Scolaris Content Display Scolaris Content Display

Intervenciones adaptadas o personalizadas para adultos con enfermedad pulmonar obstructiva crónica y al menos otra enfermedad de larga duración: una revisión de métodos mixtos

Contraer todo Desplegar todo

Antecedentes

La enfermedad pulmonar obstructiva crónica (EPOC) es una enfermedad respiratoria crónica que se caracteriza por disnea, tos y exacerbaciones recurrentes. Las personas con EPOC a menudo viven con una o más afecciones crónicas o coexistentes (comorbilidades). Las personas con EPOC más grave suelen tener un mayor número de comorbilidades, lo que las expone a un mayor riesgo de morbilidad y mortalidad.

Objetivos

Evaluar la efectividad de cualquier intervención única para la EPOC, adaptada o personalizada según la/s comorbilidad/es, en comparación con cualquier otra intervención para personas con EPOC y una o más comorbilidades frecuentes (datos cuantitativos, ECA), en los siguientes desenlaces: calidad de vida, exacerbaciones, estado funcional, ingresos hospitalarios por todas las causas y relacionados con problemas respiratorios, mortalidad, dolor y depresión y ansiedad.

Evaluar la efectividad de una intervención (simple o compleja) adaptada o personalizada para la EPOC que tenga como objetivo cambiar el tratamiento de las personas con EPOC y una o más comorbilidades frecuentes (datos cuantitativos, ECA) en comparación con la atención habitual, en los siguientes desenlaces: calidad de vida, exacerbaciones, estado funcional, ingresos hospitalarios por todas las causas y relacionados con problemas respiratorios, mortalidad, dolor y depresión y ansiedad.

Identificar temas emergentes que describan las opiniones y las experiencias de los pacientes, los cuidadores y los profesionales sanitarios al recibir o prestar atención para tratar multimorbilidades (datos cualitativos).

Métodos de búsqueda

Se realizaron búsquedas en varias bases de datos, que incluyen el Registro de ensayos del Grupo Cochrane de Vías respiratorias (Cochrane Airways), CENTRAL, MEDLINE, Embase y CINAHL, para identificar estudios aleatorizados y cualitativos relevantes. También se buscó en registros de ensayos y se realizaron búsquedas de citas. La última búsqueda se realizó en enero de 2021.

Criterios de selección

Los ensayos controlados aleatorizados (ECA) elegibles compararon a) cualquier intervención única adaptada o personalizada según la/s comorbilidad/es para la EPOC en comparación con cualquier otra intervención, o b) cualquier intervención única (simple o compleja) adaptada o personalizada para la EPOC que tenga como objetivo cambiar el tratamiento de las personas con EPOC y una o más comorbilidades, en comparación con la atención habitual. Se incluyeron estudios cualitativos o de metodología mixta para identificar los temas.

Obtención y análisis de los datos

Se utilizaron los métodos estándar de Cochrane para el análisis de los ECA. Se utilizó la herramienta Risk of bias de Cochrane para los ECA y la lista de verificación CASP para los estudios cualitativos. Se planificó utilizar la herramienta Mixed Methods Appraisal (MMAT) para evaluar el riesgo de sesgo en los estudios de métodos mixtos, pero no se encontraron tales estudios. Se utilizó GRADE y CERQual para evaluar la calidad de la evidencia cuantitativa y cualitativa respectivamente. Los desenlaces principales para esta revisión fueron la calidad de vida y las exacerbaciones.

Resultados principales

Estudios cuantitativos

Se incluyeron siete estudios (1197 participantes) en los análisis cuantitativos, con intervenciones que incluían la telemonitorización, la rehabilitación pulmonar, la optimización del tratamiento, el entrenamiento con ejercicios en el agua y la gestión de casos. Las intervenciones se compararon con la atención habitual o con un comparador activo (como el entrenamiento con ejercicios en tierra). La duración de los ensayos varió de cuatro a 52 semanas. La media de edad de los participantes varió entre los 64 y los 72 años y la gravedad de la EPOC de leve a muy grave. Los ensayos incluyeron a personas con EPOC y una comorbilidad específica (que incluyó enfermedad cardiovascular, síndrome metabólico, cáncer de pulmón, cáncer de cabeza o cuello y afecciones musculoesqueléticas), o con una o más comorbilidades de cualquier tipo.

En general, se consideró que la evidencia presentada era de certeza moderada a muy baja (GRADE), principalmente debido a la calidad metodológica de los ensayos incluidos y a la imprecisión de las estimaciones del efecto.

Intervención versus atención habitual

La calidad de vida medida por la puntuación total del St George's Respiratory Questionnaire (SGRQ) podría mejorar con la rehabilitación pulmonar adaptada en comparación con la atención habitual a las 52 semanas (diferencia de medias [DM] ‐10,85; intervalo de confianza [IC] del 95%: ‐12,66 a ‐9,04; un estudio, 70 participantes; evidencia de certeza baja). Es probable que la rehabilitación pulmonar adaptada mejore las puntuaciones de la prueba de evaluación de la EPOC (CAT) en comparación con la atención habitual a las 52 semanas (DM ‐8,02; IC del 95%: ‐9,44 a ‐6,60; un estudio, 70 participantes, evidencia de certeza moderada) y con una intervención de telesalud multicomponente a las 52 semanas (DM ‐6,90; IC del 95%: ‐9,56 a ‐4,24; evidencia de certeza moderada). No está clara la evidencia sobre los efectos de la optimización de la farmacoterapia o de las intervenciones de telemonitorización en la mejoría de la CAT en comparación con la atención habitual.

Podría haber poca o ninguna diferencia en el número de personas que presentan exacerbaciones, o en la media de las exacerbaciones con el manejo de casos en comparación con la atención habitual (OR 1,09; IC del 95%: 0,75 a 1,57; un estudio, 470 participantes; evidencia de certeza muy baja).

En el caso de los desenlaces secundarios, la prueba de marcha de seis minutos (6MWD), podría mejorar con la rehabilitación pulmonar, el ejercicio en el agua o las intervenciones multicomponentes a las 38 a 52 semanas (evidencia de certeza baja). Una intervención multicomponente podría dar lugar a un menor número de personas ingresadas en el hospital a las 17 semanas, aunque podría haber poca o ninguna diferencia en una intervención de telemonitorización. Podría haber poca o ninguna diferencia entre la intervención y la atención habitual en la mortalidad.

Intervención versus comparador activo

Se incluyó un estudio que comparó el ejercicio en el agua y en la tierra (30 participantes). No se encontró evidencia con respecto a la calidad de vida o las exacerbaciones.

Podría haber poca o ninguna diferencia entre el ejercicio en agua y en tierra para la 6MWD (DM 5 metros; IC del 95%: ‐22 a 32; 38 participantes; evidencia de certeza muy baja).

Estudios cualitativos

Un estudio cualitativo secundario (21 participantes) exploró las percepciones y experiencias de personas con EPOC y afecciones de larga duración, y de los investigadores y profesionales sanitarios que participaron en un ECA de equipos de telemonitorización.

Se identificaron varios temas, como el estado de salud, las creencias y preocupaciones, la fiabilidad del equipo, la autoeficacia, la facilidad de uso percibida, los factores que afectan la utilidad y la utilidad percibida, las actitudes y la intención, el autocuidado y los cambios en el uso de la asistencia sanitaria. La evidencia cualitativa presentada se consideró de certeza muy baja en general.

Conclusiones de los autores

Debido a la escasez de ensayos aptos, así como a la diversidad del tipo de intervención, las comorbilidades y las medidas de desenlace proporcionadas, no fue posible proporcionar una síntesis sólida de los datos. Las intervenciones de rehabilitación pulmonar o multicomponentes podrían mejorar la calidad de vida y el estado funcional (6MWD), pero la evidencia es demasiado limitada para establecer conclusiones sólidas. El mensaje clave de esta revisión es la falta de datos de los ECA sobre los tratamientos para las personas que viven con EPOC y comorbilidades.

Debido a la variación en el número y tipo de comorbilidad/es que podría tener un individuo, y la gravedad de la EPOC, se requieren estudios más grandes que informen datos de pacientes individuales para determinar estos efectos.

PICO

Population
Intervention
Comparison
Outcome

El uso y la enseñanza del modelo PICO están muy extendidos en el ámbito de la atención sanitaria basada en la evidencia para formular preguntas y estrategias de búsqueda y para caracterizar estudios o metanálisis clínicos. PICO son las siglas en inglés de cuatro posibles componentes de una pregunta de investigación: paciente, población o problema; intervención; comparación; desenlace (outcome).

Para saber más sobre el uso del modelo PICO, puede consultar el Manual Cochrane.

Estrategias para ayudar a las personas con EPOC que tienen una o más enfermedades crónicas

¿Qué es la EPOC y la comorbilidad?

La EPOC es una enfermedad frecuente causada principalmente por fumar que puede dar lugar a problemas respiratorios a largo plazo. Los síntomas incluyen dificultad para respirar y tos con producción de esputo debido al daño en las vías respiratorias y los pulmones. Las personas con EPOC podrían tener una o más afecciones a largo plazo (comorbilidades), como enfermedades cardíacas, hipertensión, diabetes, asma y cáncer de pulmón, que pueden provocar una salud deficiente. Las personas que viven con dos o más comorbilidades también se conocen como personas con multimorbilidades.

¿Por qué se hace esta revisión?

Debido a que muchas personas con EPOC viven con multimorbilidades, naturalmente las personas que participan en los ensayos clínicos tendrán multimorbilidades. Sin embargo, los resultados de estos ensayos no suelen presentarse desglosados por multimorbilidades. Las personas con comorbilidades podrían tener que adaptar las intervenciones para tener en cuenta su comorbilidad; por ejemplo, hacer ejercicios en el agua en lugar de en la tierra para que su cuerpo esté mejor sostenido. Históricamente, las revisiones del Grupo Cochrane de Vías respiratorias no han tenido en cuenta las comorbilidades de las personas, y esta revisión es un primer paso para abordarlo. Se decidió realizar una revisión centrada en las personas con EPOC y comorbilidades tras una reunión con un grupo de pacientes con EPOC, que destacó la preocupación por las comorbilidades. Tras algunas deliberaciones, se decidió incluir los dos tipos de ensayos siguientes.

1. Cualquier intervención única para la EPOC, administrada a personas con EPOC, que esté adaptada o personalizada a su comorbilidad, en comparación con la atención habitual o cualquier otra intervención.

2. Cualquier intervención destinada a cambiar el manejo de las personas con EPOC y comorbilidades, que podría ser simple (p.ej., programar las consultas de EPOC y corazón en el mismo día) o más compleja (p.ej., desarrollar un nuevo paquete de atención para la gestión de las personas con EPOC en un servicio de salud local), en comparación con la atención habitual.

Se quería saber qué tratamientos mejoran la calidad de vida y reducen las exacerbaciones de las personas que viven con EPOC y una o más comorbilidades.

También se quería conocer la opinión de las personas con EPOC, los cuidadores y los profesionales sanitarios sobre esos tratamientos.

¿Qué información se encontró?

Se realizó una búsqueda de estudios en enero de 2021. Se encontraron siete ensayos controlados aleatorizados (ECA) elegibles que incluían a 1197 personas, y un estudio cualitativo que formaba parte de uno de los ensayos aleatorizados y proporcionaba información sobre las opiniones y experiencias de las personas en el uso de equipos de telesalud. Las personas incluidas en los ensayos tenían entre 64 y 72 años, y la gravedad de su EPOC iba de leve a muy grave. Los ensayos incluían a personas con EPOC y una comorbilidad específica, como enfermedad cardiovascular o cáncer de pulmón, o bien incluían a personas con EPOC y una o más afecciones de cualquier tipo.

Resultados y conclusiones

No hay evidencia suficiente sobre las personas con EPOC y otras comorbilidades para establecer conclusiones firmes acerca de las intervenciones dirigidas a la EPOC adaptadas a la comorbilidad. La evidencia disponible indicó lo siguiente:

‐ La calidad de vida medida por la puntuación total del St George's Respiratory Questionnaire (SGRQ) podría mejorar con la rehabilitación pulmonar adaptada en comparación con la atención habitual (cabe señalar que existe una sólida base de evidencia sobre la rehabilitación pulmonar en personas con EPOC).

‐ Es probable que la rehabilitación pulmonar mejore la calidad de vida medida por las puntuaciones del cuestionario CAT (COPD assessment test) de evaluación de la EPOC en comparación con la atención habitual a las 52 semanas y con una intervención de telesalud multicomponente.

‐ No está clara la evidencia sobre los efectos de las intervenciones de optimización de la farmacoterapia o de telemonitorización en la mejoría de la CAT en comparación con la atención habitual.

‐ Podría haber poca o ninguna diferencia en el número de personas que presentan exacerbaciones, o en la media de las exacerbaciones con la atención integral en comparación con la atención habitual.

‐ En cuanto a los desenlaces secundarios, la distancia caminada por los participantes en seis minutos podría mejorar con la rehabilitación pulmonar, el ejercicio en el agua o las intervenciones multicomponentes. Una intervención multicomponente podría dar lugar a un menor número de ingresos hospitalarios, aunque podría haber poca o ninguna diferencia en una intervención de telemonitorización.

