SIGN157 Risk reduction and management of delirium A national clinical guideline March 2019 Key to evidence statements and recommendations Levels of evidence 1++ High-quality meta-analyses, systematic reviews of RCTs, or RCTs with a very low risk of bias 1+ Well-conducted meta-analyses, systematic reviews, or RCTs with a low risk of bias 1– Meta-analyses, systematic reviews, or RCTs with a high risk of bias 2++ High-quality systematic reviews of case-control or cohort studies High-quality case-control or cohort studies with a very low risk of confounding or bias and a high probability that the relationship is causal 2+ Well-conducted case-control or cohort studies with a low risk of confounding or bias and a moderate probability that the relationship is causal 2 – Case-control or cohort studies with a high risk of confounding or bias and a significant risk that the relationship is not causal 3 Non-analytic studies, eg case reports, case series 4 Expert opinion Recommendations Some recommendations can be made with more certainty than others. The wording used in the recommendations in this guideline denotes the certainty with which the recommendation is made (the ‘strength’ of the recommendation). The ‘strength’ of a recommendation takes into account the quality (level) of the evidence. Although higher-quality evidence is more likely to be associated with strong recommendations than lower-quality evidence, a particular level of quality does not automatically lead to a particular strength of recommendation. Other factors that are taken into account when forming recommendations include: relevance to the NHS in Scotland; applicability of published evidence to the target population; consistency of the body of evidence, and the balance of benefits and harms of the options. RFor ‘strong’ recommendations on interventions that ‘should’ be used, the guideline development group is confident that, for the vast majority of people, the intervention (or interventions) will do more good than harm. For ‘strong’ recommendations on interventions that ‘should not’ be used, the guideline development group is confident that, for the vast majority of people, the intervention (or interventions) will do more harm than good. RFor ‘conditional’ recommendations on interventions that should be ‘considered’, the guideline development group is confident that the intervention will do more good than harm for most patients. The choice of intervention is therefore more likely to vary depending on a person’s values and preferences, and so the healthcare professional should spend more time discussing the options with the patient. Good-practice points  Recommended best practice based on the clinical experience of the guideline development group. NICE has accredited the process used by Scottish Intercollegiate Guidelines Network to produce clinical guidelines. The accreditation term is valid until 31 March 2020 and is applicable to guidance produced using the processes described in SIGN 50: a guideline developer’s handbook, 2015 edition (www.sign.ac.uk/assets/sign50_2015.pdf). More information on accreditation can be viewed at www.nice.org.uk/accreditation Healthcare Improvement Scotland (HIS) is committed to equality and diversity and assesses all its publications for likely impact on the six equality groups defined by age, disability, gender, race, religion/belief and sexual orientation. SIGN guidelines are produced using a standard methodology that has been equality impact assessed to ensure that these equality aims are addressed in every guideline. This methodology is set out in the current version of SIGN 50, our guideline manual, which can be found at www.sign.ac.uk/sign-50.html. The EQIA assessment of the manual can be seen at www.sign.ac.uk/assets/sign50eqia.pdf. The full report in paper form and/or alternative format is available on request from the Healthcare Improvement Scotland Equality and Diversity Officer. Every care is taken to ensure that this publication is correct in every detail at the time of publication. However, in the event of errors or omissions corrections will be published in the web version of this document, which is the definitive version at all times. This version can be found on our web site www.sign.ac.uk Scottish Intercollegiate Guidelines Network Risk reduction and management of delirium A national clinical guideline March 2019 Scottish Intercollegiate Guidelines Network Gyle Square, 1 South Gyle Crescent Edinburgh EH12 9EB www.sign.ac.uk First published March 2019 978-1-909103-68-9 Citation text Scottish Intercollegiate Guidelines Network (SIGN). Risk reduction and management of delirium. Edinburgh: SIGN; 2019. (SIGN publication no. 157). [March 2019]. Available from URL: http://www.sign.ac.uk SIGN consents to the photocopying of this guideline for the purpose of implementation in NHSScotland. Contents 1 Introduction 1 1.1 The need for a guideline 1 1.2 Remit of the guideline 1 1.3 Statement of intent 3 2 Key recommendations 5 2.1 Detecting delirium 5 2.2 Risk reduction 5 2.3 Non-pharmacological treatment 5 3 Detecting delirium 6 3.1 Tools for detection and assessment 6 3.2 Tools for measuring severity of delirium 8 3.3 Tools for detecting incident delirium in hospital 8 3.4 Clinical investigations 8 4 Non-pharmacological risk reduction 11 4.1Introduction 11 4.2 Inpatient care 11 4.3 Intensive care 12 5 Pharmacological risk reduction 14 5.1 Medicines optimisation 14 5.2Antipsychotics 15 5.3Dexmedetomidine 16 5.4 Other pharmacological therapies 17 6 Non-pharmacological treatment 18 7 Pharmacological treatment 19 7.1Antipsychotics 19 7.2Dexmedetomidine 19 7.3 Acetylcholinesterase inhibitors 20 7.4Benzodiazepines 20 7.5 Urgent pharmacological intervention 20 8 Follow up 21 9 Provision of information 22 9.1 Checklist for provision of information 22 9.2 Publications from SIGN 24 9.3 Sources of further information 24 10 Implementing the guideline 26 10.1 Implementation strategy 26 10.2 Resource implications of key recommendations 26 10.3 Auditing current practice 26 10.4 Health technology assessment advice for NHSScotland 26 11 The evidence base 27 11.1 Systematic literature review 27 11.2 Recommendations for research 28 11.3 Review and updating 28 12 Development of the guideline 29 12.1Introduction 29 12.2 The guideline development group 29 12.3Acknowledgements 30 12.4 Consultation and peer review 30 Abbreviations 32 Annexes 34 References 42 1 | Introduction 1Introduction 1.1 The need for a guideline Delirium is an acute deterioration in mental functioning arising over hours or days that is triggered mainly by acute medical illness, surgery, trauma, or drugs.1 It was previously termed ‘acute confusional state’. Delirium is independently linked with poor outcomes including medical complications, falls, increased length of hospital stay, new institutionalisation, and mortality.1 It can cause significant patient and carer distress.2 The main features of delirium are acute cognitive deficits and altered level of arousal, with up to half of patients also experiencing hallucinations or delusions.3 Delirium varies in duration, mostly resolving within days, but in some people it can last weeks or months.4 Delirium is among the most common of medical emergencies. A UK study found a prevalence of 20% in adult acute general medical patients.5 The prevalence is higher in particular clinical groups, such as patients in intensive care units (ICU). It affects up to 50% who have hip fracture and up to 75% in intensive care.1 Several predisposing factors increase the risk of delirium, including older age, dementia, frailty, the presence of multiple comorbidities, male sex, sensory impairments, a history of depression, a history of delirium, and alcohol misuse.1,6 Despite its importance, there are deficiencies in care of people with delirium in Scotland. It is underdiagnosed,7 and the treatment of patients with established delirium is variable. Preventative measures can reduce the incidence of delirium,1 yet few clinical units have formal delirium risk- reduction programmes. Experience gained from quality improvement programmes in Scotland shows that advances can be made.8 There is potential to improve clinical practice by reducing variation in the standards of assessment and management of people with delirium. This new national guideline on delirium provides a critical focal point for Scotland-wide improvements in delirium care. Because delirium is so common, all healthcare staff having contact with acutely unwell patients need to assume responsibility for detecting and treating it, as well as aiming to reduce the risk of delirium occurring. Those working in the long-term care environment should be able to recognise delirium, reduce risk, and monitor those in their care to resolve delirium. 1.1.1 Patient and carer perspective Common concerns raised by patient groups and through research into patient and carer issues identified good communication with family members or carers as crucial. Family members can provide background information on patient history, changes in behaviour and early warning signs. Once diagnosed, carers need information and support to enable them to care for the patient (see section 9).9-11 1.2 Remit of the guideline 1.2.1 Overall objectives This guideline provides recommendations based on current evidence for best practice in the detection, assessment, treatment and follow up of adults with delirium, as well as reducing the risk of delirium. The guideline applies to all settings: home, long-term care, hospital, and hospice. It is important to note that, to date, much of the existing evidence and the focus of other guidelines, is in acute care settings. However, this does not preclude application of the recommendations to other settings, adapted according to clinician judgement. Person-centred care should be the focus of the implementation of this guideline. 1 Risk reduction and management of delirium The guideline excludes delirium secondary solely to alcohol and illicit substances use. It also excludes delirium in children. 1.2.2 Common comorbidities Common comorbidities which have been considered when reviewing the evidence for this guideline are: • critical illness • dementia • depression • frailty • head injury • learning disability • Parkinson’s disease • cerebrovascular disease. 1.2.3Definitions The International Classification of Diseases, version 10 (ICD-10) defines delirium as, “An aetiologically nonspecific organic cerebral syndrome characterized by concurrent disturbances of consciousness and attention, perception, thinking, memory, psychomotor behaviour, emotion, and the sleep-wake schedule. The duration is variable and the degree of severity ranges from mild to very severe.”12 According to the Diagnostic and Statistical Manual of Mental Disorders, fifth edition (DSM-5), delirium is a disturbance in attention and awareness with an additional disturbance in cognition, not explained by another pre-existing, established or evolving neurocognitive disorder or coma. The disturbance develops over a short period of time and tends to fluctuate in severity during the course of the day with evidence of direct physiological consequence of another medical condition, substance intoxication or withdrawal, or exposure to a toxin, or is due to multiple aetiologies. It’s specified as ‘acute’, lasting a few hours or days and ‘persistent’, lasting weeks or months.13 Delirium presents variably but its main characteristics are rapid onset (hours, days) of acute mental status deterioration. Patients may present with cognitive impairment, but drowsiness to the point that the patient is not speaking, severe agitation, or psychotic features such as hallucinations or delusions may be the most prominent features. Delirium is sometimes described using hyperactive, hypoactive or mixed labels depending on the level of arousal.13 Most delirium has a duration of a small number of days, but in around 20% of cases, it can persist for weeks or months.14 Delirium is known by several terms, some still in use in clinical practice. These terms include ‘acute confusional state’, ‘acute confusion’, ‘acute on chronic confusion’, and ‘acute encephalopathy’. The SIGN guideline group advocates use of the term delirium rather than alternatives to promote more consistent communication among professionals, more accurate provision of information to patients and carers, and more consistent use of detection tools and management strategies. 1.2.4 Target users of the guideline This guideline will be of interest to primary and secondary healthcare professionals, community and care home staff involved in the care of patients at risk of, or experiencing, delirium, as well as patients and carers. 2 1 | Introduction 1.3 Statement of intent This guideline is not intended to be construed or to serve as a standard of care. Standards of care are determined on the basis of all clinical data available for an individual case and are subject to change as scientific knowledge and technology advance and patterns of care evolve. Adherence to guideline recommendations will not ensure a successful outcome in every case, nor should they be construed as including all proper methods of care or excluding other acceptable methods of care aimed at the same results. The ultimate judgement must be made by the appropriate healthcare professional(s) responsible for clinical decisions regarding a particular clinical procedure or treatment plan. This judgement should only be arrived at through a process of shared decision making with the patient (or family or carers, where appropriate), covering the diagnostic and treatment choices available. It is advised, however, that significant departures from the national guideline or any local guidelines derived from it should be documented in the patient’s medical records at the time the relevant decision is taken. 1.3.1 Influence of financial and other interests It has been recognised that financial interests in, or close working relationships with, pharmaceutical companies may have an influence on the interpretation of evidence from clinical studies. It is not possible to completely eliminate any possible bias from this source, nor even to quantify the degree of bias with any certainty. SIGN requires that all those involved in the work of guideline development should declare all financial interests, whether direct or indirect, annually for as long as they are actively working with the organisation. By being explicit about the influences to which contributors are subjected, SIGN acknowledges the risk of bias and makes it possible for guideline users or reviewers to assess for themselves how likely it is that the conclusions and guideline recommendations are based on a biased interpretation of the evidence. While developing this guideline, one academic conflict of interest was declared. As the main author of the 4AT test, Co-Chair Professor Alasdair MacLullich did not participate in the process to form recommendations to address key question 1: tools for detecting delirium (see Annex 1). Signed copies of declaration of interests forms are retained by the SIGN Executive and a register of interests is available in the supporting material section for this guideline at www.sign.ac.uk 1.3.2 Prescribing of licensed medicines outwith their marketing authorisation Recommendations within this guideline are based on the best clinical evidence. Some recommendations may be for medicines prescribed outwith the marketing authorisation (MA) also known as product licence. This is known as ‘off-label’ use. Medicines may be prescribed ‘off label’ in the following circumstances: • for an indication not specified within the marketing authorisation • for administration via a different route • for administration of a different dose • for a different patient population. An unlicensed medicine is a medicine which does not have MA for medicinal use in humans. Generally ‘off-label’ prescribing of medicines becomes necessary if the clinical need cannot be met by licensed medicines within the marketing authorisation. Such use should be supported by appropriate evidence and experience.15 3 Risk reduction and management of delirium “Prescribing medicines outside the conditions of their marketing authorisation alters (and probably increases) the prescribers’ professional responsibility and potential liability.”15 The General Medical Council (GMC) recommends that when prescribing a medicine ‘off label’, doctors should:16 • be satisfied that there is no suitably licensed medicine that will meet the patient’s need. • b e satisfied that there is sufficient evidence or experience of using the medicine to show its safety and efficacy • t ake responsibility for prescribing the medicine and for overseeing the patient’s care, monitoring, and any follow-up treatment, or ensure that arrangements are made for another suitable doctor to do so. • m ake a clear, accurate and legible record of all medicines prescribed and, when not following common practice, the reasons for prescribing an unlicensed medicine. Non-medical prescribers should ensure that they are familiar with the legislative framework and their own professional prescribing standards. Prior to any prescribing, the licensing status of a medication should be checked in the summary of product characteristics (www.medicines.org.uk). The prescriber must be competent, operate within the professional code of ethics of their statutory bodies and the prescribing practices of their employers.17 1.3.3 Health technology assessment advice for NHSScotland Specialist teams within Healthcare Improvement Scotland issue a range of advice that focuses on the safe and effective use of medicines and technologies in NHSScotland. The Scottish Medicines Consortium (SMC) provides advice to NHS boards and their Area Drug and Therapeutics Committees about the status of all newly-licensed medicines, all new formulations of existing medicines and new indications for established products. NHSScotland should take account of this advice and ensure that medicines accepted for use are made available to meet clinical need where appropriate. SMC advice relevant to this guideline is summarised in section 10.4. 