Notes
Article history
The research reported in this issue of the journal was commissioned by the HTA programme as project number 02/41/06. The contractual start date was in February 2005. The draft report began editorial review in May 2009 and was accepted for publication in January 2010. As the funder, by devising a commissioning brief, the HTA programme specified the research question and study design. 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
Ipsen Ltd provided the botulinum toxin type A used by the study free of charge. They also provided sponsorship for launch meetings at study sites. The design, analysis and reporting of the study were undertaken independently of Ipsen Ltd. LS, HR, CP, FvW, GF, PS and NS have no competing interests. MB and LG use botulinum toxin regularly in clinical practice. They have received sponsorship from Ipsen Ltd to attend and teach at conferences and meetings, but have no personal financial interest in botulinum toxin or any related product.
Permissions
Copyright statement
© 2010 Queen’s Printer and Controller of HMSO. This journal is a member of and subscribes to the principles of the Committee on Publication Ethics (COPE) (http://www.publicationethics.org/). This journal 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: NETSCC, Health Technology Assessment, Alpha House, University of Southampton Science Park, Southampton SO16 7NS, UK.
2010 Queen’s Printer and Controller of HMSO
Chapter 1 Introduction
The National Institute for Health Research (NIHR) Health Technology Assessment programme identified the need to evaluate the clinical effectiveness and cost-effectiveness of using botulinum toxin to treat chronic upper limb spasticity due to stroke in adults. This report describes the work commissioned to address this issue.
Overview of stroke
Stroke is a major cause of death and disability in the UK. In England, over 900,000 people are living with the consequences of stroke, 300,000 of them are moderately or severely disabled. 1 The direct cost of stroke to the National Health Service (NHS) is £2.8B per annum, although the overall cost to the economy is much higher (£7B per annum) once informal care costs and lost productivity are taken into account. 1
Upper limb problems following stroke
Upper limb impairments such as muscle weakness, spasticity, poor co-ordination and sensory disturbance are common after stroke. These impairments alone, or in combination, can result in a range of functional limitations. Between 50% and 70% of stroke patients have long-term upper limb functional limitations2–4 and many feel that insufficient attention is paid to upper limb rehabilitation. 5 In contrast, about 80% of stroke survivors are able to walk again. 6
Spasticity
Spasticity is traditionally defined as ‘a motor disorder characterised by a velocity-dependent increase in tonic stretch reflexes (muscle tone) with exaggerated tendon jerks, resulting from hyperexcitability of the stretch reflex, as one component of the upper motor neurone syndrome. 7 A recent definition, which is broader and perhaps more clinically relevant, is ‘disordered sensori-motor control, resulting from an upper motor neurone lesion, presenting as intermittent or sustained involuntary activation of muscles’. 8 However, put simply, spasticity is involuntary overactivity of muscles as a result of damage to the brain or spinal cord.
Spasticity may cause reduced function, pain and deformity, and in the longer term may lead to the development of contracture. 9 Patients with upper limb spasticity can develop abnormal limb posturing such as the classic adducted internally rotated shoulder, flexed elbow, flexed wrist and clenched fist10 (Figure 1). These positions can make washing of the axilla, elbow crease and hand difficult, leading to hygiene problems, which in turn can lead to skin breakdown, infections and pressure sores. 10 Dressing can also be a challenge. Activities such as opening the hand for washing and putting an arm down a sleeve may need the assistance of a carer or the unaffected limb if the patient cannot carry out the task voluntarily with the affected limb. Tasks completed by another person or the unaffected limb are commonly described as ‘passive’ functional tasks. 11
Although the relationship between spasticity and motor performance remains unclear,12,13 upper limb spasticity is thought to lead to reduced ‘active’ function as overactive muscles around the shoulder and elbow limit reaching activities and spastic finger flexors impair potential finger extension. 10
Upper limb spasticity after stroke is readily recognised clinically, but studies of the prevalence of the condition are lacking. The largest prospective cohort study to date (n = 106) found that 31% of patients had upper limb spasticity at 12 months. 15 A further study (n = 95) found that 20% of stroke patients had upper limb spasticity 5 days after stroke and 18% had upper limb spasticity at 3 months. 16 Despite the lack of prevalence or prospective cohort studies, upper limb spasticity after stroke is an important clinical issue and identification and treatment of spasticity is a key component of stroke rehabilitation. 17
Because of the complex multifaceted definition, measurement of spasticity is a challenge and no tool covers all aspects of the definition. 18 Clinicians and researchers often measure resistance to passive movement (muscle tone) using a clinical assessment scale, e.g. the Modified Ashworth Scale. 19 Resistance to passive movement (muscle tone) in addition to measuring some components of spasticity such as hyperactive tonic stretch reflexes also measures muscle biomechanical/viscoelastic changes. 20 Biomechanical measures have been developed to measure resistance to passive movement21 and the stretch reflex can be quantified using neurophysiological techniques,22 but neither have been widely used in clinical practice or trials.
Botulinum toxin
Botulinum toxins are proteins produced by the bacterial species Clostridium botulinum. Seven serotypes of toxin, labelled A–G, are produced from different strains of C. botulinum. 23,24 All serotypes of toxin can cause botulism, which is a life-threatening condition involving symmetrical flaccid paralysis, autonomic dysfunction and respiratory compromise. 25
The clinical syndrome of botulism was first accurately described in 1820 when Justinus Kerner published his observations of ‘sausage poisoning’. 26 Kerner correctly hypothesised that the syndrome was caused by a biological poison that interrupted nerve conduction. Although he was unable to isolate the toxin, he did suggest that it may be possible to use it therapeutically. 26
Botulinum toxins act by blocking the release of acetylcholine from cholinergic nerves leading to blockage of transmission at the neuromuscular junction and paralysis, and blockage of cholinergic autonomic nerves with resulting autonomic disturbance. 27 When given by intramuscular injection, botulinum toxin causes local and temporary paresis. It has been suggested that botulinum toxin may also block transmission of sensory neurotransmitters providing an analgesic effect independent of muscle relaxation. 27–29 The effects of botulinum toxins are not permanent. Each serotype has a different length of activity with that of botulinum toxin type A being the longest – lasting for 3–4 months. 30
Botulinum toxin was first used clinically in 1977 when it was given by local injection to treat a patient with squint due to overactive ocular muscles. 31 Since then, botulinum toxin has been used to treat various conditions including dystonia, tremor, spasticity, achalasia, migraine, incontinence and sweating. Three preparations of botulinum toxin type A [Dysport® (Ipsen Ltd), Botox® (Allergan Inc.) and Xeomin® (MerzPharmaceuticals)] and one preparation of botulinum toxin type B [Neurobloc® (Eisai Ltd)] are currently available in the UK. 32 Only Dysport and Botox currently have a licence for treating spasticity. The potency of each product is different and doses are not interchangeable. 32,33
Adverse effects following injection with botulinum toxin are generally mild and transient. 9,34 Local reactions such as erythema, rash and oedema have been reported at the injection site. Migration of toxin into adjacent tissues can lead to weakening of surrounding muscles and autonomic effects; for example, injections into the neck can result in dysphagia and dry mouth. Systemic effects such as fatigue, malaise and flu-like symptoms are reported and systemic transport of toxin to tissues distant to those injected can also occur. Systemic transport of toxin has given cases of botulism-like illness35 or cases of muscle weakness distant from the injection site,32 but these events are very rare.
Review of the evidence for treating upper limb spasticity due to stroke with botulinum toxin
Sixteen randomised controlled trials (RCTs)36–51 and five systematic reviews52–56 evaluating the clinical effectiveness of botulinum toxin as a treatment for upper limb spasticity after stroke have been published. Eight trials44–51 and four systematic reviews53–56 were published following the start of this study in 2005. Details of our literature search strategy, methodological appraisal of the papers, overview of the studies, and a summary of the systematic reviews can be found in Appendix 1.
Trials have reported that treating upper limb spasticity due to stroke with botulinum toxin results in a measurable reduction in resistance to passive movement (muscle tone), which is evident by 1–2 weeks post-treatment. The treatment effect usually lasts for 3–4 months. Although trials varied in the dose and type of botulinum toxin used, the magnitude of initial change in muscle tone/spasticity was approximately a one-point decrease on the Modified Ashworth Scale, which reflects a clinically significant improvement. 36,38–42,44–46,48,49,51
The main benefits of spasticity reduction appear to be in terms of improved global patient/physician ratings36,38,41,42,44,51 and itemised passive functional tasks, notably hand hygiene. 37,40,42 Only one trial reported an improvement in active upper limb function. 45
Botulinum toxin has also been shown to reduce shoulder pain associated with spasticity,46,47 but its role in preventing or treating other types of upper limb pain associated with spasticity is unclear. Only one trial found that upper limb pain was reduced in those who received botulinum toxin compared with those in the control group. 45 Trials reported no unexpected adverse events, however, the event reporting system was often unclear. No trial considered the cost-effectiveness of treatment.
As the treatment effect of botulinum toxin lasts for 3–4 months, injections may need to be repeated to offer sustained benefit. There is limited evidence to support the continued use of repeated botulinum toxin injections for spasticity reduction. Two RCTs considered the impact of repeat injections demonstrating a decrease in resistance to passive movement following a second injection. 44,51 One of the trials also demonstrated sustained improvement in global ratings and patient-selected goals. 51 Six uncontrolled studies have examined the effects of repeated injections. 57–62 These studies are summarised in Appendix 1, Table 36. Repeat injections decreased muscle tone by a similar amount after each injection and improved passive functional scores by a similar amount to the initial injection. 58–60,62 Two studies measured active function58,61 and one found improvement. 61
Botulinum toxin and upper limb rehabilitation
The use of botulinum toxin to treat spasticity forms only one part of upper limb rehabilitation following stroke. Guidelines for the use of botulinum toxin in the treatment of spasticity recommend that it should be used in combination with a rehabilitation programme to achieve optimal beneficial effect. 9,17,63 It is recommended that the rehabilitation programme should consist of 2–8 weeks of physical and/or occupational therapy. 17
Limitations of previous studies
Previous trials varied in methodological quality, size, type of patients included, muscles treated with botulinum toxin, dose of botulinum toxin delivered and outcome measures used. It was often unclear how randomisation was undertaken, whether blinding was robust and follow-up complete. A number of trials developed outcome measures specifically for their studies which were not assessed for validity or reliability and which focused on passive benefits rather than active function. Participants were significantly younger (average age 52–66 years) than typical stroke patients (the average age of incident stroke is 74 years64). Studies were mostly undertaken in specialist rehabilitation centres and were small, lacking statistical power.
Studies published to date have not attempted to standardise upper limb therapy and the amount and content of the therapy received was usually poorly described. It was also often unclear what concurrent medication or additional antispasticity treatments patients received.
Justification for the current study
Botulinum toxin is increasingly used to treat upper limb spasticity due to stroke. Although botulinum toxin does reduce muscle tone and facilitates activities such as hand hygiene and dressing, the impact of this treatment upon upper limb active function is unclear. In clinical practice, botulinum toxin injections may be repeated every 3–4 months, but the effectiveness of repeat injections has not been adequately studied. It is also important that botulinum toxin is evaluated as part of a rehabilitation programme which is clearly described. In addition, cost-effectiveness of treatment is not fully established.
Multidisciplinary care on a stroke unit is currently the gold standard for stroke care regardless of age or stroke severity. 65 Evaluations of botulinum toxin should recruit participants from stroke services rather than tertiary referral centres to avoid selection bias and ensure results are applicable to routine care.
The Botulinum Toxin for the Upper Limb after Stroke (BoTULS) trial was designed to evaluate the clinical effectiveness and cost-effectiveness of botulinum toxin type A plus an upper limb therapy programme for the treatment of post-stroke upper limb spasticity.
Chapter 2 Methods
Study design
The BoTULS trial was a multicentre open-label parallel-group RCT comparing the clinical effectiveness and cost-effectiveness of botulinum toxin type A plus an upper limb therapy programme with the upper limb therapy programme alone for the treatment of upper limb spasticity due to stroke in adults.
Primary objective
-
To compare the upper limb function of participants with spasticity due to stroke who receive botulinum toxin type A injection(s) to the upper arm and/or forearm flexors/hand/shoulder girdle plus a 4-week evidence-based upper limb therapy programme (intervention group) with participants who receive the upper limb therapy programme alone (control group) 1 month after study entry. Upper limb function was assessed using the Action Research Arm Test (ARAT). 66
Secondary objectives
-
To compare the upper limb function, impairment and activity limitation of participants with spasticity due to stroke who receive botulinum toxin type A injection(s) to the upper arm and/or forearm flexors/hand/shoulder girdle plus a 4-week evidence-based upper limb therapy programme (intervention group) with participants who receive the upper limb therapy programme (control group) 1, 3 and 12 months after study entry. Upper limb function, impairment and activity limitation was assessed by: ARAT,66 Nine-Hole Peg Test,67 basic upper limb functional activity questions used in previous upper limb spasticity studies,37,39,41 Modified Ashworth Scale,19 Motricity Index,68 grip strength69 and Barthel Activities of Daily Living (ADL) Index. 70
-
To compare attainment of participant-selected upper limb goals, upper limb pain, and stroke-related quality of life/participation restriction between intervention and control groups at 1, 3 and 12 months. The following measures were used: attainment of participant-selected upper limb goals (1 month only) – Canadian Occupational Performance Measure (COPM);71 upper limb pain – numerical rating scales;72 stroke-related quality of life/participation restriction – Stroke Impact Scale,73 European Quality of Life-5 Dimensions (EQ-5D) measure of health-related quality of life74 and Oxford Handicap Scale. 75
-
To seek the experience and views of participants about treatment.
-
To compare the health-care and social services resources used by control and intervention groups during 3 months following study entry.
-
To report adverse events and compare the use of other antispasticity treatments between intervention and control groups.
-
To investigate the influence of severity of initial upper limb function and time since stroke upon the efficacy of the intervention.
Summary of design of randomised controlled trial
Figure 2 summarises the study method.
A list of all case record forms used in the study is shown in Appendix 2.
Setting
The study involved a collaborative network of 12 stroke services in the north of England. Expertise in the management of spasticity and use of botulinum toxin was provided by the regional spasticity clinic based at the International Centre for Neurorehabilitation, Newcastle upon Tyne, UK. This model, i.e. stroke units with close links to a specialist spasticity service, enabled stroke patients to access specialist care (both in terms of stroke and spasticity management) and could be replicated in other settings.
Case ascertainment
Potential participants were identified from a number of sources in each study centre which were components of locally organised stroke services (stroke unit, outpatients, day hospital and community rehabilitation teams). They were given an information leaflet and had an opportunity to discuss the study with a member of the clinical team. Training was given to clinical teams about the project and research governance. A member of the research team then arranged to see the participant to discuss the study. Consent was sought at the screening visit.
Some potential participants were not in contact with rehabilitation or stroke services. Local community stroke clubs and day centres were given information about the study and individuals were able contact a member of the study team directly.
Inclusion criteria
Adults with a stroke more than 1 month previously who had moderate/severe spasticity and reduced upper limb function who fulfilled all of following criteria were eligible:
-
age over 18 years
-
at least 1 month since stroke
-
upper limb spasticity [Modified Ashworth Scale19 > 2 at the elbow and/or spasticity at the hand, wrist or shoulder (there is no validated measure of spasticity at these sites)]
-
reduced upper limb function (ARAT66 score 0–56)
-
able to comply with the requirements of the protocol and upper limb therapy programme
-
informed consent given by participant or legal representative.
Exclusion criteria
-
Significant speech or cognitive impairment which impeded ability to perform the ARAT66 assessment.
-
Other significant upper limb impairment, e.g. fracture or frozen shoulder within 6 months, severe arthritis, amputation.
-
Evidence of fixed contracture.
-
Pregnancy or lactating.
-
Female at risk of pregnancy and not willing to take adequate precautions against pregnancy for the duration of the study.
-
Other diagnosis likely to interfere with rehabilitation or outcome assessments, e.g. registered blind, malignancy.
-
Other diagnosis which may contribute to upper limb spasticity, e.g. multiple sclerosis, cerebral palsy.
-
Contraindications to intramuscular injection.
-
Religious objections to blood products [botulinum toxin type A (Dysport) contains human albumin].
-
Contraindications to botulinum toxin type A, which include bleeding disorders, myasthenia gravis and concurrent use of aminoglycosides.
-
Use of botulinum toxin to the upper limb in the previous 3 months.
-
Known allergy or hypersensitivity to any of the test compounds.
-
Previous enrolment in this study.
Screening assessment
Having sought consent, the screening assessment was completed by a study therapist or clinical research associate. The assessment consisted of demographic details, review of medical history and medication, handedness, Abbreviated Mental Test Score,76 Sheffield Aphasia Screening Test,77 prestroke limitations (Oxford Handicap Scale),75 time since stroke, stroke type and subtype,78 self-reported current neurological impairment and activity limitation (Barthel ADL Index70), quality of life (EQ-5D74), assessment of spasticity (Modified Ashworth Scale19) and measurement of upper limb function (ARAT66). Details of antispasticity treatment and concomitant medications were recorded.
Baseline assessment
The baseline visit was undertaken within 2 weeks of the screening visit by a study therapist or clinical research associate. The inclusion/exclusion criteria were reviewed to ensure that the participant was still eligible. Participants underwent a clinical assessment and were asked to complete the following assessments: Modified Ashworth Scale,19 Motricity Index,68 grip strength,69 ARAT,66 Nine-Hole Peg Test,67 upper limb functional activity questions,37,39,41 Stroke Impact Scale73 and upper limb pain. 72 Female participants with child-bearing potential (i.e. those who were not either surgically sterile or at least 1 year post last menstrual period) had to have a negative urine pregnancy test to be included in the study. Such participants agreed to use adequate contraception throughout the study if they were randomised to receive botulinum toxin type A. Participants were randomised once the baseline assessment was completed.
Randomisation
Randomisation was by a central independent web-based randomisation service from the Clinical Trials Unit, Newcastle University, Newcastle upon Tyne, UK. Participants were stratified according to research site and level of upper limb function (ARAT 0–3, ARAT 4–28, ARAT 29–56), and randomised to intervention or control in a 1 : 1 ratio using permuted block sequences.
Botulinum toxin
Participants in the intervention group received botulinum toxin type A (Dysport). Dysport is available as a white lyophilised powder for reconstitution containing 500 units of C. botulinum type A toxin–haemagglutinin complex together with 125 µg of a 20% albumin solution and 2.5 mg lactose in a clear glass vial.
The range of muscles and dosages injected were as described in ‘The management of adults with spasticity using botulinum toxin: a guide to clinical practice’. 9 The maximum dose of botulinum toxin type A (Dysport) that could be administered at any one time point was 1000 units. All injectors were clinicians trained in the assessment and injection of botulinum toxin in the context of upper limb spasticity.
The use of aminoglycosides was prohibited during the study because they enhance the effects of botulinum toxin, thereby increasing the risk of toxicity. Clinicians were advised to use muscle relaxants with caution because the effects of botulinum toxin are enhanced by non-depolarising muscle relaxants. The international normalised ratio of participants taking warfarin was checked before injection. Information about concomitant drug use was given in the patient information sheet and in letters to consultants and general practitioners.
If further treatment was necessary at 3, 6 or 9 months, further injections were provided to those in the intervention group. At each visit a letter was sent to the participant’s stroke physician, general practitioner and physiotherapist. At the 12-month review, participants in both the intervention and control groups who required botulinum toxin were referred to the spasticity clinic.
If during the course of the trial the study therapist decided that a participant in the control group had an unacceptable degree of symptomatic spasticity, further management was discussed with the stroke physician, physiotherapist, occupational therapist and/or a member of the local or regional spasticity team and the participant could then be referred to a local spasticity service for botulinum toxin.
The upper limb rehabilitation programme
Guidelines highlight that it is important that botulinum toxin is not used in isolation but as part of a comprehensive rehabilitation programme. 9,17,63,79,80 Focal reductions in upper limb spasticity from any pharmacological intervention are unlikely to translate into sustained improvements in function or patient-selected rehabilitation goals without a targeted therapy programme.
The upper limb therapy programme was based upon available research evidence from the stroke rehabilitation and skill acquisition literature as well as clinical practice80–98 and consisted of two menus. Participants with ARAT 0–3 received menu 1, which was designed specifically for participants with no active upper limb function. Menu 1 aimed at improving and maintaining range of movement, encouraging active assisted upper limb movement in the context of functional activities, along with hand hygiene and positioning88–93. Menu 2 was for participants with some retained active upper limb movement (ARAT 4–56) and was piloted in a previous study. 99 Following stretching of soft tissues affected by spasticity, this menu specifically concentrated on task-orientated practise aimed at patient-centred goals. Upper limb goals were measured by the COPM. 71 Each menu standardised the category of tasks, the number and order of repetitions as well as the amount of feedback for each session, but within these parameters the therapist was able to tailor the specifics of each activity to the ability of the patient. Manuals and training programmes were developed for both upper limb therapy menus and all therapists were trained in the delivery of the programme.
The upper limb therapy programme was provided by study therapists and each participant received 1 hour per day, twice a week for 4 weeks, in addition to their other rehabilitation needs. The study therapist could transfer participants between menu 1 and menu 2 according to their clinical opinion. Participants were given a written exercise programme, which was based on the content of the face-to-face sessions with the therapist, to carry out by themselves or with a carer (following training) on the weekdays on which they were not attending therapy.
If the participant was already receiving rehabilitation, then the upper limb therapy programme was delivered in that setting, e.g. stroke unit, outpatients, day hospital or home. In each case, the study therapist liaised closely with the rehabilitation team to ensure the participant’s needs were addressed and therapy was well co-ordinated. At the end of the 4-week intervention period participants were given advice by the study therapist regarding maintenance of upper limb function.
Participants were reviewed by the research team every 3 months. If further therapy was required, this was provided by a study therapist. Those in the intervention group could also receive further botulinum toxin type A injections. Participants in both the intervention and control groups who had symptomatic spasticity at the 12-month follow-up appointment were referred to the spasticity clinic.
Participants who made a good recovery before completing the 4-week upper limb therapy programme could be discharged from the programme provided that they had achieved a maximum score on the ARAT66 and achieved their upper limb goals.
Outcome assessments
Outcomes were measured 1 month (+/– 3 days), 3 months (+/– 5 days) and 12 months (+/– 5 days) after study randomisation.
Each outcome assessment consisted of two stages – stage 1 outcome assessment was a self completion postal questionnaire (Barthel ADL Index,70 Oxford Handicap Scale,75 Stroke Impact Scale,73 EQ-5D,74 upper limb functional activity questions37,39,41 and resource utilisation) which was sent to participants 1 week before stage 2. Participants were asked to bring the completed proforma to their stage 2 appointment.
Stage 2 outcome assessments consisted of assessment of upper limb impairment and function (Modified Ashworth Scale,19 Motricity Index,68 grip strength,69 ARAT,66 Nine-Hole Peg Test67 and upper limb pain72) and face-to-face interview seeking participant’s experience and views of the study treatment. Information was sought about side effects, use of other antispasticity treatment and any change in the participant’s concomitant medications. Any new adverse events or changes in existing adverse events that had occurred since the previous visit were sought. The stage 1 questionnaire was checked for completeness. The stage 2 assessment was completed by a study therapist or clinical research associate.
Blinding
Outcome assessments were undertaken by an assessor who was blinded to the randomisation group. Participants and the study therapists who provided the upper limb therapy programme were not blind to the randomisation group. To enable blinding to be achieved, study therapists undertook screening and baseline assessments and provided the upper limb therapy programme in one research centre and undertook outcome assessments in adjacent centres. As it was possible for assessors to become unblinded, at each outcome assessment an evaluation of blinding was performed.
Participant withdrawal criteria
No specific withdrawal criteria were defined for the study. If a participant discontinued the study prematurely (i.e. before completion of the protocol), the primary reason for discontinuation was recorded when given. In all cases the investigator ensured that the participant received medical follow-up as necessary. Withdrawn participants were not replaced.
Study completion/early termination visit
Study completion was the last outcome visit. If a participant discontinued from the study prematurely, every effort was made to perform an early termination visit consisting of all outcome assessments. At the participant’s last study visit details of their completion of the study/withdrawal from the study were recorded. Female participants of child-bearing potential (i.e. those who were not either surgically sterile or at least 1 year post last menstrual period) in the intervention group were asked to undertake a urine pregnancy test.
Safety evaluation
Side effects of botulinum toxin type A are generally mild and transient. Local muscle weakness may occur as a result of toxin spread to nearby muscles. Five per cent experience flu-like symptoms 1 week to 10 days after injection. Pain at the injection site and a dry mouth can occur. Transient dysphagia has been reported. Anaphylaxis rarely occurs. Excessive doses may produce distant and profound neuromuscular paralysis. Respiratory support may be required where excessive doses cause paralysis of respiratory muscles.
The safety of botulinum toxin type A in the treatment of participants with upper limb spasticity post stroke was evaluated by examining the occurrence of all adverse and serious adverse events as defined by the Medicines for Human Use (Clinical Trials) Regulations, 2004. 100 Follow-up of each adverse event continued until the event or its sequelae resolved or stabilised at a level that was acceptable to the investigator.
Study schedule
Table 1 summarises the study schedule.
Time point | Screening < 2 weeks | Baseline | Visit 3 Month 1a | Visit 4 Month 3b | Visit 5 Month 6 | Visit 6 Month 9 | Visit 7 Month 12b |
---|---|---|---|---|---|---|---|
Informed consent | ✗ | ||||||
Record demographics and handedness | ✗ | ||||||
Review inclusion/exclusion criteria | ✗ | ✗ | |||||
Review medical history | ✗ | ✗ | |||||
Details of stroke | ✗ | ||||||
Prestroke limitation (inc. Oxford Handicap Scale) | ✗ | ||||||
Abbreviated Mental Test Score | ✗ | ||||||
Sheffield Aphasia Screening Test | ✗ | ||||||
Action Research Arm Test | ✗ | ✗ | ✗ | ✗ | ✗ | ||
Motricity Index | ✗ | ✗ | ✗ | ✗ | |||
Grip strength | ✗ | ✗ | ✗ | ✗ | |||
Nine-Hole Peg Test | ✗ | ✗ | ✗ | ✗ | |||
Modified Ashworth Scale | ✗ | ✗ | ✗ | ✗ | ✗ | ||
Upper limb pain | ✗ | ✗ | ✗ | ✗ | |||
Patient selects upper limb goals | ✗ | ||||||
Review upper limb goal attainment | ✗ | ||||||
Oxford Handicap Scalec | ✗ | ✗c | ✗c | ✗c | |||
Barthel ADL Indexc | ✗ | ✗c | ✗c | ✗c | |||
Upper limb functional activity questionsc | ✗ | ✗c | ✗c | ✗c | |||
Quality of life – Stroke Impact Scalec | ✗ | ✗c | ✗c | ✗c | |||
Quality of life – EQ-5Dc | ✗ | ✗c | ✗c | ✗c | |||
Resource utilisation questionsc | ✗ | ✗c | ✗c | ✗c | |||
Pregnancy testd | ✗ | ✗f | ✗f | ✗f | |||
Randomisation | ✗ | ||||||
Treatment with Dysporte | ✗ | ✗g | ✗g | ✗g | |||
Commencement of 4-week upper limb therapy programme | ✗ | ✗g | ✗g | ✗g | |||
Clinical assessment by study therapist | ✗ | ✗ | ✗ | ✗ | ✗ | ||
Concomitant medications (inc. antispasticity treatment) | ✗ | ✗ | ✗ | ✗ | ✗ | ||
Adverse events | ✗ | ✗ | ✗ | ✗ | |||
Participants’ views and experience | ✗ | ✗ |
Resource utilisation and economic evaluation
This is discussed in Chapter 4.
Data completeness
Data quality checks were performed regularly throughout the study. Missing data and data queries were discussed with the local site teams and collected/resolved as possible.
Statistical analysis
Analyses were undertaken on an ‘intention-to-treat’ basis; participants were analysed in the group to which they were randomised. Data were exported from the study microsoft access database to spss for analysis. All available data were analysed, missing data were not imputed.
The primary end point was the ARAT score at 1 month. For each participant it was determined if there had been a significant improvement in function based on the change in ARAT score from baseline. It is suggested that the minimal clinically important difference for the ARAT is 10% of its range (six points);101 however, we estimated that a smaller treatment effect would be clinically beneficial in those with poor initial upper limb function (ARAT 0–3) compared with those with some retained active function (ARAT 4–56).
A successful outcome was defined as:
-
a change of three or more points on the ARAT scale for a participant whose baseline ARAT score was between 0 and 3
-
a change of six or more points on the ARAT scale for a participant whose baseline ARAT score was between 4 and 51
-
a final ARAT score of 57 for a participant whose baseline ARAT score was 52–56.
The proportion of ‘successes’ in each group was compared using Fisher’s exact test and an interval estimate of the effect of the intervention in the form of an approximate 95% confidence interval (95% CI) for the relative risk was calculated.
Secondary outcomes providing binary data were compared using Fisher’s exact test (or chi-squared test if unable to compute exact form). Secondary outcomes providing ordinal or continuous data were compared using the Mann–Whitney U-test (exact form where possible). Two-tailed p-values are reported. All secondary outcomes were analysed using scale score at follow-up and change in scale score from baseline to follow-up. Change in score was believed to be the key analysis and is presented in the Results section. Scale score at follow-up is presented in Appendix 3.
Although the Mann–Whitney U-test gives an indication of statistical significance it does not provide any information about the magnitude of difference between the groups or whether the difference is clinically important. For some outcomes, the presentation of median values was not helpful in determining clinical importance because of the presence of skewness (changes in the tail of the distribution may not result in any change in the median score). To address this we used resampling methods (bootstrapping) to generate an interval estimate of the effect of the intervention on the group means for each outcome (changes in the tails of the distribution will affect the mean score). The 2.5 and 97.5 percentiles of the bootstrap distribution based on 10,000 replications are reported. This analysis was not prespecified in the study protocol because it was not anticipated to be necessary before viewing the results of planned non-parametric tests.
To enable comparison with previous studies, the basic upper limb functional activity questions were also analysed by comparing the proportion of participants in each randomisation group who had improved by one or more points on the scale from baseline.
As a secondary analysis, logistic regression modelling was used to estimate the effect of the intervention on the primary outcome (ARAT ‘success’) adjusting for randomisation strata (research site and baseline upper limb function).
There were two prespecified subgroup analyses. Response to treatment was compared for:
-
participants who had a stroke ≤ 1 year ago and those who had a stroke > 1 year ago
-
participants with no initial active upper limb function (with a baseline ARAT score of 0–3) and participants with some initial upper limb function (baseline ARAT 4–56).
Subgroup analysis of the primary outcome was undertaken using logistic regression procedures by adding a subgroup by treatment interaction term to a model that already included the main effects. For secondary outcomes, subgroup analysis was only undertaken where the difference between treatment groups in the main analysis had been statistically significant. For these secondary outcomes, resampling procedures were used to estimate the difference between the treatment effects in the two subgroups.
A power calculation was performed before the start of the study using prognosis based methodology. 102 A clinically important treatment effect was defined as a difference in good outcomes between intervention and control groups of 15% where a good outcome was defined as listed above for each ARAT group; it was expected to see 20% of the control group achieve good outcomes and 35% of the intervention group achieve good outcomes. Using Fleiss’s method for a binary outcome103 and inflating the sample size by 10% to allow for attrition, we needed to recruit a total sample of 332 participants to give 80% power to detect a 15% difference in good outcomes assuming a two-tailed test and a significance level of 5%. The study aimed to recruit 50% of the sample from the ARAT 0–3 group and 50% from the ARAT 4–56 group.
Ethical arrangements and research governance
Multicentre Research Ethics Committee approval was granted. For each individual centre, a site-specific approval was obtained from the appropriate local research ethics committee. Research and development approval was obtained from each participating Trust. The trial was commenced subsequent to the UK Medicines for Human Use (Clinical Trials) Regulations, 2004100 and was one of the first investigator-led trials of an investigational medical product to be undertaken in the UK following the introduction of this legislation. Regulatory approval was granted by the Medicines and Healthcare Products Regulatory Agency and the trial was conducted in accordance with the legislation, the International Conference on Harmonisation–Good Clinical Practice (ICH-GCP),104 and the Research Governance Framework for Health and Social Care. 105
A Trial Steering Committee (TSC) and Data Monitoring and Ethics Committee (DMEC) following Medical Research Council guidelines were established. 106 The TSC comprised an independent chairperson, two independent researchers (all of whom had expertise in rehabilitation research and clinical trials), a consumer representative and three members of the study team.
The DMEC was chaired by a clinical academic with expertise in RCTs of complex interventions in the field of stroke. A biostatistician with expertise in multicentre trials, a researcher experienced in running rehabilitation trials and the study statistician were also members.
An informal interim analysis was performed at the request of the DMEC following recruitment of 50% of the required sample size. Only the DMEC had access to these data. They were able to recommend discontinuation of the study if significant ethical or safety concerns arose, or if there was clear evidence of benefit (clinical or statistical).
The study was monitored for compliance with ICH-GCP by an independent monitor from the Newcastle Clinical Trials Unit.
Amendments to the study following commencement
Objectives
The initial study protocol included measurement of spasticity at the elbow by a biomechanical device that had been used in a previous pilot study. 107 This was to be used in addition to clinical measures. Unfortunately, the device was not at a stage of development where it could be used in a multicentre study.
Setting
Initially the trial was planned in four geographical areas within the UK: North Tyneside, Wansbeck, Newcastle upon Tyne and Sunderland. As a result of low recruitment rates, eight further areas were added (Gateshead, South Tyneside, Durham, Hexham, Carlisle, Bishop Auckland, Hartlepool, North Tees).
Case ascertainment
This was widened to include identification of participants from stroke clubs and day centres (in addition to clinical settings) because of initial low recruitment rates.
Inclusion criteria
Prospective studies of upper limb recovery have shown that baseline impairment is a strong predictor of outcome. To demonstrate whether botulinum toxin plus upper limb therapy can improve upper limb function (primary outcome) it was initially thought important to exclude those participants with no retained upper limb function (ARAT 0–3). Because of the initial low recruitment and following reconsideration of the evidence of effectiveness of botulinum toxin in this group, this was later reviewed and a decision was taken in conjunction with the TSC and the Health Technology Assessment programme that it would be valuable to include stroke patients with all levels of reduced upper limb function (ARAT 0–56).
The study also initially excluded participants with cognitive impairment or significant speech problems measured by the Abbreviated Mental Test Score and Sheffield Aphasia Screening Test. This was felt to be too restrictive, excluding patients who were keen to participate. This inclusion criterion was relaxed to include all participants capable of performing the ARAT and complying with the upper limb therapy programme.
Upper limb therapy programme
A second menu was developed for the upper limb therapy programme after the eligibility criteria were widened to include participants with no active upper limb function. This alternative menu was designed because the original menu contained activities that these participants would not have been able to undertake.
Statistical analysis
Inclusion of participants with lower ARAT scores required revision of the primary analysis and power calculation. In the original protocol, the primary analysis was comparison of ARAT scores and 390 participants were required to provide 80% power to detect a difference of six points on the ARAT between intervention and control groups. However, participants with a baseline ARAT of 0–3 could not be expected to improve as much as those with a baseline ARAT of 4–56. This led to the definition of successful treatment as improvement by three points on the ARAT for those with a starting ARAT of 0–3, six points by those with a starting ARAT of 4–51 and a final score of 57 for participants whose baseline ARAT was 52–56. Comparison of the proportions of successes between the groups (control/intervention) became the primary analysis. The power calculation was revised for the new binary outcome and 332 participants were required to provide 80% power to detect a 15% difference in treatment successes.
Follow-up period
Participants recruited after 2 July 2007 were followed for 3 months only. This was a pragmatic decision taken because the trial was behind schedule as a result of initial low recruitment rates. Curtailing 12-month follow-up allowed the trial to be completed within the initial study timetable.
The initial study protocol specified comparison of health-care and social services resource usage between the randomisation groups for 12 months. Because of the curtailing of 12-month follow-up, it was felt more appropriate to compare health-care and social services resource usage over 3 months to include all study participants.
Chapter 3 Results
Study recruitment
Between July 2005 and March 2008, 333 participants were recruited to the BoTULS trial. One hundred and seventy were randomised to the intervention group and 163 to the control group. Monthly and cumulative recruitment are shown in Figure 3.
Two hundred and eight (62%) participants were randomised before July 2007 and entered the trial for 12 month follow-up. The remaining 125 (38%) participants were followed for 3 months. Details of recruitment in each study site are given in Appendix 4.
Study attrition
Figure 4 shows participant flow through the trial. There were nine deaths during the study period and 12 participants withdrew. Reasons for attrition are described in Table 2.
Control (n and reasons) | Intervention (n and reasons) | |
---|---|---|
Withdrawn | ||
0–1 month | 4; wanted injection; did not want to continue; wife unwell; no reason recorded | 3; did not want injection; unwell; in another trial |
> 1–3 months | 2; wanted injection; thought no benefit | 1; unwell |
> 3–12 months | 0 | 2; unwell; no reason recorded |
Death | ||
0–1 month | 1; myocardial infarction | 0 |
> 1–3 months | 1; pneumonia | 1; pneumonia |
> 3–12 months | 4; pneumonia; further stroke; cancer; cause unknown | 2; pneumonia; further stroke |
Assessment not done | ||
1 month | 3; unwell (n = 2);administrative error | 0 |
3 months | 4; unwell (n = 3); unable to contact | 2; unwell |
12 months | 2; assessment recorded as done but data missing | 2; unable to contact |
One participant withdrew consent shortly after randomisation and asked for their data to be excluded from the analyses. Baseline data are therefore presented for 332 participants. The 1-month assessment was completed by 155/163 (95%) participants in the control group and 167/170 (98%) participants in the intervention group. The 3-month assessment was completed by 151/163 (93%) participants in the control group and 163/170 (96%) in the intervention group.
Of the 332 participants randomised into the study, 208 (63%) were enrolled for 12-month follow-up. The 12-month assessment was completed by 92/103 (89%) participants in the control group and 97/105 (92%) in the intervention group. Outcome data were missing at all time points for one study participant only.
Study population
Randomisation groups were well matched at baseline with regard to demography, stroke characteristics and comorbidity (Table 3). The median age of participants was 67 years [interquartile range (IQR) 59–74], 225 (67.8%) were male and the majority were living at home. One hundred and eighty-one participants (54.5%) were randomised within 1 year of stroke.