‐ Podría haber poca o ninguna diferencia entre la intervención y la atención habitual en la mortalidad en varios estudios.

‐ Un estudio comparó el ejercicio en el agua con el ejercicio en tierra. No se encontró evidencia con respecto a la calidad de vida o las exacerbaciones. Es posible que haya poca o ninguna diferencia entre el ejercicio en el agua y en tierra sobre la distancia caminada por los participantes en seis minutos.

‐ Un estudio cualitativo exploró las percepciones y las experiencias de las personas con EPOC y enfermedades de larga duración, así como de los investigadores y profesionales sanitarios que participaron en un ECA de equipos de telemonitorización. Se identificaron varios temas, como el estado de salud, las creencias y preocupaciones, la fiabilidad del equipo, la autoeficacia, la facilidad de uso percibida, los factores que afectan la utilidad y la utilidad percibida, las actitudes y la intención, el autocuidado y los cambios en el uso de la asistencia sanitaria.

Estudios más grandes con más personas con EPOC y comorbilidades podrían ayudar a averiguar si los enfoques adaptados pueden mejorar la salud.

Certeza de la información

En general, hubo muy pocos estudios y la mayoría eran pequeños. Esto significa que los resultados se basan en una pequeña cantidad de información. Los ensayos con intervenciones diferentes y que incluyan más personas o diferentes podrían dar un resultado distinto.

Authors' conclusions

Implications for practice

Owing to a paucity of eligible trials, as well as diversity in the intervention type, comorbidities and the outcome measures reported, we were unable to provide a robust synthesis of data. Rehabilitation or pharmacological management may improve quality of life, but evidence is based on single studies and should be interpreted with caution. We could not determine a benefit or harm of interventions on other outcomes, including exacerbations. The key take‐home message from this review and a potential area of investigation is the lack of data from RCTs on treatments for people living with COPD and comorbidities.

Implications for research

Future COPD comorbidity studies should evaluate the impact of interventions on pain. Researchers should consider adding a qualitative element to their RCT, or running a qualitative study alongside it to help understand the experiences of care for people with complex chronic conditions which may not be captured by quantitative measurement tools that are often disease‐specific. This information is important in understanding decision‐making processes about acceptance and their ability to process the information provided. It can inform the delivery of the intervention and the individuals most likely to benefit. Qualitative studies should explore the experiences of participants, carers and HCPs who take part in trials of these interventions.

It is important that the inclusion criteria for COPD trials allow the participation of people with comorbidity, to ensure their results can be applied to people with COPD living with multiple long‐term conditions. Trials should make full individual patient data (IPD) available so that the impact of interventions on outcomes in people with comorbid COPD can be assessed.

Summary of findings

Open in table viewer
Summary of findings 1. Intervention compared to usual care for COPD and at least one other long‐term condition

Intervention compared to usual care for COPD and at least one other long‐term condition

Patient or population: COPD and at least one other long‐term condition
Setting: community teaching hospital (1), hospital outpatient clinic (1), university hospital (1), tertiary public hospital (1), multi‐centre (3), single hospital (1)
Intervention: Intervention (rehabilitation, organisation of care, pharmacotherapy, multicomponent intervention)
Comparison: Usual care

Outcome domain

Intervention group

(follow‐up)

Anticipated absolute effects* (95% CI)

Relative effect

No of participants

(studies)

Certainty of the evidence

(GRADE)

Comments

Risk with usual care

Risk with intervention

Quality of life ‐ SGRQ total
Scale from: 0 to 100 (lower scores better)

 

Rehabilitation (pulmonary rehab)

(follow‐up 52 weeks)

The mean SGRQ total score was 70

MD 10.85 lower (12.66 lower to 9.04 lower)

70
(1 RCT)

(95% CI)⊕⊝⊝
LOWa,b
 

MCID for SGRQ is a change of 4 points (Jones 2005)

 

Quality of life ‐ CAT total

Scale from 0 to 40 (lower scores better)

 

 

 

Pharmacotherapy (optimised COPD treatment)

(follow‐up 52 weeks)

The mean CAT total score was 0.4

MD 0.00
(3.40 lower to 3.40 higher)

77
(1 RCT)

⊕⊕⊝⊝
LOWa,c,d

MCID for CAT total is 2 points (Kon 2014)

 

 

 

 

Rehabilitation (pulmonary rehab)

(follow‐up 52 weeks)

The mean CAT total score was 24.34

MD 8.02 lower
(9.44 lower to 6.6 lower)

70
(1 RCT)

⊕⊕⊕⊝
MODERATEa,b

Organisation of care (telemonitoring)

(follow‐up 39 weeks)

The mean CAT total score was 17.17

MD 0.41 lower
(2.19 lower to 1.37 higher)

312
(1 RCT)

⊕⊕⊝⊝
LOWc,d

Multicomponent intervention

(follow‐up 52 weeks)

The mean CAT total score was 1.6

MD 6.9 lower
(9.56 lower to 4.24 lower)

80
(1 RCT)

⊕⊕⊕⊝
MODERATEa,c

Exacerbations ‐ number of people experiencing one or more

 

Rehabilitation (case management)
(follow‐up 52 weeks)

 

 

573 per 1000

594 per 1000 (501 to 678)

OR 1.09
(0.75 to 1.57)

470
(1 RCT)

⊕⊝⊝⊝
VERY LOWe,f

Functional status‐6MWD (metres)

 

 

Rehabilitation (pulmonary rehab and water‐based exercise)

(follow‐up 38.8 weeks**)

The mean distance walked in 6 minutes was 344 metres

MD 60.4 metres higher
(44.26 higher to 76.54 higher)

100
(2 RCTs)

⊕⊕⊝⊝
LOWa,c

MCID for the 6MWT is 25 to 35 metres (Holland 2013)

 

 

Multicomponent intervention

(follow‐up 52 weeks)

The mean distance walked in six minutes was ‐15

MD 75 higher
(28.06 higher to 121.94 higher)

80
(1 RCT)

⊕⊕⊝⊝
LOWa,c

All‐cause hospital admissions ‐ people experiencing one or more

 

Organisation of care (telemonitoring)

(follow‐up 39 weeks)

 

292 per 1000

 

273 per 1000
(185 to 382)

OR 0.91 (0.55 to 1.50)

312
(1 RCT)

⊕⊕⊝⊝
LOWc

Multicomponent intervention

(follow‐up 17 weeks)

732 per 1000

 

459 per 1000
(277 to 647)

OR 0.31
(0.14 to 0.67)

112
(1 RCT)

⊕⊕⊝⊝
LOWa,c

All‐cause mortality (deaths)

 

 

 

Pharmacotherapy (optimised COPD treatment)
(follow‐up 17.6 weeks**)

170 per 1000

102 per 1000
(45 to 217)

OR 0.55
(0.23 to 1.35)

177
(2 RCTs)

⊕⊝⊝⊝
VERY LOWa,e,g

Organisation of care (case management and telemonitoring)

(follow‐up 46.7 weeks**)

102 per 1000

60 per 1000
(36 to 98)

OR 0.56
(0.33 to 0.96)

782
(2 RCTs)

⊕⊕⊝⊝
LOWe

Multicomponent intervention

(follow‐up 52 weeks)

18 per 1000

18 per 1000
(1 to 230)

OR 1.00
(0.06 to 16.39)

112
(1 RCT)

⊕⊝⊝⊝
VERY LOWa,c,h

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

**Weighted mean duration of follow‐up

6MWD: 6‐minute walk distance; CAT: COPD assessment test; CI: Confidence interval; COPD: chronic obstructive pulmonary disease; GIV: generic inverse variance; HADS: Hospital Anxiety and Depression Scale; MD: mean difference; MCID: minimally clinically important difference; OR: Odds ratio; RCT: randomised controlled trial;RR: Risk ratio; SGRQ: St George's Respiratory Questionnaire.

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

aThe evidence was downgraded by 1 for imprecision due to the optimal information size of less than 200 participants.
bThe evidence was downgraded by 1 for risk of performance bias.
cThe evidence was downgraded by 1 for risk of performance and attrition bias.
dThe evidence was downgraded by 1 for imprecision due to wide confidence intervals.
eThe evidence was downgraded by 2 for risk of performance and detection bias.
fThe evidence was downgraded by 1 for imprecision as the lower confidence interval crossed the line of no effect.
gThe evidence was downgraded by 1 for risk of attrition due to high dropout rate in the study.
hThe evidence for this outcome was downgraded by 2 for imprecision due to very wide confidence intervals.

Open in table viewer
Summary of findings 2. Intervention compared to active comparison for COPD and at least one other long‐term condition

Intervention compared to active comparison for COPD and at least one other long‐term condition

Patient or population: COPD and at least one other long‐term condition
Setting: outpatient respiratory departments from 5 hospitals (multicentre), hospital outpatient clinic (1), tertiary public hospital (1)
Intervention: Intervention (rehabilitation water‐based versus land‐based exercise programme)
Comparison: active comparison

Outcome domain

Intervention group (follow‐up)

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with active comparison

Risk with Intervention

Quality of life ‐ SGRQ total
 

Not reported
 

Quality of life ‐ CAT total
 

Not reported
 

Exacerbations
 

Not reported
 

Functional status ‐ 6MWD (metres)

Rehabilitation

(follow‐up 8 weeks)

The mean distance walked in 6 minutes was 43 metres

MD 5 metres higher
(22.21 lower to 32.21 higher)

30
(1 RCT)

⊕⊝⊝⊝
VERY LOWa,b,c

The MCID for 6MWT is 25 to 35 metres (Holland 2013)

All‐cause hospital admissions
 

Not reported
 

All‐cause mortality
 

Not reported
 

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

6MWD: 6‐minute walk distance; CAT: COPD assessment test; CI: Confidence interval; COPD: chronic obstructive pulmonary disease; MD: mean difference; MCID: minimally clinically important difference; RCT: randomised controlled trial;SGRQ: St George's Respiratory Questionnaire.

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

aThe evidence was downgraded by 1 for imprecision due to the lower confidence interval crossing 1.0.
bThe evidence was downgraded by 1 for imprecision as the optimal information size was less than 200 participants.
cThe evidence was downgraded by 2 for risk of performance and attrition bias.

Open in table viewer
Summary of findings 3. Summary of findings table for qualitative GRADE CERqual assessment

Theme

Methodological limitations

Coherence

Adequacy of data

Relevance

Dissemination bias

Overall assessment

Explanation of

CERQual assessment

Studies contributing to the review finding

Health status, beliefs and concerns

The findings were graded

as very low confidence

because of minor

concerns regarding

methodological limitations

and relevance. There were major concerns regarding

coherence and adequacy of data

Middlemass 2017

Unchanging nature of condition

Some

concernsa

Major concernsb

Major concernsb

Some

Concernsc

No concerns

Very lowc

Withdrawal of face to face communication

Some

concernsa

Major concernsb

Major concernsb

Some

Concernsc

No concerns

Very lowc

Reminder of illness and anxiety

Some

concernsa

Major concernsb

Major concernsb

Some

Concernsc

No concerns

Very lowc

Information

Subjective norms

Some

concernsa

Major concernsb

Major concernsb

Some

Concernsc

No concerns

Very lowc

Technology

Unreliable technology

Some

concernsa

Major concernsb

Major concernsb

Some

Concernsc

No concerns

Very lowc

HIT Self‐efficacy

Some

concernsa

Major concernsb

Major concernsb

Some

Concernsc

No concerns

Very lowc

Perceived usefulness

Daily monitoring of conditions

Some

concernsa

Major concernsb

Major concernsb

Some

Concernsc

No concerns

Very lowc

Factors affecting usefulness

Lack of feedback

Some

concernsa

Major concernsb

Major concernsb

Some

Concernsc

No concerns

Very lowc

Behaviour

Self‐management and healthcare utilisation

Some

concernsa

Major concernsb

No concerns

No concerns

No concerns

Very lowc

Criteria for assessment (https://www.cerqual.org/publications/)

Methodological limitations: risk to rigour see Table 1.

Coherence: how clear and cogent the fit is between the data from the primary studies and the review finding that synthesises that data. ‘Findings’ are ‘transformations’ of the underlying data into descriptions, interpretations and /or explanations of the phenomenon of interest. Findings are developed by identifying patterns in the data across primary studies

Adequacy: overall determination of the degree of richness as well as the quantity of data supporting the review finding i.e., extent to which information that the study authors provide is detailed enough to interpret the meaning and context of what is being researched.

Relevance: refers to the extent to which the body of data from primary studies supports the review finding in terms of applicability to the review question.

Dissemination bias: a systematic distortion of the phenomenon of interest due to selective dissemination of qualitative studies or the findings of qualitative studies

aThere were some concerns about the research design, and recruitment strategy to address the aims. It was not clear how the data was collected.
bThe evidence mainly because the evidence is based on one study. Due to the limited number of studies, we cannot be certain that there are any issues about whether the data fit the finding of the review. Findings do not support quantitative data.
cThe findings from the study did not answer all aspects of context specified in the review question.

Open in table viewer
1. Risk of bias assessment for qualitative studies

Study ID

Is there a statement of research aims?

Is a qualitative approach justified?

Was the research design appropriate to address the aims?

Was the recruitment strategy appropriate to address the aims?

Were data collected in a way that addressed the research issue?

Was the researcher/participant relationship adequately considered?