4 2 | Key recommendations 2 Key recommendations The following recommendations were highlighted by the guideline development group as the key clinical recommendations that should be prioritised for implementation. 2.1 Detecting delirium RThe 4AT tool should be used for identifying patients with probable delirium in emergency department and acute hospital settings. 99 here delirium is detected, the diagnosis of delirium should be clearly documented and W coded to aid transfers of care (eg handover notes, referral and discharge letters). 2.2 Risk reduction RThe following components should be considered as part of a package of care for patients at risk of developing delirium: • orientation and ensuring patients have their glasses and hearing aids • promoting sleep hygiene • early mobilisation • pain control • prevention, early identification and treatment of postoperative complications • maintaining optimal hydration and nutrition • regulation of bladder and bowel function • provision of supplementary oxygen, if appropriate. RAll patients at risk of delirium should have a medication review conducted by an experienced healthcare professional. 2.3 Non-pharmacological treatment RHealthcare professionals should follow established pathways of good care to manage patients with delirium. • F irst consider acute, life-threatening causes of delirium, including low oxygen level, low blood pressure, low glucose level, and drug intoxication or withdrawal. ystematically identify and treat potential causes (medications, acute illness, etc), noting • S that multiple causes are common. ptimise physiology, management of concurrent conditions, environment (reduce noise), • O medications, and natural sleep, to promote brain recovery. • S pecifically detect, assess causes of, and treat agitation and/or distress, using non- pharmacological means only if possible (see section 7 for pharmacological treatment). • C ommunicate the diagnosis to patients and carers, encourage involvement of carers and provide ongoing engagement and support. • A im to prevent complications of delirium such as immobility, falls, pressure sores, dehydration, malnourishment, isolation. • Monitor for recovery and consider specialist referral if not recovering. • Consider follow up (see section 8). 5 Risk reduction and management of delirium 3 Detecting delirium 3.1 Tools for detection and assessment Delirium is frequently missed in routine clinical care and lack of detection is associated with poor outcomes.18,19 Delirium detection should ideally be undertaken at the earliest opportunity. Numerous assessment tools have been developed to help identify probable delirium in patients in a variety of settings, which can then prompt a more accurate diagnosis and consideration of underlying causes. For practical reasons, for implementation and acceptability to patients, assessment tools should be brief, require little or no training and be appropriate to the clinical setting.20 The sensitivity of the tool is also important, as it is vital not to miss delirium. A commonly used tool, the Confusion Assessment Method (CAM) and its variants have been reported as useful tools for detecting delirium.18,21,22 However, sensitivity and specificity varied broadly, possibly due to the need for users to have training and knowledge of delirium and its differential diagnoses. The CAM-ICU has particularly broad use within ICU settings but has the 2+ same limitations.19,23 The 4 As Test (Arousal, Attention, Abbreviated Mental Test 4, Acute change) 2++ (4AT) was developed, validated and widely implemented in Scotland in non-ICU settings. It does 2+ not require specific training, is brief and easy to use and has wide applicability in various clinical settings.24,25 It performed well for sensitivity and patient completion rate compared to other similar tools within the same patient group.20,26 The 4AT is also applicable as an assessment tool in older emergency department attendees.27 Other tools had significant disadvantages over CAM and 4AT, such as longer assessment time, poorer sensitivity and/or specificity, and/or relative lack of validation in published studies (see Table 1). 2+ The 13-item Delirium Observation Screening Scale (DOS) had good specificity and sensitivity but 2++ requires assessment over three shift periods and its authors have suggested it is geared more towards detection of hyperactive delirium, whereas hypoactive is more common in practice.28-30 The CAM-ICU and Intensive Care Delirium Screening Checklist (ICDSC) have been developed and 2+ validated in ICU settings, and may be better suited than other tests for use in intensive care.31 In all cases, a positive assessment should be followed by additional assessment and diagnosis against ICD-10 or DSM-5 criteria by a suitably trained clinician.12,13 It is important to be aware that delirium may still occur in the absence of a positive test result because the condition fluctuates. Healthcare staff should not rely on the result of a single assessment during hospital admission. Assessment of the patient’s capacity to make decisions about fundamental health and personal care must also be taken into consideration. If the person is deemed to be incapacitated appropriate documentation (eg in Scotland, Adults with Incapacity Act, Section 47 part 5 certificate with accompanying treatment plan) must be completed. Table 1 summarises the commonly used and validated brief delirium assessment tools. There is a wide range of sensitivities and specificities with the different tools as well as in the time taken to complete assessment. Some are designed for assessment at first presentation, and others for monitoring for incident delirium. A tool with high sensitivity that requires no training or very little time to perform, and with additional advantages (for example, suitable for patients with dementia), will be important in clinical practice to ensure all cases of delirium are identified. In the case of uncertainty over whether delirium or dementia or both are present in a patient, it is best to assume it is delirium unless there is clarification from the patient’s notes or from family members that the mental state is clearly in keeping with their usual mental state. 6 Tool Time taken Training Staff Settings Reported Reported Delirium Suitable for Suitable for (min) Required Sensitivity Specificity severity monitoring detecting % % rating DSD 4AT20,24,26,27,30,32 <2 No Any Multiple 86–100 65–82 No No Yes AMT20,26 2 No Any Medical 75–87 61–64 No No No CAM and 3–10 Yes Any Multiple 46–94 63–100 No† No Yes variants18,20,22,24,30,33 CAM-ICU19,22,30,31 <5 Yes Any ICU 28–100 53–99 No Yes No DOS (13-item)*28-30 5 Minimal Any Multiple 89–100 87–97 Yes Yes No DRS- R-9830,34,35 20 Yes Psychiatry Multiple 57–93 82–98 Yes No Yes ICDSC19,31 7–10 Minimal Any ICU 73–97 69–97 Yes Yes No Table 1: Overview of delirium assessment tools MMSE36 5 Minimal Any Multiple 76–91 51–84 Yes No No Nu-DESC30 <5 No Any Multiple 32–96 69–92 No No No RADAR37 <1 No Any Multiple 43–84 64–78 No Yes No mRASS33,38 1 No Any Multiple 65–75 82–90 Yes Yes Yes SQiD20 <1 No Any Medical 77–91 56–71 No Yes No Suitability for monitoring refers to the use of a tool daily or more for screening for incident delirium. †with the exception of CAM-S *DOS requires assessment over three shifts so time to detection is three days. It is geared towards assessment of hyperactive delirium. Abbreviations: AMT – Abbreviated Mental Test; CAM – Confusion Assessment Method; DSD – delirium superimposed on dementia; DRS-98-R – Delirium Rating Scale; DOS - Delirium Observation Screening Scale; ICDSC – Intensive Care Delirium Screening Checklist; Nu-DESC – Nursing Delirium Screening Scale; MMSE – Mini Mental State Examination; RADAR – Recognising those in intensive care. This need not preclude their use in other settings, such as primary care. assessed using an assessment tool, but studies are in older people (aged 65 or over) in hospital and 7 Evidence was not identified to determine exactly which groups of patients should be routinely 3 | Detecting delirium Acute Delirium As part of your Routine; mRASS – Modified Richmond Agitation-Sedation Scale; SQiD – Single Question to Identify Delirium Risk reduction and management of delirium RThe 4AT tool should be used for identifying patients with probable delirium in emergency department and acute hospital settings. RUse of the 4AT tool could be considered for use in community or other settings for identifying patients with probable delirium. RFor intensive care unit settings, CAM-ICU or ICDSC should be considered to help identify patients with probable delirium. 99 A formal assessment and diagnosis must be made by a suitably trained clinician whenever patients with probable delirium are identified. 99 here delirium is detected, patients and their family/carers should be informed of the W diagnosis (see section 9). 99 here delirium is detected, the diagnosis of delirium should be clearly documented to aid W transfers of care (eg handover notes, referral and discharge letters). 3.2 Tools for measuring severity of delirium Monitoring patients diagnosed with delirium for changes in severity or response to treatment may help predict the full clinical impact.39,40 Insufficient evidence was identified to recommend a 3 particular tool for monitoring purposes. Table 1 lists tools that assess severity. Selection of a tool should take into consideration time required and ease of use. The 13-item DOS, mRASS, MMSE, DRS-R-98 and ICDSC can be considered as tools for the purpose of monitoring severity of delirium in suitable clinical areas (see Table 1). 3.3 Tools for detecting incident delirium in hospital Insufficient evidence was identified to recommend a particular tool for regular monitoring purposes for detection of incident delirium after initial assessment in hospital. Selection of a tool should take into consideration time required and ease of use. Although theoretically any tool listed in Table 1 in section 3.1 could be used, the RADAR, 13 item DOS, mRASS, CAM-ICU and SQiD are most suitable for monitoring purposes in suitable clinical areas (see Table 1). 3.4 Clinical investigations Many conditions can trigger delirium. There is often more than one contributory factor in an individual person.41 A major part of treating people with delirium is treating the underlying precipitants or causes. A structured approach should be taken to identify, where possible, the issues contributing to delirium for an individual (see Annex 2). These include obtaining a good history from the person, a collateral or informant history, clinical examination (including a neurological examination), basic and then targeted investigations. There is little evidence supporting the use of basic investigations because a fundamental standard of care is assumed in trials, and trials have not been conducted comparing, for example, carrying out a full blood count or not, in a person with delirium. 8 3 | Detecting delirium This section examines the available evidence for advanced investigations which are more invasive or expensive or where a condition may be identified which significantly alters the management of a person (eg identifying stroke, subdural haemorrhage or non-convulsive status epilepticus). If there is suspicion of significant illness (eg meningitis, encephalitis or sepsis) causing the delirium, these should be managed appropriately. Strategies for such an approach and systems of care are outlined in the Royal College of Physicians’ ‘Acute Care Toolkit 3: Acute medical care for frail older people’ and in the Healthcare Improvement Scotland TIME Bundle (see Annex 3).42 3.4.1 Brain imaging The aim of brain imaging is to identify stroke, haemorrhage, trauma or structural abnormality, such as a tumour, as causes of delirium. The diagnostic yield of computed tomography (CT) in determining the cause of delirium is low, but may be indicated in some high-risk patients.43 For patients with 3 pre-existing cognitive impairment who have other identified conditions that can precipitate delirium, such as dehydration or infection, brain imaging is unlikely to change management.44 Observational, mostly retrospective, studies identified abnormal brain imaging with CT in people aged over 70 years presenting with acute confusion and: • new focal neurological signs44-48 (defined as acute onset dysphasia, visual field defect, pyramidal or cerebellar signs).46 There should be awareness that dysphasia (a focal sign) may be mistaken for confusion (a global brain dysfunction)49 3 4 • presenting after a fall46,48 • a reduced level of consciousness46,47 • a head injury (in patients of any age)50 • taking anticoagulant therapy.48 Cerebral atrophy is more likely in patients presenting with delirium than without.51 This in itself, 3 however, is not a useful finding in making a diagnosis of delirium or changing medical management. RCT brain scan should not be used routinely but should be considered in patients presenting to hospital with delirium in the presence of: • new focal neurological signs • a reduced level of consciousness (not adequately explained by another cause) • a history of recent falls • a head injury (patients of any age) • anticoagulation therapy. 99 Consideration should be given to imaging patients with non-resolving delirium where no clear cause is identified or there are features to suggest primary central nervous system pathology. 9 Risk reduction and management of delirium 3.4.2Electroencephalogram Currently electroencephalogram (EEG) is not performed routinely in patients with delirium, however, three retrospective studies from an epilepsy research group suggest that the incidence of epileptic activity and non-convulsive status epilepticus (NCSE) are higher than recognised in 3 patients with delirium. One study found that 80% of patients with NCSE had delirium attributed initially to another cause.52 In one prospective study EEG was carried out in 44 patients aged 60 years or older with “confusion of unknown origin” (defined as unexplained confusion following screening investigations, including CT scan and blood sample screening, carried out in an emergency unit). Patients with a known 3 history of epilepsy or in whom seizure-related confusion was highly suspected were excluded. Seven (15.9%) patients had EEG changes consistent with NCSE. Clinical indications of NCSE, with statistical significance, were rapid onset (<24 hours), lack of response towards simple commands and female gender.53 Continuous EEG monitoring is more sensitive than single EEG assessment at identifying epileptic activities and NCSE (28% v 6%).52 3 Further evidence is needed to determine the efficacy of routine use of EEG in patients presenting with confusion. RElectroencephalogram should be considered when there is a suspicion of epileptic activity or non-convulsive status epilepticus as a cause of a patient’s delirium. 3.4.3 Lumbar puncture Only one small study from the 1980s was identified on the use of lumbar puncture in the assessment of patients with delirium. It concluded that most patients with fever and delirium have a cause other than infection in the central nervous system (80 of 81 samples were negative for bacterial 3 growth). Given the age of the trial, viral polymerase chain reaction (PCR) testing is unlikely to have been performed.54 Lumbar puncture is not a straightforward procedure and such an invasive investigation may cause further distress to someone who may be confused or agitated. There is also a risk of adverse events, such as infection, causing spinal haematoma, cerebrospinal fluid (CSF) leak or low pressure CSF headache.55 99 Lumbar puncture should not be performed routinely on patients presenting with delirium. 