI onset | Beta | 0.16 | 104 | 668 |
25 | Probability patient given antiviral treatment | presents < 48 hours | NA | 0 | 0 | – |
26 | Probability patient receives oseltamivir | prescribed antiviral | NA | 0.89 | 0.89 | – |
27 | Probability patient receives zanamivir | prescribed antiviral | NA | 0.11 | 0.11 | – |
28 | Probability adverse events – oseltamivir treatment | Beta | 0.02 | 1.72 | 86.11 |
29 | Probability adverse events – zanamivir treatment | Beta | 0.01 | 10.41 | 800.78 |
30 | Probability complication – no treatment | Beta | 0.08 | 6509 | 85,248 |
31 | Odds ratio complication – oseltamivir treatment | Lognormal | 0.40 | –1 | 0 |
32 | Odds ratio complication – zanamivir treatment | Lognormal | 0.70 | –0.36 | 0.16 |
33 | Probability complication is respiratory | Dirichlet | 0.87 | 5637 | 6515 |
34 | Probability complication is cardiac | Dirichlet | 0 | 12 | 6515 |
35 | Probability complication is CNS | Dirichlet | 0.02 | 102 | 6515 |
36 | Probability complication is renal | Dirichlet | 0 | 10 | 6515 |
37 | Probability complication is otitis media | Dirichlet | 0.08 | 501 | 6515 |
38 | Probability complication is other | Dirichlet | 0.04 | 253 | 6515 |
39 | Probability respiratory complication is pneumonia | Beta | 0.04 | 237 | 5636 |
40 | Probability patient receives antibiotics | no complication | Beta | 0.42 | 19811 | 47,169 |
41 | Probability patient receives antibiotics | complication | Beta | 0.81 | 6983 | 8579 |
42 | Probability of influenza death | complication | Beta | 0.01 | 33 | 6437 |
Cost/resource parameters | |||||
43 | Cost of amantadine prophylaxis course (without vaccine) | NA | £4.80 | £4.80 | – |
44 | Cost of amantadine prophylaxis course (with vaccine) | NA | £4.80 | £4.80 | – |
45 | Cost of oseltamivir prophylaxis course | NA | £16.36 | £16.36 | – |
46 | Cost of zanamivir prophylaxis course | NA | £24.55 | £24.55 | – |
47 | Cost of oseltamivir treatment course | NA | £16.36 | £16.36 | – |
48 | Cost of zanamivir treatment course | NA | £24.55 | £24.55 | – |
49 | Days per course – amantadine prophylaxis | NA | £10 | £10 | – |
50 | Days per course – amantadine prophylaxis (prior vaccination) | NA | 10 | 10 | – |
51 | Days per course – oseltamivir prophylaxis | NA | 10 | 10 | – |
52 | Days per course – zanamivir prophylaxis | NA | 10 | 10 | – |
53 | Acquisition cost for vaccination | NA | £5.63 | £5.63 | – |
54 | Administration cost for vaccination | NA | £25 | £25 | – |
55 | Cost of attendance at GP surgery consultation | NA | £25 | £25 | – |
56 | Cost of attendance at GP home visit | NA | £69 | £69 | – |
57 | Cost of attendance at A&E | NA | £95.56 | £95.56 | – |
58 | Probability A&E attendance | patient presents (no complication) | Beta | 0.03 | 8.35 | 270.11 |
59 | Probability GP attendance | patient presents (no complication) | NA | 0.97 | 0.97 | – |
60 | Probability home GP visit | GP presentation (no complication) | Beta | 0.08 | 56 | 674 |
61 | Probability A&E attendance | patient presents (complication) | Beta | 0.03 | 8.35 | 270.11 |
62 | Probability GP attendance | patient presents (complication) | NA | 0.97 | 0.97 | – |
63 | Probability home GP visit | GP presentation (complication) | Beta | 0.08 | 56 | 674 |
64 | Cost of uncomplicated influenza presentation | NA | £30.73 | £30.73 | – |
65 | Cost of complicated influenza presentation | NA | £30.73 | £30.73 | – |
66 | Cost of antibiotics course | NA | £6.80 | £6.80 | – |
67 | Cost of anti-emetics course (metaclopramide 7-day course) | NA | £1.69 | £1.69 | – |
68 | Cost of managing adverse events – vaccination | NA | £25 | £25 | – |
69 | Cost of managing adverse events – amantadine prophylaxis | NA | £25 | £25 | – |
70 | Cost of inpatient episode | Gamma | £261.17 | £261.17 | 5.16 |
71 | Probability hospitalisation no treatment | complication | Beta | 0.11 | 5 | 46 |
72 | Probability ICU care | complication | Beta | 0.05 | 22 | 453 |
73 | Inpatient LOS (days) | Gamma | 11.90 | 16 | 1 |
74 | Cost of ITU day | Normal | £1345.39 | £1345.39 | £31.95 |
75 | ITU LOS (days) | Gamma | 28 | 11.60 | 2.41 |
76 | Expected cost of hospitalisation | NA | £4937.39 | £4937.39 | – |
HRQoL parameters | |||||
77 | 21-day QALYs for influenza case – no treatment | Beta | 0.04 | 4146 | 100,000 |
78 | 21-day QALYs for influenza case – oseltamivir treatment | Beta | 0.04 | 4247 | 100,000 |
79 | 21-day QALYs for influenza case – zanamivir treatment | Beta | 0.04 | 4247 | 100,000 |
80 | QALY loss for influenza case – no treatment | NA | 0.01 | 0.01 | – |
81 | QALY loss for influenza case – oseltamivir treatment | NA | 0.01 | 0.01 | – |
82 | QALY loss for influenza case – zanamivir treatment | NA | 0.01 | 0.01 | – |
83 | Utility decrement – adverse events | Beta | 0.20 | 200 | 1000 |
84 | Duration adverse events | Gamma | 0.01 | 25 | 0 |
85 | Utility decrement respiratory complication | Lognormal | 0.15 | –1.90 | 0.41 |
86 | Utility decrement cardiac complication | Lognormal | 0.37 | –0.99 | 0.14 |
87 | Utility decrement CNS complication | Lognormal | 0.37 | –0.99 | 0.14 |
88 | Utility decrement renal complication | Lognormal | 0.37 | –0.99 | 0.14 |
89 | Utility decrement otitis media complication | Lognormal | 0.15 | –1.90 | 0.41 |
90 | Utility decrement other complication | Lognormal | 0.37 | –0.99 | 0.14 |
91 | Duration respiratory complication (years) | Gamma | 0.03 | 9.46 | 0.98 |
92 | Duration cardiac complication (years) | Gamma | 0.03 | 9.46 | 0.98 |
93 | Duration CNS complication (years) | Gamma | 0.03 | 9.46 | 0.98 |
94 | Duration renal complication (years) | Gamma | 0.03 | 9.46 | 0.98 |
95 | Duration otitis media complication (years) | Gamma | 0.03 | 9.73 | 0.96 |
96 | Duration other complication (years) | Gamma | 0.03 | 9.46 | 0.98 |
97 | Utility general population 0–24 | Normal | 0.94 | 0.94 | 0.01 |
98 | Utility general population 25–34 | Normal | 0.93 | 0.93 | 0.01 |
99 | Utility general population 35–44 | Normal | 0.91 | 0.91 | 0.01 |
100 | Utility general population 45–54 | Normal | 0.85 | 0.85 | 0.01 |
101 | Utility general population 55–64 | Normal | 0.80 | 0.80 | 0.01 |
102 | Utility general population 65–74 | Normal | 0.78 | 0.78 | 0.01 |
103 | Utility general population > 75 | Normal | 0.73 | 0.73 | 0.02 |
104 | Percentage population female | NA | 0.51 | 0.51 | – |
105 | QALY loss for premature death | NA | 13.37 | 13.37 | – |
106 | Discount rate for QALYs | NA | 3.50% | 3.50% | – |
No. | Parameter description | Distribution | Mean | Parameter 1 | Parameter 2 |
---|---|---|---|---|---|
Baseline event probabilities (disease) | |||||
1 | Baseline attack rate for influenza | Beta | 0.09 | 180 | 2051 |
2 | Probability ILI is influenza within epidemic period | Beta | 0.50 | 622 | 1256 |
3 | Probability influenza A strain is dominant | Beta | 0.75 | 9 | 12 |
4 | Probability influenza is influenza A in influenza A dominant seasons | Beta | 0.86 | 740 | 859 |
5 | Probability influenza is influenza A in influenza B dominant years | Beta | 0.30 | 83 | 281 |
6 | Probability influenza is influenza A | NA | 0.72 | 0.72 | – |
7 | Duration of influenza epidemic (days) | Gamma | 40 | 32.65 | 1.23 |
Effectiveness parameters (prevention) | |||||
8 | RR for influenza – vaccine | Lognormal | 0.35 | –1.05 | 0.17 |
9 | RR for influenza – amantadine prophylaxis | Lognormal | 0.10 | –2.26 | 0.60 |
10 | RR for influenza – oseltamivir prophylaxis | Lognormal | 0.19 | –1.66 | 0.44 |
11 | RR for influenza – zanamivir prophylaxis | Lognormal | 0.21 | –1.56 | 0.24 |
12 | Probability of amantadine resistance | Beta | 0.37 | 73.40 | 199 |
13 | Probability influenza case occurs within epidemic | Beta | 1 | 1 | 1125 |
14 | Probability influenza case avoidable – amantadine | NA | 1 | 1 | – |
15 | Probability influenza case avoidable – amantadine (vaccination) | NA | 1 | 1 | – |
16 | Probability influenza case avoidable – oseltamivir | NA | 1 | 1 | – |
17 | Percentage of influenza cases avoidable – zanamivir | NA | 1 | 1 | – |
Adverse events/withdrawals (prophylaxis) | |||||
18 | Probability adverse event – vaccination | Beta | 0.02 | 2 | 100 |
19 | Probability adverse event – amantadine prophylaxis | Beta | 0.05 | 10 | 200 |
20 | Probability withdrawal – amantadine prophylaxis | Beta | 0.15 | 0.37 | 2.55 |
21 | Probability withdrawal – oseltamivir prophylaxis | Beta | 0.02 | 1.72 | 86.11 |
22 | Probability withdrawal – zanamivir prophylaxis | Beta | 0.01 | 10.41 | 800.78 |
ILI event probabilities (treatment) | |||||
23 | Probability patient with ILI presents | Beta | 0.25 | 5 | 20 |
24 | Probability patient presents within 48 hours of ILI onset | Beta | 0.16 | 104 | 668 |
25 | Probability patient given antiviral treatment | presents < 48 hours | NA | 1 | 1 | – |
26 | Probability patient receives oseltamivir | prescribed antiviral | NA | 0.89 | 0.89 | – |
27 | Probability patient receives zanamivir | prescribed antiviral | NA | 0.11 | 0.11 | – |
28 | Probability adverse events – oseltamivir treatment | Beta | 0.02 | 1.72 | 86.11 |
29 | Probability adverse events – zanamivir treatment | Beta | 0.01 | 10.41 | 800.78 |
30 | Probability complication – no treatment | Beta | 0.12 | 2166 | 17,597 |
31 | Odds ratio complication – oseltamivir treatment | Lognormal | 0.40 | –1 | 0 |
32 | Odds ratio complication – zanamivir treatment | Lognormal | 0.49 | –0.71 | 0.38 |
33 | Probability complication is respiratory | Dirichlet | 0.89 | 1942 | 2172 |
34 | Probability complication is cardiac | Dirichlet | 0.01 | 30 | 2172 |
35 | Probability complication is CNS | Dirichlet | 0.01 | 16 | 2172 |
36 | Probability complication is renal | Dirichlet | 0 | 6 | 2172 |
37 | Probability complication is otitis media | Dirichlet | 0.05 | 111 | 2172 |
38 | Probability complication is other | Dirichlet | 0.03 | 67 | 2172 |
39 | Probability respiratory complication is pneumonia | Beta | 0.03 | 62 | 1941 |
40 | Probability patient receives antibiotics | no complication | Beta | 0.42 | 19811 | 47,169 |
41 | Probability patient receives antibiotics | complication | Beta | 0.81 | 6983 | 8579 |
42 | Probability of influenza death | complication | Beta | 0.01 | 16 | 2142 |
Cost/resource parameters | |||||
43 | Cost of amantadine prophylaxis course (without vaccine) | NA | £4.80 | £4.80 | – |
44 | Cost of amantadine prophylaxis course (with vaccine) | NA | £4.80 | £4.80 | – |
45 | Cost of oseltamivir prophylaxis course | NA | £16.36 | £16.36 | – |
46 | Cost of zanamivir prophylaxis course | NA | £24.55 | £24.55 | – |
47 | Cost of oseltamivir treatment course | NA | £16.36 | £16.36 | – |
48 | Cost of zanamivir treatment course | NA | £24.55 | £24.55 | – |
49 | Days per course – amantadine prophylaxis | NA | £10 | £10 | – |
50 | Days per course – amantadine prophylaxis (prior vaccination) | NA | 10 | 10 | – |
51 | Days per course – oseltamivir prophylaxis | NA | 10 | 10 | – |
52 | Days per course – zanamivir prophylaxis | NA | 10 | 10 | – |
53 | Acquisition cost for vaccination | NA | £5.63 | £5.63 | – |
54 | Administration cost for vaccination | NA | £25 | £25 | – |
55 | Cost of attendance at GP surgery consultation | NA | £25 | £25 | – |
56 | Cost of attendance at GP home visit | NA | £69 | £69 | – |
57 | Cost of attendance at A&E | NA | £95.56 | £95.56 | – |
58 | Probability A&E attendance | patient presents (no complication) | Beta | 0.03 | 8.35 | 270.11 |
59 | Probability GP attendance | patient presents (no complication) | NA | 0.97 | 0.97 | – |
60 | Probability home GP visit | GP presentation (no complication) | Beta | 0.08 | 56 | 674 |
61 | Probability A&E attendance | patient presents (complication) | Beta | 0.03 | 8.35 | 270.11 |
62 | Probability GP attendance | patient presents (complication) | NA | 0.97 | 0.97 | – |
63 | Probability home GP visit | GP presentation (complication) | Beta | 0.08 | 56 | 674 |
64 | Cost of uncomplicated influenza presentation | NA | £30.73 | £30.73 | – |
65 | Cost of complicated influenza presentation | NA | £30.73 | £30.73 | – |
66 | Cost of antibiotics course | NA | £6.80 | £6.80 | – |
67 | Cost of anti-emetics course (metaclopramide 7-day course) | NA | £1.69 | £1.69 | – |
68 | Cost of managing adverse events – vaccination | NA | £25 | £25 | – |
69 | Cost of managing adverse events – amantadine prophylaxis | NA | £25 | £25 | – |
70 | Cost of inpatient episode | Gamma | £261.17 | £261.17 | 5.16 |
71 | Probability hospitalisation no treatment | complication | Beta | 0.16 | 15 | 95 |
72 | Probability ICU care | complication | Beta | 0.05 | 22 | 453 |
73 | Inpatient LOS (days) | Gamma | 11.90 | 16 | 1 |
74 | Cost of ITU day | Normal | £1345.39 | £1345.39 | £31.95 |
75 | ITU LOS (days) | Gamma | 28 | 11.60 | 2.41 |
76 | Expected cost of hospitalisation | NA | £4937.39 | £4937.39 | – |
HRQoL parameters | |||||
77 | 21-day QALYs for influenza case – no treatment | Beta | 0.03 | 2820 | 100,000 |
78 | 21-day QALYs for influenza case – oseltamivir treatment | Beta | 0.03 | 2977 | 100,000 |
79 | 21-day QALYs for influenza case – zanamivir treatment | Beta | 0.03 | 2977 | 100,000 |
80 | QALY loss for influenza case – no treatment | NA | 0.02 | 0.02 | – |
81 | QALY loss for influenza case – oseltamivir treatment | NA | 0.02 | 0.02 | – |
82 | QALY loss for influenza case – zanamivir treatment | NA | 0.02 | 0.02 | – |
83 | Utility decrement – adverse events | Beta | 0.20 | 200 | 1000 |
84 | Duration adverse events | Gamma | 0.01 | 25 | 0 |
85 | Utility decrement respiratory complication | Lognormal | 0.15 | –1.90 | 0.41 |
86 | Utility decrement cardiac complication | Lognormal | 0.37 | –0.99 | 0.14 |
87 | Utility decrement CNS complication | Lognormal | 0.37 | –0.99 | 0.14 |
88 | Utility decrement renal complication | Lognormal | 0.37 | –0.99 | 0.14 |
89 | Utility decrement otitis media complication | Lognormal | 0.15 | –1.90 | 0.41 |
90 | Utility decrement other complication | Lognormal | 0.37 | –0.99 | 0.14 |
91 | Duration respiratory complication (years) | Gamma | 0.03 | 12.60 | 0.85 |
92 | Duration cardiac complication (years) | Gamma | 0.03 | 12.60 | 0.85 |
93 | Duration CNS complication (years) | Gamma | 0.03 | 12.60 | 0.85 |
94 | Duration renal complication (years) | Gamma | 0.03 | 12.60 | 0.85 |
95 | Duration otitis media complication (years) | Gamma | 0.03 | 9.73 | 0.96 |
96 | Duration other complication (years) | Gamma | 0.03 | 12.60 | 0.85 |
97 | Utility general population 0–24 | Normal | 0.94 | 0.94 | 0.01 |
98 | Utility general population 25–34 | Normal | 0.93 | 0.93 | 0.01 |
99 | Utility general population 35–44 | Normal | 0.91 | 0.91 | 0.01 |
100 | Utility general population 45–54 | Normal | 0.85 | 0.85 | 0.01 |
101 | Utility general population 55–64 | Normal | 0.80 | 0.80 | 0.01 |
102 | Utility general population 65–74 | Normal | 0.78 | 0.78 | 0.01 |
103 | Utility general population > 75 | Normal | 0.73 | 0.73 | 0.02 |
104 | Percentage population female | NA | 0.51 | 0.51 | – |
105 | QALY loss for premature death | NA | 13.37 | 13.37 | – |
106 | Discount rate for QALYs | NA | 3.50% | 3.50% | – |
No. | Parameter description | Distribution | Mean | Parameter 1 | Parameter 2 |
---|---|---|---|---|---|
Baseline event probabilities (disease) | |||||
1 | Baseline attack rate for influenza | Beta | 0.09 | 180 | 2051 |
2 | Probability ILI is influenza within epidemic period | Beta | 0.50 | 622 | 1256 |
3 | Probability influenza A strain is dominant | Beta | 0.75 | 9 | 12 |
4 | Probability influenza is influenza A in influenza A dominant seasons | Beta | 0.86 | 740 | 859 |
5 | Probability influenza is influenza A in influenza B dominant years | Beta | 0.30 | 83 | 281 |
6 | Probability influenza is influenza A | NA | 0.72 | 0.72 | – |
7 | Duration of influenza epidemic (days) | Gamma | 40 | 32.65 | 1.23 |
Effectiveness parameters (prevention) | |||||
8 | RR for influenza – vaccine | Lognormal | 0.42 | –0.87 | 0.23 |
9 | RR for influenza – amantadine prophylaxis | Lognormal | 0.10 | –2.26 | 0.60 |
10 | RR for influenza – oseltamivir prophylaxis | Lognormal | 0.19 | –1.66 | 0.44 |
11 | RR for influenza – zanamivir prophylaxis | Lognormal | 0.21 | –1.56 | 0.24 |
12 | Probability of amantadine resistance | Beta | 0.37 | 73.40 | 199 |
13 | Probability influenza case occurs within epidemic | Beta | 1 | 1 | 1125 |
14 | Probability influenza case avoidable – amantadine | NA | 1 | 1 | – |
15 | Probability influenza case avoidable – amantadine (vaccination) | NA | 1 | 1 | – |
16 | Probability influenza case avoidable – oseltamivir | NA | 1 | 1 | – |
17 | Percentage of influenza cases avoidable – zanamivir | NA | 1 | 1 | – |
Adverse events/withdrawals (prophylaxis) | |||||
18 | Probability adverse event – vaccination | Beta | 0.02 | 2 | 100 |
19 | Probability adverse event – amantadine prophylaxis | Beta | 0.05 | 10 | 200 |
20 | Probability withdrawal – amantadine prophylaxis | Beta | 0.15 | 0.37 | 2.55 |
21 | Probability withdrawal – oseltamivir prophylaxis | Beta | 0.02 | 1.72 | 86.11 |
22 | Probability withdrawal – zanamivir prophylaxis | Beta | 0.01 | 10.41 | 800.78 |
ILI event probabilities (treatment) | |||||
23 | Probability patient with ILI presents | Beta | 0.25 | 5 | 20 |
24 | Probability patient presents within 48 hours of ILI onset | Beta | 0.11 | 18.5 | 164 |
25 | Probability patient given antiviral treatment | presents < 48 hours | NA | 1 | 1 | – |
26 | Probability patient receives oseltamivir | prescribed antiviral | NA | 0.89 | 0.89 | – |
27 | Probability patient receives zanamivir | prescribed antiviral | NA | 0.11 | 0.11 | – |
28 | Probability adverse events – oseltamivir treatment | Beta | 0.02 | 1.72 | 86.11 |
29 | Probability adverse events – zanamivir treatment | Beta | 0.01 | 10.41 | 800.78 |
30 | Probability complication – no treatment | Beta | 0.09 | 942 | 10,145 |
31 | Odds ratio complication – oseltamivir treatment | Lognormal | 0.40 | –1 | 0 |
32 | Odds ratio complication – zanamivir treatment | Lognormal | 0.70 | –0.36 | 0.16 |
33 | Probability complication is respiratory | Dirichlet | 0.86 | 820 | 948 |