Have ethical issues been taken into consideration?

Was the data analysis sufficiently rigorous?

Was there a clear statement of findings?

Overall assessment

Middlemass 2017

Yes

Yes

Can't tell

Yes

Can't tell

No

Yes

Yes

Yes

Some concerns

To test the HITAM and see if it could be used to increase the adoption of HIT. HIT has been shown to reduce mortality. Many with LTC are over 60 and HIT is not always accepted.

Pre‐ and post‐interviews to ascertain perceptions/experience on the HITAM elements. However, no rationale for adopting a qualitative and framework approach.

The study design (instrumental, collective case design) is not justified.

Selection process for interviews included those who had taken part in the pilot and those assigned to the Rx. One was not interviewed and was unwell.

Setting of interviews (home or telephone) was not justified. Not clear if interviewers were in‐depth or semi‐structured. Not justified why interviews were chosen rather than focus groups. Interview schedule was used and is made available. Interviews were audio‐recorded. Data saturation is not mentioned.

The interviewer's role is not described or examined.

Ethical approval obtained and informed written consent to take part in two taped interviewers to ascertain views if using the equipment.

Process of analysis is well described, although unsure how the framework was formed. Quotes provided to support themes. Contrasting data are taken into account and described.

Explicit well organised findings and multiple researchers involved in the process.

Abbreviations: HITAM: Health Information Technology Acceptance Model; LTC: long term condition.

Background

Description of condition

It is estimated that the global population of people aged 60 and over will triple to 2.1 billion by 2050, with an increase of 32% in more developed countries, and 10% to 19% in less developed countries (United Nations 2013). As more people live longer, the number of chronic physical conditions that they may have are likely to increase (Academy of Medical Sciences Report 2018; Garin 2016).

The term 'multimorbidity' is defined as the co‐existence of two or more chronic conditions, neither (or none) of which are considered to be an index condition (Academy of Medical Sciences Report 2018). Multimorbidity is associated with increasingly poor health outcomes (including reduced quality of life; impaired functional status; weakened physical and mental health; increased risk of re‐admission to hospital; and mortality) (Barnett 2012; Holland 2016; NICE 2018).

Prevalence of multimorbidity on a global level may be difficult to determine, as access to health care and diagnosis of chronic conditions vary from country to country (Academy of Medical Sciences Report 2018). However, one cross‐sectional study has recently shown that the prevalence of multimorbidity increases from over 40% to 70% in those aged 60 to 69 years across several low‐, middle‐ and high‐income countries (China, Finland, Ghana, India, Mexico, Poland, Russia, South Africa and Spain) (Garin 2016). It is estimated that approximately one in four people in the UK live with two or more long‐term conditions, rising to two‐thirds in people aged 65 and over (Barnett 2012; NHS England 2018; Salisbury 2018).

Chronic obstructive pulmonary disease (COPD) is the name for a group of lung diseases, including bronchitis and emphysema. COPD occurs in adults (aged 35 years and over), and is characterised by chronic airflow obstruction that interferes with normal breathing and is not fully reversible (World Health Organization 2018). Chronic airflow obstruction is defined by spirometry in which "the volume of air forcibly exhaled from the point of maximal inspiration (forced vital capacity, FVC), and the volume of air exhaled during the first second (forced expiratory volume in one second, FEV₁)" are measured. A FEV₁/FVC ratio of less than 0.70 is an indicator for airway obstruction (GOLD 2021). Diagnosis of COPD is considered in people experiencing day‐to‐day symptoms such as coughing, breathlessness (dyspnoea), wheezing, frequent chest infections and is confirmed with spirometry ‐ a post‐bronchodilator FEV1/FVC ratio below 70% indicates airflow limitation (GOLD 2021). People may also experience periodic exacerbations (flare‐ups) that punctuate the disease course. Risk factors for COPD include smoking and environmental exposures leading to abnormalities of the airways and alveoli (World Health Organization 2018; GOLD 2021).

Comorbidity is defined as any distinct clinical entity that may occur during the clinical course of a patient with the index disease under study. The focus of this review is COPD as the index disease. COPD is associated with a high prevalence of comorbidities (Smith 2014), and it is common for people with COPD to have more than one co‐existing long‐term health condition that can vary in nature and severity (Cavailles 2013; Holland 2016). People with more severe COPD (GOLD stage D) are likely to have a higher number of comorbidities (Raherison 2018), which puts them at a higher risk of mortality compared to people with mild or moderate COPD, or those without COPD and co‐existing long‐term health conditions (Divo 2012; Hanlon 2018; Mannino 2008).

Common long‐term conditions that co‐exist with COPD are cardiovascular disease, hypertension, diabetes, asthma, and lung cancer (Hillas 2015). People may also live with long‐term condition system complexes such as frailty and chronic pain (Andenes 2018; Holland 2016). In this review we will treat pain as an outcome, rather than a condition. These long‐term conditions may or may not be related to COPD.

In this review, we focus on people with COPD living with one or more long‐term physical conditions (also referred to as comorbidities of COPD) (Holland 2016; Smith 2016). We did not plan to include people with conditions caused by COPD treatments, such as pneumonia, or ongoing conditions such as learning disability, sensory impairment such as sight or hearing loss, and alcohol and substance misuse.

Description of interventions

Interventions (treatments) for people with COPD are either aimed at helping them to manage the symptoms of COPD in day‐to‐day life, or are treatment of exacerbations (flare‐ups). For treating the symptoms, there are drugs including inhaled therapies (such as long‐acting beta₂‐agonists, long‐acting muscarinic antagonists, and inhaled corticosteroids), phosphodiesterases and antibiotics, as well as physical interventions such as pulmonary rehabilitation, physical therapy (e.g. exercise), ventilation (e.g. non‐invasive ventilation (NIV)). For treating exacerbations, there are inhaled therapies, antibiotics and ventilation.

In this review we looked at COPD interventions which target the comorbidity, and interventions for the overall management of people with COPD and one or more comorbidities.

We created a framework from the GOLD 2021 guidelines and the Cochrane Airways subtopic list, from which we intend to create an evidence (gap) map and use it as a basis for the analysis (Table 2).

Open in table viewer
Table 2. Framework and map of interventions identified from included studies

Interventions (identified fromGOLD 2021 guideline andCochrane Airways subtopic list.)

Study

Interventions

Evidence type (quantitative, qualitative or mixed methods)

Comorbidities

  • Reducing risk factors

    • Smoking cessation

  • Vaccination

    • Pneumococcal

    • Influenza

  • Pharmacotherapies

    • Short‐acting inhalers (e.g. SABA)

    • Long‐acting inhalers (e.g. LABA, LAMA, ICS)

    • Phosphodiesterase‐4 (PDE4) inhibitors (e.g. Roflumilast)

    • Mucolytic agents

    • Combination inhalers and triple therapy

    • Methyl xanthines

    • Oral corticosteroids

    • Antibiotics

    • Statins

    • Alpha‐1 antitrypsin augmentation therapy

    • Biomarker mediated therapy

EUCTR2010‐021412‐42‐GB

Inhaler optimisation and best supportive care vs UC

Quantitative

All had lung cancer

Gottlieb 2020

Management: optimising COPD treatment vs UC

Quantitative

All had lung cancer or head and neck cancer, and some had other comorbidities including ischaemic heart diseases, heart failure, depression/anxiety, osteoporosis, cerebrovascular disease, diabetes

  • Rehabilitation

    • Pulmonary rehabilitation

    • Exercise therapy (e.g. upper limb exercise, ongoing physical exercise after pulmonary rehabilitation)

    • Complementary therapies (e.g. active mind‐body therapy, Tai chi, singing)

Budnevskiy 2015

Pulmonary rehabilitation vs UC

Quantitative

All had metabolic syndrome

McNamara 2013b

Water‐based exercise training vs land‐based exercise training vs UC

Quantitative

One or more physical comorbidities: musculoskeletal, or neurological, or obesity

  • Self‐management

  • Organisation of care

    • Support services (e.g. social care, specialist respiratory nurse)

    • Integrated care

    • Telehealthcare

    • Digital management interventions

    • Home care

    • Integrated disease management (e.g. disease management programming)

    • Interventions to promote or increase adherence to PR or other treatments

Rose 2018

Case management vs UC

Quantitative

Two or more comorbidities: cardiovascular disease, including coronary artery disease, hypertension and congestive heart failure, diabetes, depression, osteopenia, osteoporosis, and gastro‐oesophageal reflux disease.

Walker 2018

Middlemass 2017

Telemonitoring vs UC

Quantitative

Qualitative

One or more non‐pulmonary comorbidity: congestive heart failure or ischaemic heart disease or both, hypertension, osteoporosis, hyperlipidaemia, osteoporosis, sleep‐related disordered breathing.

  • Other treatments

    • Oxygen therapy and ventilatory support (e.g. NIV, ambulatory oxygen)

    • Nutritional support

    • Lung volume reduction surgery

    • Lung transplantation

    • Supportive, palliative, end of life and hospice care

    • Psychotherapy

    • interventions for sexual dysfunction

    • Self‐help groups

  • Multicomponent intervention

Bernocchi 2018

Maintenance rehabilitation and telemonitoring vs UC. Following inpatient rehabilitation participants randomised to personalised discharge plus nurse telephone support and telemonitoring, plus physiotherapist personalised rehabilitation vs UC

Quantitative

All had cardiovascular disease

UC: usual care

How will the intervention work?

Long‐term conditions other than COPD may interfere with the delivery of the COPD intervention. An example of people with comorbid COPD engaging with an intervention differently from those with COPD alone is seen in pulmonary rehabilitation, one of the more effective treatments for people with COPD (McCarthy 2015). Researchers have shown that people with comorbid COPD are more likely than people with COPD alone to either decline to enrol for treatment or, once enrolled, to drop out of the programme or not attend sessions regularly (Fischer 2009; Hayton 2013; Keating 2011). People are more likely to drop out of pulmonary rehabilitation programmes as a result of symptoms of other comorbidities (Blackstock 2018). Furthermore, researchers have evaluated the impact of co‐existing conditions on outcomes of a pulmonary rehabilitation intervention for people with COPD which showed that, depending on the co‐existing condition, pulmonary rehabilitation outcomes can be positive or negative (Carreiro 2013; Crisafulli 2008; Holland 2016; Walsh 2013). Targeted interventions can help people take part in pulmonary rehabilitation programmes: a targeted water‐based exercise component of a pulmonary rehabilitation programme was shown to be more effective than land‐based exercise (McNamara 2013a). We therefore intend to summarise evidence from randomised controlled trials (RCTs) which target a COPD intervention to take account of another comorbidity.

People with multimorbidities may be taking multiple drugs for each individual condition; for example, prescribed drugs, over‐the‐counter treatments, herbal remedies or dietary supplements. This is called polypharmacy and 16.4% of people over the age of 65 years are estimated to be taking 10 or more treatments each day (Duerden 2013; Guthrie 2012). This can lead to unfavourable drug interactions and practical issues with remembering to take so many medications in a day. We will include interventions which help people adapt to taking multiple medications; we will not, however, be looking at polypharmacy interventions which aim to optimise a person's drugs and reduce harmful drug interactions.

People with multimorbidities may also have to see many healthcare professionals (HCPs) to help them with various different elements of their different long‐term conditions. We will include trials which aim to streamline (or simplify) this care in some way, to make it easier or better for the patient. These might include, for example, putting a patient under the care of one particular consultant who works across several hospital departments, thereby providing a holistic package of care. It may include a hospital putting together an integrated disease management programme — a map of a patient's journey for managing their condition in a particular location. We will also consider simpler interventions such as running COPD and cardiovascular clinics on the same day to reduce the number of attendances at hospital.

Anxiety and depression are common in people with COPD. Pharmacological and psychological interventions aimed at treating the anxiety and depression are explored in a suite of Cochrane Reviews (Pollok 2018; Pollok 2019; Usmani 2011; Usmani 2017). Because the interventions are treating the comorbidity rather than the COPD, we do not include them in this review. We will also not include studies of people with COPD who have symptoms of depression or anxiety as the sole comorbidity.

We have been unable to update a draft logic model that we presented in the protocol for this review (Figure 1, Janjua 2019), due to limited evidence found. As a result, we were unable to take this forward to our Cochrane Airways Patient Advisory Group and Programme Grant Steering Group for their consideration.


Study flow diagram for randomised controlled trials

Study flow diagram for randomised controlled trials

Why is it important to do this review?

Most clinical trials are designed to involve people with one condition, and exclude people with multimorbidities — which may represent well over half of people who live with COPD. Systematic reviews, including Cochrane Reviews, have therefore also traditionally focused on these patients, rather than including a sample representing the true population. This means that most studies may not be applicable to people with more than one chronic condition: for example, trials may only enrol people with COPD and may exclude people with asthma or heart disease. This means that we cannot be confident about applying the results of the trial to people with COPD and asthma, or people with COPD and heart disease.

There is a substantial health burden for people living with COPD and multimorbidities, with associated cost implications due to an increased need for hospital utilisation compared to those who only have one condition (Chen 2017). People living with multimorbidity may also have to manage several symptoms, to adhere to multiple drug regimens and various lifestyle recommendations, all while attending appointments with different HCPs (Smith 2016). Healthcare services experience higher demands as people with multimorbidities require more frequent complex care (Barnett 2012; Rijken 2018), and these services can be fragmented (Smith 2016).