Control (n = 162)a | Intervention (n = 170)a | |
---|---|---|
Sex: n (%) | ||
Male | 115 (71.0) | 110 (64.7) |
Female | 47 (29.0) | 60 (35.3) |
Age: median (IQR) | ||
All | 66 (59.8 to 72.3) | 67 (58.8 to 74) |
Male | 67 (61 to 73) | 68 (59 to 74) |
Female | 64 (54 to 72) | 67 (56.3 to 74) |
Current residence: n (%) | ||
Own house | 133 (82.1) | 129 (75.9) |
Living with family/friends | 0 (0.0) | 4 (2.4) |
Sheltered | 3 (1.9) | 4 (2.4) |
Residential care/nursing home | 7 (4.3) | 5 (2.9) |
Hospital | 19 (11.7) | 28 (16.5) |
Stroke type: n (%) | ||
Infarct | 131 (81.9) | 140 (82.8) |
Intracerebral haemorrhage | 21 (13.1) | 25 (14.8) |
Subarachnoid haemorrhage | 3 (1.9) | 2 (1.2) |
Unknown | 7 (4.3) | 3 (1.8) |
Stroke subtype: n (%) | ||
Total anterior circulation stroke | 68 (42.0) | 75 (44.1) |
Partial anterior circulation stroke | 61 (37.7) | 57 (33.5) |
Lacunar stroke | 26 (16.0) | 33 (19.4) |
Posterior circulation stroke | 3 (1.9) | 2 (1.2) |
Uncertain | 4 (2.5) | 3 (1.8) |
Time from stroke to randomisation: median (IQR) days | 280 (148.8 to 1145.8) | 324 (128.5 to 1387.5) |
Time from stroke to randomisation: n (%) | ||
1–6 months | 49 (30.2) | 60 (35.3) |
> 6 months to 1 year | 43 (26.5) | 29 (17.1) |
> 1–2 years | 19 (11.7) | 16 (9.4) |
> 2–5 years | 29 (17.9) | 31 (18.2) |
5+ years | 22 (13.6) | 34 (20.0) |
Comorbidity: n (%) | ||
Previous stroke/transient ischaemic attack | 48 (29.6) (n = 162) | 49 (28.8) (n = 170) |
Ischaemic heart disease | 36 (22.4) (n = 161) | 39 (23.1) (n = 169) |
Peripheral arterial occlusive disease | 8 (5.0) (n = 160) | 6 (3.6) (n = 168) |
Diabetes mellitus | 22 (13.6) (n = 162) | 22 (13.1) (n = 168) |
Hypertension | 119 (73.5) (n = 162) | 124 (74.3) (n = 167) |
Hyperlipidaemia | 103 (64.4) (n = 160) | 111 (65.7) (n = 169) |
Atrial fibrillation | 21 (13.3) (n = 158) | 24 (14.5) (n = 166) |
Randomisation groups were well matched for the distribution, severity and current treatment of upper limb spasticity (Table 4). The majority of participants had spasticity present throughout the upper limb and the median score on the Modified Ashworth Scale at the elbow was two. Previous treatment with botulinum toxin was not an exclusion criterion provided that it was given more than 3 months before study entry and potential participants were prepared to temporarily relinquish further upper limb injection(s) should they be randomised to the control group. Botulinum toxin treatment had been previously received by 27 (16.7%) of the control group compared with 21 (12.4%) of the intervention group. Randomisation groups were also well matched for other measures of upper limb impairment (Table 4).
Control (n = 162) | Intervention (n = 170) | |
---|---|---|
Upper limb affected by spasticity: n (%) | ||
Right | 65 (40.1) | 75 (44.1) |
Left | 97 (59.9) | 95 (55.9) |
Dominant hand affected: n (%) | ||
Yes | 64 (39.5) | 80 (47.1) |
No | 98 (60.5) | 90 (52.9) |
Area affected by spasticity: n (%) | ||
Shoulder | 95 (58.6) | 111 (65.3) |
Elbow | 156 (96.3) | 161 (94.7) |
Wrist | 141 (87.0) | 141 (82.9) |
Hand | 140 (86.4) | 138 (81.2) |
Distribution of spasticity: n (%) | ||
Shoulder and elbow | 9 (5.6) | 15 (8.8) |
Elbow and wrist | 8 (4.9) | 2 (1.2) |
Wrist and hand | 5 (3.1) | 4 (2.4) |
Shoulder and elbow and wrist | 1 (0.6) | 9 (5.3) |
Shoulder and elbow and hand | 4 (2.5) | 4 (2.4) |
Elbow and wrist and hand | 47 (29.0) | 43 (25.3) |
Shoulder and elbow and wrist and hand | 80 (49.4) | 81 (47.6) |
Other | 8 (4.9) | 12 (7.1) |
Antispasticity treatment: n (%) | ||
Total | 67 (41.4) | 64 (37.6) |
Dantrolene | 2 (1.2) | 3 (1.8) |
Baclofen | 16 (9.9) | 20 (11.8) |
Tizanidine | 0 (0.0) | 3 (1.8) |
Gabapentin | 16 (9.9) | 14 (8.2) |
Methocarbanol | 1 (0.6) | 0 (0.0) |
Shoulder brace | 1 (0.6) | 0 (0.0) |
Upper limb sling | 0 (0.0) | 2 (1.2) |
Thumb strap | 1 (0.6) | 0 (0.0) |
Elasticated glove | 1 (0.6) | 0 (0.0) |
Functional electrical stimulation machine | 1 (0.6) | 1 (0.6) |
Upper limb splint | 42 (25.9) | 36 (21.2) |
Botulinum toxin treatment > 3 months before study entry: n (%) | ||
Total | 27 (16.7) | 21a (12.4) |
Arm | 12 (44.4) | 4 (19.0) |
Leg | 7 (25.9) | 5 (23.8) |
Both arm and leg | 8 (29.6) | 11 (52.4) |
Modified Ashworth Scale at elbow: n (%) | ||
0 | 7 (4.3) | 8 (4.7) |
1 | 16 (9.9) | 20 (11.8) |
1 + | 53 (32.7) | 44 (25.9) |
2 | 57 (35.2) | 68 (40.0) |
3 | 28 (17.3) | 30 (17.6) |
4 | 1 (0.6) | 0 (0.0) |
Median (IQR) | 2 (1 + to 2) | 2 (1 + to 2) |
Motricity index: median (IQR) | ||
Arm | 40 (29 to 62) | 40 (24 to 61) |
Total | 47 (34.5 to 64.3) | 50 (33.5 to 64.5) |
Grip strength (kg): median (IQR) | 1.8 (0.0 to 6.0) | 0.7 (0.0 to 5.0) |
One hundred and eighty-four participants (55.4%) had no active upper limb function (ARAT 0–3) and 148 (44.6%) had some retained active function (ARAT 4–56) at randomisation. The median initial ARAT in both intervention and control groups was three (Table 5). Most participants had no or poor dexterity and were unable to complete the Nine-Hole Peg Test. Participants experienced moderate difficulty with upper limb functional activities such as putting their arm through a sleeve or opening the hand to clean the palm and the majority were unable to use cutlery as a result of their stroke. The control group had a lower level of participation restriction when assessed using the Oxford Handicap Scale, but it is unlikely that this possible imbalance at baseline is clinically important or had an impact upon outcome assessments.
Control (n = 162)a | Intervention (n = 170)a | |
---|---|---|
ARAT: median (IQR) | ||
Total | 3 (3 to 16) | 3 (3 to 16) |
Grasp | 0 (0 to 5) | 0 (0 to 5) |
Grip | 0 (0 to 4) | 0 (0 to 3) |
Pinch | 0 (0 to 0) | 0 (0 to 0) |
Gross | 3 (3 to 5) | 3 (2.8 to 4) |
Nine-Hole Peg Test (pegs placed in 50 seconds): median (IQR) | 0 (0 to 0) | 0 (0 to 0) |
Upper limb functional activities: median (IQR) | ||
Put arm through sleeve | 3 (2 to 4) (n = 142) | 3 (2 to 4) (n = 159) |
Open the hand for cleaning your palm | 3 (2 to 4) (n = 142) | 3 (2 to 4) (n = 159) |
Open the hand for cutting fingernails | 2 (1 to 4) (n = 141) | 2 (1 to 3.3) (n = 158) |
Use cutlery | 1 (1 to 1) (n = 140) | 1 (1 to 1) (n = 155) |
Barthel ADL Index: median (IQR) | 15 (10 to 17) (n = 162) | 15 (10 to 17) (n = 170) |
Stroke Impact Scale: median (IQR) | ||
Strength | 31.3 (18.8 to 43.8) (n = 141) | 31.3 (18.8 to 43.8) (n = 157) |
Memory | 82.1 (64.3 to 92.9) (n = 143) | 78.6 (57.1 to 92.9) (n = 159) |
Emotion | 66.7 (52.8 to 80.6) (n = 141) | 66.7 (55.6 to 77.8) (n = 159) |
Communication | 89.3 (67.9 to 100) (n = 142) | 85.7 (60.7 to 100.0) (n = 159) |
ADL | 42.5 (32.5 to 57.5) (n = 142) | 40.0 (30.0 to 55.0) (n = 159) |
Mobility | 50.0 (30.6 to 69.4) (n = 139) | 47.2 (25.0 to 64.3) (n = 158) |
Hand function | 0.0 (0.0 to 10.0) (n = 139) | 0.0 (0.0 to 15.0) (n = 158) |
Participation / handicap | 40.6 (21.9 to 65.6) (n = 141) | 34.4 (15.6 to 53.1) (n = 158) |
Physical domain | 33.3 (22.7 to 42.5) (n = 143) | 30.5 (20.1 to 43.1) (n = 159) |
Stroke recovery | 50.0 (30.0 to 60.0) (n = 136) | 40.0 (25.0 to 53.5) (n = 157) |
EQ-5D: median (IQR) | ||
Mobility | 2 (2 to 2) (n = 162) | 2 (2 to 2) (n = 170) |
Self-care | 2 (2 to 2) (n = 162) | 2 (2 to 2) (n = 170) |
Usual activities | 3 (2 to 3) (n = 162) | 3 (2 to 3) (n = 170) |
Pain/discomfort | 2 (1 to 2) (n = 162) | 2 (1 to 2) (n = 170) |
Anxiety/depression | 2 (1 to 2) (n = 162) | 2 (1 to 2) (n = 170) |
Good/bad health scale | 60 (50 to 70) (n = 161) | 60 (50 to 70) (n = 169) |
Oxford Handicap Scale: median (IQR) | 3 (3 to 4) | 4 (3 to 4) |
The median rating for pain was moderate in both groups. The median pain score was 5/10 in both the intervention and control groups (Table 6).
Primary outcome
Improved upper limb function (predefined treatment success on the ARAT) at 1 month was achieved by 30/154 (19.5%) participants in the control group and 42/167 (25.1%) in the intervention group. This difference was not significant (p = 0.232). The relative risk of having a ‘successful treatment’ in the intervention group compared with the control group was 1.3 (95% CI 0.9 to 2.0).
Secondary outcomes
Impairment
Changes in impairment from baseline to 1, 3 and 12 months are shown in Tables 7a and 7b. At 1 month, muscle tone at the elbow (Modified Ashworth Scale) decreased in the intervention group compared with the control group; the median change from baseline to 1 month in the control group was zero; the median change in the intervention group was – 1 (p < 0.001). The corresponding differences in change in muscle tone between intervention and control groups seen at 3 and 12 months were not statistically significant.
Control | Intervention | p-value | |
---|---|---|---|
Modified Ashworth Scale at elbow: median change (IQR) | |||
1 month | 0 (– 1 to 1) (n = 154) | – 1 (– 1 to 0) (n = 167) | 0.001 |
3 months | 0 (– 1 to 0) (n = 151) | 0 (– 1 to 0) (n = 163) | 0.145 |
12 months | 0 (– 1 to 1) (n = 91) | 0 (– 1 to 0) (n = 97) | 0.333 |
Motricity index: median change (IQR) | |||
Arm | |||
1 month | 0 (– 9 to 11) (n =153) | 3 (– 5 to 11) (n = 167) | 0.138 |
3 months | 0 (– 6 to 11) (n = 151) | 4 (– 4 to 14) (n = 164) | 0.055 |
12 months | 5 (– 3 to 11) (n = 92) | 5 (0 to 13) (n = 97) | 0.597 |
Total | |||
1 month | 1.5 (– 5.5 to 8.8) (n = 153) | 2.5 (– 4.5 to 11) (n = 167) | 0.277 |
3 months | 0 (– 6.6 to 9) (n = 151) | 4 (– 4.5 to 11.5) (n = 162) | 0.042 |
12 months | 2 (– 5.9 to 9.4) (n = 92) | 3 (– 2.5 to 9.5) (n = 97) | 0.588 |
Grip strength (kg): median change (IQR) | |||
1 month | 0.0 (– 0.4 to 2.0) (n = 154) | 0.0 (– 0.7 to 1.3) (n =167) | 0.233 |
3 months | 0.0 (– 0.7 to 2.0) (n = 151) | 0.0 (– 0.0 to 2.7) (n = 163) | 0.139 |
12 months | 0.5 (– 0.5 to 3.9) (n = 92) | 0.0 (– 0.2 to 3.0) (n = 97) | 0.764 |
Control | Intervention | Difference | |
---|---|---|---|
Modified Ashworth Scale at elbow: mean change (95% CI) | |||
1 month | – 0.1 (– 0.2 to 0.1) | – 0.6 (– 0.8 to – 0.4) | – 0.5 (– 0.8 to – 0.3) |
3 months | – 0.1 (– 0.3 to 0.1) | – 0.3 (– 0.4 to – 0.1) | – 0.2 (– 0.5 to 0.1) |
12 months | – 0.2 (– 0.5 to 0.1) | – 0.3 (– 0.5 to – 0.1) | – 0.1 (– 0.4 to 0.2) |
Motricity Index: mean change (95% CI) | |||
Arm | |||
1 month | 1.4 (– 0.9 to 3.7) | 3.6 (1.5 to 5.7) | 2.2 (– 0.9 to 5.4) |
3 months | 1.7 (– 0.6 to 4.1) | 5.2 (2.8 to 7.6) | 3.5 (0.1 to 6.8) |
12 months | 3.6 (0.7 to 6.4) | 6.1 (3.5 to 8.8) | 2.5 (– 1.4 to 6.3) |
Total | |||
1 month | 1.4 (– 0.3 to 3.1) | 2.6 (0.9 to 4.3) | 1.2 (– 1.2 to 3.7) |
3 months | 1.2 (– 0.7 to 3.1) | 4.3 (2.5 to 6.2) | 3.2 (0.4 to 5.8) |
12 months | 2.4 (0.1 to 4.8) | 3.6 (1.6 to 5.8) | 1.3 (– 1.9 to 4.4) |
Grip strength (kg): mean change (95% CI) | |||
1 month | 0.6 (0.2 to 1.1) | 0.5 (0.0 to 1.0) | – 0.2 (– 0.8 to 0.5) |
3 months | 0.9 (0.4 to 1.5) | 1.9 (1.1 to 2.8) | 1.0 (0.0 to 2.0) |
12 months | 1.6 (0.7 to 2.6) | 1.5 (0.7 to 2.4) | – 0.1 (– 1.4 to 1.1) |
The differences between the groups for change in upper limb motor impairment (Motricity Index) from baseline to 1 or 12 months were not significant. However, at 3 months the intervention group had improved by a median of four compared with a median of zero in the control group. This difference approached statistical significance when groups were compared using the Mann–Whitney U-test (p = 0.055). Examination of the difference between the mean change in the groups confirmed a similar magnitude of effect (3.5, 95% CI 0.1 to 6.8), which was statistically significant. Total motor impairment was also improved at 3 months in the intervention group compared with the control group (p = 0.042). There were no significant differences between intervention and control groups for change in grip strength at any assessment.
Upper limb function and activity limitation
At 3 months, predefined treatment success (improvement in upper limb function) on the ARAT was achieved by 37/151 (24.5%) participants in the control group and 54/161 (33.5%) in the intervention group (p = 0.083). At 12 months treatment success was achieved by 27/92 (29.3%) participants in the control group and 36/97 (37.1%) in the intervention group (p = 0.282). The relative risk of having a ‘successful treatment’ in the intervention group compared with the control group was 1.4 (95% CI 0.9 to 1.9) at 3 months and 1.3 (95% CI 0.8 to 1.9) at 12 months.
The effect of botulinum toxin upon upper limb function was also examined by analysing change in ARAT score from baseline to each assessment (Tables 8a and 8b). No significant differences were seen between intervention and control groups at 1 or 12 months. At 3 months, although the median change in upper limb function in both randomisation groups was zero, the Mann–Whitney U-test reached statistical significance (p = 0.049). Examination of the difference between the mean change in the groups showed that the intervention group had improved by a mean of 1.8 (95% CI 0.4 to 3.2) points on the ARAT compared with the control group. Although this demonstrates improved upper limb function in the intervention group compared with the control group, the size of the improvement is small and therefore of doubtful clinical significance.
Control | Intervention | p-value | |
---|---|---|---|
ARAT: median change (IQR) | |||
1 month | 0 (0 to 3) (n = 154) | 0 (0 to 4) (n = 167) | 0.427 |
3 months | 0 (0 to 3) (n = 151) | 0 (0 to 5) (n = 161) | 0.049 |
12 months | 0 (0 to 3) (n = 92) | 1 (0 to 5) (n = 97) | 0.227 |
Nine-Hole Peg Test (pegs placed in 50s): median change (IQR) | |||
1 month | 0 (0 to 0) (n = 155) | 0 (0 to 0) (n = 166) | 0.150 |
3 months | 0 (0 to 0) (n = 151) | 0 (0 to 0) (n = 162) | 0.062 |
12 months | 0 (0 to 0) (n = 92) | 0 (0 to 0) (n = 97) | 0.498 |
Upper limb functional activities: median change (IQR) | |||
Put arm through sleeve | |||
1 month | 0 ( to 0.5 to 1) (n = 125) | 0 (0 to 1) (n = 144) | 0.004 |
3 months | 0 (0 to 1) (n = 122) | 0 (0 to 1) (n = 142) | 0.127 |
12 months | 0 ( to 1 to 1) (n = 79) | 0 ( to 0.3 to 1) (n = 86) | 0.956 |
Open the hand for cleaning your palm | |||
1 month | 0 (0 to 1) (n = 124) | 0 (0 to 1) (n = 143) | 0.071 |
3 months | 0 (– 1 to 1) (n = 122) | 0 (–1 to 1) (n = 142) | 0.047 |
12 months | 0 (– 1 to 1) (n = 79) | 0 (0 to 1.3) (n = 86) | 0.029 |
Open the hand for cutting fingernails | |||
1 month | 0 (0 to 0.5) (n = 125) | 0 (– 1 to 1) (n = 143) | 0.526 |
3 months | 0 (– 1 to 1) (n = 122) | 0 (– 1 to 1) (n = 141) | 0.342 |
12 months | 0 (– 1 to 1) (n = 78) | 0 (– 0.3 to 2) (n = 86) | 0.097 |
Use cutlery | |||
1 month | 0 (0 to 0) (n = 123) | 0 (0 to 0) (n = 141) | 0.376 |
3 months | 0 (0 to 0) (n = 120) | 0 (0 to 0) (n = 140) | 0.595 |
12 months | 0 (0 to 0) (n = 77) | 0 (0 to 0) (n = 83) | 0.066 |
Barthel ADL Index: median change (IQR) | |||
1 month | 0 (– 2 to 1) (n = 134) | 0 (– 2 to 1) (n = 142) | 0.335 |
3 months | 0 (– 2 to 1) (n = 130) | 0 (– 2 to 2) (n = 143) | 0.260 |
12 months | – 1 (– 2 to 1) (n = 75) | – 1 (– 3 to 1.3) (n = 82) | 0.833 |
Control | Intervention | Difference | |
---|---|---|---|
ARAT: mean change (95% CI) | |||
1 month | 1.5 (0.8 to 2.2) | 2.2 (1.3 to 3.0) | 0.7 (– 0.4 to 1.8) |
3 months | 1.3 (0.4 to 2.1) | 3.0 (2.0 to 4.2) | 1.8 (0.4 to 3.2) |
12 months | 2.0 (– 0.5 to 0.1) | –3.1 (1.7 to 4.5) | 1.1 (– 0.7 to 2.9) |
Nine-Hole Peg Test (pegs placed in 50s): mean change (95% CI) | |||
1 month | 0.1 (– 0.1 to 0.3) | 0.3 (0.0 to 0.5) | 0.2 (– 0.1 to 0.5) |
3 months | 0.1 (– 0.1 to 0.3) | 0.5 (0.2 to 0.9) | 0.5 (0.1 to 0.8) |
12 months | 0.2 (0.0 to 0.3) | 0.3 (0.0 to 0.7) | 0.1 (– 0.2 to 0.6) |
Upper limb functional activities: mean change (95% CI) | |||
Put arm through sleeve | |||
1 month | 0.0 (– 0.2 to 0.2) | 0.4 (0.2 to 0.6) | 0.4 (0.1 to 0.6) |
3 months | 0.1 (– 0.1 to 0.3) | 0.3 (0.1 to 0.5) | 0.2 (– 0.1 to 0.5) |
12 months | 0.1 (– 0.2 to 0.3) | 0.1 (– 0.2 to 0.3) | 0.0 (– 0.4 to 0.4) |
Open the hand for cleaning your palm | |||
1 month | 0.1 (– 0.1 to 0.3) | 0.4 (0.2 to 0.6) | 0.3 (0.0 to 0.6) |
3 months | 0.0 (– 0.3 to 0.2) | 0.3 (0.0 to 0.5) | 0.3 (– 0.1 to 0.7) |
12 months | – 0.1 (– 0.4 to 0.2) | 0.4 (0.1 to 0.8) | 0.5 (0.0 to 1.0) |
Open the hand for cutting fingernails | |||
1 month | 0.1 (– 0.1 to 0.4) | 0.2 (– 0.1 to 0.4) | 0.1 (– 0.3 to 0.4) |
3 months | 0.0 (– 0.3 to 0.2) | 0.1 (– 0.2 to 0.3) | 0.1 (– 0.3 to 0.5) |
12 months | 0.0 (– 0.4 to 0.4) | 0.3 (– 0.1 to 0.7) | 0.3 (– 0.2 to 0.9) |
Use cutlery | |||
1 month | 0.1 (0.0 to 0.2) | 0.2 (0.1 to 0.3) | 0.1 (– 0.1 to 0.3) |
3 months | 0.2 (0.0 to 0.3) | 0.2 (0.1 to 0.4) | 0.1 (– 0.1 to 0.3) |
12 months | – 0.1 (– 0.3 to 0.1) | 0.2 (0.0 to 0.2) | 0.3 (0 to 0.5) |
Barthel ADL Index: mean change (IQR) | |||
1 month | – 0.6 (– 1.0 to 0.2) | – 0.4 (– 0.8 to 0.1) | 0.2 (– 0.4 to 0.8) |
3 months | – 0.3 (– 0.8 to 0.1) | 0.0 (– 0.5 to 0.4) | 0.3 (– 0.3 to 0.9) |
12 months | – 0.5 (– 1.1 to 0.2) | – 0.4 (– 1 to 0.2) | 0.1 (– 0.8 to 1.0) |
Although the median change in score from baseline to 1 month for the ability put the affected arm through a sleeve was zero in both the control and intervention groups, comparison of scores reached statistical significance (p = 0.004). Examination of the difference between the mean change in the groups showed that the intervention group had improved by 0.4 (95% CI 0.1 to 0.6) compared with the control group (Tables 8a and 8b). Similarly, at 3 and 12 months there were statistically significant differences between intervention and control groups in ability to open the hand for cleaning the palm despite the median change from baseline being zero in both groups. Further examination of the data showed that the intervention group had improved by 0.3 (95% CI – 0.1 to 0.7) compared with the control group at 3 months and by 0.5 (95% CI 0.0 to 1.0) at 12 months. As self-reported arm function was assessed using an ordinal scale of 1 (unable to perform) to 5 (no difficulty), a mean improvement of 0.3–0.5 is of doubtful clinical importance.
To enable comparison with previous studies, responses to basic upper limb functional activity questions were also analysed by comparing the proportion of participants in each randomisation group who had improved by one or more points on the scale from baseline (Table 8c). For the ability to dress a sleeve, this improvement was seen for 65/144 (45.1%) of participants in the intervention group compared with 38/125 (30.4%) in the control group at 1 month (p = 0.017). No significant differences were seen at 3 and 12 months. For opening the hand to clean the palm and opening the hand to cut fingernails, significant differences in favour of the intervention group were seen at 1, 3 and 12 months. No significant differences were seen between the groups for improvement in ability to use cutlery.
Overall, at 1 month 109/144 (75.7%) of the intervention group and 79/125 (63.2%) of the control group had improved by at least one point on any of the four tasks (p = 0.033). At 3 months the corresponding proportions were 102/142 (71.8%) of the intervention group and 71/122 (58.2%) of the control group (p = 0.027). No significant difference was seen at 12 months [intervention group 59/86 (68.6%), control group 51/79 (64.6%), p = 0.622].
Control | Intervention | p-value | Relative risk Intervention: Control (95% CI) | |
---|---|---|---|---|
Dressing sleeve improvement by ≥ 1: n (%) | ||||
1 month | 38 (30.4) (n = 125) | 65 (45.1) (n = 144) | 0.017 | 1.5 (1.1 to 2.0) |
3 months | 39 (32.0) (n = 122) | 62 (43.7) (n = 142) | 0.057 | 1.4 (1.0 to 1.9) |
12 months | 32 (40.5) (n = 79) | 30 (34.9) (n = 86) | 0.521 | 0.9 (0.6 to 1.3) |
Opening hand for cleaning palm improvement of ≥ 1: n (%) | ||||
1 month | 41 (33.1) (n = 124) | 65 (45.5) (n = 143) | 0.045 | 1.4 (1.0 to 1.8) |
3 months | 34 (27.9) (n = 122) | 64 (45.1) (n = 142) | 0.005 | 1.6 (1.2 to 2.3) |
12 months | 25 (31.6) (n = 79) | 41 (47.7) (n = 86) | 0.040 | 1.5 (1.0 to 2.2) |
Opening the hand for cutting nails improvement of ≥ 1: n (%) | ||||
1 month | 31 (24.8) (n = 125) | 52 (36.6) (n = 142) | 0.047 | 1.5 (1.0 to 2.2) |
3 months | 31 (25.4) (n = 122) | 52 (36.9) (n = 141) | 0.048 | 1.5 (1.0 to 2.2) |
12 months | 21 (26.9) (n = 78) | 39 (45.3) (n = 86) | 0.016 | 1.7 (1.1 to 2.6) |
Ability to use cutlery improvement of ≥ 1: n (%) | ||||
1 month | 22 (17.9) (n = 123) | 31 (22.0) (n = 141) | 0.444 | 1.2 (0.8 to 2.0) |
3 months | 25 (20.8) (n = 120) | 31 (22.1) (n = 140) | 0.880 | 1.1 (0.7 to 1.7) |
12 months | 10 (13.0) (n = 77) | 17 (20.5) (n = 83) | 0.291 | 1.6 (0.8 to 3.2) |
Stroke-related quality of life/participation restriction
Tables 9a and 9b show stroke-related quality of life/participation restriction at 1, 3 and 12 months. For the EQ-5D question about pain and discomfort, although the median change in score from baseline to 3 months was zero in both groups, comparison of scores between the groups reached statistical significance (p = 0.025). Examination of the difference between mean change in the groups showed that the intervention group had improved (decreased) their score by 0.2 points compared with the control group. Similarly, the median change in score from baseline to 3 months on the Oxford Handicap Scale was zero in both groups, but comparison of scores between the groups reached statistical significance (p = 0.015). The mean change in the groups showed that the intervention had improved (decreased) their score by 0.3 points compared with the control group.
Control | Intervention | p-value | |
---|---|---|---|
Stroke Impact Scale domains: median change (IQR) | |||
Strength | |||
1 month | 0.0 (– 6.3 to 12.5) (n = 124) | 0.0 (– 6.3 to 12.5) (n = 142) | 0.544 |
3 months | 0.0 (– 12.5 to 6.3) (n = 117) | 0.0 (– 12.5 to 6.3) (n = 140) | 0.784 |
12 months | 0.0 (– 12.5 to 12.5) (n = 76) | 0.0 (– 12.5 to 10.9) (n = 84) | 0.408 |
Memory | |||
1 month | 0.0 (– 7.1 to 7.1) (n = 125) | 0.0 (– 6.3 to 7.1) (n = 144) | 0.204 |
3 months | 0.0 (– 10.7 to 10.7) (n = 122) | 0.0 (– 7.1 to 10.7) (n = 143) | 0.674 |
12 months | – 3.6 (– 14.3 to 0.0) (n = 79) | 0.0 (– 10.7 to 7.1) (n = 86) | 0.060 |
Emotion | |||
1 month | 0.0 (– 5.6 to 8.3) (n = 123) | 0.0 (– 8.3 to 8.3) (n = 143) | 0.831 |
3 months | 0.0 (– 8.3 to 8.3) (n = 120) | 0.0 (– 12.6 to 8.3) (n = 140) | 0.771 |
12 months | – 5.6 (– 13.9 to 5.6) (n = 78) | 0.0 (– 8.3 to 8.3) (n = 86) | 0.092 |
Communication | |||
1 month | 0.0 (– 7.1 to 3.6) (n = 125) | 0.0 (– 3.6 to 3.6) (n = 144) | 0.519 |
3 months | 0.0 (– 7.1 to 3.6) (n = 122) | 0.0 (– 3.6 to 7.1) (n = 141) | 0.115 |
12 months | – 3.6 (– 10.7 to 3.6) (n = 79) | 0.0 (– 7.1 to 7.1) (n = 86) | 0.023 |
ADL | |||
1 month | 0.0 (– 7.5 to 5.0) (n = 125) | 0.3 (– 5.0 to 10.0) (n = 143) | 0.078 |
3 months | 0.0 (– 7.5 to 7.5) (n = 122) | 0.0 (– 6.3 to 7.5) (n = 142) | 0.642 |
12 months | – 2.5 (– 10.0 to 5.0) (n = 79) | 0.0 (– 7.5 to 10.0) (n = 85) | 0.189 |
Mobility | |||
1 month | 0.0 (– 8.3 to 5.6) (n = 124) | 0.0 (– 5.6 to 8.3) (n = 142) | 0.217 |
3 months | 2.8 (– 8.3 to 11.1) (n = 119) | 2.8 (– 5.6 to 8.3) (n = 140) | 0.686 |
12 months | – 1.4 (– 11.1 to 8.3) (n = 78) | 0.0 (– 8.8 to 8.3) (n = 86) | 0.686 |
Hand function | |||
1 month | 0.0 (0.0–10.0) (n = 124) | 0.0 (0.0 to 10.0) (n = 142) | 0.387 |
3 months | 0.0 (0.0–5.0) (n = 120) | 0.0 (0.0 to 10.0) (n = 141) | 0.908 |
12 months | 0.0 (– 2.5 to 0.0) (n = 77) | 0.0 (0.0 to 11.3) (n = 86) | 0.096 |
Participation/Handicap | |||
1 month | – 2.3 (– 12.5 to 9.4) (n = 124) | 0.0 (– 9.4 to 13.5) (n = 142) | 0.122 |
3 months | 0.0 (– 18.7 to 14.5) (n = 122) | 3.1 (– 9.4 to 21.9) (n = 140) | 0.091 |
12 months | 0.0 (– 18.0 to 12.5) (n = 76) | 3.1 (– 12.5 to 18.8) (n = 85) | 0.241 |
Physical domain | |||
1 month | 1.0 (– 5.0 to 6.2) (n = 126) | 2.0 (– 2.7 to 7.5) (n = 144) | 0.125 |
3 months | 1.1 (– 5.6 to 7.1) (n = 123) | 2.0 (– 4.6 to 7.0) (n = 143) | 0.790 |
12 months | – 1.8 (– 6.8 to 5.0) (n = 79) | – 0.4 (– 7.1 to 6.9) (n = 86) | 0.534 |
Stroke recovery | |||
1 month | 0.0 (– 10.0 to 5.0) (n = 121) | 0.0 (– 12.5 to 10.0) (n = 141) | 0.352 |
3 months | 0.0 (– 10.0 to 10.0) (n = 117) | 0.0 (– 10.0 to 15.0) (n = 141) | 0.464 |
12 months | 0.0 (– 20.0 to 10.0) (n = 74) | 0.0 (– 20.0 to 20.0) (n =86 ) | 0.369 |
EQ-5D: median change (IQR) | |||
Mobility | |||
1 month | 0 (0 to 0) (n = 138) | 0 (0 to 0) (n = 151) | 0.914 |
3 months | 0 (0 to 0) (n = 134) | 0 (0 to 0) (n = 151) | 0.445 |
12 months | 0 (0 to 0) (n = 83) | 0 (0 to 0) (n = 87) | 0.542 |
Self-care | |||
1 month | 0 (0 to 0) (n = 138) | 0 (0 to 0) (n = 151) | 0.255 |
3 months | 0 (0 to 0) (n = 134) | 0 (0 to 0) (n = 152) | 0.256 |
12 months | 0 (0 to 0) (n = 82) | 0 (0 to 0) (n = 87) | 0.576 |
Usual activities | |||
1 month | 0 (0 to 0) (n = 138) | 0 (0 to 0) (n = 149) | 0.764 |
3 months | 0 (0 to 0) (n = 134) | 0 (0 to 0) (n = 151) | 0.311 |
12 months | 0 (0 to 0) (n = 83) | 0 (0 to 0) (n = 86) | 0.443 |
Pain/discomfort | |||
1 month | 0 (0 to 0) (n = 137) | 0 (0 to 0) (n = 150) | 0.247 |
3 months | 0 (0 to 0) (n = 133) | 0 (0 to 0) (n = 152) | 0.025 |
12 months | 0 (0 to 1) (n = 83) | 0 (0 to 0) (n = 87) | 0.270 |
Anxiety/depression | |||
1 month | 0 (0 to 0) (n = 133) | 0 (0 to 0) (n = 149) | 0.138 |
3 months | 0 (0 to 0) (n = 132) | 0 (0 to 0) (n = 151) | 0.818 |
12 months | 0 (0 to 1) (n = 81) | 0 (– 1 to 0) (n =86 ) | 0.002 |
Good/bad health scale | |||
1 month | – 5 (– 20 to 10) (n = 135) | – 1 (– 18 to 10) (n = 149) | 0.663 |
3 months | – 1 (– 22.5 to 15) (n = 133) | – 5 (– 20 to 10) (n = 148) | 0.755 |
12 months | 0 (– 25.8 to 18.8) (n = 80) | – 5 (– 20 to 10) (n = 84) | 0.749 |
Oxford Handicap Scale: median change (IQR) | |||
1 month | 0 (– 0.5 to 0) (n = 137) | 0 (–1 to 0) (n = 152) | 0.359 |
3 months | 0 (0 to 0) (n = 133) | 0 (–1 to 0) (n = 151) | 0.015 |
12 months | 0 (– 1 to 0) (n = 83) | 0 (–1 to 0) (n = 87) | 0.045 |
Control | Intervention | Difference | |
---|---|---|---|
Stroke Impact Scale domains: mean change (95% CI) | |||
Strength | |||
1 month | 0.9 (– 1.6 to 3.4) | 1.7 (– 1.1 to 4.4) | 0.8 (– 2.9 to 4.6) |
3 months | – 1.6 (– 5.1 to 1.8) | – 0.2 (– 3.4 to 3.0) | 1.4 (– 3.3 to 6.2) |
12 months | 0.2 (– 4.2 to 4.5) | – 2.2 (– 6.5 to 2.2) | – 2.3 (– 8.3 to 4.0) |
Memory | |||
1 month | – 1.1 (– 3.7 to 1.5) | 1.3 (– 1.6 to 4.2) | 2.4 (– 1.5 to 6.3) |
3 months | – 2.0 (– 5.0 to 1.0) | – 0.8 (– 2.3 to 4.0) | 2.8 (– 1.5 to 7.2) |
12 months | – 5.6 (– 9.6 to – 1.5) | – 1.8 (– 5.6 to 1.8) | 3.8 (– 1.7 to 9.2) |
Emotion | |||
1 month | 0.6 (– 2.0 to 3.1) | 0.3 (– 1.8 to 2.4) | – 0.3 (– 3.6 to 3.0) |
3 months | – 0.1 (– 2.8 to 2.6) | – 1.0 (– 3.4 to 1.5) | – 0.9 (– 4.6 to 2.8) |
12 months | – 3.5 (– 6.9 to – 0.1) | – 1.0 (– 4.0 to 1.9) | 2.5 (– 2.0 to 7.0) |
Communication | |||
1 month | – 1.6 (– 3.6 to 0.4) | 0.6 (– 1.7 to 2.8) | 2.1 (– 0.9 to 5.1) |
3 months | – 2.4 (– 5.3 to 0.3) | 0.3 (– 2.2 to 2.7) | 4.7 (1.1 to 8.5) |
12 months | – 4.2 (– 8.1 to – 0.5) | 1.2 (– 2.4 to 4.7) | 5.3 (0.2 to 10.6) |
ADL | |||
1 month | – 1.6 (– 3.8 to 0.5) | 1.8 (– 0.5 to 4.1) | 3.4 (0.4 to 6.6) |
3 months | – 1.0 (– 3.7 to 1.4) | 2.5 (0.0 to 5.0) | 1.4 (– 2.2 to 4.9) |
12 months | – 2.4 (– 5.5 to 0.7) | 0.8 (– 2.3 to 3.8) | 3.2 (– 1.1 to 7.5) |
Mobility | |||
1 month | – 1.0 (– 3.6 to 1.7) | 1.1 (– 1.4 to 3.5) | 2.1 (– 1.5 to 5.7) |
3 months | 1.7 (– 1.3 to 4.7) | 2.9 (– 0.5 to 6.2) | 0.8 (– 3.1 to 4.6) |
12 months | – 2.0 (– 5.4 to 1.4) | – 0.8 (– 3.9 to 2.2) | 1.2 (– 3.5 to 5.8) |
Hand function | |||
1 month | 3.3 (0.2 to 6.1) | 4.7 (1.3 to 8.0) | 1.5 (– 3.0 to 5.8) |
3 months | 3.2 (– 0.5 to 6.8) | 5.0 (– 0.5 to 10.4) | – 0.3 (– 5.2 to 4.7) |
12 months | – 0.9 (– 5.7 to 3.6) | 4.6 (1.0 to 8.5) | 5.6 (– 0.2 to 11.6) |
Participation/Handicap | |||
1 month | – 2.8 (– 7.8 to 2.3) | 1.2 (– 4.4 to 6.8) | 4.0 (– 3.5 to 11.6) |
3 months | – 2.0 (– 6.5 to 2.6) | 1.4 (– 0.6 to 3.4) | 7.0 (– 0.1 to 14.2) |
12 months | – 1.7 (– 7.6 to 4.2) | 4.2 (– 2.4 to 10.7) | 5.9 (– 3.0 to 14.7 |
Physical domain | |||
1 month | 0.6 (– 1.0 to 2.1) | 2.1 (0.2 to 3.8) | 1.5 (– 1.0 to 3.9) |
3 months | 0.9 (– 1.2 to 3.1) | 1.4 (– 0.6 to 3.4) | 0.4 (– 2.6 to 3.4) |
12 months | – 1.2 (– 3.8 to 1.2) | 0.5 (– 1.9 to 2.9) | 1.7 (– 1.8 to 5.2) |
Stroke recovery | |||
1 month | – 2.1 (– 4.6 to 0.3) | – 0.6 (– 3.8 to 2.5) | 1.6 (– 2.5 to 5.5) |
3 months | – 0.8 (– 3.7 to 2.1) | 2.0 (– 1.3 to 5.4) | 2.8 (– 1.5 to 7.2) |
12 months | – 2.1 (– 6.8 to 2.7) | 0.5 (– 4.5 to 5.7) | 2.6 (– 4.2 to 9.4) |
EQ-5D: mean change (95% CI) | |||
Mobility | |||
1 month | 0.0 (– 0.1 to 0.1) | 0.0 (– 0.1 to 0.1) | 0.0 (– 0.1 to 0.1) |
3 months | 0.0 (– 0.1 to 0.0) | 0.0 (– 0.1 to 0.0) | 0.0 (– 0.1 to 0.1) |
12 months | 0.0 (– 0.1 to 0.1) | 0.0 (0.0 to 0.1) | 0.0 (– 0.1 to 0.2) |
Self-care | |||
1 month | 0.1 (0.1 to 0.2) | 0.1 (0.0 to 0.2) | – 0.1 (– 0.2 to 0.0) |
3 months | 0.1 (0.1 to 0.2) | 0.1 (0.0 to 0.2) | – 0.1 (– 0.2 to 0.0) |
12 months | 0.1 (0.0 to 0.3) | 0.1 (0.0 to 0.2) | – 0.1 (– 0.2 to 0.1) |
Usual activities | |||
1 month | 0.0 (– 0.1 to 0.1) | 0.0 (– 0.1 to 0.1) | 0.0 (– 0.1 to 0.2) |
3 months | 0.0 (– 0.1 to 0.1) | 0.0 (– 0.1 to 0.2) | 0.1 (– 0.1 to 0.2) |
12 months | 0.0 (– 0.1 to 0.2) | 0.1 (0.0 to 0.2) | 0.1 (– 0.1 to 0.3) |
Pain/discomfort | |||
1 month | 0.1 (0.0 to 0.2) | 0.0 (– 0.1 to 0.1) | – 0.1 (– 0.2 to 0.1) |
3 months | 0.1 (0.0 to 0.2) | – 0.1 (– 0.2 to 0.0) | – 0.2 (– 0.3 to 0.0) |
12 months | 0.1 (0.0 to 0.3) | 0.0 (– 0.1 to 0.2) | – 0.1 (– 0.3 to 0.1) |
Anxiety/depression | |||
1 month | 0.0 (– 0.1 to 0.1) | – 0.1 (– 0.2 to 0.0) | – 0.1 (– 0.3 to 0.0) |
3 months | – 0.1 (– 0.2 to 0.1) | 0.0 (– 0.1 to 0.1) | 0.0 (– 0.1 to 0.2) |
12 months | 0.1 (0.0 to 0.3) | – 0.2 (– 0.3 to – 0.1) | – 0.3 (– 0.5 to – 0.1) |
Good/bad health scale | |||
1 month | – 5.2 (– 9.4 to – 0.9) | – 4.9 (– 8.6 to – 1.4) | 0.2 (– 5.4 to 5.7) |
3 months | – 4.9 (– 9.4 to – 0.5) | – 5.6 (– 9.6 to – 1.6) | – 0.7 (– 6.6 to 5.3) |
12 months | – 2.7 (– 9.3 to 4.0) | – 4.8 (– 10.6 to 0.9) | – 2.1 (– 10.9 to 6.5) |
Oxford Handicap Scale: mean change (95% CI) | |||
1 month | 0.0 (– 0.2 to 0.1) | – 0.1 (– 0.3 to 0.0) | – 0.1 (– 0.3 to 0.1) |
3 months | 0.0 (– 0.2 to 0.1) | – 0.3 (– 0.4 to – 0.1) | – 0.3 (– 0.5 to – 0.1) |
12 months | 0.0 (– 0.2 to 0.2) | – 0.3 (– 0.5 to – 0.1) | – 0.3 (– 0.6 to 0.0) |
Statistically significant differences were also found between the groups for change in participation restriction from baseline to 12 months as measured by the communication domain of the Stroke Impact Scale, the EQ-5D question about anxiety and depression, and the Oxford Handicap Scale. On all occasions where significant differences were found, however, the magnitude of the treatment effect was small and of doubtful clinical importance.