34 | Probability complication is cardiac | Dirichlet | 0.01 | 10 | 948 |
35 | Probability complication is CNS | Dirichlet | 0.02 | 22 | 948 |
36 | Probability complication is renal | Dirichlet | 0.01 | 6 | 948 |
37 | Probability complication is otitis media | Dirichlet | 0.02 | 22 | 948 |
38 | Probability complication is other | Dirichlet | 0.07 | 68 | 948 |
39 | Probability respiratory complication is pneumonia | Beta | 0.13 | 106 | 819 |
40 | Probability patient receives antibiotics | no complication | Beta | 0.55 | 8544 | 15,620 |
41 | Probability patient receives antibiotics | complication | Beta | 0.80 | 1527 | 1916 |
42 | Probability of influenza death | complication | Beta | 0.11 | 110 | 981 |
Cost/resource parameters | |||||
43 | Cost of amantadine prophylaxis course (without vaccine) | NA | £4.80 | £4.80 | – |
44 | Cost of amantadine prophylaxis course (with vaccine) | NA | £4.80 | £4.80 | – |
45 | Cost of oseltamivir prophylaxis course | NA | £16.36 | £16.36 | – |
46 | Cost of zanamivir prophylaxis course | NA | £24.55 | £24.55 | – |
47 | Cost of oseltamivir treatment course | NA | £16.36 | £16.36 | – |
48 | Cost of zanamivir treatment course | NA | £24.55 | £24.55 | – |
49 | Days per course – amantadine prophylaxis | NA | £10 | £10 | – |
50 | Days per course – amantadine prophylaxis (prior vaccination) | NA | 10 | 10 | – |
51 | Days per course – oseltamivir prophylaxis | NA | 10 | 10 | – |
52 | Days per course – zanamivir prophylaxis | NA | 10 | 10 | – |
53 | Acquisition cost for vaccination | NA | £5.63 | £5.63 | – |
54 | Administration cost for vaccination | NA | £25 | £25 | – |
55 | Cost of attendance at GP surgery consultation | NA | £25 | £25 | – |
56 | Cost of attendance at GP home visit | NA | £69 | £69 | – |
57 | Cost of attendance at A&E | NA | £95.56 | £95.56 | – |
58 | Probability A&E attendance | patient presents (no complication) | Beta | 0.03 | 8.35 | 270.11 |
59 | Probability GP attendance | patient presents (no complication) | NA | 0.97 | 0.97 | – |
60 | Probability home GP visit | GP presentation (no complication) | Beta | 0.38 | 62 | 165 |
61 | Probability A&E attendance | patient presents (complication) | Beta | 0.03 | 8.35 | 270.11 |
62 | Probability GP attendance | patient presents (complication) | NA | 0.97 | 0.97 | – |
63 | Probability home GP visit | GP presentation (complication) | Beta | 0.38 | 62 | 165 |
64 | Cost of uncomplicated influenza presentation | NA | £43.20 | £43.20 | – |
65 | Cost of complicated influenza presentation | NA | £43.20 | £43.20 | – |
66 | Cost of antibiotics course | NA | £6.80 | £6.80 | – |
67 | Cost of anti-emetics course (metaclopramide 7-day course) | NA | £1.69 | £1.69 | – |
68 | Cost of managing adverse events – vaccination | NA | £25 | £25 | – |
69 | Cost of managing adverse events – amantadine prophylaxis | NA | £25 | £25 | – |
70 | Cost of inpatient episode | Gamma | £261.17 | £261.17 | 5.16 |
71 | Probability hospitalisation no treatment | complication | Beta | 0.16 | 15 | 95 |
72 | Probability ICU care | complication | Beta | 0.05 | 22 | 453 |
73 | Inpatient LOS (days) | Gamma | 15 | 25 | 1 |
74 | Cost of ITU day | Normal | £1345.39 | £1345.39 | £31.95 |
75 | ITU LOS (days) | Gamma | 28 | 11.60 | 2.41 |
76 | Expected cost of hospitalisation | NA | £5747.01 | £5747.01 | – |
HRQoL parameters | |||||
77 | 21-day QALYs for influenza case – no treatment | Beta | 0.03 | 2820 | 100,000 |
78 | 21-day QALYs for influenza case – oseltamivir treatment | Beta | 0.03 | 2977 | 100,000 |
79 | 21-day QALYs for influenza case – zanamivir treatment | Beta | 0.03 | 2977 | 100,000 |
80 | QALY loss for influenza case – no treatment | NA | 0.02 | 0.02 | – |
81 | QALY loss for influenza case – oseltamivir treatment | NA | 0.01 | 0.01 | – |
82 | QALY loss for influenza case – zanamivir treatment | NA | 0.01 | 0.01 | – |
83 | Utility decrement – adverse events | Beta | 0.20 | 200 | 1000 |
84 | Duration adverse events | Gamma | 0.01 | 25 | 0 |
85 | Utility decrement respiratory complication | Lognormal | 0.15 | –1.90 | 0.41 |
86 | Utility decrement cardiac complication | Lognormal | 0.37 | –0.99 | 0.14 |
87 | Utility decrement CNS complication | Lognormal | 0.37 | –0.99 | 0.14 |
88 | Utility decrement renal complication | Lognormal | 0.37 | –0.99 | 0.14 |
89 | Utility decrement otitis media complication | Lognormal | 0.15 | –1.90 | 0.41 |
90 | Utility decrement other complication | Lognormal | 0.37 | –0.99 | 0.14 |
91 | Duration respiratory complication (years) | Gamma | 0.03 | 13.15 | 0.83 |
92 | Duration cardiac complication (years) | Gamma | 0.03 | 13.15 | 0.83 |
93 | Duration CNS complication (years) | Gamma | 0.03 | 13.15 | 0.83 |
94 | Duration renal complication (years) | Gamma | 0.03 | 13.15 | 0.83 |
95 | Duration otitis media complication (years) | Gamma | 0.03 | 9.73 | 0.96 |
96 | Duration other complication (years) | Gamma | 0.03 | 13.15 | 0.83 |
97 | Utility general population 0–24 | Normal | 0.94 | 0.94 | 0.01 |
98 | Utility general population 25–34 | Normal | 0.93 | 0.93 | 0.01 |
99 | Utility general population 35–44 | Normal | 0.91 | 0.91 | 0.01 |
100 | Utility general population 45–54 | Normal | 0.85 | 0.85 | 0.01 |
101 | Utility general population 55–64 | Normal | 0.80 | 0.80 | 0.01 |
102 | Utility general population 65–74 | Normal | 0.78 | 0.78 | 0.01 |
103 | Utility general population > 75 | Normal | 0.73 | 0.73 | 0.02 |
104 | Percentage population female | NA | 0.51 | 0.51 | – |
105 | QALY loss for premature death | NA | 2.95 | 2.95 | – |
106 | Discount rate for QALYs | NA | 3.50% | 3.50% | – |
No. | Parameter description | Distribution | Mean | Parameter 1 | Parameter 2 |
---|---|---|---|---|---|
Baseline event probabilities (disease) | |||||
1 | Baseline attack rate for influenza | Beta | 0.09 | 180 | 2051 |
2 | Probability ILI is influenza within epidemic period | Beta | 0.50 | 622 | 1256 |
3 | Probability influenza A strain is dominant | Beta | 0.75 | 9 | 12 |
4 | Probability influenza is influenza A in influenza A dominant seasons | Beta | 0.86 | 740 | 859 |
5 | Probability influenza is influenza A in influenza B dominant years | Beta | 0.30 | 83 | 281 |
6 | Probability influenza is influenza A | NA | 0.72 | 0.72 | – |
7 | Duration of influenza epidemic (days) | Gamma | 40 | 32.65 | 1.23 |
Effectiveness parameters (prevention) | |||||
8 | RR for influenza – vaccine | Lognormal | 0.42 | –0.87 | 0.23 |
9 | RR for influenza – amantadine prophylaxis | Lognormal | 0.10 | –2.26 | 0.60 |
10 | RR for influenza – oseltamivir prophylaxis | Lognormal | 0.19 | –1.66 | 0.44 |
11 | RR for influenza – zanamivir prophylaxis | Lognormal | 0.21 | –1.56 | 0.24 |
12 | Probability of amantadine resistance | Beta | 0.37 | 73.40 | 199 |
13 | Probability influenza case occurs within epidemic | Beta | 1 | 1 | 1125 |
14 | Probability influenza case avoidable – amantadine | NA | 1 | 1 | – |
15 | Probability influenza case avoidable – amantadine (vaccination) | NA | 1 | 1 | – |
16 | Probability influenza case avoidable – oseltamivir | NA | 1 | 1 | – |
17 | Percentage of influenza cases avoidable – zanamivir | NA | 1 | 1 | – |
Adverse events/withdrawals (prophylaxis) | |||||
18 | Probability adverse event – vaccination | Beta | 0.02 | 2 | 100 |
19 | Probability adverse event – amantadine prophylaxis | Beta | 0.05 | 10 | 200 |
20 | Probability withdrawal – amantadine prophylaxis | Beta | 0.15 | 0.37 | 2.55 |
21 | Probability withdrawal – oseltamivir prophylaxis | Beta | 0.02 | 1.72 | 86.11 |
22 | Probability withdrawal – zanamivir prophylaxis | Beta | 0.01 | 10.41 | 800.78 |
ILI event probabilities (treatment) | |||||
23 | Probability patient with ILI presents | Beta | 0.25 | 5 | 20 |
24 | Probability patient presents within 48 hours of ILI onset | Beta | 0.11 | 18.5 | 164 |
25 | Probability patient given antiviral treatment | presents < 48 hours | NA | 1 | 1 | – |
26 | Probability patient receives oseltamivir | prescribed antiviral | NA | 0.89 | 0.89 | – |
27 | Probability patient receives zanamivir | prescribed antiviral | NA | 0.11 | 0.11 | – |
28 | Probability adverse events – oseltamivir treatment | Beta | 0.02 | 1.72 | 86.11 |
29 | Probability adverse events – zanamivir treatment | Beta | 0.01 | 10.41 | 800.78 |
30 | Probability complication – no treatment | Beta | 0.12 | 908 | 7407 |
31 | Odds ratio complication – oseltamivir treatment | Lognormal | 0.40 | –1 | 0 |
32 | Odds ratio complication – zanamivir treatment | Lognormal | 0.49 | –0.71 | 0.38 |
33 | Probability complication is respiratory | Dirichlet | 0.83 | 755 | 914 |
34 | Probability complication is cardiac | Dirichlet | 0.07 | 60 | 914 |
35 | Probability complication is CNS | Dirichlet | 0.03 | 24 | 914 |
36 | Probability complication is renal | Dirichlet | 0.01 | 13 | 914 |
37 | Probability complication is otitis media | Dirichlet | 0.01 | 12 | 914 |
38 | Probability complication is other | Dirichlet | 0.05 | 50 | 914 |
39 | Probability respiratory complication is pneumonia | Beta | 0.13 | 97 | 754 |
40 | Probability patient receives antibiotics | no complication | Beta | 0.55 | 8544 | 15,620 |
41 | Probability patient receives antibiotics | complication | Beta | 0.80 | 1527 | 1916 |
42 | Probability of influenza death | complication | Beta | 0.12 | 114 | 936 |
Cost/resource parameters | |||||
43 | Cost of amantadine prophylaxis course (without vaccine) | NA | £4.80 | £4.80 | – |
44 | Cost of amantadine prophylaxis course (with vaccine) | NA | £4.80 | £4.80 | – |
45 | Cost of oseltamivir prophylaxis course | NA | £16.36 | £16.36 | – |
46 | Cost of zanamivir prophylaxis course | NA | £24.55 | £24.55 | – |
47 | Cost of oseltamivir treatment course | NA | £16.36 | £16.36 | – |
48 | Cost of zanamivir treatment course | NA | £24.55 | £24.55 | – |
49 | Days per course – amantadine prophylaxis | NA | £10 | £10 | – |
50 | Days per course – amantadine prophylaxis (prior vac) | NA | 10 | 10 | – |
51 | Days per course – oseltamivir prophylaxis | NA | 10 | 10 | – |
52 | Days per course – zanamivir prophylaxis | NA | 10 | 10 | – |
53 | Acquisition cost for vaccination | NA | £5.63 | £5.63 | – |
54 | Administration cost for vaccination | NA | £25 | £25 | – |
55 | Cost of attendance at GP surgery consultation | NA | £25 | £25 | – |
56 | Cost of attendance at GP home visit | NA | £69 | £69 | – |
57 | Cost of attendance at A&E | NA | £95.56 | £95.56 | – |
58 | Probability A&E attendance | patient presents (no complication) | Beta | 0.03 | 8.35 | 270.11 |
59 | Probability GP attendance | patient presents (no complication) | NA | 0.97 | 0.97 | – |
60 | Probability home GP visit | GP presentation (no complication) | Beta | 0.38 | 62 | 165 |
61 | Probability A&E attendance | patient presents (complication) | Beta | 0.03 | 8.35 | 270.11 |
62 | Probability GP attendance | patient presents (complication) | NA | 0.97 | 0.97 | – |
63 | Probability home GP visit | GP presentation (complication) | Beta | 0.38 | 62 | 165 |
64 | Cost of uncomplicated influenza presentation | NA | £43.20 | £43.20 | – |
65 | Cost of complicated influenza presentation | NA | £43.20 | £43.20 | – |
66 | Cost of antibiotics course | NA | £6.80 | £6.80 | – |
67 | Cost of anti-emetics course (metaclopramide 7 day course) | NA | £1.69 | £1.69 | – |
68 | Cost of managing adverse events – vaccination | NA | £25 | £25 | – |
69 | Cost of managing adverse events – amantadine prophylaxis | NA | £25 | £25 | – |
70 | Cost of inpatient episode | Gamma | £261.17 | £261.17 | 5.16 |
71 | Probability hospitalisation no treatment | complication | Beta | 0.16 | 15 | 95 |
72 | Probability ICU care | complication | Beta | 0.05 | 22 | 453 |
73 | Inpatient LOS (days) | Gamma | 15 | 25 | 1 |
74 | Cost of ITU day | Normal | £1345.39 | £1345.39 | £31.95 |
75 | ITU LOS (days) | Gamma | 28 | 11.60 | 2.41 |
76 | Expected cost of hospitalisation | NA | £5747.01 | £5747.01 | – |
HRQoL parameters | |||||
77 | 21-day QALYs for influenza case – no treatment | Beta | 0.03 | 2820 | 100,000 |
78 | 21-day QALYs for influenza case – oseltamivir treatment | Beta | 0.03 | 2977 | 100,000 |
79 | 21-day QALYs for influenza case – zanamivir treatment | Beta | 0.03 | 2977 | 100,000 |
80 | QALY loss for influenza case – no treatment | NA | 0.02 | 0.02 | – |
81 | QALY loss for influenza case – oseltamivir treatment | NA | 0.01 | 0.01 | – |
82 | QALY loss for influenza case – zanamivir treatment | NA | 0.01 | 0.01 | – |
83 | Utility decrement – adverse events | Beta | 0.20 | 200 | 1000 |
84 | Duration adverse events | Gamma | 0.01 | 25 | 0 |
85 | Utility decrement respiratory complication | Lognormal | 0.15 | –1.90 | 0.41 |
86 | Utility decrement cardiac complication | Lognormal | 0.37 | –0.99 | 0.14 |
87 | Utility decrement CNS complication | Lognormal | 0.37 | –0.99 | 0.14 |
88 | Utility decrement renal complication | Lognormal | 0.37 | –0.99 | 0.14 |
89 | Utility decrement otitis media complication | Lognormal | 0.15 | –1.90 | 0.41 |
90 | Utility decrement other complication | Lognormal | 0.37 | –0.99 | 0.14 |
91 | Duration respiratory complication (years) | Gamma | 0.03 | 13.13 | 0.83 |
92 | Duration cardiac complication (years) | Gamma | 0.03 | 13.13 | 0.83 |
93 | Duration CNS complication (years) | Gamma | 0.03 | 13.13 | 0.83 |
94 | Duration renal complication (years) | Gamma | 0.03 | 13.13 | 0.83 |
95 | Duration otitis media complication (years) | Gamma | 0.03 | 9.73 | 0.96 |
96 | Duration other complication (years) | Gamma | 0.03 | 13.13 | 0.83 |
97 | Utility general population 0–24 | Normal | 0.94 | 0.94 | 0.01 |
98 | Utility general population 25–34 | Normal | 0.93 | 0.93 | 0.01 |
99 | Utility general population 35–44 | Normal | 0.91 | 0.91 | 0.01 |
100 | Utility general population 45–54 | Normal | 0.85 | 0.85 | 0.01 |
101 | Utility general population 55–64 | Normal | 0.80 | 0.80 | 0.01 |
102 | Utility general population 65–74 | Normal | 0.78 | 0.78 | 0.01 |
103 | Utility general population > 75 | Normal | 0.73 | 0.73 | 0.02 |
104 | Percentage population female | NA | 0.51 | 0.51 | – |
105 | QALY loss for premature death | NA | 2.95 | 2.95 | – |
106 | Discount rate for QALYs | NA | 3.50% | 3.50% | – |
Appendix 8 Cost-effectiveness acceptability curves (base-case analysis)
Cost-effectiveness acceptability curves for seasonal prophylaxis
Appendix 9 Cost-effectiveness acceptability curves (incorporating proposed price reduction for zanamivir)
Cost-effectiveness acceptability curves for seasonal prophylaxis
Cost-effectiveness acceptability curves for post-exposure prophylaxis
Glossary
- Attack rate
- A cumulative incidence rate in a population over time, such as in the circumstances of an epidemic
- Dominated (simple)
- Where a given treatment alternative is less effective and more expensive than its comparator
- Dominated (extended)
- The state when a strategy under study is both less effective and more costly than a linear combination of two other strategies with which it is mutually exclusive
- Meta-analysis
- A statistical method by which the results of a number of studies are pooled to give a combined summary statistic
- Post-exposure prophylaxis
- Prophylaxis initiated in response to close contact of an individual with another suspected as suffering from influenza; treatment typically lasts 7–10 days following presumed exposure
- Protective efficacy
- 1 minus the RR value, expressed as a percentage
- Relative risk (RR)
- Ratio of the probability of an event occurring in an exposed group relative to a non-exposed or control group
- Seasonal prophylaxis
- Prophylaxis initiated in response to known circulation of influenza within the community; treatment typically lasts for 6 weeks
- Symptomatic, laboratory-confirmed influenza
- Cases of influenza in which illness is clinically confirmed according to presence of symptoms indicative of influenza and with evidence of infection by the influenza virus, as determined by laboratory methods
List of abbreviations
- A&E
- accident and emergency
- ARI
- acute respiratory illness
- BNF
- British National Formulary
- CEAC
- cost-effectiveness acceptability curve
- CI
- confidence interval
- CNS
- central nervous system
- COPD
- chronic obstructive pulmonary disease
- EISS
- European Influenza Surveillance Scheme
- EQ-5D
- EuroQol-5D
- GI
- gastrointestinal
- GP
- general practitioner
- GPRD
- General Practice Research Database
- GSK
- GlaxoSmithKline
- HAI
- haemagglutination inhibition assay
- HPA
- Health Protection Agency
- HRG
- Health-care Resource Group
- HRQoL
- health-related quality of life
- HTA
- Health Technology Assessment
- HUI
- Health Utilities Index
- ICER
- incremental cost-effectiveness ratio
- ICU
- intensive care unit
- ILI
- influenza-like illness
- ITT
- intention-to-treat
- ITU
- intensive therapy unit
- MVH
- Measurement and Valuation of Health
- NAMCS
- National Ambulatory Medical Care Survey
- NI
- neuraminidase inhibitor
- NICE
- National Institute for Health and Clinical Excellence
- ONS
- Office for National Statistics
- PCR
- polymerase chain reaction
- PE
- protective efficacy
- PSSRU
- Personal Social Services Research Unit
- Px
- prophylaxis
- QALY
- quality-adjusted life-year
- QUOROM
- quality of reporting of meta-analyses
- RCGP
- Royal College of General Practitioners
- RCT
- randomised controlled trial
- RR
- relative risk
- RSV
- respiratory syncytial virus
- SAVE
- simulating anti-influenza value and effectiveness
- SE
- standard error
- SLCI
- symptomatic, laboratory-confirmed influenza
- SPC
- Summary of Product Characteristics
- TTO
- time trade-off
- VAS
- visual analogue scale
- WHO
- World Health Organization
All abbreviations that have been used in this report are listed here unless the abbreviation is well known (e.g. NHS), or it has been used only once, or it is a non-standard abbreviation used only in figures/tables/appendices, in which case the abbreviation is defined in the figure legend or in the notes at the end of the table.