Policy‐makers are increasingly aware that overall care for people with multimorbidities needs to be patient‐centred (i.e. care that takes a person's needs into account, either via individual preference, or by involving the person in making decisions about their care) and integrated (i.e. organisations and staff working together to provide seamless care through processes that are flexible and continuous) (Rijken 2016).

In addition, guidance for managing multimorbidities is limited because of the potential exclusion from clinical trials of people living with multimorbidity. The systematic failure of clinical trials to include people living with multimorbidity leads to care strategies that may not be suitable or helpful for most people with COPD (Wyatt 2014). For example, multiple prescriptions (polypharmacy) can lead to potential interactions between conditions and medication resulting in inadequate and complex choices of treatment in terms of benefit and harm (Sinnot 2013; Muth 2018), or fragmented and poorly co‐ordinated care packages can lead to complications such as over‐hospitalisation when managing patients with multimorbidities (Sinnot 2013; Rijken 2016).

We have decided to undertake this review because the Cochrane Airways Patient Advisory Group and Programme Grant Steering Group considered this to be an important topic to be reviewed for a programme of Cochrane Airways Reviews funded by the National Institute for Health Research. The patients and HCPs agreed that the systematic review should report information about the clinical effectiveness of interventions, and the views and experiences of those involved in managing multimorbidities and COPD, and identify gaps in the evidence. The review will address issues that are important for people with COPD who have co‐existing conditions, as well as for HCPs and policy‐makers. A scoping search of quantitative and qualitative evidence conducted prior to initiation of the protocol showed potentially eligible studies.

In comparison with a previous Cochrane Review (Smith 2016), we decided to take a mixed‐methods approach to evaluate the evidence that exists for people living with COPD and at least one other chronic condition in this review, because of concerns that interventions begun with the best intentions may not always be helpful for patients. Combining both quantitative and qualitative data can provide a better understanding of why some interventions are successful and why others less so. This approach helps to add richness and depth to quantitative findings, which is not methodologically possible to do when interpreting quantitative data alone. It can identify areas where quantitative research may be lacking but appears to be important to patients, carers or health professionals (as identified from qualitative research). We identified studies conducted in a community or hospital setting and combined both quantitative data (numerical data from clinical trials), and qualitative data (non‐numerical data from, for example, semi‐structured interviews, focus group discussions and patient, carer or health professional observations). To illustrate: we are aware of a local example where people with COPD and heart disease have been put under a co‐ordinated care regimen, but the patients have said they prefer separate appointments because they are shorter, and they like having a reason to get out of the house.

We deliberately left the types of intervention very broad (compared to Smith 2016), to reflect the reality of people living with COPD and other long‐term health conditions in trying to make sense of a sparse literature, who nonetheless need to make decisions about how to manage their own symptoms and daily life. The interventions aimed to address COPD rather than the full extent of multimorbidity.

Objectives

  • To assess the effectiveness of any single intervention for COPD adapted or tailored to their comorbidity(s) compared to any other intervention for people with COPD and one or more common comorbidities (quantitative data, RCTs) in terms of the following outcomes: Quality of life, exacerbations, functional status, all‐cause and respiratory‐related hospital admissions, mortality, pain, and depression and anxiety.

  • To assess the effectiveness of an adapted or tailored single COPD intervention (simple or complex) that is aimed at changing the management of people with COPD and one or more common comorbidities (quantitative data, RCTs) compared to usual care in terms of the following outcomes: Quality of life, exacerbations, functional status, all‐cause and respiratory‐related hospital admissions, mortality, pain, and depression and anxiety.

  • To identify emerging themes that describe the views and experiences of patients, carers and healthcare professionals when receiving or providing care to manage comorbidities (qualitative data).

  • To use a mixed‐methods approach to combine quantitative and qualitative data resulting from the first three objectives, provided that we find relevant data. If we find that we are unable to combine quantitative data and qualitative textual themes, we will present the data and themes separately.

Methods

Types of studies

We included the following study designs to address the objectives of this review.

  • Randomised controlled trials (RCTs) to assess effectiveness of interventions (quantitative data).

  • Qualitative studies of any design, including in‐depth interviews, semi‐structured interviews, focus group discussion, observations, and surveys that explore views, opinions and experiences of people with comorbid COPD, their carers and health professionals involved in provision of care.

  • Mixed‐methods studies (RCTs that also include a qualitative study as part of their investigations).

Types of participants

We included adults with a primary diagnosis of COPD of any severity (e.g. Global Initiative for Chronic Obstructive Lung Disease (GOLD), or American Thoracic Society (ATS) criteria), with at least one other long‐term condition (e.g. asthma, coronary heart disease, diabetes, atrial fibrillation, heart failure, hypertension, stroke/transient Ischaemic attack, lung cancer or osteoporosis (Barnett 2012)).

We included people with COPD who also had anxiety or depression or both, but this was not permitted to be the only comorbidity.

We included studies involving carers and healthcare professionals (HCPs) when receiving or providing care to manage comorbidities.

Types of interventions

We included the following interventions for quantitative studies.

  • Any single intervention for COPD delivered to people with COPD adapted to or targeting their comorbidity (or comorbidities) (e.g. participants receiving a pulmonary rehabilitation programme tailored to take account of their comorbidities by delivering the exercise component in water rather than out of water (McNamara 2013a)) compared to any other intervention. We envisage these interventions being broken into the following categories of interventions.

    • Pulmonary rehabilitation.

    • Self‐management.

    • Exercise or other physical therapy.

    • Ventilation.

    • Pharmacotherapy (e.g. change of inhaler).

  • Any intervention aimed at changing the management of people with COPD and one (or more) co‐existing long‐term condition(s), which could be simple (e.g. scheduling COPD and heart clinics on the same day) or more complex (e.g. developing an integrated care package for management of people with COPD in a particular hospital and providing training to staff to deliver it), compared to routine care (or usual care, control, or no intervention). We envisage these interventions being broken into the following categories of interventions.

    • Organisation‐wide interventions (such as introducing a new care pathway).

    • Simple changes within the organisation (such as scheduling relevant clinics on the same day).

    • Interventions across a wider area (such as integration between GP, hospital and pharmacy).

We included interventions delivered in primary (community) or secondary (hospital) care.

We excluded studies of interventions for the comorbidity (e.g. heart surgery).

We excluded studies of pharmacological or psychological interventions that targeted anxiety or depression or both rather than COPD (previous Cochrane Reviews have, for example, included COPD patients with either anxiety (Usmani 2017), depression (Pollok 2018; Pollok 2019), or both anxiety and depression (Usmani 2011)).

We excluded interventions that were designed to reduce the number of prescribed medicines or interactions between them (polypharmacy), but we planned to include interventions that aim to help people to manage polypharmacy.

We excluded interventions delivered to HCPs.

We included qualitative studies that explored the experiences of participants, carers and HCPs, taking part in the above interventions.

Types of outcome measures

We included the following outcomes for quantitative analysis.

Primary outcomes

  • Quality of life (e.g. St. George's Respiratory Questionnaire (SGRQ), COPD assessment test (CAT))

  • Exacerbations (as defined by trialists, depending on the data available, we extracted either number of participants experiencing one or more exacerbation, or the exacerbation rate, or both)

Secondary outcomes

  • Functional status (6‐minute walk distance (6MWD)/incremental shuttle walk test (ISWT))

  • All‐cause hospital admissions (also as a proxy for use of services, e.g. reduction of use)

  • Respiratory hospital admissions

  • Mortality (all causes)

  • Pain (as reported in trials)

  • Anxiety symptoms (measured by e.g. Hospital Anxiety and Depression Scale (HADS))

  • Depression symptoms (measured by e.g. HADS)

For the planned qualitative synthesis, our outcomes were themes arising from the data.

We included studies regardless of whether they report our predefined outcomes.

There was no minimum duration for the interventions, and we used data reported for the last follow‐up point.

Search methods for identification of studies

Electronic searches

We searched for studies in June 2019, February 2020, and January 2021 in the following databases and trials registries:

  • Cochrane Airways Register of Trials through the CRS, from inception onwards;

  • Cochrane Central Register of Controlled Trials (CENTRAL, in the Cochrane Library) through the Cochrane Register of Studies (CRS), from inception onwards;

  • MEDLINE Ovid SP from 1946 onwards;

  • Embase Ovid from 1974 onwards;

  • PsycINFO Ovid Sp from 1974 onwards;

  • CINAHL EBSCO (Cumulative Index to Nursing and Allied Health Literature) from 1937 onwards;

  • Web of Science Core Collection from 1970 onwards;

  • US National Institutes of Health Ongoing Trials Register ClinicalTrials.gov;

  • World Health Organization International Clinical Trials Registry Platform (ICTRP).

Searches for qualitative and quantitative studies were run separately using appropriate study design filters. We combined search terms for the target population with the Cochrane Highly Sensitive Search Strategy to identify reports of RCTs (Lefebvre 2021), and terms from the search strategies developed and tested by DeJean 2016 to find reports of qualitative studies. The search was developed in MEDLINE by ES, with input from the other authors, and was peer‐reviewed by another Information Specialist using the PRESS checklist (McGowan 2016). The MEDLINE search strategy was adapted appropriately for use in the other databases. We did not apply any language limits, and we did not limit the search strategy by population characteristics such as age, gender, or race. Details of the database search strategies, search dates, and the number of results retrieved are presented in Appendix 1.

We initially searched all databases from their inception to June 2019. We updated the searches in February 2020 and January 2021 in a reduced number of databases (Appendix 2) following an analysis of the individual database yield of eligible study references.

Searching other resources

We conducted a forwards and backwards citation search in Web of Science for each included study on 15 May 2020 (RCTs) and 5 June 2020 (qualitative studies).

We checked the reference lists of related review articles for additional references. We used the Epistemonikos database to search for relevant systematic reviews (www.epistemonikos.org).

We searched for errata or retractions from included studies published in full text on PubMed on 4 December 2020.

Data collection and analysis

Selection of studies

We retrieved many search results, and we therefore used Cochrane's 'Screen4Me' workflow to help assess the results of our search for RCTs. Screen4Me comprises three components: known assessments — a service that matches records in the search results to records that have already been screened in Cochrane Crowd and have been labelled as 'RCT' or as 'Not an RCT'; the RCT classifier — a machine‐learning model that distinguishes RCTs from non‐RCTs (Marshall 2018); and if appropriate, Cochrane Crowd — Cochrane's citizen science platform where 'the crowd' help to identify and describe health evidence (Noel‐Storr 2020).

Following use of the Screen4Me workflow, two of three review authors (SJ, ES and ED) screened each title and abstract of the remaining search results independently and coded them as 'retrieve' (eligible or potentially eligible/unclear) or 'do not retrieve'. We retrieved the full‐text study reports of all potentially eligible studies and two of three review authors (SJ, ES and ED) independently screened each full text for inclusion, and recorded the reasons for exclusion of ineligible studies.

We resolved any disagreement through discussion or, if required, we consulted a third review author (SH). We identified and excluded duplicates and collated multiple reports of the same study so that each study, rather than each report, is the unit of interest in the review. We recorded the selection process in sufficient detail to complete a PRISMA flow diagram and Characteristics of excluded studies table (Moher 2009).

Data extraction and management

Three authors (SJ, ED, ES) screened citations in Covidence. One review author (SJ) piloted the data extraction form on at least one quantitative and one qualitative study before we extracted data from the rest of the included studies. We extracted data into a Microsoft Excel spreadsheet.

Quantitative studies

Three review authors extracted the following study characteristics from included studies.

  • Methods: study design, total duration of study, details of any 'run‐in' period, number of study centres and location, study setting, withdrawals and date of study.

  • Participants: N, mean age, age range, gender, severity of condition, diagnostic criteria, baseline lung function, smoking history, inclusion criteria and exclusion criteria.

  • Interventions: intervention, comparison, concomitant medications and excluded medications.

  • Outcomes: primary and secondary outcomes specified and collected (e.g. confidence intervals, P values, measurement scales used), and time points reported. Definitions used to diagnose an exacerbation were sought and recorded.

  • Notes: funding for studies and notable conflicts of interest of trial authors.

Three review authors (SJ, ES and ED) independently extracted outcome data from included studies. We planned to note in the Characteristics of included studies table if outcome data were not reported in a usable way. We resolved disagreements by consensus or by involving a third person/review author (SH). One review author (ED) transferred quantitative data into the Review Manager 5 (RevMan 5) file (Review Manager 2020). We double‐checked that data are entered correctly by comparing the data presented in the review with the study reports. A second review author (SJ) spot‐checked study characteristics for accuracy against the study report, and results were also checked by the Cochrane Airways Group statistician.

Qualitative studies

In order to capture context, two review authors (from SJ, ES, ED) extracted the following study characteristics from included studies.

  • Study details: country, study type (e.g. focus group, semi‐structured interviews, structured interviews, surveys), dates, source of funding, objectives.

  • Participants: N, mean age, age range, gender, severity of condition, diagnostic criteria, baseline lung function, smoking history, inclusion criteria and exclusion criteria.