Upper limb pain
No significant differences were found in change in pain rating on either pain scale from baseline to 1 month or 3 months (Tables 10a and 10b). However, pain rating did decrease from baseline to 12 months in the intervention group compared with the control group on both of the pain scales. For the pain rating scale 0–10, the median change was a decrease of two points in the intervention group compared with zero points in the control group (p = 0.004).
Control | Intervention | p-value | |
---|---|---|---|
Pain description: median change (IQR) | |||
1 month | 0 (0 to 1) (n = 133) | 0 (0 to 1) (n = 133) | 0.119 |
3 months | 0 (0 to 1) (n = 133) | 0 (0 to 2) (n = 133) | 0.259 |
12 months | 0 (0 to 1) (n = 133) | 0 (0 to 2) (n = 133) | 0.036 |
Pain score (0–10): median change (IQR) | |||
1 month | 0 (– 3 to 0) (n = 15) | 0 (– 4 to 0) (n = 167) | 0.600 |
3 months | 0 (– 4 to 1) (n = 149) | 0 (– 4 to 0) (n = 163) | 0.269 |
12 months | 0 (– 2.8 to 1) (n = 92) | – 2 (– 5 to 0) (n = 97) | 0.004 |
Control | Intervention | Difference | |
---|---|---|---|
Pain description: mean change (95% CI) | |||
1 month | 0.3 (0.1 to 0.6) | 0.6 (0.4 to 0.8) | 0.2 (– 0.1 to 0.5) |
3 months | 0.4 (0.2 to 0.7) | 0.6 (0.4 to 0.8) | 0.2 (– 0.1 to 0.5) |
12 months | 0.4 (0.1 to 0.6) | 0.8 (0.5 to 1.1) | 0.5 (0.1 to 0.8) |
Pain score (0–10): mean change (95% CI) | |||
1 month | – 1.1 (– 1.6 to –0.6) | – 1.3 (– 1.9 to – 0.8) | – 0.3 (– 1.0 to 0.5) |
3 months | – 1.2 (– 1.8 to –0.6) | – 1.6 (– 2.2 to 1.1) | – 0.4 (– 1.2 to 0.3) |
12 months | – 0.8 (– 1.5 to 0.1) | – 2.2 (– 2.9 to – 1.4) | – 1.4 (– 2.4 to – 0.3) |
Additional antispasticity treatment
Antispasticity medication and physical treatments such as splints were used infrequently and use was similar in both groups (Table 11). At 1 month, 3 months and 12 months, 41%, 39% and 36% were using some form of antispasticity treatment, respectively.
Control | Intervention | p-value | |
---|---|---|---|
Antispasticity treatments: n (%) | |||
1 month | 67 (43.2) (n = 155) | 65 (38.9) (n = 167) | 0.496 |
3 months | 62 (41.1) (n = 151) | 62 (38.0) (n = 163) | 0.644 |
12 months | 33 (35.9) (n = 92) | 36 (37.1) (n = 97) | 0.881 |
Dantrolene | |||
1 month | 2 (1.3) | 3 (1.8) | 1.000 |
3 months | 3 (2.0) | 3 (1.8) | 0.500 |
12 months | 0 (0.0) | 1 (1.0) | 1.000 |
Baclofen | |||
1 month | 15 (9.7) | 22 (13.2) | 0.383 |
3 months | 17 (11.3) | 23 (14.1) | 0.500 |
12 months | 11 (12.0) | 14 (14.4) | 0.671 |
Tizanidine | |||
1 month | 0 (0.0) | 2 (1.2) | 0.499 |
3 months | 0 (0.0) | 2 (1.2) | 0.499 |
12 months | 0 (0.0) | 1 (1.0) | 1.000 |
Gabapentin | |||
1 month | 17 (11.0) | 16 (9.6) | 0.716 |
3 months | 17 (11.3) | 15 (9.2) | 0.580 |
12 months | 11 (12.0) | 10 (10.3) | 0.818 |
Methocarbanol | |||
1 month | 1 (0.6) | 0 (0.0) | 0.481 |
3 months | 1 (0.7) | 0 (0.0) | 0.481 |
12 months | 0 (0.0) | 0 (0.0) | – |
Thumb strap | |||
1 month | 1 (0.6) | 0 (0.0) | 0.481 |
3 months | 0 (0.0) | 0 (0.0) | – |
12 months | 0 (0.0) | 0 (0.0) | – |
Elasticated glove | |||
1 month | 1 (0.6) | 0 (0.0) | 0.481 |
3 months | 1 (0.7) | 0 (0.0) | 0.481 |
12 months | 0 (0.0) | 0 (0.0) | – |
Functional electrical stimulation machine | |||
1 month | 1 (0.6) | 1 (0.6) | 1.000 |
3 months | 2 (1.3) | 1 (0.6) | 0.610 |
12 months | 0 (0.0) | 1 (1.0) | 1.000 |
TENS machine | |||
1 month | 1 (0.6) | 3 (1.8) | 0.249 |
3 months | 0 (0.0) | 1 (0.6) | 1.000 |
12 months | 0 (0.0) | 0 (0.0) | – |
Upper limb splint | |||
1 month | 45 (29.0) | 39 (23.4) | 0.256 |
3 months | 35 (23.2) | 32 (19.6) | 0.492 |
12 months | 16 (17.4) | 18 (18.6) | 0.852 |
Outcome assessment timing
One-month outcome assessments were scheduled for completion 28 +/– 3 days after randomisation. In the control group, 78.1% of assessments were performed within this time window and in the intervention group it was 81.4%. The median time from randomisation to the 1-month outcome was 30 days (IQR 28–31 days) for both intervention and control groups. Three-month assessments were scheduled for 84 +/– 5 days after study entry. In the control group, 81.6% were completed in the time window and in the intervention group it was 85.5%. The median time from randomisation to the 3-month assessment was 85 days (IQR 83.5–87 days) in the control group and 85 days (IQR 83–87 days) in the intervention group. Twelve-month assessments were scheduled for 336 +/– 5 days after study entry. In the control group, 68.5% of assessments were performed on time and in the intervention group it was 81.4%. The median time from randomisation to 12-month assessment was 337 days (IQR 334–341.5 days) in the control group and 336 days (IQR 335.75–339 days) in the intervention group.
Blinding of outcome assessments
The study aimed for outcome assessments to be performed by an assessor who was blinded to the randomisation group. This was achieved by having assessments carried out by a therapist who had not been involved in providing the upper limb therapy programme for that participant. As participants were not blinded it was possible for the therapist to become aware of the randomisation group. Correct identification of treatment group by the outcome assessor occurred for 36.1% (95% CI 30.8 to 41.6), 44.1% (95% CI 38.5 to 49.8) and 58.6% (95% CI 51.2 to 65.8), respectively, at 1, 3 and 12 months.
Patient-selected goals
The COPM71 was used to identify and measure patient-selected goals. Participants were asked at the first session of the upper limb therapy programme to identify and prioritise activities they would like to be able to undertake in three occupational areas – self-care, productivity and leisure. Each of these areas was subdivided into: (1) self-care: personal care, functional mobility, community management; (2) productivity: paid/unpaid work, household management, play/school; and (3) leisure: quiet recreation, active recreation and socialising. Participants were then asked to select up to five activities they would like to be able to undertake by the end of the therapy programme. These activities were then a focus for work during therapy sessions.
The COPM was completed by 145/163 (89.0%) participants in the control group and 155/170 (91.2%) in the intervention group. Participants in both groups tended to select self-care activities and the majority of these were related to personal care. The other common activity choices were household management and quiet recreation. The median number of final activities chosen in the control group was four (IQR 3–5) and in the intervention group was four (IQR 3–5).
According to the standard COPM protocol, participants scored each final activity out of 10 for both performance and satisfaction. A total baseline performance and satisfaction score was calculated (scores were added and divided by the number of activities). At 1 month, the COPM assessment was completed by rescoring the final activities. Changes in performance and satisfaction were calculated (Table 12).
Control (n = 145) | Intervention (n = 155) | p-value | |
---|---|---|---|
Baseline performance: median (IQR) | 1.7 (1.0 to 2.8) | 2.0 (1.0 to 3.0) | 0.143 |
Post-treatment performance: median (IQR) | 4.7 (3.1 to 6.7) | 4.8 (3.4 to 6.0) | 0.985 |
Change in performance: median (IQR) | 2.3 (1.3 to 4.0) | 2.3 (1.2 to 3.8) | 0.535 |
Baseline satisfaction: median (IQR) | 1.6 (1.0 to 2.8) | 2.0 (1.0 to 3.3) | 0.152 |
Post-treatment satisfaction: median (IQR) | 4.8 (3.0 to 6.8) | 4.8 (3.5 to 6.2) | 0.792 |
Change in satisfaction: median (IQR) | 2.4 (1.0 to 4.4) | 2.3 (1.0 to 4.0) | 0.342 |
As the COPM was part of the upper limb therapy programme, the assessment was undertaken by the treating physiotherapist rather than a therapist who was blinded to the randomisation group. There were no statistically significant differences between the groups for baseline performance or satisfaction scores. Following treatment, performance and satisfaction increased in both groups but there were no statistically significant differences between the groups. Both performance and satisfaction increased by approximately two points in both groups.
Views about the study upper limb therapy programme
Participant views about the study upper limb therapy programme were sought by including four questions in the 1-month assessment: how did you find the upper limb therapy programme; what was good; what was not so good; other comments.
To analyse responses a simple coding framework was developed for each question. Responses were coded blinded to the randomisation group. A relative risk calculation was performed to compare the coded responses between the groups.
Table 13 shows comments about how participants found the upper limb therapy programme. Approximately half of the responders (50.7% control, 56.9% intervention) in both randomisation groups provided very general comments about how ‘excellent’ or ‘good’ they thought the programme had been. A further 37.2% of the control group responders and 35.6% of the intervention group felt that they had gained benefit from the therapy programme. Less favourable comments about therapy were provided by 18/148 (12.1%) in the control group and 12/160 (7.5%) in the intervention group; these were divided into responses about no benefit from the programme, the programme being too difficult or the programme being not challenging enough. There were no statistically significant differences between the groups for the type of comments made.
Code | Control (n = 148) | Intervention (n = 160) | Relative risk Intervention: Control (95% CI) | Examples |
---|---|---|---|---|
Excellent programme: n (%) | 29 (19.6) | 36 (22.5) | 1.1 (0.7 to 1.8) |
Control: excellent great treatment Intervention: the programme was terrific |
Good programme: n (%) | 46 (31.1) | 55 (34.4) | 1.1 (0.8 to 1.5) |
Control: it was good Intervention: fine, enjoyed it |
Benefit from programme: n (%) | 55 (37.2) | 57 (35.6) | 1.0 (0.7 to 1.3) |
Control: very helpful and glad to have treatment focused on arm Intervention: felt it was beneficial, has made progress |
No benefit from programme: n (%) | 5 (3.4) | 3 (1.9) | 0.6 (0.1 to 2.3) |
Control: no benefit from stretches or functional programme Intervention: worth trying but not really any good |
Difficult programme: n (%) | 8 (5.4) | 7 (4.4) | 0.8 (0.3 to 2.2) |
Control: difficult at first but got better with practice Intervention: difficult but managed |
Programme not challenging: n (%) | 5 (3.4) | 2 (1.3) | 0.4 (0.1 to 1.9) |
Control: didn’t think it was good as just repeating things Intervention: thought it was too easy |
Table 14 shows the comments of the participants about the positive aspects of the upper limb therapy programme. Nearly 50% of responders in both groups provided a comment about benefit they thought had been received from the programme, either upper limb benefit or more general benefit. Similarly, approximately 50% of responders in both groups provided a comment about an aspect of the therapy they thought had been good. These comments were about stretching, exercises and functional tasks. The relative risk calculations demonstrated that people in the intervention group were more likely to comment on contact with therapists than those in the control group (relative risk 2.3, 95% CI 1.2 to 4.2). This was the only statistically significant difference between the groups for the type of comment.
Code | Control (n = 120) | Intervention (n = 132) | Relative risk Intervention: Control (95% CI) | Examples |
---|---|---|---|---|
Gained arm benefits: n (%) | 53 (44.2) | 60 (45.5) | 1.0 (0.8 to 1.4) |
Control: being able to put things in left hand Intervention: started to get arm moving |
Gained general benefits: n (%) | 4 (3.3) | 3 (2.3) | 0.7 (0.2 to 3.0) |
Control: given confidence Intervention: felt better and more relaxed afterwards |
Therapy – stretches: n (%) | 20 (16.7) | 21 (15.9) | 1.0 (0.5 to 1.7) |
Control: enjoyed having the stretching done Intervention: the stretches reduced the upper limb stiffness and pain |
Therapy – exercises: n (%) | 21 (17.5) | 20 (15.2) | 0.9 (0.5 to 1.5) |
Control: enjoyed the exercises Intervention: exercises made arm feel more supple |
Therapy – functional tasks: n (%) | 21 (17.5) | 17 (12.9) | 0.7 (0.4 to 1.3) |
Control: being able to take jar lids and bottle lids off Intervention: being able to choose everyday tasks to practice |
Contact with therapist: n (%) | 12 (10.0) | 30 (22.7) | 2.3 (1.2 to 4.2) |
Control: enjoyed one-to-one sessions with study therapist Intervention: physio flexible to my needs |
Organisation of programme, programme as whole: n (%) | 18 (15.0) | 11 (8.3) | 0.6 (0.3 to 1.1) |
Control: the whole programme helped Intervention: very impressed with it all |
Negative comments: n (%) | 3 (2.5) | 2 (1.5) | 0.6 (0.1 to 3.6) |
Control: nothing in particular Intervention: painful shoulder limited movement |
When asked to comment about what was not so good about the therapy programme (Table 15), approximately 60% of responders in both groups said that they had no negative comments. However, 15.2% in the control group and 18.0% in the intervention group described difficulties they had found with the programme. Pain was experienced as a result of the upper limb therapy programme by 25/192 (13%). There were no statistically significant differences between the groups for the type of comments made.
Code | Control (n = 92) | Intervention (n = 100) | Relative risk Intervention: Control (95% CI) | Examples |
---|---|---|---|---|
Nothing bad: n (%) | 56 (60.9) | 58 (58.0) | 1.0 (0.8 to 1.2) |
Control: felt everything therapist did was beneficial Intervention: no negative points |
Programme difficult: n (%) | 7 (7.6) | 8 (8.0) | 1.1 (0.4 to 2.8) |
Control: found exercises difficult and frustrating Intervention: sometimes very difficult |
Specific things in programme difficult: n (%) | 7 (7.6) | 10 (10.0) | 1.3 (0.5 to 3.3) |
Control: could not manage glove Intervention: doing buttons was very difficult |
Programme caused pain: n (%) | 10 (10.9) | 15 (15.0) | 1.4 (0.7 to 2.9) |
Control: a little painful at times on stretching Intervention: pain during exercises |
Programme tiring: n (%) | 1 (1.1) | 6 (6.0) | 5.5 (0.7 to 45.5) |
Control: a bit tiring Intervention: sometimes got tired if he pushed himself too much |
No benefit from programme: n (%) | 5 (5.4) | 0 (0.0) | Control: lack of progress | |
Programme too short: n (%) | 6 (6.5) | 3 (3.0) | 0.5 (0.1 to 1.8) |
Control: too short a programme Intervention: would like more in that format” |
One hundred and fourteen participants (35.4%) provided additional comments (Table 16). The majority of these responses, 66.0% in the control group and 77.0% in the intervention group, were further positive comments about the therapy programme or therapy staff. A few responders said they were pleased to be provided with the opportunity to take part in a research study and others gave altruistic comments about wanting to help future stroke patients. About 20% of responses in both groups were less favourable comments. These were mainly about feeling the programme was too short or wanting additional treatment. There were no statistically significant differences between the groups for the types of comments made.
Code | Control (n = 53) | Intervention (n = 61) | Relative risk Intervention: Control (95% CI) |
Example |
---|---|---|---|---|
Benefit from programme: n (%) | 19 (35.8) | 26 (42.6) | 1.2 (0.7 to 1.9) |
Control: like the feeling of achieving tasks that he thought he could no longer do Intervention: very helpful, improved a lot |
Enjoyment of programme: n (%) | 9 (17.0) | 8 (13.1) | 0.8 (0.3 to 1.9) |
Control: really good, I liked it, it has given me something to work on Intervention: enjoyed the therapy |
Positive comments about staff: n (%) | 7 (13.2) | 13 (21.3) | 1.6 (0.7 to 3.7) |
Control: enjoyed working with therapist Intervention: therapist very encouraging with exercises |
Opportunity to take part: n (%) | 5 (9.4) | 1 (1.6) | 0.2 (0.02 to 1.4) |
Control: pleased to have participated Intervention: appreciative of opportunity to take part |
Altruism for future stroke survivors: n (%) | 1 (1.9) | 2 (3.3) | 1.7 (0.2 to 18.5) |
Control: grateful, glad to have helped Intervention: hoping the research will help people in future |
Negative comments: n (%) | 12 (22.6) | 11 (18.0) | 0.8 (0.4 to 1.7) |
Control: didn’t feel it was long enough Intervention: needs more exercises for fingers |
Trial treatments
Botulinum toxin
Participants in the intervention group received botulinum toxin type A injections to the upper limb immediately following study entry, plus repeat injections at 3 , 6 and 9 months if clinically indicated and they remained in the study for 12-month follow-up.
An initial set of injections was received by 164/170 (96.5%) intervention group participants. Six intervention group participants did not receive the planned initial treatment; three withdrew from the study before treatment, two became unwell following randomisation and a decision was taken not use botulinum toxin (although they did receive upper limb therapy) and one participant had insufficient hypertonicity when seen in the study injection clinic. The latter participant was believed to have variable tone, having been assessed as having increased tone at the screening visit. She remained in the intervention group for the purpose of analysis.
At 3, 6 and 9 months, further injections were received by 71/105 (67.7%), 64/105 (61.0%) and 54/105 (51.4%) intervention group participants, respectively. Muscles treated and botulinum toxin type A doses used are shown in Table 17. Muscles treated were also grouped as limb areas (Table 18). The median (IQR) total botulinum toxin type A doses per participant at initial treatment, 3 months, 6 months and 9 months were 200 units (100–300), 300 units (150–400), 300 units (150–450) and 300 units (188–450), respectively.
Muscle | Participants injected n (%) | Dose (units) median (IQR) |
---|---|---|
Flexor digitorum superficialis | ||
Initial | 90 (54.9) (n = 164) | 100 (50 to 100) |
3 months | 50 (70.4) (n = 71) | 100 (100 to 100) |
6 months | 46 (71.9) (n = 64) | 100 (100 to 100) |
9 months | 39 (72.2) (n = 54) | 100 (100 to 100) |
Flexor digitorum profundus | ||
Initial | 63 (38.4) (n = 164) | 100 (50 to 100) |
3 months | 37 (52.1) (n = 71) | 100 (100 to 100) |
6 months | 39 (60.9) (n = 64) | 100 (100 to 120) |
9 months | 35 (64.8) (n = 54) | 100 (100 to 100) |
Flexor pollicis longus | ||
Initial | 6 (3.7) (n = 164) | 100 (72.5 to 112.5) |
3 months | 5 (7.0) (n = 71) | 80 (50 to 100) |
6 months | 7 (10.9) (n = 64) | 50 (50 to 100) |
9 months | 7 (13.0) (n = 54) | 50 (50 to 80) |
Forearm flexors | ||
Initial | 17 (10.4) (n = 164) | 200 (200 to 300) |
3 months | 7 (9.9) (n = 71) | 300 (100 to 300) |
6 months | 4 (6.3) (n = 64) | 200 (100 to 300) |
9 months | 0 (0.0) (n = 54) | 0 (0 to 0) |
Flexor carpis ulnaris | ||
Initial | 57 (34.8) (n = 164) | 100 (50 to 100) |
3 months | 29 (40.8) (n = 71) | 100 (100 to 100) |
6 months | 31 (48.4) (n = 64) | 100 (100 to 100) |
9 months | 29 (53.7) (n = 54) | 100 (100 to 100) |
Flexor carpis radialis | ||
Initial | 10 (6.1) (n = 164) | 50 (28.8 to 100) |
3 months | 3 (4.2) (n = 71) | 100 (100 to 100) |
6 months | 1 (1.6) (n = 64) | 100 (100 to 100) |
9 months | 1 (1.9) (n = 54) | 100 (100 to 100) |
Biceps brachii | ||
Initial | 125 (76.2) (n = 164) | 100 (50 to 100) |
3 months | 55 (77.5) (n = 71) | 100 (100 to 100) |
6 months | 47 (73.4) (n = 64) | 100 (100 to 150) |
9 months | 41 (75.9) (n = 54) | 100 (100 to 175) |
Brachioradialis | ||
Initial | 25 (15.2) (n = 164) | 100 (50 to 100) |
3 months | 13 (18.3) (n = 71) | 100 (100 to 100) |
6 months | 8 (12.5) (n = 64) | 100 (100 to 100) |
9 months | 5 (9.3) (n = 54) | 100 (100 to 150) |
Pronator teres | ||
Initial | 2 (1.2) (n = 164) | 100 (100 to 100) |
3 months | 0 (0.0) (n = 71) | 0 (0 to 0) |
6 months | 0 (0.0) (n = 64) | 0 (0 to 0) |
9 months | 0 (0.0) (n = 54) | 0 (0 to 0) |
Pectoralis major | ||
Initial | 9 (5.5) (n = 164) | 100 (50 to 100) |
3 months | 5 (7.0) (n = 71) | 100 (50 to 100) |
6 months | 1 (1.6) (n = 64) | 200 (200 to 200) |
9 months | 2 (3.7) (n = 54) | 150 (100 to 200) |
Limb area | Participants injected n (%) | Dose (units) median (IQR) |
Hand only (FDS and/or FDP and/or FPL) | ||
---|---|---|
Initial | 19 (11.6) (n = 164) | 100 (50 to 300) |
3 months | 9 (12.7) (n = 71) | 150 (100 to 250) |
6 months | 8 (12.5) (n = 64) | 100 (100 to 188) |
9 months | 5 (9.3) (n = 54) | 200 (100 to 200) |
Wrist only (FCU and/or FCR) | ||
Initial | 3 (1.8) (n = 164) | 50 (50 to 125)a |
3 months | 1 (1.4) (n = 71) | 10 (100 to 100) |
6 months | 3 (4.7) (n = 64) | 100 (100 to 100) |
9 months | 2 (3.7) (n = 54) | 100 (100 to 100) |
Elbow only (Biceps and/or Brachioradialis) | ||
Initial | 37 (22.6) (n = 164) | 100 (50 to 100) |
3 months | 10 (14.1) (n = 71) | 100 (50 to 163) |
6 months | 6 (9.4) (n = 64) | 100 (88 to 113) |
9 months | 6 (11.1) (n = 54) | 100 (88 to 225) |
Shoulder only (Pectoralis major) | ||
Initial | 2 (1.2) (n = 164) | 75 (50 to 100) |
3 months | 0 (0.0) (n = 71) | – |
6 months | 0 (0.0) (n = 64) | – |
9 months | 1 (1.9) (n = 54) | 100 (100 to 100) |
Hand and wrist | ||
Initial | 13 (7.9) (n = 164) | 300 (175 to 300) |
3 months | 6 (8.5) (n = 71) | 300 (275 to 325) |
6 months | 6 (9.4) (n = 64) | 300 (300 to 338) |
9 months | 5 (9.3) (n = 54) | 400 (300 to 425) |
Hand and elbow | ||
Initial | 45 (27.4) (n = 164) | 300 (150 to 400) |
3 months | 21 (29.6) (n = 71) | 300 (300 to 500) |
6 months | 19 (29.7) (n = 64) | 400 (200 to 500) |
9 months | 13 (24.1) (n = 54) | 300 (250 to 425) |
Wrist and elbow | ||
Initial; | 7 (4.3) (n = 164) | 100 (100 to 300) |
3 month | 2 (2.8) (n = 71) | 150 (100 to 200) |
6 month | 3 (4.7) (n = 64) | 150 (125 to 175)a |
9 month | 5 (9.3) (n = 54) | 150 (125 to 200) |
Hand and wrist and elbow | ||
Initial | 29 (17.7) (n = 164) | 400 (250 to 400) |
3 months | 17 (23.9) (n = 71) | 400 (400 to 500) |
6 months | 18 (28.1) (n = 64) | 450 (400 to 613) |
9 months | 16 (29.6) (n = 54) | 450 (400 to 575) |
Hand and wrist and elbow and shoulder | ||
Initial | 4 (2.4) (n = 164) | 500 (363 to 600) |
3 months | 2 (2.8) (n = 71) | 700 (600 to 800) |
6 months | 0 (0.0) (n = 64) | – |
9 months | 0 (0.0) (n = 54) | – |
Other | ||
Initial | 5 (3.0) (n = 164) | 200 (150 to 300) |
3 months | 3 (4.2) (n = 71) | 150 (150 to 250)a |
6 months | 1 (1.6) (n = 64) | 700 (700 to 700) |
9 months | 1 (1.9) (n = 54) | 850 (850 to 850) |
The trial protocol allowed for participants in the control group to be considered for treatment with botulinum toxin injections if they had an ‘unacceptable degree of symptomatic spasticity’. In addition, it was possible for trial participants to be referred to routine spasticity/botulinum toxin services by local health-care providers outside the study.
In the initial 3-month study period botulinum toxin was received by 4/163 (2.5%) control group participants. Only one participant in the control group received treatment before the study 1-month outcome assessment. Eight (7.8%) control group participants commenced botulinum toxin treatment after 3 months. Figure 5 shows the use of botulinum toxin in both intervention and control groups during the study period. All participants treated with botulinum toxin in the control group remained in the control group for the purposes of analyses.
At the end of the study period (3 or 12 months) participants in both groups whom research therapists felt would benefit from botulinum toxin treatment were referred to local spasticity services. Following the last outcome assessment 87/170 (51.2%) in the intervention group and 67/163 (41.1%) in the control group were referred to a spasticity service for botulinum toxin.
Upper limb therapy
Participants in both groups received upper limb therapy immediately following study entry for 1 hour twice per week for 4 weeks. Participants were reassessed for further therapy at 3, 6 and 9 months. The therapy programme for the study consisted of two menus. Menu 1 was designed for participants with no active upper limb function at trial entry (ARAT 0–3) and menu 2 was designed for participants with some retained function (ARAT 4–56). At the end of the study participants were clinically reviewed and those requiring further therapy were referred to local services.
In the control group, 159/163 (97.5%) participants received initial study therapy and in the intervention group this was 168/170 (98.8%). The six participants who did not receive therapy withdrew from the study before treatment could commence. Table 19 shows the therapy menu used and number of sessions delivered in each group. Therapy data were missing for one participant in the control group, although the study therapist confirmed that he was treated. Participants in both groups received a median of eight treatment sessions in the 4 weeks postrandomisation [110 (70%) participants in the control group and 129 (77%) participants in the intervention group received the full eight treatment sessions following randomisation]. There were no statistically significant differences between the groups for the therapy menu used or number of therapy sessions received.
Control (n = 158) | Intervention (n = 168) | p-value | |
---|---|---|---|
Therapy programme: n (%) | |||
Menu 1 (ARAT 0–3) | 91 (57.6) | 97 (57.7) | 1.000 |
Menu 2 (ARAT 4–56) | 67 (42.4) | 71 (42.3) | |
Number of sessions (maximum 8): median (IQR) | 8 (7 to 8) | 8 (8 to 8) | 0.210 |
One hundred and five participants randomised to the intervention group and 103 randomised to the control group entered the study for a 12-month follow-up and were therefore eligible for further therapy if necessary following reassessment at 3, 6 and 9 months. The number of participants completing clinical review at each time point is shown in Table 20.
Control | Intervention | |
---|---|---|
Completing clinical review: n (%) | ||
3 months | 92 (89.3) | 101 (96.1) |
6 months | 91 (88.3) | 102 (97.1) |
9 months | 90 (87.4) | 99 (94.3) |
The design of the therapy programme was such that once it had been taught it could be practised without the need for face-to-face contact with a therapist. Therefore, following review, the majority of participants were encouraged to continue with the programme previously demonstrated. Unfortunately, data regarding the amount and content of therapy received at 3, 6 and 9 months was of poor quality and was not suitable for analysis.
Trial safety evaluation
For clinical trials using investigational medicinal products, trial safety is evaluated by examining the occurrence of all adverse events as defined by the Medicines for Human Use (Clinical Trials) Regulations. 100 All serious adverse events were assessed for causality and expectedness, and were immediately reported to the trial sponsor. Systems were in place for expedited reporting of suspected unexpected serious adverse reactions to the appropriate bodies. Annual safety reports were submitted to the Medicines and Healthcare Products Regulatory Agency and the Multicentre Research Ethics Committee as required.
Safety data were analysed according to treatment received. Serious adverse events occurred in 34/156 (21.7%) participants who had not received botulinum toxin type A during the study and in 36/176 (20.5%) participants who had received botulinum toxin type A treatment. However, for two participants who received botulinum toxin type A during the study, the serious adverse events occurred before receipt of injections so they were included in the ‘no botulinum toxin’ group for the purposes of the analyses.
Fifty serious adverse events were reported from the ‘no botulinum toxin’ group and 52 from the ‘received botulinum toxin’ group. These events were summarised and categorised blinded to treatment received (Table 21). The most commonly reported serious adverse events were neurological events, including further strokes and seizures. Several musculoskeletal and respiratory events were also reported from both groups. There were no statistically significant differences between the groups for any serious adverse event type.
Serious adverse event | No botulinum toxin (n = 158) | Received botulinum toxin (n = 174) | Botulinum toxin relative risk (95% CI) | ||
---|---|---|---|---|---|
No of participants (%) | Total number of events | No of participants (%) | Total number of events | ||
Cardiac | 4 (2.5) | 4 | 3 (1.7) | 3 | 0.7 (0.2 to 3.0) |
Arrhythmia | 3 | 1 | |||
Cardiac failure | 0 | 1 | |||
Chest pain unknown cause | 0 | 1 | |||
Myocardial infarction | 1 | 0 | |||
Endocrine | 0 (0.0) | 0 | 1 (0.6) | 2 | |
Unstable diabetes mellitus | 0 | 2 | |||
Gastrointestinal | 2 (1.3) | 2 | 2 (1.1) | 2 | 0.9 (0.1 to 6.4) |
Clostridium difficile diarrhoea | 0 | 1 | |||
Gastrointestinal bleed | 1 | 1 | |||
Irritable bowel syndrome | 1 | 0 | |||
Musculoskeletal | 5 (3.2) | 5 | 10 (5.7) | 11 | 1.8 (0.6 to 5.2) |
Fracture following fall | 3 | 7 | |||
Soft tissue injury following fall | 0 | 2 | |||
Fracture following road traffic accident | 1 | 0 | |||
Chronic osteomyelitis | 1 | 0 | |||
Decreased mobility ? cause | 0 | 1 | |||
Elective musculoskeletal surgery | 0 | 1 | |||
Neurological | 19 (12.0) | 22 | 10 (5.7) | 11 | 0.5 (0.2 to 1.0) |
Seizure | 11 | 6 | |||
New stroke | 7 | 5 | |||
Transient ischaemic attack | 1 | 0 | |||
Elective neurosurgery | 1 | 0 | |||
Headache | 1 | 0 | |||
Possible tardive dyskinesia | 0 | 1 | |||
Psychiatric | 0 (0.0) | 0 | 1 (0.6) | 1 | |
Self-harm | 0 | 1 | |||
Respiratory | 6 (3.8) | 8 | 9 (5.2) | 11 | 1.4 (0.5 to 3.7) |
Chest infection | 8 | 8 | |||
Elective respiratory surgery | 0 | 2 | |||
Non-infective exacerbation of chronic obstructive airway disease | 0 | 1 | |||
Urinary tract | 4 (2.5) | 4 | 7 (4.0) | 7 | 1.6 (0.5 to 5.3) |
Urinary tract infection (UTI) | 2 | 3 | |||
UTI and renal failure | 0 | 1 | |||
Urinary retention/catheter issue | 2 | 3 | |||
Miscellaneous | 5 (3.2) | 5 | 4 (2.3) | 4 | 0.7 (0.2 to 2.7) |
Burn | 1 | 0 | |||
Cellulitis | 1 | 1 | |||
Deep vein thrombosis | 1 | 0 | |||
Death, cause unknown | 1 | 0 | |||
Dysphagia unknown cause | 0 | 1 | |||
Elective sinus surgery | 0 | 1 | |||
Metastatic cancer | 1 | 0 | |||
Vasovagal event | 0 | 1 |
All serious adverse events were assessed for their relationship to the study drug at the time of reporting. Only one event was reported as potentially related to botulinum toxin type A. This was an event recorded as dysphagia of unclear cause. As this is a known adverse reaction to botulinum toxin type A it was reported as a suspected serious adverse reaction. However, when further information became available about this event, it was assessed as unrelated to the study drug. No suspected unexpected serious adverse reactions were reported
Adverse events occurred in 70/156 (44.9%) participants who had not received botulinum toxin type A during the study and 90/176 (51.1%) participants who had received botulinum toxin type A treatment. However, for the participants who received botulinum toxin type A during the study, six had events that occurred before receipt of toxin and three had events that occurred both before and after botulinum toxin injection(s). For the purposes of the analyses those that had events before they received botulinum toxin type A were included in the ‘no botulinum’ group. Participants that had events both before and after botulinum toxin type A were included in the ‘received botulinum toxin’ group, which was a pragmatic decision as analyses were performed in exclusive categories.
One hundred and fourteen adverse events were reported from the ‘no botulinum toxin’ group and 147 from the ‘received botulinum toxin’ group. Adverse event descriptions were summarised and categorised blinded to treatment received (Table 22). It was not intended to calculate relative risk for any of the individual adverse events (only the categories), but on examining the data it appeared that there were several more ‘chest infections’ and ‘general malaise/flu-like/cold symptoms’ in the ‘received botulinum toxin’ group. A relative risk estimate was therefore calculated for these. For chest infection, the botulinum toxin relative risk was 1.4 (95% CI 0.5 to 3.5) and for general malaise/flu-like/cold it was 7.6 (95% CI 1.8 to 32.3). General malaise/flu-like/cold symptoms are recognised side effects of botulinum toxin type A.
Adverse event | No botulinum toxin (n = 162) | Received botulinum toxin (n = 170) | Botulinum toxin relative risk (95% CI) | ||
---|---|---|---|---|---|
No of participants (%) | Total number of events | No of participants (%) | Total number of events | ||
Cardiac | 8 (4.9) | 9 | 8 (4.7) | 9 | 1.0 (0.4 to 2.5) |
Ear | 1 (0.6) | 1 | 2 (1.2) | 2 | 1.9 (0.2 to 20.8) |
Eye | 2 (1.2) | 2 | 3 (1.8) | 3 | 1.4 (0.2 to 8.5) |
Endocrine | 1 (0.6) | 1 | 1 (0.6) | 1 | 1.0 (0.1 to 15.1) |
Gastrointestinal | 4 (2.5) | 4 | 10 (5.9) | 12 | 2.4 (0.8 to 7.5) |
Haematological | 1 (0.6) | 2 | 3 (1.8) | 3 | 2.9 (0.3 to 27.0) |
Musculoskeletal | 8 (4.9) | 10 | 13 (7.6) | 16 | 1.5 (0.7 to 3.6) |
Neurological | 10 (6.2) | 10 | 6 (3.5) | 6 | 0.6 (0.2 to 1.5) |
Psychiatric | 2 (1.2) | 3 | 5 (2.9) | 4 | 2.4 (0.5 to 12.0) |
Respiratory | 8 (4.9) | 9 | 12 (7.1) | 13 | 1.4 (0.6 to 3.4) |
Skin | 11 (6.8) | 12 | 10 (5.9) | 11 | 0.9 (0.4 to 2.0) |
Urinary tract | 8 (4.9) | 9 | 9 (5.3) | 9 | 1.1 (0.4 to 2.7) |
Miscellaneous | 32 (19.8) | 42 | 43 (25.3) | 58 | 1.3 (0.9 to 1.9) |
As with serious adverse events, all adverse events were assessed for their relationship to botulinum toxin type A at the time of reporting. In addition, adverse events were also assessed for their relationship to study upper limb therapy. Twenty-eight events were reported as possibly or probably related to botulinum toxin type A and 16 events were reported as possibly or probably related to study upper limb therapy (Table 23). All other events were assessed as not related to study botulinum toxin or study therapy.
Event | Potentially related to botulinum toxin type A | Potentially related to upper limb therapy |
---|---|---|
Gastrointestinal | 3 | 0 |
Diarrhoea | 2 | – |
Abdominal pain unknown cause | 1 | – |
Musculoskeletal | 1 | 3 |
Muscle sprain | 1 | 1 |
Frozen shoulder | 0 | 1 |
Cramp | 0 | 1 |
Neurological | 2 | 2 |
Headache | 2 | 0 |
Paraesthesia | 0 | 2 |
Respiratory | 2 | 0 |
Chest infection | 1 | – |
Shortness of breath/cough | 1 | – |
Skin | 2 | 0 |
Rash | 2 | – |
Urinary tract | 1 | 0 |
Urinary frequency/incontinence | 1 | – |
Miscellaneous | 17 | 11 |
General malaise/flu like/cold | 14 | 2 |
Fall +/– minor injury | 1 | 0 |
Dizziness | 1 | 0 |
Perspiration | 1 | 0 |
Upper limb pain | 0 | 7 |
Pain – other | 0 | 1 |
Minor injury | 0 | 1 |
Secondary analysis of primary outcome
A logistic regression model was used to adjust the primary outcome (predefined treatment success on the ARAT at 1 month) for randomisation strata factors (Table 24). Adjustment for research centre and baseline upper limb function (ARAT 0–3, 4–28, 29–56) had very little impact on the magnitude of the estimated effect of botulinum toxin; the relative odds of a ‘treatment success’ changed from 1.39 to 1.41.