Notes
Health Technology Assessment reports published to date
-
Home parenteral nutrition: a systematic review.
By Richards DM, Deeks JJ, Sheldon TA, Shaffer JL.
-
Diagnosis, management and screening of early localised prostate cancer.
A review by Selley S, Donovan J, Faulkner A, Coast J, Gillatt D.
-
The diagnosis, management, treatment and costs of prostate cancer in England and Wales.
A review by Chamberlain J, Melia J, Moss S, Brown J.
-
Screening for fragile X syndrome.
A review by Murray J, Cuckle H, Taylor G, Hewison J.
-
A review of near patient testing in primary care.
By Hobbs FDR, Delaney BC, Fitzmaurice DA, Wilson S, Hyde CJ, Thorpe GH, et al.
-
Systematic review of outpatient services for chronic pain control.
By McQuay HJ, Moore RA, Eccleston C, Morley S, de C Williams AC.
-
Neonatal screening for inborn errors of metabolism: cost, yield and outcome.
A review by Pollitt RJ, Green A, McCabe CJ, Booth A, Cooper NJ, Leonard JV, et al.
-
Preschool vision screening.
A review by Snowdon SK, Stewart-Brown SL.
-
Implications of socio-cultural contexts for the ethics of clinical trials.
A review by Ashcroft RE, Chadwick DW, Clark SRL, Edwards RHT, Frith L, Hutton JL.
-
A critical review of the role of neonatal hearing screening in the detection of congenital hearing impairment.
By Davis A, Bamford J, Wilson I, Ramkalawan T, Forshaw M, Wright S.
-
Newborn screening for inborn errors of metabolism: a systematic review.
By Seymour CA, Thomason MJ, Chalmers RA, Addison GM, Bain MD, Cockburn F, et al.
-
Routine preoperative testing: a systematic review of the evidence.
By Munro J, Booth A, Nicholl J.
-
Systematic review of the effectiveness of laxatives in the elderly.
By Petticrew M, Watt I, Sheldon T.
-
When and how to assess fast-changing technologies: a comparative study of medical applications of four generic technologies.
A review by Mowatt G, Bower DJ, Brebner JA, Cairns JA, Grant AM, McKee L.
-
Antenatal screening for Down’s syndrome.
A review by Wald NJ, Kennard A, Hackshaw A, McGuire A.
-
Screening for ovarian cancer: a systematic review.
By Bell R, Petticrew M, Luengo S, Sheldon TA.
-
Consensus development methods, and their use in clinical guideline development.
A review by Murphy MK, Black NA, Lamping DL, McKee CM, Sanderson CFB, Askham J, et al.
-
A cost–utility analysis of interferon beta for multiple sclerosis.
By Parkin D, McNamee P, Jacoby A, Miller P, Thomas S, Bates D.
-
Effectiveness and efficiency of methods of dialysis therapy for end-stage renal disease: systematic reviews.
By MacLeod A, Grant A, Donaldson C, Khan I, Campbell M, Daly C, et al.
-
Effectiveness of hip prostheses in primary total hip replacement: a critical review of evidence and an economic model.
By Faulkner A, Kennedy LG, Baxter K, Donovan J, Wilkinson M, Bevan G.
-
Antimicrobial prophylaxis in colorectal surgery: a systematic review of randomised controlled trials.
By Song F, Glenny AM.
-
Bone marrow and peripheral blood stem cell transplantation for malignancy.
A review by Johnson PWM, Simnett SJ, Sweetenham JW, Morgan GJ, Stewart LA.
-
Screening for speech and language delay: a systematic review of the literature.
By Law J, Boyle J, Harris F, Harkness A, Nye C.
-
Resource allocation for chronic stable angina: a systematic review of effectiveness, costs and cost-effectiveness of alternative interventions.
By Sculpher MJ, Petticrew M, Kelland JL, Elliott RA, Holdright DR, Buxton MJ.
-
Detection, adherence and control of hypertension for the prevention of stroke: a systematic review.
By Ebrahim S.
-
Postoperative analgesia and vomiting, with special reference to day-case surgery: a systematic review.
By McQuay HJ, Moore RA.
-
Choosing between randomised and nonrandomised studies: a systematic review.
By Britton A, McKee M, Black N, McPherson K, Sanderson C, Bain C.
-
Evaluating patient-based outcome measures for use in clinical trials.
A review by Fitzpatrick R, Davey C, Buxton MJ, Jones DR.
-
Ethical issues in the design and conduct of randomised controlled trials.
A review by Edwards SJL, Lilford RJ, Braunholtz DA, Jackson JC, Hewison J, Thornton J.
-
Qualitative research methods in health technology assessment: a review of the literature.
By Murphy E, Dingwall R, Greatbatch D, Parker S, Watson P.
-
The costs and benefits of paramedic skills in pre-hospital trauma care.
By Nicholl J, Hughes S, Dixon S, Turner J, Yates D.
-
Systematic review of endoscopic ultrasound in gastro-oesophageal cancer.
By Harris KM, Kelly S, Berry E, Hutton J, Roderick P, Cullingworth J, et al.
-
Systematic reviews of trials and other studies.
By Sutton AJ, Abrams KR, Jones DR, Sheldon TA, Song F.
-
Primary total hip replacement surgery: a systematic review of outcomes and modelling of cost-effectiveness associated with different prostheses.
A review by Fitzpatrick R, Shortall E, Sculpher M, Murray D, Morris R, Lodge M, et al.
-
Informed decision making: an annotated bibliography and systematic review.
By Bekker H, Thornton JG, Airey CM, Connelly JB, Hewison J, Robinson MB, et al.
-
Handling uncertainty when performing economic evaluation of healthcare interventions.
A review by Briggs AH, Gray AM.
-
The role of expectancies in the placebo effect and their use in the delivery of health care: a systematic review.
By Crow R, Gage H, Hampson S, Hart J, Kimber A, Thomas H.
-
A randomised controlled trial of different approaches to universal antenatal HIV testing: uptake and acceptability. Annex: Antenatal HIV testing – assessment of a routine voluntary approach.
By Simpson WM, Johnstone FD, Boyd FM, Goldberg DJ, Hart GJ, Gormley SM, et al.
-
Methods for evaluating area-wide and organisation-based interventions in health and health care: a systematic review.
By Ukoumunne OC, Gulliford MC, Chinn S, Sterne JAC, Burney PGJ.
-
Assessing the costs of healthcare technologies in clinical trials.
A review by Johnston K, Buxton MJ, Jones DR, Fitzpatrick R.
-
Cooperatives and their primary care emergency centres: organisation and impact.
By Hallam L, Henthorne K.
-
Screening for cystic fibrosis.
A review by Murray J, Cuckle H, Taylor G, Littlewood J, Hewison J.
-
A review of the use of health status measures in economic evaluation.
By Brazier J, Deverill M, Green C, Harper R, Booth A.
-
Methods for the analysis of quality-of-life and survival data in health technology assessment.
A review by Billingham LJ, Abrams KR, Jones DR.
-
Antenatal and neonatal haemoglobinopathy screening in the UK: review and economic analysis.
By Zeuner D, Ades AE, Karnon J, Brown J, Dezateux C, Anionwu EN.
-
Assessing the quality of reports of randomised trials: implications for the conduct of meta-analyses.
A review by Moher D, Cook DJ, Jadad AR, Tugwell P, Moher M, Jones A, et al.
-
‘Early warning systems’ for identifying new healthcare technologies.
By Robert G, Stevens A, Gabbay J.
-
A systematic review of the role of human papillomavirus testing within a cervical screening programme.
By Cuzick J, Sasieni P, Davies P, Adams J, Normand C, Frater A, et al.
-
Near patient testing in diabetes clinics: appraising the costs and outcomes.
By Grieve R, Beech R, Vincent J, Mazurkiewicz J.
-
Positron emission tomography: establishing priorities for health technology assessment.
A review by Robert G, Milne R.
-
The debridement of chronic wounds: a systematic review.
By Bradley M, Cullum N, Sheldon T.
-
Systematic reviews of wound care management: (2) Dressings and topical agents used in the healing of chronic wounds.
By Bradley M, Cullum N, Nelson EA, Petticrew M, Sheldon T, Torgerson D.
-
A systematic literature review of spiral and electron beam computed tomography: with particular reference to clinical applications in hepatic lesions, pulmonary embolus and coronary artery disease.
By Berry E, Kelly S, Hutton J, Harris KM, Roderick P, Boyce JC, et al.
-
What role for statins? A review and economic model.
By Ebrahim S, Davey Smith G, McCabe C, Payne N, Pickin M, Sheldon TA, et al.
-
Factors that limit the quality, number and progress of randomised controlled trials.
A review by Prescott RJ, Counsell CE, Gillespie WJ, Grant AM, Russell IT, Kiauka S, et al.
-
Antimicrobial prophylaxis in total hip replacement: a systematic review.
By Glenny AM, Song F.
-
Health promoting schools and health promotion in schools: two systematic reviews.
By Lister-Sharp D, Chapman S, Stewart-Brown S, Sowden A.
-
Economic evaluation of a primary care-based education programme for patients with osteoarthritis of the knee.
A review by Lord J, Victor C, Littlejohns P, Ross FM, Axford JS.
-
The estimation of marginal time preference in a UK-wide sample (TEMPUS) project.
A review by Cairns JA, van der Pol MM.
-
Geriatric rehabilitation following fractures in older people: a systematic review.
By Cameron I, Crotty M, Currie C, Finnegan T, Gillespie L, Gillespie W, et al.
-
Screening for sickle cell disease and thalassaemia: a systematic review with supplementary research.
By Davies SC, Cronin E, Gill M, Greengross P, Hickman M, Normand C.
-
Community provision of hearing aids and related audiology services.
A review by Reeves DJ, Alborz A, Hickson FS, Bamford JM.
-
False-negative results in screening programmes: systematic review of impact and implications.
By Petticrew MP, Sowden AJ, Lister-Sharp D, Wright K.
-
Costs and benefits of community postnatal support workers: a randomised controlled trial.
By Morrell CJ, Spiby H, Stewart P, Walters S, Morgan A.
-
Implantable contraceptives (subdermal implants and hormonally impregnated intrauterine systems) versus other forms of reversible contraceptives: two systematic reviews to assess relative effectiveness, acceptability, tolerability and cost-effectiveness.
By French RS, Cowan FM, Mansour DJA, Morris S, Procter T, Hughes D, et al.
-
An introduction to statistical methods for health technology assessment.
A review by White SJ, Ashby D, Brown PJ.
-
Disease-modifying drugs for multiple sclerosis: a rapid and systematic review.
By Clegg A, Bryant J, Milne R.
-
Publication and related biases.
A review by Song F, Eastwood AJ, Gilbody S, Duley L, Sutton AJ.
-
Cost and outcome implications of the organisation of vascular services.
By Michaels J, Brazier J, Palfreyman S, Shackley P, Slack R.
-
Monitoring blood glucose control in diabetes mellitus: a systematic review.
By Coster S, Gulliford MC, Seed PT, Powrie JK, Swaminathan R.
-
The effectiveness of domiciliary health visiting: a systematic review of international studies and a selective review of the British literature.
By Elkan R, Kendrick D, Hewitt M, Robinson JJA, Tolley K, Blair M, et al.
-
The determinants of screening uptake and interventions for increasing uptake: a systematic review.
By Jepson R, Clegg A, Forbes C, Lewis R, Sowden A, Kleijnen J.
-
The effectiveness and cost-effectiveness of prophylactic removal of wisdom teeth.
A rapid review by Song F, O’Meara S, Wilson P, Golder S, Kleijnen J.
-
Ultrasound screening in pregnancy: a systematic review of the clinical effectiveness, cost-effectiveness and women’s views.
By Bricker L, Garcia J, Henderson J, Mugford M, Neilson J, Roberts T, et al.
-
A rapid and systematic review of the effectiveness and cost-effectiveness of the taxanes used in the treatment of advanced breast and ovarian cancer.
By Lister-Sharp D, McDonagh MS, Khan KS, Kleijnen J.
-
Liquid-based cytology in cervical screening: a rapid and systematic review.
By Payne N, Chilcott J, McGoogan E.
-
Randomised controlled trial of non-directive counselling, cognitive–behaviour therapy and usual general practitioner care in the management of depression as well as mixed anxiety and depression in primary care.
By King M, Sibbald B, Ward E, Bower P, Lloyd M, Gabbay M, et al.
-
Routine referral for radiography of patients presenting with low back pain: is patients’ outcome influenced by GPs’ referral for plain radiography?
By Kerry S, Hilton S, Patel S, Dundas D, Rink E, Lord J.
-
Systematic reviews of wound care management: (3) antimicrobial agents for chronic wounds; (4) diabetic foot ulceration.
By O’Meara S, Cullum N, Majid M, Sheldon T.
-
Using routine data to complement and enhance the results of randomised controlled trials.
By Lewsey JD, Leyland AH, Murray GD, Boddy FA.
-
Coronary artery stents in the treatment of ischaemic heart disease: a rapid and systematic review.
By Meads C, Cummins C, Jolly K, Stevens A, Burls A, Hyde C.
-
Outcome measures for adult critical care: a systematic review.
By Hayes JA, Black NA, Jenkinson C, Young JD, Rowan KM, Daly K, et al.
-
A systematic review to evaluate the effectiveness of interventions to promote the initiation of breastfeeding.
By Fairbank L, O’Meara S, Renfrew MJ, Woolridge M, Sowden AJ, Lister-Sharp D.
-
Implantable cardioverter defibrillators: arrhythmias. A rapid and systematic review.
By Parkes J, Bryant J, Milne R.
-
Treatments for fatigue in multiple sclerosis: a rapid and systematic review.
By Brañas P, Jordan R, Fry-Smith A, Burls A, Hyde C.
-
Early asthma prophylaxis, natural history, skeletal development and economy (EASE): a pilot randomised controlled trial.
By Baxter-Jones ADG, Helms PJ, Russell G, Grant A, Ross S, Cairns JA, et al.
-
Screening for hypercholesterolaemia versus case finding for familial hypercholesterolaemia: a systematic review and cost-effectiveness analysis.
By Marks D, Wonderling D, Thorogood M, Lambert H, Humphries SE, Neil HAW.
-
A rapid and systematic review of the clinical effectiveness and cost-effectiveness of glycoprotein IIb/IIIa antagonists in the medical management of unstable angina.
By McDonagh MS, Bachmann LM, Golder S, Kleijnen J, ter Riet G.