  • Methods: sampling, setting (e.g. community or hospital), data collection (e.g. how the authors conducted the study, length of interviews, whether interviews were recorded, use of interview guide, data collected until achievement of thematic saturation), data analysis (e.g. method of analysis of transcripts, framework used, coding, thematic map).

  • Results: authors' interpretations, quotes from participants provided by authors.

Assessment of risk of bias

Quantitative studies

Three review authors (SJ, ES and ED) assessed risks of bias independently for each study using the criteria outlined in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2011). We resolved any disagreements by discussion or by involving another author (SH). We assessed the risk of bias according to the following domains.

  • Random sequence generation.

  • Allocation concealment.

  • Blinding of participants and personnel.

  • Blinding of outcome assessment.

  • Incomplete outcome data.

  • Selective outcome reporting.

  • Other potential bias.

We judged each potential source of bias as high, low or unclear and provided a quote from the study report together with a justification for our judgement in the risk of bias table. We summarised the risk of bias judgements across different studies for each of the domains listed. We considered blinding separately for different key outcomes where necessary (e.g. for unblinded outcome assessment, risk of bias for all‐cause mortality may be very different from a patient‐reported quality‐of‐life scale). We planned to note where information on risk of bias was from unpublished data or correspondence with a trialist, but we decided not to contact trial authors for clarification or unpublished data. When considering treatment effects, we took into account the risk of bias for the studies that contribute to that outcome via GRADE assessment of the certainty of the evidence (see below).

We presented a risk of bias table for all studies.

Qualitative studies

Two review authors (SJ, SH) assessed risk of bias independently for each study using the criteria outlined by the Cochrane Quality and Intervention Methods Group (Hannes 2011). We resolved any disagreements by discussion or by involving another review author (AH). We assessed the risk of bias according to the following criteria.

  • Quality of reporting (explicitness in reporting of all study aspects).

  • Methodological rigour (validity and reliability of study design and process).

  • Conceptual depth and breadth (are reported aims, rationale or theory reflected in the study design, process and findings?).

We used the Critical Appraisal Skills Programme (CASP) checklist (Critical Appraisal Skills Programme 2018) to assess risk of bias. We present risk of bias of studies in a table.

We assessed the risk of bias after the identification of relevant data to help us make judgements about the relative strength of messages in the included research.

Mixed‐methods studies

We planned to use the Mixed Methods Appraisal Tool (MMAT) to assess risk of bias (Hong 2018; Pluye 2011). Two review authors (SJ and ED) would have assessed risk of bias independently for each study using the criteria outlined by the MMAT. We planned to resolve any disagreements by discussion or by involving another author (SH). We planned to assess the risk of bias according to the following criteria.

  • Is there an adequate rationale for using a mixed‐methods design to address the research question?

  • Are the different components of the study effectively integrated to answer the research question?

  • Are the outputs of the integration of qualitative and quantitative components adequately addressed?

  • Are divergences and inconsistencies between quantitative and qualitative results adequately addressed?

  • Do the different components of the study adhere to the quality criteria of each tradition of the methods involved?

Measurement of treatment effect

Quantitative treatment effects

We analysed dichotomous data as an odds ratios (OR) and continuous data as the mean difference (MD) or standardised mean difference (SMD). Where data from rating scales were combined in a meta‐analysis, we ensured they were entered with a consistent direction of effect (e.g. lower scores always indicate improvement). We undertook meta‐analyses only where this was meaningful: that is, if the treatments, participants and the underlying clinical question were similar enough for pooling to make sense. Where we encountered substantial statistical or clinical heterogeneity, we presented data in graphs, but did not pool them. We described skewed data narratively (for example, as medians and interquartile ranges for each group).

Where multiple trial arms were reported in a single study, we included only the relevant arms for either comparison. Where two comparisons (e.g. intervention A versus intervention B) were combined in the same meta‐analysis, we planned to either combine the active arms or halve the control group to avoid double‐counting, but this was not necessary. If adjusted analyses were available (ANOVA or ANCOVA), we used these as a preference in our meta‐analyses. If both change from baseline and endpoint scores were available for continuous data, we used change from baseline unless there was low correlation between measurements in individuals. Where studies reported outcomes at multiple time points, we used endpoint data. We used intention‐to‐treat (ITT) or 'full analysis set' analyses where they were reported (i.e. those where data have been imputed for participants who were randomly assigned but did not complete the study) instead of completer or per protocol analyses.

Unit of analysis issues

Quantitative analysis

For dichotomous outcomes, we used participants, rather than events, as the unit of analysis. Where rate ratios were reported in a study, we planned to analyse them on this basis. We planned to meta‐analyse data from cluster‐RCTs where data had been adjusted (or could be adjusted by us), to account for the clustering.

Dealing with missing data

We planned to contact investigators or study sponsors in order to verify key study characteristics and to obtain missing numerical outcome data (e.g. when a study is identified as an abstract only), however when conducting the review, we decided not to contact authors for missing data.

Assessment of certainty of evidence

Quantitative data

We used the five GRADE considerations (risk of bias, consistency of effect, imprecision, indirectness, and publication bias) to assess the quality of the body of evidence as it relates to the following outcomes: Quality of life (SGRQ and CAT), exacerbations, functional status (6MWD), all‐cause hospital admissions, all‐cause mortality, anxiety and depression (unfortunately we did not specify these a priori). We used the methods and recommendations described in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2021), using GRADEpro GDT software (GRADEpro GDT; Guyatt 2008). We justified all decisions to downgrade the quality of studies in the footnotes of the table, and made comments to aid the reader's understanding of the results where necessary. We presented GRADE findings in a summary of findings table.

Qualitative data

We used the GRADE Confidence in the Evidence from Reviews of Qualitative Research (CERQual) approach to assess our confidence in the evidence of effectiveness arising from studies evaluating interventions (Lewin 2015). One review author (SJ) did this independently and Jane Noyes (from the Cochrane Qualitative and Implementation methods group) checked the completed assessment separately. This assessment allowed us to make judgements on the following four domains.

  • Methodological limitations of included studies.

  • Relevance of contributing studies to the research question.

  • Coherence of study findings.

  • Adequacy of data supporting the study findings.

We used the methods and recommendations described in Chapter 21 of the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2021). We summarised findings of the four domains for each outcome in a CERQual Qualitative Evidence Profile (Lewin 2015). We rated the overall assessment of confidence of evidence as high, moderate, low or very low. We presented CERQual findings in a summary of findings table (Glenton 2020). We justified all decisions to downgrade the quality of studies in the footnotes of the table, and we made comments to aid the reader's understanding of the review where necessary.

Assessment of heterogeneity

Quantitative data

We used the I2 statistic to measure heterogeneity among the studies in each analysis where possible. We conducted a meta‐analysis using a random‐effects model, as the interventions were varied. We explored possible causes of heterogeneity.

We considered the following I2 ranges in the analyses (Deeks 2021).

  • 0% to 40%: might not be important

  • 30% to 60%: may represent moderate heterogeneity

  • 50% to 90%: may represent substantial heterogeneity

  • 75% to 100%: considerable heterogeneity

We checked all data where we encountered substantial statistical heterogeneity. We did not pool data where there was substantial heterogeneity.

Assessment of reporting bias

Quantitative data

If we had been able to pool more than 10 studies, we planned to create and examine a funnel plot to explore possible small‐study and publication biases.

Data synthesis

Quantitative data

We used RevMan 5 and RevManWeb to perform quantitative data syntheses (meta‐analyses) (Review Manager 2020); and where data for population or interventions were similar, we pooled such data. Where we felt it was not appropriate to pool data, we present the data in forest plots to show the range of effect sizes where possible. Where we found clinical heterogeneity within the studies we identified, we grouped studies according to interventions and outcomes, and used the random‐effects model in the analyses (Ioannidis 2008). Where it was not possible to perform a meta‐analysis, we considered presenting data graphically and narratively using recommendations in the latest version of the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2021).

Qualitative data

Where studies were similar in design we planned to synthesise their data using a thematic analysis (Thomas 2008). This method would have allowed us to identify important or recurrent themes that arise from studies and summarise them under thematic headings. We planned to tabulate information, allowing identification of prominent themes and offer structured ways of dealing with the data in each theme. This type of synthesis would have helped us to identify emerging themes that described the experiences of participants, carers and HCPs when receiving or providing care to manage comorbid COPD.

We planned to initially analyse carer and HCP data separately to identify, for example, conflicting views or experiences. If we had found that the views and experiences had been similar, we planned to combine the two subgroups in subsequent syntheses.

Combining quantitative and qualitative data

We planned to use the methods and recommendations described in the Cochrane Handbook for Systematic Reviews of Interventions, and methods outlined by the Cochrane Qualitative and Implementation Methods Group (Higgins 2021 and Harden 2018, respectively). There are two main approaches to integrating qualitative and quantitative data: sequential and convergent. The sequential approach involves synthesising qualitative and quantitative analyses separately, followed by using common frameworks to integrate the findings across syntheses. We planned to use the sequential approach to integrate qualitative data to explain quantitative findings. We planned to analyse quantitative data at the first stage, followed by synthesis of qualitative data in the second stage.

Integrating the qualitative syntheses with the quantitative analyses can be achieved by using a 'matrix' to compare and contrast findings across both types of evidence. The matrix will help to identify gaps in the evidence. This approach can help us to understand why heterogeneity exists that we may find in the quantitative analyses. Other approaches include the development of a 'logic model' by providing a common framework within which both quantitative and qualitative evidence can contribute (Harden 2018). We acknowledge that the methods for integration are dependent on the quantity of studies and extracted evidence available, and quality of description within included studies (e.g. intervention content, context, and study findings). For divergent data (qualitative data that does not match the quantitative data), we aimed to resolve divergence (deviation of the qualitative data from the quantitative data, or vice versa (Erzberger 1997)) where possible by providing a narrative explanation according to research and knowledge of the topic area.

Subgroup and investigation of heterogeneity

We investigated clinical heterogeneity in the quantitative studies. We planned to perform subgroup analyses using the following prespecified groups, but we used the subgroup function in RevMan to present data by intervention type, and did not complete the following subgroup analysis due to lack of a sufficient number of studies.

  • Number of comorbidities in addition to COPD (≤ 2 conditions versus ≥ 3 conditions).

  • Duration of intervention (< 3 months versus ≥ 3 months).

We planned to use the following outcomes in the subgroup analyses.

  • Quality of life.

  • Functional status.

  • Hospital admissions.

  • Exacerbations.

We planned to use the formal test for subgroup interactions in Review Manager 5 (Review Manager 2020).

Sensitivity analysis

We planed to carry out sensitivity analyses excluding studies in which one or more risk of bias domains is judged to be at high risk of bias.

Assessment of bias conducting the systematic review

We conducted the review according to our published protocol and justified any deviations from it in the 'Differences between protocol and review' section.

Review author reflexivity

We maintained a reflexive stance throughout the stages of the review process, from study selection to data syntheses. Progress was discussed regularly among the team and decisions made were discussed critically. As a review author team, our expertise has been listed in Contributions of authors. Based on our collective and individual experiences as clinicians, academics and researchers, we anticipated that the findings of our review would identify a combination of organisational, professional and individual factors influencing the implementation of targeted interventions and approaches to care for people with COPD to manage comorbidities. ED has overseen progress of the review, a process that has allowed her to document and reflect on any decisions made.

Results

Description of studies

Results of the search

Quantitative studies

The literature search run on 8 January 2021 for reports of RCTs identified a total of 17,069 search results after duplicates were removed. We used Cochrane’s Screen4Me workflow to help identify potential reports of randomised trials: 486 records were excluded by Cochrane Crowd Known Assessments, 6635 records were excluded by the Cochrane RCT Classifier, and 4528 records were excluded by Cochrane Crowd screeners After this initial assessment, we screened the remaining 5420 records and excluded 5252 records after reading the titles and abstracts. We obtained 168 articles for full‐text review, excluded 142 of these with reasons (see Characteristics of excluded studies), and identified three studies that require further assessment (Characteristics of studies awaiting classification).

We included seven studies (19 references) and found a further four ongoing studies (Characteristics of ongoing studies). See Figure 1 for an overview of the study selection process.

Qualitative studies

The search for qualitative studies, run on 8 January 2021, identified a total of 9370 search results after duplicates were removed. We excluded 9257 records after reading the titles and abstracts, and obtained 113 articles for full‐text assessment. We excluded 108 records with reasons (Characteristics of excluded studies) and identified two ongoing studies (four references), one of which is an ongoing RCT with a planned qualitative element (Characteristics of ongoing studies).

We included one study in the qualitative synthesis. See Figure 2 for an overview of the study selection process for qualitative studies.


Study flow diagram for qualitative studies

Study flow diagram for qualitative studies

Included studies

Quantitative

We included seven studies in the quantitative synthesis. An overview of the study characteristics is given in Table 3.

Open in table viewer
Table 3. Summary of interventions

Study

Duration (weeks)

Intervention

Comparison

Setting

Provider

Materials/method

Delivery

Tailoring

Pharmacotherapy

EUCTR2010‐021412‐42‐GB

4

Pharmacotherapy: inhaler optimisation and best supportive care

UC

Outpatient

NR

Evohaler 100 μg, Spiriva 18 mg via Handihaler, fluticasone propionate 500 μg

Inhaler

Considered the poor prognosis of patients with lung cancer: intervention group treated with maximum inhaled therapy

Gottlieb 2020

25

Management: optimising COPD treatment

UC

Outpatient clinic

Pulmonary physician

Two visits at 12 and 24 weeks in an outpatient clinic where the adjustment of COPD treatment was considered

Dialogue with the participants

Adjustment of COPD treatment considered at each visit

Rehabilitation

Budnevskiy 2015

52

Pulmonary rehabilitation

UC

NR

NR

Series of seminars covering treatment and prevention of COPD and MS. Physical training: therapeutic exercises.