10 4 | Non-pharmacological risk reduction 4 Non-pharmacological risk reduction 4.1Introduction Delirium is often multifactorial. Prevention may merge with treatment when non-pharmacological practices are used. Risk reduction should therefore be considered throughout the patient’s care. Many of the acute factors triggering delirium or lowering the threshold of risk are modifiable. Targeting these modifiable factors forms the basis of reducing the risk of delirium. Up to 50% of delirium in hospitalised patients arises after hospital admission.1,56 Categories of risk reduction include preventing physiological derangements such as dehydration and hypoxia, maintaining sleep, reducing psychological stress through communication and managing the environment, and correcting sensory impairments when possible. These non-pharmacological strategies have often been delivered in multicomponent packages, and trials of such packages form the majority of the evidence. Because of limited resources, targeting higher-risk patients (eg older people, or those with cognitive impairments) for specific delirium risk-reduction strategies is commonly advocated.57,58 To date, these strategies are considered distinct from the use of medication to reduce the risk of delirium (see section 5), and are advocated in pathways and guidelines based on expert opinion.58-60 Non-pharmacological practices should be tried before pharmacological interventions are considered. 4.2 Inpatient care Studies in a variety of patients and settings (acute and perioperative) have found multicomponent interventions to be effective in reducing incidence of delirium.61-64 Meta-analysis of seven studies found that compared to usual care there was a significant reduction in incidence of delirium with multicomponent interventions, with a relative risk (RR) of 0.73, 95% confidence interval (CI) 0.63 to 0.85.62 Pooled analysis in a Cochrane review also reported a reduction in incidence of delirium, (RR) 0.69, 95% CI 0.59 to 0.81 compared to usual care.63 Interventions included in multicomponent care varied, but consisted of some of the following; physiotherapy, reorientation, early mobilisation, 2++ 1++ identification and treatment of underlying causes or postoperative complications, pain control, 1+ regulation of bowel and bladder function, hydration and nutrition, and oxygen delivery.61-63 Such interventions are considered to be good fundamental care.63 Comprehensive geriatric care, defined as a specialist geriatric orthopaedics team providing comprehensive medical assessment, management and initiation of rehabilitation, was also associated with lower incidence of delirium during the hospital stay and at one month.65 Most of the studies identified in the systematic reviews were medium or low quality. Use of a checklist may help to embed good fundamental care and reduce incidence of delirium in patients after an operation.62,63,66 Educating relatives or carers to deliver non-pharmacological multicomponent interventions, such as reorientation, can also reduce the incidence of delirium.9 1+ 1++ A systematic review identified one randomised controlled trial (RCT) that reported an 8% reduction 2++ in the incidence of delirium in those patients cared for by relatives who were educated in 4 delivering a reorientating intervention versus care as usual, RR 0.42, 95% CI 0.19 to 0.92.61 Advice on information to provide to family and carers, and suggestions of ways to help are in section 9. Expert consensus recommends the use of multicomponent interventions as fundamental good 4 practice.58,67,68 Pathways for good practice for risk reduction and management are in Annexes 3 and 4. 11 Risk reduction and management of delirium RThe following components should be considered as part of a package of care for patients at risk of developing delirium: • orientation and ensuring patients have their glasses and hearing aids • promoting sleep hygiene • early mobilisation • pain control • prevention, early identification and treatment of postoperative complications • maintaining optimal hydration and nutrition • regulation of bladder and bowel function • provision of supplementary oxygen, if appropriate. 99 Ward moves should be avoided wherever possible for patients at risk of delirium. 99 rior to surgery patients and carers should be advised of the risk of developing delirium, P to alleviate distress and help with management if it does occur. 99 here possible, assistance should be sought from a patient’s relatives and carers to deliver W care to reduce the risk of delirium developing. 4.2.1 Anaesthetic management Using monitoring to avoid episodes of deep anaesthesia in patients aged over 60 under general anaesthesia for surgery lasting more than one hour can significantly reduce the risk of developing postoperative delirium. Two RCTs have shown a reduction in incidence of 16.7% in the monitoring group versus 21.4% in the control group and 15.6%, intervention, versus 24.1%, control.69,70 1++ A substudy from a large RCT showed a risk reduction that did not reach statistical significance (18.8% 1+ in the intervention group and 28% in the control group), however, meta-analysis of the three trials and one further study of bispectral index-guided sedation reported an odds ratio (OR) of 0.56, 95% CI 0.42 to 0.73.71 None of the studies included patients with dementia, emergency anaesthesia or surgery for hip fracture in older patients. RDepth of anaesthesia should be monitored in all patients aged over 60 years under general anaesthesia for surgery expected to last for more than one hour, with the aim of avoiding excessively deep anaesthesia. 4.3 Intensive care A number of studies of non-pharmacological interventions in ICU settings were identified.72-77 Interventions included acupuncture, mirror therapies, and range of motion exercises. Most of the studies were underpowered. The largest trial addressed the use of dynamic light therapy to 1+ 1– reduce the incidence and duration of delirium in patients in ICU.77 It did not find the therapy to be more effective than placebo. Due to the heterogeneity of interventions and populations no single intervention for patients in ICU can be recommended. A systematic review of eight studies of a multicomponent care approach reported benefit in five of the studies.78 The other three studies showed no difference between the treatment and control groups. However, the multicomponent care approach is considered as standard good practice (see 1+ section 4.2), and the effect of multimodal therapy may not be as evident as in other patient groups, given that critically ill patients exhibit ongoing risk factors for much of their critical care admission. 12 4 | Non-pharmacological risk reduction The use of earplugs, either alone or along with eye shades and other noise-reducing strategies to promote sleep in ICUs, was associated with a reduction in the incidence of delirium, RR 0.59, 95% CI 0.44 to 0.78, in a systematic review of five low-quality studies (832 patients).79 Suitability 2++ for earplugs should be considered on an individual basis as there may be a risk of exacerbating confusion in some patients. RThe use of earplugs should be considered as part of a sleep-promotion strategy in intensive care. 13 Risk reduction and management of delirium 5 Pharmacological risk reduction 5.1 Medicines optimisation Delirium has numerous causes that interact in any one person to cause delirium. Several classes of medication can increase the likelihood of delirium occurring, and the probability that a drug will precipitate delirium should be considered when prescribing, particularly in those at increased risk 1++ of delirium.61,80,81 Observational evidence suggests that exposure to certain medicines increases 2+ the odds of delirium developing and that medication review can decrease rates of delirium.80-83 The following is an approach to medication review and prescribing in people who are experiencing, or are at increased risk of, delirium, and covers three broad areas. • A ny changes in medications, including over-the-counter and herbal medications. Commencement of new medications, changes in dosage of medication or abrupt withdrawal of medication could result in delirium.84,85 • C hanges in how the body handles and is affected by medication. The natural physiology of 3 ageing can result in medication, which has been beneficial without side effects for years, 4 2+ now causing or contributing to delirium. The same can also be said for acute derangements in physiology seen with illness.84 • D elirium risk should be considered when assessing the risks and benefits of commencing a new medication.81,84 It is impossible to cover all medication that may cause or contribute to delirium but some medication particularly associated with delirium merits discussion. Benzodiazepines markedly increase the odds of delirium developing in a variety of settings (OR 3.0 95% CI 1.3 to 6.8) and should not be used unless in specific circumstances such as management 1++ of alcohol withdrawal or acute seizure management.81 Opiates can also cause delirium but they remain a vital class of drug for treating pain. It is important to remember that pain in itself can precipitate delirium. A systematic review cited an OR of delirium associated with treatment with opioids of 2.5 (95% CI 1.2 to 5.2).81 However, the main opioid in the review, which carries the highest risk, is pethidine (OR 2.7, 95% CI 1.3 to 5.5), which 1++ is anticholinergic and is rarely used in the UK. More commonly used opioids such as morphine (OR of delirium 1.2, 95% CI 0.6 to 2.4) and fentanyl (OR of delirium 1.5, 95% CI 0.6 to 4.2) were not significantly associated with delirium.81 The opioid with the lowest odds of causing delirium was oxycodone but this still had a confidence interval that crossed 1 (OR 0.7, 95% CI 0.3 to 1.6).81 Probably the most important factor when using opiate analgesia is titrating to the minimal effective dose to achieve pain control and minimise side effects. If opiates are used it is also important to be holistic and prescribe laxatives to prevent constipation which can contribute to delirium. Optimising the dose of analgesic and sedative drugs in a critical care setting is advocated.86 Daily sedative interruption or nurse-protocolised sedation to facilitate spontaneous breathing trials and 4 daily mobilisation have been associated with improved outcomes, although not directly delirium.86 Although delirium is not specifically covered in the NHSScotland polypharmacy guideline, the guideline contains information on medication to avoid or reduce in older people, some of which is aimed at reducing falls by reducing medications that can cause delirium. Medications recommended 4 to avoid, stop or reduce the dose if possible include tricyclic antidepressants, anticholinergic medications, benzodiazepines, antihistamines and tramadol. The guideline also contains practical information on how to safely reduce chronic medication with potential for withdrawal such as benzodiazepines.87 14 5 | Pharmacological risk reduction RAll patients at risk of delirium should have a medication review conducted by an experienced healthcare professional. 99 reas with patients at high risk of delirium, such as trauma orthopaedic wards, should have A protocols for commonly required medication (eg analgesia and anti-emesis) that contain choices for first-line treatments which minimise the risk of causing delirium. 5.2Antipsychotics Some, low-quality, studies suggest that prophylactic antipsychotic medication may be beneficial for the prevention of postoperative delirium in patients undergoing cardiac, general, elective joint replacement and hip fracture surgery.88-91 One systematic review did not support its use.92 Results 1+ in this review may have been skewed by the inclusion of a controlled trial in which an imbalance 1++ in the age of participants could have been a confounding factor. A Cochrane review concluded that there was no evidence of benefit for the use of haloperidol, but olanzapine compared to placebo, reduced the incidence of delirium (RR 0.36, 95% CI 0.24 to 0.52).63 A systematic review of several small studies evaluated the use of haloperidol prophylaxis in ICU or surgical patients compared to placebo and found that haloperidol prophylaxis did not decrease 1++ the incidence of delirium overall or in subgroup analysis of the patient groups (ICU or surgery).93 A systematic review of six RCTs concluded that there appears to be a greater benefit from antipsychotic prophylaxis in patients at higher risk of delirium.88 If delirium did occur, prophylaxis did not reduce the severity or duration, length of hospital stay or mortality.88 Two systematic 1++ reviews cited one RCT which found that although prophylactic haloperidol did not reduce incidence of delirium it was associated with a reduction in delirium duration and severity.88,94 Two small RCTs concluded that daily doses of ≥5 mg haloperidol may reduce the incidence of 1++ delirium in surgical patients.93 Overall no optimal regime for perioperative antipsychotic use was determined from the studies. A large RCT, designed primarily to investigate the effect of prophylactic haloperidol on survival of critically ill patients, also measured delirium incidence as a secondary outcome. No difference 1++ was found between the haloperidol and placebo groups: delirium incidence (mean difference 1.5%, 95% CI -3.6% to 6.7%).95 There is insufficient evidence to determine whether antipsychotic prophylaxis is effective in other hospital inpatients.92,94,96,97 One Dutch multicentre RCT (n=242) examined the efficacy of prophylactic 1++ oral haloperidol in patients ≥70 years of age at risk of delirium, acutely admitted to hospital for 1+ 1– both medical and surgical emergencies. It concluded that haloperidol prophylaxis did not lower delirium incidence during the first seven days of admission in this patient group.98 Common side effects of antipsychotics include constipation, movement disorders, QTc prolongation, reduced seizure threshold, urinary retention and neuroleptic malignant syndrome.15 One of the secondary outcomes of the Dutch RCT was to examine haloperidol safety. No significant differences were found between oral haloperidol and placebo for changes to QTc interval.98 Similarly, three 1+ other RCTs, in a systematic review, reported no difference in QTc prolongation or extrapyramidal 1++ side effects with haloperidol prophylaxis compared to placebo.93 Otherwise, adverse effects were rarely noted, but this could be due to lack of reporting in the studies included in the systematic reviews. No antipsychotics are licensed for the prophylaxis of delirium. There is insufficient evidence of benefit to recommend the use of antipsychotic prophylaxis in patients at risk of developing delirium. 15 Risk reduction and management of delirium 5.3Dexmedetomidine Dexmedetomidine has been utilised in a perioperative and critical care setting. A meta-analysis identified 14 trials of medium to low quality, incorporating 3,029 patients in ICU (general and postoperative).99 Dexmedetomidine was compared to other therapies (propofol, midazolam or morphine) or placebo to assess reduction of the incidence of delirium, agitation and confusion. 1+ Overall, analysis was associated with a significant reduction in the incidence of delirium with dexmedetomidine versus controls, RR 0.68, 95% CI 0.49 to 0.96.99 However, not all studies showed a statistically significant benefit, and the greatest effect was evident when midazolam was used as the comparator. An RCT of 90 patients undergoing non-invasive ventilation in ICU found dexmedetomidine to be superior to haloperidol or placebo (3/30 patients given dexmedetomidine developed delirium compared to 10/30 given haloperidol and 13/30 in the placebo group; p=0.014).100 However, 1– patients in both the haloperidol and particularly the placebo group received significantly larger amounts of supplementary sedatives and analgesics including midazolam. A two-centre RCT allocated 100 critically ill patients without a diagnosis of delirium to receive either a nocturnal dexmedetomidine infusion or placebo. Sedatives were halved during the drug infusion period and opiates were unchanged. Nocturnal dexmedetomidine was associated with 1++ a greater proportion of patients who remained delirium free during their ICU stay (80% v 54%); RR 0.44, 95% CI 0.23 to 0.82.101 Results of a trial on the use of dexmedetomidine in patients who are mechanically ventilated in a general ICU setting (Early Goal Directed Sedation Compared with Standard Care in Mechanically Ventilated Patients in Intensive Care (SPICE III)) are awaited. A systematic review and meta-analysis of 18 RCTs with 3,309 patients, assessed the efficacy of perioperative dexmedetomidine on delirium incidence in both adult cardiac and non-cardiac surgical patients.102 Findings indicated dexmedetomidine reduced postoperative delirium for the entire surgical population (OR 0.35; 95% CI 0.24 to 0.51). Subgroup analyses of the nine cardiac surgery studies and nine other surgical studies showed a benefit in both patient groups (OR 0.41; 95% CI 0.26 to 0.63) and (OR 0.33; CI 0.18 to 0.59) respectively. Studies were heterogeneous in their size, dose and timing of dexmedetomidine administration and in particular the control drug given. Propofol, midazolam and different opiates were used as comparators, in addition to placebo. 