-
A randomised controlled trial of prehospital intravenous fluid replacement therapy in serious trauma.
By Turner J, Nicholl J, Webber L, Cox H, Dixon S, Yates D.
-
Intrathecal pumps for giving opioids in chronic pain: a systematic review.
By Williams JE, Louw G, Towlerton G.
-
Combination therapy (interferon alfa and ribavirin) in the treatment of chronic hepatitis C: a rapid and systematic review.
By Shepherd J, Waugh N, Hewitson P.
-
A systematic review of comparisons of effect sizes derived from randomised and non-randomised studies.
By MacLehose RR, Reeves BC, Harvey IM, Sheldon TA, Russell IT, Black AMS.
-
Intravascular ultrasound-guided interventions in coronary artery disease: a systematic literature review, with decision-analytic modelling, of outcomes and cost-effectiveness.
By Berry E, Kelly S, Hutton J, Lindsay HSJ, Blaxill JM, Evans JA, et al.
-
A randomised controlled trial to evaluate the effectiveness and cost-effectiveness of counselling patients with chronic depression.
By Simpson S, Corney R, Fitzgerald P, Beecham J.
-
Systematic review of treatments for atopic eczema.
By Hoare C, Li Wan Po A, Williams H.
-
Bayesian methods in health technology assessment: a review.
By Spiegelhalter DJ, Myles JP, Jones DR, Abrams KR.
-
The management of dyspepsia: a systematic review.
By Delaney B, Moayyedi P, Deeks J, Innes M, Soo S, Barton P, et al.
-
A systematic review of treatments for severe psoriasis.
By Griffiths CEM, Clark CM, Chalmers RJG, Li Wan Po A, Williams HC.
-
Clinical and cost-effectiveness of donepezil, rivastigmine and galantamine for Alzheimer’s disease: a rapid and systematic review.
By Clegg A, Bryant J, Nicholson T, McIntyre L, De Broe S, Gerard K, et al.
-
The clinical effectiveness and cost-effectiveness of riluzole for motor neurone disease: a rapid and systematic review.
By Stewart A, Sandercock J, Bryan S, Hyde C, Barton PM, Fry-Smith A, et al.
-
Equity and the economic evaluation of healthcare.
By Sassi F, Archard L, Le Grand J.
-
Quality-of-life measures in chronic diseases of childhood.
By Eiser C, Morse R.
-
Eliciting public preferences for healthcare: a systematic review of techniques.
By Ryan M, Scott DA, Reeves C, Bate A, van Teijlingen ER, Russell EM, et al.
-
General health status measures for people with cognitive impairment: learning disability and acquired brain injury.
By Riemsma RP, Forbes CA, Glanville JM, Eastwood AJ, Kleijnen J.
-
An assessment of screening strategies for fragile X syndrome in the UK.
By Pembrey ME, Barnicoat AJ, Carmichael B, Bobrow M, Turner G.
-
Issues in methodological research: perspectives from researchers and commissioners.
By Lilford RJ, Richardson A, Stevens A, Fitzpatrick R, Edwards S, Rock F, et al.
-
Systematic reviews of wound care management: (5) beds; (6) compression; (7) laser therapy, therapeutic ultrasound, electrotherapy and electromagnetic therapy.
By Cullum N, Nelson EA, Flemming K, Sheldon T.
-
Effects of educational and psychosocial interventions for adolescents with diabetes mellitus: a systematic review.
By Hampson SE, Skinner TC, Hart J, Storey L, Gage H, Foxcroft D, et al.
-
Effectiveness of autologous chondrocyte transplantation for hyaline cartilage defects in knees: a rapid and systematic review.
By Jobanputra P, Parry D, Fry-Smith A, Burls A.
-
Statistical assessment of the learning curves of health technologies.
By Ramsay CR, Grant AM, Wallace SA, Garthwaite PH, Monk AF, Russell IT.
-
The effectiveness and cost-effectiveness of temozolomide for the treatment of recurrent malignant glioma: a rapid and systematic review.
By Dinnes J, Cave C, Huang S, Major K, Milne R.
-
A rapid and systematic review of the clinical effectiveness and cost-effectiveness of debriding agents in treating surgical wounds healing by secondary intention.
By Lewis R, Whiting P, ter Riet G, O’Meara S, Glanville J.
-
Home treatment for mental health problems: a systematic review.
By Burns T, Knapp M, Catty J, Healey A, Henderson J, Watt H, et al.
-
How to develop cost-conscious guidelines.
By Eccles M, Mason J.
-
The role of specialist nurses in multiple sclerosis: a rapid and systematic review.
By De Broe S, Christopher F, Waugh N.
-
A rapid and systematic review of the clinical effectiveness and cost-effectiveness of orlistat in the management of obesity.
By O’Meara S, Riemsma R, Shirran L, Mather L, ter Riet G.
-
The clinical effectiveness and cost-effectiveness of pioglitazone for type 2 diabetes mellitus: a rapid and systematic review.
By Chilcott J, Wight J, Lloyd Jones M, Tappenden P.
-
Extended scope of nursing practice: a multicentre randomised controlled trial of appropriately trained nurses and preregistration house officers in preoperative assessment in elective general surgery.
By Kinley H, Czoski-Murray C, George S, McCabe C, Primrose J, Reilly C, et al.
-
Systematic reviews of the effectiveness of day care for people with severe mental disorders: (1) Acute day hospital versus admission; (2) Vocational rehabilitation; (3) Day hospital versus outpatient care.
By Marshall M, Crowther R, Almaraz- Serrano A, Creed F, Sledge W, Kluiter H, et al.
-
The measurement and monitoring of surgical adverse events.
By Bruce J, Russell EM, Mollison J, Krukowski ZH.
-
Action research: a systematic review and guidance for assessment.
By Waterman H, Tillen D, Dickson R, de Koning K.
-
A rapid and systematic review of the clinical effectiveness and cost-effectiveness of gemcitabine for the treatment of pancreatic cancer.
By Ward S, Morris E, Bansback N, Calvert N, Crellin A, Forman D, et al.
-
A rapid and systematic review of the evidence for the clinical effectiveness and cost-effectiveness of irinotecan, oxaliplatin and raltitrexed for the treatment of advanced colorectal cancer.
By Lloyd Jones M, Hummel S, Bansback N, Orr B, Seymour M.
-
Comparison of the effectiveness of inhaler devices in asthma and chronic obstructive airways disease: a systematic review of the literature.
By Brocklebank D, Ram F, Wright J, Barry P, Cates C, Davies L, et al.
-
The cost-effectiveness of magnetic resonance imaging for investigation of the knee joint.
By Bryan S, Weatherburn G, Bungay H, Hatrick C, Salas C, Parry D, et al.
-
A rapid and systematic review of the clinical effectiveness and cost-effectiveness of topotecan for ovarian cancer.
By Forbes C, Shirran L, Bagnall A-M, Duffy S, ter Riet G.
-
Superseded by a report published in a later volume.
-
The role of radiography in primary care patients with low back pain of at least 6 weeks duration: a randomised (unblinded) controlled trial.
By Kendrick D, Fielding K, Bentley E, Miller P, Kerslake R, Pringle M.
-
Design and use of questionnaires: a review of best practice applicable to surveys of health service staff and patients.
By McColl E, Jacoby A, Thomas L, Soutter J, Bamford C, Steen N, et al.
-
A rapid and systematic review of the clinical effectiveness and cost-effectiveness of paclitaxel, docetaxel, gemcitabine and vinorelbine in non-small-cell lung cancer.
By Clegg A, Scott DA, Sidhu M, Hewitson P, Waugh N.
-
Subgroup analyses in randomised controlled trials: quantifying the risks of false-positives and false-negatives.
By Brookes ST, Whitley E, Peters TJ, Mulheran PA, Egger M, Davey Smith G.
-
Depot antipsychotic medication in the treatment of patients with schizophrenia: (1) Meta-review; (2) Patient and nurse attitudes.
By David AS, Adams C.
-
A systematic review of controlled trials of the effectiveness and cost-effectiveness of brief psychological treatments for depression.
By Churchill R, Hunot V, Corney R, Knapp M, McGuire H, Tylee A, et al.
-
Cost analysis of child health surveillance.
By Sanderson D, Wright D, Acton C, Duree D.
-
A study of the methods used to select review criteria for clinical audit.
By Hearnshaw H, Harker R, Cheater F, Baker R, Grimshaw G.
-
Fludarabine as second-line therapy for B cell chronic lymphocytic leukaemia: a technology assessment.
By Hyde C, Wake B, Bryan S, Barton P, Fry-Smith A, Davenport C, et al.
-
Rituximab as third-line treatment for refractory or recurrent Stage III or IV follicular non-Hodgkin’s lymphoma: a systematic review and economic evaluation.
By Wake B, Hyde C, Bryan S, Barton P, Song F, Fry-Smith A, et al.
-
A systematic review of discharge arrangements for older people.
By Parker SG, Peet SM, McPherson A, Cannaby AM, Baker R, Wilson A, et al.
-
The clinical effectiveness and cost-effectiveness of inhaler devices used in the routine management of chronic asthma in older children: a systematic review and economic evaluation.
By Peters J, Stevenson M, Beverley C, Lim J, Smith S.
-
The clinical effectiveness and cost-effectiveness of sibutramine in the management of obesity: a technology assessment.
By O’Meara S, Riemsma R, Shirran L, Mather L, ter Riet G.
-
The cost-effectiveness of magnetic resonance angiography for carotid artery stenosis and peripheral vascular disease: a systematic review.
By Berry E, Kelly S, Westwood ME, Davies LM, Gough MJ, Bamford JM, et al.
-
Promoting physical activity in South Asian Muslim women through ‘exercise on prescription’.
By Carroll B, Ali N, Azam N.
-
Zanamivir for the treatment of influenza in adults: a systematic review and economic evaluation.
By Burls A, Clark W, Stewart T, Preston C, Bryan S, Jefferson T, et al.
-
A review of the natural history and epidemiology of multiple sclerosis: implications for resource allocation and health economic models.
By Richards RG, Sampson FC, Beard SM, Tappenden P.
-
Screening for gestational diabetes: a systematic review and economic evaluation.
By Scott DA, Loveman E, McIntyre L, Waugh N.
-
The clinical effectiveness and cost-effectiveness of surgery for people with morbid obesity: a systematic review and economic evaluation.
By Clegg AJ, Colquitt J, Sidhu MK, Royle P, Loveman E, Walker A.
-
The clinical effectiveness of trastuzumab for breast cancer: a systematic review.
By Lewis R, Bagnall A-M, Forbes C, Shirran E, Duffy S, Kleijnen J, et al.
-
The clinical effectiveness and cost-effectiveness of vinorelbine for breast cancer: a systematic review and economic evaluation.
By Lewis R, Bagnall A-M, King S, Woolacott N, Forbes C, Shirran L, et al.
-
A systematic review of the effectiveness and cost-effectiveness of metal-on-metal hip resurfacing arthroplasty for treatment of hip disease.
By Vale L, Wyness L, McCormack K, McKenzie L, Brazzelli M, Stearns SC.
-
The clinical effectiveness and cost-effectiveness of bupropion and nicotine replacement therapy for smoking cessation: a systematic review and economic evaluation.
By Woolacott NF, Jones L, Forbes CA, Mather LC, Sowden AJ, Song FJ, et al.
-
A systematic review of effectiveness and economic evaluation of new drug treatments for juvenile idiopathic arthritis: etanercept.
By Cummins C, Connock M, Fry-Smith A, Burls A.
-
Clinical effectiveness and cost-effectiveness of growth hormone in children: a systematic review and economic evaluation.
By Bryant J, Cave C, Mihaylova B, Chase D, McIntyre L, Gerard K, et al.
-
Clinical effectiveness and cost-effectiveness of growth hormone in adults in relation to impact on quality of life: a systematic review and economic evaluation.
By Bryant J, Loveman E, Chase D, Mihaylova B, Cave C, Gerard K, et al.
-
Clinical medication review by a pharmacist of patients on repeat prescriptions in general practice: a randomised controlled trial.
By Zermansky AG, Petty DR, Raynor DK, Lowe CJ, Freementle N, Vail A.
-
The effectiveness of infliximab and etanercept for the treatment of rheumatoid arthritis: a systematic review and economic evaluation.
By Jobanputra P, Barton P, Bryan S, Burls A.
-
A systematic review and economic evaluation of computerised cognitive behaviour therapy for depression and anxiety.
By Kaltenthaler E, Shackley P, Stevens K, Beverley C, Parry G, Chilcott J.
-
A systematic review and economic evaluation of pegylated liposomal doxorubicin hydrochloride for ovarian cancer.
By Forbes C, Wilby J, Richardson G, Sculpher M, Mather L, Reimsma R.
-
A systematic review of the effectiveness of interventions based on a stages-of-change approach to promote individual behaviour change.
By Riemsma RP, Pattenden J, Bridle C, Sowden AJ, Mather L, Watt IS, et al.
-
A systematic review update of the clinical effectiveness and cost-effectiveness of glycoprotein IIb/IIIa antagonists.
By Robinson M, Ginnelly L, Sculpher M, Jones L, Riemsma R, Palmer S, et al.
-
A systematic review of the effectiveness, cost-effectiveness and barriers to implementation of thrombolytic and neuroprotective therapy for acute ischaemic stroke in the NHS.
By Sandercock P, Berge E, Dennis M, Forbes J, Hand P, Kwan J, et al.
-
A randomised controlled crossover trial of nurse practitioner versus doctor-led outpatient care in a bronchiectasis clinic.
By Caine N, Sharples LD, Hollingworth W, French J, Keogan M, Exley A, et al.
-
Clinical effectiveness and cost – consequences of selective serotonin reuptake inhibitors in the treatment of sex offenders.
By Adi Y, Ashcroft D, Browne K, Beech A, Fry-Smith A, Hyde C.
-
Treatment of established osteoporosis: a systematic review and cost–utility analysis.
By Kanis JA, Brazier JE, Stevenson M, Calvert NW, Lloyd Jones M.
-
Which anaesthetic agents are cost-effective in day surgery? Literature review, national survey of practice and randomised controlled trial.
By Elliott RA Payne K, Moore JK, Davies LM, Harper NJN, St Leger AS, et al.
-
Screening for hepatitis C among injecting drug users and in genitourinary medicine clinics: systematic reviews of effectiveness, modelling study and national survey of current practice.
By Stein K, Dalziel K, Walker A, McIntyre L, Jenkins B, Horne J, et al.
-
The measurement of satisfaction with healthcare: implications for practice from a systematic review of the literature.
By Crow R, Gage H, Hampson S, Hart J, Kimber A, Storey L, et al.
-
The effectiveness and cost-effectiveness of imatinib in chronic myeloid leukaemia: a systematic review.
By Garside R, Round A, Dalziel K, Stein K, Royle R.
-
A comparative study of hypertonic saline, daily and alternate-day rhDNase in children with cystic fibrosis.
By Suri R, Wallis C, Bush A, Thompson S, Normand C, Flather M, et al.
-
A systematic review of the costs and effectiveness of different models of paediatric home care.
By Parker G, Bhakta P, Lovett CA, Paisley S, Olsen R, Turner D, et al.
-
How important are comprehensive literature searches and the assessment of trial quality in systematic reviews? Empirical study.
By Egger M, Jüni P, Bartlett C, Holenstein F, Sterne J.
-
Systematic review of the effectiveness and cost-effectiveness, and economic evaluation, of home versus hospital or satellite unit haemodialysis for people with end-stage renal failure.
By Mowatt G, Vale L, Perez J, Wyness L, Fraser C, MacLeod A, et al.
-
Systematic review and economic evaluation of the effectiveness of infliximab for the treatment of Crohn’s disease.
By Clark W, Raftery J, Barton P, Song F, Fry-Smith A, Burls A.
-
A review of the clinical effectiveness and cost-effectiveness of routine anti-D prophylaxis for pregnant women who are rhesus negative.
By Chilcott J, Lloyd Jones M, Wight J, Forman K, Wray J, Beverley C, et al.
-
Systematic review and evaluation of the use of tumour markers in paediatric oncology: Ewing’s sarcoma and neuroblastoma.
By Riley RD, Burchill SA, Abrams KR, Heney D, Lambert PC, Jones DR, et al.
-
The cost-effectiveness of screening for Helicobacter pylori to reduce mortality and morbidity from gastric cancer and peptic ulcer disease: a discrete-event simulation model.
By Roderick P, Davies R, Raftery J, Crabbe D, Pearce R, Bhandari P, et al.
-
The clinical effectiveness and cost-effectiveness of routine dental checks: a systematic review and economic evaluation.
By Davenport C, Elley K, Salas C, Taylor-Weetman CL, Fry-Smith A, Bryan S, et al.
-
A multicentre randomised controlled trial assessing the costs and benefits of using structured information and analysis of women’s preferences in the management of menorrhagia.
By Kennedy ADM, Sculpher MJ, Coulter A, Dwyer N, Rees M, Horsley S, et al.
-
Clinical effectiveness and cost–utility of photodynamic therapy for wet age-related macular degeneration: a systematic review and economic evaluation.
By Meads C, Salas C, Roberts T, Moore D, Fry-Smith A, Hyde C.
-
Evaluation of molecular tests for prenatal diagnosis of chromosome abnormalities.
By Grimshaw GM, Szczepura A, Hultén M, MacDonald F, Nevin NC, Sutton F, et al.
-
First and second trimester antenatal screening for Down’s syndrome: the results of the Serum, Urine and Ultrasound Screening Study (SURUSS).
By Wald NJ, Rodeck C, Hackshaw AK, Walters J, Chitty L, Mackinson AM.
-
The effectiveness and cost-effectiveness of ultrasound locating devices for central venous access: a systematic review and economic evaluation.
By Calvert N, Hind D, McWilliams RG, Thomas SM, Beverley C, Davidson A.
-
A systematic review of atypical antipsychotics in schizophrenia.
By Bagnall A-M, Jones L, Lewis R, Ginnelly L, Glanville J, Torgerson D, et al.
-
Prostate Testing for Cancer and Treatment (ProtecT) feasibility study.
By Donovan J, Hamdy F, Neal D, Peters T, Oliver S, Brindle L, et al.
-
Early thrombolysis for the treatment of acute myocardial infarction: a systematic review and economic evaluation.
By Boland A, Dundar Y, Bagust A, Haycox A, Hill R, Mujica Mota R, et al.
-
Screening for fragile X syndrome: a literature review and modelling.
By Song FJ, Barton P, Sleightholme V, Yao GL, Fry-Smith A.
-
Systematic review of endoscopic sinus surgery for nasal polyps.
By Dalziel K, Stein K, Round A, Garside R, Royle P.
-
Towards efficient guidelines: how to monitor guideline use in primary care.
By Hutchinson A, McIntosh A, Cox S, Gilbert C.
-
Effectiveness and cost-effectiveness of acute hospital-based spinal cord injuries services: systematic review.
By Bagnall A-M, Jones L, Richardson G, Duffy S, Riemsma R.