Group seminars

Exercises took into account concomitant MS

Gurgun 2013

8

Pulmonary rehabilitation and nutritional support

PR or UC

Outpatient clinic

Dietician

Dietary counselling and oral supplementation in combination with exercise training

Supervised exercise training, dietary counselling

Daily nutrition intake of the patient was checked

McNamara 2013b

8

Water‐based exercise training

Land‐based exercise training or UC

Hospital hydrotherapy pool

Physiotherapist

Exercise in a hydrotherapy pool: warm‐up, lower limb endurance, upper limb endurance, and cool down

Supervised exercise

Training intensity measured, participants chose the most comfortable level of water immersion

Organisation of care

Rose 2018

52

Case management

UC

Outpatient

Case‐manager

Education based on 'Living Well with COPD', ongoing communication with physician and hospital specialist

Education session at enrolment, telephone consultations

Individualised action plan

Walker 2018

39

Telemonitoring

UC

Outpatient

Study nurse

CHROMED monitoring platform

Wearable device

Respiratory alert triggered contact with the nurse if worsening was detected

Multicomponent intervention

Bernocchi 2018

17

Inpatient rehabilitation, personalised discharge, nurse telephone support and telemonitoring, physiotherapist personalised maintenance rehabilitation

Inpatient rehabilitation, UC

Participants' home

Nurse; physiotherapist

Exercise programme, mini‐ergometer, pedometer and diary. Participants provided with a pulse oximeter, and a portable one‐lead ECG

PT instruction; weekly phone call with NT; weekly phone call with PT

Personalised exercise programme

Abbreviations: COPD: Chronic obstructive pulmonary disease; MS: metabolic syndrome; NT: Nurse Tutor; PT:Physiotherapist tutor; UC: usual care

Participants

Participants in included studies had COPD that ranged from mild to very severe. The studies involved people with a range of comorbidities. Three studies accepted people with COPD plus one or more of a range of specified comorbidities (McNamara 2013b; Rose 2018; Walker 2018). The participants in McNamara 2013b had either musculoskeletal or neurological comorbidities, or obesity. Participants in Rose 2018 had two or more comorbidities. The most common comorbid conditions were cardiovascular disease, diabetes, osteopenia/osteoporosis, and depression. Walker 2018 included participants with one or more comorbid conditions including hyperlipidaemia, diabetes, congestive heart failure and/or ischaemic heart disease, sleep‐disordered breathing and osteoporosis. The other four studies included people with COPD plus one specific comorbidity: lung cancer (EUCTR2010‐021412‐42‐GB); lung cancer, or head and neck cancer (Gottlieb 2020); cardiovascular disease (Bernocchi 2018), and metabolic syndrome (Budnevskiy 2015). See Table 4 for more information.

Open in table viewer
Table 4. Summary of baseline characteristics

Study

COPD severity

Comorbidities (%)

Intervention

Comorbidities (%)

Control

Ethnicity

Male (%)

Age (Mean, SD)

Intervention

Control

Intervention

Control

Bernocchi 2018

Mild to very severe

Cardiovascular disease (100%)

Cardiovascular disease (100%)

NR

88

75

71 (9)

70 (9.5)

Budnevskiy 2015

Moderate

Metabolic syndrome (100%)

Metabolic syndrome (100%)

NR

66

71

NR

NR

EUCTR2010‐021412‐42‐GB

NR

Lung cancer (100%)

Lung cancer (100%)

NR

34

38

68 (59 to 75)*

67 (61 to 71)*

Gottlieb 2020

Mild to severe

Lung cancer (84%); head‐and‐neck cancer (16%); Ischaemic heart disease/heart failure (12.3%); Depression/anxiety (3.5%); Osteoporosis (12.3%); Cerebrovascular disease (14%); Diabetes (14%)

Lung cancer (82.5%); head‐and‐neck cancer (17.5%); ischaemic heart disease/heart failure (10.5%); Depression/anxiety (3.5%); Osteoporosis (10.5%); Cerebrovascular disease (8.8%);
Diabetes (8.8%)

NR

58

69

67.8 (8.3)

67.2 (8.1)

McNamara 2013b

NR

Water‐based group:

Musculoskeletal (50%);
neurological (5.5%);
obesity (44.5%)

Land‐based group:

Musculoskeletal (65%);
neurological (5%);
obesity (30%)

Musculoskeletal (47%);
neurological (0%);
obesity (53%)

NR

Water‐based group: 28

Land‐based group: 50

47

Water‐based group: 72 (10)

Land‐based group: 73(7)

70 (9)

Rose 2018

Moderate to severe

Two or more comorbidities: cardiovascular disease (75%); diabetes (18%); depression (17%); Osteopenia and osteoporosis (30%); Gastro‐oesophageal reflux disease (14%); Hypothyroidism (9%); Osteoarthritis (9%); Glaucoma and cataracts (9%); Cachexia and malnutrition (10%); Chronic kidney disease (7%); Anxiety (6%); Peripheral muscle dysfunction (6%); Obstructive sleep apnoea (5%); Lung cancer (6); Cerebrovascular accident (3%)

Two or more comorbidities: cardiovascular disease (76%); diabetes (22%); depression (20%); Osteopenia and osteoporosis (29%); Gastro‐oesophageal reflux disease (12%); Hypothyroidism (9%); Osteoarthritis (9%); Glaucoma and cataracts (9%); Cachexia and malnutrition (8%); Chronic kidney disease (7%); Anxiety (7%); Peripheral muscle dysfunction (6%); Obstructive sleep apnoea (6%); Lung cancer (6%); Cerebrovascular accident (4%)

NR

50

44

71 (9.2)

71 (9.7)

Walker 2018

Mild to very severe

One or more non‐pulmonary comorbidities: Congestive heart failure (12%); ischaemic heart disease (25%; Congestive heart failure plus ischaemic heart disease (12%); hypertension (72%); Sleep‐related disordered breathing (11%); Osteoporosis (17%); Hyperlipidemia (53%)

One or more non‐pulmonary comorbidities: Congestive heart failure (8%); ischaemic heart disease (23%; Congestive heart failure plus ischaemic heart disease (13%); hypertension (68%); Sleep‐related disordered breathing (6%); Osteoporosis (15%); Hyperlipidemia (58%)

NR

66

66

71 (66.0 to 75.8)*

71 (65.3 to 76.0)*

*median, interquartile range

Abbreviations: NR: not reported

The proportion of men in the studies ranged from 28% to 88%. Mean age ranged from 67 to 72 year. Ethnicity was not reported in the studies.

Interventions

Two studies delivered pharmacological interventions. EUCTR2010‐021412‐42‐GB compared inhaler optimisation plus usual care, with usual care alone. EUCTR2010‐021412‐42‐GB tailored the intervention by considering the poor prognosis of participants with co‐existing lung cancer, and the intervention group was given the maximum inhaled therapy. Gottlieb 2020 optimised the COPD treatment for the intervention group while the control group received usual care; the tailoring element was that the therapy was re‐considered at each six‐monthly visit.

Two studies delivered a rehabilitation intervention. Budnevskiy 2015 compared a pulmonary rehabilitation programme, where the exercises took into account metabolic syndrome comorbidity, and included education, physical training and nutritional recommendations, versus usual care. McNamara 2013b compared water‐based exercise training with land‐based exercise training or usual care. The tailoring element was that the exercises were carried out in water, which provides more support to people's bodies, and participants were able to choose the most comfortable level of water immersion. We used both comparisons in the review in separate comparisons (water‐ vs land‐based training and water‐based training versus usual care).

Two studies were categorised under organisation of care. Rose 2018 assigned case‐managers to deliver case management and this group was compared to usual care. Case‐managers tailored the intervention to the individual by providing an individualised action plan. Walker 2018 compared telemonitoring with usual care. Participants in the treatment arm were given a wearable device and used the CHROMED monitoring platform, and there was a nested qualitative study that collected feedback from participants, researchers and HCPs in the study (Middlemass 2017).

All participants in Bernocchi 2018 received inpatient rehabilitation, then people were randomised to a package of care (including personalised discharge, nurse telephone support and telemonitoring, physiotherapist‐personalised maintenance rehabilitation) or usual care.

Setting

Four of the seven studies were conduced in single‐centre hospital‐based settings from Australia (McNamara 2013b), Denmark (Gottlieb 2020), Russia (Budnevskiy 2015), and United Kingdom (EUCTR2010‐021412‐42‐GB). Three were multicentre studies, one based in two community teaching hospitals in Canada (Rose 2018), one based in three centres (respiratory, cardiology and telemedicine) in Italy (Bernocchi 2018), and one in multiple centres in Estonia, Slovenia, Spain, Sweden and United Kingdom (Walker 2018).

Study design

Six of the studies were parallel RCTs, plus one RCT (Walker 2018), which included a nested qualitative study of a subset of the participants and people involved in running the study (Middlemass 2017).

Trial duration

The duration of the trials ranged from 4 to 52 weeks (Table 5). We used the data from the latest endpoint in those studies that reported multiple time points during the intervention duration (Bernocchi 2018; EUCTR2010‐021412‐42‐GB; Gottlieb 2020; Rose 2018). The remaining studies reported outcome data at the end of the intervention (Budnevskiy 2015; McNamara 2013b; Walker 2018). None of the studies reported follow‐up data after the intervention was stopped.

Open in table viewer
Table 5. Summary of relevant quantitative study outcomes

Study ID

Outcome domain

Outcome measure

End point time (weeks)

Bernocchi 2018

All‐cause hospital admissions

Number of events

17

Functional status

6MWT

Functional status

PASE

Mortality (all‐causes)

Number of events

Quality of life

MLHFQ

Quality of life

CAT

Quality of life

Dyspnoea MRC

Respiratory hospital admissions

Number of events

Functional status

6MWT

Quality of life

CAT (total)

Quality of life

SGRQ (total)

Quality of life

Dyspnoea MRC

Budnevskiy 2015

Functional status

6MWT

52 weeks

Quality of life

CCQ

Quality of life

CAT (total)

Quality of life

SGRQ (total)

EUCTR2010‐021412‐42‐GB

Adverse events

Number of events

4

Mortality (all‐causes)

Number of events

Quality of life

Dyspnoea

Gottlieb 2020

Mortality (all‐causes)

Number of events

25

Quality of life

CAT (total)

McNamara 2013b

Anxiety symptoms

HADS‐A (anxiety)

8

Depression symptoms

HADS‐D (depression)

Functional status

ESWT

Functional status

6MWT

Functional status

ISWT

Functional status

ESWT

Quality of life

CRDQ dyspnoea

Rose 2018

All‐cause hospital admissions

Mean & SD

All‐cause hospital admissions

Risk difference

52

Anxiety symptoms

HADS‐A (anxiety)

Depression symptoms

HADS‐D (depression)

Exacerbations

Exacerbation (ED visit)

Mortality (all‐causes)

Number of events

Quality of life

SGRQ (total)

Walker 2018

All‐cause hospital admissions

Hospitalisation rate

39

All‐cause hospital admissions

Number of people experiencing one or more events

Exacerbations

Exacerbation rate moderate exacerbations

Mortality (all‐causes)

Number of events

Quality of life

CAT (total)

Quality of life

PHQ‐9

Quality of life

EQ‐5D utility

Quality of life

EQ‐5D VAS

6MWT: 6‐Minute Walk Test; CAT: COPD Assessment Test; CCQ: COPD clinical questionnaire; CRQ: Chronic Respiratory Disease Questionnaire; CSES: Coping Self‐Efficacy Scale; EQ‐5D: EuroQuol‐5D; ESWT: Endurance Shuttle Walk Test; HADS: Hospital Anxiety and Depression Scale; ICFS: Identity‐Consequence Fatigue Score; ISWT: Incremental Shuttle Walk Test; MLHFQ: Minnesota Living with Heart Failure Questionnaire; MRC: Medical Research Council; PASE: Physical Activity Profile; PHQ‐9: Patient Health Questionnaire‐9; PIH: Partners in Health scale; SGRQ: St George's Respiratory Questionnaire; VAS: Visual Analogue Scale.

Outcomes

The studies reported a range of outcomes, summarised in Table 5. Briefly, measures of all‐cause hospitalisations were reported by Bernocchi 2018, Rose 2018 and Walker 2018. Two studies reported functional status (Bernocchi 2018; McNamara 2013b). All seven studies reported one or more measures of quality of life (Bernocchi 2018; Budnevskiy 2015; EUCTR2010‐021412‐42‐GB; Gottlieb 2020; McNamara 2013b; Rose 2018; Walker 2018) (Table 5).

Qualitative

We included one study in the qualitative synthesis. An overview of the study characteristics is given in Characteristics of included studies table.