1++ Trials comparing dexmedetomidine directly with placebo were not analysed separately. The largest study included in the review was in a Chinese population, involving 700 patients given either dexmedetomidine or placebo postoperatively on arrival in ICU. Delirium was significantly lower in the group receiving dexmedotimidine (OR 0.35, 95% CI 0.22 to 0.54). However, patients who were mechanically ventilated were sedated with propofol or midazolam to achieve a set RASS. Patients in the placebo group received more supplemental sedatives and analgesics compared to the dexmedetomidine group, which may have increased their delirium risk.102 Two randomised placebo-controlled trials, one each in patients undergoing cardiac and non- cardiac surgery, showed no reduction in delirium when comparing dexmedetomidine with normal 1++ saline.103,104 Bradycardia and hypotension are known side effects of dexmedetomidine, secondary to its intrinsic effects as an alpha2-receptor agonist. Dexmedetomidine has been associated with an increased incidence of bradycardia in patients undergoing cardiac surgery (OR 1.89; 95% CI 1.11 to 3.2) but this did not appear to be an issue in other surgical groups.102 Neither was there consistent evidence 1++ linking hypotension to dexmedetomidine. Caution should be taken when considering the use of dexmedetomidine, particularly in patients with low cardiac output states and consideration given to either omitting or using a lower loading dose prior to starting a continuous infusion.105 16 5 | Pharmacological risk reduction There remains controversy over whether dexmedetomidine can reduce the incidence of delirium in both critically ill patients and those in the perioperative setting. Many of the trials which indicate a benefit have used other sedative agents, including benzodiazepines in the control group. It remains unclear if dexmedetomidine can inherently reduce delirium or merely reduce the need for delirogenic drugs. Since there are potential physiological concerns relating to the widespread adoption of dexmedetomidine for prophylaxis, in addition to cost implications, dexmedetomidine cannot be recommended for the prevention of delirium. Large randomised placebo-controlled trials and cost-effectiveness studies are warranted. 5.4 Other pharmacological therapies One systematic review included four RCTs which reported the incidence of postoperative delirium when comparing ketamine to placebo. Overall, the incidence of postoperative delirium did not differ between the control and intervention groups. The quality of each RCT was low, so the review 1++ concluded that the effect of ketamine on postoperative delirium is unclear. The largest RCT reported an increase in postoperative hallucinations and nightmares with ketamine use.106 Systematic reviews identified four RCTs on the use of melatonin to prevent delirium in medical and 1++ surgical settings.63,107,108 Results were inconclusive. One small RCT in 88 patients found a melatonin 1+ receptor agonist reduced the incidence (24.4% v 46.5%) and duration (0.78 v 1.4 days) of delirium 2+ in critically ill patients.109 Results from two large RCT are awaited. 17 Risk reduction and management of delirium 6 Non-pharmacological treatment Other guidelines, narrative reviews and expert opinion on the treatment of patients with established delirium focus mainly on treating the presumed causes of the delirium, and other aspects of care such as treating distress and agitation.1,58,60,110 Few trials have been conducted testing such approaches. There is insufficient high-quality evidence to determine the efficacy of formal packages 4 of non-pharmacological interventions in reducing the severity or duration of delirium when 2++ 1++ it does occur.58,61 Meta-analyses did not find a significant difference in the reduction of duration of delirium with multicomponent care or comprehensive geriatric care, compared to usual care.62,65 One RCT did not find benefit from the use of cognitive-stimulating interventions in patients with delirium superimposed on dementia.111 Therefore, guidance on treatment of people with delirium relies on expert consensus, which advocates multicomponent interventions as fundamental good practice.58,67,68 In Scotland a comprehensive pathway, incorporating the Triggers, Investigate, Manage, Engage (TIME) bundle, which covers the first two hours of care, and the Scottish Delirium Association (SDA) delirium 4 management pathway provide protocols for good care (see Annexes 3 and 4). NICE recommends treating the causes, effectively communicating with the patient, providing a suitable care environment, and specifically addressing distress.58 RHealthcare professionals should follow established pathways of good care to manage patients with delirium. • F irst consider acute, life-threatening causes of delirium, including low oxygen level, low blood pressure, low glucose level, and drug intoxication or withdrawal. ystematically identify and treat potential causes (medications, acute illness, etc), noting • S that multiple causes are common. ptimise physiology, management of concurrent conditions, environment (reduce noise), • O medications, and natural sleep, to promote brain recovery. • S pecifically detect, assess causes of, and treat agitation and/or distress, using non- pharmacological means only if possible (see section 7 for pharmacological treatment). • C ommunicate the diagnosis to patients and carers, encourage involvement of carers, and provide ongoing engagement and support. • A im to prevent complications of delirium such as immobility, falls, pressure sores, dehydration, malnourishment, isolation. • Monitor for recovery and consider specialist referral if not recovering. • Consider follow up (see section 8). 99 Promote cognitive engagement, mobilisation, and other rehabilitation strategies. Advice on information to provide to family and carers, and suggestions of ways to help are in section 9. 18 7 | Pharmacological treatment 7 Pharmacological treatment 7.1Antipsychotics Studies of the efficacy of antipsychotics are heterogenous and inconclusive. Most are small and rated as low or very low quality.92,96,112,113 One meta-analysis concluded that antipsychotics should not be used in non-ICU settings for the treatment of patients with delirium, while another concluded that antipsychotics were superior to placebo or usual care in reducing delirium severity scale 2++ scores.92,112 A Cochrane review concluded that antipsychotics did not reduce delirium severity, 1+ resolve symptoms or alter mortality in the acute care setting.113 The Cochrane review also identified a large RCT of patients receiving palliative cancer care, which found that patients treated with either risperidone or haloperidol had worse delirium symptom scores than those receiving placebo.113 Comparisons of haloperidol and other antipsychotics did not find any antipsychotic to be more 1+ effective than another.96,112,114 Two RCTs comparing the efficacy of haloperidol and quetiapine 2++ reported conflicting results.112,115 No serious side effects were reported in the studies of haloperidol and overall adverse effects were poorly or rarely reported.94,96 Haloperidol was associated with a higher incidence of extrapyramidal side effects and dystonias than second generation antipsychotics, although this may be due to the high dose of haloperidol used in the trials.112,114 The Cochrane review concluded that extrapyramidal 2++ symptoms were not more frequent with antipsychotics compared to non-antipsychotics and there 2+ was no difference between typical and atypical antipsychotics.94 Haloperidol is contraindicated in combination with any drug that is associated with QTc prolongation.116 If it is used with other QT prolonging drugs, treatment is rendered unlicensed. Advise on prescribing unlicensed medicines can be found in section 1.3.2. If commenced, antipsychotics prescribed for delirium should be reviewed on a daily basis and stopped as soon as the clinical situation allows, typically within 1–2 days. In situations where it is deemed safer to continue antipsychotic therapy for delirium beyond discharge or transfer from hospital, a clear plan for early medication review and follow up in the community should be agreed. Pooled subgroup analysis of two small trials of patients in ICU with delirium found use of antipsychotics to be marginally superior to placebo in response rate at the studies’ endpoint (risk ratio 0.25, 95% CI 0.06 to 1.02). Second generation antipsychotics were superior to haloperidol in 1++ reducing delirium severity scores in patients in ICU (standardised mean difference (SMD) -0.52, 95% CI -0.85 to -0.19). There was no difference in discontinuation rates or adverse events.112 A systematic review identified five studies, one of which reported that quetiapine reduced the duration of delirium (1 day v 4.5 days) compared to placebo in 36 patients.97 None of the studies 1+ reported a reduction in length of stay, or mortality. Because the studies identified are underpowered, further, larger trials are needed before recommendations can be made on the use of antipsychotics for the treatment of patients in ICU with delirium. 7.2Dexmedetomidine A small RCT on the use of dexmedetomidine in patients with agitated delirium receiving mechanical ventilation in ICU reported secondary outcomes of a reduction in delirium (23.3 hours v 40 hours 1+ with placebo) and reduced the length of ICU stay.117 19 Risk reduction and management of delirium 7.3 Acetylcholinesterase inhibitors One systematic review included six small RCTs, comparing either donezepil or rivastigmine to placebo in patients older than 60 years. Five of the trials were in surgical settings. Four of the seven studies found no benefit from acetylcholinesterase inhibitors and overall the studies were not 1+ sufficiently powered to detect a difference between the intervention medication and placebo. Four of the seven studies found acetylcholinesterase inhibitors to have similar tolerability to placebo.118 A systematic review identified one RCT (104 participants) which reported longer duration of delirium and longer length of hospital stay in patients with delirium in ICU given a combination of haloperidol and rivastigmine compared to those given haloperidol and placebo.118 There 1+ were three times as many deaths among patients receiving the haloperidol and rivastigmine combination.118 There is insufficient evidence to draw conclusions on the efficacy and safety of the use of acetylcholinesterase inhibitors for the treatment of patients with delirium. 7.4Benzodiazepines In a systematic review only one small trial (n=30) was identified on the use of lorazepam in the treatment of patients with delirium. The trial, in patients with acquired immune deficiency syndrome 2+ (AIDS) in a hospital setting, found no benefit from lorazepam and treatment was stopped early due to intolerable side effects.114 7.5 Urgent pharmacological intervention While the evidence for pharmacological treatment is insufficient to support a recommendation, expert opinion supports a role for medication in specific situations such as in patients in intractable distress, and where the safety of the patient and others is compromised (see Annex 4). 20 8 | Follow up 8 Follow up Older patients who develop delirium may have undiagnosed underlying dementia or mild cognitive impairment.119,120 Delirium is also associated with an increased rate of cognitive decline after the 3 episode of delirium.119,120 The majority of studies identified found that delirium is a risk factor for 2++ future cognitive decline.121-124 Longer duration of delirium has been linked to worse global cognition at three and 12 months’ follow up.122 A systematic review of non-comparative prospective studies concluded that people may develop depression after experiencing delirium.125 The length of time before people experience depression post-delirium in ICU varied between studies, with some reporting no association between delirium and depression at three months, but higher rates of depression and worse mental health status at 2+ 12 months, and others reporting depression at three, four, six and 12 months.125,126 Other studies did 3 not find a significant association between delirium, post-traumatic stress disorder (PTSD), anxiety or depression.127-129 In these studies the patient groups were younger (mean ages 42, 61 and 62 compared to mean age >80 years in the majority of studies in the systematic review).125,127-129 The studies addressed a variety of population groups, in acute and ICU settings, and used different measures for delirium, mental and cognitive impairment and depression. RHealthcare professionals should be aware that older people may have pre-existing cognitive impairment which may have been undetected, or exacerbated in the context of delirium. Appropriate cognitive and functional assessment should be considered. Timing of this assessment must take into account persistent delirium. RIn patients who have experienced delirium in ICU consideration should be given to follow up for psychological sequelae including cognitive impairment. 99 atient records should be coded to highlight a previous episode of delirium so that hospital P staff are aware of the increased risk on readmission. 99 Ensure that delirium is noted in the discharge letter for the primary care team. 99 All patients who have had delirium should be reviewed by the primary care team. 21 Risk reduction and management of delirium 9 Provision of information This section reflects the issues likely to be of most concern to patients and their carers. These points are provided for use by health professionals when discussing delirium with patients and carers and in guiding the development of locally-produced information materials. 9.1 Checklist for provision of information This section gives examples of the information patients/carers may find helpful at the key stages of the patient journey. The checklist was designed by members of the guideline development group based on their experience and their understanding of the evidence base. The checklist is neither exhaustive nor exclusive. If a patient is at risk of delirium Identify the family and/or main carer of the patient. •Ensure that the patient’s contact details are on file. If the patient lacks capacity, ascertain whether a family member or carer has Power of Attorney/Guardianship over welfare. Explain to the patient and the family/carer about delirium: • Delirium is common amongst hospitalised patients especially following an operation. • Acute triggers of delirium include: - infection, dehydration, severe constipation, urinary retention, and pain - critical illness - surgery especially heart and hip operations - side effects of new medicines or medicines withdrawal. • Those most at risk are: - older people - older people on multiple medicines -people with dementia, Parkinson's disease, stroke or pre-existing cognitive impairment - people who are hearing or visually impaired. Ask family/carers to alert medical staff if they notice any change to their relative's normal behaviour. Ask the patient and family/carers to complete a ‘Getting to know me’ form (see section 9.3.2), or similar, to help healthcare staff to take care of the person’s specific needs. Ask family/carers to help, if they feel able to do so, to reduce the risk of delirium developing by doing the following: • ensure hearing aids, glasses and dentures are available at all times • talk to and keep the patient informed in short, simple sentences • check that the patient has understood you and be prepared to repeat if necessary • keep a calendar and/or clock within view • bring in some familiar objects from home to the hospital to keep next to the bed side • if required, encourage the patient to eat and drink. 