-
Prioritisation of health technology assessment. The PATHS model: methods and case studies.
By Townsend J, Buxton M, Harper G.
-
Systematic review of the clinical effectiveness and cost-effectiveness of tension-free vaginal tape for treatment of urinary stress incontinence.
By Cody J, Wyness L, Wallace S, Glazener C, Kilonzo M, Stearns S, et al.
-
The clinical and cost-effectiveness of patient education models for diabetes: a systematic review and economic evaluation.
By Loveman E, Cave C, Green C, Royle P, Dunn N, Waugh N.
-
The role of modelling in prioritising and planning clinical trials.
By Chilcott J, Brennan A, Booth A, Karnon J, Tappenden P.
-
Cost–benefit evaluation of routine influenza immunisation in people 65–74 years of age.
By Allsup S, Gosney M, Haycox A, Regan M.
-
The clinical and cost-effectiveness of pulsatile machine perfusion versus cold storage of kidneys for transplantation retrieved from heart-beating and non-heart-beating donors.
By Wight J, Chilcott J, Holmes M, Brewer N.
-
Can randomised trials rely on existing electronic data? A feasibility study to explore the value of routine data in health technology assessment.
By Williams JG, Cheung WY, Cohen DR, Hutchings HA, Longo MF, Russell IT.
-
Evaluating non-randomised intervention studies.
By Deeks JJ, Dinnes J, D’Amico R, Sowden AJ, Sakarovitch C, Song F, et al.
-
A randomised controlled trial to assess the impact of a package comprising a patient-orientated, evidence-based self- help guidebook and patient-centred consultations on disease management and satisfaction in inflammatory bowel disease.
By Kennedy A, Nelson E, Reeves D, Richardson G, Roberts C, Robinson A, et al.
-
The effectiveness of diagnostic tests for the assessment of shoulder pain due to soft tissue disorders: a systematic review.
By Dinnes J, Loveman E, McIntyre L, Waugh N.
-
The value of digital imaging in diabetic retinopathy.
By Sharp PF, Olson J, Strachan F, Hipwell J, Ludbrook A, O’Donnell M, et al.
-
Lowering blood pressure to prevent myocardial infarction and stroke: a new preventive strategy.
By Law M, Wald N, Morris J.
-
Clinical and cost-effectiveness of capecitabine and tegafur with uracil for the treatment of metastatic colorectal cancer: systematic review and economic evaluation.
By Ward S, Kaltenthaler E, Cowan J, Brewer N.
-
Clinical and cost-effectiveness of new and emerging technologies for early localised prostate cancer: a systematic review.
By Hummel S, Paisley S, Morgan A, Currie E, Brewer N.
-
Literature searching for clinical and cost-effectiveness studies used in health technology assessment reports carried out for the National Institute for Clinical Excellence appraisal system.
By Royle P, Waugh N.
-
Systematic review and economic decision modelling for the prevention and treatment of influenza A and B.
By Turner D, Wailoo A, Nicholson K, Cooper N, Sutton A, Abrams K.
-
A randomised controlled trial to evaluate the clinical and cost-effectiveness of Hickman line insertions in adult cancer patients by nurses.
By Boland A, Haycox A, Bagust A, Fitzsimmons L.
-
Redesigning postnatal care: a randomised controlled trial of protocol-based midwifery-led care focused on individual women’s physical and psychological health needs.
By MacArthur C, Winter HR, Bick DE, Lilford RJ, Lancashire RJ, Knowles H, et al.
-
Estimating implied rates of discount in healthcare decision-making.
By West RR, McNabb R, Thompson AGH, Sheldon TA, Grimley Evans J.
-
Systematic review of isolation policies in the hospital management of methicillin-resistant Staphylococcus aureus: a review of the literature with epidemiological and economic modelling.
By Cooper BS, Stone SP, Kibbler CC, Cookson BD, Roberts JA, Medley GF, et al.
-
Treatments for spasticity and pain in multiple sclerosis: a systematic review.
By Beard S, Hunn A, Wight J.
-
The inclusion of reports of randomised trials published in languages other than English in systematic reviews.
By Moher D, Pham B, Lawson ML, Klassen TP.
-
The impact of screening on future health-promoting behaviours and health beliefs: a systematic review.
By Bankhead CR, Brett J, Bukach C, Webster P, Stewart-Brown S, Munafo M, et al.
-
What is the best imaging strategy for acute stroke?
By Wardlaw JM, Keir SL, Seymour J, Lewis S, Sandercock PAG, Dennis MS, et al.
-
Systematic review and modelling of the investigation of acute and chronic chest pain presenting in primary care.
By Mant J, McManus RJ, Oakes RAL, Delaney BC, Barton PM, Deeks JJ, et al.
-
The effectiveness and cost-effectiveness of microwave and thermal balloon endometrial ablation for heavy menstrual bleeding: a systematic review and economic modelling.
By Garside R, Stein K, Wyatt K, Round A, Price A.
-
A systematic review of the role of bisphosphonates in metastatic disease.
By Ross JR, Saunders Y, Edmonds PM, Patel S, Wonderling D, Normand C, et al.
-
Systematic review of the clinical effectiveness and cost-effectiveness of capecitabine (Xeloda®) for locally advanced and/or metastatic breast cancer.
By Jones L, Hawkins N, Westwood M, Wright K, Richardson G, Riemsma R.
-
Effectiveness and efficiency of guideline dissemination and implementation strategies.
By Grimshaw JM, Thomas RE, MacLennan G, Fraser C, Ramsay CR, Vale L, et al.
-
Clinical effectiveness and costs of the Sugarbaker procedure for the treatment of pseudomyxoma peritonei.
By Bryant J, Clegg AJ, Sidhu MK, Brodin H, Royle P, Davidson P.
-
Psychological treatment for insomnia in the regulation of long-term hypnotic drug use.
By Morgan K, Dixon S, Mathers N, Thompson J, Tomeny M.
-
Improving the evaluation of therapeutic interventions in multiple sclerosis: development of a patient-based measure of outcome.
By Hobart JC, Riazi A, Lamping DL, Fitzpatrick R, Thompson AJ.
-
A systematic review and economic evaluation of magnetic resonance cholangiopancreatography compared with diagnostic endoscopic retrograde cholangiopancreatography.
By Kaltenthaler E, Bravo Vergel Y, Chilcott J, Thomas S, Blakeborough T, Walters SJ, et al.
-
The use of modelling to evaluate new drugs for patients with a chronic condition: the case of antibodies against tumour necrosis factor in rheumatoid arthritis.
By Barton P, Jobanputra P, Wilson J, Bryan S, Burls A.
-
Clinical effectiveness and cost-effectiveness of neonatal screening for inborn errors of metabolism using tandem mass spectrometry: a systematic review.
By Pandor A, Eastham J, Beverley C, Chilcott J, Paisley S.
-
Clinical effectiveness and cost-effectiveness of pioglitazone and rosiglitazone in the treatment of type 2 diabetes: a systematic review and economic evaluation.
By Czoski-Murray C, Warren E, Chilcott J, Beverley C, Psyllaki MA, Cowan J.
-
Routine examination of the newborn: the EMREN study. Evaluation of an extension of the midwife role including a randomised controlled trial of appropriately trained midwives and paediatric senior house officers.
By Townsend J, Wolke D, Hayes J, Davé S, Rogers C, Bloomfield L, et al.
-
Involving consumers in research and development agenda setting for the NHS: developing an evidence-based approach.
By Oliver S, Clarke-Jones L, Rees R, Milne R, Buchanan P, Gabbay J, et al.
-
A multi-centre randomised controlled trial of minimally invasive direct coronary bypass grafting versus percutaneous transluminal coronary angioplasty with stenting for proximal stenosis of the left anterior descending coronary artery.
By Reeves BC, Angelini GD, Bryan AJ, Taylor FC, Cripps T, Spyt TJ, et al.
-
Does early magnetic resonance imaging influence management or improve outcome in patients referred to secondary care with low back pain? A pragmatic randomised controlled trial.
By Gilbert FJ, Grant AM, Gillan MGC, Vale L, Scott NW, Campbell MK, et al.
-
The clinical and cost-effectiveness of anakinra for the treatment of rheumatoid arthritis in adults: a systematic review and economic analysis.
By Clark W, Jobanputra P, Barton P, Burls A.
-
A rapid and systematic review and economic evaluation of the clinical and cost-effectiveness of newer drugs for treatment of mania associated with bipolar affective disorder.
By Bridle C, Palmer S, Bagnall A-M, Darba J, Duffy S, Sculpher M, et al.
-
Liquid-based cytology in cervical screening: an updated rapid and systematic review and economic analysis.
By Karnon J, Peters J, Platt J, Chilcott J, McGoogan E, Brewer N.
-
Systematic review of the long-term effects and economic consequences of treatments for obesity and implications for health improvement.
By Avenell A, Broom J, Brown TJ, Poobalan A, Aucott L, Stearns SC, et al.
-
Autoantibody testing in children with newly diagnosed type 1 diabetes mellitus.
By Dretzke J, Cummins C, Sandercock J, Fry-Smith A, Barrett T, Burls A.
-
Clinical effectiveness and cost-effectiveness of prehospital intravenous fluids in trauma patients.
By Dretzke J, Sandercock J, Bayliss S, Burls A.
-
Newer hypnotic drugs for the short-term management of insomnia: a systematic review and economic evaluation.
By Dündar Y, Boland A, Strobl J, Dodd S, Haycox A, Bagust A, et al.
-
Development and validation of methods for assessing the quality of diagnostic accuracy studies.
By Whiting P, Rutjes AWS, Dinnes J, Reitsma JB, Bossuyt PMM, Kleijnen J.
-
EVALUATE hysterectomy trial: a multicentre randomised trial comparing abdominal, vaginal and laparoscopic methods of hysterectomy.
By Garry R, Fountain J, Brown J, Manca A, Mason S, Sculpher M, et al.
-
Methods for expected value of information analysis in complex health economic models: developments on the health economics of interferon-β and glatiramer acetate for multiple sclerosis.
By Tappenden P, Chilcott JB, Eggington S, Oakley J, McCabe C.
-
Effectiveness and cost-effectiveness of imatinib for first-line treatment of chronic myeloid leukaemia in chronic phase: a systematic review and economic analysis.
By Dalziel K, Round A, Stein K, Garside R, Price A.
-
VenUS I: a randomised controlled trial of two types of bandage for treating venous leg ulcers.
By Iglesias C, Nelson EA, Cullum NA, Torgerson DJ, on behalf of the VenUS Team.
-
Systematic review of the effectiveness and cost-effectiveness, and economic evaluation, of myocardial perfusion scintigraphy for the diagnosis and management of angina and myocardial infarction.
By Mowatt G, Vale L, Brazzelli M, Hernandez R, Murray A, Scott N, et al.
-
A pilot study on the use of decision theory and value of information analysis as part of the NHS Health Technology Assessment programme.
By Claxton K, Ginnelly L, Sculpher M, Philips Z, Palmer S.
-
The Social Support and Family Health Study: a randomised controlled trial and economic evaluation of two alternative forms of postnatal support for mothers living in disadvantaged inner-city areas.
By Wiggins M, Oakley A, Roberts I, Turner H, Rajan L, Austerberry H, et al.
-
Psychosocial aspects of genetic screening of pregnant women and newborns: a systematic review.
By Green JM, Hewison J, Bekker HL, Bryant, Cuckle HS.
-
Evaluation of abnormal uterine bleeding: comparison of three outpatient procedures within cohorts defined by age and menopausal status.
By Critchley HOD, Warner P, Lee AJ, Brechin S, Guise J, Graham B.
-
Coronary artery stents: a rapid systematic review and economic evaluation.
By Hill R, Bagust A, Bakhai A, Dickson R, Dündar Y, Haycox A, et al.
-
Review of guidelines for good practice in decision-analytic modelling in health technology assessment.
By Philips Z, Ginnelly L, Sculpher M, Claxton K, Golder S, Riemsma R, et al.
-
Rituximab (MabThera®) for aggressive non-Hodgkin’s lymphoma: systematic review and economic evaluation.
By Knight C, Hind D, Brewer N, Abbott V.
-
Clinical effectiveness and cost-effectiveness of clopidogrel and modified-release dipyridamole in the secondary prevention of occlusive vascular events: a systematic review and economic evaluation.
By Jones L, Griffin S, Palmer S, Main C, Orton V, Sculpher M, et al.
-
Pegylated interferon α-2a and -2b in combination with ribavirin in the treatment of chronic hepatitis C: a systematic review and economic evaluation.
By Shepherd J, Brodin H, Cave C, Waugh N, Price A, Gabbay J.
-
Clopidogrel used in combination with aspirin compared with aspirin alone in the treatment of non-ST-segment- elevation acute coronary syndromes: a systematic review and economic evaluation.
By Main C, Palmer S, Griffin S, Jones L, Orton V, Sculpher M, et al.
-
Provision, uptake and cost of cardiac rehabilitation programmes: improving services to under-represented groups.
By Beswick AD, Rees K, Griebsch I, Taylor FC, Burke M, West RR, et al.
-
Involving South Asian patients in clinical trials.
By Hussain-Gambles M, Leese B, Atkin K, Brown J, Mason S, Tovey P.
-
Clinical and cost-effectiveness of continuous subcutaneous insulin infusion for diabetes.
By Colquitt JL, Green C, Sidhu MK, Hartwell D, Waugh N.
-
Identification and assessment of ongoing trials in health technology assessment reviews.
By Song FJ, Fry-Smith A, Davenport C, Bayliss S, Adi Y, Wilson JS, et al.
-
Systematic review and economic evaluation of a long-acting insulin analogue, insulin glargine
By Warren E, Weatherley-Jones E, Chilcott J, Beverley C.
-
Supplementation of a home-based exercise programme with a class-based programme for people with osteoarthritis of the knees: a randomised controlled trial and health economic analysis.
By McCarthy CJ, Mills PM, Pullen R, Richardson G, Hawkins N, Roberts CR, et al.
-
Clinical and cost-effectiveness of once-daily versus more frequent use of same potency topical corticosteroids for atopic eczema: a systematic review and economic evaluation.
By Green C, Colquitt JL, Kirby J, Davidson P, Payne E.
-
Acupuncture of chronic headache disorders in primary care: randomised controlled trial and economic analysis.
By Vickers AJ, Rees RW, Zollman CE, McCarney R, Smith CM, Ellis N, et al.
-
Generalisability in economic evaluation studies in healthcare: a review and case studies.
By Sculpher MJ, Pang FS, Manca A, Drummond MF, Golder S, Urdahl H, et al.
-
Virtual outreach: a randomised controlled trial and economic evaluation of joint teleconferenced medical consultations.
By Wallace P, Barber J, Clayton W, Currell R, Fleming K, Garner P, et al.
-
Randomised controlled multiple treatment comparison to provide a cost-effectiveness rationale for the selection of antimicrobial therapy in acne.
By Ozolins M, Eady EA, Avery A, Cunliffe WJ, O’Neill C, Simpson NB, et al.
-
Do the findings of case series studies vary significantly according to methodological characteristics?
By Dalziel K, Round A, Stein K, Garside R, Castelnuovo E, Payne L.
-
Improving the referral process for familial breast cancer genetic counselling: findings of three randomised controlled trials of two interventions.
By Wilson BJ, Torrance N, Mollison J, Wordsworth S, Gray JR, Haites NE, et al.
-
Randomised evaluation of alternative electrosurgical modalities to treat bladder outflow obstruction in men with benign prostatic hyperplasia.
By Fowler C, McAllister W, Plail R, Karim O, Yang Q.
-
A pragmatic randomised controlled trial of the cost-effectiveness of palliative therapies for patients with inoperable oesophageal cancer.
By Shenfine J, McNamee P, Steen N, Bond J, Griffin SM.
-
Impact of computer-aided detection prompts on the sensitivity and specificity of screening mammography.
By Taylor P, Champness J, Given- Wilson R, Johnston K, Potts H.
-
Issues in data monitoring and interim analysis of trials.
By Grant AM, Altman DG, Babiker AB, Campbell MK, Clemens FJ, Darbyshire JH, et al.
-
Lay public’s understanding of equipoise and randomisation in randomised controlled trials.
By Robinson EJ, Kerr CEP, Stevens AJ, Lilford RJ, Braunholtz DA, Edwards SJ, et al.
-
Clinical and cost-effectiveness of electroconvulsive therapy for depressive illness, schizophrenia, catatonia and mania: systematic reviews and economic modelling studies.
By Greenhalgh J, Knight C, Hind D, Beverley C, Walters S.
-
Measurement of health-related quality of life for people with dementia: development of a new instrument (DEMQOL) and an evaluation of current methodology.
By Smith SC, Lamping DL, Banerjee S, Harwood R, Foley B, Smith P, et al.
-
Clinical effectiveness and cost-effectiveness of drotrecogin alfa (activated) (Xigris®) for the treatment of severe sepsis in adults: a systematic review and economic evaluation.
By Green C, Dinnes J, Takeda A, Shepherd J, Hartwell D, Cave C, et al.
-
A methodological review of how heterogeneity has been examined in systematic reviews of diagnostic test accuracy.
By Dinnes J, Deeks J, Kirby J, Roderick P.
-
Cervical screening programmes: can automation help? Evidence from systematic reviews, an economic analysis and a simulation modelling exercise applied to the UK.
By Willis BH, Barton P, Pearmain P, Bryan S, Hyde C.
-
Laparoscopic surgery for inguinal hernia repair: systematic review of effectiveness and economic evaluation.
By McCormack K, Wake B, Perez J, Fraser C, Cook J, McIntosh E, et al.
-
Clinical effectiveness, tolerability and cost-effectiveness of newer drugs for epilepsy in adults: a systematic review and economic evaluation.
By Wilby J, Kainth A, Hawkins N, Epstein D, McIntosh H, McDaid C, et al.
-
A randomised controlled trial to compare the cost-effectiveness of tricyclic antidepressants, selective serotonin reuptake inhibitors and lofepramine.
By Peveler R, Kendrick T, Buxton M, Longworth L, Baldwin D, Moore M, et al.
-
Clinical effectiveness and cost-effectiveness of immediate angioplasty for acute myocardial infarction: systematic review and economic evaluation.
By Hartwell D, Colquitt J, Loveman E, Clegg AJ, Brodin H, Waugh N, et al.
-
A randomised controlled comparison of alternative strategies in stroke care.
By Kalra L, Evans A, Perez I, Knapp M, Swift C, Donaldson N.
-
The investigation and analysis of critical incidents and adverse events in healthcare.
By Woloshynowych M, Rogers S, Taylor-Adams S, Vincent C.
-
Potential use of routine databases in health technology assessment.
By Raftery J, Roderick P, Stevens A.
-
Clinical and cost-effectiveness of newer immunosuppressive regimens in renal transplantation: a systematic review and modelling study.
By Woodroffe R, Yao GL, Meads C, Bayliss S, Ready A, Raftery J, et al.