Effects included in logistic regression modela | Estimated effect of botulinum toxinb | ||
---|---|---|---|
Fixed | Random | ORc | 95% Cl |
None | None | 1.39 | 0.82 to 2.36 |
None | Centre | 1.39 | 0.82 to 2.36 |
Initial ARAT group | Centre | 1.41 | 0.82 to 2.42 |
Preplanned subgroup analyses
The effect of time since stroke and severity of initial upper limb function upon the primary outcome and statistically significant secondary outcomes was addressed.
Time since stroke
Response to treatment was compared for participants who joined the study within 1 year following stroke (n = 181) and those who were recruited after 1 year (n = 151).
Fitting a logistic regression model, participants recruited within 1 year of stroke were more likely to experience ‘treatment success’ than participants recruited more than 1 year after stroke, but the difference was not statistically significant (odds ratio 1.6; 95% CI 0.92 to 2.79; p = 0.09). Fitting an interaction between randomised treatment and time since stroke did not improve the fit of the model (p = 0.69).
For secondary outcomes (Table 25), although there were some significant differences between intervention and control groups within the subgroups, there were no significant differences between the subgroups for any outcome.
Control | Intervention | Difference | Difference between subgroups | |
---|---|---|---|---|
Modified Ashworth Score at elbow: mean change 1 month (95% CI) | ||||
≤ 1 year | – 0.1 (– 0.3 to 0.2) | – 0.6 (– 0.8 to – 0.4) | – 0.5 (– 0.9 to – 0.2) | 0.0 (– 0.5 to 0.5) |
> 1 year | 0.0 (– 0.3 to 0.2) | – 0.6 (– 0.8 to – 0.3) | – 0.5 (– 0.9 to – 0.2) | |
Motricity Index: mean change 3 months (95% CI) | ||||
Arm | ||||
≤ 1 year | – 0.6 (– 3.5 to 2.3) | 5.4 (1.9 to 8.7) | 6.0 (1.5 to 10.5) | – 5.7 (– 12.3 to 1.0) |
> 1 year | 4.8 (1.2 to 8.5) | 5.1 (1.9 to 8.6) | 0.3 (– 4.6 to 5.3) | |
Total | ||||
≤ 1 year | – 0.3 (– 2.6 to 2.0) | 4.8 (2.1 to 7.6) | 5.1 (1.6 to 8.7) | – 4.4 (– 9.7 to 0.8) |
> 1 year | 3.1 (0.0 to 6.2) | 3.8 (1.3 to 6.4) | 0.7 (– 3.3 to 4.7) | |
ARAT: mean change 3 months (95% CI) | ||||
≤ 1 year | 1.8 (0.7 to 3.1) | 3.5 (1.8 to 5.3) | 1.6 (– 0.4 to 3.7) | 0.5 (– 2.3 to 3.4) |
> 1 year | 0.5 (– 1.0 to 1.8) | 2.6 (1.4 to 3.9) | 2.1 (0.3 to 4.1) | |
Upper limb functional activities: mean change (95% CI) | ||||
Put arm through sleeve | ||||
1 month | ||||
≤ 1 year | – 0.1 (– 0.4 to 0.2) | 0.4 (0.2 to 0.7) | 0.5 (0.2 to 0.9) | – 0.4 (– 0.9 to 0.1) |
> 1 year | 0.2 (0.0 to 0.4) | 0.3 (0.0 to 0.6) | 0.1 (– 0.2 to 0.5) | |
Open the hand for cleaning your palm | ||||
1 month | ||||
≤ 1 year | 0.1 (– 0.2 to 0.4) | 0.3 (0.0 to 0.6) | 0.2 (– 0.2 to 0.6) | 0.2 (– 0.4 to 0.7) |
> 1 year | 0.2 (– 0.2 to 0.5) | 0.5 (0.2 to 0.8) | 0.4 (– 0.1 to 0.8) | |
3 months | ||||
≤ 1 year | 0.1 (– 0.3 to 0.5) | 0.2 (– 0.2 to 0.6) | 0.1 (– 0.4 to 0.6) | 0.4 (– 0.3 to 1.2) |
> 1 year | – 0.2 (– 0.5 to 0.2) | 0.4 (0.1 to 0.7) | 0.5 (0.1 to 1.0) | |
12 months | ||||
≤ 1 year | 0.0 (– 0.5 to 0.5) | 0.2 (– 0.3 to 0.8) | 0.3 (– 0.5 to 1.0) | 0.5 (– 0.5 to 1.4) |
> 1 year | – 0.2 (– 0.6 to 0.3) | 0.6 (0.1 to 1.0) | 0.7 (0.1 to 1.4) | |
Open the hand for cutting fingernails | ||||
1 month | ||||
≤ 1 year | 0.1 (0.0 to 0.2) | 0.2 (0.0 to 0.4) | 0.1 (– 0.1 to 0.3) | – 0.1 (– 0.4 to 0.3) |
> 1 year | 0.1 (– 0.1 to 0.3) | 0.2 (0.0 to 0.4) | 0.1 (– 0.2 to 0.3) | |
3 months | ||||
≤ 1 year | 0.2 (0.0 to 0.4) | 0.3 (0.2 to 0.5) | 0.1 (– 0.2 to 0.4) | – 0.1 (– 0.5 to 0.3) |
> 1 year | 0.1 (– 0.2 to 0.3) | 0.1 (– 0.1 to 0.3) | 0.1 (– 0.3 to 0.4) | |
12 months | ||||
≤ 1 year | – 0.1 (– 0.4 to 0.3) | 0.3 (– 0.1 to 0.6) | 0.3 (– 0.2 to 0.8) | – 0.1 (– 0.7 to 0.5) |
> 1 year | – 0.1 (– 0.3 to 0.2) | 0.2 (0.0 to 0.4) | 0.2 (– 0.1 to 0.6) | |
Pain description: mean change 12 months (95% CI) | ||||
≤ 1 year | 0.4 (– 0.1 to 0.8) | 0.9 (0.5 to 1.3) | 0.5 (– 0.1 to 1.2) | – 0.2 (– 1.0 to 0.6) |
> 1 year | 0.4 (0.0 to 0.7) | 0.7 (0.4 to 1.1) | 0.4 (– 0.1 to 0.9) | |
Pain score (0– 10): mean change 12 months (95% CI) | ||||
≤ 1 year | – 0.8 (– 2.0 to 0.4) | – 2.4 (– 3.4 to – 1.3) | – 1.5 (– 3.2 to 0.1) | 0.4 (– 1.7 to 2.4) |
> 1 year | – 0.8 (– 1.6 to 0.0) | – 2.0 (– 3.0 to – 1.0) | – 1.2 (– 2.5 to 0.1) |
Severity of initial upper limb function
Data were analysed according to baseline ARAT score: no active upper limb function ARAT 0–3 (n = 184) and some retained active upper limb function ARAT 4–56 (n = 145).
Fitting a logistic regression model, participants with some retained active upper limb function (ARAT 4–56) were more likely to experience ‘treatment success’ than participants with no retained upper limb function (ARAT 0–3) (odds ratio 2.41; 95% CI 1.40 to 4.14) but on fitting an interaction between randomised treatment and baseline ARAT score, the model was not significant (p = 0.81).
For secondary outcomes (Table 26), there were no significant differences between the subgroups for the estimated effect of the intervention on changes in muscle tone (1 month), arm strength (3 months), performance of basic upper limb functional activities (1, 3 and 12 months) or pain rating (12 months). However, for the change in ARAT score from baseline to 3 months the estimated effect of the intervention was 0.3 (95% CI – 0.5 to 1.1) and 3.6 (95% CI 0.7 to 6.5) in the ARAT 0–3 and ARAT 4–56 subgroups, respectively. The difference in effect between the subgroups was 3.3 (95% CI 0.3 to 6.3).
Control | Intervention | Difference | Difference between subgroups | |
---|---|---|---|---|
Modified Ashworth Score at elbow: mean change 1 month (95% CI) | ||||
0–3 | 0.2 (– 0.1 to 0.4) | – 0.5 (– 0.8 to – 0.3) | – 0.7 (– 1.0 to – 0.3) | 0.3 (– 0.2 to 0.8) |
4–56 | – 0.3 (– 0.6 to – 0.1) | – 0.7 (– 0.9 to – 0.4) | – 0.4 (– 0.7 to 0.0) | |
Motricity Index: mean change 3 months (95% CI) | ||||
Arm | ||||
0–3 | – 0.2 (– 3.4 to 3.0) | 5.5 (2.2 to 9.0) | 5.7 (1.1 to 10.4) | – 5.1 (– 11.7 to 1.5) |
4–56 | 4.3 (1.1 to 7.6) | 4.9 (1.6 to 8.2) | 0.6 (– 4.1 to 5.3) | |
Total | ||||
0–3 | – 0.2 (– 2.5 to 2.1) | 4.8 (2.2 to 7.4) | 5.0 (1.5 to 8.5) | – 4.2 (– 9.6 to 1.3) |
4–56 | 3.0 (– 0.1 to 6.1) | 3.8 (1.2 to 6.4) | 0.8 (– 3.2 to 4.9) | |
ARAT: mean change 3 months (95% CI) | ||||
0–3 | 0.8 (0.4 to 1.3) | 1.1 (0.6 to 1.8) | 0.3 (– 0.5 to 1.1) | 3.3 (0.3 to 6.3) |
4–56 | 1.8 (– 0.1 to 3.7) | 5.4 (3.3 to 7.6) | 3.6 (0.7 to 6.5) | |
Upper limb functional activities mean change (95% CI) | ||||
Put arm through sleeve 1 month | ||||
1 month | ||||
0–3 | – 0.1 (– 0.4 to 0.1) | 0.3 (0.0 to 0.5) | 0.4 (0.0 to 0.8) | – 0.1 (– 0.6 to 0.4) |
4–56 | 0.2 (0.0 to 0.4) | 0.5 (0.2 to 0.7) | 0.3 (0.0 to 0.6) | |
Open the hand for cleaning your palm | ||||
1 month | ||||
0–3 | 0.1 (– 0.3 to 0.4) | 0.3 (0.0 to 0.6) | 0.3 (– 0.2 to 0.7) | 0.0 (– 0.5 to 0.6) |
4–56 | 0.2 (– 0.1 to 0.5) | 0.5 (0.2 to 0.8) | 0.3 (– 0.1 to 0.7) | |
3 months | ||||
0–3 | – 0.1 (– 0.5 to 0.3) | 0.2 (– 0.2 to 0.5) | 0.2 (– 0.3 to 0.8) | 0.2 (– 0.6 to 0.9) |
4–56 | 0.0 (– 0.3 to 0.4) | 0.4 (0.1 to 0.8) | 0.4 (– 0.1 to 0.9) | |
12 months | ||||
0–3 | – 0.2 (– 0.7 to 0.3) | 0.4 (– 0.2 to 1.1) | 0.7 (– 0.1 to 1.5) | – 0.3 (– 1.3 to 0.6) |
4–56 | 0.1 (– 0.4 to 0.5) | 0.4 (0.1 to 0.8) | 0.3 (– 0.2 to 0.9) | |
Open the hand for cutting fingernails | ||||
1 month | ||||
0–3 | 0.1 (– 0.1 to 0.2) | 0.0 (– 0.2 to 0.2) | – 0.1 (– 0.3 to 0.2) | 0.3 (0.0 to 0.7) |
4–56 | 0.1 (0.0 to 0.3) | 0.4 (0.2 to 0.6) | 0.3 (0.0 to 0.5) | |
3 months | ||||
0–3 | 0.0 (– 0.2 to 0.3) | 0.1 (– 0.1 to 0.2) | 0.0 (– 0.2 to 0.3) | 0.0 (– 0.4 to 0.5) |
4–56 | 0.3 to 0.1 to 0.6) | 0.4 (0.2 to 0.6) | 0.1 (– 0.2 to 0.4) | |
12 months | ||||
0–3 | – 0.2 (– 0.5 to 0.0) | 0.2 (0.0 to 0.5) | 0.4 (0.1 to 0.8) | – 0.4 (– 1.0 to 0.1) |
4–56 | 0.2 (– 0.2 to 0.5) | 0.2 (– 0.1 to 0.4) | 0.0 (– 0.4 to 0.4) | |
Pain description: mean change 12 months (95% CI) | ||||
0–3 | 0.3 (– 0.1 to 0.7) | 0.8 (0.3 to 1.2) | 0.5 (– 0.1 to 1.1) | – 0.1 (– 0.9 to 0.7) |
4–56 | 0.5 (0.1 to 0.8) | 0.9 (0.5 to 1.2) | 0.4 (– 0.1 to 0.9) | |
Pain score (0– 10): mean change 12 months (95% CI) | ||||
0–3 | – 0.5 (– 1.5 to 0.5) | – 2.1 (– 3.2 to – 1.0) | – 1.6 (– 3.1 to – 0.2) | 0.6 (– 1.4 to 2.8) |
4–56 | – 1.3 (– 2.4 to – 0.2) | – 2.2 (– 3.2 to – 1.3) | – 1.0 (– 2.4 to 0.5) |
Summary
Table 27 summarises the main results of the RCT.
1 month | 3 months | 12 months | |
---|---|---|---|
Modified Ashworth Scale at elbow | ✓ | ✗ | ✗ |
Motricity Index | |||
Arm | ✗ | ✓ | ✗ |
Total | ✗ | ✓ | ✗ |
Grip strength (kg) | ✗ | ✗ | ✗ |
ARAT predefined success | ✗ | ✗ | ✗ |
ARAT change from baseline | ✗ | ? | ✗ |
Nine-Hole Peg Test (pegs placed in 50s) | ✗ | ✗ | ✗ |
Upper limb functional activities | |||
Put arm through sleeve | ? | ✗ | ✗ |
Open the hand for cleaning your palm | ✗ | ? | ? |
Open the hand for cutting fingernails | ✗ | ✗ | ✗ |
Use cutlery | ✗ | ✗ | ✗ |
Improvement on upper limb functional activities of ≥ 1 | |||
Put arm through sleeve | ✓ | ✗ | ✗ |
Open the hand for cleaning your palm | ✓ | ✓ | ✓ |
Open the hand for cutting fingernails | ✓ | ✓ | ✓ |
Use cutlery | ✗ | ✗ | ✗ |
Barthel ADL Index | ✗ | ✗ | ✗ |
Stroke Impact Scale domains | |||
Strength | ✗ | ✗ | ✗ |
Memory | ✗ | ✗ | ✗ |
Emotion | ✗ | ✗ | ✗ |
Communication | ✗ | ✗ | ? |
ADL | ✗ | ✗ | ✗ |
Mobility | ✗ | ✗ | ✗ |
Hand function | ✗ | ✗ | ✗ |
Participation / Handicap | ✗ | ✗ | ✗ |
Physical domain | ✗ | ✗ | ✗ |
Stroke recovery | ✗ | ✗ | ✗ |
EQ-5D | |||
Mobility | ✗ | ✗ | ✗ |
Self-care | ✗ | ✗ | ✗ |
Usual activities | ✗ | ✗ | ✗ |
Pain / discomfort | ✗ | ? | ✗ |
Anxiety / depression | ✗ | ✗ | ? |
Good/bad health scale | ✗ | ✗ | ✗ |
Oxford Handicap Scale | ✗ | ? | ? |
Pain description | ✗ | ✗ | ✓ |
Pain score (0–10) | ✗ | ✗ | ✓ |
Chapter 4 Economic evaluation
The aim of the economic evaluation was to assess the cost-effectiveness of botulinum toxin type A injection(s) plus upper limb therapy relative to upper limb therapy alone. The economic evaluation follows the technology appraisal guidelines used by the National Institute for Health and Clinical Excellence (NICE) and as such adopts the perspective of the UK National Health Service and Social Services. 108 The time horizon for the analysis was 3 months from randomisation, with all costs reported in 2007 prices.
Assessment of costs
Participants’ use of resources was categorised under four general headings: (1) upper limb therapy sessions with or without botulinum toxin type A; (2) other antispasticity medication; (3) management of adverse events attributable to botulinum toxin type A and/or upper limb therapy requiring a hospital contact; and (4) other health-care and social services resource use. A breakdown of the individual items of resource use under these headings and their unit costs are presented in Table 28.
Resource | Unit cost (2007 prices) | Source of unit cost data |
---|---|---|
Upper limb therapy and botulinum toxin | ||
Therapist | £40 per session | Curtis (2007)109 |
Botulinum toxin | £153.21 per 500-unit vial | BNF (2007)110 |
Other antispasticity medication | ||
Gabapentin | £96.73 per month | BNF (2007)110 |
Baclofen | £9.13 per month | BNF (2007)110 |
Tizanidine | £74.83 per month | BNF (2007)110 |
Dantrolene | £33.76 per month | BNF (2007)110 |
Methocarbamol | £7.60 per month | BNF (2007)110 |
Other health-care and social services | ||
Day hospital | £83 per place per day | Curtis (2007)109 |
Home care services | £19 per contacta | Curtis (2007)109 |
Private home help | £11.33 per contacta | Curtis (2007)109 |
Day centre | £147 per attendance | Curtis (2007)109 |
Meals on wheels | £3.63 per meal | Curtis (2007)109 |
Laundry service | £3 per wash | Assumptionb |
General practitioner | £36 per consultation | Curtis (2007)109 |
Practice nurse | £9 per consultation | Curtis (2007)109 |
District nurse | £24 per home visit | Curtis (2007)109 |
Health visitor | £36 per home visit | Curtis (2007)109 |
Physiotherapist | £40 per contacta | Curtis (2007)109 |
Occupational therapist | £40 per contacta | Curtis (2007)109 |
Speech and language therapist | £40 per contacta | Curtis (2007)109 |
Dietician | £32 per contacta | Curtis (2007)109 |
Chiropodist | £18 per contacta | Curtis (2007)109 |
Social worker | £34 per contacta | Curtis (2007)109 |
Clinical psychologist | £67 per contacta | Curtis (2007)109 |
Continence advisor | £24 per home visit | Curtis (2007)109 |
Bath attendant | £11.33 per contacta | Assumptionc |
Orthotist | £40 per contacta | Assumptiond |
With respect to therapy sessions and botulinum toxin type A, data were collected in the study case record forms on the number of therapy sessions each participant received and the number of 500-unit vials of botulinum toxin type A they used. Each therapy session was staffed by one therapist and lasted for 1 hour. Unit cost data were obtained from Curtis 2007109 and the British National Formulary (BNF). 110
Data on other antispasticity medication were collected in the study case record forms. The forms recorded which drugs the participants were on at baseline, 1 and 3 months. For most participants, the length of time they spent taking a particular drug was straightforward to calculate insofar as if no changes to medications were noted, they were assumed to be taking the drug until such time as a change was noted. Problems arose when, for example, a participant was not taking a drug at baseline, but was noted as taking a drug at a later follow-up time point. Since the precise time at which the participant began taking the drug was not recorded, it was assumed that such participants began taking the drug at the mid-point between baseline and the time at which it was first noted they were taking the drug, i.e. 2 weeks for the 1-month follow-up, and 2 months for the 3-month follow-up. In the few cases where follow-up data were missing, it was assumed that participants remained on the drugs they were taking at baseline.
Another problem regarding antispasticity medication was failure to record the drug dosage that participants were taking. To deal with this, it was assumed that participants were taking the standard dose stipulated in the BNF 2007. 110 If a standard dose was not stated, then it was assumed that participants were taking the maximum recommended dose.
Data on adverse events attributable to botulinum toxin type A and/or therapy which led to a hospital contact were obtained from the study adverse event monitoring forms and from participant responses to specific resource use questions included in the participant assessment questionnaires. A distinction was made between outpatient attendances and events requiring an inpatient stay. Although there were cases of participants encountering hospital services as a consequence of their initial stroke, clinical review showed that none of the hospital contacts were attributable to either therapy or botulinum toxin type A. As a result, hospital resource use due to adverse events did not form part of the cost-effectiveness analysis.
Data on other health-care and social services resource use were obtained from the participant assessment questionnaires, which were administered at baseline, 1 and 3 months. These included questions on participants’ use of health-care and social services, such as day hospitals and day centres, and health-care and social services professional contacts, such as general practitioners and social workers. Unit cost data were mainly obtained from Curtis 2007. 109 Where unit cost data were not available, assumptions were made (see notes accompanying Table 28).
The resource use questions asked participants to report resource use for the 1-month period prior to completion of the questionnaire. This means that participant-reported resource use data were comprehensive for months 1 and 3, but that extrapolations had to be made for month 2. The extrapolation method adopted in the base-case analysis was to assume resource use in month 2 was the same as that in month 3. The impact on the results of alternatively assuming that resource use in month 2 is the same as in month 1 is investigated in the sensitivity analysis.
Assessment of outcome
Participant health-related quality of life was assessed using the EQ-5D,111 which was included in the participant assessment questionnaires. Differences between the randomisation groups at follow-up with respect to EQ-5D scores were investigated using multiple regression analysis of covariance. The average follow-up score of the EQ-5D (the mean of the 1-, 3- and 12-month assessments) was estimated and included as the dependent variable in a linear regression model. Covariates in the model were the ‘baseline EQ-5D score’ and the ‘randomisation group’ (coded 0 for therapy alone and 1 for botulinum toxin type A plus therapy). The regression coefficient estimate for randomisation group represents the difference in mean EQ-5D follow-up scores between the therapy alone and the botulinum toxin plus therapy groups after adjustment for baseline EQ-5D values.
Participant responses to the EQ-5D questionnaire were converted to health-state utility values using the UK tariff values. 112 These values were then multiplied by duration in each health state to estimate quality-adjusted life-years (QALYs). QALYs were estimated using an area under the curve (AUC) approach. 113 To illustrate this approach, consider an individual whose baseline and 3-month quality of life weights are 0.6 and 0.8, respectively. When located on a two-dimensional plane where the y-axis corresponds to the quality of life weight and the x-axis corresponds to time in years, the AUC that joins these two points defines a trapezium, the area of which is equal to the number of QALYs enjoyed by the patient in the 3 months since randomisation. The area of a trapezium with a base width of 3 months (one-quarter of a year) and whose sides are defined as a and b is equal to ½ × ¼ (a + b). In this example, a = 0.6 and b = 0.8. Hence, the AUC corresponds to 0.175 QALYs.
To estimate QALYs, participant responses at baseline and 3 months were used to map out the AUC. An alternative approach would be to use the baseline, 1-month and 3-month points. Although this latter approach provides a more precise estimate of QALYs, the inclusion of the 1-month values increases missing values and consequently decreases the number of participants on which the estimation is based. Therefore, the first approach was used in the base-case analysis, with the possible impacts on the results of the second approach being investigated in the sensitivity analysis.
Assessment of cost-effectiveness
To assess the relative cost-effectiveness of botulinum toxin type A plus upper limb therapy relative to upper limb therapy alone, data on cost and outcome were brought together to estimate an incremental cost-effectiveness ratio (ICER). Specifically, the incremental cost per QALY gained of botulinum toxin type A plus therapy relative to therapy alone was estimated.
The ICER for botulinum toxin type A plus therapy can be located on the cost-effectiveness plane (Figure 6), which is a two-dimensional space in which the origin represents the comparator intervention – in this case therapy alone. The x-axis represents the average difference in effectiveness per participant between botulinum toxin type A plus therapy and therapy alone, while the y-axis represents the average difference in cost per participant between the interventions. The four quadrants are conventionally referred to as points on the compass, namely north-west (NW), north-east (NE), south-west (SW) and south-east (SE). The ICER can be plotted as a point on this plane, with the slope of the line from the origin to the ICER representing the value of the ICER. Treatments with ICERs located in the NW quadrant (more costly, less effective) are said to be dominated by the comparator treatment, whereas treatments with ICERs located in the SE quadrant (less costly, more effective) are said to dominate the comparator treatment. In practice, most new treatments locate in the NE quadrant where increased effectiveness is achieved at increased cost. In this instance the decision to adopt the new treatment will depend upon whether the ICER lies below the acceptable ceiling ratio of the decision-maker. If the decision-maker’s willingness to pay for a unit of effectiveness (λ) is greater than the ICER, then on efficiency grounds the treatment should be recommended for adoption.
The point estimate of the ICER is subject to uncertainty and it is therefore important that this uncertainty is taken into account. Because of the problems associated with estimating CIs for ratio statistics, the approach of non-parametric bootstrapping is adopted to represent the uncertainty surrounding the ICER estimate. 114 A cost-effectiveness acceptability curve (CEAC), which summarises the evidence in support of botulinum toxin type A plus therapy being cost-effective for a range of values of λ, is also presented. The probabilistic interpretation of this curve should be from a Bayesian perspective. In effect, the CEAC provides information to decision-makers on the level of uncertainty associated with a potential decision to recommend the use of a new or additional intervention. For example, a 0.82 probability of an intervention being cost-effective at a ceiling ratio of £20,000 per QALY implies an error probability (i.e. the probability of making a wrong decision) of 0.18 (1 – 0.82). In making a decision regarding the potential recommendation of a new intervention, the decision-maker must weigh up these probabilities against one another. Alternatively, instead of deciding whether or not to recommend the new intervention on the basis of the currently available evidence, the decision-maker may demand an expected value of perfect information analysis to compare the expected cost of the uncertainty with the value of conducting further research to reduce the uncertainty (see Claxton et al. 115 for more details on expected value of perfect information analysis).
In addition to addressing the uncertainty surrounding the point estimate of the ICER, sensitivity analysis was undertaken to investigate the impact on the results of making alternative assumptions and varying key parameters. As part of the sensitivity analysis, to take into account missing data, an additional set of analyses were carried out using the technique of multiple imputation in which missing data were imputed using the norm package116. Five data sets were imputed using age, sex, place of residence, Barthel ADL score and time between stroke and randomisation as explanatory variables. A point estimate of the ICER and the accompanying CEAC for botulinum toxin type A plus therapy were estimated.
Results of the outcome assessments
Table 29 shows the mean EQ-5D scores over time for the botulinum toxin type A plus therapy and therapy alone groups.
Time point | Mean (SD) EQ-5D scores and number of participants providing a response (n) | |||
---|---|---|---|---|
Therapy alone | Botulinum toxin type A plus therapy | |||
Mean (SD) | n | Mean (SD) | n | |
Baseline | 0.3322 (0.2962) | 162 | 0.3206 (0.2964) | 170 |
1 month | 0.3041 (0.2992) | 134 | 0.3245 (0.2956) | 144 |
3 months | 0.3206 (0.2963) | 133 | 0.3478 (0.2920) | 150 |
12 months | 0.2727 (0.3078) | 86 | 0.3195 (0.2942) | 88 |
Regression analysis of covariance indicated that there was no significant difference between the groups with respect to mean follow-up EQ-5D scores after adjusting for baseline values.
Results of the cost-effectiveness analysis
The base-case analysis was based on 283 participants who provided EQ-5D responses at baseline and 3 months, of whom 150 were in the intervention group and 133 were in the control group.
There was no significant difference in the mean number of upper limb therapy sessions received by participants in the botulinum toxin type A plus therapy and therapy alone groups (7.64 versus 7.56, respectively; p = 0.46). With respect to use of botulinum toxin type A, the average number of vials used by each patient in the intervention group was 1.01. The numbers of participants taking other antispasticity drugs were 38 in the botulinum toxin type A plus therapy group and 31 in the therapy alone group, with the difference between the groups not being significant (χ2 = 0.157; p = 0.692).
A breakdown of other health-care and social services resource use among participants is presented in Table 30.
Item of resource use | Mean (SD) number of contacts among patients reporting a contact (n) | Mean difference in number of contacts (95% CI of difference) | |||
---|---|---|---|---|---|
Therapy alone | Botulinum toxin type A plus therapy | ||||
Mean (SD) | n | Mean (SD) | n | ||
Day hospital | 9.4 (9.0) | 25 | 3.1 (2.7) | 21 | 6.3 (2.4 to 10.2) |
Home-care services | 87.5 (75.7) | 45 | 114.6 (76.5) | 56 | – 27.1 (– 57.3 to 3.2) |
Private home help | 32.7 (53.5) | 17 | 38.9 (60.1) | 16 | – 6.2 (– 46.6 to 34.1) |
Day centre | 16.3 (8.3) | 25 | 14.1 (10.3) | 24 | 2.2 (– 3.1 to 7.6) |
Meals on wheels | 6.0 | 1 | 56.0 (38.6) | 3 | – 50.0 (– 241.7 to 141.7) |
Laundry service | 14.7 (18.5) | 3 | 18.0 (11.1) | 3 | – 3.3 (– 38.0 to 31.3) |
General practitioner | 2.8 (1.4) | 60 | 2.8 (1.9) | 86 | 0.0 (– 0.6 to 0.6) |
Practice nurse | 2.6 (1.8) | 24 | 2.9 (2.9) | 44 | – 0.3 (– 1.6 to 1.0) |
District nurse | 8.3 (14.6) | 27 | 4.0 (5.0) | 30 | 4.3 (– 1.8 to 10.2) |
Health visitora | 1 | 1.0 | 1 | ||
Physiotherapist | 13.3 (12.7) | 74 | 12.0 (10.5) | 87 | 1.3 (– 2.46 to 4.8) |
Occupational therapist | 8.2 (8.8) | 26 | 8.5 (9.1) | 38 | – 0.3 (– 4.9 to 4.2) |
Speech and language therapist | 4.6 (5.3) | 28 | 8.9 (9.7) | 20 | – 4.3 (– 9.2 to 0.6) |
Dietician | 2.2 (1.2) | 6 | 13.0 (15.6) | 2 | – 10.8 (– 149.5 to 127.8) |
Chiropodist | 2.0 (1.2) | 24 | 2.3 (1.1) | 40 | – 0.3 (– 0.9 to 0.3) |
Social worker | 2.8 (1.7) | 17 | 2.5 (2.0) | 35 | 0.3 (– 0.8 to 1.5) |
Clinical psychologist | 3.1 (2.5) | 8 | 2.5 (1.3) | 4 | 0.6 (– 2.4 to 3.7) |
Continence advisor | 1.8 (1.0) | 4 | 1.0 | 1 | 0.8 (– 2.7 to 4.2) |
Bath attendant | 19.5 (18.9) | 13 | 18.2 (11.4) | 13 | 1.3 (– 11.3 to 14.0) |
Orthotist | 1.00 | 1 | 2.7 (1.2) | 3 | – 1.7 (– 7.4 to 4.1) |
Chi-squared tests of differences in the proportion of participants in each randomisation group reporting a contact reveal significantly more practice nurse and social worker contacts among participants in the botulinum toxin type A plus therapy group (χ2 = 5.115; p = 0.024 and χ2 = 4.586; p = 0.032, respectively) and significantly more continence advisor contacts among participants in the therapy alone group (χ2 = 4.319; p = 0.038).
The only significant difference in the mean number of contacts among participants reporting a contact is with respect to day hospital contacts, with participants in the therapy alone group having significantly more contacts on average (9.4 versus 3.1, respectively; 95% CI of the difference, 2.42 to 10.19).
Table 31 shows the contribution of botulinum toxin type A costs, upper limb therapy costs, other antispasticity medication costs, and other health-care and social services costs to the overall mean cost per participant.
Breakdown of overall mean cost per participant | Mean (SD) cost per participant (£) | Mean difference in costs (£) (95% CI of difference) | |
---|---|---|---|
Therapy alone | Botulinum toxin type A plus therapy | ||
Overall | 1796 (1944) | 2170 (2007) | – 374 (– 837 to 90) |
Botulinum toxin | 3a (23) | 154 (28) | – 151 (– 157 to – 145) |
Upper limb therapy | 300 (45) | 303 (41) | – 3 (– 13 to 7) |
Antispasticity medication | 37 (93) | 38 (90) | – 1 (– 22 to 21) |
Otherb | 1456 (1923) | 1675 (2001) | – 219 (– 679 to 242) |
The overall mean cost per participant was higher in the botulinum toxin type A plus therapy group, although the difference was not significant. There were also no significant differences between the groups with respect to upper limb therapy costs, antispasticity medication costs and other health-care and social services costs. The biggest contributor to total costs for both groups was the cost of other health-care and social services contacts, accounting for 81% in the therapy alone group and 77% in the botulinum toxin type A plus therapy group.
Table 32 shows the point estimate of the ICER of botulinum toxin type A plus therapy relative to therapy alone.
Intervention | Cost (£) | QALYs | Incremental cost (£) | Incremental QALYs | ICER (£/QALY) |
---|---|---|---|---|---|
Therapy alone | 1796 | 0.081 | – | – | – |
Botulinum toxin plus therapy | 2170 | 0.085 | 374 | 0.004 | 93,500 |
Botulinum toxin type A plus therapy was associated with an incremental cost of £374 and an incremental QALY gain of 0.004, compared with therapy alone. When combined, these data gave an ICER for botulinum toxin type A plus therapy of £93,500 per QALY gained.
Bootstrapping the point estimate of the ICER for botulinum toxin type A plus therapy resulted in 27% of the replications being located in the NE quadrant of the cost-effectiveness plane (more costly, more effective), 21% being located in the SE quadrant (less costly, more effective), and 7% being located in the SW quadrant (less costly, less effective). The largest proportion of the replications (45%) is located in the NW quadrant, where botulinum toxin type A plus upper limb therapy is more costly and less effective, and therefore dominated by, upper limb therapy alone.
Figure 7 shows the CEAC for botulinum toxin type A plus therapy relative to therapy alone. The probabilities that botulinum toxin type A plus therapy is cost-effective at ceiling ratios of £10,000, £20,000, £50,000 and £100,000 per QALY are 0.29, 0.36, 0.41 and 0.42, respectively.
Sensitivity analysis
The impact on the results of the following sensitivity analyses were explored:
-
making an alternative extrapolation assumption for participant reported resource use
-
rerunning the analysis using data from participants with complete EQ-5D data at baseline and at 1 and 3 months
-
allowing the cost of botulinum toxin type A to fall to zero
-
a best–worst QALY analysis in which the impact of alternative assumptions regarding the timing of health-state changes is explored
-
rerunning the analysis following multiple imputation of missing data.
The impact on the results of assuming that participant-reported resource use in month 2 is the same as that in month 1 was minimal. Under this assumption the incremental cost of botulinum toxin type A plus therapy increased by £14 to £388, which resulted in the point estimate of the ICER increasing from £93,500 per QALY gained to £97,000 per QALY gained.
The results of the remaining sensitivity analyses, along with those of the base-case analysis, are summarised in Table 33.
Cost-effectiveness analysis scenario | IC (£) | IQ (QALYs) | ICER (£) | Probability that botulinum toxin type A plus therapy is cost-effective at threshold ratio: | |||
---|---|---|---|---|---|---|---|
£10,000 | £20,000 | £50,000 | £100,000 | ||||
Base-case dataa | 374 | 0.004 | 93,500 | 0.29 | 0.36 | 0.41 | 0.42 |
Complete EQ-5D datab | 482 | 0.007 | 68,857 | 0.29 | 0.34 | 0.40 | 0.43 |
Base-case data and zero cost for botulinum toxin | 223 | 0.004 | 55,750 | 0.31 | 0.39 | 0.42 | 0.43 |
Base-case data and best–worst QALY assumptions | 374 | 0.006 | 62,333 | 0.29 | 0.35 | 0.43 | 0.45 |
Missing data imputed using multiple imputation | 430 | 0.005 | 86,000 | 0.34 | 0.39 | 0.42 | 0.46 |
Complete EQ-5D data
Rerunning the analysis to include only those participants for whom complete EQ-5D data were available (i.e. responses at baseline and at 1 and 3 months) had little impact on the results. The numbers of participants included were 248, of whom 116 were in the botulinum toxin type A plus therapy group and 132 were in the therapy alone group. The average cost per patient and QALYs enjoyed for therapy alone were £1773 and 0.079 QALYs, respectively. The corresponding figures for botulinum toxin type A plus therapy were £2255 and 0.086 QALYs, respectively. Hence, compared to the base-case analysis, botulinum toxin type A plus therapy was associated with a higher incremental cost (£482 versus £374) and a bigger incremental QALY gain (0.007 versus 0.004), compared with therapy alone. The resultant ICER from combining these data was lower than that in the base-case analysis (£68,857 versus £93,500 per QALY gained, respectively).
Bootstrapping the point estimate of the ICER for botulinum toxin type A plus therapy resulted in 27%, 20%, 7% and 46% of the replications being located in the NE, SE, SW and NW quadrants of the cost-effectiveness plane, respectively.
Figure 8 shows the CEAC for botulinum toxin type A plus therapy relative to therapy alone for participants with complete EQ-5D data at 3 months. The probabilities that botulinum toxin type A plus therapy is cost-effective at ceiling ratios of £10,000, £20,000, £50,000 and £100,000 per QALY are 0.29, 0.34, 0.40 and 0.43, respectively.
Base-case data and zero cost for botulinum toxin
The impact on the results of allowing the cost of botulinum toxin type A to fall was investigated by considering the extreme assumption that the cost of botulinum toxin type A was zero. Under this assumption the average cost per patient and QALYs enjoyed for therapy alone were £1793 and 0.081 QALYs, respectively. The corresponding figures for botulinum toxin type A plus therapy were £2016 and 0.085 QALYs, respectively. Hence, despite the extreme assumption of a zero cost of botulinum toxin type A, botulinum toxin type A plus therapy still had a positive ICER compared with therapy alone, with the ICER having fallen from a base-case value of £93,500 to £55,750.
Bootstrapping the new point estimate of the ICER for botulinum toxin type A plus therapy resulted in 26%, 22%, 8% and 44% of the replications being located in the NE, SE, SW and NW quadrants of the cost-effectiveness plane, respectively.
Figure 9 shows the CEAC for botulinum toxin type A plus therapy relative to therapy alone assuming a zero cost for botulinum toxin type A. The probabilities that botulinum toxin type A plus therapy is cost-effective at ceiling ratios of £10,000, £20,000, £50,000 and £100,000 per QALY are 0.31, 0.39, 0.42 and 0.43, respectively.
Base-case data and best–worst QALY assumptions
The AUC approach to estimating QALYs described above assumes that the rate of change in health status between any two points (in this case, EQ-5D tariff values) is linear. For example, if the baseline value is 0.5 and the 3 months value is 0.8, then it is assumed that after 1 month, the health-state value is 0.6, and after 2 months it is 0.7. This is the method most commonly employed in AUC analyses in the literature. 113 However, many other assumptions could be made, each of which may have an impact on the results. In light of the relatively high ICERs associated with botulinum toxin type A plus therapy, it was decided to investigate the impact on the results of the timing of the health-state changes favouring the use of botulinum toxin type A. Specifically, it was assumed in the botulinum toxin type A plus therapy group that participants moved into the health-state value reported at 3 months almost immediately (approximately 3 days or 0.1 of a month) after baseline. This can be regarded as a best-case outcome scenario for botulinum toxin type A. On the flip side, a worst-case outcome scenario for the therapy alone group was defined whereby it was assumed that participants in this group remained in their baseline reported health state for 2.9 months, at which point they moved into the health state value reported at 3 months. The results of these assumptions were that the average number of QALYs enjoyed by botulinum toxin type A plus therapy participants increased from 0.085 in the base case to 0.087, while the average number of QALYs enjoyed by participants in the therapy alone group remained at 0.081. Combining the incremental QALY gain of 0.006 with the incremental cost of £374 gives a best–worst QALY scenario ICER for botulinum toxin type A plus therapy of £62,333.