22 9 | Provision of information If a patient develops delirium Explain to the patient and family/carers that delirium is a change in mental state that often starts suddenly but usually improves when the physical condition improves and the underlying cause gets better. Discuss treatment options and possible side effects with the patient and/or carer. Provide the family/carer with appropriate information leaflets. It is important for carers and relatives to participate and work together with the clinical team in hospital or home to clear delirium and give the affected person the best chance of getting back to good health. Explain that the person affected with delirium may show many different types of change. The patient may: • be less aware of their surroundings • be unable to speak clearly or follow conversations • have dreams which can sometimes be frightening and can carry on when they wake up • hear voices or noises which may not be present (auditory hallucinations) • see objects or people that are not present or in different context (visual hallucinations) • get upset that other people are trying to harm them • be agitated or restless, unable to sit still, and have an increased risk of having a fall • be sleepy and slow to move and respond • be reluctant to eat or drink • have a temporary change in personality • have all or some of the above and that could quickly change • have worse symptoms in the evenings or overnight. Suggest completing a diary so that if the person with delirium cannot remember what has happened the carer can fill in the blanks and help make sense of the experience once the person is starting to feel better. Let the family/carer know how to help someone with delirium: They can help by reassuring and reorienting the patient, eg: • ensure hearing aids, glasses and dentures are available at all times • have a gentle and friendly approach, smiling and providing reassurance • talk and keep the patient informed in short, simple sentences • check that the patient has understood you and be prepared to repeat if necessary • familiarity helps, so try to make sure that someone the patient knows well is with them • try not to agree with any incorrect ideas but disagree with tact and change the subject • keep a calendar and/or clock within view and give reminders of the surroundings. • bring in some familiar objects from home to the hospital to keep next to the bed side • remind the patient to eat and drink and assist if required. The key is to remain calm and help the affected person feel calm and in control. At discharge following an acute episode of delirium Liaise with the family/carers regarding discharge arrangements. Discuss with family/carers whether they need extra support. Some patients may still be recovering, not be entirely themselves or be less able than usual to carry out their daily activities. Inform carers of their right to have a new or updated adult carer support plan. Ensure that support is in place before the patient is discharged to their home. If there are concerns about cognitive impairment in the following months, advise the patient/carers to see their general practitioner (GP). 23 Risk reduction and management of delirium 9.2 Publications from SIGN SIGN patient versions of guidelines are documents that ‘translate’ guideline recommendations and their rationales, originally developed for healthcare professionals, into a form that is more easily understood and used by patients and the public. They are intended to: • h elp patients and carers understand what the latest evidence supports around diagnosis, treatment and self care • e mpower patients to participate fully in decisions around management of their condition in discussion with healthcare professionals • highlight for patients where there are areas of uncertainty. A copy of the patient version of this guideline is available from www.sign.ac.uk/assets/patient- publications 9.3 Sources of further information 9.3.1Websites Scottish Delirium Association www.scottishdeliriumassociation.com The Scottish Delirium Association consists of healthcare professionals working to share best practice in delirium by providing education, promoting research and raising awareness of the condition. Critical Care Recovery www.criticalcarerecovery.com This is a website developed by the NHS to offer information, advice and support on recovery after intensive care. Minded for Families www.mindedforfamilies.org.uk This is a website with videos and information to raise awareness of what delirium is and help older people and those around them with how to deal with it. 9.3.2 Information leaflets THINK Delirium www.knowledge.scot.nhs.uk/media/CLT/ResourceUploads/4052742/20141007%20Delirium%20 leaflet%20(web).pdf This is patient information leaflet developed by Healthcare Improvement Scotland in collaboration with NHS boards. Alzheimers Scotland www.alzheimers.org.uk/info/20029/daily_living/370/delirium This is a patient information leaflet on the effects of delirium, treatment and what happens afterwards. It also includes patient experiences. www.alzscot.org/information_and_resources/information_sheet/3472_getting_to_know_me The ‘Getting to Know Me’ form was designed for patients with dementia. It is completed with information about the person’s likes and dislikes and needs and is held with the patient notes to allow staff to provide the best care possible. 24 9 | Provision of information Dementia UK www.dementiauk.org/delirium This web-based leaflet describes the symptoms of delirium and gives suggestions of what carers can do to help a person with delirium. Marie Curie www.mariecurie.org.uk/professionals/palliative-care-knowledge-zone/symptom-control/delirium This web-based leaflet focuses on delirium occurring towards the end of life. It describes causes, and offers advice on what carers can do and when to seek expert help. NHS website www.nhs.uk/conditions/confusion This leaflet provides advice on when to contact a GP or phone an ambulance if someone is showing signs of delirium. Royal College of Psychiatrists www.rcpsych.ac.uk/mental-health/problems-disorders/delirium This leaflet provides information on signs and symptoms, treatment and what may happen after a person has had delirium. 9.3.3 Telephone helplines Alzheimers Scotland 0808 808 3000 helpline@alzscot.org This helpline provides information and emotional support to people with dementia, their families, friends and professionals. Dementia UK 0800 888 6678 This helpline is available to support to carers or anyone with dementia. 25 Risk reduction and management of delirium 10 Implementing the guideline This section provides advice on the resource implications associated with implementing the key clinical recommendations, and advice on audit as a tool to aid implementation. 10.1 Implementation strategy Implementation of national clinical guidelines is the responsibility of each NHS board and care provider and is an essential part of clinical governance. Mechanisms should be in place to educate staff and review care provided against the guideline recommendations. The reasons for any differences should be assessed and addressed where appropriate. Local arrangements should then be made to implement the national guideline in individual hospitals, units and practices. Implementation of this guideline will be encouraged and supported by SIGN. 10.2 Resource implications of key recommendations No recommendations are considered likely to reach the £5 million threshold which warrants resource impact analysis. 10.3 Auditing current practice A first step in implementing a clinical practice guideline is to gain an understanding of current clinical practice. Audit tools designed around guideline recommendations can assist in this process. Audit tools should be comprehensive but not time consuming to use. Successful implementation and audit of guideline recommendations requires good communication between staff and multidisciplinary team working. To assist with the implementation of this guideline the guideline development group has identified the following as key points to audit: The percentage of: • at-risk patients assessed using the 4AT tool • p atients in a critical care environment assessed using CAM-ICU or ICDSC (taking account of appropriate sedation level) • p atients with confirmed delirium who are recorded and coded with delirium, and the diagnosis is included in discharge summaries to the GP • patients who have medication review and medications stopped as a result • patients followed up by the primary care team after experiencing delirium • operations compliant with depth of anaesthesia monitoring. The quality of care for older patients with delirium can be measured against the Healthcare Improvement Scotland Care of Older People in Hospital standards.130 10.4 Health technology assessment advice for NHSScotland In May 2012 the SMC accepted dexmedetomidine hydrochloride for sedation in adult patients in intensive care unit requiring a sedation level not deeper than arousal in response to verbal stimulation (corresponding to RASS 0 to -3). 26 11 | The evidence base 11 The evidence base 11.1 Systematic literature review The evidence base for this guideline was synthesised in accordance with SIGN methodology. A systematic review of the literature was carried out using an explicit search strategy devised by a SIGN Evidence and Information Scientist. Databases searched include Medline, Embase, Cinahl, PsycINFO and the Cochrane Library. The year range covered was 2012–2017. Internet searches were carried out on various websites including the US National Guidelines Clearinghouse. The main searches were supplemented by material identified by individual members of the development group. Each of the selected papers was evaluated by two Evidence and Information Scientists using standard SIGN methodological checklists before conclusions were considered as evidence by the guideline development group. The search strategies are available on the SIGN website, www.sign.ac.uk 11.1.1 Literature search for patient and carer issues or concerns At the start of the guideline development process, a SIGN Evidence and Information Scientist conducted a literature search for qualitative and quantitative studies that addressed patient and carer issues of relevance to patients with delirium and their carers. Databases searched include Medline, Embase, Cinahl and PsycINFO, and the results were summarised by the SIGN Patient and Public Involvement Advisor and presented to the guideline development group. 11.1.2 Literature search for cost-effectiveness evidence The guideline development group identified key questions with potential cost-effectiveness implications, based on the following criteria, where it was judged particularly important to gain an understanding of the additional costs and benefits of different treatment strategies: • treatments which may have a significant resource impact • opportunities for significant disinvestment or resource release • the potential need for significant service redesign • cost-effectiveness evidence could aid implementation of a recommendation. A systematic literature search for economic evidence for these questions was carried out by a SIGN Evidence and Information Scientist covering the years 2012–2017. Databases searched include Medline, Embase and NHS Economic Evaluation Database (NHS EED). Each of the selected papers was evaluated by a Health Economist, and considered for clinical relevance by guideline group members. Interventions are considered to be cost effective if they fall below the commonly-accepted UK threshold of £20,000 per Quality-Adjusted Life Year (QALY). 27 Risk reduction and management of delirium 11.2 Recommendations for research The guideline development group was not able to identify sufficient evidence to answer all of the key questions asked in this guideline (see Annex 1). The following areas for further research have been identified. • R esearch into detection and management of people with delirium in primary care, community and long-term care settings. • V alidation of tools for routine monitoring of patients with delirium with clarification of the frequency of using these tools and their impact on outcomes and cost effectiveness. • S tudies of the practicalities and diagnostic yield of performing EEG in adults presenting with delirium. • R CTs on the efficacy of depth of anaesthesia monitoring in reducing postoperative delirium in patients with dementia undergoing surgery and patients undergoing emergency surgery or trauma orthopaedic surgery. • T rials of multicomponent interventions for the treatment of patients with delirium in general hospital settings. • L arge multicentre trials detailing a package of non-pharmacological interventions in the ICU with evidence of implementation. • R CTs on the efficacy and safety of antipsychotics in reducing the risk of delirium in patients in ICU or other hospital settings. • R CTs on the efficacy and safety of haloperidol in the reduction of the severity and duration of delirium in non-ICU settings. • R CTs on the efficacy and safety of antipsychotics, benzodiazepines or dexmedetomidine in the reduction of the severity and duration of delirium in patients in ICU. • S tudies on the impact of follow-up clinics and community mental health support for people who have experienced delirium, in improving first-year mortality. 11.3 Review and updating This guideline was issued in 2019 and will be considered for review in three years. The review history, and any updates to the guideline in the interim period, will be noted in the review report, which is available in the supporting material section for this guideline on the SIGN website: www.sign.ac.uk Comments on new evidence that would update this guideline are welcome and should be sent to the SIGN Executive, Gyle Square, 1 South Gyle Crescent, Edinburgh, EH12 9EB (email: sign@sign.ac.uk). 28 12 | Development of the guideline 12 Development of the guideline 12.1Introduction SIGN is a collaborative network of clinicians, other healthcare professionals and patient organisations and is part of Healthcare Improvement Scotland. SIGN guidelines are developed by multidisciplinary groups of practising healthcare professionals using a standard methodology based on a systematic review of the evidence. Further details about SIGN and the guideline development methodology are contained in ‘SIGN 50: A Guideline Developer’s Handbook’, available at www.sign.ac.uk This guideline was developed according to the 2015 edition of SIGN 50. 12.2 The Guideline Development Group Dr Ajay Macharouthu Consultant in Liaison Psychiatry for Elderly, University (Co-Chair) Hospital Crosshouse, Kilmarnock Professor Alasdair MacLullich Professor of Geriatric Medicine, University of Edinburgh, (Co-Chair) Honorary Consultant in Geriatric Medicine, Royal Infirmary of Edinburgh Dr Anthony ByrneConsultant Physician, Forth Valley Royal Hospital, Larbert Dr Sweyn GarriochConsultant Anaesthetist, Borders General Hospital, Melrose Dr Michael GötzConsultant Psychiatrist, Forth Valley Royal Hospital, Larbert Ms Maureen Huggins Carer representative, Dumfries Dr Julie MardonConsultant in Emergency Medicine, University Hospital Crosshouse, Kilmarnock Dr Sharon MulhernConsultant Clinical Lead – Neuropsychology, Ayrshire Central Hospital, Irvine Mr Brian O’TooleHealth Economist, Healthcare Improvement Scotland Dr Gautamananda RayConsultant Physician, Royal Alexandra Hospital, Paisley Ms Daisy VE Sandeman Clinical Nurse Practitioner, Royal Infirmary of Edinburgh Mrs Lynne SmithEvidence and Information Scientist, Healthcare Improvement Scotland Dr Roy Soiza Consultant Physician, Aberdeen Royal Infirmary Ms Christine SteelAHP Consultant (Dementia), West Glasgow Ambulatory Care Hospital Ms Ailsa Stein Programme Manager, SIGN Ms Alyson WarrenLead Pharmacist, Care of the Elderly, Raigmore Hospital, Inverness Dr Elizabeth WilsonConsultant in Critical Care Medicine and Anaesthesia, Royal Infirmary of Edinburgh Dr Maria Wybrew General Practitioner, Thurso and Halkirk Medical Practice The membership of the guideline development group was confirmed following consultation with the member organisations of SIGN. All members of the guideline development group made declarations of interest. A register of interests is available in the supporting material section for this guideline at www.sign.ac.uk 29 Risk reduction and management of delirium Guideline development and literature review expertise, support and facilitation were provided by SIGN Executive and Healthcare Improvement Scotland staff. All members of the SIGN Executive make yearly declarations of interest. A register of interests is available on the contacts page of the SIGN website www.sign.ac.uk Euan Bremner Project Officer, SIGN Executive Karen GrahamPatient and Public Involvement Adviser, Healthcare Improvement Scotland Administration Officer, SIGN Executive Domenico Romano Publications Designer, SIGN Executive Gaynor Rattray Guideline Co-ordinator, SIGN Executive Dr Carolyn SleithEvidence and Information Scientist, Healthcare Improvement Scotland 12.3Acknowledgements SIGN is grateful to the following former members of the guideline development group and others who have contributed to the development of the guideline. Ms Karen MartinMental Health Development Co-ordinator, Carers Trust, Glasgow Mr James McKillop Patient representative, Glasgow Ms Rachael Wybrew Medical student, Birmingham 12.4 Consultation and peer review 12.4.1 National open meeting A national open meeting is the main consultative phase of SIGN guideline development, at which the guideline development group presents its draft recommendations for the first time. The national open meeting for this guideline was held on 21 June 2018 and was attended by 123 representatives of all the key specialties relevant to the guideline. The draft guideline was also available on the SIGN website for a limited period at this stage to allow those unable to attend the meeting to contribute to the development of the guideline. 