-
A systematic review and economic evaluation of alendronate, etidronate, risedronate, raloxifene and teriparatide for the prevention and treatment of postmenopausal osteoporosis.
By Stevenson M, Lloyd Jones M, De Nigris E, Brewer N, Davis S, Oakley J.
-
A systematic review to examine the impact of psycho-educational interventions on health outcomes and costs in adults and children with difficult asthma.
By Smith JR, Mugford M, Holland R, Candy B, Noble MJ, Harrison BDW, et al.
-
An evaluation of the costs, effectiveness and quality of renal replacement therapy provision in renal satellite units in England and Wales.
By Roderick P, Nicholson T, Armitage A, Mehta R, Mullee M, Gerard K, et al.
-
Imatinib for the treatment of patients with unresectable and/or metastatic gastrointestinal stromal tumours: systematic review and economic evaluation.
By Wilson J, Connock M, Song F, Yao G, Fry-Smith A, Raftery J, et al.
-
Indirect comparisons of competing interventions.
By Glenny AM, Altman DG, Song F, Sakarovitch C, Deeks JJ, D’Amico R, et al.
-
Cost-effectiveness of alternative strategies for the initial medical management of non-ST elevation acute coronary syndrome: systematic review and decision-analytical modelling.
By Robinson M, Palmer S, Sculpher M, Philips Z, Ginnelly L, Bowens A, et al.
-
Outcomes of electrically stimulated gracilis neosphincter surgery.
By Tillin T, Chambers M, Feldman R.
-
The effectiveness and cost-effectiveness of pimecrolimus and tacrolimus for atopic eczema: a systematic review and economic evaluation.
By Garside R, Stein K, Castelnuovo E, Pitt M, Ashcroft D, Dimmock P, et al.
-
Systematic review on urine albumin testing for early detection of diabetic complications.
By Newman DJ, Mattock MB, Dawnay ABS, Kerry S, McGuire A, Yaqoob M, et al.
-
Randomised controlled trial of the cost-effectiveness of water-based therapy for lower limb osteoarthritis.
By Cochrane T, Davey RC, Matthes Edwards SM.
-
Longer term clinical and economic benefits of offering acupuncture care to patients with chronic low back pain.
By Thomas KJ, MacPherson H, Ratcliffe J, Thorpe L, Brazier J, Campbell M, et al.
-
Cost-effectiveness and safety of epidural steroids in the management of sciatica.
By Price C, Arden N, Coglan L, Rogers P.
-
The British Rheumatoid Outcome Study Group (BROSG) randomised controlled trial to compare the effectiveness and cost-effectiveness of aggressive versus symptomatic therapy in established rheumatoid arthritis.
By Symmons D, Tricker K, Roberts C, Davies L, Dawes P, Scott DL.
-
Conceptual framework and systematic review of the effects of participants’ and professionals’ preferences in randomised controlled trials.
By King M, Nazareth I, Lampe F, Bower P, Chandler M, Morou M, et al.
-
The clinical and cost-effectiveness of implantable cardioverter defibrillators: a systematic review.
By Bryant J, Brodin H, Loveman E, Payne E, Clegg A.
-
A trial of problem-solving by community mental health nurses for anxiety, depression and life difficulties among general practice patients. The CPN-GP study.
By Kendrick T, Simons L, Mynors-Wallis L, Gray A, Lathlean J, Pickering R, et al.
-
The causes and effects of socio-demographic exclusions from clinical trials.
By Bartlett C, Doyal L, Ebrahim S, Davey P, Bachmann M, Egger M, et al.
-
Is hydrotherapy cost-effective? A randomised controlled trial of combined hydrotherapy programmes compared with physiotherapy land techniques in children with juvenile idiopathic arthritis.
By Epps H, Ginnelly L, Utley M, Southwood T, Gallivan S, Sculpher M, et al.
-
A randomised controlled trial and cost-effectiveness study of systematic screening (targeted and total population screening) versus routine practice for the detection of atrial fibrillation in people aged 65 and over. The SAFE study.
By Hobbs FDR, Fitzmaurice DA, Mant J, Murray E, Jowett S, Bryan S, et al.
-
Displaced intracapsular hip fractures in fit, older people: a randomised comparison of reduction and fixation, bipolar hemiarthroplasty and total hip arthroplasty.
By Keating JF, Grant A, Masson M, Scott NW, Forbes JF.
-
Long-term outcome of cognitive behaviour therapy clinical trials in central Scotland.
By Durham RC, Chambers JA, Power KG, Sharp DM, Macdonald RR, Major KA, et al.
-
The effectiveness and cost-effectiveness of dual-chamber pacemakers compared with single-chamber pacemakers for bradycardia due to atrioventricular block or sick sinus syndrome: systematic review and economic evaluation.
By Castelnuovo E, Stein K, Pitt M, Garside R, Payne E.
-
Newborn screening for congenital heart defects: a systematic review and cost-effectiveness analysis.
By Knowles R, Griebsch I, Dezateux C, Brown J, Bull C, Wren C.
-
The clinical and cost-effectiveness of left ventricular assist devices for end-stage heart failure: a systematic review and economic evaluation.
By Clegg AJ, Scott DA, Loveman E, Colquitt J, Hutchinson J, Royle P, et al.
-
The effectiveness of the Heidelberg Retina Tomograph and laser diagnostic glaucoma scanning system (GDx) in detecting and monitoring glaucoma.
By Kwartz AJ, Henson DB, Harper RA, Spencer AF, McLeod D.
-
Clinical and cost-effectiveness of autologous chondrocyte implantation for cartilage defects in knee joints: systematic review and economic evaluation.
By Clar C, Cummins E, McIntyre L, Thomas S, Lamb J, Bain L, et al.
-
Systematic review of effectiveness of different treatments for childhood retinoblastoma.
By McDaid C, Hartley S, Bagnall A-M, Ritchie G, Light K, Riemsma R.
-
Towards evidence-based guidelines for the prevention of venous thromboembolism: systematic reviews of mechanical methods, oral anticoagulation, dextran and regional anaesthesia as thromboprophylaxis.
By Roderick P, Ferris G, Wilson K, Halls H, Jackson D, Collins R, et al.
-
The effectiveness and cost-effectiveness of parent training/education programmes for the treatment of conduct disorder, including oppositional defiant disorder, in children.
By Dretzke J, Frew E, Davenport C, Barlow J, Stewart-Brown S, Sandercock J, et al.
-
The clinical and cost-effectiveness of donepezil, rivastigmine, galantamine and memantine for Alzheimer’s disease.
By Loveman E, Green C, Kirby J, Takeda A, Picot J, Payne E, et al.
-
FOOD: a multicentre randomised trial evaluating feeding policies in patients admitted to hospital with a recent stroke.
By Dennis M, Lewis S, Cranswick G, Forbes J.
-
The clinical effectiveness and cost-effectiveness of computed tomography screening for lung cancer: systematic reviews.
By Black C, Bagust A, Boland A, Walker S, McLeod C, De Verteuil R, et al.
-
A systematic review of the effectiveness and cost-effectiveness of neuroimaging assessments used to visualise the seizure focus in people with refractory epilepsy being considered for surgery.
By Whiting P, Gupta R, Burch J, Mujica Mota RE, Wright K, Marson A, et al.
-
Comparison of conference abstracts and presentations with full-text articles in the health technology assessments of rapidly evolving technologies.
By Dundar Y, Dodd S, Dickson R, Walley T, Haycox A, Williamson PR.
-
Systematic review and evaluation of methods of assessing urinary incontinence.
By Martin JL, Williams KS, Abrams KR, Turner DA, Sutton AJ, Chapple C, et al.
-
The clinical effectiveness and cost-effectiveness of newer drugs for children with epilepsy. A systematic review.
By Connock M, Frew E, Evans B-W, Bryan S, Cummins C, Fry-Smith A, et al.
-
Surveillance of Barrett’s oesophagus: exploring the uncertainty through systematic review, expert workshop and economic modelling.
By Garside R, Pitt M, Somerville M, Stein K, Price A, Gilbert N.
-
Topotecan, pegylated liposomal doxorubicin hydrochloride and paclitaxel for second-line or subsequent treatment of advanced ovarian cancer: a systematic review and economic evaluation.
By Main C, Bojke L, Griffin S, Norman G, Barbieri M, Mather L, et al.
-
Evaluation of molecular techniques in prediction and diagnosis of cytomegalovirus disease in immunocompromised patients.
By Szczepura A, Westmoreland D, Vinogradova Y, Fox J, Clark M.
-
Screening for thrombophilia in high-risk situations: systematic review and cost-effectiveness analysis. The Thrombosis: Risk and Economic Assessment of Thrombophilia Screening (TREATS) study.
By Wu O, Robertson L, Twaddle S, Lowe GDO, Clark P, Greaves M, et al.
-
A series of systematic reviews to inform a decision analysis for sampling and treating infected diabetic foot ulcers.
By Nelson EA, O’Meara S, Craig D, Iglesias C, Golder S, Dalton J, et al.
-
Randomised clinical trial, observational study and assessment of cost-effectiveness of the treatment of varicose veins (REACTIV trial).
By Michaels JA, Campbell WB, Brazier JE, MacIntyre JB, Palfreyman SJ, Ratcliffe J, et al.
-
The cost-effectiveness of screening for oral cancer in primary care.
By Speight PM, Palmer S, Moles DR, Downer MC, Smith DH, Henriksson M, et al.
-
Measurement of the clinical and cost-effectiveness of non-invasive diagnostic testing strategies for deep vein thrombosis.
By Goodacre S, Sampson F, Stevenson M, Wailoo A, Sutton A, Thomas S, et al.
-
Systematic review of the effectiveness and cost-effectiveness of HealOzone® for the treatment of occlusal pit/fissure caries and root caries.
By Brazzelli M, McKenzie L, Fielding S, Fraser C, Clarkson J, Kilonzo M, et al.
-
Randomised controlled trials of conventional antipsychotic versus new atypical drugs, and new atypical drugs versus clozapine, in people with schizophrenia responding poorly to, or intolerant of, current drug treatment.
By Lewis SW, Davies L, Jones PB, Barnes TRE, Murray RM, Kerwin R, et al.
-
Diagnostic tests and algorithms used in the investigation of haematuria: systematic reviews and economic evaluation.
By Rodgers M, Nixon J, Hempel S, Aho T, Kelly J, Neal D, et al.
-
Cognitive behavioural therapy in addition to antispasmodic therapy for irritable bowel syndrome in primary care: randomised controlled trial.
By Kennedy TM, Chalder T, McCrone P, Darnley S, Knapp M, Jones RH, et al.
-
A systematic review of the clinical effectiveness and cost-effectiveness of enzyme replacement therapies for Fabry’s disease and mucopolysaccharidosis type 1.
By Connock M, Juarez-Garcia A, Frew E, Mans A, Dretzke J, Fry-Smith A, et al.
-
Health benefits of antiviral therapy for mild chronic hepatitis C: randomised controlled trial and economic evaluation.
By Wright M, Grieve R, Roberts J, Main J, Thomas HC, on behalf of the UK Mild Hepatitis C Trial Investigators.
-
Pressure relieving support surfaces: a randomised evaluation.
By Nixon J, Nelson EA, Cranny G, Iglesias CP, Hawkins K, Cullum NA, et al.
-
A systematic review and economic model of the effectiveness and cost-effectiveness of methylphenidate, dexamfetamine and atomoxetine for the treatment of attention deficit hyperactivity disorder in children and adolescents.
By King S, Griffin S, Hodges Z, Weatherly H, Asseburg C, Richardson G, et al.
-
The clinical effectiveness and cost-effectiveness of enzyme replacement therapy for Gaucher’s disease: a systematic review.
By Connock M, Burls A, Frew E, Fry-Smith A, Juarez-Garcia A, McCabe C, et al.
-
Effectiveness and cost-effectiveness of salicylic acid and cryotherapy for cutaneous warts. An economic decision model.
By Thomas KS, Keogh-Brown MR, Chalmers JR, Fordham RJ, Holland RC, Armstrong SJ, et al.
-
A systematic literature review of the effectiveness of non-pharmacological interventions to prevent wandering in dementia and evaluation of the ethical implications and acceptability of their use.
By Robinson L, Hutchings D, Corner L, Beyer F, Dickinson H, Vanoli A, et al.
-
A review of the evidence on the effects and costs of implantable cardioverter defibrillator therapy in different patient groups, and modelling of cost-effectiveness and cost–utility for these groups in a UK context.
By Buxton M, Caine N, Chase D, Connelly D, Grace A, Jackson C, et al.
-
Adefovir dipivoxil and pegylated interferon alfa-2a for the treatment of chronic hepatitis B: a systematic review and economic evaluation.
By Shepherd J, Jones J, Takeda A, Davidson P, Price A.
-
An evaluation of the clinical and cost-effectiveness of pulmonary artery catheters in patient management in intensive care: a systematic review and a randomised controlled trial.
By Harvey S, Stevens K, Harrison D, Young D, Brampton W, McCabe C, et al.
-
Accurate, practical and cost-effective assessment of carotid stenosis in the UK.
By Wardlaw JM, Chappell FM, Stevenson M, De Nigris E, Thomas S, Gillard J, et al.
-
Etanercept and infliximab for the treatment of psoriatic arthritis: a systematic review and economic evaluation.
By Woolacott N, Bravo Vergel Y, Hawkins N, Kainth A, Khadjesari Z, Misso K, et al.
-
The cost-effectiveness of testing for hepatitis C in former injecting drug users.
By Castelnuovo E, Thompson-Coon J, Pitt M, Cramp M, Siebert U, Price A, et al.
-
Computerised cognitive behaviour therapy for depression and anxiety update: a systematic review and economic evaluation.
By Kaltenthaler E, Brazier J, De Nigris E, Tumur I, Ferriter M, Beverley C, et al.
-
Cost-effectiveness of using prognostic information to select women with breast cancer for adjuvant systemic therapy.
By Williams C, Brunskill S, Altman D, Briggs A, Campbell H, Clarke M, et al.
-
Psychological therapies including dialectical behaviour therapy for borderline personality disorder: a systematic review and preliminary economic evaluation.
By Brazier J, Tumur I, Holmes M, Ferriter M, Parry G, Dent-Brown K, et al.
-
Clinical effectiveness and cost-effectiveness of tests for the diagnosis and investigation of urinary tract infection in children: a systematic review and economic model.
By Whiting P, Westwood M, Bojke L, Palmer S, Richardson G, Cooper J, et al.
-
Cognitive behavioural therapy in chronic fatigue syndrome: a randomised controlled trial of an outpatient group programme.
By O’Dowd H, Gladwell P, Rogers CA, Hollinghurst S, Gregory A.
-
A comparison of the cost-effectiveness of five strategies for the prevention of nonsteroidal anti-inflammatory drug-induced gastrointestinal toxicity: a systematic review with economic modelling.
By Brown TJ, Hooper L, Elliott RA, Payne K, Webb R, Roberts C, et al.
-
The effectiveness and cost-effectiveness of computed tomography screening for coronary artery disease: systematic review.
By Waugh N, Black C, Walker S, McIntyre L, Cummins E, Hillis G.
-
What are the clinical outcome and cost-effectiveness of endoscopy undertaken by nurses when compared with doctors? A Multi-Institution Nurse Endoscopy Trial (MINuET).
By Williams J, Russell I, Durai D, Cheung W-Y, Farrin A, Bloor K, et al.
-
The clinical and cost-effectiveness of oxaliplatin and capecitabine for the adjuvant treatment of colon cancer: systematic review and economic evaluation.
By Pandor A, Eggington S, Paisley S, Tappenden P, Sutcliffe P.
-
A systematic review of the effectiveness of adalimumab, etanercept and infliximab for the treatment of rheumatoid arthritis in adults and an economic evaluation of their cost-effectiveness.
By Chen Y-F, Jobanputra P, Barton P, Jowett S, Bryan S, Clark W, et al.
-
Telemedicine in dermatology: a randomised controlled trial.
By Bowns IR, Collins K, Walters SJ, McDonagh AJG.
-
Cost-effectiveness of cell salvage and alternative methods of minimising perioperative allogeneic blood transfusion: a systematic review and economic model.
By Davies L, Brown TJ, Haynes S, Payne K, Elliott RA, McCollum C.
-
Clinical effectiveness and cost-effectiveness of laparoscopic surgery for colorectal cancer: systematic reviews and economic evaluation.
By Murray A, Lourenco T, de Verteuil R, Hernandez R, Fraser C, McKinley A, et al.
-
Etanercept and efalizumab for the treatment of psoriasis: a systematic review.
By Woolacott N, Hawkins N, Mason A, Kainth A, Khadjesari Z, Bravo Vergel Y, et al.
-
Systematic reviews of clinical decision tools for acute abdominal pain.
By Liu JLY, Wyatt JC, Deeks JJ, Clamp S, Keen J, Verde P, et al.
-
Evaluation of the ventricular assist device programme in the UK.
By Sharples L, Buxton M, Caine N, Cafferty F, Demiris N, Dyer M, et al.
-
A systematic review and economic model of the clinical and cost-effectiveness of immunosuppressive therapy for renal transplantation in children.
By Yao G, Albon E, Adi Y, Milford D, Bayliss S, Ready A, et al.
-
Amniocentesis results: investigation of anxiety. The ARIA trial.
By Hewison J, Nixon J, Fountain J, Cocks K, Jones C, Mason G, et al.
-
Pemetrexed disodium for the treatment of malignant pleural mesothelioma: a systematic review and economic evaluation.
By Dundar Y, Bagust A, Dickson R, Dodd S, Green J, Haycox A, et al.
-
A systematic review and economic model of the clinical effectiveness and cost-effectiveness of docetaxel in combination with prednisone or prednisolone for the treatment of hormone-refractory metastatic prostate cancer.
By Collins R, Fenwick E, Trowman R, Perard R, Norman G, Light K, et al.
-
A systematic review of rapid diagnostic tests for the detection of tuberculosis infection.
By Dinnes J, Deeks J, Kunst H, Gibson A, Cummins E, Waugh N, et al.
-
The clinical effectiveness and cost-effectiveness of strontium ranelate for the prevention of osteoporotic fragility fractures in postmenopausal women.
By Stevenson M, Davis S, Lloyd-Jones M, Beverley C.
-
A systematic review of quantitative and qualitative research on the role and effectiveness of written information available to patients about individual medicines.
By Raynor DK, Blenkinsopp A, Knapp P, Grime J, Nicolson DJ, Pollock K, et al.
-
Oral naltrexone as a treatment for relapse prevention in formerly opioid-dependent drug users: a systematic review and economic evaluation.
By Adi Y, Juarez-Garcia A, Wang D, Jowett S, Frew E, Day E, et al.
-
Glucocorticoid-induced osteoporosis: a systematic review and cost–utility analysis.
By Kanis JA, Stevenson M, McCloskey EV, Davis S, Lloyd-Jones M.
-
Epidemiological, social, diagnostic and economic evaluation of population screening for genital chlamydial infection.