Bootstrapping the point estimate of the ICER for botulinum toxin type A plus therapy resulted in 30%, 19%, 9% and 42% of the replications being located in the NE, SE, SW and NW quadrants of the cost-effectiveness plane, respectively.
Figure 10 shows the CEAC for botulinum toxin type A plus therapy relative to therapy alone for the best–worst QALY scenario. The probabilities that botulinum toxin type A plus therapy is cost-effective at ceiling ratios of £10,000, £20,000, £50,000 and £100,000 per QALY are 0.29, 0.35, 0.43 and 0.45, respectively.
Missing data imputed using multiple imputation
Imputing missing data using multiple imputation resulted in estimates of average cost per patient and QALYs enjoyed for therapy alone of £1807 and 0.077 QALYs, respectively. The corresponding figures for botulinum toxin type A plus therapy were £2237 and 0.082 QALYs, respectively. Compared with the base-case analysis, therefore, botulinum toxin type A plus therapy was associated with a higher incremental cost (£430 versus £374) and a bigger incremental QALY gain (0.005 versus 0.004), compared with therapy alone. The resultant ICER from combining these data was lower than that in the base-case analysis (£86,000 versus £93,500 per QALY gained, respectively).
Bootstrapping the point estimate of the ICER for botulinum toxin type A plus therapy over the five imputed data sets resulted in 28%, 20%, 11% and 41% of the replications being located in the NE, SE, SW and NW quadrants of the cost-effectiveness plane, respectively.
Figure 11 shows the CEAC for botulinum toxin type A plus therapy relative to therapy alone when missing data have been imputed. The probabilities that botulinum toxin type A plus therapy is cost-effective at ceiling ratios of £10,000, £20,000, £50,000 and £100,000 per QALY are 0.34, 0.39, 0.42 and 0.46, respectively.
Chapter 5 Discussion
Key findings
Primary outcome measure
Patients with upper limb spasticity due to stroke who were treated with botulinum toxin type A plus an upper limb therapy programme achieved similar levels of improvement in upper limb function to those who were treated with the upper limb therapy programme alone at 1 month after study entry. Improved arm function (predefined success on the Action Research Arm Test) was achieved by 19.5% of participants in the control group and 25.1% in the intervention group (p = 0.232). The relative risk of having a ‘successful treatment’ in the intervention group compared with the control group was 1.3 (95% CI 0.9 to 2.0). There was no significant improvement in ‘treatment success’ with botulinum toxin when the analysis was adjusted for randomisation strata factors (research site and baseline upper limb function).
Secondary outcome measures
Impairment
Botulinum toxin type A plus the upper limb therapy programme reduced muscle tone at the elbow by one point on the Modified Ashworth Scale at 1 month, but no significant differences were seen between the randomisation groups at 3 or 12 months. The reduction in muscle tone is consistent with previous studies, which reported that botulinum toxin reduced muscle tone at the elbow by one point on the Modified Ashworth Scale and the treatment lasted for approximately 3–4 months. 36,39–41,44,45 A reduction of one point on this scale is generally accepted as a clinically important difference.
Participants who were treated with botulinum toxin showed improvement in upper limb strength (Motricity Index) at 3 months, but no differences between the randomisation groups were found at other time points. This is the first time that improvement in upper limb strength has been demonstrated in an RCT of treatment of upper limb spasticity due to stroke with botulinum toxin.
No differences were seen in grip strength between randomisation groups. Improved grip strength was reported in one study with a dose of 75 units of botulinum toxin (Botox). 36 Reduced grip strength was reported in one trial following treatment with 1000 units of botulinum toxin (Dysport), which was a much higher dose than used in our study. 40
Upper limb function and activity limitation
BoTULS included a number of measures of upper limb function and activity limitation at each assessment. Results suggest that treatment with botulinum toxin type A is unlikely to improve active arm function in the majority of patients after stroke, but may improve the ability to carry out specific basic upper limb functional activities.
No differences in predefined success on the ARAT were seen at 3 or 12 months. However, at 3 months, although the median change in ARAT from baseline was zero in both randomisation groups, this result was statistically significant. The mean difference was 1.8 (95% CI 0.4 to 3.3) in favour of those treated with botulinum toxin. Although this does demonstrate improved active arm function following treatment with botulinum toxin, the magnitude of change is small and unlikely to be clinically meaningful.
A small trial45 (n = 50) published after BoTULS commenced found no improvement in upper limb function measured by the ARAT when a dose of 350 units of botulinum toxin (Dysport) was used. A dose of 500 units was associated with an improvement of 9.27 points at 8 weeks (p = 0.024) and 10.0 points at 24 weeks (p = 0.019) compared with placebo. The study also found that a higher dose of botulinum toxin (1000 units) significantly reduced active upper limb function, presumably as a result of weakness.
The relative contributions of spasticity and motor weakness to reduced function are debated. 12,13 Although there are those who advocate spasticity as an important component of reduced upper limb function, others believe that the main problem is motor weakness. As we have not shown improvement in active function despite improvement in muscle tone at 1 month, and only a small change in active function at 3 months, this study supports the argument that spasticity is of less importance. However, we may have failed to detect important differences because the impact of treatment may relate to practising activities over a longer period. Lack of sustained improvement at 12 months may be because a significant proportion of participants did not receive repeated botulinum toxin type A or upper limb therapy, or lack of power for detecting change at this time point.
Improvements in some specific basic upper limb activities in favour of those who received botulinum toxin type A were seen at 1, 3 and 12 months. When change in score from baseline was analysed, improvements were seen in dressing the sleeve at 1 month and opening the hand to clean the palm at 3 and 12 months. These differences were small and of uncertain clinical significance. To further understand these data and to enable comparison with other studies, we compared the proportion of participants who improved by one or more points from baseline at 1, 3 and 12 months. This showed a clear benefit in favour of the intervention group for opening the hand to clean the palm and opening the hand to cut nails at all time points. Improvement in dressing the sleeve was only seen at 1 month and no differences were seen between randomisation groups in terms of using cutlery at any time point.
Six previous RCTs have assessed similar upper limb functional activities and four reported improvements in participants receiving botulinum toxin. 37,40,42,51 Two studies reported magnitude of change from baseline in activities which was similarly small, but concluded that these were clinically important findings. 40,42 Two studies reported the proportion of participants who had improved by one point or more and demonstrated improvement with botulinum toxin for these tasks. 42,51
As spasticity is believed to limit activities such as opening the hand and dressing a sleeve,10 it follows that decreasing spasticity with botulinum toxin should result in improvement. It may seem inconsistent that such activities showed improvement when arm function measured by the ARAT did not improve. However, the measurement tool used to assess the basic upper limb activities asked about ability to undertake tasks but did not distinguish whether they were performed by the affected arm, with assistance from the non-affected arm, or by a carer. This means that the questions may measure passive and/or active function, whereas the ARAT measures only active function. The improvement in the specific upper limb activities demonstrated in this trial is likely to reflect a combination of improvement in both passive and/or active function. The lack of improvement in the use of cutlery may be because this requires improvement in active function.
No differences were seen in dexterity (Nine-Hole Peg Test) or ADL (Barthel ADL Index) at any time point. Four previous studies have used the Barthel ADL Index to measure activity limitation. 39,41,45,49 One reported significant improvements at 8 and 24 weeks in patients who received 350 or 500 units of botulinum toxin (Dysport). 45 Compared with placebo, participants who received 350 units of botulinum toxin improved by 7.0 points at 8 weeks (p = 0.012) and 14.0 points at 24 weeks (p < 0.001). Those who received 500 units improved by 20.6 points and 28.3 points at 8 (p < 0.001) and 24 weeks (p < 0.001), respectively.
Stroke-related quality of life/participation restriction
There were statistically significant improvements in the Oxford Handicap Scale at 3 and 12 months in favour of those who had received botulinum toxin. However, the differences were small and of doubtful clinical significance. In addition, those who received botulinum toxin type A had improved scores on the anxiety/depression component of EQ-5D, but this change was also small and of doubtful clinical significance.
Four previous trials have assessed quality of life/participation restriction, but no improvements were demonstrated after treatment of upper limb spasticity with botulinum toxin. 36,44,49,51
Pain
Our results suggest that botulinum toxin type A can reduce pain in patients with upper limb spasticity due to stroke. Those who received botulinum toxin type A experienced less pain at 3 months, as measured by the EQ-5D, but this difference was small and of doubtful clinical significance. However, a clinically important decrease in pain was found at 12 months.
Previous studies have shown that botulinum toxin reduces upper limb pain at 8 and 24 weeks45 and that it can also be used to treat shoulder pain associated with spasticity. 46,47
Upper limb pain is common after stroke and it is not unusual for sites and intensity to fluctuate, especially during the first 6 months. Identification of a single cause is often difficult. 117 As spasticity is believed to cause pain, decreasing spasticity may relieve pain. In addition, recent evidence suggests that botulinum toxin may have a direct analgesic effect by blocking transmission of neurotransmitters involved in pain pathways. 27 BoTULS found that pain was decreased at 12 months but not at 1 or 3 months, whereas muscle tone at the elbow was decreased at 1 month but not at 3 or 12 months. Hence, pain was decreased in the absence of a decrease in muscle tone. This suggests that pain reduction may be through a mechanism other than spasticity reduction, or alternatively, as muscle tone was only evaluated at the elbow, it is possible that spasticity was decreased at other joints but this was not recorded. It may be also be the result of the avoidance of late complications of spasticity, such as spasm and contracture.
Patient-selected goals
Treatment with botulinum toxin type A did not lead to improvement in achieving upper limb rehabilitation goals. Two previous trials asked participants to select and score a principle target of treatment from a list of upper limb functional activities (including dressing a sleeve, opening the hand for cleaning the palm) and demonstrated improvement in this activity after botulinum toxin treatment. 42,51 Two previous trials asked participants to identify individual treatment goals41,51 and one demonstrated improved achievement of the goals following treatment with botulinum toxin. 51
Adverse events
There were no significant differences in the number and type of serious adverse events between participants who received botulinum toxin type A and those who did not receive botulinum toxin type A injections. Only one serious adverse event (dysphagia) was potentially related to botulinum toxin type A. Although the number of adverse events was similar between participants who received botulinum toxin type A and those who did not receive injections, there was a higher incidence of general malaise/flu-like/cold symptoms in those who received botulinum toxin type A with a relative risk of 7.6 (95% CI 1.8 to 32.3).
Although the overall numbers of serious adverse events and adverse events reported in this study were higher than in other trials, this probably reflects the robust reporting system employed in line with the Medicines for Human Use (Clinical Trials) Regulations. 100 The few events likely to be related to botulinum toxin type A were in keeping with those reported in other studies and data compiled by the manufacturing company. 32
Subgroup analyses
Time since stroke
There were no significant differences between patients treated within 1 year of stroke and those treated more than 1 year after stroke for the estimated effect of the intervention on any outcome. The hypothesis that time after stroke to treatment with botulinum toxin type A may influence effectiveness was therefore not supported. However, the study was not powered for subgroup analyses and we may have failed to detect important differences.
Severity of initial upper limb function
Participants with some retained function at baseline who were treated with botulinum toxin type A (ARAT 4–56) had a significantly greater improvement in ARAT score at 3 months (change from baseline) when compared with those with no retained function at baseline. This suggests that the improvement in ARAT score at 3 months in the main analysis was predominantly due to participants in the 4–56 group. However, the magnitude of difference between the subgroups (3.3; 95% CI 0.3 to 6.3) was small and this finding is of uncertain clinical significance. It is possible that there is a small group of patients for whom botulinum toxin type A and upper limb therapy can improve active arm function, but we have not been able to demonstrate this convincingly.
There were no other differences between participants with some retained function at baseline and those with no retained function for the estimated effect of botulinum toxin type A on other outcomes.
Economic evaluation
The base-case ICER for botulinum toxin type A plus therapy was £93,500, which is well in excess of the £20,000 threshold value used by NICE. 118 Estimation of the CEAC for botulinum toxin type A plus therapy indicated that there was only a 0.36 probability of its being cost-effective at a ceiling ratio of £20,000 per QALY.
Sensitivity analysis using participants with complete EQ-5D data at 3 months produced an improved point estimate for the ICER of £68,857, but this was still over three times the NICE threshold value. The CEAC for these participants suggests that the probability of botulinum toxin type A plus therapy being cost-effective at £20,000 per QALY is actually lower than in the base case (0.34 versus 0.36, respectively).
Sensitivity analysis on the cost of botulinum toxin type A revealed that even if the cost of the drug were zero, the point estimate of the ICER still exceeded the NICE threshold value (£55,750 versus £20,000, respectively), and the probability of botulinum toxin type A plus therapy being cost-effective at £20,000 was little changed from the base case (0.39 versus 0.36, respectively).
Altering the assumptions regarding the timing of the health-state changes following treatment so that they favoured botulinum toxin type A plus therapy (best–worst QALY analysis) resulted in a lower ICER than in the base-case analysis (£62,333 versus £93,500, respectively). However, the probability that botulinum toxin plus therapy was cost-effective at £20,000 per QALY was actually lower than in the base-case analysis (0.35 versus 0.36, respectively).
Imputing missing data using multiple imputation resulted in an ICER of £86,000 for botulinum toxin plus therapy and a probability of 0.39 of its being cost-effective at £20,000 per QALY.
In summary, even the lowest point estimate of the ICER for botulinum toxin type A plus therapy was over two and a half times the NICE cost-effectiveness threshold value (£55,750 versus £20,000, respectively), and the probability of its being cost-effective at the threshold value did not exceed 0.39, regardless of the assumptions made. The economic analysis therefore provides no evidence to suggest that botulinum toxin type A plus therapy is a cost-effective alternative to therapy alone.
Methodological issues
Study setting
A multicentre RCT is the gold standard study design to assess the effectiveness of an intervention and increase the generalisability of the results. Of the previous 16 trials which have evaluated the effectiveness of using botulinum toxin to treat upper limb spasticity due to stroke, seven were multicentre studies. 36,38,39,41,42,44,51 In BoTULS, for logistical reasons, all centres were in the north of England, but the range of stroke services involved was typical of stroke care across the UK. The majority of botulinum toxin type A injections were given at a single clinic at the regional rehabilitation centre, with a small number being given at one study site. This does have an impact upon the generalisability of results although both clinics followed national guidelines for the range of muscles and dosages of botulinum toxin injected. 9 We had originally intended that botulinum toxin type A injections would be given at each study site by a local clinician or the study clinical research associate, but only two clinicians had training and expertise in giving botulinum toxin and most preferred to refer patients centrally. This involved study participants travelling to the regional rehabilitation centre, which is the usual service model currently used to deliver botulinum toxin treatment in the UK.
Case ascertainment
Unfortunately, it was not possible to describe the proportion of stroke patients who were screened for the study because this was undertaken in a number of settings by a large number of clinicians. Referral to the study was on an ad hoc basis so it was not possible to identify and screen the prevalent population of stroke patients for upper limb spasticity across such a large geographical area. Screening logs were kept but it was not possible to keep an accurate record. To have achieved this would have required significant resource and would have contributed relatively little to our findings, although it would have helped to identify the proportion of prevalent stroke patients who may have benefitted from treatment.
Study therapists had close links with clinical services and we feel that our notification systems enabled us to identify the majority of patients with upper limb spasticity who were in contact with stroke and rehabilitation services. As long-term follow-up and support for stroke patients is limited,1 there were likely to be a number of patients with upper limb spasticity who were not in contact with stroke or rehabilitation services who did not have the opportunity to take part in the study.
Study participants
Recruitment of participants from stroke services avoided a potential participant selection bias, which may have occurred if participants were recruited from specialist rehabilitation services. For 8/16 previously published studies37,38,40,43,45,47,48,51 recruitment was from rehabilitation clinics and for the other 8/16 studies36,39,41,42,44,46,49,50 the setting was unclear. None recruited participants from the wider stroke population.
As there was uncertainty about which patients with upper limb spasticity due to stroke may benefit from botulinum toxin our eligibility criteria were broad and inclusive. Although the incidence and prevalence of upper limb spasticity post stroke is unclear, we feel that study participants were typical of stroke patients who experience upper limb spasticity post stroke in the UK. Study participants may have had less severe spasticity than patients who participated in previous studies (Modified Ashworth Score at the elbow at baseline was two in BoTULS compared with more than two in most previous studies). As we were keen to include participants with retained active upper limb function, this was not surprising as it is unusual to see severe spasticity in this group of patients. The pattern of upper limb spasticity was similar to that seen in clinical practice and described in published studies.
The study design did not include an upper limit of time since stroke because we were keen to have inclusion criteria that reflected the spectrum of patients with upper limb spasticity following stroke seen in clinical practice. In terms of time since stroke, the study population was therefore heterogeneous. As we hypothesised that the effect of treatment may be influenced by time since stroke the study design included a preplanned subgroup analysis of this variable. Previous studies also recruited some patients a number of years after stroke and some have shown benefit. 38,40,42,44,49
Study participants were well matched at baseline with respect to all key variables. The median age of study participants was 67 years, which is younger than the median age at which stroke occurs (74 years64). The higher proportion of male participants recruited (67.8%) may reflect the higher prevalence of stroke in males compared to females. 119,120.
Measurement of spasticity
Spasticity is difficult to define and measure. 18 Although the Modified Ashworth Scale is used to measure spasticity in clinical practice and research studies, it measures muscle tone rather than spasticity. The scale has been validated to measure muscle tone at the elbow121 and although it has been used in some studies to measure tone at other upper limb joints it is unclear how this was undertaken. As there is no validated measure of spasticity at the shoulder, wrist, or fingers, study therapists, who all had extensive experience of stroke rehabilitation, used their clinical judgement to determine the presence of spasticity at joints other than the elbow. As no validated measure exists, the severity of spasticity at these joints was not rated.
Neurophysiological techniques22 and biomechanical measures21 of spasticity have been developed, but are not yet at the stage where they can be used in routine clinical practice or multicentre studies.
Randomisation
One of the strengths of BoTULS is that the methods of randomisation and group allocation concealment employed were robust. Randomisation and allocation concealment were adequately reported in only 8/16 previous studies. 40,41,43,44,46–48,51
Botulinum toxin
In contrast to several other trials evaluating the use of botulinum toxin to treat upper limb spasticity after stroke, the dose and pattern of botulinum toxin type A injections delivered were according to the spasticity pattern of participants and determined by the injecting clinician rather than fixed in a trial protocol. The doses for individual muscles or muscle groups used were in accordance with published guidelines,9 which were developed by an expert panel. Some previous studies have been criticised for using fixed injection protocols, which may not target the most appropriate muscles for treatment gain. 11 By allowing an experienced clinician to determine the dosage and pattern of use of botulinum toxin type A for individual patients we have evaluated how botulinum toxin type A is currently used in clinical practice. Studies of inter- or intra-clinician use of botulinum toxin dosages or patterns of use of botulinum toxin in clinical practice are not available.
Localisation of muscle for injection was by surface anatomy. Although electromyography can be used to identify muscles for treatment, injection site placement by surface anatomy is considered acceptable as botulinum toxin spreads locally to active muscles. 63 Only five of the 16 previously published studies report use of electromyography. 36,37,43,46,47
Our median initial dose of botulinum toxin type A (Dysport) was 200 units (IQR 100–300). This compares with doses of 500–1500 units of botulinum toxin (Dysport) used in previous studies. The dosages of Dysport and Botox are not interchangeable and studies that have used Botox have used dosages ranging from 50 to 360 units. Our study could be criticised for not having used a high enough dose of botulinum toxin type A, but although dosages used were lower than in previous studies, the same level of reduction in spasticity (as measured by the Modified Ashworth Scale at the elbow) was achieved. We can therefore be confident that sufficient botulinum toxin was given to achieve a reduction in muscle tone. The lower dosage of botulinum toxin type A in BoTULS compared with previous studies is likely to reflect the fact that patients with less severe levels of spasticity were included. Guidelines recommend dose adjustment according to the level of spasticity. 9 The pattern of muscles injected was in keeping with common spasticity patterns in the post-stroke upper limb and was similar to published studies. 36,37,39–45,49
Participants in the intervention group were assessed by a study therapist to decide whether they should be referred for further botulinum toxin type A injections at 3, 6 and 9 months. All study therapists were experienced in stroke rehabilitation and had experience in treating patients with spasticity. As spasticity is a chronic condition it was surprising that not all participants received repeat injections and unfortunately we did not record the reasons for this decision. In retrospect, clear criteria for repeat injections should have been developed. We did consider reviewing all participants at the regional spasticity clinic at 3, 6 and 9 months, but this would not have been practical. Failure to show sustained benefit from botulinum toxin type A treatment may be because a significant proportion of participants did not receive 3-monthly repeat injections.
The median dosage of botulinum toxin type A was higher for repeat injections. This may be because participants with more severe spasticity were referred for repeat injections [the median Modified Ashworth Scale score at elbow at 3 months for participants receiving repeat injections was 2 (IQR 1+ to 2) compared with 1+ (IQR 1–2) for those who did not receive injections]. A clear dose–response for treating upper limb spasticity due to stroke with botulinum toxin has not been established and in clinical practice where minimal benefit is obtained from the initial injections a higher dose is usually subsequently used. Uncontrolled studies of repeat injections have used up to 1000 units of Dysport or 50–400 units of Botox.
Although the study protocol did allow for participants with an unacceptable degree of symptomatic spasticity to be referred for treatment with botulinum toxin during the study period, the crossover rate was low.
Upper limb therapy
A standardised therapy programme is one of the strengths of BoTULS as most previous studies have neither described nor quantified the amount of therapy received. The programme was based upon best available evidence and a manual and training programme were developed. Unlike many rehabilitation studies we were able to describe the amount and type of treatment received. As study therapists were trained centrally, delivery and content of the programme should not have differed across the study sites. The therapy programme was provided 2 hours per week for 1 month and for most participants this was likely to be more therapy than they would have received with ‘usual care’. Further upper limb therapy was available at 3, 6 and 9 months at the discretion of the study therapists. As with repeat botulinum toxin injections, we feel that specific criteria for repeat therapy would have been useful and informative. It was unfortunate that data regarding the amount and content of therapy at 3, 6 and 9 months were not suitable for analysis and this was a study limitation.
There was considerable debate in establishing the study about whether upper limb therapy should be ‘usual care’ or a standardised upper limb therapy programme. We felt that to maximise the potential effectiveness of botulinum toxin type A, a standardised therapy programme should be provided. ‘Usual care’ is very variable and difficult to measure and if we had adopted this approach and the study results had been neutral then we could be criticised for not evaluating best practice in relation to botulinum toxin type A and uncertainty would remain about the effectiveness of treating upper limb spasticity due to stroke with botulinum toxin. We did collect information about contact with rehabilitation services at each assessment but as with most rehabilitation RCTs we did not collect detailed information about the therapy received in addition to the study upper limb therapy programme. As this treatment could have been provided by a number of services within each study site it would have been impossible to collect reliable data without intensive effort and significant additional resource.
In designing the study we did consider a factorial design of botulinum toxin type A versus no botulinum toxin type A and enhanced therapy versus ‘usual care’. However, the sample size required to address this issue was over 1000 and we did not feel that this was achievable.
Participants in both intervention and control groups made positive comments about the programme and 44.8% felt that they had gained benefit in terms of upper limb function as a result of the therapy programme. Most found that the upper limb therapy programme met their needs and expectations although it was too easy or too difficult for a small number of participants. Aspects of the programme may need to be modified as 13% experienced pain during stretching or exercises, although discomfort may be unavoidable in some cases and indeed may lead to longer-term benefit. The predominant negative comment about the programme was that it was too short and many participants were keen to have further additional treatment.
Improvements in several outcomes were seen for both intervention and control groups. At 1, 3 and 12 months predefined ARAT success was achieved by 19.5%, 24.2% and 29.3% of participants in the control group, respectively. This improvement was higher than anticipated and may reflect benefit from the upper limb therapy programme, which incorporated repetitive task-specific practice. However, a recent systemic review examining the effects of repetitive task practice on functional ability after stroke showed that improvement in upper limb function did not reach statistical significance. 122
Outcome measures
As the focus of BoTULS was to look at the effect of botulinum toxin type A upon upper limb function in patients with upper limb spasticity due to stroke, the ARAT was selected as the primary outcome measure. The ARAT has been widely used in rehabilitation research and has been shown to be valid, reliable and sensitive to change in stroke patients. 123,124 Previous studies have predominantly assessed specific basic functional activities, e.g. putting the arm through a sleeve, or used global disability scales as functional outcomes measures. Of the 16 previous studies only nine defined a primary outcome measure38–41,43,44,47,50,51 and only one was a functional assessment. 40
Defining a clinically meaningful change in arm function is difficult and the judgements upon which the decision is based can always be challenged. Improvement in arm function is influenced by a number of parameters including time since stroke and severity of the initial neurological deficit and the perspectives of patients, carers and clinicians may differ. More recovery would be expected for participants randomised early after stroke and for those who had some active upper limb function at baseline. Initially, BoTULS included only participants with some active upper limb function and treatment success was based upon an improvement of the ARAT score by six points, which has been suggested as the minimal clinically important change for the ARAT. 101 When the eligibility criteria were widened to include patients with no active upper limb function (ARAT 0–3) we felt that an improvement of three points would be a meaningful clinical improvement in this group because they have a poorer prognosis for recovery of active function.
It can also be argued that the ARAT is not the optimum instrument to measure outcome in patients with no active upper limb function as many patients might be expected to show little improvement. Of participants who had no active upper limb function at baseline (ARAT 0–3), 67.4% in the control group and 77.2% in the intervention group demonstrated no change on the ARAT at 1 month. Assessment of specific basic upper limb functional activities, e.g. putting hand into a sleeve, opening the hand for washing, may be a more appropriate primary outcome measure for this group. Measures of these basic upper limb functional activities were included as secondary outcomes and we were able to demonstrate improvement for a number of these. In clinical practice and research an outcome measure needs to be able to measure change across the spectrum of patients to whom treatment is given and we do not feel that current measures of upper limb function fulfil this criterion.
The secondary outcome measures chosen are widely used in rehabilitation research. The COPM was included as an outcome measure because of the recognition that improvement in patient-selected goals may provide a more meaningful treatment evaluation than standard outcome measures. 54 However, the goals set may or may not have been realistic and achievable.
Timing of outcome measurements
Botulinum toxin type A takes approximately 1 week to achieve maximal effect and wears off after 3–4 months. Therefore, the 1-month outcome assessment should have captured a maximal treatment effect directly due to the botulinum toxin type A. At 3 months the effects of the initial injection would be wearing off so any benefit that relied upon a direct treatment effect of botulinum toxin type A may have been reduced. As injections could be repeated at 3, 6 and 9 months, then the 12-month outcome assessment may also have been at a time when any treatment effect was declining. As there may be benefits that are sustained when the effect of the toxin wears off, it was not unreasonable to look at the intermediate and long-term effects at these times. It would have been useful to have included further assessments at 4 months, 7 months and 10 months to study the effects of repeat injections because at these times the treatment effect would likely be greater; however, this would have needed additional resources and increased participant burden.
Blinding
BoTULS did not use placebo injections so participants and the study therapists who delivered the upper limb therapy programme were not blinded to the randomisation group, which was a source of potential bias. Some participants were disappointed that they did not receive botulinum toxin and may have felt disadvantaged. Although therapists were trained to deliver a standard therapy programme it is possible that there were inadvertent differences in content between participants in the intervention and control groups. Every effort was made to ensure that the study therapists who undertook outcome assessments were blinded to the patients’ randomisation group. As participants were unblinded, it was possible for outcome assessors to become unintentionally unblinded during conversation. The assessor was unblinded for 36.1% of primary outcome assessments and it is difficult to know how this could have been reduced because participants were asked not to mention the treatment they had received at each outcome assessment.
In planning the study, the use of placebo injections was discussed at length. It was decided that they should not be used because the study was a pragmatic trial and because it was felt unethical to subject participants to sham injections.
Study dropout and data quality
One of the strengths of this study is the quality of the data. Follow-up levels were high at all assessment points and levels of missing data were low.
Statistical considerations
The study achieved the prespecified sample size of 332 participants so we can be confident that we have not missed an important treatment effect upon our primary outcome measure. However, it could be argued that our prespecified level of successful treatment was too ambitious.
BoTULS is the largest RCT to evaluate the role of botulinum toxin in the treatment of upper limb spasticity due to stroke undertaken to date. Sample sizes of previous RCTs range from 15 to 126 participants (median n = 39) and the total number of participants who have been randomised in studies of botulinum toxin in the treatment of upper limb spasticity due to stroke is 785. This rises to 1118 with our data. Only 9/16 of the previous studies reported a power calculation39–42,44,46–48,51 and seven achieved their prespecified sample size. 39–42,44,47,51
Due to the nature of the data collected in the study many statistical comparisons were made during the analyses. As multiple statistical testing could have been responsible for some of the statistically significant findings, we have been cautious in the interpretation of the results. Sample size calculations were not undertaken for secondary outcome measures or for subgroup analyses so we may have failed to detect some important treatment effects. In addition, as 12-month follow-up was curtailed, 12-month outcomes were available for 57% of participants and therefore we may have failed to detect an important treatment effect because of the reduced sample size at this stage.
We have undertaken an intention-to-treat analysis as described in our protocol. We have not undertaken an on-treatment analysis because 96.5% of participants in the intervention group received treatment with botulinum toxin type A as planned.
Economic evaluation
The main limitation of the cost-effectiveness analysis was the relatively short time horizon over which it was conducted. The rationale for adopting a 3-month time horizon rather than 12 months was the loss of participant responses due to curtailment of 12-month follow-up. The proportion of participants providing EQ-5D data at 12 months was 52.4%. This is considerably lower than the corresponding figures for baseline, 1 month and 3 months, which were 100%, 83.7% and 85.2%, respectively.
One option would have been to impute the missing 12-month data, but this would have meant that even if the estimation of QALYs was restricted to the baseline and 12-month EQ-5D values only, data would have to be imputed for almost half the sample. If the intermediate EQ-5D values were also used in the QALY estimation, then data for more than half the sample would need to be imputed. For these reasons, a decision was made to conduct the analysis over 3 months where levels of missing data were much lower. This is not to say that there would not be benefits in extrapolating costs and outcomes over a longer time period. This could be done using economic modelling, which would incorporate data from the trial and other relevant sources to estimate cost-effectiveness beyond the time horizon of the trial. Although such an exercise is beyond the scope of this study, it would be a potentially worthwhile piece of future research.
Chapter 6 Conclusions
The addition of botulinum toxin type A to an upper limb therapy programme to treat spasticity due to stroke did not enhance improvement in upper limb function when assessed by the prespecified primary outcome measure.
Botulinum toxin type A reduced muscle tone and the level of reduction was similar to that in previous studies.
Treatment with botulinum toxin type A was associated with increased upper limb strength at 3 months but this was not sustained until 12 months.
Treatment with botulinum toxin type A resulted in improvements in basic upper limb functional activities related to specific tasks, e.g. dressing a sleeve, opening the hand for cleaning the palm or opening the hand for cutting fingernails, which were sustained until 12 months for some activities.
Participants treated with botulinum toxin type A demonstrated a small improvement in active upper limb function at 3 months when this was analysed as change in ARAT score from baseline. This was predominantly because of improvement in participants with some retained function at baseline. However, the size of the improvement was small and this finding is of uncertain clinical significance. Improvement in upper limb function was not sustained at 12 months.
It is of interest that participants in both randomisation groups made improvements in upper limb function over a 12-month period.
Treatment with botulinum toxin type A did not result in any important improvements in stroke-related quality of life.
Botulinum toxin type A appeared to have a long-term benefit in terms of pain reduction for patients with upper limb spasticity due to stroke.
Failure to show sustained benefit at 12 months may have been because a significant proportion of participants did not receive 3-monthly repeat injections, the timing of outcome measurement in relation to repeat injections, or lack of power to detect change at this time.
The side effects of botulinum toxin type A were minor and predominantly of flu-like illness.
The addition of botulinum toxin type A to an upper limb therapy programme for the treatment of upper limb spasticity due to stroke was not cost-effective at 3 months.
Implications for clinical practice
National clinical guidelines for the management of spasticity in adults using botulinum toxin were updated in January 2009. 63 Our study has followed the guidelines by ensuring that botulinum toxin was not used in isolation, but was part of a multidisciplinary approach to the management of spasticity.
Our results will help to inform clinicians regarding the outcomes that may be improved by treating upper limb spasticity due to stroke with botulinum toxin type A. Management of spasticity should include clearly agreed goals for treatment and for our study the primary goal was improvement in upper limb function. The guidelines suggest that active functional gain is an appropriate goal in some cases. However, our primary analysis and preplanned subgroup analyses did not find any significant clinical improvement in active upper limb function. Other goals suggested in the guidelines include improvement of basic upper limb tasks and pain reduction, and our results support these choices. Although repeated treatment is commonly used in clinical practice and suggested in the guidelines, our study was not able to demonstrate sustained improvement for all outcomes.
Guidelines suggest that botulinum toxin ‘has the potential to reduce the overall cost of ongoing care in people with severe spasticity through the prevention of contracture and deformity, and improved ease of care and handling’. Botulinum toxin type A was not a cost-effective treatment for the patients included in our study, but we acknowledge that we did not examine the longer-term consequences.
Implications for research
We have demonstrated that it is possible to undertake an investigator-led multicentre study of an investigational medicinal product and standardised therapy programme in the UK. Only a small number of multicentre stroke rehabilitation studies have been published to date, although since the Stroke Research Network was established in 2005 numbers are increasing.
Further research is needed to increase our understanding of the natural history and clinical impact of spasticity following stroke, and to explain the relationship between spasticity and functional limitation. Studies are needed to improve the measurement of spasticity and to develop valid measures for all upper limb joints.
Further work is required to establish the optimum dosage and pattern of injections of botulinum toxin type A to treat upper limb spasticity due to stroke and to define the efficacy of repeat injections. The timing of outcome measures should relate to the time when botulinum toxin type A is likely to be optimally effective and measure outcome in the longer term.
There is a need for patients, clinicians and the research community to clearly define important clinical outcomes for patients with upper limb impairment. Further work is needed to develop robust person-centred outcome measures that can be used in national or international multicentre studies.
A national register to record the clinical details of patients who receive treatment with botulinum toxin, their goals and outcomes would be helpful to assist the development of RCTs to identify which patients benefit from this treatment.
Acknowledgements
The BoTULS trial research costs were funded by the NIHR Health Technology Assessment programme. Additional treatment costs to provide the upper limb therapy programme were available from an NHS subvention. Ipsen Ltd provided the botulinum toxin type A (Dysport) free of charge.
We would like to acknowledge the contribution of Dr Tony Field, Consultant in Rehabilitation Medicine, Hunters Moor Regional Rehabilitation Centre, Newcastle upon Tyne, who was involved in designing and planning the project. He sadly died in 2005.
We would also like to acknowledge the following for their contribution:
-
Patients who participated in the trial.
-
Study therapists: Lydia Aird, Louise Baxter, Lianne Brkic, Caroline Deacon, Hilary Ford, Heather Hunter, Nina Lishman, Paul McNeillie, Dadirayi Mhiripiri, Janet Nesbitt and Frances Sapsford.
-
Co-ordinating centre team: Jessie Cowe, Katharine Forster, Linda Goulbourne, Anne Harrison, Joseph Hoben, Debbie Jones and Ruth Wood.
-
Principal investigators: Professor David Barer (Gateshead Health NHS Trust), Dr David Bruce (North Tees and Hartlepool NHS Trust), Dr Tim Cassidy (City Hospitals Sunderland NHS Foundation Trust and Gateshead Health NHS Trust), Dr Paul Davies (North Cumbria Acute Hospitals NHS Trust), Dr Philip Earnshaw (County Durham and Darlington NHS Foundation Trust), Dr Akif Gani (Newcastle Primary Care Trust), Professor Chris Gray (City Hospitals Sunderland NHS Foundation Trust), Dr Ali Mehrzard (County Durham and Darlington NHS Foundation Trust) and Dr Jon Scott (South Tyneside NHS Foundation Trust).
-
Staff at the International Centre for Neurorehabilitation, Newcastle upon Tyne: Dr John Macfarlane, Lesley Kidd, Debbie Best and Julie Long.
-
Sandra Stark and Julia Johnson (International Centre for Neurorehabilitation, Newcastle upon Tyne), who assisted with the upper limb therapy programme design.
-
Professor Garth Johnson for advice about measurement of spasticity.
-
DMEC external members: Professor Martin Dennis, (chairperson) (University of Edinburgh), Professor Ian Ford (University of Glasgow) and Professor Marion Walker (University of Nottingham).
-
TSC external members: Professor Peter Langhorne (chairperson) (University of Glasgow), Professor Bipin Bhakta (University of Leeds), Mrs Beryl Fairless (lay member) and Professor Anne Forster (University of Leeds).
-
Ipsen Ltd who provided advice, support and the botulinum toxin type A (Dysport).
-
UK Stroke Research Network: Karin Bond, Ellen Brown, Claire Garner and Penny Williams.
Contribution of authors
Dr Lisa Shaw (Clinical Research Associate) was responsible for day-to-day management of the study, delivery of botulinum toxin type A injections and drafting the report. Professor Helen Rodgers (Professor of Stroke Care and chief investigator) designed the RCT and was responsible for the overall management of the study and drafting the report. Dr Christopher Price (Consultant Stroke Physician and Clinical Senior Lecturer) designed the RCT and undertook the initial literature review. Dr Frederike van Wijck (Senior Lecturer, Physiotherapy) contributed to the design of the RCT, designed the upper limb therapy programme, and trained the research therapists. Dr Phil Shackley (Senior Lecturer, Health Economics) designed and conducted the economic analyses. Dr Nick Steen (Principle Research Associate, statistics) was responsible for the trial statistics. Professor Michael Barnes (Professor of Neurological Rehabilitation) contributed to the design of the RCT and provided expertise in spasticity management and the use of botulinum toxin. Professor Gary Ford (Professor of Clinical Pharmacology) contributed to the design of the RCT. Dr Laura Graham (Consultant in Rehabilitation Medicine) provided expertise in spasticity management and the use of botulinum toxin, and was involved in delivery of botulinum toxin type A injections. All authors have contributed to analyses, interpretation of results and drafts of the report.
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
- National Audit Office . Reducing Brain Damage: Faster Access to Better Stroke Care 2005.
- Heller A, Wade DT, Wood VA, Sunderland A, Hewer RL, Ward E. Arm function after stroke: measurement and recovery over the first three months. J Neurol Neurosurg Psychiatry 1987;50:714-19.
- Skilbeck CE, Wade DT, Hewer RL, Wood VA. Recovery after stroke. J Neurol Neurosurg Psychiatry 1983;46:5-8.
- Broeks JG, Lankhorst GJ, Rumping K, Prevo AJ. The long-term outcome of arm function after stroke: results of a follow-up study. Disab Rehab 1999;21:357-64.
- Wade D. Editorial. Clin Rehab 1997;11:91-2.
- Friedman P. Gait recovery after hemiplegic stroke. Int Disab Stud 1990;12:119-22.
- Lance JW, Feldman RG, Young RR, Koella WP. Spasticity: Disordered Motor Control. Chicago, IL: Symposium Specialists Inc.; 1980.
- Pandyan AD, Gregoric M, Barnes MP, Wood D, Van Wijck F, Burridge J, et al. Spasticity: clinical perceptions, neurological realities and meaningful measurement. Disab Rehab 2005;27:2-6.
- Barnes MP, Bhakta BB, Moore P, Richardson D, Salisbury C, Thornton H, et al. The management of adults with spasticity using botulinum toxin: a guide to clinical practice. London: Radius Healthcare; 2001.