12.4.2 Specialist review This guideline was reviewed in draft form by the following independent expert referees, who were asked to comment primarily on the comprehensiveness and accuracy of interpretation of the evidence base supporting the recommendations in the guideline. The guideline group addresses every comment made by an external reviewer, and must justify any disagreement with the reviewers’ comments. A report of the peer review comments and responses is available in the supporting material section for this guideline on the SIGN website. All expert referees made declarations of interest and further details of these are available on request from the SIGN Executive. SIGN is very grateful to all of these experts for their contribution to the guideline. Dr Nikhil Agrawal General Practitioner, Southside Road Surgery, Inverness Dr Jonathan Antrobus Consultant Anaesthetist, Borders General Hospital, Melrose Dr Janet Bennison Consultant Geriatrician, Borders General Hospital, Melrose Dr Claire Copeland Consultant Physician, Forth Valley Royal Hospital, Larbert 30 12 | Development of the guideline Dr Duncan GrayAssociate Specialist Geriatrician, Raigmore Hospital, Inverness Dr Conrad Harvey General Practitioner, Ayrshire Central Hospital, Irvine Ms Lynne IrvineAlzheimer Scotland Dementia Nurse Consultant, NHS Grampian Dr Simon McAreeConsultant in Anaesthesia and Intensive Care, Borders General Hospital, Melrose Mr Scott MurrayOlder Adults Liaison Psychiatry Nurse Specialist, Borders General Hospital, Melrose Dr Terence QuinnSenior Lecturer and Honorary Consultant Physician, University of Glasgow Dr Saif RazviConsultant Neurologist, Queen Elizabeth University Hospital, Glasgow Dr Pushkar ShahConsultant Neurologist, Queen Elizabeth University Hospital, Glasgow Ms Sandra ShieldsAlzheimer Scotland Dementia Nurse Consultant, NHS Greater Glasgow and Clyde Ms Helen Skinner Alzheimer Scotland Dementia Nurse Consultant, NHS Fife 12.4.3 SIGN editorial group As a final quality control check, the guideline is reviewed by an editorial group comprising the relevant specialty representatives on SIGN Council to ensure that the specialist reviewers’ comments have been addressed adequately and that any risk of bias in the guideline development process as a whole has been minimised. The editorial group for this guideline was as follows. All members of SIGN Council make yearly declarations of interest. A register of interests is available on the SIGN Council Membership page of the SIGN website www.sign.ac.uk Dr Jenny Bennison Royal College of General Practitioners Dr Roberta James SIGN Programme Lead; Co-Editor Professor John Kinsella Chair of SIGN; Co-Editor Dr Jane Morris Royal College of Psychiatrists Ms Jacqueline Thompson Royal College of Nursing 31 Risk reduction and management of delirium Abbreviations 4AT 4 As Test (Arousal, Attention, Abbreviated Mental Test 4, Acute change Test) AIDS acquired immune deficiency syndrome AMT Abbreviated Mental Test CAM Confusion Assessment Method CI confidence interval CRP C-reactive protein CSF cerebrospinal fluid CT computed tomography CXR chest X-ray DOS Delirium Observation Screening Scale DRS-98-R Delirium Rating Scale DSD delirium superimposed on dementia DSM-5 Diagnostic and Statistical Manual of Mental Disorders, fifth edition ECGelectrocardiogram EEGelectroencephalogram FBC full blood count GMC General Medical Council GP general practitioner ICD-10 International Classification of Diseases, version 10 ICU intensive care unit ICDSC Intensive Care Delirium Screening Checklist LFT liver function test MA marketing authorisation MMSE Mini Mental State Examination mRASS Modified Richmond Agitation-Sedation Scale NCSE non-convulsive status epilepticus NICE National Institute for Health and Care Excellence Nu-DESC Nursing Delirium Screening Scale OR odds ratio PCR polymerase chain reaction PTSD post-traumatic stress disorder 32 Abbreviations QALY quality-adjusted life year RADAR Recognising Acute Delirium As part of your Routine RASS Richmond Agitation – Sedation Scale RCT randomised controlled trial RR relative risk SDA Scottish Delirium Association SIGN Scottish Intercollegiate Guidelines Network SMC Scottish Medicines Consortium SMD standardised mean difference SPICE IIIearly goal directed sedation compared with standard care in mechanically ventilated patients in intensive care SQiD Single Question to Identify Delirium TCD transcranial doppler TIME Triggers, Investigate, Manage, Engage USUltrasound UTI urinary tract infection 33 Risk reduction and management of delirium Annex 1 Key questions used to develop the guideline This guideline is based on a series of structured key questions that define the target population, the intervention, diagnostic test, or exposure under investigation, the comparison(s) used and the outcomes used to measure efficacy, effectiveness, or risk. These questions form the basis of the systematic literature search. Section(s) Key question 3.1 1. What tool(s) should be used to detect delirium and when? Population: Adults at risk of delirium Assessment tools: a. 4 As Test (Arousal, Attention, Abbreviated Mental Test 4, Acute change Test) (4AT) b. Confusion Assessment Method Instrument (CAM) c. 3D Confusion Assessment Method Instrument (3D-CAM) d. Delirium Observation Screening Scale (DOS) e. Single Question to Identify Delirium (SQID) f. Memorial Delirium Assessment Scale (MDAS) g. Recognizing Acute Delirium As part of your Routine (RADAR) h. Delirium Rating Scale – 98 – Revised (DRS-98-R) i. Intensive Care Delirium Screening Checklist (ICD-SC) j. CAM for the intensive care unit (CAM-ICU) k. Richmond agitation sedation score (RASS); Modified RASS l. Family Confusion Assessment Method Instrument (FAM-CAM) m. Brief Confusion Assessment Method Instrument (B-CAM) n. Nursing Delirium Screening Scale (Nu-DESC) o. Organic Brain Syndrome scale (OBS) p. Mini Mental State Examination Comparison: Diagnostic and Statistical Manual (DSM-5) or International Classification of Diseases (ICD-10) defined diagnosis; between tools Outcomes: Sensitivity, specificity, evidence of adherence in clinical practical 3.2, 3.3 2.What tool(s) should be used for monitoring purposes and when should they be used? Population: Adults at risk of delirium Assessment tools: a. 4 As Test (Arousal, Attention, Abbreviated Mental Test 4, Acute change Test) (4AT) b. Confusion Assessment Method Instrument (CAM) c. 3D Confusion Assessment Method Instrument (3D-CAM) d. Delirium Observation Screening Scale (DOS) e. Single Question to Identify Delirium (SQiD) f. Memorial Delirium Assessment Scale (MDAS) g. Recognizing Acute Delirium As part of your Routine (RADAR) 34 Annexes h. Delirium Rating Scale – 98 – Revised (DRS-98-R) i. Intensive Care Delirium Screening Checklist (ICDSC) j. Confusion Assessment Method Instrument for the intensive care unit (CAM-ICU) k. Family Confusion Assessment Method Instrument (FAM-CAM) l. Confusion Assessment Method Instrument (B-CAM) m. Nursing Delirium Screening Scale (Nu-DESC) n. Organic Brain Syndrome (OBS) scale Comparison: Diagnostic and Statistical Manual (DSM-5) or International Classification of Diseases (ICD-10) defined diagnosis; between tools Outcomes: Sensitivity, specificity, evidence of adherence in clinical practical 3.4 3. What (other) investigations are useful when assessing a patient for delirium? Population: Adults with suspected delirium Intervention: a. imaging (CT or MRI scans) b. lumbar puncture c. electroencephalogram (EEG) d. testing for antibodies for autoimmune encephalitis e. toxicology screening Comparison: Usual care Outcomes: Sensitivity, specificity, cost effectiveness 4, 5 4. What risk reduction strategies for patients at risk of delirium are effective? Population: Patients at risk of developing delirium Interventions: Multicomponent interventions – non-pharmacological and pharmacological Non-pharmacological: a. proactive screening of delirium and pre-existing cognitive impairment including dementia b.hydration c. catheterisation avoidance d. sensory impairment e.constipation f. sleep hygiene and promotion g. falls prevention and mobility h. providing means of communication i. impact of ward moves (incl ‘boarding’) j. environmental factors Pharmacological: a. medication reconciliation b. pain relief c. antipsychotics and benzodiazepines (medical and surgical patients) d. sedation for night-time sleep Comparison: usual care Outcomes: Incidence of delirium (hospital acquired), prevalence of delirium (community acquired), duration of delirium, severity of delirium 35 Risk reduction and management of delirium 6 5.What are the most effective non-pharmacological strategies for managing patients with delirium? Population: people with delirium Interventions: Multicomponent non-pharmacological interventions (Staff) behavioural adaptations: • calm non-confrontational manner •reassurance •reorientation • distraction/de-escalation techniques • one-to-one nursing • cognitive stimulation Environmental adaptations: • single room • well-lit area • clear signs re: day, time, season, place • familiar objects • family input • minimise bed moves • activities and occupational therapy • address sensory impairment • sleep promotion • facilitate mobility Address specific causes of stress: •pain •hunger • feeling too hot/too cold • thirst/dry mouth • urinary retention • specific fears • not understanding what is happening • hallucinations, delusions, aggression, agitation, and wandering/searching Comparison: Usual care, pharmacological therapies Outcomes: Mortality, complete response, duration of delirium, severity of delirium, distress in delirium, length of hospital stay, loss of independent living/new institutionalisation, reduction in depression and anxiety, reduced dementia risk, worsening of dementia, reduction in long- term effects, reduction in falls, cost effectiveness 36 Annexes 7 6.What are the most effective pharmacological strategies for managing patients with delirium? Population: Patients with delirium Consider hyperactive versus hypoactive delirium. Consider subpopulations: 1. Parkinson’s disease 2. delirium superimposed on dementia 3. patients already taking long-term medication Interventions: a.antipsychotics b.benzodiazepines c. acetylcholinesterase inhibitors d.melatonin e.antidepressants f.dexmedetomidine g.clonidine h.propanolol i. withdrawal of medicines which may be causing the delirium Comparison: Usual care, between therapies Outcomes: Mortality, complete response, duration of delirium, severity of delirium, length of hospital stay, loss of independent living/new institutionalisation, increased dementia risk, worsening of dementia, adverse events, reduction in long-term effects, cost effectiveness 8 7.What follow-up care should patients receive after experiencing delirium? Population: patients who have had delirium Interventions: Screening for: a.dementia b. functional psychiatry disorders – post-traumatic stress disorder, depression Comparison: usual care Outcomes: incidence of dementia after delirium, incidence of psychiatric disorders 37 Risk reduction and management of delirium Annex 2 Investigations for underlying causes of delirium The majority of people with delirium are older adults, often with a vulnerability to delirium due to underlying neurological disease (eg dementia, cerebrovascular disease, Parkinson’s Disease). In each there is commonly more than one precipitating factor. Identifying these factors and addressing those that are modifiable underpin the treatment of a person with delirium. A good clinical history taking into account premorbid illness, cognition and level of function gives key information. However, the person at risk of delirium may not be able to provide reliable information themselves due to confusion or diminished attentiveness. A collateral history from the person’s family or carers should be obtained to confirm and supplement information provided by the person. This collateral history should be sought at the earliest opportunity. Relatives will often accompany the unwell person when initially assessed in hospital or at home. Some additional time obtaining this information at an early stage can assist rapid identification and treatment of precipitants. A full clinical examination should be undertaken including neurological examination to identify focal signs and musculoskeletal examination to look for evidence of injury. Confusion and agitation resulting in poor co-operation or understanding of instructions may make examination difficult. Severe illness should be identified and rapidly treated as an urgent priority (see Annex 3). This should include assessment of basic observations, blood oxygen saturations, and blood glucose with near-patient testing to exclude hypoglycaemia. Intoxication due to medication should be considered in every case. The information obtained from history and investigation will guide further investigation. Some investigations would be considered general and applicable to most patients, while others are targeted to specific clues from history and examination. Investigations will also depend on the setting, whether the person is in hospital or at home. These tests are commonly done but this list is not entirely comprehensive. Blood tests: • R enal function (urea and electrolytes) – to identify dehydration, acute kidney injury, chronic kidney disease, hyponatraemia. • Full blood count (FBC) – to identify anaemia, macrocystosis, elevated white cell count • C-reactive protein (CRP) – to identify inflammation/infection • L iver function tests (LFT) – can identify liver dysfunction which could identify biliary infection, malignant disease, encephalopathy • Calcium – hypercalcaemia can cause confusion, and requires further investigation • Blood cultures – where there is evidence of infection (eg fever or sepsis) • Thyroid function – thyroid dysfunction can cause confusion • Vitamin B12 and folate – consider if there are concerns about nutrition or macrocytosis on full blood count. Electrocardiogram (ECG) • this may identify clinically silent myocardial ischaemia or arrhythmia which may be significant (such as atrial fibrillation). 38 Annexes Radiological imaging • C hest X-ray (CXR) – should be done if there are symptoms or signs of chest pathology such as infection. It should be remembered that clinical examination may not reveal all pathology, such as a tumour, and should be considered. • Musculoskeletal X-rays – target where there is evidence of injury or suspicion of fracture. • Other imaging should be guided by history, examination and initial investigations. Other tests • Identify hypoxia using pulse oximetry • U rine dipstick and culture – a negative urine dipstick can be useful, in that urinary tract infection (UTI) would be less likely, but a positive dipstick does not necessarily mean infection. Asymptomatic bacteruria can also exist in the elderly and delirium may mean that the person is unable to give a history of symptoms of UTI. This may cloud the situation and treatment of suspected urinary tract infection should be based on clinical grounds and probability. • Bedside ultrasound (US) bladder scan – to identify urinary retention • Pain assessment tools. This is not a comprehensive list of tests which could be done and investigation should be targeted from information obtained initially and built on as the clinical situation evolves. Section 3 recommends investigations which are evidence based. Where there is consideration of central nervous system pathology as a cause of confusion or delirium, targeted investigations may be appropriate including brain imaging, lumbar puncture, EEG, auto-antibody testing (such as for autoimmune encephalitis – antivoltage-gated potassium channel antibodies, anti-NMDA antibodies). 39 Risk reduction and management of delirium Annex 3 TIME bundle delirium management protocol68 40 Annexes Annex 4 Scottish Delirium Association delirium management pathway68 Developed in collaboration with DELIRIUM MANAGEMENT COMPREHENSIVE PATHWAY History of Acute Change – Think Delirium Risk Factors for Delirium  Acute illness  Dementia  Age over 70 years  Frailty This pathway does  Sensory Impairment  Polypharmacy  Recent anaesthetic/surgery  Catheterised  Recent discharge from acute hospital  Use of opioids, benzodiazepines or anticholinergics NOT relate to  Restraint  Depression  History of alcohol misuse  Acute or chronic pain alcohol or substance misuse. Clinical suspicion of delirium or “local tool” positive [e.g. 4AT or CAM] If this is suspected [Screening tools can be negative in the presence of delirium – use clinical judgement] use appropriate local pathway. This pathway is appropriate for Act on acute, severe causes e.g. sepsis, hypoxia, hypoglycemia, medication intoxication adult patients (18 years & over) The clinical team should take an informant history and assess capacity to consent to treatment. Delirium is If the patient is unable to consent to treatment complete an AWI Section 47 (consent to treatment) frequently form. Treatment plan to be discussed with the patients informant/power of attorney (attach undetected. certificate to the treatment plan). This pathway is Be aware that not exhaustive  An informant should be contacted to provide information about the history of cognitive impairment and functional ability, in addition to the history of current illness. patients with Other causes of delirium  The informant should be asked to clarify and quantify alcohol intake and recent changes to delirium may exist and additional or prescribed medication, falls, hydration & nutrition and identify current social support. have paranoid alternative assessments, ideas/delusions: investigations,  If there is no informant then contact the patient’s GP/social work/carers/care home. management strategies  Use the IQCODE or AD8 to assist with informant history  Identify current social support risk assess and or therapies may be manage necessary for an appropriately. individual patient. Assess with local tool & record baseline cognitive function. Clinical judgement &  AMT4 AMT10 MOCA GPCOG  Assess memory, mood, perception, sleep patterns, thinking decisions should be made by the Do a full physical examination including detailed neurological examination, speech assessment, and level of appropriate responsible arousal. Look for local signs of sepsis (e.g. bladder, lungs, skin), constipation and consider PR exam. healthcare professional. DOCUMENT DIAGNOSIS OF DELIRIUM & SUSPECTED CAUSES; REVISE AS APPROPRIATE Medication Review Investigation Optimise Management of  Review age appropriateness Dictated by the history and examination findings Co-morbidity  Any drugs recently started/stopped?  