By Low N, McCarthy A, Macleod J, Salisbury C, Campbell R, Roberts TE, et al.
-
Methadone and buprenorphine for the management of opioid dependence: a systematic review and economic evaluation.
By Connock M, Juarez-Garcia A, Jowett S, Frew E, Liu Z, Taylor RJ, et al.
-
Exercise Evaluation Randomised Trial (EXERT): a randomised trial comparing GP referral for leisure centre-based exercise, community-based walking and advice only.
By Isaacs AJ, Critchley JA, See Tai S, Buckingham K, Westley D, Harridge SDR, et al.
-
Interferon alfa (pegylated and non-pegylated) and ribavirin for the treatment of mild chronic hepatitis C: a systematic review and economic evaluation.
By Shepherd J, Jones J, Hartwell D, Davidson P, Price A, Waugh N.
-
Systematic review and economic evaluation of bevacizumab and cetuximab for the treatment of metastatic colorectal cancer.
By Tappenden P, Jones R, Paisley S, Carroll C.
-
A systematic review and economic evaluation of epoetin alfa, epoetin beta and darbepoetin alfa in anaemia associated with cancer, especially that attributable to cancer treatment.
By Wilson J, Yao GL, Raftery J, Bohlius J, Brunskill S, Sandercock J, et al.
-
A systematic review and economic evaluation of statins for the prevention of coronary events.
By Ward S, Lloyd Jones M, Pandor A, Holmes M, Ara R, Ryan A, et al.
-
A systematic review of the effectiveness and cost-effectiveness of different models of community-based respite care for frail older people and their carers.
By Mason A, Weatherly H, Spilsbury K, Arksey H, Golder S, Adamson J, et al.
-
Additional therapy for young children with spastic cerebral palsy: a randomised controlled trial.
By Weindling AM, Cunningham CC, Glenn SM, Edwards RT, Reeves DJ.
-
Screening for type 2 diabetes: literature review and economic modelling.
By Waugh N, Scotland G, McNamee P, Gillett M, Brennan A, Goyder E, et al.
-
The effectiveness and cost-effectiveness of cinacalcet for secondary hyperparathyroidism in end-stage renal disease patients on dialysis: a systematic review and economic evaluation.
By Garside R, Pitt M, Anderson R, Mealing S, Roome C, Snaith A, et al.
-
The clinical effectiveness and cost-effectiveness of gemcitabine for metastatic breast cancer: a systematic review and economic evaluation.
By Takeda AL, Jones J, Loveman E, Tan SC, Clegg AJ.
-
A systematic review of duplex ultrasound, magnetic resonance angiography and computed tomography angiography for the diagnosis and assessment of symptomatic, lower limb peripheral arterial disease.
By Collins R, Cranny G, Burch J, Aguiar-Ibáñez R, Craig D, Wright K, et al.
-
The clinical effectiveness and cost-effectiveness of treatments for children with idiopathic steroid-resistant nephrotic syndrome: a systematic review.
By Colquitt JL, Kirby J, Green C, Cooper K, Trompeter RS.
-
A systematic review of the routine monitoring of growth in children of primary school age to identify growth-related conditions.
By Fayter D, Nixon J, Hartley S, Rithalia A, Butler G, Rudolf M, et al.
-
Systematic review of the effectiveness of preventing and treating Staphylococcus aureus carriage in reducing peritoneal catheter-related infections.
By McCormack K, Rabindranath K, Kilonzo M, Vale L, Fraser C, McIntyre L, et al.
-
The clinical effectiveness and cost of repetitive transcranial magnetic stimulation versus electroconvulsive therapy in severe depression: a multicentre pragmatic randomised controlled trial and economic analysis.
By McLoughlin DM, Mogg A, Eranti S, Pluck G, Purvis R, Edwards D, et al.
-
A randomised controlled trial and economic evaluation of direct versus indirect and individual versus group modes of speech and language therapy for children with primary language impairment.
By Boyle J, McCartney E, Forbes J, O’Hare A.
-
Hormonal therapies for early breast cancer: systematic review and economic evaluation.
By Hind D, Ward S, De Nigris E, Simpson E, Carroll C, Wyld L.
-
Cardioprotection against the toxic effects of anthracyclines given to children with cancer: a systematic review.
By Bryant J, Picot J, Levitt G, Sullivan I, Baxter L, Clegg A.
-
Adalimumab, etanercept and infliximab for the treatment of ankylosing spondylitis: a systematic review and economic evaluation.
By McLeod C, Bagust A, Boland A, Dagenais P, Dickson R, Dundar Y, et al.
-
Prenatal screening and treatment strategies to prevent group B streptococcal and other bacterial infections in early infancy: cost-effectiveness and expected value of information analyses.
By Colbourn T, Asseburg C, Bojke L, Philips Z, Claxton K, Ades AE, et al.
-
Clinical effectiveness and cost-effectiveness of bone morphogenetic proteins in the non-healing of fractures and spinal fusion: a systematic review.
By Garrison KR, Donell S, Ryder J, Shemilt I, Mugford M, Harvey I, et al.
-
A randomised controlled trial of postoperative radiotherapy following breast-conserving surgery in a minimum-risk older population. The PRIME trial.
By Prescott RJ, Kunkler IH, Williams LJ, King CC, Jack W, van der Pol M, et al.
-
Current practice, accuracy, effectiveness and cost-effectiveness of the school entry hearing screen.
By Bamford J, Fortnum H, Bristow K, Smith J, Vamvakas G, Davies L, et al.
-
The clinical effectiveness and cost-effectiveness of inhaled insulin in diabetes mellitus: a systematic review and economic evaluation.
By Black C, Cummins E, Royle P, Philip S, Waugh N.
-
Surveillance of cirrhosis for hepatocellular carcinoma: systematic review and economic analysis.
By Thompson Coon J, Rogers G, Hewson P, Wright D, Anderson R, Cramp M, et al.
-
The Birmingham Rehabilitation Uptake Maximisation Study (BRUM). Homebased compared with hospital-based cardiac rehabilitation in a multi-ethnic population: cost-effectiveness and patient adherence.
By Jolly K, Taylor R, Lip GYH, Greenfield S, Raftery J, Mant J, et al.
-
A systematic review of the clinical, public health and cost-effectiveness of rapid diagnostic tests for the detection and identification of bacterial intestinal pathogens in faeces and food.
By Abubakar I, Irvine L, Aldus CF, Wyatt GM, Fordham R, Schelenz S, et al.
-
A randomised controlled trial examining the longer-term outcomes of standard versus new antiepileptic drugs. The SANAD trial.
By Marson AG, Appleton R, Baker GA, Chadwick DW, Doughty J, Eaton B, et al.
-
Clinical effectiveness and cost-effectiveness of different models of managing long-term oral anti-coagulation therapy: a systematic review and economic modelling.
By Connock M, Stevens C, Fry-Smith A, Jowett S, Fitzmaurice D, Moore D, et al.
-
A systematic review and economic model of the clinical effectiveness and cost-effectiveness of interventions for preventing relapse in people with bipolar disorder.
By Soares-Weiser K, Bravo Vergel Y, Beynon S, Dunn G, Barbieri M, Duffy S, et al.
-
Taxanes for the adjuvant treatment of early breast cancer: systematic review and economic evaluation.
By Ward S, Simpson E, Davis S, Hind D, Rees A, Wilkinson A.
-
The clinical effectiveness and cost-effectiveness of screening for open angle glaucoma: a systematic review and economic evaluation.
By Burr JM, Mowatt G, Hernández R, Siddiqui MAR, Cook J, Lourenco T, et al.
-
Acceptability, benefit and costs of early screening for hearing disability: a study of potential screening tests and models.
By Davis A, Smith P, Ferguson M, Stephens D, Gianopoulos I.
-
Contamination in trials of educational interventions.
By Keogh-Brown MR, Bachmann MO, Shepstone L, Hewitt C, Howe A, Ramsay CR, et al.
-
Overview of the clinical effectiveness of positron emission tomography imaging in selected cancers.
By Facey K, Bradbury I, Laking G, Payne E.
-
The effectiveness and cost-effectiveness of carmustine implants and temozolomide for the treatment of newly diagnosed high-grade glioma: a systematic review and economic evaluation.
By Garside R, Pitt M, Anderson R, Rogers G, Dyer M, Mealing S, et al.
-
Drug-eluting stents: a systematic review and economic evaluation.
By Hill RA, Boland A, Dickson R, Dündar Y, Haycox A, McLeod C, et al.
-
The clinical effectiveness and cost-effectiveness of cardiac resynchronisation (biventricular pacing) for heart failure: systematic review and economic model.
By Fox M, Mealing S, Anderson R, Dean J, Stein K, Price A, et al.
-
Recruitment to randomised trials: strategies for trial enrolment and participation study. The STEPS study.
By Campbell MK, Snowdon C, Francis D, Elbourne D, McDonald AM, Knight R, et al.
-
Cost-effectiveness of functional cardiac testing in the diagnosis and management of coronary artery disease: a randomised controlled trial. The CECaT trial.
By Sharples L, Hughes V, Crean A, Dyer M, Buxton M, Goldsmith K, et al.
-
Evaluation of diagnostic tests when there is no gold standard. A review of methods.
By Rutjes AWS, Reitsma JB, Coomarasamy A, Khan KS, Bossuyt PMM.
-
Systematic reviews of the clinical effectiveness and cost-effectiveness of proton pump inhibitors in acute upper gastrointestinal bleeding.
By Leontiadis GI, Sreedharan A, Dorward S, Barton P, Delaney B, Howden CW, et al.
-
A review and critique of modelling in prioritising and designing screening programmes.
By Karnon J, Goyder E, Tappenden P, McPhie S, Towers I, Brazier J, et al.
-
An assessment of the impact of the NHS Health Technology Assessment Programme.
By Hanney S, Buxton M, Green C, Coulson D, Raftery J.
-
A systematic review and economic model of switching from nonglycopeptide to glycopeptide antibiotic prophylaxis for surgery.
By Cranny G, Elliott R, Weatherly H, Chambers D, Hawkins N, Myers L, et al.
-
‘Cut down to quit’ with nicotine replacement therapies in smoking cessation: a systematic review of effectiveness and economic analysis.
By Wang D, Connock M, Barton P, Fry-Smith A, Aveyard P, Moore D.
-
A systematic review of the effectiveness of strategies for reducing fracture risk in children with juvenile idiopathic arthritis with additional data on long-term risk of fracture and cost of disease management.
By Thornton J, Ashcroft D, O’Neill T, Elliott R, Adams J, Roberts C, et al.
-
Does befriending by trained lay workers improve psychological well-being and quality of life for carers of people with dementia, and at what cost? A randomised controlled trial.
By Charlesworth G, Shepstone L, Wilson E, Thalanany M, Mugford M, Poland F.
-
A multi-centre retrospective cohort study comparing the efficacy, safety and cost-effectiveness of hysterectomy and uterine artery embolisation for the treatment of symptomatic uterine fibroids. The HOPEFUL study.
By Hirst A, Dutton S, Wu O, Briggs A, Edwards C, Waldenmaier L, et al.
-
Methods of prediction and prevention of pre-eclampsia: systematic reviews of accuracy and effectiveness literature with economic modelling.
By Meads CA, Cnossen JS, Meher S, Juarez-Garcia A, ter Riet G, Duley L, et al.
-
The use of economic evaluations in NHS decision-making: a review and empirical investigation.
By Williams I, McIver S, Moore D, Bryan S.
-
Stapled haemorrhoidectomy (haemorrhoidopexy) for the treatment of haemorrhoids: a systematic review and economic evaluation.
By Burch J, Epstein D, Baba-Akbari A, Weatherly H, Fox D, Golder S, et al.
-
The clinical effectiveness of diabetes education models for Type 2 diabetes: a systematic review.
By Loveman E, Frampton GK, Clegg AJ.
-
Payment to healthcare professionals for patient recruitment to trials: systematic review and qualitative study.
By Raftery J, Bryant J, Powell J, Kerr C, Hawker S.
-
Cyclooxygenase-2 selective non-steroidal anti-inflammatory drugs (etodolac, meloxicam, celecoxib, rofecoxib, etoricoxib, valdecoxib and lumiracoxib) for osteoarthritis and rheumatoid arthritis: a systematic review and economic evaluation.
By Chen Y-F, Jobanputra P, Barton P, Bryan S, Fry-Smith A, Harris G, et al.
-
The clinical effectiveness and cost-effectiveness of central venous catheters treated with anti-infective agents in preventing bloodstream infections: a systematic review and economic evaluation.
By Hockenhull JC, Dwan K, Boland A, Smith G, Bagust A, Dundar Y, et al.
-
Stepped treatment of older adults on laxatives. The STOOL trial.
By Mihaylov S, Stark C, McColl E, Steen N, Vanoli A, Rubin G, et al.
-
A randomised controlled trial of cognitive behaviour therapy in adolescents with major depression treated by selective serotonin reuptake inhibitors. The ADAPT trial.
By Goodyer IM, Dubicka B, Wilkinson P, Kelvin R, Roberts C, Byford S, et al.
-
The use of irinotecan, oxaliplatin and raltitrexed for the treatment of advanced colorectal cancer: systematic review and economic evaluation.
By Hind D, Tappenden P, Tumur I, Eggington E, Sutcliffe P, Ryan A.
-
Ranibizumab and pegaptanib for the treatment of age-related macular degeneration: a systematic review and economic evaluation.
By Colquitt JL, Jones J, Tan SC, Takeda A, Clegg AJ, Price A.
-
Systematic review of the clinical effectiveness and cost-effectiveness of 64-slice or higher computed tomography angiography as an alternative to invasive coronary angiography in the investigation of coronary artery disease.
By Mowatt G, Cummins E, Waugh N, Walker S, Cook J, Jia X, et al.
-
Structural neuroimaging in psychosis: a systematic review and economic evaluation.
By Albon E, Tsourapas A, Frew E, Davenport C, Oyebode F, Bayliss S, et al.
-
Systematic review and economic analysis of the comparative effectiveness of different inhaled corticosteroids and their usage with long-acting beta2 agonists for the treatment of chronic asthma in adults and children aged 12 years and over.
By Shepherd J, Rogers G, Anderson R, Main C, Thompson-Coon J, Hartwell D, et al.
-
Systematic review and economic analysis of the comparative effectiveness of different inhaled corticosteroids and their usage with long-acting beta2 agonists for the treatment of chronic asthma in children under the age of 12 years.
By Main C, Shepherd J, Anderson R, Rogers G, Thompson-Coon J, Liu Z, et al.
-
Ezetimibe for the treatment of hypercholesterolaemia: a systematic review and economic evaluation.
By Ara R, Tumur I, Pandor A, Duenas A, Williams R, Wilkinson A, et al.
-
Topical or oral ibuprofen for chronic knee pain in older people. The TOIB study.
By Underwood M, Ashby D, Carnes D, Castelnuovo E, Cross P, Harding G, et al.
-
A prospective randomised comparison of minor surgery in primary and secondary care. The MiSTIC trial.
By George S, Pockney P, Primrose J, Smith H, Little P, Kinley H, et al.
-
A review and critical appraisal of measures of therapist–patient interactions in mental health settings.
By Cahill J, Barkham M, Hardy G, Gilbody S, Richards D, Bower P, et al.
-
The clinical effectiveness and cost-effectiveness of screening programmes for amblyopia and strabismus in children up to the age of 4–5 years: a systematic review and economic evaluation.
By Carlton J, Karnon J, Czoski-Murray C, Smith KJ, Marr J.
-
A systematic review of the clinical effectiveness and cost-effectiveness and economic modelling of minimal incision total hip replacement approaches in the management of arthritic disease of the hip.
By de Verteuil R, Imamura M, Zhu S, Glazener C, Fraser C, Munro N, et al.
-
A preliminary model-based assessment of the cost–utility of a screening programme for early age-related macular degeneration.
By Karnon J, Czoski-Murray C, Smith K, Brand C, Chakravarthy U, Davis S, et al.
-
Intravenous magnesium sulphate and sotalol for prevention of atrial fibrillation after coronary artery bypass surgery: a systematic review and economic evaluation.
By Shepherd J, Jones J, Frampton GK, Tanajewski L, Turner D, Price A.
-
Absorbent products for urinary/faecal incontinence: a comparative evaluation of key product categories.
By Fader M, Cottenden A, Getliffe K, Gage H, Clarke-O’Neill S, Jamieson K, et al.
-
A systematic review of repetitive functional task practice with modelling of resource use, costs and effectiveness.
By French B, Leathley M, Sutton C, McAdam J, Thomas L, Forster A, et al.
-
The effectiveness and cost-effectivness of minimal access surgery amongst people with gastro-oesophageal reflux disease – a UK collaborative study. The reflux trial.
By Grant A, Wileman S, Ramsay C, Bojke L, Epstein D, Sculpher M, et al.
-
Time to full publication of studies of anti-cancer medicines for breast cancer and the potential for publication bias: a short systematic review.
By Takeda A, Loveman E, Harris P, Hartwell D, Welch K.
-
Performance of screening tests for child physical abuse in accident and emergency departments.
By Woodman J, Pitt M, Wentz R, Taylor B, Hodes D, Gilbert RE.
-
Curative catheter ablation in atrial fibrillation and typical atrial flutter: systematic review and economic evaluation.
By Rodgers M, McKenna C, Palmer S, Chambers D, Van Hout S, Golder S, et al.
-
Systematic review and economic modelling of effectiveness and cost utility of surgical treatments for men with benign prostatic enlargement.
By Lourenco T, Armstrong N, N’Dow J, Nabi G, Deverill M, Pickard R, et al.
-
Immunoprophylaxis against respiratory syncytial virus (RSV) with palivizumab in children: a systematic review and economic evaluation.
By Wang D, Cummins C, Bayliss S, Sandercock J, Burls A.
-
Deferasirox for the treatment of iron overload associated with regular blood transfusions (transfusional haemosiderosis) in patients suffering with chronic anaemia: a systematic review and economic evaluation.
By McLeod C, Fleeman N, Kirkham J, Bagust A, Boland A, Chu P, et al.
-
Thrombophilia testing in people with venous thromboembolism: systematic review and cost-effectiveness analysis.
By Simpson EL, Stevenson MD, Rawdin A, Papaioannou D.
-
Surgical procedures and non-surgical devices for the management of non-apnoeic snoring: a systematic review of clinical effects and associated treatment costs.
By Main C, Liu Z, Welch K, Weiner G, Quentin Jones S, Stein K.
-
Continuous positive airway pressure devices for the treatment of obstructive sleep apnoea–hypopnoea syndrome: a systematic review and economic analysis.
By McDaid C, Griffin S, Weatherly H, Durée K, van der Burgt M, van Hout S, Akers J, et al.
-
Use of classical and novel biomarkers as prognostic risk factors for localised prostate cancer: a systematic review.
By Sutcliffe P, Hummel S, Simpson E, Young T, Rees A, Wilkinson A, et al.
-
The harmful health effects of recreational ecstasy: a systematic review of observational evidence.