- Mayer NH, Esquenazi A, Childers MK. Common patterns of clinical motor dysfunction. Muscle Nerve 1997;20:s21-s35.
- Sheean GL. Botulinum treatment of spasticity: why is it so difficult to show a functional benefit?. Curr Opin Neurol 2001;14:771-6.
- Ada L, O’Dwyer N, O’Neill E. Relation between spasticity, weakness and contracture of the elbow flexors and upper limb activity after stroke: an observational study. Disab Rehab 2006;28:891-7.
- Leonard CT, Gardipee KA, Koontz JR, Anderson J-H, Wilkins SA. Correlation between impairment and motor performance during reaching tasks in subjects with spastic hemiparesis. J Rehab Med 2006;38:243-9.
- Brashear A, Mayer NH. Spasticity and other forms of muscle overactivity in the upper motor neuron syndrome. New York, NY: We Move; 2009.
- Watkins CL, Leathley MJ, Gregson JM, Moore AP, Smith TL, Sharma AK. Prevalence of spasticity post stroke. Clin Rehab 2002;16:515-22.
- Sommerfeld DK, Eek EUB, Svensson A-K, Holmqvist LW, von Arbin MH. Spasticity after stroke: its occurrence and association with motor impairments and activity limitations. Stroke 2004;35:134-9.
- Intercollegiate Stroke Working Party . National Clinical Guideline for Stroke 2008.
- Burridge JH, Wood DE, Hermens HJ, Voerman GE, Johnson GR, van Wijck F, et al. Theoretical and methodological considerations in the measurement of spasticity. Disab Rehab 2005;27:69-80.
- Bohannon RW, Smith MB. Interrater reliability of a modified Ashworth scale of muscle spasticity. Physical Ther 1987;67:206-7.
- Johnson GR, Barnes MP, Johnson GR. Upper motor neurone syndrome and spasticity. Cambridge: Cambridge University Press; 2001.
- Wood DE, Burridge JH, van Wijck FM, McFadden C, Hitchcock RA, Pandyan AD, et al. Biomechanical approaches applied to the lower and upper limb for the measurement of spasticity: a systematic review of the literature. Disab Rehab 2005;27:19-32.
- Voerman GE, Gregoric M, Hermens HJ. Neurophysiological methods for the assessment of spasticity: the Hoffmann reflex, the tendon reflex, and the stretch reflex. Disab Rehab 2005;27:33-68.
- Collins MD, East AK. Phylogeny and taxonomy of the food-borne pathogen Clostridium botulinum and its neurotoxins. J Appl Microbiol 1998;84:5-17.
- Goonetilleke A, Harris JB. Clostridial neurotoxins. J Neurol Neurosurg Psychiatry 2004;75:35-9.
- Sobel J. Botulism. Clin Infect Dis 2005;41:1167-73.
- Erbguth FJ. From poison to remedy: the chequered history of botulinum toxin. J Neural Transm 2008;115:559-65.
- Dolly JO, Aoki KR. The structure and mode of action of different botulinum toxins. Eur J Neurol 2006;13:1-9.
- Aoki KR. Review of a proposed mechanism for the antinociceptive action of botulinum toxin type A. Neurotoxicology 2005;26:785-93.
- Mense S. Neurobiological basis for the use of botulinum toxin in pain therapy. J Neurol 2004;251:I1-7.
- Rossetto O, Montecucco C, Moore P, Naumann M. Handbook of botulinum toxin treatment. Oxford: Blackwell Science Ltd; 2003.
- Naumann M, Toyka KV, Moore P, Moore P, Naumann M. Handbook of botulinum toxin treatment. Oxford: Blackwell Science Ltd; 2003.
- Electronic Medicines Compendium n.d. www.emc.medicines.org.uk.
- Dressler D, Benecke R. Pharmacology of therapeutic botulinum toxin preparations. Disab Rehab 2007;29:1761-8.
- Naumann M, Albanese A, Heinen F, Molenaers G, Relja M. Safety and efficacy of botulinum toxin type A following long-term use. Eur J Neurol 2006;13:35-40.
- Brin MF. Botulinum toxin: chemistry, pharmacology, toxicity, and immunology. Muscle Nerve Suppl 1997;6:S146-68.
- Simpson DM, Alexander DN, O’Brien CF, Tagliati M, Aswad AS, Leon JM, et al. Botulinum toxin type A in the treatment of upper extremity spasticity: a randomized, double-blind, placebo-controlled trial. Neurology 1996;46:1306-10.
- Hesse S, Reiter F, Konrad M, Jahnke MT. Botulinum toxin type A and short-term electrical stimulation in the treatment of upper limb flexor spasticity after stroke: a randomized, double-blind, placebo-controlled trial. Clin Rehab 1998;12:381-8.
- Smith SJ, Ellis E, White S, Moore AP. A double-blind placebo-controlled study of botulinum toxin in upper limb spasticity after stroke or head injury. Clin Rehab 2000;14:5-13.
- Bakheit AM, Thilmann AF, Ward AB, Poewe W, Wissel J, Muller J, et al. A randomized, double-blind, placebo-controlled, dose-ranging study to compare the efficacy and safety of three doses of botulinum toxin type A (Dysport) with placebo in upper limb spasticity after stroke. Stroke 2000;31:2402-6.
- Bhakta BB, Cozens JA, Chamberlain MA, Bamford JM. Impact of botulinum toxin type A on disability and carer burden due to arm spasticity after stroke: a randomised double blind placebo controlled trial. J Neurol Neurosurg Psychiatry 2000;69:217-21.
- Bakheit AM, Pittock S, Moore AP, Wurker M, Otto S, Erbguth F, et al. A randomized, double-blind, placebo-controlled study of the efficacy and safety of botulinum toxin type A in upper limb spasticity in patients with stroke. Eur J Neurol 2001;8:559-65.
- Brashear A, Gordon MF, Elovic E, Kassicieh VD, Marciniak C, Do M, et al. Intramuscular injection of botulinum toxin for the treatment of wrist and finger spasticity after a stroke. N Engl J Med 2002;347:395-400.
- Brashear A, McAfee AL, Kuhn ER, Fyffe J. Botulinum toxin type B in upper-limb poststroke spasticity: a double-blind, placebo-controlled trial. Arch Physical Med Rehab 2004;85:705-9.
- Childers MK, Brashear A, Jozefczyk P, Reding M, Alexander D, Good D, et al. Dose-dependent response to intramuscular botulinum toxin type A for upper-limb spasticity in patients after a stroke. Arch Physical Med Rehab 2004;85:1063-9.
- Suputtitada A, Suwanwela NC. The lowest effective dose of botulinum A toxin in adult patients with upper limb spasticity. Disab Rehab 2005;27:176-84.
- Yelnik AP, Colle FM, Bonan IV, Vicaut E. Treatment of shoulder pain in spastic hemiplegia by reducing spasticity of the subscapular muscle: a randomised, double blind, placebo controlled study of botulinum toxin A. J Neurol Neurosurg Psychiatry 2007;78:845-8.
- Marco E, Duarte E, Vila J, Tejero M, Guillen A, Boza R, et al. Is botulinum toxin type A effective in the treatment of spastic shoulder pain in patients after stroke? A double-blind randomized clinical trial. J Rehab Med 2007;39:440-7.
- Kong K-H, Neo J-J, Chua KS. A randomized controlled study of botulinum toxin A in the treatment of hemiplegic shoulder pain associated with spasticity. Clin Rehab 2007;21:28-35.
- Jahangir AW, Tan HJ, Norlinah MI, Nafisah WY, Ramesh S, Hamidon BB, et al. Intramuscular injection of botulinum toxin for the treatment of wrist and finger spasticity after stroke. Med J Malaysia 2007;62:319-22.
- de Boer KS, Arwert HJ, de Groot JH, Meskers CGM, Mishre ADR, Arendzen JH. Shoulder pain and external rotation in spastic hemiplegia do not improve by injection of botulinum toxin A into the subscapular muscle. J Neurol Neurosurg Psychiatry 2008;79:581-3.
- McCrory P, Turner-Stokes L, Baguley IJ, De Graaff S, Katrak P, Sandanam J, et al. Botulinum toxin A for treatment of upper limb spasticity following stroke: a multi-centre randomized placebo-controlled study of the effects on quality of life and other person-centred outcomes. J Rehab Med 2009;41:536-44.
- van Kuijk AA, Geurts ACH, Bevaart BJW, van Limbeek J. Treatment of upper extremity spasticity in stroke patients by focal neuronal or neuromuscular blockade: a systematic review of the literature. J Rehab Med 2002;34:51-6.
- Cardoso E, Rodrigues B, Lucena R, Oliveira IR, Pedreira G, Melo A. Botulinum toxin type A for the treatment of the upper limb spasticity after stroke: a meta-analysis. Arquiv Neuro-Psiquiatria 2005;63:30-3.
- Garces K, McCormick A, McGahan L, Skidmore B. Botulinum Toxin A for Upper and Lower Limb Spasticity: A Systematic Review 2005.
- Rosales RL, Chua-Yap AS. Evidence-based systematic review on the efficacy and safety of botulinum toxin-A therapy in post-stroke spasticity. J Neural Transm 2008;115:617-23.
- Elia A, Filippini G, Calandrella D, Albanese A. Botulinum neurotoxins for post stroke spasticity in adults: a systematic review. Movement Disord 2009;24:801-12.
- Rodriquez AA, McGinn M, Chappell R. Botulinum toxin injection of spastic finger flexors in hemiplegic patients. Am J Physical Med Rehab 2000;79:44-7.
- Lagalla G, Danni M, Reiter F, Ceravolo MG, Provinciali L. Post-stroke spasticity management with repeated botulinum toxin injections in the upper limb. Am J Physical Med Rehab 2000;79:377-84.
- Gordon MF, Brashear A, Elovic E, Kassicieh D, Marciniak C, Liu J, et al. Repeated dosing of botulinum toxin type A for upper limb spasticity following stroke. Neurology 2004;63:1971-3.
- Bakheit AMO, Fedorova NV, Skoromets AA, Timerbaeva SL, Bhakta BB, Coxon L. The beneficial antispasticity effect of botulinum toxin type A is maintained after repeated treatment cycles. J Neurol Neurosurg Psychiatry 2004;75:1558-61.
- Cardoso E, Pedreira G, Prazeres A, Ribeiro N, Melo A. Does botulinum toxin improve the function of the patient with spasticity after stroke?. Arquiv Neuro-Psiquiatria 2007;65:592-5.
- Elovic EP, Brashear A, Kaelin D, Liu J, Millis SR, Barron R, et al. Repeated treatments with botulinum toxin type a produce sustained decreases in the limitations associated with focal upper-limb poststroke spasticity for caregivers and patients. Archiv Physical Med Rehab 2008;89:799-806.
- Royal College of Physicians, British Society of Rehabilitation Medicine, Chartered Society of Physiotherapy, Association of Chartered Physiotherapists Interested in Neurology . Spasticity in Adults: Management Using Botulinum Toxin. National Guidelines. 2009.
- Rothwell PM, Coull AJ, Giles MF, Howard SC, Silver LE, Bull LM, et al. Change in stroke incidence, mortality, case-fatality, severity, and risk factors in Oxfordshire, UK from 1981 to 2004 (Oxford Vascular Study). Lancet 2004;363:1925-33.
- Stroke Unit Trialists Collaboration . Organised inpatient (stroke unit) care for stroke. Cochrane Database Syst Rev 2007.
- Lyle RC. A performance test for assessment of upper limb function in physical rehabilitation treatment and research. Int J Rehab Res 1981;4:483-92.
- Sharpless JW. Mossman’s A problem-orientated approach to stroke rehabilitation. Illinois: Charles C Thomas; 1982.
- Demeurisse G, Demol O, Robaye E. Motor evaluation in vascular hemiplegia. Eur Neurol 1980;19:382-9.
- Sunderland A, Tinson D, Bradley L, Hewer RL. Arm function after stroke. An evaluation of grip strength as a measure of recovery and a prognostic indicator. J Neurol Neurosurg Psychiatry 1989;52:1267-72.
- Mahoney FI, Barthel DW. Functional evaluation: the Barthel Index. Maryland State Med J 1965;14:61-5.
- Law M, Baptiste S, McColl M, Opzoomer A, Polatajko H, Pollock N. The Canadian Occupational Performance Measure: an outcome measure for occupational therapy. Can J Occup Ther – Rev Can Ergother 1990;57:82-7.
- Price DD, Bush FM, Long S, Harkins SW. A comparison of pain measurement characteristics of mechanical visual analogue and simple numerical rating scales. Pain 1994;56:217-26.
- Duncan PW, Bode RK, Min Lai S, Perera S. Rasch analysis of a new stroke-specific outcome scale: the Stroke Impact Scale. Arch Physical Med Rehab 2003;84:950-63.
- Euroquol Group . Euroquol – a new facility for the measurement of health-related quality of life. Health Policy 1990;16:199-20.
- Bamford JM, Sandercock PA, Warlow CP, Slattery J. Interobserver agreement for the assessment of handicap in stroke patients. Stroke 1989;20.
- Hodkinson HM. Evaluation of a mental test score for assessment of mental impairment in the elderly. Age and Ageing 1972;1:233-9.
- Al-Khawaja I, Wade DT, Collin CF. Bedside screening for aphasia: a comparison of two methods. J Neurol 1996;243:201-4.
- Bamford J, Sandercock P, Dennis M, Burn J, Warlow C. Classification and natural history of clinically identifiable subtypes of cerebral infarction. Lancet 1991;337:1521-6.
- Royal College of Physicians . Guidelines for the Use of Botulinum Toxin (BTX) in the Management of Spasticity in Adults 2002.
- Intercollegiate Stroke Working Party . National Clinical Guideline for Stroke 2004.
- Wade D. Goal planning in stroke rehabilitation: evidence. Topics Stroke Rehab 1999;6:37-42.
- Pomeroy V, Tallis R. Restoring movement and functional ability after stroke: now and the future. Physiotherapy 2002;88:3-17.
- van der Lee JH, Snels IA, Beckerman H, Lankhorst GJ, Wagenaar RC, Bouter LM. Exercise therapy for arm function in stroke patients: a systematic review of randomized controlled trials. Clin Rehab 2001;15:20-31.
- Kwakkel G, Wagenaar R, Twisk J, Lankhorst G, Koetsier J. Intensity of leg and arm training after primary middle-cerebral-artery stroke: a randomised trial. Lancet 1999;354:191-6.
- Dean CM, Richards CL, Malouin F. Task-related circuit training improves performance of locomotor tasks in chronic stroke: a randomized, controlled pilot trial. Arch Physical Med Rehab 2000;81:409-17.
- Dean CM, Shepherd RB. Task-related training improves performance of seated reaching tasks after stroke. A randomized controlled trial. Stroke 1997;28:722-8.
- Carr J, Shepherd RB. Stroke Rehabilitation. Guidelines for exercise training to optimise motor skill. London: Butterworth Heinemann; 2003.
- Barnes MP, Johnson GR. Upper motor neurone syndrome and spasticity. Cambridge: Cambridge University Press; 2001.
- Barnes M, Dobkin B, Bogousslavsky J. Recovery after stroke. Cambridge: Cambridge University Press; 2005.
- Edwards S. Neurological physiotherapy. Edinburgh: Churchill Livingstone; 2002.
- Lannin NA, Herbert RD. Is hand splinting effective for adults following stroke? A systematic review and methodologic critique of published research. Clin Rehab 2003;17:807-16.
- Scottish Intercollegiate Guidelines Network (SIGN) . Guidelines on Stroke n.d. www.sign.ac.uk (accessed 9 November 2005).
- Stokes M. Physical management in neurological rehabilitation. Edinburgh: Elsevier Mosby; 2005.
- Van Peppen RPS, Kwakkel G, Wood-Dauphinee S, Hendriks HJM, Van der Wees PJ, Dekker J. The impact of physical therapy on functional outcomes after stroke: what’s the evidence?. Clin Rehab 2004;18:833-62.
- Hanlon RE. Motor learning following unilateral stroke. Arch Phys Med Rehab 1996;77:811-15.
- Magill RA. Motor learning: concepts and applications. Boston, MA: McGraw Hill; 2001.
- Shea JB, Morgan RL. Contextual interference effects on the acquisition, retention and transfer of a motor skill. J Exp Psychol: Learn Mem Cogn 1979;5:179-87.
- Duncan PW. Synthesis of intervention trials to improve motor recovery following stroke. Topics Stroke Rehab 1997;3:1-20.
- van Wijck F, Mackenzie J, Hooper J, Barnes M, Johnson G. Improving arm function in patients with chronic spasticity: combining botulinum toxin with a task orientated motor learning programme. Int J Rehab Res 2004;27.
- Medicines for Human Use (Clinical Trials) Regulations 2004. www.uk-legislation.hmso.gov.uk/si/si2004/20041031.htm.
- van der Lee JH, Wagenaar RC, Lankhorst GJ, Vogelaar TW, Deville WL, Bouter LM. Forced use of the upper extremity in chronic stroke patients: results from a single-blind randomized clinical trial. Stroke 1999;30:2369-75.
- Mendelow AD, Gregson BA, Fernandes HM, Murray GD, Teasdale GM, Hope DT, et al. Early surgery versus initial conservative treatment in patients with spontaneous supratentorial intracerebral haematomas in the International Surgical Trial in Intracerebral Haemorrhage (STICH): a randomised trial. Lancet 2005;365:387-97.
- Fleiss JL. Statistical methods for rates and proportions. New York, NY: Wiley; 1981.
- International Conference on Harmonisation . Harmonised Tripartite Guideline for Good Clinical Practice n.d. www.ich.org.
- Department of Health . Research Governance Framework for Health and Social Care 2005.
- MRC . Guidelines for Good Clinical Practice in Clinical Trails 1998.
- Pandyan AD, Vuadens P, van Wijck FMJ, Stark S, Johnson GR, Barnes MP. Are we underestimating the clinical efficacy of botulinum toxin (type A)? Quantifying changes in spasticity, strength and upper limb function after injections of Botox to the elbow flexors in a unilateral stroke population. Clin Rehab 2002;16:654-60.
- National Institute for Clinical Excellence . Guide to the Methods of Technology Appraisal 2004.
- Curtis L. Unit costs of health and social care. Canterbury: PSSRU, University of Kent; 2007.
- British National Formulary. London: BMJ Publishing Group Ltd and RPS Publishing; 2007.
- Brooks R. EuroQol: the current state of play. Health Policy 1996;37:53-72.
- Dolan P. Modeling valuations for EuroQol health states. Medical Care 1997;35:1095-108.
- Richardson G, Manca A. Calculation of quality adjusted life years in the published literature: a review of methodology and transparency. Health Econ 2004;13:1203-10.
- O’Brien B, Briggs AH. Analysis of uncertainty in health care cost-effectiveness studies: an introduction to statistical issues and methods. Stat Meth Med Res 2002;11:455-68.
- Claxton K, Fenwick E, Sculpher M, Jones A. The Elgar companion to health economics. Cheltenham: Edward Elgar Publishing; 2006.
- Schafer JL. Analysis of incomplete multivariate data. London: Chapman and Hall; 1997.
- Price C. Treatment of shoulder and upper limb pain after stroke: an obstacle course for evidence based practice. Rev Clin Gerontol 2003;13:321-33.
- Rawlins MD, Culyer AJ. National Institute for Clinical Excellence and its value judgments. BMJ 2004;329:224-7.
- Geddes JML, Fear J, Tennant A, Pickering A, Hillman M, Chamberlain MA. Prevalence of self reported stroke in a population in northern England. J Epidemiol Commun Health 1996;50:140-3.
- O’Mahony PG, Thomson RG, Dobson R, Rodgers H, James OFW. The prevalence of stroke and associated disability. J Publ Health Med 1999;21:166-71.
- Pandyan AD, Johnson GR, Price CI, Curless RH, Barnes MP, Rodgers H. A review of the properties and limitations of the Ashworth and modified Ashworth Scales as measures of spasticity. Clin Rehab 1999;13:373-83.
- French B, Thomas LH, Leathley MJ, Sutton CJ, McAdam J, Forster A, et al. Repetitive task training for improving functional ability after stroke. Cochrane Database Syst Rev 2007.
- Platz T, Pinkowski C, van Wijck F, Kim I-H, di Bella P, Johnson G. Reliability and validity of arm function assessment with standardized guidelines for the Fugl–Meyer Test, Action Research Arm Test and Box and Block Test: a multicentre study. Clin Rehab 2005;19:404-11.
- Lang CE, Wagner JM, Dromerick AW, Edwards DF. Measurement of upper extremity function after stroke: properties of the Action Research Arm Test. Arch Phys Med Rehabil 2006;87:1605-10.
- SIGN . Methodology Checklist 2: Randomised Controlled Trials 2004. www.sign.ac.uk.
Appendix 1 Additional literature review documents
Literature search strategy
Literature search strategy for studies of botulinum toxin for treatment of upper limb spasticity following stroke.
The following databases were accessed:
-
MEDLINE (1950 to July 2009) via Ovid
-
PubMed
-
EMBASE (1980 to July 2009) via Ovid
-
CINAHL (1982 to July 2009) via Ovid
-
EBM reviews via Ovid.
The following search terms were used and combined as the databases allowed:
-
stroke/cerebrovascular accident
-
upper limb/arm
-
spasticity
-
botulinum toxin.
The limits ‘human’ and ‘english’ were applied.
Database results were manually scanned and potentially relevant abstracts were reviewed. Full papers were selected from potentially relevant abstracts and were fully reviewed to determine appropriateness of inclusion. References of retrieved studies were also reviewed and any further potentially relevant papers were obtained and assessed.
Methodological appraisal of randomised controlled trials
Randomised controlled trials were methodologically appraised using the Scottish Intercollegiate Guidelines Network (SIGN) checklist for RCTs. 125 Each methodological question was answered with one of the following:
-
well covered
-
adequately addressed
-
poorly addressed
-
not reported (mentioned, but insufficient detail to allow assessment to be made)
-
not addressed (not mentioned or indicated this aspect of study design was ignored)
-
not applicable.
Results are shown in Table 34.
Simpson 199636 | Hesse 199837 | Smith 200038 | Bakheit 200039 | Bhakta 200040 | Bakheit 200141 | Brashear 200242 | Brashear 200443 | |
---|---|---|---|---|---|---|---|---|
Appropriate and clearly focused question | Well covered | Well covered | Well covered | Well covered | Well covered | Well covered | Well covered | Well covered |
Randomisation method | Not reported | Not reported | Not reported | Not reported | Well covered | Well covered | Not reported | Well covered |
Adequate concealment method | Not addressed | Not addressed | Not addressed | Not addressed | Well covered | Well covered | Not addressed | Well covered |
Blinded subjects and investigators | Adequately addressed | Adequately addressed | Adequately addressed | Adequately addressed | Well covered | Well covered | Adequately addressed | Well covered |
Groups are similar at baseline | Adequately addressed | Adequately addressed | Well covered | Adequately addressed | Adequately addressed | Well covered | Adequately addressed | Adequately addressed |
Only difference between groups is the intervention | Poorly addressed | Poorly addressed | Poorly addressed | Poorly addressed | Poorly addressed | Not addressed | Not addressed | Poorly addressed |
Relevant outcomes measured in a valid and reliable way | Poorly addressed | Poorly addressed | Poorly addressed | Poorly addressed | Poorly addressed | Poorly addressed | Adequately addressed | Poorly addressed |
% recruited dropped out before study completed | 5% | Not addressed | Not addressed | 1% | 7% | 1% | 3% | 6% |
Intention-to-treat analysis | Not addressed | Not addressed | Not addressed | Well covered | Not addressed | Well covered | Not addressed | Not addressed |
Results comparable for all sites | Not addressed | Not applicable | Not addressed | Not addressed | Not applicable | Not addressed | Not addressed | Not applicable |
How well does study minimise bias | – | – | – | – | + | + | – | – |
Does methodology mean overall effect due to the intervention? | Possibly | Possibly | Possibly | Possibly | Probably | Probably | Possibly | Possibly |
Overview of randomised controlled trials
An overview of each trial is shown in Table 35.
Author and year | Purpose of study | Eligibility | Exclusions | Number of participants | Intervention/Control | Outcome measures | Follow-up | Statistically significant results |
---|---|---|---|---|---|---|---|---|
Simpson 199636 | To evaluate effectiveness of botulinum toxin in treating upper extremity spasticity in chronic stroke patients |
At least 9 months post stroke Mean 2.5 on AS at elbow and wrist with minimum 2 at either joint Stable clinical course for 2 months before study Willing to maintain ongoing spasticity treatment |
Fixed contracture Previous treatment with botulinum Neurolytic or surgical procedures in the study limb Neuromuscular disease |
39 |
10 placebo 9, 75 u Botox 9, 150 u Botox 9, 300 u Botox (only give numbers analysed) All had biceps, FCU, FCR |
AS, FIM, Rand 36-Item Health Survey, Fugl–Meyer Scale, caregiver dependency, function and pain assessment, motor task/function rating scale, grip strength, Global Assessment of Spasticity Scale | 2, 4, 6, 10, 16 weeks |
Decreased AS score at elbow and wrist at weeks 2, 4, 6 in high-dose group compared with placebo Decreased AS at wrist at week 6 in low-dose group compared with placebo Improvement on physicians and patients global assessment in the high- and low-dose groups at weeks 4 and 6 Increased grip strength in low-dose group at weeks 6 and 16 |
Hesse 199837 | To investigate whether the combined approach of botulinum toxin and electrical stimulation was more effective than botulinum toxin alone in the treatment of chronic upper limb spasticity after stroke |
6–12 months post stroke MAS at least 3 at elbow, wrist and fingers No function of study arm |
Fixed contracture Previous treatment with botulinum Neurolytic or surgical procedures in the study limb Severe impairments of cognition and communication |
24 |
6 placebo 6 placebo + ES 6, 1000 u Dysport 6, 1000 u Dysport + ES All had biceps, brachioradialis, FCU, FCR, FDP, FDS |
MAS, limb position at rest, 3 functional activities, pain | 2, 6, 12 weeks | Cleaning the palm better in BTX + ES group compared with BTX alone or placebo alone at all time points |
Smith 200038 | To assess dose–response relationships to a single dose of botulinum toxin in upper limb spasticity associated with stroke or head injury |
At least 1 year post stroke or head injury Significant disabling spasticity Non function or partial function of study arm |
Fixed contracture | 21 |
6 placebo 6, 500 u Dysport 7, 1000 u Dysport 6, 1500 u Dysport (25 in total as four people re randomised at end of follow-up period) Muscles injected according to spasticity |
MAS, passive and active range of movement, time to dress upper body, FAT, finger curl, global clinical assessment score, gait (if mobile) | 2, 6, 12 weeks |
Decreased wrist and finger MAS score, increased passive range of movement and greater number of patients improved on global assessment at week 6 when all botulinum toxin groups combined and compared to placebo Individual groups: 500 u group MAS score decreased compared to placebo at week 6. 1500 u group passive range of movement better at weeks 6 and 12 |
Bakheit 200039 | To define an effective and safe dose of Dysport for the treatment of upper limb spasticity in stroke patients |
At least 3 months post stroke MAS 2 or more at elbow, wrist, fingers |
Muscle contracture Previous treatment with botulinum, phenol or alcohol nerve blocks |
83 |
20 placebo 22, 500 u Dysport 22, 1000 u Dysport 19, 1500 u Dysport All had biceps, FDS, FDP, FCU, FCR |
MAS, range of movement, pain, Rivermead Motor Assessment, Barthel ADL index, three functional activities | 2, 4, 8, 12, 16 weeks |
Decreased MAS score at any joint in all BTX groups compared with placebo at week 4 Decreased MAS score over 16 weeks for elbow and wrist in all BTX groups and fingers in 1000 u group |
Bhakta 200040 | To investigate whether reduction in spasticity after botulinum toxin treatment translates into reduction in disability and carer burden |
At least 6 months post stroke Elbow or finger MAS > 2 Non-function of study arm Moderate difficulty with two out of eight items on the disability scale |
Functionally useful arm Previous botulinum toxin or phenol treatment |
40 |
20 placebo 20, 1000 u Dysport Muscles injected according to spasticity |
Patient disability scale, carer burden scale, MAS, range of movement, pain, muscle power, grip strength | 2, 6, 12 weeks |
Improved patient disability scale score in BTX group at 2 and 6 weeks compared with placebo. Improved carer burden out to 12 weeks Decreased MAS score at fingers at all time points in BTX group compared with placebo and decreased MAS at elbow at week 2 Decreased grip strength in BTX group compared with placebo at week 6 |
Bakheit 200141 | To study the efficacy and safety of botulinum toxin in the treatment of upper limb spasticity caused by stroke |
At least 3 months post stroke MAS 2 or more in at least two of elbow, wrist, fingers and 1+ in remaining area |
Muscle contracture Previous treatment with alcohol or phenol nerve blocks Treatment with BTX in previous 6 months |
59 |
32 placebo 27, 1000 u Dysport All had biceps, FDS, FDP, FCU, FCR |
MAS, range of movement, pain, Barthel ADL index, three functional activities, goal attainment and subjective global assessment | 4, 8, 12, 16 weeks |
Decreased MAS score at any joint in BTX group compared with placebo at week 4. Decreased MAS score over 16 weeks for fingers and wrist in BTX group compared with placebo Improved passive range of movement at the elbow over 16 weeks in BTX group compared with placebo Numbers improving on global assessment greater in BTX group at all time points compared with placebo at week 16 |
Brashear 200242 | Assess the effects of one set of injections of botulinum toxin on muscle tone and measures of disability with respect to self-care, limb position, pain |
At least 6 months post stroke Wrist AS 3 or more, fingers AS 2 or more 2 or 3 on disability assessment scale |
Fixed contracture Profound muscle atrophy Previous or planned treatment of limb with botulinum, alcohol, phenol, surgery Intrathecal baclofen Change in medication for spasticity in previous 3 months Pregnancy |
126 |
62 placebo 64, 200–200 u Botox All had FCU, FCR, FDP, FDS and some FPL, AP |
AS, disability assessment scale, global assessment scale | 1, 4, 6, 8, 12 weeks |
Disability assessment scale principle target score improved in BTX group than placebo at all time points Greater number of patients improved by at least one point on the disability scale in BTX group compared with placebo week 6 Decreased AS score in BTX group compared with placebo at all time points Greater number of patients improving by at least one point on global assessment in BTX group at all time points |
Brashear 200443 | To determine whether botulinum toxin type B is effective in controlling upper limb spasticity |
At least 6 months post stroke AS 2 or more at elbow, wrist and fingers |
Unstable medical illness Previous treatment with botulinum, phenol or surgery to the affected limb |
15 |
5 placebo 10 botulinum toxin type B All had biceps, FCU, FCR, FDS, FDP |
AS, physician, patient and therapist global assessment of change, range of movement, nine-hole peg test, Jebsen test of hand function | 2, 4, 8, 12, 16 weeks (+ 12 weeks further open label) |
Decreased AS score at wrist at week 2 compared with placebo All MAS and global scores improved in the open label f/u |
Childers 200444 | To test the hypothesis that intramuscular botulinum toxin reduces excessive muscle tone in a dose-dependent manner in the elbow, wrist and fingers after stroke |
At least 6 weeks post stroke Wrist AS 3 or more, elbow AS 2 or more |
Fixed contracture Profound muscle atrophy of affected limb Previous treatment with botulinum, phenol or surgery Plaster casting for spasticity FEV1 < 65% predicted Diagnosis myasthenia gravis or other condition that could interfere with study Sensitivity to components of botulinum Pregnancy |
91 |
26 placebo 21, 90 u Botox 23, 180 u Botox 21, 360 u Botox All had biceps, FCU, FCR, FDP, FDS second injection given at > 12 weeks if AS 2 or more at wrist and/or elbow same dose given as first injection |
Trial’s own MAS, pain score and functional disability measure. SF-36, FIM, physician and patients global assessment | 1, 2, 3, 4, 5, 6, 9, 12, 18, 24 weeks |
After first injection Decreased AS score at wrist in all BTX groups compared with placebo at most time points out to week 9 Decreased AS score at elbow in the BTX group compared with placebo out to week nine Global response score higher in 360 u and 180 u groups compared with placebo at some time points After second injection Number of participants to decrease AS score by a least one point higher in the 360 u group compared with placebo at weeks 18 and 24 |
Suputtitada 200545 | To define the lowest effective dose and safety of botulinum toxin in the treatment of adult patients with upper limb spasticity |
Any cause spasticity (but only stroke included) Upper limb spasticity Medically stable No cognitive deficits Not independent ADLs Had already had rehabilitation programme for 6 weeks |
Fixed contracture Complete plegia Previous phenol or surgery to affected muscles Hypersensitivity to BTX Myasthenia gravis or other disorders where BTX contraindicated Aminoglycoside use Pregnancy |
50 |
15 placebo 15, 350 u Dysport 15, 500 u Dysport 5, 1000 u Dysport All had biceps, FCU, FCR, FDP, FDS |
MAS, ARAT, Barthel ADL index, visual analogue pain scale | 2, 4, 8, 16, 24 weeks |
Mean MAS score decreased in all botulinum toxin groups at week 8 compared with placebo Improved VAS score compared with placebo at weeks 8 and 24 Improved ARAT score in 500 u group compared with placebo at weeks 8 and 24. Decreased ARAT score in 1000 u compared with placebo at weeks 8 and 24 Increased Barthel ADL Index in 350 and 500 u groups at weeks 8 and 24 |
Yelnik 200746 | To assess the beneficial effect of injection of botulinum toxin into the subscapularis muscle on shoulder pain |
Any time limit after stroke MAS at least 1+ at shoulder and elbow Shoulder external rotation > 30° |
Retraction of muscle at elbow, wrist or fingers Previous traumatic or neurological disease of the shoulder Previous BTX or alcohol to subscapularis Myasthenia gravis Pregnancy |
20 |
10 placebo 10, 500 u Dysport All had subscapularis |
10-point pain VAS, MAS, passive range of movement | 1,2, 4 weeks |
Decreased pain in BTX group compared with placebo at week 4 Improved lateral rotation compared with placebo at weeks 2 and 4 Decreased MAS at fingers compared with placebo at week 4 |
Marco 200747 | To determine the efficacy of botulinum toxin type A for the treatment of spastic shoulder pain in patients after stroke |
> 18 years old > 3 months post stroke MAS at least 3 but limb area/s unclear Pain VAS at least 40 mm |
Mild hemiparesis Concomitant shoulder pathology Pacemaker Peripheral nervous system disease Hypersensitivity to botulinum toxin Pregnancy |
31 |
15 placebo 16, 500 u Dysport All had pectoralis major All had TENS machine |
100-point pain VAS, MAS, passive range of movement | 1, 3, 12, 24 weeks |
Decreased pain compared with placebo at all time points Improved external rotation compared with placebo at all time points |
Kong 200748 | To assess the effects of botulinum toxin type A on hemiplegic shoulder pain associated with spasticity |
Aged 18–80 > 3 months post stroke MAS at least 2 at shoulder and elbow Pain VAS at least 4 |
Shoulder pain or surgery before stroke Aphasia/cognitive impairment Reflex sympathetic dystrophy Post-stroke central pain Neuromuscular disease Previous treatment BTX |
17 |
9 placebo 8, 500 u Dysport All had pectoralis major and biceps |
10-point pain VAS, MAS, passive range of movement | 4, 8, 12 weeks | Decreased MAS score at shoulder and elbow compared with placebo at week 4 |
Jahangir 200749 | To assess the effectiveness, safety and impact of botulinum toxin on activities of daily living and quality of life in post-stroke hand spasticity in Malaysian patients |
> 21 years old At least 1 year post stroke MAS at least 2 at wrist and fingers |
Fixed contracture Profound muscle atrophy Prior BTX or nerve blocks Treatment agents affecting neuromuscular transmission Neuromuscular disease Injection site infection Systemic infection Pregnancy or lactation |
52 |
25 placebo 27, 80 u Botox All had FCR, FCU, FDS, FDP |
MAS, Barthel ADL Index, Euroqol EQ-5D | 1, 12 weeks | Decreased MAS score at wrist and fingers compared with placebo at 1 and 12 weeks |
de Boer 200850 | To study the effect of botulinum toxin type A in the subscapular muscle on shoulder pain and humerus rotation |
> 18 years old Any time after stroke MAS at least 1 at elbow Pain VAS at least 40 50% restricted external rotation relative to unaffected arm |
Inability to complete VAS INR > 3 Glenohumeral infiltration in last 4 weeks Shoulder pathology |
21 |
11 placebo 10, 50 u Botox All had subscapular muscle |
100-point pain VAS, passive range of movement | 6, 12 weeks | Nil |
McCrory 200951 | To examine the effect of botulinum toxin type A on quality of life and person-centred outcomes in patients with upper limb spasticity following stroke |
> 18 years old > 6 months post stroke MAS at least 2 in two out of three of elbow, wrist, fingers and 1+ remaining area Cognitive and communicative ability to give written consent |
Severe contracture Other neurological impairment Concurrent treatment with aminoglycoside antibiotics Botulinum toxin within last 4 months Previous with phenol or intrathecal baclofen for arm spasticity |
96 |
42 placebo 54, 750–1000 u Dysport Muscles injected according to spasticity pattern Second injections given at week 12, muscles and dosages according to response to first injection |
Assessment of quality of life measure 100-point pain VAS, Hospital Anxiety and Depression rating scale, goal attainment, MAS, Modified Motor Assessment Scale, patient disability and carer-burden scale (with a principal target of treatment), global assessment of benefit |
8, 20 weeks (global benefit only 12, 24 weeks) |
Decreased MAS score across all joints at weeks 8 and 20 in BTX group compared with placebo Higher proportion of participants with improved score for principle target of treatment at weeks 8 and 20. Goal attainment score higher at week 20 Higher proportion of participants with global benefit at week 12, 24 |
Summary of systematic reviews
van Kuijk et al. 200252
This systematic review was about the use of any focal treatment for upper limb spasticity after stroke. Four RCTs evaluating the effect of botulinum toxin injections were included. 36–39 No meta-analysis was performed. The authors concluded that there was evidence to support a decrease in muscle tone and increase in passive range of movement following botulinum toxin treatment.
Cardoso et al. 200553
This systematic review included five RCTs evaluating the effect of botulinum toxin as a treatment for post-stroke upper limb spasticity. 36,38,39,41,42 Modified Ashworth Scale scores and patient/physician global assessment scale scores were combined as possible for meta-analysis. The authors concluded that there was evidence to support improvement in both muscle tone and global response following treatment.
Garces et al. 200554
This systematic review was about the use of botulinum toxin in both upper and lower limb spasticity in various conditions. Six RCTs36,37,39–42 and three published abstracts regarding botulinum toxin for upper limb spasticity after stroke were included. Muscle tone scores and range of movement evaluations were combined where possible for meta-analysis. Effects on function/disability (including patient and carer disability scales, global assessment scales) and pain were also reviewed, but data could not be combined. The meta-analysis showed that muscle tone was reduced after treatment, but there was no improvement in range of movement. There was no evidence to support pain decrease after treatment but trials had shown function/disability was improved after treatment.
Rosales and Chua-Yap 200855
This systematic review examined botulinum toxin for the upper and lower limb after stroke. Seven upper limb studies were included. 36–39,41,42,44 Modified Ashworth Scale scores and global assessment scale scores from both upper and lower limb studies were extracted for a combined meta-analysis. Both muscle tone and global assessment were improved after botulinum toxin treatment. Other outcome measures were not analysed.
Elia et al. 200956
The effect of botulinum toxin on muscle tone, global ratings, functional disability, pain, quality of life and adverse events in both upper and lower limb spasticity following stroke were evaluated in this systematic review. Ten upper limb RCTs were included. 36–45 Muscle tone scores and global rating scores were combined for meta-analysis and improvements were demonstrated after botulinum toxin treatment. The remaining outcome measures could not be combined. The authors concluded that botulinum toxin can reduce upper limb spasticity, but effects on functional ability are unclear.