U&E / LFT / FBC / Glucose / CRP For example;  Dose changes to medication?  Calcium / Phosphate  Respiratory disease  Compliance/concordance issues with medication?  Thyroid function  Diabetes mellitus  Carefully consider ongoing needs for:  Oxygen saturation / arterial blood gases  Cardiac disease / heart failure Opioids / benzodiazepines / antipsychotics /  ECG  Thyroid disease antispasmodics / antiepileptics / antihistamines /  Chest X-ray antihypertensives (especially if hypotension) /  Parkinson’s disease  Urinalysis / urine culture corticosteroids / tricyclic antidepressants / digoxin /  Cerebrovascular disease  Blood / sputum / stool culture as appropriate antiparkinsonian medication  CT brain if anti-coagulated (urgent), head  Avoid abrupt withdrawal of drugs with dependence injury, focal neurological signs, or persistent potential or possible discontinuationsyndrome. symptoms. Environmental & General Measures Treatment of Delirium Symptoms  Approach patient calmly and gently from the front Relax visiting times - use family to reassure and support care Hypoactive delirium is common in older patients  Sleep chart; maintain daytime wakefulness with activities AVOID Treat psychotic symptoms if distressing  Allow patients to mobilise as much as possible in an area which Consider additional staff Bed moves has been deemed safe given confusion/fallsrisk.  If patient’s symptoms threaten their safety or the safety of others Unnecessary  Ensure glasses and hearing aids are working, treat ear wax use low dose of one medication (start low – go slow method) and interventions  Ensure adequate diet taken, keep daily food & fluid charts review every 24 hours Hypoxia  Regularly reassure and re-orientate (use clocks & calendars) Consider capacity to consent to treatment (AWI Section 47) Dehydration Constipation  Ensure buzzer close to patient and respond promptly to calls Medications for unmanageable agitation/distress: Catheterisation  Listen to the patient’s expression of needs o * Haloperidol 0.5-1mg orally (max 2mg/24 hours)  Reduce noise (e.g. monitors and alarms) and background noise o * Haloperidol 0.5mg IM (max 2mg/24 hours) (* Haloperidol is  If language or hearing problems, consider an interpreter contra-indicated in combination with QTc prolonging drugs, which makes  Refer to advocacy as appropriate e.g. if patient detained it unlicensed and local “off label” policy should be followed) under Mental Health (Care and Treatment) (Scotland) Act o Or atypical antipsychotic at low dose, for example, Risperidone 0.25 mgs daily, maximum 1mg in 24 hours Do not use if signs of Parkinsonism or Lewy Body Dementia If antipsychotics are contra-indicated (as above), There are Medical & Nursing Management Lorazepam 0.5-1mg orally (max 2mg/24 hrs); Midazolam 2.5mg IM often multiple Treat underlying causes (max 7.5mg/24 hours). Younger patients may need higher drug doses  causes of  Infection/sepsis, urinary retention, constipation, hypotension, pain, delirium but dehydration, hypoxia, hypoglycaemia, hyponatraemia in up to  Ensure O2 saturation > 95% (except in COPD - type 2 respiratory failure)  Explain diagnosis to patient & carer and provide information leaflet Triggers for Referral to Liaison Psychiatry 30% of cases no cause is  Use Butterfly scheme / “Getting to know me” / “This is me” / “Forget me not”  Severe agitation or distress not responding to  Assess and monitor pain (e.g. by using the Abbey Pain scale or similar) standard measures above found  Consider if swallow safe  Doubt about diagnosis  If detention under the Mental Health Act is being considered Psychiatric services may also hold useful information Patient Improving Repeat delirium screening when on background cognition and mental health.  Reduce and discontinue antipsychotic treatment clinically indicated until two  Repeat cognitive assessment successive daily negatives.  Consider post-delirium distress (eg. recall of delusional states) Improvement may also be seen with Patient NOT Improving  Encourage patients to share their experience with healthcare staff improving cognition or sleep pattern. After one week or if severe delirium, refer to the appropriate local specialist Ongoing Cognitive Impairment No Ongoing Cognitive Impairment Delirium can persist for weeks or  Document diagnosis of delirium on discharge letter to GP  Document diagnosis of delirium on discharge letter to GP months after the cause is treated  High risk of recurrent delirium requiring prompt treatment  High risk of recurrent delirium requiring prompt treatment  Follow Cognitive Impairment Pathway  Increased risk of dementia in the future in older people Version 1.03 FINAL – Oct 2018; Review by Oct 2020 41 Risk reduction and management of delirium References 16General Medical Council (GMC). Good practice in prescribing and managing medicines and devices. [cited 29 Nov 2018]. Available from url: https://www.gmc-uk.org/ethical-guidance/ ethical-guidance-for-doctors/prescribing-and-managing- medicines-and-devices 1Marcantonio ER. Delirium in hospitalized older adults. N Engl J Med 2017;377(15):1456-66. 17Medicines and Healthcare products Regulatory Agency. Off-label or unlicensed use of medicines: prescribers' 2Partridge JS, Martin FC, Harari D, Dhesi JK. The delirium responsibilities. Drug safety update 2009;2(9):6-7. experience: what is the effect on patients, relatives and staff and what can be done to modify this? Int J Geriatr 18LaMantia MA, Messina FC, Hobgood CD, Miller DK. Screening Psychiatry 2013;28(8):804-12. for delirium in the emergency department: a systematic review. Annals of Emergency Medicine 2014;63(5):551-60.e2. 3Tieges Z, Evans JJ, Neufeld KJ, MacLullich AM. The neuropsychology of delirium: advancing the science 19Neto AS, Nassar AP, Jr., Cardoso SO, Manetta JA, Pereira VGM, of delirium assessment . Int J Geriatr Psychiatry Esposito DC, et al. Delirium screening in critically ill patients: 2018;33(11):1501-11. a systematic review and meta-analysis. Critical Care Medicine 2012;40(6):1946-51. 4Cole MG, Bailey R, Bonnycastle M, McCusker J, Fung S, Ciampi A, et al. Partial and no recovery from delirium in older 20Hendry K, Quinn TJ, Evans J, Scortichini V, Miller H, Burns J, et hospitalized adults: frequency and baseline risk factors. J al. Evaluation of delirium screening tools in geriatric medical Am Geriatr Soc 2015;63(11):2340-8. inpatients: a diagnostic test accuracy study. Age & Ageing 2016;45(6):832-7. 5Pendlebury ST, Lovett NG, Smith SC, Dutta N, Bendon C, Lloyd-Lavery A, et al. Observational, longitudinal study of 21Han JH, Wilson A, Graves AJ, Shintani A, Schnelle JF, Ely delirium in consecutive unselected acute medical admissions: EW. A quick and easy delirium assessment for nonphysician age-specific rates and associated factors, mortality and re- research personnel. American Journal of Emergency Medicine admission. BMJ Open 2015;5(11):e007808. 2016;34(6):1031-6. 6Persico I, Cesari M, Morandi A, Haas J, Mazzola P, Zambon 22Shi Q, Warren L, Saposnik G, MacDermid JC. Confusion A, et al. Frailty and delirium in older adults: a systematic assessment method: a systematic review and meta-analysis of review and meta-analysis of the literature. J Am Geriatr Soc diagnostic accuracy. Neuropsychiatric Disease and Treatment 2018;66(10):2022-30. 2013;9:1359-70. 7Harrison JK, Garrido AG, Rhynas SJ, Logan G, MacLullich AM, 23Nishimura K, Yokoyama K, Yamauchi N, Koizumi M, Harasawa MacArthur J, et al. New institutionalisation following acute N, Yasuda T, et al. Sensitivity and specificity of the Confusion hospital admission: a retrospective cohort study. Age Ageing Assessment Method for the Intensive Care Unit (CAM-ICU) and 2017;46(2):238-44. the Intensive Care Delirium Screening Checklist (ICDSC) for detecting post-cardiac surgery delirium: a single-center study 8Bond P, Goudie K. Identifying and managing patients in Japan. Heart & Lung 2016;45(1):15-20. with delirium in acute care settings. Nurs Older People 2015;27(9):28-32. 24De J, Wand AP. Delirium screening: a systematic review of delirium screening tools in hospitalized patients. The 9Bull MJ, Boaz L, Jerme M. Educating family caregivers for Gerontologist 2015;55(6):1079-99. older adults about delirium: a systematic review. Worldviews on Evidence-Based Nursing 2016;13(3):232-40. 25De J, Wand AP, Smerdely PI, Hunt GE. Validating the 4A's test in screening for delirium in a culturally diverse geriatric inpatient 10Day J, Higgins I. Adult family member experiences during population. International Journal of Geriatric Psychiatry. an older loved one's delirium: a narrative literature review. 2017;32(12):1322-9. Journal of Clinical Nursing 2015;24(11-12):1447-56. 26Lees R, Corbet S, Johnston C, Moffitt E, Shaw G, Quinn TJ. Test 11Dewar B, Bond P, Millar M, Goudie K. Staff, patients and accuracy of short screening tests for diagnosis of delirium or families experiences of giving and receiving care during cognitive impairment in an acute stroke unit setting. Stroke an episode of delirium in an acute hospital care setting. 2013;44(11):3078-83. Edinburgh: Healthcare Improvement Scotland; 2013. 27O'Sullivan D, Brady N, Manning E, O'Shea E, O'Grady S, O'Regan 12World Health Organization (WHO). The ICD-10 Classification N, et al. Validation of the 6-Item cognitive impairment test and of Mental and Behavioural Disorder. Diagnostic criteria for the 4AT test for combined delirium and dementia screening research. [cited 29 Nov 2018]. Available from url: https:// in older emergency department attendees. Age Ageing www.who.int/classifications/icd/en/GRNBOOK.pdf 2018;47(1):61-8. 13American Psychiatric Association. Diagnostic and Statistical 28Detroyer E, Clement PM, Baeten N, Pennemans M, Decruyenaere Manual of Mental Disorders. Fifth edition. Arlington, VA: M, Vandenberghe J, et al. Detection of delirium in palliative American Psychiatric Association; 2013. care unit patients: a prospective descriptive study of the 14Cole MG. Persistent delirium in older hospital patients. Curr delirium observation screening scale administered by bedside Opin Psychiatry 2010;23(3):250-4. nurses. Palliative Medicine 2014;28(1):79-86. 15BMJ Group and the Royal Pharmaceutical Society. British 29Gavinski K, Carnahan R, Weckmann M. Validation of the National Formulary (BNF): Guidance on Prescribing. delirium observation screening scale in a hospitalized older [cited 29 Nov 2018]. Available from url: https://www. population. Journal of Hospital Medicine. 2016;11(7):494-7. medicinescomplete.com/mc/bnf/current/PHP97234- 30van Velthuijsen EL, Zwakhalen SMG, Warnier RMJ, Mulder guidance-on-prescribing.htm WJ, Verhey FRJ, Kempen GIJM. Psychometric properties and feasibility of instruments for the detection of delirium in older hospitalized patients: a systematic review. International Journal of Geriatric Psychiatry 2016;31(9):974-89. 42 References 31Gusmao-Flores D, Salluh JI, Chalhub RT, Quarantini LC. The 45Hardy JE, Brennan N. Computerized tomography of the brain confusion assessment method for the intensive care unit (CAM- for elderly patients presenting to the emergency department ICU) and intensive care delirium screening checklist (ICDSC) with acute confusion. Emergency Medicine Australasia for the diagnosis of delirium: a systematic review and meta- 2008;20(5):420-4. analysis of clinical studies. Critical Care 2012;16 (4):R115. 46Lai MMY, Wong Tin Niam DM. Intracranial cause of delirium: 32Infante MT, Pardini M, Balestrino M, Finocchi C, Malfatto computed tomography yield and predictive factors. Internal L, Bellelli G, et al. Delirium in the acute phase after stroke: Medicine Journal 2012;42(4):422-7. comparison between methods of detection. Neurological 47Naughton BJ, Moran M, Ghaly Y, Michalakes C. Computed Sciences 2017;38(6):1101-4. tomography scanning and delirium in elder patients. Academic 33Morandi A, McCurley J, Vasilevskis EE, Fick DM, Bellelli G, Lee Emergency Medicine 1997;4(12):1107-10. P, et al. Tools to detect delirium superimposed on dementia: a 48Vijayakrishnan R, Ramasubramanian A, Dhand S. Utility of systematic review. Journal of the American Geriatrics Society head CT scan for acute inpatient delirium. Hospital Topics 2012;60(11):2005-13. 2015;93(1):9-12. 34Sepulveda E, Franco JG, Trzepacz PT, Gaviria AM, Vinuelas 49Johnson MH. Assessing confused patients. J Neurol Neurosurg E, Palma J, et al. Performance of the delirium rating scale- Psychiatry 2001;71 Suppl 1:i7-12. revised-98 against different delirium diagnostic criteria in a population with a high prevalence of dementia. Psychosomatics 50Scottish Intercollegiate Guidelines Network (SIGN). Early 2015;56(5):530-41. management of patients with a head injury. Edinburgh: SIGN; 2009. (SIGN publication no. 110). [cited 29 Nov 2018]. 35Whittamore KH, Goldberg SE, Gladman JRF, Bradshaw LE, Available from url: https://www.sign.ac.uk/sign-110-early- Jones RG, Harwood RH. The diagnosis, prevalence and outcome management-of-patients-with-a-head-injury.html of delirium in a cohort of older people with mental health problems on general hospital wards. International Journal of 51Koponen H, Hurri L, Stenback U, Mattila E, Soininen H, Riekkinen Geriatric Psychiatry 2014;29(1):32-40. PJ. Computed tomography findings in delirium. Journal of Nervous & Mental Disease 1989;177(4):226-31. 36Mitchell AJ, Shukla D, Ajumal HA, Stubbs B, Tahir TA. The Mini- Mental State Examination as a diagnostic and screening test 52Naeije G, Depondt C, Meeus C, Korpak K, Pepersack T, Legros B. for delirium: systematic review and meta-analysis. General EEG patterns compatible with nonconvulsive status epilepticus Hospital Psychiatry 2014;36(6):627-33. are common in elderly patients with delirium: a prospective study with continuous EEG monitoring. Epilepsy & Behavior 37Voyer P, Champoux N, Desrosiers J, Landreville P, McCusker 2014;36:18-21. J, Monette J, et al. Recognizing acute delirium as part of your routine [RADAR]: a validation study. BMC Nursing 2015;14:19. 53Veran O, Kahane P, Thomas P, Hamelin S, Sabourdy C, Vercueil L. De novo epileptic confusion in the elderly: a 1-year prospective 38Grossmann FF, Hasemann W, Kressig RW, Bingisser R, study. Epilepsia 2010;51(6):1030-5. Nickel CH. Performance of the modified Richmond Agitation Sedation Scale in identifying delirium in older emergency 54Warshaw G, Tanzer F. The effectiveness of lumbar puncture in department patients. American Journal of Emergency the evaluation of delirium and fever in the hospitalized elderly. Medicine. 2017;35(9):1324-6. Archives of Family Medicine 1993;2(3):293-7. 39Vasunilashorn SM, Guess J, Ngo L, Fick D, Jones RN, Schmitt EM, 55Engelborghs S, Niemantsverdriet E, Struyfs H, Blennow K, et al. Derivation and validation of a severity scoring method Brouns R, Comabella M, et al. Consensus guidelines for lumbar for the 3-minute diagnostic interview for confusion assessment puncture in patients with neurological diseases. Alzheimers method-defined delirium. Journal of the American Geriatrics Dement (Amst) 2017;8:111-26. Society 2016;64(8):1684-9. 56Siddiqi N, House AO, Holmes JD. Occurrence and outcome of 40Vasunilashorn SM, Marcantonio ER, Gou Y, Pisani MA, Travison delirium in medical in-patients: a systematic literature review. TG, Schmitt EM, et al. Quantifying the severity of a delirium Age Ageing 2006;35(4):350-64. episode throughout hospitalization: the combined importance of intensity and duration. Journal of General Internal Medicine 57National Institute for Health and Clinical Excellence (NICE). 2016;31(10):1164-71. Delirium in adults. London: NICE; 2014. (NICE quality standard QS63). [cited 29 Nov 2018]. Available from url: https://www. 41Laurila JV, Laakkonen ML, Tilvis RS, Pitkala KH. Predisposing nice.org.uk/guidance/qs63 and precipitating factors for delirium in a frail geriatric population. J Psychosom Res 2008;65(3):249-54. 