Participants

Twenty‐one participants with severe COPD aged between 60 and 99 years and their partners or relatives if available, from the pilot study and a subset of the main intervention group in Walker 2018 were included in Middlemass 2017. Middlemass 2017 did not report the participants' comorbidities. However, the CHROMED RCT linked to this publication included people with COPD who had one or more comorbidities (congestive heart failure and/or ischaemic heart disease hypertension, sleep‐related disordered breathing, osteoporosis, or hyperlipidaemia) (Walker 2018).

Interventions

Middlemass 2017 explored participants' perceptions of using telehealth equipment at home (those who were assigned to the intervention arm of Walker 2018).

Setting

Middlemass 2017 was a nested qualitative study linked to Walker 2018, and was based in participants' homes in Estonia, Italy, Norway, Slovenia, Spain, Sweden and United Kingdom (two sites).

Study design

Middlemass 2017 was an instrumental, collective study design that used qualitative interviews. They conducted a framework thematic analysis, using the Health Information Technology Acceptance Model (HITAM) as a guide.

Duration of study

Qualitative interviews were conducted once, shortly after the equipment was installed, and at the end of the study at 39 weeks.

Themes

A range of themes are summarised in Table 6.

Open in table viewer
Table 6. First and second order constructs of qualitative studies

Study

Aims

Main themes and example quotes provided in study report

Author comments provided in the study report

Conclusions ‐ review author team interpretation of study results

Middlemass 2017, UK

To explore the usefulness of the HITAM for understanding acceptance of HIT in older people (≥ 60 years age) with COPD and associated heart diseases

Health status, beliefs and concerns

Unchanging nature of condition: Patients accepted that their chronic condition was unlikely to change and that ageing and (eventual) death was inevitable: "I'm getting older and I'm not going to get any better. I haven't got young genes to repair everything. So, if I can pummel along the way I am, I'll accept it".

Acceptance of illness: Some patients had accepted the life‐restricting (and sometimes life‐threatening) limitations of their LTCs

Beliefs about condition influence motivation to engage in HIT

Withdrawal of face‐to‐face communication: "But I would hope they would still do their person‐ to‐person contact [and] that they wouldn't just forget."

Concern of losing face to face contact: Patients were concerned that they would lose face‐to‐face communication with their HCP when using TM

HCP face to face interaction is re‐assuring that their condition is actively being monitored

Reminder of illness and anxiety :"This is reminding me every day, then I should think I wonder what my reading is, how good it is or how bad it is and I thought no, get away from illness you know. Every time as soon I started thinking about it, I started thinking about my illness..."

Fear of illness: Patients perception of telemonitoring was linked to fear of reminders of how serious their condition was, which led to them not continuing with home monitoring

Belief that HIT causes anxiety about condition which can lead to non‐adherence/

Information

Subjective norms:"I think if my very close relatives…and if the GP said it is essential… I would say I'm definitely going ahead with it"

Increased motivation to comply with HIT: Close relatives and GP influenced and increased individuals' perceptions of using HIT

Input from HCP and relatives increases acceptance to use HIT

Technology

Unreliable technology:"...a couple of times it didn't go through very well, but that was an internet problem".

Unreliable technology: Poor internet connectivity and data transmission in rural areas led to generation of technical alerts, which led to the study research nurse visiting the patient to find out what the problem was.

HIT can be beneficial for those who cannot visit HCP face to face, however, this can be limited by Internet connectivity

HIT Self‐efficacy:"The very first time I really got panicked. But then the next day when I did it, it was easier, but I was at the start of a chest infection, which did affect me… It helped my husband stood beside me and was chatting saying yeah you're doing fine, not long to go, just a little bit of encouragement"

Increased self‐efficacy: Both HCPs and patients' significant others were key to them using the TM equipment.

Patients' relatives and HCPs can help to reduce apprehension of using HIT in the initial stages

Perceived usefulness

Daily monitoring of conditions: "...I feel more comfortable knowing that somebody's checking it all the time, you know they're looking at it every day..."

Confidence of daily monitoring: Patients perception of being linked to a HCP checking data and ready to act on change in health status led them to feel safe about using HIT.

Patients' perceptions are dependent on knowing that HCP involvement is linked to HIT

Factors affecting usefulness

Lack of feedback: "I'm in a vacuum. I'm doing something, I'm sending it off to you, [but] there's no feedback..."

Lack of feedback: there was lack of two‐way communication between the patient and HCP

Lack of feedback from HCP resulted in reduced perception of usefulness of HIT

Behaviour

Self‐management and health care utilisation: Patients felt that their condition stabilised after joining the study and did not need to go to the GP so often: "I've been less to the surgery... Because I think it's helped me sort everything out. I'm much better on the medication I'm on now for my blood pressure."

Improved self‐management and reduced need to see the GP: Patients' condition stabilised whilst enrolled in the study, and GP visits also declined.

HIT may lead to changes in behaviour towards improving patients' self‐management and a reduced need to visit the GP

Abbreviations: HITAM: Health Information Technology Acceptance Model

Risk of bias in included studies — quantitative

See Figure 3 for a summary of the risk of bias judgments.


Risk of bias summary

Risk of bias summary

Allocation

We judged five studies to be at low risk of bias for random sequence generation (Bernocchi 2018; Gottlieb 2020; McNamara 2013b; Rose 2018; Walker 2018), while two were unclear (Budnevskiy 2015; EUCTR2010‐021412‐42‐GB). We judged three studies to be at low risk of bias for allocation concealment (Bernocchi 2018; McNamara 2013b; Walker 2018), while four were judged to be at unclear risk of bias (Budnevskiy 2015; EUCTR2010‐021412‐42‐GB; Gottlieb 2020; Rose 2018).

Blinding

Where the interventions involved a complex intervention such as pulmonary rehabilitation, case management or telemonitoring, it was not possible to blind participants or personnel because they would know whether they were receiving the intervention or not and were therefore judged to be at high risk of bias. This applied to most of the studies. The two studies that involved optimisation of medication (EUCTR2010‐021412‐42‐GB; Gottlieb 2020), were run as open‐label studies and so were also judged to be at high risk of bias.

Blinding of outcome assessors only protects studies if the participants are also blinded, or if outcomes are objective or not self‐reported or both. So while we have awarded some low risk of bias, they may not be protected. Three studies explained that they blinded outcome assessors, earning them a low risk of bias assessment for this domain (Bernocchi 2018; McNamara 2013b; Walker 2018). While the reporting was not clear enough to make a judgement for two studies (Budnevskiy 2015; Gottlieb 2020), two studies did not blind the outcome assessors so they were judged to be at high risk of bias (EUCTR2010‐021412‐42‐GB; Rose 2018).

Incomplete outcomes data

Incomplete outcome data relates to attrition or withdrawal from the study. Two studies had zero to low levels of dropouts (EUCTR2010‐021412‐42‐GB; Rose 2018) and were judged at low risk of bias for this domain. One study did not provide any information on dropouts or how many people completed the study, so we judged the domain as unclear risk (Budnevskiy 2015). The remaining four studies were at high risk of bias for the following reasons; Bernocchi 2018 reported more dropouts in the control group compared to the intervention group (37.5% versus 19.6%); Gottlieb 2020 had a high loss to follow‐up in both intervention and control groups (28% and 33% respectively), and McNamara 2013b reported more dropouts (25%) in the land‐based training group because of exacerbation of comorbidity or loss of interest in the study. Sixteen per cent of participants in the water‐based training group dropped out due to skin tear or general fatigue. Walker 2018 had high and unbalanced dropout rates and 5% withdrew as they could not use the equipment. Rose 2018 reported that missing data were problematic for their secondary outcomes measured at 52 weeks.

Selective reporting

Four studies were at low risk of bias because protocols were available and outcomes specified were reported in the full text (EUCTR2010‐021412‐42‐GB; McNamara 2013b; Rose 2018; Walker 2018). One study was at unclear risk of bias because there was no published protocol (Gottlieb 2020) and two studies were at unclear risk because they did not report a standard deviation (SD) to accompany a mean for some outcomes (Budnevskiy 2015; Bernocchi 2018). We did not contact the authors because we did not think that clarifying this information would change the outcome of the review.

Other bias

We did not observe any other risks of bias.

Risk of bias in included studies — Qualitative

See Table 1 for critical appraisal of qualitative studies.

Overall, there were some concerns about risk of bias in Middlemass 2017. We could not be sure if the study design was justified in addressing the aims of the study. Similarly, methods for collecting the data may not have addressed the research question because it was not clear if the interviews were in‐depth or semi‐structured. There was no justification provided for using interviews rather than focus groups. Authors did not provide details on data saturation. The researcher and participant relationship was not considered, as the role of the interviewer was not examined. We did not have any concerns with ethical issues as they had been taken into consideration. The data analysis was rigorous and the process was well described. There was a clear statement of findings which were explicit and well organised.

Effects of the interventions ‐ quantitative

For an overall summary, see summary of findings Table 1 and summary of findings Table 2.

Intervention versus usual care

For this comparison we included seven studies with 1177 participants.

Quality of life (primary outcome)

Quality of life was reported in several studies using a range of measures: St George’s Respiratory Questionnaire (SGRQ), COPD Assessment Test (CAT), Chronic Respiratory Disease Questionnaire (CRQ), Minnesota Living with Heart Failure Questionnaire (MLHFQ) and EuroQuol‐5D VAS (EQ‐5D).

In this comparison, we used water‐based versus usual care for McNamara 2013b.

SGRQ

Two studies reported the SGRQ total (Budnevskiy 2015; Rose 2018). Budnevskiy 2015, a rehabilitation study of 70 participants, reported SGRQ total scores at 52 weeks follow‐up. Uncertain evidence suggests that pulmonary rehabilitation may result in a decrease (improvement) in SGRQ score compared to usual care (mean difference (MD) −10.85, 95% confidence interval (CI) −12.66 to −9.04; low‐certainty evidence; Analysis 1.1; summary of findings Table 1).

The second study (Rose 2018) provided outpatient case management and reported on the SGRQ total score at 52 weeks follow‐up. The mean difference of 4 was reported with a very tight confidence interval This is likely to overestimate the benefit of the intervention. There were also some discrepancies between the published and supplemental data. We therefore decided not to present the results graphically.

CAT

Four studies from four different categories of intervention reported the CAT total score; these were not pooled because the interventions were too diverse (Analysis 1.2).

One rehabilitation study (Budnevskiy 2015) showed that pulmonary rehabilitation probably improves quality CAT score at 52 weeks follow‐up (MD −8.02, 95% CI −9.44 to −6.60; moderate‐certainty evidence; Analysis 1.2; summary of findings Table 1). The multicomponent telehealth intervention trialled in Bernocchi 2018 probably improves CAT score at 17 weeks follow‐up (MD −6.90, 95% CI −9.56 to −4.24; moderate‐certainty evidence; Analysis 1.2; summary of findings Table 1). Evidence is uncertain about effects of pharmacotherapy optimisation or telemonitoring interventions on quality of life as there is little to no difference in effect compared to usual care at 25 weeks (MD 0.00, 95% CI −3.40 to 3.40) or at 39 weeks follow‐up (MD −0.41, 95% CI −2.19 to 1.37) respectively (Walker 2018; Gottlieb 2020) (low‐certainty evidence; Analysis 1.2; summary of findings Table 1).

CRQ domains (dyspnoea, fatigue, emotion, mastery)

One study reported results from the CRQ which was presented as the four subdomains rather than an overall domain (McNamara 2013b; Analysis 1.3). Water‐based exercise training may offer a slight improvement in quality of life, as measured by CRQ dyspnoea (MD 3.25, 95% CI 0.90 to 5.60; 1 study, 33 participants), fatigue (MD 4.70, 95% CI 2.40 to 7.00), and emotion (MD 3.10, 95% CI 0.10 to 6.10) domains, compared with usual care at eight weeks follow‐up. There may be little of no difference in the mastery domain (MD 1.90, 95% CI −0.20 to 4.00).

MLHFQ

One study (Bernocchi 2018) reported on the MLHFQ and showed that a targeted multicomponent intervention may result in a small but clinically insignificant improvement in this outcome at 17 weeks follow‐up (MD −10.06, 95% CI −16.27 to −3.85; 92 participants; Analysis 1.4). The minimal clinically important difference (MCID) is 45 (Behlouli 2009).

EQ‐5D

One study reported on the EQ‐5D, but reported two scores, the VAS and the utility domain (Walker 2018). For both scores on the standardised mean difference (SMD) scale, there is little to no difference in the effect of targeted interventions on VAS (SMD −0.02, 95% CI −0.24 to 0.20; 303 participants; Analysis 1.5) or utility scores (MD −0.01, 95% CI −0.23 to 0.21; 303 participants) at 39 weeks follow‐up.

Exacerbations (primary outcome)

One study reported exacerbations as the number of people experiencing one or more exacerbations resulting in an emergency department (ED) visit (Rose 2018). Case management may result in little to no difference in the number of people experiencing exacerbations at 52 weeks follow‐up compared to usual care, with the evidence very uncertain (OR 1.09, 95% 0.75 to 1.57; 470 participants; very low‐certainty evidence; Analysis 1.6; summary of findings Table 1). Rose 2018 also reported mean exacerbations per person, which showed case management may result in little to no difference in effect at 52 weeks follow‐up, regardless of the issue of missing data reported (Analysis 1.7).