Overview of uncontrolled studies of repeated botulinum toxin injections
An overview of each study is shown in Table 36.
Author and year | Purpose of study | Eligibility | Exclusions | Number of subjects | Treatment | Outcome measures | Follow up | Statistically significant results |
---|---|---|---|---|---|---|---|---|
Rodriquez 200057 | Assess the outcomes of botulinum toxin injection of spastic finger flexors followed by intensive training of finger extensors |
Chronic hemiplegia (all were stroke) Clinical spasticity of long fingers flexors Trace finger extension when wrist flexed Full passive range of movement of finger flexors Adequate cognition for home programme No lower motor neurone disease |
None given | 14 |
50 mouse u into FDS and FDP. Preparation of botulinum not detailed No criteria given for repeat injections Six participants given second injection at mean 5.7 months after first injection Two participants given third injection at mean 5.5 months after second |
MAS Grip strength Clonus score Finger extension (by video) |
Mean 3.5 days after first injection. Mean 1.7 days after second injection Mean 1 day after third |
Improved MAS, clonus and video of finger extension after first injections No significant differences after second injections |
Lagalla 200058 | Evaluate long-term ability of repeated botulinum treatment of upper limb spasticity after stroke to accomplish technical, functional and caregiver objectives |
Spasticity after stroke present for at least 6 months Lack of efficiency of drug treatment |
Fixed contracture Previous treatment with alcoholic neurolytic agents Concurrent treatment with antispastic drugs Severe concomitant disease |
34 |
50–300 u Botox depending on size of muscles and severity of spasticity. Muscles injected according to spasticity pattern Repeat injections given when tone and ROM returned to baseline |
MAS ROM FAT Pt/carer goals assessment |
4 weeks following injection Minimum six injection cycles |
Improved MAS at elbow, wrist and fingers compared with baseline after each injection Improved ROM at each joint after each injection compared with baseline No difference between scores across injection cycles |
Gordon 200459 | Evaluate long-term efficacy and safety of botulinum in post-stroke spasticity (follow-up of patients involved in Brashear et al. 2002 study42) |
At least 6 months post stroke Wrist AS 3 or more, fingers AS 2 or more 2 or 3 on disability assessment scale |
Fixed contracture Profound muscle atrophy Previous or planned treatment of limb with botulinum, alcohol, phenol, surgery Intrathecal baclofen Change in medication for spasticity in previous 3 months Pregnancy |
111 |
200–240 u Botox distributed between FCU, FCR, FDP, FDS in all participants with some having additional FPL and AP Repeat injections given once 12 weeks had passed and wrist AS 2 or more, finger AS 1 or more. Same muscles injected and doses used |
Disability assessment scale AS, botulinum antibody level. |
Every 6 weeks for 42 weeks Up to 4 injection cycles |
Improved DAS and AS scores compared with baseline after each injection cycle No differences in scores across the cycles |
Bakheit 200460 | To study the efficacy, safety and incidence of botulinum antibody formation with repeated treatments with botulinum in post-stroke upper limb spasticity |
At least 3 months post stroke MAS 2 or more in at least 2 of elbow, wrist, fingers and 1+ in remaining area 10 or more on disability and carer burden rating scale |
Muscle contracture Previous treatment with alcohol or phenol nerve blocks Treatment with BTX in previous 3 months Treatment with intrathecal baclofen |
51 |
1000 u Dysport distributed between biceps, FCR, FCU, FDS and FDP for initial injections Repeat injections at week 12 if MAS back to baseline, otherwise at week 16. Same muscles injected but dose at discretion of clinician |
MAS ROM Pain Disability and carer burden rating scale Global assessment Goal attainment scale Antibody level |
4 weeks after each injection 3 injection cycles |
Decrease of at least one point on the MAS across at least one joint achieved by 100%, 98% and 98% of participants following first, second and third injections, respectively Improvements on disability and carer scale after each injection Improvements in ROM after each injection Approx 50% of patients achieved their goals after each injection 90% of patients reported global benefit |
Cardoso 200761 | To establish whether individualised botulinum toxin type A injections improve upper limb function in post-stroke patients |
> 18 years old Between 6 months and 5 years post stroke Spastic hemiparesis |
Fibrotic retraction of affected muscles Liver, blood or kidney disease Aphasia or dementia Pregnancy or lactation |
20 |
Dysport dosed and delivered according to spasticity pattern Repeat injections at week 16. No criteria given |
Fugl–Meyer Scale MAS FIM Passive range of movement |
16, 32 weeks |
Improved MAS scores at both 16 and 32 weeks Improved ROM at both 16 and 32 weeks Arm sections of Fugl–Meyer and FIM improved at both 16 and 32 weeks |
Elovic 200862 | To assess the safety and effects of repeated administration of botulinum toxin type A in patients with post stroke spasticity |
> 21 years old > 6 months post stroke ‘Need’ for a minimum of 200 u Botox into wrist or finger flexors |
Fixed contracture Profound atrophy affected arm Neuromuscular disorders Infection/dermatological condition at injection site Inflammation of study limb limiting movement Past/planned treatment with phenol, alcohol, surgery, intrathecal baclofen, casting Hypersensitivity to botulinum toxin |
279 |
200–400 u Botox distributed at discretion of study investigators Repeated treatments at minimum of 12-weekly intervals if ‘need’ for at least 200 u Botox into wrist or finger muscles |
DAS MAS Stroke-adapted sickness impact profile, EQ-5D VAS Botulinum toxin antibody level |
Every 6 weeks for 54 weeks |
Improved DAS ‘principal target’ scores from baseline at all time points Improved MAS scores at the wrist, fingers and thumb from baseline at all time points Improved MAS scores at the elbow in participants receiving at least 250 u at all time points Improved scores on the stroke-adapted sickness impact profile and EQ-5D at all time points from baseline |
Appendix 2 List of case record forms
The following case record forms were used in the study:
-
patient information sheet
-
consent form 1 (potentially interested in study, contact details can be given to study team)
-
consent form 2 (consent to take part in study)
-
screening visit
-
baseline visit
-
randomisation (included initial botulinum toxin injection details for intervention group)
-
therapy manual and data collection forms menu 1
-
therapy manual and data collection forms menu 2
-
Canadian Occupational Performance Measure
-
1-month outcome visit
-
1-month questionnaire
-
3-month outcome visit
-
3-month questionnaire
-
12-month outcome visit
-
12-month questionnaire
-
clinical review form (review regarding further therapy/botulinum toxin at 3, 6, 9, 12 months)
-
end of study form
-
withdrawal of consent form
-
adverse event form
-
serious adverse event form.
Copies of case record forms are available from Professor Helen Rodgers: e-mail: Helen.Rodgers@ncl.ac.uk
Appendix 3 Outcome scale scores at follow-up
Impairment
Control | Intervention | p-value | |
---|---|---|---|
Modified Ashworth Score at elbow: n (%) | |||
0 | 7 (4.5) (n = 154) | 11 (6.6) (n = 167) | < 0.001 |
1 | 25 (16.2) (n = 154) | 59 (35.3) (n = 167) | |
1+ | 48 (31.2) (n = 154) | 35 (21.0) (n = 167) | |
2 | 40 (26.0) (n = 154) | 47 (28.1) (n = 167) | |
3 | 33 (21.4) (n = 154) | 15 (9.0) (n = 167) | |
4 | 1 (0.6) (n = 154) | 0 (0.0) (n = 167) | |
Median (IQR) | 1+ (1+ to 2) | 1+ (1 to 2) | |
Grip strength: median (IQR) | 2.0 (0.0 to 6.5) (n = 154) | 1.3 (0.0 to 5.3) (n = 167) | 0.092 |
Motricity Index: median (IQR) | |||
Arm | 46.5 (29 to 66) (n = 154) | 40 (26 to 66) (n = 167) | 0.790 |
Leg | 54 (43 to 76) (n = 154) | 65 (43 to 76) (n = 167) | 0.410 |
Total | 49 (36.3 to 66.3) (n = 153) | 52.5 (33.5 to 71) (n = 167) | 0.751 |
Control | Intervention | Difference | |
---|---|---|---|
Modified Ashworth Score at elbow: mean (95% CI) a | 2.5 (2.3 to 2.6) | 2.0 (1.8 to 2.1) | – 0.5 (– 0.7 to – 0.2) |
Grip strength (kg): mean (95% CI) | 4.7 (3.7 to 5.8) | 4.0 (3.1 to 5.0) | – 0.7 (– 2.1 to 0.7) |
Motricity Index: mean (95% CI) | |||
Arm | 44.4 (40.4 to 48.4) | 43.9 (39.9 to 47.8) | – 0.6 (– 6.2 to 5.1) |
Total | 50.5 (47.2 to 53.8) | 51.1 (47.7 to 54.5) | 0.6 (– 4.2 to 5.4) |
Control | Intervention | p-value | |
---|---|---|---|
Modified Ashworth Score at elbow: n (%) | |||
0 | 6 (4.0) (n = 151) | 9 (5.5) (n = 163) | 0.145 |
1 | 30 (19.9) (n = 151) | 37 (22.7) (n = 163) | |
1+ | 37 (24.5) (n = 151) | 47 (28.8) (n = 163) | |
2 | 47 (31.1) (n = 151) | 44 (27.0) (n = 163) | |
3 | 30 (19.9) (n = 151) | 23 (14.1) (n = 163) | |
4 | 1 (0.7) (n = 151) | 3 (1.8) (n = 163) | |
Median (IQR) | 2 (1+ to 2) | 1+ (1 to 2) | |
Grip strength: median (IQR) | 2.0 (0.0 to 7.0) (n = 151) | 2.0 (0.0 to 6.7) (n = 163) | 0.874 |
Motricity Index: median (IQR) | |||
Arm | 40 (29 to 67) (n = 151) | 40 (29 to 66) (n = 163) | 0.862 |
Leg | 54 (43 to 76) (n = 150) | 65 (43 to 76) (n = 162) | 0.093 |
Total | 47 (35.9 to 70.1) (n = 150) | 54.3 (36 to 69.5) (n = 162) | 0.256 |
Control | Intervention | Difference | |
---|---|---|---|
Modified Ashworth Score at elbow: mean (95% CI) a | 2.5 (2.3 to 2.6) | 2.3 (2.1 to 2.4) | – 0.2 (– 0.4 to – 0.1) |
Grip strength (kg): mean (95% CI) | 5.0 (3.9 to 6.1) | 5.4 (4.2 to 6.6) | 0.4 (– 1.2 to 2.0) |
Motricity Index: mean (95% CI) | |||
Arm | 43.8 (39.7 to 48.0) | 45.7 (41.6 to 49.7) | 1.9 (– 4.0 to 7.7) |
Total | 49.5 (46.0 to 52.9) | 53.2 (50.0 to 56.6) | 3.8 (– 1.0 to 8.5) |
Control | Intervention | p-value | |
---|---|---|---|
Modified Ashworth Score at elbow: n (%) | |||
0 | 6 (6.6) (n = 91) | 6 (6.2) (n = 97) | 0.508 |
1 | 25 (27.5) (n = 91) | 21 (21.6) (n = 97) | |
1+ | 13 (14.3) (n = 91) | 22 (22.7) (n = 97) | |
2 | 22 (24.2) (n = 91) | 34 (35.1) (n = 97) | |
3 | 21 (23.1) (n = 91) | 14 (14.4) (n = 97) | |
4 | 4 (4.4) (n = 91) | 0 (0.0) (n = 97) | |
Median (IQR) | 2 (1–3) | 1+ (1–2) | |
Grip strength: median (IQR) | 3.0 (0.0 to 8.9) (n = 92) | 2.0 (0.0 to 9.8) (n = 97) | 0.484 |
Motricity Index: median (IQR) | |||
Arm | 44 (29 to 66) (n = 92) | 48 (29 to 66.5) (n = 97) | 0.737 |
Leg | 54 (38.3 to 76) (n = 92) | 62 (46.5 to 76) (n = 97) | 0.488 |
Total | 49.5 (36.1 to 70.3) (n = 92) | 54.5 (38.5 to 69.8) (n = 97) | 0.535 |
Control | Intervention | Difference | |
---|---|---|---|
Modified Ashworth Score at elbow: mean (95% CI) a | 2.4 (2.1 to 2.7) | 2.3 (2.1 to 2.5) | – 0.1 (– 0.5 to 0.3) |
Grip strength (kg): mean (95% CI) | 5.8 (4.4 to 7.3) | 5.4 (4.0 to 6.9) | – 0.4 (– 2.4 to 1.6) |
Motricity Index: mean (95% CI) | |||
Arm | 45.5(40.6 to 50.6) | 47.1 (42.1 to 52.2) | 1.6 (– 5.7 to 8.7) |
Total | 51.5 (47.1 to 56.0) | 53.0 (48.8 to 57.0) | 1.5 (– 4.7 to 7.4) |
Upper limb function and activity limitation
Control | Intervention | p-value | |
---|---|---|---|
ARAT: median (IQR) | |||
Total | 4 (3 to 18) (n = 154) | 3 (3 to 16) (n = 167) | 0.754 |
Grasp | 0 (0 to 6) (n = 154) | 0 (0 to 5) (n = 167) | 0.979 |
Grip | 0 (0 to 4.3) (n = 154) | 0 (0 to 4) (n = 167) | 0.965 |
Pinch | 0 (0 to 1) (n = 154) | 0 (0 to 2) (n = 167) | 0.401 |
Gross | 3 (3 to 5) (n = 154) | 3 (3 to 5) (n = 167) | 0.485 |
Nine-Hole Peg Test (pegs placed in 50 seconds): median (IQR) | 0 (0 to 0) (n = 155) | 0 (0 to 0) (n = 166) | 0.979 |
Upper limb functional activities: median (IQR) | |||
Put arm through sleeve | 3 (2 to 4) (n = 138) | 3 (2 to 4) (n = 152) | 0.255 |
Open the hand for cleaning your palm | 3 (2 to 4) (n = 137) | 3 (2 to 4) (n = 151) | 0.538 |
Open the hand for cutting fingernails | 2 (1 to 4) (n = 138) | 3 (1 to 4) (n = 151) | 0.480 |
Use cutlery | 1 (1 to 2) (n = 135) | 1 (1 to 2) (n = 149) | 0.746 |
Barthel ADL Index: median (IQR) | 14 (10 to 17) (n = 134) | 14.5 (9 to 17) (n = 142) | 0.873 |
Barthel ADL Index groups: n (%) | |||
0–4 | 9 (6.7) (n = 134) | 10 (7) (n = 142) | 0.265 |
5–9 | 21 (15.7) (n = 134) | 27 (19) (n = 142) | |
10–14 | 47 (35.1) (n = 134) | 34 (23.9) (n = 142) | |
15–19 | 48 (35.8) (n = 134) | 64 (45.1) (n = 142) | |
20 | 9 (6.7) (n = 134) | 7 (4.9) (n = 142) |
Control | Intervention | Difference | |
---|---|---|---|
ARAT: mean (95% CI) | 11.9 (9.5 to 14.3) | 11.5 (9.4 to 13.8) | – 0.3 (– 3.6 to 2.9) |
Nine-Hole Peg Test (pegs placed in 50s): mean (95% CI) | 1.2 (0.8 to 1.6) | 1.1 (0.7 to 1.5) | – 0.1 (– 0.6 to 0.5) |
Upper limb functional activities: mean (95% CI) | |||
Put arm through sleeve | 3.0 (2.8 to 3.2) | 3.1 (3.0 to 3.3) | 0.1 (– 0.1 to 0.4) |
Open the hand for cleaning your palm | 3.2 (3.0 to 3.4) | 3.3 (3.1 to 3.5) | 0.1 (– 0.2 to 0.4) |
Open the hand for cutting fingernails | 2.5 (2.3 to 2.8) | 2.6 (2.3 to 2.8) | 0.0 (– 0.3 to 0.4) |
Use cutlery | 1.5 (1.3 to 1.7) | 1.6 (1.4 to 1.7) | 0.0 (– 0.2 to 0.3) |
Barthel ADL Index: mean (95% CI) | 13.2 (12.3 to 14.0) | 13.2 (12.4 to 14.0) | 0.0 (– 1.1 to 1.2) |
Control | Intervention | p-value | |
---|---|---|---|
ARAT: median (IQR) | |||
Total | 3 (3 to 18) (n = 151) | 4 (3 to 18.5) (n = 161) | 0.759 |
Grasp | 0 (0 to 6) (n = 151) | 0 (0 to 6) (n = 161) | 0.427 |
Grip | 0 (0 to 4) (n = 151) | 0 (0 to 5) (n = 161) | 0.228 |
Pinch | 0 (0 to 2) (n = 151) | 0 (0 to 1.5) (n = 161) | 0.663 |
Gross | 3 (3 to 5) (n = 151) | 3 (3 to 5) (n = 161) | 0.704 |
Nine-Hole Peg Test (pegs placed in 50 seconds): median (IQR) | 0 (0 to 0) (n = 151) | 0 (0 to 0) (n = 162) | 0.529 |
Upper limb functional activities: median (IQR) | |||
Put arm through sleeve | 3 (2 to 4) (n = 134) | 3 (2 to 4) (n = 151) | 0.577 |
Open the hand for cleaning your palm | 3 (2 to 4) (n = 134) | 3 (2 to 4) (n = 151) | 0.413 |
Open the hand for cutting fingernails | 2 (1 to 3) (n = 134) | 2 (1 to 4) (n = 151) | 0.620 |
Use cutlery | 1 (1 to 2) (n = 132) | 1 (1 to 2) (n = 151) | 0.692 |
Barthel ADL Index: median (IQR) | 14.5 (10 to 17) (n = 130) | 15 (11 to 17) (n = 143) | 0.967 |
Barthel ADL Index groups: n (%) | |||
0–4 | 6 (4.6) (n = 130) | 8 (5.6) (n = 143) | 0.980 |
5–9 | 24 (18.5) (n = 130) | 23 (16.1) (n = 143) | |
10–14 | 35 (26.9) (n = 130) | 40 (28.0) (n = 143) | |
15–19 | 55 (42.3) (n = 130) | 62 (43.4) (n = 143) | |
20 | 10 (7.7) (n = 130) | 10 (7) (n = 143) |
Control | Intervention | Difference | |
---|---|---|---|
ARAT: mean (95% CI) | 11.4 (9.2 to 13.7) | 12.5 (10.2 to 15.2) | 1.2 (– 2.2 to 4.7) |
Nine-Hole Peg Test (pegs placed in 50s): mean (95% CI) | 1.1 (0.7 to 1.5) | 1.4 (1.0 to 1.9) | 0.3 (– 0.3 to 0.9) |
Upper limb functional activities: mean (95% CI) | |||
Put arm through sleeve | 3.1 (2.9 to 3.3) | 3.1 (2.9 to 3.3) | 0.1 (– 0.2 to 0.3) |
Open the hand for cleaning your palm | 3.0 (2.8 to 3.2) | 3.2 (3.0 to 3.4) | 0.2 (– 0.2 to 0.5) |
Open the hand for cutting fingernails | 2.3 (2.0 to 2.5) | 2.4 (2.1 to 2.6) | 0.1 (– 0.2 to 0.4) |
Use cutlery | 1.5 (1.4 to 1.7) | 1.5 (1.4 to 1.7) | 0.0 (– 0.2 to 0.2) |
Barthel ADL Index: mean (95% CI) | 13.4 (12.6 to 14.2) | 13.4 (12.6 to 14.2) | 0.0 (– 1.2 to 1.1) |
Control | Intervention | p-value | |
---|---|---|---|
ARAT: median (IQR) | |||
Total | 4 (3 to 20) (n = 92) | 4 (3 to 23) (n = 97) | 0.329 |
Grasp | 0 (0 to 6) (n = 92) | 1 (0 to 6) (n = 97) | 0.288 |
Grip | 0 (0 to 5) (n = 92) | 0 (0 to 6) (n = 97) | 0.416 |
Pinch | 0 (0 to 1.8) (n = 92) | 0 (0 to 3) (n = 97) | 0.319 |
Gross | 3 (3 to 5) (n = 92) | 3 (3 to 5) (n = 97) | 0.497 |
Nine-Hole Peg Test (pegs placed in 50 seconds): median (IQR) | 0 (0 to 0) (n = 92) | 0 (0 to 0) (n = 97) | 0.833 |
Upper limb functional activities: median (IQR) | |||
Put arm through sleeve | 3 (2 to 4) (n = 83) | 3 (2 to 4) (n = 87) | 0.303 |
Open the hand for cleaning your palm | 3 (2 to 4) (n = 83) | 3 (2 to 5) (n = 87) | 0.026 |
Open the hand for cutting fingernails | 2 (1 to 3) (n = 83) | 3 (1 to 4) (n = 87 | 0.036 |
Use cutlery | 1 (1 to 2) (n = 83) | 1 (1 to 2) (n = 87) | 0.124 |
Barthel ADL Index: median (IQR) | 14 (12 to 17) (n = 75) | 14 (9 to 17) (n = 82) | 0.767 |
Barthel ADL Index groups: n (%) | |||
0–4 | 3 (4.0) (n = 75) | 3 (3.7) (n = 82) | 0.372 |
5–9 | 9 (12.0) (n = 75) | 19 (23.2) (n = 82) | |
10–14 | 26 (34.7) (n = 75) | 21 (25.6) (n = 82) | |
15–19 | 33 (44.4) (n = 75) | 33 (40.2) (n = 82) | |
20 | 4 (5.3) (n = 75) | 6 (7.3) (n = 82) |
Control | Intervention | Difference | |
---|---|---|---|
ARAT: mean (95% CI) | 11.9 (9.0 to 15.1) | 13.6 (10.6 to 16.5) | 1.6 (– 2.7 to 6.1) |
Nine-Hole Peg Test (pegs placed in 50s): mean (95% CI) | 1.2 (0.7 to 1.8) | 1.3 (0.7 to 1.9) | 0.0 (– 0.7 to 0.8) |
Upper limb functional activities: mean (95% CI) | |||
Put arm through sleeve | 2.9 (2.6 to 3.1) | 3.1 (2.8 to 3.3) | 0.2 (– 0.2 to 0.5) |
Open the hand for cleaning your palm | 2.9 (2.6 to 3.2) | 3.4 (3.1 to 3.7) | 0.5 (0.1 to 0.9) |
Open the hand for cutting fingernails | 2.2 (1.9 to 2.5) | 2.7 (2.4 to 3.0) | 0.5 (0.1 to 0.9) |
Use cutlery | 1.4 (1.2 to 1.5) | 1.6 (1.4 to 1.8) | 0.3 (0.0 to 0.5) |
Barthel ADL Index: mean (95% CI) | 13.7 (12.8 to 14.7) | 13.4 (12.4 to 14.4) | – 0.3 (– 1.7 to 1.1) |
Stroke-related quality of life/participation restriction
Control | Intervention | p-value | |
---|---|---|---|
Stroke Impact Scale domains: median (IQR) | |||
Strength | 34.4 (18.8 to 50.0) (n = 138) | 31.3 (18.8 to 50.0) (n = 150) | 0.767 |
Memory | 75.0 (60.7 to 92.9) (n = 137) | 75.0 (64.3 to 92.9) (n = 152) | 0.981 |
Emotion | 66.7 (55.6 to 80.6) (n = 136) | 66.7 (55.6 to 75.0) (n = 151) | 0.413 |
Communication | 89.3 (64.3 to 100) (n = 138) | 85.7 (57.1 to 96.4) (n = 152) | 0.156 |
ADL | 42.5 (30.0 to 57.5) (n = 138) | 42.5 (30.0 to 57.5) (n = 151) | 0.876 |
Mobility | 50.0 (29.2 to 63.9) (n = 137) | 44.4 (27.8 to 63.9) (n = 151) | 0.618 |
Hand function | 0.0 (0.0 to 25.0) (n = 138) | 0.0 (0.0 to 20.0) (n = 151) | 0.740 |
Participation/Handicap | 37.5 (18.8 to 62.5) (n = 137) | 34.4 (17.9 to 57.1) (n = 151) | 0.452 |
Physical domain | 34.0 (24.1 to 45.6) (n = 138) | 32.4 (20.5 to 45.5) (n = 152) | 0.728 |
Stroke recovery | 40.0 (30.0 to 57.5) (n = 137) | 40.0 (25.0 to 50.0) (n = 151) | 0.585 |
EQ-5D: median (IQR) | |||
Mobility | 2 (2 to 2) (n = 138) | 2 (2 to 2) (n = 151) | 0.746 |
Self-care | 2 (2 to 2) (n = 138) | 2 (2 to 2) (n = 151) | 0.540 |
Usual activities | 3 (2 to 3) (n = 138) | 3 (2 to 3) (n = 149) | 0.440 |
Pain/discomfort | 2 (2 to 2) (n = 137) | 2 (1 to 2) (n = 150) | 0.181 |
Anxiety/depression | 2 (1 to 2) (n = 133) | 2 (1 to 2) (n = 149) | 0.377 |
Good/bad health scale | 50 (40 to 65) (n = 135) | 50 (40 to 70) (n = 150) | 0.401 |
Oxford Handicap Scale: median (IQR) | 3 (3 to 4) (n = 137) | 3 (3 to 4) (n = 152) | 0.169 |
Control | Intervention | Difference | |
---|---|---|---|
Stroke Impact Scale domains: mean (95% CI) | |||
Strength | 33.7 (30.4 to 37.0) | 33.7 (30.8 to 36.8) | 0.0 (– 4.4 to 4.5) |
Memory | 74.5 (70.5 to 78.5) | 74.5 (70.9 to 77.8) | – 0.1 (– 5.2 to 5.1) |
Emotion | 66.9 (63.9 to 69.7) | 65.2 (62.8 to 67.7) | – 1.7 (– 5.4 to 2.2) |
Communication | 78.2 (73.8 to 82.5) | 75.7 (71.4 to 79.8) | – 2.5 (– 8.5 to 3.6) |
ADL | 42.5 (39.1 to 45.9) | 43.3 (39.9 to 46.7) | 0.8 (– 3.9 to 5.5) |
Mobility | 48.0 (43.7 to 52.4) | 47.3 (43.3 to 51.4) | – 0.7 (– 6.6 to 5.4) |
Hand function | 12.7 (9.6 to 16.1) | 15.2 (11.7 to 19.0) | 2.5 (– 2.3 to 7.3) |
Participation/Handicap | 41.5 (36.7 to 46.3) | 38.3 (33.9 to 42.9) | – 3.1 (– 9.7 to 3.6) |
Physical domains | 34.0 (31.2 to 36.8) | 34.5 (31.7 to 37.3) | 0.5 (– 3.6 to 4.4) |
Stroke recovery | 43.1 (39.9 to 46.3) | 40.5 (37.4 to 43.7) | – 2.6 (– 7.1 to 1.9) |
EQ-5D: mean (95% CI) | |||
Mobility | 2.0 (2.0 to 2.1) | 2.1 (2.0 to 2.1) | 0.0 (– 0.1 to 0.1) |
Self-care | 2.1 (2.0 to 2.2) | 2.1 (2.0 to 2.2) | 0.0 (– 0.2 to 0.1) |
Usual activities | 2.5 (2.4 to 2.6) | 2.5 (2.4 to 2.6) | 0.0 (– 0.1 to 0.2) |
Pain/discomfort | 1.9 (1.8 to 2.0) | 1.8 (1.7 to 1.9) | – 0.1 (– 0.2 to 0.0) |
Anxiety/depression | 1.7 (1.6 to 1.8) | 1.6 (1.6 to 1.7) | – 0.1 (– 0.2 to 0.1) |
Good/bad health scale | 53.4 (50.1 to 56.8) | 55.0 (52.0 to 58.0) | 1.6 (– 2.9 to 6.2) |
Oxford Handicap Scale: mean (95% CI) | 3.4 (3.2 to 3.5) | 3.5 (3.3 to 3.7) | 0.1 (– 0.1 to 0.3) |
Control | Intervention | p-value | |
---|---|---|---|
Stroke Impact Scale domains: median (IQR) | |||
Strength | 31.3 (12.5 to 50.0) (n = 130) | 31.3 (18.8 to 43.8) (n = 151) | 0.966 |
Memory | 78.6 (59.8 to 96.4) (n = 133) | 78.6 (64.3 to 92.9) (n = 152) | 0.668 |
Emotion | 65.3 (55.6 to 77.8) (n = 132) | 63.9 (54.6 to 75.0) (n = 149) | 0.422 |
Communication | 85.7 (60.7 to 100.0) (n = 134) | 86.6 (66.1 to 100.0) (n = 150) | 0.468 |
ADL | 42.5 (30.0 to 57.5) (n = 134) | 40.0 (28.1 to 55.0) (n = 151) | 0.663 |
Mobility | 52.8 (30.6 to 70.4) (n = 133) | 47.2 (30.6 to 67.2) (n = 150) | 0.455 |
Hand function | 0.0 (0.0 to 15.0) (n = 134) | 0.0 (0.0 to 20.0) (n = 151) | 0.673 |
Participation/Handicap | 34.7 (15.6 to 62.5) (n = 134) | 39.1 (18.5 to 62.5) (n = 150) | 0.874 |
Physical domain | 34.6 (21.8 to 44.9) (n = 134) | 31.0 (20.7 to 44.6) (n = 152) | 0.683 |
Stroke recovery | 40.0 (30.0 to 60.0) (n = 133) | 40.0 (30.0 to 60.0) (n = 152) | 0.844 |
EQ-5D: median (IQR) | |||
Mobility | 2 (2 to 2) (n = 134) | 2 (2 to 2) (n = 151) | 0.525 |
Self-care | 2 (2 to 2) (n = 134) | 2 (2 to 2) (n = 152) | 0.325 |
Usual activities | 2 (2 to 3) (n = 134) | 3 (2 to 3) (n = 151) | 0.605 |
Pain/discomfort | 2 (2 to 2) (n = 133) | 2 (1 to 2) (n = 152) | 0.008 |
Anxiety/depression | 2 (1 to 2) (n = 132) | 2 (1 to 2) (n = 151) | 0.139 |
Good/bad health scale | 50 (40 to 70) (n = 133) | 50 (40 to 70) (n = 149) | 0.734 |
Oxford Handicap Scale: median (IQR) | 3 (3 to 4) (n = 133) | 3 (3 to 4) (n = 151) | 0.928 |
Control | Intervention | Difference | |
---|---|---|---|
Stroke Impact Scale domain: mean (95% CI) | |||
Strength | 31.2 (27.2 to 35.2) | 32.1 (28.7 to 35.4) | 0.9 (– 4.3 to 6.1) |
Memory | 73.6 (68.8 to 78.2) | 75.1 (71.6 to 78.6) | 1.5 (– 4.3 to 7.5) |
Emotion | 67.3 (64.5 to 70.2) | 63.9 (61.2 to 66.6) | – 3.4 (– 7.4 to 0.5) |
Communication | 76.4 (71.8 to 80.9) | 79.5 (75.5 to 83.2) | 3.0 (– 2.8 to 9.1) |
ADL | 43.0 (39.5 to 46.5) | 43.0 (39.8 to 46.2) | – 0.1 (– 4.8 to 4.8) |
Mobility | 50.4 (45.8 to 55.1) | 49.1 (45.0 to 53.2) | – 1.3 (– 7.6 to 5.0) |
Hand function | 12.2 (8.6 to 16.0) | 13.4 (10.1 to 16.9) | 1.3 (– 3.8 to 6.2) |
Participation/Handicap | 40.4 (35.7 to 45.1) | 40.8 (36.1 to 45.4) | 0.3 (– 6.3 to 7.0) |
Physical domains | 33.9 (30.7 to 37.0) | 34.0 (31.1 to 36.9) | 0.2 (– 4.1 to 4.4) |
Stroke recovery | 43.8 (40.2 to 47.5) | 43.5 (40.3 to 46.7) | – 0.3 (– 5.2 to 4.5) |
EQ-5D: mean (95% CI) | |||
Mobility | 2.0 (1.9 to 2.1) | 2.0 (2.0 to 2.1) | 0.0 (– 0.1 to 0.1) |
Self-care | 2.1 (2.0 to 2.2) | 2.1 (2.0 to 2.2) | – 0.1 (– 0.2 to 0.1) |
Usual activities | 2.5 (2.4 to 2.6) | 2.5 (2.4 to 2.6) | 0.0 (– 0.1 to 0.1) |
Pain/discomfort | 1.9 (1.8 to 2.0) | 1.7 (1.7 to 1.8) | – 0.2 (– 0.3 to 0.0) |
Anxiety/depression | 1.6 (1.5 to 1.7) | 1.7 (1.6 to 1.8) | 0.1 (0.0 to 0.2) |
Good/bad health scale | 53.8 (50.2 to 57.5) | 54.3 (51.2 to 57.4) | 0.5 (– 4.3 to 5.3) |
Oxford Handicap Scale: mean (95% CI) | 3.4 (3.3 to 3.6) | 3.4 (3.2 to 3.5) | – 0.1 (– 0.3 to 0.2) |
Control | Intervention | p-value | |
---|---|---|---|
Stroke Impact Scale domains: median (IQR) | |||
Strength | 25.0 (12.5 to 43.8) (n = 80) | 31.3 (12.5 to 43.8) (n = 86) | 0.675 |
Memory | 75.0 (53.6 to 92.9) (n = 83) | 82.1 (64.3 to 92.9) (n = 87) | 0.328 |
Emotion | 63.9 (52.8 to 75.7) (n = 82) | 66.7 (55.6 to 75.0) (n = 87) | 0.964 |
Communication | 85.7 (60.7 to 100.0) (n = 83) | 89.3 (67.9 to 96.4) (n = 87) | 0.754 |
ADL | 45.0 (30.0 to 55.6) (n = 83) | 41.3 (30.0 to 57.5) (n = 86) | 0.660 |
Mobility | 52.8 (30.6 to 66.7) (n = 83) | 47.2 (27.8 to 69.4) (n = 87) | 0.806 |
Hand function | 0.0 (0.0 to 10.0) (n = 83) | 0.0 (0.0 to 25.0) (n = 87) | 0.130 |
Participation/Handicap | 40.6 (18.8 to 61.6) (n = 81) | 39.1 (17.9 to 63.3) (n = 86) | 0.978 |
Physical domain | 30.7 (23.1 to 44.1) (n = 83) | 32.6 (19.5 to 47.2) (n = 87) | 0.648 |
Stroke recovery | 40.0 (22.5 to 55.0) (n = 81) | 40.0 (30.0 to 60.0) (n = 87) | 0.266 |
EQ-5D: median (IQR) | |||
Mobility | 2 (2 to 2) (n = 83) | 2 (2 to 2) (n = 87) | 0.381 |
Self-care | 2 (2 to 2) (n = 82) | 2 (2 to 2) (n = 87) | 0.589 |
Usual activities | 2 (2 to 3) (n = 83) | 2.5 (2 to 3) (n = 86) | 0.765 |
Pain/discomfort | 2 (2 to 2) (n = 83) | 2 (2 to 2) (n = 87) | 0.588 |
Anxiety/depression | 2 (1 to 2) (n = 81) | 2 (1 to 2) (n = 86) | 0.167 |
Good/bad health scale | 52.5 (40 to 70) (n = 80) | 60 (40 to 70) (n = 85) | 0.583 |
Oxford Handicap Scale: median (IQR) | 3 (3 to 4) (n = 83) | 3 (3 to 4) (n = 87) | 0.119 |
Control | Intervention | Difference | |
---|---|---|---|
Stroke Impact Scale domains: mean (95% CI) | |||
Strength | 29.7 (25.5 to 34.0) | 31.5 (27.3 to 35.9) | 1.9 (– 4.2 to 7.9) |
Memory | 71.1 (65.8 to 76.3) | 75.0 (70.3 to 79.6) | 3.9 (– 3.2 to 11.0) |
Emotion | 64.7 (60.9 to 68.4) | 63.7 (60.3 to 67.1) | – 0.9 (– 6.1 to 4.0) |
Communication | 77.9 (72.3 to 83.2) | 79.1 (73.9 to 84.1) | 1.2 (– 6.1 to 8.7) |
ADL | 41.8 (37.5 to 46.1) | 44.3 (40.3 to 48.3) | 2.5 (– 3.4 to 8.4) |
Mobility | 49.1 (43.9 to 54.2) | 48.1 (42.9 to 53.3) | – 1.1 (– 8.4 to 6.3) |
Hand function | 8.3 (5.1 to 11.9) | 15.1 (10.3 to 20.1) | 6.8 (0.8 to 12.7) |
Participation/Handicap | 41.4 (35.4 to 47.4) | 41.8 (35.6 to 48.0) | 0.4 (– 8.3 to 9.1) |
Physical domains | 31.9 (28.5 to 35.1) | 34.5 (30.8 to 38.4) | 2.7 (– 2.2 to 7.7) |
Stroke recovery | 40.6 (36.0 to 45.1) | 44.0 (39.5 to 48.5) | 3.4 (– 3.0 to 9.7) |
EQ-5D: mean (95% CI) | |||
Mobility | 2.0 (2.0 to 2.1) | 2.1 (2.0 to 2.2) | 0.1 (– 0.1 to 0.2) |
Self-care | 2.1 (2.0 to 2.3) | 2.1 (2.0 to 2.2) | – 0.1 (– 0.2 to 0.1) |
Usual activities | 2.5 (2.3 to 2.6) | 2.5 (2.4 to 2.6) | 0.0 (– 0.1 to 0.2) |
Pain/discomfort | 1.9 (1.8 to 2.0) | 1.9 (1.7 to 2.0) | – 0.1 (– 0.2 to 0.1) |
Anxiety/depression | 1.8 (1.7 to 2.0) | 1.7 (1.5 to 1.8) | – 0.2 (– 0.3 to 0.0) |
Good/bad health scale | 54.2 (48.7 to 59.6) | 57.1 (52.5 to 61.6) | 2.9 (– 4.2 to 10.0) |
Oxford Handicap Scale: mean (95% CI) | 3.5 (3.3 to 3.7) | 3.3 (3.0 to 3.5) | – 0.2 (– 0.5 to 0.1) |
Upper limb pain
Control | Intervention | p-value | |
---|---|---|---|
Pain description (excruciating, severe, moderate, mild, none): median (IQR) | 4 (mild) (3 to 5) (n = 155) | 4 (mild) (3 to 5) (n = 167) | 0.066 |
Pain score (0–10): median (IQR) | 4 (0 to 6) (n = 153) | 2 (0 to 6) (n = 167) | 0.216 |
Control | Intervention | Difference | |
---|---|---|---|
Pain description (excruciating, severe, moderate, mild, none): mean (95% CI) | 3.7 (3.5 to 3.9) | 4.0 (3.8 to 4.1) | 0.3 (0.0 to 0.5) |
Pain score (0–10): mean (95% CI) | 3.5 (3.0 to 4.0) | 3.0 (2.5 to 3.5) | – 0.5 (– 1.2 to 0.2) |
Control (n = 151) | Intervention (n = 163) | p-value | |
---|---|---|---|
Pain description (excruciating, severe, moderate, mild, none): median (IQR) | 4 (3 to 5) (mild) (n = 149) | 5 (3 to 5) (none) (n = 163) | 0.068 |
Pain score (0–10): median (IQR) | 3 (0 to 6) (n = 148) | 0 (0 to 5) (n = 162) | 0.047 |
Control | Intervention | Difference | |
---|---|---|---|
Pain description (excruciating, severe, moderate, mild, none): mean (95% CI) | 3.7 (3.5 to 3.9) | 4.0 (3.8 to 4.1) | 0.3 (0.0 to 0.5) |
Pain score (0–10): mean (95% CI) | 3.4 (3.0 to 4.0) | 2.7 (2.2 to 3.2) | – 0.8 (– 1.5 to 0.0) |
Control (n = 92) | Intervention (n = 97) | p-value | |
---|---|---|---|
Pain description (excruciating, severe, moderate, mild, none): median (IQR) | 4 (mild) (3 to 5) | 5 (none) (3.5 to 5) | 0.001 |
Pain score (0–10): median (IQR) | 4 (0 to 7) | 0 (0 to 4) | < 0.001 |
Control | Intervention | Difference | |
---|---|---|---|
Pain description (excruciating, severe, moderate, mild, none): mean (95% CI) | 3.7 (3.4 to 3.9) | 4.2 (4.0 to 4.5) | 0.6 (0.2 to 0.9) |
Pain score (0–10): mean (95% CI) | 3.6 (2.9 to 4.3) | 1.9 (1.4 to 2.5) | –1.7 (– 2.6 to – 0.8) |
Appendix 4 Site recruitment
Twelve research sites participated in the BoTULS study. For logistical reasons, some sites were joined or divided to create recruitment areas as shown in Table 61. There were differences in recruitment from each area because of time of site/area initiation and study therapist availability. The study was initially planned in four recruitment areas (North Tyneside, Wansbeck area, Newcastle upon Tyne and Sunderland) but because of low recruitment rates the additional sites/areas were added.