By Rogers G, Elston J, Garside R, Roome C, Taylor R, Younger P, et al.
-
Systematic review of the clinical effectiveness and cost-effectiveness of oesophageal Doppler monitoring in critically ill and high-risk surgical patients.
By Mowatt G, Houston G, Hernández R, de Verteuil R, Fraser C, Cuthbertson B, et al.
-
The use of surrogate outcomes in model-based cost-effectiveness analyses: a survey of UK Health Technology Assessment reports.
By Taylor RS, Elston J.
-
Controlling Hypertension and Hypotension Immediately Post Stroke (CHHIPS) – a randomised controlled trial.
By Potter J, Mistri A, Brodie F, Chernova J, Wilson E, Jagger C, et al.
-
Routine antenatal anti-D prophylaxis for RhD-negative women: a systematic review and economic evaluation.
By Pilgrim H, Lloyd-Jones M, Rees A.
Health Technology Assessment Programme
-
Director, NIHR HTA Programme, Professor of Clinical Pharmacology, University of Liverpool
-
Director, Medical Care Research Unit, University of Sheffield
Prioritisation Strategy Group
-
Director, NIHR HTA Programme, Professor of Clinical Pharmacology, University of Liverpool
-
Director, Medical Care Research Unit, University of Sheffield
-
Dr Bob Coates, Consultant Advisor, NCCHTA
-
Dr Andrew Cook, Consultant Advisor, NCCHTA
-
Dr Peter Davidson, Director of Science Support, NCCHTA
-
Professor Robin E Ferner, Consultant Physician and Director, West Midlands Centre for Adverse Drug Reactions, City Hospital NHS Trust, Birmingham
-
Professor Paul Glasziou, Professor of Evidence-Based Medicine, University of Oxford
-
Dr Nick Hicks, Director of NHS Support, NCCHTA
-
Dr Edmund Jessop, Medical Adviser, National Specialist, National Commissioning Group (NCG), Department of Health, London
-
Ms Lynn Kerridge, Chief Executive Officer, NETSCC and NCCHTA
-
Dr Ruairidh Milne, Director of Strategy and Development, NETSCC
-
Ms Kay Pattison, Section Head, NHS R&D Programme, Department of Health
-
Ms Pamela Young, Specialist Programme Manager, NCCHTA
HTA Commissioning Board
-
Director, NIHR HTA Programme, Professor of Clinical Pharmacology, University of Liverpool
-
Director, Medical Care Research Unit, University of Sheffield
-
Senior Lecturer in General Practice, Department of Primary Health Care, University of Oxford
-
Professor Ann Ashburn, Professor of Rehabilitation and Head of Research, Southampton General Hospital
-
Professor Deborah Ashby, Professor of Medical Statistics, Queen Mary, University of London
-
Professor John Cairns, Professor of Health Economics, London School of Hygiene and Tropical Medicine
-
Professor Peter Croft, Director of Primary Care Sciences Research Centre, Keele University
-
Professor Nicky Cullum, Director of Centre for Evidence-Based Nursing, University of York
-
Professor Jenny Donovan, Professor of Social Medicine, University of Bristol
-
Professor Steve Halligan, Professor of Gastrointestinal Radiology, University College Hospital, London
-
Professor Freddie Hamdy, Professor of Urology, University of Sheffield
-
Professor Allan House, Professor of Liaison Psychiatry, University of Leeds
-
Dr Martin J Landray, Reader in Epidemiology, Honorary Consultant Physician, Clinical Trial Service Unit, University of Oxford?
-
Professor Stuart Logan, Director of Health & Social Care Research, The Peninsula Medical School, Universities of Exeter and Plymouth
-
Dr Rafael Perera, Lecturer in Medical Statisitics, Department of Primary Health Care, Univeristy of Oxford
-
Professor Ian Roberts, Professor of Epidemiology & Public Health, London School of Hygiene and Tropical Medicine
-
Professor Mark Sculpher, Professor of Health Economics, University of York
-
Professor Helen Smith, Professor of Primary Care, University of Brighton
-
Professor Kate Thomas, Professor of Complementary & Alternative Medicine Research, University of Leeds
-
Professor David John Torgerson, Director of York Trials Unit, University of York
-
Professor Hywel Williams, Professor of Dermato-Epidemiology, University of Nottingham
-
Ms Kay Pattison, Section Head, NHS R&D Programmes, Research and Development Directorate, Department of Health
-
Dr Morven Roberts, Clinical Trials Manager, Medical Research Council
Diagnostic Technologies & Screening Panel
-
Professor of Evidence-Based Medicine, University of Oxford
-
Consultant Paediatrician and Honorary Senior Lecturer, Great Ormond Street Hospital, London
-
Professor Judith E Adams, Consultant Radiologist, Manchester Royal Infirmary, Central Manchester & Manchester Children’s University Hospitals NHS Trust, and Professor of Diagnostic Radiology, Imaging Science and Biomedical Engineering, Cancer & Imaging Sciences, University of Manchester
-
Ms Jane Bates, Consultant Ultrasound Practitioner, Ultrasound Department, Leeds Teaching Hospital NHS Trust
-
Dr Stephanie Dancer, Consultant Microbiologist, Hairmyres Hospital, East Kilbride
-
Professor Glyn Elwyn, Primary Medical Care Research Group, Swansea Clinical School, University of Wales
-
Dr Ron Gray, Consultant Clinical Epidemiologist, Department of Public Health, University of Oxford
-
Professor Paul D Griffiths, Professor of Radiology, University of Sheffield
-
Dr Jennifer J Kurinczuk, Consultant Clinical Epidemiologist, National Perinatal Epidemiology Unit, Oxford
-
Dr Susanne M Ludgate, Medical Director, Medicines & Healthcare Products Regulatory Agency, London
-
Dr Anne Mackie, Director of Programmes, UK National Screening Committee
-
Dr Michael Millar, Consultant Senior Lecturer in Microbiology, Barts and The London NHS Trust, Royal London Hospital
-
Mr Stephen Pilling, Director, Centre for Outcomes, Research & Effectiveness, Joint Director, National Collaborating Centre for Mental Health, University College London
-
Mrs Una Rennard, Service User Representative
-
Dr Phil Shackley, Senior Lecturer in Health Economics, School of Population and Health Sciences, University of Newcastle upon Tyne
-
Dr W Stuart A Smellie, Consultant in Chemical Pathology, Bishop Auckland General Hospital
-
Dr Nicholas Summerton, Consultant Clinical and Public Health Advisor, NICE
-
Ms Dawn Talbot, Service User Representative
-
Dr Graham Taylor, Scientific Advisor, Regional DNA Laboratory, St James’s University Hospital, Leeds
-
Professor Lindsay Wilson Turnbull, Scientific Director of the Centre for Magnetic Resonance Investigations and YCR Professor of Radiology, Hull Royal Infirmary
-
Dr Tim Elliott, Team Leader, Cancer Screening, Department of Health
-
Dr Catherine Moody, Programme Manager, Neuroscience and Mental Health Board
-
Dr Ursula Wells, Principal Research Officer, Department of Health
Pharmaceuticals Panel
-
Consultant Physician and Director, West Midlands Centre for Adverse Drug Reactions, City Hospital NHS Trust, Birmingham
-
Professor in Child Health, University of Nottingham
-
Mrs Nicola Carey, Senior Research Fellow, School of Health and Social Care, The University of Reading
-
Mr John Chapman, Service User Representative
-
Dr Peter Elton, Director of Public Health, Bury Primary Care Trust
-
Dr Ben Goldacre, Research Fellow, Division of Psychological Medicine and Psychiatry, King’s College London
-
Mrs Barbara Greggains, Service User Representative
-
Dr Bill Gutteridge, Medical Adviser, London Strategic Health Authority
-
Dr Dyfrig Hughes, Reader in Pharmacoeconomics and Deputy Director, Centre for Economics and Policy in Health, IMSCaR, Bangor University
-
Professor Jonathan Ledermann, Professor of Medical Oncology and Director of the Cancer Research UK and University College London Cancer Trials Centre
-
Dr Yoon K Loke, Senior Lecturer in Clinical Pharmacology, University of East Anglia
-
Professor Femi Oyebode, Consultant Psychiatrist and Head of Department, University of Birmingham
-
Dr Andrew Prentice, Senior Lecturer and Consultant Obstetrician and Gynaecologist, The Rosie Hospital, University of Cambridge
-
Dr Martin Shelly, General Practitioner, Leeds, and Associate Director, NHS Clinical Governance Support Team, Leicester
-
Dr Gillian Shepherd, Director, Health and Clinical Excellence, Merck Serono Ltd
-
Mrs Katrina Simister, Assistant Director New Medicines, National Prescribing Centre, Liverpool
-
Mr David Symes, Service User Representative
-
Dr Lesley Wise, Unit Manager, Pharmacoepidemiology Research Unit, VRMM, Medicines & Healthcare Products Regulatory Agency
-
Ms Kay Pattison, Section Head, NHS R&D Programme, Department of Health
-
Mr Simon Reeve, Head of Clinical and Cost-Effectiveness, Medicines, Pharmacy and Industry Group, Department of Health
-
Dr Heike Weber, Programme Manager, Medical Research Council
-
Dr Ursula Wells, Principal Research Officer, Department of Health
Therapeutic Procedures Panel
-
Consultant Physician, North Bristol NHS Trust
-
Professor of Psychiatry, Division of Health in the Community, University of Warwick, Coventry
-
Professor Jane Barlow, Professor of Public Health in the Early Years, Health Sciences Research Institute, Warwick Medical School, Coventry
-
Ms Maree Barnett, Acting Branch Head of Vascular Programme, Department of Health
-
Mrs Val Carlill, Service User Representative
-
Mrs Anthea De Barton-Watson, Service User Representative
-
Mr Mark Emberton, Senior Lecturer in Oncological Urology, Institute of Urology, University College Hospital, London
-
Professor Steve Goodacre, Professor of Emergency Medicine, University of Sheffield
-
Professor Christopher Griffiths, Professor of Primary Care, Barts and The London School of Medicine and Dentistry
-
Mr Paul Hilton, Consultant Gynaecologist and Urogynaecologist, Royal Victoria Infirmary, Newcastle upon Tyne
-
Professor Nicholas James, Professor of Clinical Oncology, University of Birmingham, and Consultant in Clinical Oncology, Queen Elizabeth Hospital
-
Dr Peter Martin, Consultant Neurologist, Addenbrooke’s Hospital, Cambridge
-
Dr Kate Radford, Senior Lecturer (Research), Clinical Practice Research Unit, University of Central Lancashire, Preston
-
Mr Jim Reece Service User Representative
-
Dr Karen Roberts, Nurse Consultant, Dunston Hill Hospital Cottages
-
Dr Phillip Leech, Principal Medical Officer for Primary Care, Department of Health
-
Ms Kay Pattison, Section Head, NHS R&D Programme, Department of Health
-
Dr Morven Roberts, Clinical Trials Manager, Medical Research Council
-
Professor Tom Walley, Director, NIHR HTA Programme, Professor of Clinical Pharmacology, University of Liverpool
-
Dr Ursula Wells, Principal Research Officer, Department of Health
Disease Prevention Panel
-
Medical Adviser, National Specialist, National Commissioning Group (NCG), London
-
Director, NHS Sustainable Development Unit, Cambridge
-
Dr Elizabeth Fellow-Smith, Medical Director, West London Mental Health Trust, Middlesex
-
Dr John Jackson, General Practitioner, Parkway Medical Centre, Newcastle upon Tyne
-
Professor Mike Kelly, Director, Centre for Public Health Excellence, NICE, London
-
Dr Chris McCall, General Practitioner, The Hadleigh Practice, Corfe Mullen, Dorset
-
Ms Jeanett Martin, Director of Nursing, BarnDoc Limited, Lewisham Primary Care Trust
-
Dr Julie Mytton, Locum Consultant in Public Health Medicine, Bristol Primary Care Trust
-
Miss Nicky Mullany, Service User Representative
-
Professor Ian Roberts, Professor of Epidemiology and Public Health, London School of Hygiene & Tropical Medicine
-
Professor Ken Stein, Senior Clinical Lecturer in Public Health, University of Exeter
-
Dr Kieran Sweeney, Honorary Clinical Senior Lecturer, Peninsula College of Medicine and Dentistry, Universities of Exeter and Plymouth
-
Professor Carol Tannahill, Glasgow Centre for Population Health
-
Professor Margaret Thorogood, Professor of Epidemiology, University of Warwick Medical School, Coventry
-
Ms Christine McGuire, Research & Development, Department of Health
-
Dr Caroline Stone, Programme Manager, Medical Research Council
Expert Advisory Network
-
Professor Douglas Altman, Professor of Statistics in Medicine, Centre for Statistics in Medicine, University of Oxford
-
Professor John Bond, Professor of Social Gerontology & Health Services Research, University of Newcastle upon Tyne
-
Professor Andrew Bradbury, Professor of Vascular Surgery, Solihull Hospital, Birmingham
-
Mr Shaun Brogan, Chief Executive, Ridgeway Primary Care Group, Aylesbury
-
Mrs Stella Burnside OBE, Chief Executive, Regulation and Improvement Authority, Belfast
-
Ms Tracy Bury, Project Manager, World Confederation for Physical Therapy, London
-
Professor Iain T Cameron, Professor of Obstetrics and Gynaecology and Head of the School of Medicine, University of Southampton
-
Dr Christine Clark, Medical Writer and Consultant Pharmacist, Rossendale
-
Professor Collette Clifford, Professor of Nursing and Head of Research, The Medical School, University of Birmingham
-
Professor Barry Cookson, Director, Laboratory of Hospital Infection, Public Health Laboratory Service, London
-
Dr Carl Counsell, Clinical Senior Lecturer in Neurology, University of Aberdeen
-
Professor Howard Cuckle, Professor of Reproductive Epidemiology, Department of Paediatrics, Obstetrics & Gynaecology, University of Leeds
-
Dr Katherine Darton, Information Unit, MIND – The Mental Health Charity, London
-
Professor Carol Dezateux, Professor of Paediatric Epidemiology, Institute of Child Health, London
-
Mr John Dunning, Consultant Cardiothoracic Surgeon, Papworth Hospital NHS Trust, Cambridge
-
Mr Jonothan Earnshaw, Consultant Vascular Surgeon, Gloucestershire Royal Hospital, Gloucester
-
Professor Martin Eccles, Professor of Clinical Effectiveness, Centre for Health Services Research, University of Newcastle upon Tyne
-
Professor Pam Enderby, Dean of Faculty of Medicine, Institute of General Practice and Primary Care, University of Sheffield
-
Professor Gene Feder, Professor of Primary Care Research & Development, Centre for Health Sciences, Barts and The London School of Medicine and Dentistry
-
Mr Leonard R Fenwick, Chief Executive, Freeman Hospital, Newcastle upon Tyne
-
Mrs Gillian Fletcher, Antenatal Teacher and Tutor and President, National Childbirth Trust, Henfield
-
Professor Jayne Franklyn, Professor of Medicine, University of Birmingham
-
Mr Tam Fry, Honorary Chairman, Child Growth Foundation, London
-
Professor Fiona Gilbert, Consultant Radiologist and NCRN Member, University of Aberdeen
-
Professor Paul Gregg, Professor of Orthopaedic Surgical Science, South Tees Hospital NHS Trust
-
Bec Hanley, Co-director, TwoCan Associates, West Sussex
-
Dr Maryann L Hardy, Senior Lecturer, University of Bradford
-
Mrs Sharon Hart, Healthcare Management Consultant, Reading
-
Professor Robert E Hawkins, CRC Professor and Director of Medical Oncology, Christie CRC Research Centre, Christie Hospital NHS Trust, Manchester
-
Professor Richard Hobbs, Head of Department of Primary Care & General Practice, University of Birmingham
-
Professor Alan Horwich, Dean and Section Chairman, The Institute of Cancer Research, London
-
Professor Allen Hutchinson, Director of Public Health and Deputy Dean of ScHARR, University of Sheffield
-
Professor Peter Jones, Professor of Psychiatry, University of Cambridge, Cambridge
-
Professor Stan Kaye, Cancer Research UK Professor of Medical Oncology, Royal Marsden Hospital and Institute of Cancer Research, Surrey
-
Dr Duncan Keeley, General Practitioner (Dr Burch & Ptnrs), The Health Centre, Thame
-
Dr Donna Lamping, Research Degrees Programme Director and Reader in Psychology, Health Services Research Unit, London School of Hygiene and Tropical Medicine, London
-
Mr George Levvy, Chief Executive, Motor Neurone Disease Association, Northampton
-
Professor James Lindesay, Professor of Psychiatry for the Elderly, University of Leicester
-
Professor Julian Little, Professor of Human Genome Epidemiology, University of Ottawa
-
Professor Alistaire McGuire, Professor of Health Economics, London School of Economics
-
Professor Rajan Madhok, Medical Director and Director of Public Health, Directorate of Clinical Strategy & Public Health, North & East Yorkshire & Northern Lincolnshire Health Authority, York
-
Professor Alexander Markham, Director, Molecular Medicine Unit, St James’s University Hospital, Leeds
-
Dr Peter Moore, Freelance Science Writer, Ashtead
-
Dr Andrew Mortimore, Public Health Director, Southampton City Primary Care Trust
-
Dr Sue Moss, Associate Director, Cancer Screening Evaluation Unit, Institute of Cancer Research, Sutton
-
Professor Miranda Mugford, Professor of Health Economics and Group Co-ordinator, University of East Anglia
-
Professor Jim Neilson, Head of School of Reproductive & Developmental Medicine and Professor of Obstetrics and Gynaecology, University of Liverpool
-
Mrs Julietta Patnick, National Co-ordinator, NHS Cancer Screening Programmes, Sheffield
-
Professor Robert Peveler, Professor of Liaison Psychiatry, Royal South Hants Hospital, Southampton
-
Professor Chris Price, Director of Clinical Research, Bayer Diagnostics Europe, Stoke Poges
-
Professor William Rosenberg, Professor of Hepatology and Consultant Physician, University of Southampton
-
Professor Peter Sandercock, Professor of Medical Neurology, Department of Clinical Neurosciences, University of Edinburgh
-
Dr Susan Schonfield, Consultant in Public Health, Hillingdon Primary Care Trust, Middlesex
-
Dr Eamonn Sheridan, Consultant in Clinical Genetics, St James’s University Hospital, Leeds
-
Dr Margaret Somerville, Director of Public Health Learning, Peninsula Medical School, University of Plymouth
-
Professor Sarah Stewart-Brown, Professor of Public Health, Division of Health in the Community, University of Warwick, Coventry
-
Professor Ala Szczepura, Professor of Health Service Research, Centre for Health Services Studies, University of Warwick, Coventry
-
Mrs Joan Webster, Consumer Member, Southern Derbyshire Community Health Council
-
Professor Martin Whittle, Clinical Co-director, National Co-ordinating Centre for Women’s and Children’s Health, Lymington