58National Institute for Health and Clinical Excellence (NICE). Delirium: diagnosis, prevention and management. London: 42Royal College of Physicians. Acute care toolkit 3: Acute medical NICE; 2010. (NICE guideline CG103). [cited 29 Nov 2018]. care for frail older people. [cited 29 Nov 2018]. Available from Available from url: https://www.nice.org.uk/guidance/cg103 url: https://www.rcplondon.ac.uk/guidelines-policy/acute-care- toolkit-3-acute-medical-care-frail-older-people 59Aldecoa C, Bettelli G, Bilotta F, Sanders RD, Audisio R, Borozdina A, et al. European Society of Anaesthesiology evidence-based 43Theisen-Toupal J, Breu AC, Mattison MLP, Arnaout R. Diagnostic and consensus-based guideline on postoperative delirium. Eur yield of head computed tomography for the hospitalized J Anaesthesiol 2017;34(4):192-214. medical patient with delirium. Journal of Hospital Medicine (Online) 2014;9(8):497-501. 60Australian Commission on Safety and Quality in Health Care. Delirium Clinical Care Standard. Sydney: ACSQHC; 44Hufschmidt A, Shabarin V. Diagnostic yield of cerebral imaging 2016. [cited 29 Nov 2018]. Available from url: https://www. in patients with acute confusion. Acta Neurologica Scandinavica safetyandquality.gov.au/our-work/clinical-care-standards/ 2008;118(4):245-50. delirium-clinical-care-standard/ 43 Risk reduction and management of delirium 61Abraha I, Trotta F, Rimland JM, Cruz-Jentoft A, Lozano-Montoya 75Matsumoto-Miyazaki J, Ushikoshi H, Miyata S, Miyazaki I, Soiza RL, et al. Efficacy of non-pharmacological interventions N, Nawa T, Okada H, et al. Acupuncture and traditional to prevent and treat delirium in older patients: a systematic herbal medicine therapy prevent delirium in patients with overview. The SENATOR project ONTOP Series. PLoS ONE cardiovascular disease in intensive care units. American 2015;10(6):e0123090. Journal of Chinese Medicine 2017;45(2):255-68. 62Martinez F, Tobar C, Hill N. Preventing delirium: should non- 76Moon KJ, Lee SM. The effects of a tailored intensive care unit pharmacological, multicomponent interventions be used? A delirium prevention protocol: a randomized controlled trial. systematic review and meta-analysis of the literature. Age & International journal of nursing studies 2015;52(9):1423-32. Ageing 2015;44(2):196-204. 77Simons KS, Laheij RJF, van den Boogaard M, Moviat MAM, 63Siddiqi N, Harrison JK, Clegg A, Teale EA, Young J, Taylor J, et Paling AJ, Polderman FN, et al. Dynamic light application al. Interventions for preventing delirium in hospitalised non- therapy to reduce the incidence and duration of delirium in ICU patients. Cochrane Database of Systematic Reviews 2016, intensive-care patients: a randomised controlled trial. The Issue 3. Lancet Respiratory Medicine 2016;4(3):194-202. 64Stenvall M, Berggren M, Lundstrom M, Gustafson Y, Olofsson 78Collinsworth AW, Priest EL, Campbell CR, Vasilevskis EE, B. A multidisciplinary intervention program improved the Masica AL. A review of multifaceted care approaches for the outcome after hip fracture for people with dementia - prevention and mitigation of delirium in intensive care units. subgroup analyses of a randomized controlled trial. Archives Journal of Intensive Care Medicine 2016;31(2):127-41. of Gerontology & Geriatrics 2012;54(3):e284-9. 79Litton E, Carnegie V, Elliott R, Webb SA. The efficacy of earplugs 65Wang Y, Tang J, Zhou F, Yang L, Wu J. Comprehensive as a sleep hygiene strategy for reducing delirium in the ICU: a geriatric care reduces acute perioperative delirium in elderly systematic review and meta-analysis. Critical Care Medicine patients with hip fractures: a meta-analysis. Medicine 2016;44(5):992-9. 2017;96(26):e7361. 80Clegg A, Siddiqi N, Heaven A, Young J, Holt R. Interventions for 66Freter S, Koller K, Dunbar M, MacKnight C, Rockwood K. preventing delirium in older people in institutional long-term Translating delirium prevention strategies for elderly adults care. Cochrane Database of Systematic Reviews 2014, Issue 1. with hip fracture into routine clinical care: a pragmatic 81Clegg A, Young JB. Which medications to avoid in people clinical trial. Journal of the American Geriatrics Society at risk of delirium: a systematic review. Age and Ageing 2017;65(3):567-73. 2011;40(1):23-9. 67Abraha I, Rimland JM, Trotta F, Pierini V, Cruz-Jentoft A, Soiza 82Jeong YM, Lee E, Kim KI, Chung JE, In Park H, Lee BK, et R, et al. Non-pharmacological interventions to prevent or treat al. Association of pre-operative medication use with post- delirium in older patients: clinical practice recommendations operative delirium in surgical oncology patients receiving the SENATOR-ONTOP series. Journal of Nutrition, Health and comprehensive geriatric assessment. BMC Geriatr 2016;16:134. Aging 2016;20(9):927-36. 83Weinstein SM, Poultsides L, Baaklini LR, Morwald EE, Cozowicz 68Healthcare Improvement Scotland. Think Delirium. Improving C, Saleh JN, et al. Postoperative delirium in total knee and hip the care for older people. Delirium toolkit. [cited 29 Nov arthroplasty patients: a study of perioperative modifiable risk 2018]. Available from url: https://www.whatdotheyknow.com/ factors. Br J Anaesth 2018;120(5):999-1008. request/238377/response/593545/attach/4/Delirium%20 toolkit.pdf 84Alagiakrishnan K, Wiens CA. An approach to drug induced delirium in the elderly. Postgrad Med J 2004;80(945):388-93. 69Radtke FM, Franck M, Lendner J, Kruger S, Wernecke KD, Spies CD. Monitoring depth of anaesthesia in a randomized 85Blum D, Maldonado J, Meyer E, Lansberg M. Delirium following trial decreases the rate of postoperative delirium but not abrupt discontinuation of fluoxetine. Clin Neurol Neurosurg postoperative cognitive dysfunction. British Journal of 2008;110(1):69-70. Anaesthesia 2013;110 Suppl 1:i98-105. 86Devlin JW, Skrobik Y, Gelinas C, Needham DM, Slooter AJC, 70Chan MT, Cheng BC, Lee TM, Gin T. BIS-guided anesthesia Pandharipande PP, et al. Clinical practice guidelines for the decreases postoperative delirium and cognitive decline. prevention and management of pain, agitation/sedation, Journal of Neurosurgical Anesthesiology 2013;25(1):33-42. delirium, immobility, and sleep disruption in adult patients in the ICU. Crit Care Med 2018;46(9):e825-e73. 71Whitlock EL, Torres BA, Lin N, Helsten DL, Nadelson MR, Mashour GA, et al. Postoperative delirium in a substudy of 87Scottish Government Polypharmacy Model of Care Group. cardiothoracic surgical patients in the BAG-RECALL clinical Polypharmacy Guidance, Realistic Prescribing, 3rd edition trial. Anesthesia & Analgesia 2014;118(4):809-17. Scottish Government 2018. [cited 29 Nov 2018]. Available from url: https://www.therapeutics.scot.nhs.uk/wp-content/ 72Alvarez EA, Garrido MA, Tobar EA, Prieto SA, Vergara SO, Briceno uploads/2018/09/Polypharmacy-Guidance-2018.pdf CD, et al. Occupational therapy for delirium management in elderly patients without mechanical ventilation in an intensive 88Fok MC, Sepehry AA, Frisch L, Sztramko R, Borger van der care unit: a pilot randomized clinical trial. Journal of Critical Burg BLS, Vochteloo AJH, et al. Do antipsychotics prevent Care. 2017;37:85-90. postoperative delirium? A systematic review and meta- analysis. International Journal of Geriatric Psychiatry 73Giraud K, Pontin M, Sharples LD, Fletcher P, Dalgleish T, Eden 2015;30(4):333-44. A, et al. Use of a structured mirrors intervention does not reduce delirium incidence but may improve factual memory 89Fukata S, Kawabata Y, Fujishiro K, Kitagawa Y, Kuroiwa K, encoding in cardiac surgical ICU patients aged over 70 years: Akiyama H, et al. Haloperidol prophylaxis for preventing a pilot time-cluster randomized controlled trial. Frontiers in aggravation of postoperative delirium in elderly patients: Aging Neuroscience 2016;8:228. a randomized, open-label prospective trial. Surgery Today 2017;47(7):815-26. 74Karadas C, Ozdemir L. The effect of range of motion exercises on delirium prevention among patients aged 65 and over in intensive care units. Geriatric Nursing 2016;37(3):180-5. 44 References 90Teslyar P, Stock VM, Wilk CM, Camsari U, Ehrenreich MJ, 104Li X, Yang J, Nie XL, Zhang Y, Li XY, Li LH, et al. Impact of Himelhoch S. Prophylaxis with antipsychotic medication dexmedetomidine on the incidence of delirium in elderly reduces the risk of post-operative delirium in elderly patients: patients after cardiac surgery: a randomized controlled trial. a meta-analysis. Psychosomatics: Journal of Consultation and PloS one 2017;12(2):e0170757. Liaison Psychiatry 2013;54(2):124-31. 105Geng J, Qian J, Cheng H, Ji F, Liu H. The influence of perioperative 91Zhang H, Lu Y, Liu M, Zou Z, Wang L, Xu FY, et al. Strategies dexmedetomidine on patients undergoing cardiac surgery: for prevention of postoperative delirium: a systematic review a meta-analysis. PLoS One 2016;11(4):e0152829. and meta-analysis of randomized trials. Critical Care 2013;17 106H ovaguimian F, Tschopp C, Beck-Schimmer B, Puhan M. (2):R47. Intraoperative ketamine administration to prevent delirium 92Neufeld KJ, Yue J, Robinson TN, Inouye SK, Needham DM. or postoperative cognitive dysfunction: A systematic review Antipsychotic medication for prevention and treatment of and meta-analysis. Acta Anaesthesiologica Scandinavica delirium in hospitalized adults: a systematic review and 2018;62(9):1182-93. meta-analysis. Journal of the American Geriatrics Society 107Chakraborti D, Tampi DJ, Tampi RR. Melatonin and melatonin 2016;64(4):705-14. agonist for delirium in the elderly patients. American Journal 93Shen YZ, Peng K, Zhang J, Meng XW, Ji FH. Effects of haloperidol of Alzheimer's Disease and Other Dementias 2015;30(2): on delirium in adult patients: a systematic review and meta- 119-29. analysis. Medical Principles & Practice 2018;27(3):250-9. 108C hen S, Shi L, Liang F, Xu L, Desislava D, Wu Q, et al. 94Devlin JW, Al-Qadhee NS, Skrobik Y. Pharmacologic prevention Exogenous melatonin for delirium prevention: a meta-analysis and treatment of delirium in critically ill and non-critically of randomized controlled trials. Molecular Neurobiology ill hospitalised patients: a review of data from prospective, 2016;53(6):4046-53. randomised studies. Best Practice and Research Clinical 109Nishikimi M, Numaguchi A, Takahashi K, Miyagawa Y, Matsui Anaesthesiology 2012;26(3):289-309. K, Higashi M, et al. Effect of administration of ramelteon, a 95van den Boogaard M, Slooter AJC, Bruggemann RJM, melatonin receptor agonist, on the duration of stay in the ICU: Schoonhoven L, Beishuizen A, Vermeijden JW, et al. Effect of a single-center randomized placebo-controlled trial. Critical haloperidol on survival among critically ill adults with a high Care Medicine 2018;46(7):1099-105. risk of delirium: the REDUCE randomized clinical trial. JAMA 110Oh ES, Fong TG, Hshieh TT, Inouye SK. Delirium in older 2018;319(7):680-90. persons: advances in diagnosis and treatment. JAMA 96Schrijver EJM, de Graaf K, de Vries OJ, Maier AB, Nanayakkara 2017;318(12):1161-74. PWB. Efficacy and safety of haloperidol for in-hospital delirium 111Kolanowski A, Fick D, Litaker M, Mulhall P, Clare L, Hill N, et prevention and treatment: a systematic review of current al. Effect of cognitively stimulating activities on symptom evidence. European Journal of Internal Medicine 2016;27: management of delirium superimposed on dementia: a 14-23. randomized controlled trial. Journal of the American Geriatrics 97Serafim RB, Bozza FA, Soares M, do Brasil PEAA, Tura BR, Ely Society 2016;64(12):2424-32. EW, et al. Pharmacologic prevention and treatment of delirium 112K ishi T, Hirota T, Matsunaga S, Iwata N. Antipsychotic in intensive care patients: a systematic review. Journal of medications for the treatment of delirium: a systematic review Critical Care 2015;30(4):799-807. and meta-analysis of randomised controlled trials. Journal of 98Schrijver EJM, de Vries OJ, van de Ven PM, Bet PM, Kamper AM, Neurology, Neurosurgery & Psychiatry 2016;87(7):767-74. Diepeveen SHA, et al. Haloperidol versus placebo for delirium 113Burry L, Mehta S, Perreault Marc M, Luxenberg Jay S, Siddiqi prevention in acutely hospitalised older at risk patients: a N, Hutton B, et al. Antipsychotics for treatment of delirium multi-centre double-blind randomised controlled clinical trial. in hospitalised non-ICU patients. Cochrane Database of Age & Ageing 2018;47(1):48-55. Systematic Reviews 2018, Issue 6. 99Pasin L, Landoni G, Nardelli P, Belletti A, Di Prima AL, Taddeo D, 114Perrar KM, Golla H, Voltz R. Pharmacological treatment of et al. Dexmedetomidine reduces the risk of delirium, agitation delirium in palliative care patients. A systematic literature and confusion in critically ill patients: a meta-analysis of review. Schmerz 2013;27(2):190-8. randomized controlled trials. Journal of Cardiothoracic & Vascular Anesthesia 2014;28(6):1459-66. 115Grover S, Mahajan S, Chakrabarti S, Avasthi A. Comparative effectiveness of quetiapine and haloperidol in delirium: a single 100A bdelgalel EF. Dexmedetomidine versus haloperidol for blind randomized controlled study. World journal of psychiatry prevention of delirium during non-invasive mechanical 2016(3):365-71. ventilation. Egypt J Anaesthes 2016;32(4):473-81. 116e lectronic Medicines Compendium (eMC). Summaries of 101Skrobik Y, Duprey MS, Hill NS, Devlin JW. Low-dose nocturnal Product Characteristics: Haldol Decanoate. [cited 29 Nov dexmedetomidine prevents ICU delirium. A randomized, 2018]. Available from url: https://www.medicines.org.uk/emc/ placebo-controlled trial. American journal of respiratory and product/968/smpc#CONTRAINDICATIONS critical care medicine 2018;197(9):1147-56. 117Reade MC, Eastwood GM, Bellomo R, Bailey M, Bersten A, 102Duan X, Coburn M, Rossaint R, Sanders RD, Waesberghe JV, Cheung B, et al. Effect of dexmedetomidine added to standard Kowark A. Efficacy of perioperative dexmedetomidine on care on ventilator-free time in patients with agitated delirium: postoperative delirium: systematic review and meta-analysis a randomized clinical trial. JAMA 2016;315(14):1460-8. with trial sequential analysis of randomised controlled trials. British Journal of Anaesthesia 2018;121(2):384-97. 118Tampi RR, Tampi DJ, Ghori AK. Acetylcholinesterase inhibitors for delirium in older adults. American Journal of Alzheimer's 103Deiner S, Luo X, Lin HM, Sessler DI, Saager L, Sieber FE, et al. Disease and other Dementias 2015;31(4):305-10. Intraoperative infusion of dexmedetomidine for prevention of postoperative delirium and cognitive dysfunction in elderly patients undergoing major elective noncardiac surgery: a randomized clinical trial. JAMA Surgery 2017;152(8):e171505. 45 Risk reduction and management of delirium 119Jackson TA, MacLullich AMJ, Gladman JRF, Lord JM, Sheehan B. Undiagnosed long-term cognitive impairment in acutely hospitalised older medical patients with delirium: a prospective cohort study. Age & Ageing 2016;45(4):493-9. 120Krogseth M, Watne LO, Juliebø V, Skovlund E, Engedal K, Frihagen F, et al. Delirium is a risk factor for further cognitive decline in cognitively impaired hip fracture patients. Archives of Gerontology and Geriatrics 2016;64:38-44. 121Saczynski JS, Marcantonio ER, Quach L, Fong TG, Gross A, Inouye SK, et al. Cognitive trajectories after postoperative delirium. The New England Journal of Medicine 2012;367(1):30-9. 122Salluh JIF, Wang H, Schneider EB, Nagaraja N, Yenokyan G, Damluji A, et al. Outcome of delirium in critically ill patients: systematic review and meta-analysis. BMJ 2015;350:h2538. 123Witlox J, Slor CJ, Jansen RW, Kalisvaart KJ, van Stijn MF, Houdijk AP, et al. The neuropsychological sequelae of delirium in elderly patients with hip fracture three months after hospital discharge. International Psychogeriatrics 2013;25(9):1521-31. 124Wolters AE, Peelen LM, Veldhuijzen DS, Zaal IJ, Lange DW, Pasma W, et al. Long-term self-reported cognitive problems after delirium in the intensive care unit and the effect of systemic inflammation. Journal of the American Geriatrics Society 2017;65(4):786-91. 125Langan C, Sarode DP, Russ TC, Shenkin SD, Carson A, MacLullich AM. Psychiatric symptomatology after delirium: a systematic review. Psychogeriatrics 2017;17(5):327-35. 126J ackson JC, Pandharipande PP, Girard TD, Brummel NE, Thompson JL, Hughes CG, et al. Depression, post-traumatic stress disorder, and functional disability in survivors of critical illness in the BRAIN-ICU study: a longitudinal cohort study. The Lancet Respiratory Medicine 2014;2(5):369-79. 127Abraham CM, Obremskey WT, Song Y, Jackson JC, Ely EW, Archer KR. Hospital delirium and psychological distress at 1 year and health-related quality of life after moderate-to-severe traumatic injury without intracranial hemorrhage. Archives of Physical Medicine & Rehabilitation 2014;95(12):2382-9. 128S venningsen H, Egerod I, Christensen D, Tonnesen EK, Frydenberg M, Videbech P. Symptoms of posttraumatic stress after intensive care delirium. BioMed Research International 2015:876947. 129Wolters AE, Peelen LM, Welling MC, Kok L, de Lange DW, Cremer OL, et al. Long-term mental health problems after delirium in the ICU. Critical Care Medicine 2016;44(10):1808-13. 130Healthcare Improvement Scotland. Care of older people in hospital standards. [cited 29 Nov 2018]. Available from url: http://www.healthcareimprovementscotland.org/our_work/ person-centred_care/resources/opah_standards.aspx 46 978-1-909103-68-9 SIGN157 Healthcare Improvement Scotland Edinburgh Office Glasgow Office Gyle Square Delta House 1 South Gyle Crescent 50 West Nile Street Edinburgh Glasgow EH12 9EB G1 2NP 0131 623 4300 0141 225 6999 www.sign.ac.uk