Research site | Recruitment area | First patient recruited | Total recruitment |
---|---|---|---|
Northumbria Healthcare NHS Foundation Trust (North Tyneside General Hospital) | North Tyneside | July 2005 | 37 |
Newcastle Primary Care Trust | Newcastle upon Tyne | October 2005 | 44 |
Newcastle upon Tyne Hospitals NHS Foundation Trust | |||
Northumberland, Tyne and Wear NHS Trust | |||
Northumbria Healthcare NHS Foundation Trust (Wansbeck, Hexham, Morpeth, Blyth, Alnwick hospitals) | Wansbeck, Morpeth, Blyth and Alnwick | November 2005 | 41 |
Hexham | April 2007 | 5 | |
City Hospitals Sunderland NHS Foundation Trust | Sunderland | January 2006 | 69 |
South Tyneside NHS Foundation Trust | South Tyneside | October 2006 | 34 |
Gateshead Health NHS Foundation Trust | Gateshead | November 2006 | 11 |
Durham and Darlington NHS Foundation Trust (Durham hospitals) | Durham | November 2006 | 26 |
North Cumbria University Hospitals NHS Trust | Carlisle | February 2007 | 15 |
Durham and Darlington NHS Foundation Trust (Bishop Auckland General Hospital) | Bishop Auckland | June 2007 | 25 |
North Tees and Hartlepool NHS Foundation Trust | Hartlepool | September 2007 | 16 |
North Tees | October 2007 | 10 |
Appendix 5 Study dissemination
Protocol
The trial protocol (or summary of protocol) is available from:
-
NIHR Health Technology Assessment programme website: http://www.ncchta.org/project/1408.asp.
-
ISRCTN website: http://www.controlled-trials.com/ISRCTN78533119/78533119.
-
UK Clinical Research Network website: http://public.ukcrn.org.uk/search/StudyDetail.aspx?StudyID=2185.
-
The Lancet website: http://www.thelancet.com/protocol-reviews/07PRT-5979.
Study methods
Study method publication
Rodgers H, Shaw L, Price C, van Wijck F, Barnes M, Graham L, et al. Study design and methods of the BoTULS trial: a randomised controlled trial to evaluate the clinical effect and cost effectiveness of treating upper limb spasticity due to stroke with botulinum toxin type A. Trials 2008;9:59. URL: http://www.trialsjournal.com/content/9/1/59.
Study method presentations
-
British Geriatric Society Northern Regional meeting 2005 (poster).
-
British Association of Stroke Physicians meeting 2006 (poster).
-
European Stroke Conference 2006 (poster).
-
World Congress of Rehabilitation 2006 (poster).
-
UK Stroke Forum 2006 (poster).
-
Stroke Research Network meetings 2007 and 2008 (invited posters).
-
North East Stroke Research Network meeting 2007 (invited poster).
-
Society for Research in Rehabilitation meeting 2008 (invited presentation).
Botulinum toxin reviews
Shaw L, Rodgers H. Botulinum toxin type A for upper limb spasticity after stroke. Expert Rev Neurother 2009;9:1713–25.
Shaw L and Rodgers H. Botulinum toxin to treat spasticity after stroke. Stroke Matters 2010;6:12–13.
Study results
A summary of the results was sent to study participants.
Study results publications
Shaw LC, Price CIM, van Wijck FMJ, Shackley P, Steen N, Barnes MP, et al. Botulinum Toxin for the Upper Limb after Stroke (BoTULS) Trial: effect upon impairment, activity limitation and pain. Stroke, in press.
Shackley P, Shaw LC, Price CIM, van Wijck FMJ, Barnes MP, Graham LA, et al. Cost-effectiveness of treating upper limb spasticity due to stroke with botulinum toxin type A: results from the Botulinum Toxin for the Upper Limb after Stroke (BoTULS) trial. Submitted for publication.
Study results presentations
-
Local investigators and study team meeting July 2008.
-
World Stroke Congress 2008 (poster).
-
Association of North of England Physicians meeting 2008 (platform presentation).
-
UK Stroke Forum 2008 (invited presentation).
-
North East Stroke Research Network meeting 2009 (invited presentation).
-
Society for Research in Rehabilitation meeting 2009 (platform presentation).
-
British Geriatric Society Northern Regional meeting 2009 (platform presentation).
-
European Stroke Conference 2009 (platform presentation).
-
British Society of Rehabilitation Medicine meeting 2009 (platform presentation).
-
UK Stroke Forum 2009 (platform presentation).
-
Society for Research in Rehabilitation meeting 2010 (platform presentation).
-
European Stroke Conference 2010 (poster).
List of abbreviations
- ADL
- activities of daily living
- AP
- adductor pollicis
- ARAT
- Action Research Arm Test
- AS
- Ashworth Scale
- AUC
- area under curve
- BNF
- British National Formulary
- BoTULS
- Botulinum Toxin for the Upper Limb after Stroke
- BTX
- botulinum toxin
- CEAC
- cost-effectiveness acceptability curve
- CI
- confidence interval
- COPM
- Canadian Occupational Performance Measure
- DAS
- Disability Assessment Scale
- DMEC
- Data Monitoring and Ethics Committee
- EQ-5D
- European Quality of Life-5 Dimensions
- FAT
- Frenchay Arm Test
- FCR
- flexor carpi radialis
- FCU
- flexor carpi ulnaris
- FDP
- flexor digitorum profundus
- FDS
- flexor digitorum superficialis
- FIM
- Functional Independence Measure
- FPL
- flexor pollicis longus
- ICER
- incremental cost-effectiveness ratio
- ICH-GCP
- International Conference on Harmonisation – Good Clinical Practice
- IQR
- interquartile range
- MAS
- Modified Ashworth Scale
- NE
- north-east
- NHS
- National Health Service
- NICE
- National Institute for Health and Clinical Excellence
- NIHR
- National Institute for Health Research
- NW
- north-west
- QALY
- quality-adjusted life-year
- RCT
- randomised controlled trial
- ROM
- range of movement
- SE
- south-east
- SF-36
- Short Form questionnaire-36 items
- SIGN
- Scottish Intercollegiate Guidelines Network
- SW
- south-west
- TENS
- transcutaneous electrical nerve stimulation
- TSC
- Trial Steering Committee
- VAS
- visual analogue scale
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, Riemsma 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.
-
Amantadine, oseltamivir and zanamivir for the prophylaxis of influenza (including a review of existing guidance no. 67): a systematic review and economic evaluation.
By Tappenden P, Jackson R, Cooper K, Rees A, Simpson E, Read R, et al.
-
Improving the evaluation of therapeutic interventions in multiple sclerosis: the role of new psychometric methods.
By Hobart J, Cano S.
-
Treatment of severe ankle sprain: a pragmatic randomised controlled trial comparing the clinical effectiveness and cost-effectiveness of three types of mechanical ankle support with tubular bandage. The CAST trial.
By Cooke MW, Marsh JL, Clark M, Nakash R, Jarvis RM, Hutton JL, et al. , on behalf of the CAST trial group.
-
Non-occupational postexposure prophylaxis for HIV: a systematic review.
By Bryant J, Baxter L, Hird S.
-
Blood glucose self-monitoring in type 2 diabetes: a randomised controlled trial.
By Farmer AJ, Wade AN, French DP, Simon J, Yudkin P, Gray A, et al.
-
How far does screening women for domestic (partner) violence in different health-care settings meet criteria for a screening programme? Systematic reviews of nine UK National Screening Committee criteria.
By Feder G, Ramsay J, Dunne D, Rose M, Arsene C, Norman R, et al.
-
Spinal cord stimulation for chronic pain of neuropathic or ischaemic origin: systematic review and economic evaluation.
By Simpson, EL, Duenas A, Holmes MW, Papaioannou D, Chilcott J.
-
The role of magnetic resonance imaging in the identification of suspected acoustic neuroma: a systematic review of clinical and cost-effectiveness and natural history.
By Fortnum H, O’Neill C, Taylor R, Lenthall R, Nikolopoulos T, Lightfoot G, et al.
-
Dipsticks and diagnostic algorithms in urinary tract infection: development and validation, randomised trial, economic analysis, observational cohort and qualitative study.
By Little P, Turner S, Rumsby K, Warner G, Moore M, Lowes JA, et al.
-
Systematic review of respite care in the frail elderly.
By Shaw C, McNamara R, Abrams K, Cannings-John R, Hood K, Longo M, et al.
-
Neuroleptics in the treatment of aggressive challenging behaviour for people with intellectual disabilities: a randomised controlled trial (NACHBID).
By Tyrer P, Oliver-Africano P, Romeo R, Knapp M, Dickens S, Bouras N, et al.
-
Randomised controlled trial to determine the clinical effectiveness and cost-effectiveness of selective serotonin reuptake inhibitors plus supportive care, versus supportive care alone, for mild to moderate depression with somatic symptoms in primary care: the THREAD (THREshold for AntiDepressant response) study.
By Kendrick T, Chatwin J, Dowrick C, Tylee A, Morriss R, Peveler R, et al.
-
Diagnostic strategies using DNA testing for hereditary haemochromatosis in at-risk populations: a systematic review and economic evaluation.
By Bryant J, Cooper K, Picot J, Clegg A, Roderick P, Rosenberg W, et al.
-
Enhanced external counterpulsation for the treatment of stable angina and heart failure: a systematic review and economic analysis.
By McKenna C, McDaid C, Suekarran S, Hawkins N, Claxton K, Light K, et al.
-
Development of a decision support tool for primary care management of patients with abnormal liver function tests without clinically apparent liver disease: a record-linkage population cohort study and decision analysis (ALFIE).
By Donnan PT, McLernon D, Dillon JF, Ryder S, Roderick P, Sullivan F, et al.
-
A systematic review of presumed consent systems for deceased organ donation.
By Rithalia A, McDaid C, Suekarran S, Norman G, Myers L, Sowden A.
-
Paracetamol and ibuprofen for the treatment of fever in children: the PITCH randomised controlled trial.
By Hay AD, Redmond NM, Costelloe C, Montgomery AA, Fletcher M, Hollinghurst S, et al.
-
A randomised controlled trial to compare minimally invasive glucose monitoring devices with conventional monitoring in the management of insulin-treated diabetes mellitus (MITRE).
By Newman SP, Cooke D, Casbard A, Walker S, Meredith S, Nunn A, et al.
-
Sensitivity analysis in economic evaluation: an audit of NICE current practice and a review of its use and value in decision-making.
By Andronis L, Barton P, Bryan S.
-
Trastuzumab for the treatment of primary breast cancer in HER2-positive women: a single technology appraisal.
By Ward S, Pilgrim H, Hind D.
-
Docetaxel for the adjuvant treatment of early node-positive breast cancer: a single technology appraisal.
By Chilcott J, Lloyd Jones M, Wilkinson A.
-
The use of paclitaxel in the management of early stage breast cancer.
By Griffin S, Dunn G, Palmer S, Macfarlane K, Brent S, Dyker A, et al.
-
Rituximab for the first-line treatment of stage III/IV follicular non-Hodgkin’s lymphoma.
By Dundar Y, Bagust A, Hounsome J, McLeod C, Boland A, Davis H, et al.
-
Bortezomib for the treatment of multiple myeloma patients.
By Green C, Bryant J, Takeda A, Cooper K, Clegg A, Smith A, et al.
-
Fludarabine phosphate for the firstline treatment of chronic lymphocytic leukaemia.
By Walker S, Palmer S, Erhorn S, Brent S, Dyker A, Ferrie L, et al.
-
Erlotinib for the treatment of relapsed non-small cell lung cancer.
By McLeod C, Bagust A, Boland A, Hockenhull J, Dundar Y, Proudlove C, et al.
-
Cetuximab plus radiotherapy for the treatment of locally advanced squamous cell carcinoma of the head and neck.
By Griffin S, Walker S, Sculpher M, White S, Erhorn S, Brent S, et al.
-
Infliximab for the treatment of adults with psoriasis.
By Loveman E, Turner D, Hartwell D, Cooper K, Clegg A
-
Psychological interventions for postnatal depression: cluster randomised trial and economic evaluation. The PoNDER trial.
By Morrell CJ, Warner R, Slade P, Dixon S, Walters S, Paley G, et al.
-
The effect of different treatment durations of clopidogrel in patients with non-ST-segment elevation acute coronary syndromes: a systematic review and value of information analysis.
By Rogowski R, Burch J, Palmer S, Craigs C, Golder S, Woolacott N.
-
Systematic review and individual patient data meta-analysis of diagnosis of heart failure, with modelling of implications of different diagnostic strategies in primary care.
By Mant J, Doust J, Roalfe A, Barton P, Cowie MR, Glasziou P, et al.
-
A multicentre randomised controlled trial of the use of continuous positive airway pressure and non-invasive positive pressure ventilation in the early treatment of patients presenting to the emergency department with severe acute cardiogenic pulmonary oedema: the 3CPO trial.
By Gray AJ, Goodacre S, Newby DE, Masson MA, Sampson F, Dixon S, et al. , on behalf of the 3CPO study investigators.
-
Early high-dose lipid-lowering therapy to avoid cardiac events: a systematic review and economic evaluation.
By Ara R, Pandor A, Stevens J, Rees A, Rafia R.
-
Adefovir dipivoxil and pegylated interferon alpha for the treatment of chronic hepatitis B: an updated systematic review and economic evaluation.
By Jones J, Shepherd J, Baxter L, Gospodarevskaya E, Hartwell D, Harris P, et al.
-
Methods to identify postnatal depression in primary care: an integrated evidence synthesis and value of information analysis.
By Hewitt CE, Gilbody SM, Brealey S, Paulden M, Palmer S, Mann R, et al.
-
A double-blind randomised placebo-controlled trial of topical intranasal corticosteroids in 4- to 11-year-old children with persistent bilateral otitis media with effusion in primary care.
By Williamson I, Benge S, Barton S, Petrou S, Letley L, Fasey N, et al.
-
The effectiveness and cost-effectiveness of methods of storing donated kidneys from deceased donors: a systematic review and economic model.
By Bond M, Pitt M, Akoh J, Moxham T, Hoyle M, Anderson R.
-
Rehabilitation of older patients: day hospital compared with rehabilitation at home. A randomised controlled trial.
By Parker SG, Oliver P, Pennington M, Bond J, Jagger C, Enderby PM, et al.
-
Breastfeeding promotion for infants in neonatal units: a systematic review and economic analysis.
By Renfrew MJ, Craig D, Dyson L, McCormick F, Rice S, King SE, et al.
-
The clinical effectiveness and cost-effectiveness of bariatric (weight loss) surgery for obesity: a systematic review and economic evaluation.
By Picot J, Jones J, Colquitt JL, Gospodarevskaya E, Loveman E, Baxter L, et al.
-
Rapid testing for group B streptococcus during labour: a test accuracy study with evaluation of acceptability and cost-effectiveness.
By Daniels J, Gray J, Pattison H, Roberts T, Edwards E, Milner P, et al.
-
Screening to prevent spontaneous preterm birth: systematic reviews of accuracy and effectiveness literature with economic modelling.
By Honest H, Forbes CA, Durée KH, Norman G, Duffy SB, Tsourapas A, et al.
-
The effectiveness and cost-effectiveness of cochlear implants for severe to profound deafness in children and adults: a systematic review and economic model.
By Bond M, Mealing S, Anderson R, Elston J, Weiner G, Taylor RS, et al.
-
Gemcitabine for the treatment of metastatic breast cancer.
By Jones J, Takeda A, Tan SC, Cooper K, Loveman E, Clegg A.
-
Varenicline in the management of smoking cessation: a single technology appraisal.
By Hind D, Tappenden P, Peters J, Kenjegalieva K.
-
Alteplase for the treatment of acute ischaemic stroke: a single technology appraisal.
By Lloyd Jones M, Holmes M.
-
Rituximab for the treatment of rheumatoid arthritis.
By Bagust A, Boland A, Hockenhull J, Fleeman N, Greenhalgh J, Dundar Y, et al.
-
Omalizumab for the treatment of severe persistent allergic asthma.
By Jones J, Shepherd J, Hartwell D, Harris P, Cooper K, Takeda A, et al.
-
Rituximab for the treatment of relapsed or refractory stage III or IV follicular non-Hodgkin’s lymphoma.
By Boland A, Bagust A, Hockenhull J, Davis H, Chu P, Dickson R.
-
Adalimumab for the treatment of psoriasis.
By Turner D, Picot J, Cooper K, Loveman E.
-
Dabigatran etexilate for the prevention of venous thromboembolism in patients undergoing elective hip and knee surgery: a single technology appraisal.
By Holmes M, C Carroll C, Papaioannou D.
-
Romiplostim for the treatment of chronic immune or idiopathic thrombocytopenic purpura: a single technology appraisal.
By Mowatt G, Boachie C, Crowther M, Fraser C, Hernández R, Jia X, et al.
-
Sunitinib for the treatment of gastrointestinal stromal tumours: a critique of the submission from Pfizer.
By Bond M, Hoyle M, Moxham T, Napier M, Anderson R.
-
Vitamin K to prevent fractures in older women: systematic review and economic evaluation.
By Stevenson M, Lloyd-Jones M, Papaioannou D.
-
The effects of biofeedback for the treatment of essential hypertension: a systematic review.
By Greenhalgh J, Dickson R, Dundar Y.
-
A randomised controlled trial of the use of aciclovir and/or prednisolone for the early treatment of Bell’s palsy: the BELLS study.
By Sullivan FM, Swan IRC, Donnan PT, Morrison JM, Smith BH, McKinstry B, et al.
-
Lapatinib for the treatment of HER2-overexpressing breast cancer.
By Jones J, Takeda A, Picot J, von Keyserlingk C, Clegg A.
-
Infliximab for the treatment of ulcerative colitis.
By Hyde C, Bryan S, Juarez-Garcia A, Andronis L, Fry-Smith A.
-
Rimonabant for the treatment of overweight and obese people.
By Burch J, McKenna C, Palmer S, Norman G, Glanville J, Sculpher M, et al.
-
Telbivudine for the treatment of chronic hepatitis B infection.
By Hartwell D, Jones J, Harris P, Cooper K.
-
Entecavir for the treatment of chronic hepatitis B infection.
By Shepherd J, Gospodarevskaya E, Frampton G, Cooper, K.
-
Febuxostat for the treatment of hyperuricaemia in people with gout: a single technology appraisal.
By Stevenson M, Pandor A.
-
Rivaroxaban for the prevention of venous thromboembolism: a single technology appraisal.
By Stevenson M, Scope A, Holmes M, Rees A, Kaltenthaler E.
-
Cetuximab for the treatment of recurrent and/or metastatic squamous cell carcinoma of the head and neck.
By Greenhalgh J, Bagust A, Boland A, Fleeman N, McLeod C, Dundar Y, et al.
-
Mifamurtide for the treatment of osteosarcoma: a single technology appraisal.
By Pandor A, Fitzgerald P, Stevenson M, Papaioannou D.
-
Ustekinumab for the treatment of moderate to severe psoriasis.
By Gospodarevskaya E, Picot J, Cooper K, Loveman E, Takeda A.
-
Endovascular stents for abdominal aortic aneurysms: a systematic review and economic model.
By Chambers D, Epstein D, Walker S, Fayter D, Paton F, Wright K, et al.
-
Clinical and cost-effectiveness of epoprostenol, iloprost, bosentan, sitaxentan and sildenafil for pulmonary arterial hypertension within their licensed indications: a systematic review and economic evaluation.
By Chen Y-F, Jowett S, Barton P, Malottki K, Hyde C, Gibbs JSR, et al.
-
Cessation of attention deficit hyperactivity disorder drugs in the young (CADDY) – a pharmacoepidemiological and qualitative study.
By Wong ICK, Asherson P, Bilbow A, Clifford S, Coghill D, R DeSoysa R, et al.
-
ARTISTIC: a randomised trial of human papillomavirus (HPV) testing in primary cervical screening.
By Kitchener HC, Almonte M, Gilham C, Dowie R, Stoykova B, Sargent A, et al.
-
The clinical effectiveness of glucosamine and chondroitin supplements in slowing or arresting progression of osteoarthritis of the knee: a systematic review and economic evaluation.
By Black C, Clar C, Henderson R, MacEachern C, McNamee P, Quayyum Z, et al.
-
Randomised preference trial of medical versus surgical termination of pregnancy less than 14 weeks’ gestation (TOPS).
By Robson SC, Kelly T, Howel D, Deverill M, Hewison J, Lie MLS, et al.
-
Randomised controlled trial of the use of three dressing preparations in the management of chronic ulceration of the foot in diabetes.
By Jeffcoate WJ, Price PE, Phillips CJ, Game FL, Mudge E, Davies S, et al.
-
VenUS II: a randomised controlled trial of larval therapy in the management of leg ulcers.
By Dumville JC, Worthy G, Soares MO, Bland JM, Cullum N, Dowson C, et al.
-
A prospective randomised controlled trial and economic modelling of antimicrobial silver dressings versus non-adherent control dressings for venous leg ulcers: the VULCAN trial
By Michaels JA, Campbell WB, King BM, MacIntyre J, Palfreyman SJ, Shackley P, et al.
-
Communication of carrier status information following universal newborn screening for sickle cell disorders and cystic fibrosis: qualitative study of experience and practice.
By Kai J, Ulph F, Cullinan T, Qureshi N.
-
Antiviral drugs for the treatment of influenza: a systematic review and economic evaluation.
By Burch J, Paulden M, Conti S, Stock C, Corbett M, Welton NJ, et al.
-
Development of a toolkit and glossary to aid in the adaptation of health technology assessment (HTA) reports for use in different contexts.
By Chase D, Rosten C, Turner S, Hicks N, Milne R.
-
Colour vision testing for diabetic retinopathy: a systematic review of diagnostic accuracy and economic evaluation.
By Rodgers M, Hodges R, Hawkins J, Hollingworth W, Duffy S, McKibbin M, et al.
-
Systematic review of the effectiveness and cost-effectiveness of weight management schemes for the under fives: a short report.
By Bond M, Wyatt K, Lloyd J, Welch K, Taylor R.
-
Are adverse effects incorporated in economic models? An initial review of current practice.
By Craig D, McDaid C, Fonseca T, Stock C, Duffy S, Woolacott N.
-
Multicentre randomised controlled trial examining the cost-effectiveness of contrast-enhanced high field magnetic resonance imaging in women with primary breast cancer scheduled for wide local excision (COMICE).
By Turnbull LW, Brown SR, Olivier C, Harvey I, Brown J, Drew P, et al.
-
Bevacizumab, sorafenib tosylate, sunitinib and temsirolimus for renal cell carcinoma: a systematic review and economic evaluation.
By Thompson Coon J, Hoyle M, Green C, Liu Z, Welch K, Moxham T, et al.
-
The clinical effectiveness and cost-effectiveness of testing for cytochrome P450 polymorphisms in patients with schizophrenia treated with antipsychotics: a systematic review and economic evaluation.
By Fleeman N, McLeod C, Bagust A, Beale S, Boland A, Dundar Y, et al.
-
Systematic review of the clinical effectiveness and cost-effectiveness of photodynamic diagnosis and urine biomarkers (FISH, ImmunoCyt, NMP22) and cytology for the detection and follow-up of bladder cancer.
By Mowatt G, Zhu S, Kilonzo M, Boachie C, Fraser C, Griffiths TRL, et al.
-
Effectiveness and cost-effectiveness of arthroscopic lavage in the treatment of osteoarthritis of the knee: a mixed methods study of the feasibility of conducting a surgical placebo-controlled trial (the KORAL study).
By Campbell MK, Skea ZC, Sutherland AG, Cuthbertson BH, Entwistle VA, McDonald AM, et al.
-
A randomised 2 × 2 trial of community versus hospital pulmonary rehabilitation for chronic obstructive pulmonary disease followed by telephone or conventional follow-up.
By Waterhouse JC, Walters SJ, Oluboyede Y, Lawson RA.
-
The effectiveness and cost-effectiveness of behavioural interventions for the prevention of sexually transmitted infections in young people aged 13–19: a systematic review and economic evaluation.
By Shepherd J, Kavanagh J, Picot J, Cooper K, Harden A, Barnett-Page E, et al.
-
Dissemination and publication of research findings: an updated review of related biases.
By Song F, Parekh S, Hooper L, Loke YK, Ryder J, Sutton AJ, et al.
-
The effectiveness and cost-effectiveness of biomarkers for the prioritisation of patients awaiting coronary revascularisation: a systematic review and decision model.
By Hemingway H, Henriksson M, Chen R, Damant J, Fitzpatrick N, Abrams K, et al.
-
Comparison of case note review methods for evaluating quality and safety in health care.
By Hutchinson A, Coster JE, Cooper KL, McIntosh A, Walters SJ, Bath PA, et al.
-
Clinical effectiveness and cost-effectiveness of continuous subcutaneous insulin infusion for diabetes: systematic review and economic evaluation.
By Cummins E, Royle P, Snaith A, Greene A, Robertson L, McIntyre L, et al.
-
Self-monitoring of blood glucose in type 2 diabetes: systematic review.
By Clar C, Barnard K, Cummins E, Royle P, Waugh N.
-
North of England and Scotland Study of Tonsillectomy and Adeno-tonsillectomy in Children (NESSTAC): a pragmatic randomised controlled trial with a parallel non-randomised preference study.
By Lock C, Wilson J, Steen N, Eccles M, Mason H, Carrie S, et al.
-
Multicentre randomised controlled trial of the clinical and cost-effectiveness of a bypass-surgery-first versus a balloon-angioplasty-first revascularisation strategy for severe limb ischaemia due to infrainguinal disease. The Bypass versus Angioplasty in Severe Ischaemia of the Leg (BASIL) trial.
By Bradbury AW, Adam DJ, Bell J, Forbes JF, Fowkes FGR, Gillespie I, et al.
-
A randomised controlled multicentre trial of treatments for adolescent anorexia nervosa including assessment of cost-effectiveness and patient acceptability – the TOuCAN trial.
By Gowers SG, Clark AF, Roberts C, Byford S, Barrett B, Griffiths A, et al.
-
Randomised controlled trials for policy interventions: a review of reviews and meta-regression.
By Oliver S, Bagnall AM, Thomas J, Shepherd J, Sowden A, White I, et al.
-
Paracetamol and selective and non-selective non-steroidal anti-inflammatory drugs (NSAIDs) for the reduction of morphine-related side effects after major surgery: a systematic review.
By McDaid C, Maund E, Rice S, Wright K, Jenkins B, Woolacott N.
-
A systematic review of outcome measures used in forensic mental health research with consensus panel opinion.
By Fitzpatrick R, Chambers J, Burns T, Doll H, Fazel S, Jenkinson C, et al.
-
The clinical effectiveness and cost-effectiveness of topotecan for small cell lung cancer: a systematic review and economic evaluation.
By Loveman E, Jones J, Hartwell D, Bird A, Harris P, Welch K, et al.
-
Antenatal screening for haemoglobinopathies in primary care: a cohort study and cluster randomised trial to inform a simulation model. The Screening for Haemoglobinopathies in First Trimester (SHIFT) trial.
By Dormandy E, Bryan S, Gulliford MC, Roberts T, Ades T, Calnan M, et al.
-
Early referral strategies for management of people with markers of renal disease: a systematic review of the evidence of clinical effectiveness, cost-effectiveness and economic analysis.
By Black C, Sharma P, Scotland G, McCullough K, McGurn D, Robertson L, et al.
-
A randomised controlled trial of cognitive behaviour therapy and motivational interviewing for people with Type 1 diabetes mellitus with persistent sub-optimal glycaemic control: A Diabetes and Psychological Therapies (ADaPT) study.
By Ismail K, Maissi E, Thomas S, Chalder T, Schmidt U, Bartlett J, et al.
-
A randomised controlled equivalence trial to determine the effectiveness and cost–utility of manual chest physiotherapy techniques in the management of exacerbations of chronic obstructive pulmonary disease (MATREX).
By Cross J, Elender F, Barton G, Clark A, Shepstone L, Blyth A, et al.
-
A systematic review and economic evaluation of the clinical effectiveness and cost-effectiveness of aldosterone antagonists for postmyocardial infarction heart failure.
By McKenna C, Burch J, Suekarran S, Walker S, Bakhai A, Witte K, et al.
-
Avoiding and identifying errors in health technology assessment models: qualitative study and methodological review.
By Chilcott JB, Tappenden P, Rawdin A, Johnson M, Kaltenthaler E, Paisley S, et al.
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, NETSCC, HTA
-
Dr Andrew Cook, Consultant Advisor, NETSCC, HTA
-
Dr Peter Davidson, Director of Science Support, NETSCC, HTA
-
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, NETSCC, HTA
-
Dr Edmund Jessop, Medical Adviser, National Specialist, National Commissioning Group (NCG), Department of Health, London
-
Ms Lynn Kerridge, Chief Executive Officer, NETSCC and NETSCC, HTA
-
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, NETSCC, HTA
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 Programme, Department of Health
-
Dr Morven Roberts, Clinical Trials Manager, Medical Research Council
Diagnostic Technologies and 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
-
Mr A S Arunkalaivanan, Honorary Senior Lecturer, University of Birmingham and Consultant Urogynaecologist and Obstetrician, City Hospital
-
Dr Dianne Baralle, Consultant & Senior Lecturer in Clinical Genetics, Human Genetics Division & Wessex Clinical Genetics Service, Southampton, University of Southampton
-
Dr Stephanie Dancer, Consultant Microbiologist, Hairmyres Hospital, East Kilbride
-
Dr Ron Gray, Consultant, National Perinatal Epidemiology Unit, Institute of Health Sciences, University of Oxford
-
Professor Paul D Griffiths, Professor of Radiology, Academic Unit of Radiology, University of Sheffield
-
Mr Martin Hooper, Service User Representative
-
Professor Anthony Robert Kendrick, Professor of Primary Medical Care, University of Southampton
-
Dr Susanne M Ludgate, Director, Medical Devices Agency, London
-
Dr Anne Mackie, Director of Programmes, UK National Screening Committee
-
Dr David Mathew Service User Representative
-
Dr Michael Millar, Lead Consultant in Microbiology, Department of Pathology & Microbiology, Barts and The London NHS Trust, Royal London Hospital
-
Mr Stephen Pilling, Director, Centre for Outcomes, Research & Effectiveness, University College London
-
Mrs Una Rennard, Service User Representative
-
Ms Jane Smith, Consultant Ultrasound Practitioner, Ultrasound Department, Leeds Teaching Hospital NHS Trust, Leeds
-
Dr W Stuart A Smellie, Consultant, Bishop Auckland General Hospital
-
Professor Lindsay Wilson Turnbull, Scientific Director of the Centre for Magnetic Resonance Investigations and YCR Professor of Radiology, Hull Royal Infirmary
-
Dr Alan J Williams, Consultant in General Medicine, Department of Thoracic Medicine, The Royal Bournemouth Hospital
-
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
Disease Prevention Panel
-
Medical Adviser, National Specialist Commissioning Advisory Group (NSCAG), Department of Health
-
Professor of Epidemiology, University of Warwick Medical School, Coventry
-
Dr Robert Cook Clinical Programmes Director, Bazian Ltd, London
-
Dr Elizabeth Fellow-Smith, Medical Director, West London Mental Health Trust, Middlesex
-
Dr Colin Greaves Senior Research Fellow, Peninsular Medical School (Primary Care)
-
Dr John Jackson, General Practitioner, Parkway Medical Centre, Newcastle upon Tyne
-
Dr Russell Jago, Senior Lecturer in Exercise, Nutrition and Health, Centre for Sport, Exercise and Health, University of Bristol
-
Dr Chris McCall, General Practitioner, The Hadleigh Practice, Corfe Mullen, Dorset
-
Miss Nicky Mullany, Service User Representative
-
Dr Julie Mytton, Locum Consultant in Public Health Medicine, Bristol Primary Care Trust
-
Professor Irwin Nazareth, Professor of Primary Care and Director, Department of Primary Care and Population Sciences, University College London
-
Professor Ian Roberts, Professor of Epidemiology and Public Health, London School of Hygiene & Tropical Medicine
-
Professor Carol Tannahill, Glasgow Centre for Population Health
-
Mrs Jean Thurston, Service User Representative
-
Professor David Weller, Head, School of Clinical Science and Community Health, University of Edinburgh
-
Ms Christine McGuire, Research & Development, Department of Health
-
Ms Kay Pattison NHS R&D Programme/DH, Leeds
-
Dr Caroline Stone, Programme Manager, Medical Research Council
External Devices and Physical Therapies Panel
-
Consultant Physician North Bristol NHS Trust, Bristol
-
Reader in Wound Healing and Director of Research, University of Leeds, Leeds
-
Professor Bipin Bhakta Charterhouse Professor in Rehabilitation Medicine, University of Leeds, Leeds
-
Mrs Penny Calder Service User Representative
-
Professor Paul Carding, Professor of Voice Pathology, Newcastle Hospital NHS Trust, Newcastle
-
Dr Dawn Carnes, Senior Research Fellow, Barts and the London School of Medicine and Dentistry, London
-
Dr Emma Clark, Clinician Scientist Fellow & Cons. Rheumatologist, University of Bristol, Bristol
-
Mrs Anthea De Barton-Watson, Service User Representative
-
Professor Christopher Griffiths, Professor of Primary Care, Barts and the London School of Medicine and Dentistry, London
-
Dr Shaheen Hamdy, Clinical Senior Lecturer and Consultant Physician, University of Manchester, Manchester
-
Dr Peter Martin, Consultant Neurologist, Addenbrooke’s Hospital, Cambridge
-
Dr Lorraine Pinnigton, Associate Professor in Rehabilitation, University of Nottingham, Nottingham
-
Dr Kate Radford, Division of Rehabilitation and Ageing, School of Community Health Sciences. University of Nottingham, Nottingham
-
Mr Jim Reece, Service User Representative
-
Professor Maria Stokes, Professor of Neuromusculoskeletal Rehabilitation, University of Southampton, Southampton
-
Dr Pippa Tyrrell, Stroke Medicine, Senior Lecturer/Consultant Stroke Physician, Salford Royal Foundation Hospitals’ Trust, Salford
-
Dr Sarah Tyson, Senior Research Fellow & Associate Head of School, University of Salford, Salford
-
Dr Nefyn Williams, Clinical Senior Lecturer, Cardiff University, Cardiff
-
Dr Phillip Leech, Principal Medical Officer for Primary Care, Department of Health , London
-
Ms Kay Pattison, Section Head R&D, DH, Leeds
-
Dr Morven Roberts, Clinical Trials Manager, MRC, London
-
Dr Ursula Wells PRP, DH, London
Interventional Procedures Panel
-
Consultant Surgeon & Honorary Clinical Lecturer, University of Sheffield
-
Mr David P Britt, Service User Representative, Cheshire
-
Mr Sankaran ChandraSekharan, Consultant Surgeon, Colchester Hospital University NHS Foundation Trust
-
Professor Nicholas Clarke, Consultant Orthopaedic Surgeon, Southampton University Hospitals NHS Trust
-
Mr Seamus Eckford, Consultant in Obstetrics & Gynaecology, North Devon District Hospital
-
Professor David Taggart, Consultant Cardiothoracic Surgeon, John Radcliffe Hospital
-
Dr Matthew Hatton, Consultant in Clinical Oncology, Sheffield Teaching Hospital Foundation Trust
-
Dr John Holden, General Practitioner, Garswood Surgery, Wigan
-
Dr Nadim Malik, Consultant Cardiologist/ Honorary Lecturer, University of Manchester
-
Mr Hisham Mehanna, Consultant & Honorary Associate Professor, University Hospitals Coventry & Warwickshire NHS Trust
-
Dr Jane Montgomery, Consultant in Anaesthetics and Critical Care, South Devon Healthcare NHS Foundation Trust
-
Dr Simon Padley, Consultant Radiologist, Chelsea & Westminster Hospital
-
Dr Ashish Paul, Medical Director, Bedfordshire PCT
-
Dr Sarah Purdy, Consultant Senior Lecturer, University of Bristol
-
Mr Michael Thomas, Consultant Colorectal Surgeon, Bristol Royal Infirmary
-
Professor Yit Chiun Yang, Consultant Ophthalmologist, Royal Wolverhampton Hospitals NHS Trust
-
Mrs Isabel Boyer, Service User Representative, London
Pharmaceuticals Panel
-
Professor in Child Health, University of Nottingham
-
Unit Manager, Pharmacoepidemiology Research Unit, VRMM, Medicines & Healthcare Products Regulatory Agency
-
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
-
Professor Robin Ferner, Consultant Physician and Director, West Midlands Centre for Adverse Drug Reactions, City Hospital NHS Trust, Birmingham
-
Dr Ben Goldacre, Research Fellow, Division of Psychological Medicine and Psychiatry, King’s College London
-
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
-
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
-
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
Psychological and Community Therapies Panel
-
Professor of Psychiatry, University of Warwick
-
Professor Jane Barlow, Professor of Public Health in the Early Years, Health Sciences Research Institute, Warwick Medical School
-
Dr Sabyasachi Bhaumik, Consultant Psychiatrist, Leicestershire Partnership NHS Trust
-
Mrs Val Carlill, Service User Representative, Gloucestershire
-
Dr Steve Cunningham, Consultant Respiratory Paediatrician, Lothian Health Board
-
Dr Anne Hesketh, Senior Clinical Lecturer in Speech and Language Therapy, University of Manchester
-
Dr Yann Lefeuvre, GP Partner, Burrage Road Surgery, London
-
Dr Jeremy J Murphy, Consultant Physician & Cardiologist, County Durham & Darlington Foundation Trust
-
Mr John Needham, Service User, Buckingmashire
-
Ms Mary Nettle, Mental Health User Consultant, Gloucestershire
-
Professor John Potter, Professor of Ageing and Stroke Medicine, University of East Anglia
-
Dr Greta Rait, Senior Clinical Lecturer and General Practitioner, University College London
-
Dr Paul Ramchandani, Senior Research Fellow/Cons. Child Psychiatrist, University of Oxford
-
Dr Howard Ring, Consultant & University Lecturer in Psychiatry, University of Cambridge
-
Dr Karen Roberts, Nurse/Consultant, Dunston Hill Hospital, Tyne and Wear
-
Dr Karim Saad, Consultant in Old Age Psychiatry, Coventry & Warwickshire Partnership Trust
-
Dr Alastair Sutcliffe, Senior Lecturer, University College London
-
Dr Simon Wright, GP Partner, Walkden Medical Centre, Manchester
-
Ms Kay Pattison, Section Head, R&D, DH, Leeds
-
Dr Morven Roberts, Clinical Trials Manager, MRC, London
-
Professor Tom Walley, HTA Programme Director, Liverpool
-
Dr Ursula Wells, Policy Research Programme, DH, London
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