Functional status (secondary outcome)

Three rehabilitation studies reported at least one outcome relating to functional status (Bernocchi 2018; Budnevskiy 2015; McNamara 2013b). The specific outcomes reported were the six‐minute walk distance (6MWD), incremental shuffle walk test (ISWT), and endurance shuttle walk test (ESWT).

6MWD

Data from two studies reported that tailored rehabilitation interventions (pulmonary rehabilitation and water‐based exercise) are likely to result in a large increase in 6MWD at mean 38.8 weeks follow‐up (MD 60.40 metres, 95% CI 44.26 to 76.54; I2 = 0%; 2 studies, 100 participants; low‐certainty evidence; Analysis 1.8; summary of findings Table 1). One study reported that a multicomponent intervention is likely to result in a large increase in 6MWD at 52 weeks follow‐up, with evidence uncertain (MD 75.00 metres, 95% CI 28.06 to 121.94; 80 participants; low‐certainty evidence; Analysis 1.8; summary of findings Table 1). The MCID for the 6MWT is 25 to 35 meters (Holland 2013).

ISWT

One study (McNamara 2013b) with 30 participants reported that a water‐based exercise may improve ISWT compared to usual care at eight weeks follow‐up (MD 50 metres, 95% CI 20 to 80; Analysis 1.9). However, with the small number of participants, we were uncertain about the results.

ESWT

One study with 30 participants reported ESWT at eight weeks follow‐up (McNamara 2013b). The results favoured the water‐based exercise group compared to usual care (MD 371 metres, 95% CI 120 to 622; Analysis 1.10), although with so few participants and a wide confidence interval, we are uncertain about the results.

All‐cause hospital admissions (secondary outcome)
Number of people experiencing one or more hospitalisations (all‐cause)

Two studies reported the number of people experiencing one or more hospitalisations for any cause (Bernocchi 2018; Walker 2018). We did not pool the data owing to variations in the interventions. The evidence is very uncertain about the effects of telemonitoring compared to usual care at 39 weeks (OR 0.91, 95% CI 0.55 to 1.50; 312 participants; low‐certainty evidence; Analysis 1.11; summary of findings Table 1). A multicomponent intervention may reduce the number of people experiencing hospitalisations at 17 weeks, but evidence is uncertain (OR 0.31, 95% CI 0.14 to 0.67; 112 participants; low‐certainty evidence; Analysis 1.11; summary of findings Table 1). Although the sample size is small, the trial was sufficiently powered (80% with a P value < 0.05), which was reported in the publication methods (Bernocchi 2018). The study is at risk of bias and should be interpreted with caution.

Mean hospitalisations per person

One study reported the mean number of hospitalisations per person (Rose 2018). There may be little to no difference in case management on mean hospitalisations compared to usual care at 52 weeks (MD −0.10, 95% CI −0.40 to 0.20; 470 participants; Analysis 1.12).

Respiratory hospital admissions (secondary outcome)

One study reported the number people with respiratory‐related hospital admissions (Bernocchi 2018). There may be little to no difference in a multicomponent intervention on respiratory hospital admissions compared to usual care at 17 weeks (OR 0.49, 95% CI 0.17 to 1.44; 112 participants; Analysis 1.13).

Mortality — all‐cause (secondary outcome)

Five studies reported the number of deaths during the trial (Bernocchi 2018; EUCTR2010‐021412‐42‐GB; Gottlieb 2020; Rose 2018; Walker 2018). Overall there were 90 deaths in a total of 1071 participants (8.4%).

Evidence from two pharmacotherapy studies showed that optimising pharmacotherapy may result in little to no difference in mortality, with a pooled OR of 0.55 (95% CI 0.23 to 1.35; I2 = 0%; 2 studies, 177 participants; very low‐certainty evidence; Analysis 1.14; summary of findings Table 1).

Evidence from two studies showed that effects of organisation‐of‐care interventions (case‐management and telemonitoring) may result in fewer deaths compared to usual care at mean 46.7 weeks follow‐up (OR 0.56, 95% CI 0.33 to 0.96; ; I2 = 0%; 2 studies, 782 participants; low‐certainty evidence; Analysis 1.14; summary of findings Table 1).

Evidence from one multicomponent intervention study (Bernocchi 2018) was very uncertain (OR 1.00, 95% CI 0.06 to 16.39; 112 participants; very low‐certainty evidence; Analysis 1.14; summary of findings Table 1). Two studies did not report mortality as an outcome.

Pain (secondary outcome)

None of the studies reported on pain.

Anxiety (secondary outcome)

Two studies reported results from the HADS‐anxiety questionnaire. Water‐based exercise may have little or no effect on anxiety at eight weeks follow‐up compared to no exercise, and we are uncertain about the evidence (MD −1.00, 95% CI −3.50 to 1.50; 1 study, 33 participants; Analysis 1.15). A case‐management intervention may have an effect on anxiety at 52 weeks, (Rose 2018) but we are very uncertain about the evidence due to risk of bias. The confidence interval was very tight, and there were discrepancies between the results in the main text and supplementary information. We have therefore decided not to present the results.

Depression (secondary outcome)

Two studies reported results from the HADS‐depression questionnaire (McNamara 2013b; Rose 2018). Water‐based exercise may have little to no effect on the HADS‐Depression score at eight weeks follow‐up compared with usual care (Analysis 1.16). Again there were discrepancies in the reporting of results in Rose 2018, so data are not presented here.

Intervention versus active comparator

In this comparison, we used water‐based exercises versus land‐based exercises for the McNamara 2013b study (30 participants).

Quality of life (primary outcome)
CRQ domains (dyspnoea, fatigue, emotion, mastery)

One study reported results from the four domains of the CRQ (McNamara 2013b). Water‐based exercise training may result in little to no difference in the dyspnoea domain, compared with land‐based exercise training (MD 1.70, 95% CI −0.65 to 4.05; 1 study, 38 participants, Analysis 2.1), emotion domain (MD 2.90, 95% CI −0.10 to 5.90), and the mastery domain (MD 1.10, 95% CI −0.90 to 3.10). A possible small difference in the fatigue domain was reported (MD 3.10, 95% CI 0.80 to 5.40).

Exacerbations (primary outcome)

None of the studies reported on exacerbations.

Functional status (secondary outcome)

One rehabilitation study reported 6MWD, ISWT and ESWT as change from baseline to eight weeks (McNamara 2013b).

6MWD

One study reported the 6MWD as change from baseline (McNamara 2013b). The evidence is very uncertain about the effect of water‐based exercise compared with land‐based exercise. The interventions may result in little to no difference in 6MWD (MD 5.00 meters, 95% CI −22.21 to 32.21; very low‐certainty evidence, Analysis 2.2; summary of findings Table 2).

ESWT

McNamara 2013b reported ESWT as change from baseline within group. McNamara 2013b indicated a potentially large difference favouring water‐based exercise, but there is a wide confidence interval around the effect estimate, including the possibility of no effect (MD 2.04 meters, 95% CI −7.16 to 415.16; Analysis 2.3).

ISWT

McNamara 2013b reported ISWT as change from baseline within the group. The results favoured water‐based exercise compared to land‐based exercise and the confidence interval is wide (MD 36.00, 95% CI 1.46 to 70.54; Analysis 2.4).

All‐cause hospital admissions (secondary outcome)

None of the studies reported on hospitalisations.

Respiratory hospital admissions (secondary outcome)

None of the studies reported on hospitalisations.

Mortality — all‐cause (secondary outcome)

None of the studies reported on mortality.

Pain (secondary outcome)

No studies reported this outcome.

Anxiety (secondary outcome)

One study reported results from the HADS‐anxiety questionnaire at eight weeks (McNamara 2013b). Water‐based exercise may have little or no effect on anxiety compared to land‐based exercise (Analysis 2.5). The number of cases of anxiety were not reported in the studies.

Depression (secondary outcome)

One study reported results from the HADS‐depression questionnaire at eight weeks (McNamara 2013b). Water‐based exercise may have little or no effect on depression compared to land‐based exercise (Analysis 2.6). The number of cases of depression was not reported in the studies.

Effects of interventions — qualitative

One study (Middlemass 2017) reported qualitative data from interviews of a subset of participants (n = 21) in a telemonitoring trial (Walker 2018). We present the results, including quotes from participants and personnel and judgements from the trial authors from Middlemass 2017 in Table 6. Because there was only one qualitative study, we were unable to draw out themes across several studies.

Some participants said they were able to accept their condition, aging and inevitable death. While some people expressed that the telemonitoring machine was a daily reminder of their condition and got them into negative thinking, others felt positive about their data being sent to their HCP, having the sense that people were looking after them from afar, and others reported that their friends and relatives were happy that their condition was being monitored. Some technical issues were identified by participants, including issues with WiFi, and some people would have liked to have seen the screen with the readings while they were using the machine. The results were sent straight to the HCP, and some participants would have liked access to their own data so they could monitor themselves and give themselves the confidence to do things when their health was better. Some people went to the GP more often after getting the health information technology (HIT) device, and others felt that they were going to the GP less often because they were able to "sort everything out". For quotes and author and systematic review interpretations see Table 6.

The certainty of thematic evidence identified from Middlemass 2017 was rated as very low, with some concerns about study methodology and relevance, and major concerns about coherence, adequacy of data and dissemination (Table 1; summary of findings Table 3). As we only found one qualitative study, we were not able to synthesise findings and examine emerging common themes.

Integration of qualitative and quantitative data

As we only identified one eligible qualitative study (Middlemass 2017), it was not possible to carry out an integrated synthesis of qualitative and quantitative data. This study has given us some insight into the barriers and facilitators to participating in the CHROMED telemonitoring intervention, as described above.

Discussion

Summary of main results

Owing to a paucity of studies, as well as the diversity in the intervention type, comorbidities and reported outcome measures, we were unable to provide a robust synthesis of data. Instead the review pulls together the disparate data available for this population in a series of tables and forest plots. There is insufficient evidence from high‐quality studies to clearly determine benefits of these interventions. The key take‐home message from this review is the lack of data from RCTs on treatments for people living with comorbid COPD.

We planned to update a preliminary logic model that we developed in the protocol for this review (Janjua 2019), intended to explain how the interventions affected people with COPD. However, due to the paucity of the evidence, we were unable to update the logic model in collaboration with our COPD patient steering group.

Overall completeness and applicability of evidence

This review aimed to look at trials involving people living with COPD and one or more comorbidities that investigated COPD interventions specifically tailored to the comorbidity. We were interested in the tailoring aspect of the interventions, but unfortunately were unable to locate many studies addressing this group. An alternative way of looking at people with COPD and comorbidities would be to search for trials aimed at treating people with COPD and a specific comorbidity, such as coronary heart disease. This may lead to more trials being included and perhaps this would be of more help to certain users (e.g. if the tailoring aspect is of less interest).

This review, or trials eligible for it, may not be the best way to consider the evidence for interventions for this population. Given the high prevalence of multimorbidity in this patient population (GOLD 2021Hillas 2015), many, if not all, COPD trials include people with comorbidities. It might be better for RCTs to report individual patient data together with more information about the individual's comorbidities, which would allow for analysis of outcomes in trials based on an individual patient's health.

We took data from the last reported time point in the trials, so none of the data relate to the impact of trials after the intervention has finished.

We chose to exclude studies where the only comorbidity was depression, anxiety, or both. This may have meant that people with depression and anxiety are underrepresented in the review. The implications of this are discussed below.

None of the studies evaluated impact on pain. Pain is emerging as an important symptom to consider in people with COPD, and targeted rehabilitation interventions are often rationalised by the presence of pain ‐ for example, exercises done with the support of water rather than on land, where impact can cause pain. Sixty‐six per cent of older people with COPD live with pain compared to 25% of the general population, and pain is cited as a common reason for dropping out of pulmonary rehabilitation programmes (Harrison 2017).

We were surprised by the apparent lack of qualitative studies. However, our inclusion criteria for qualitative studies focused very specifically on participants who had taken part in a tailored intervention, and their experiences of it. A broader inclusion criterion of qualitative studies investigating the experiences of people living with comorbid COPD, their carers, and HCPs more generally may have yielded more studies, and given some important information on the challenges of living with comorbid COPD and what the most important issues are for patients, carers and healthcare professionals. This in turn may help to inform the design of future interventions and evidence syntheses.

Certainty of the evidence

Quantitative ‐ intervention versus usual care

The certainty of evidence in this comparison ranged from very low to moderate (summary of findings Table 1). SGRQ evidence was rated as low certainty due to imprecision (small sample size) and risk of bias due to lack of blinding. Evidence for the CAT total score was rated as moderate certainty for rehabilitation and multicomponent interventions, but low certainty for pharmacotherapy and organisation‐of‐care interventions. Exacerbations were rated as very low certainty, due to lack of blinding, as well as imprecision (confidence intervals crossing the line of no effect). Functional status as measured by the six‐minute walk distance, was rated as low certainty due to imprecision (small sample size) and risk of bias (lack of blinding). Evidence for all‐cause hospital admissions was rated as low certainty, regardless of the intervention, mainly due to imprecision (small sample size) and risk of bias (lack of blinding). Both HADS‐A and HADS‐D were rated as very low to low, due to imprecision (confidence intervals crossing the line of no effect), and risk of bias (lack of blinding).

Quantitative ‐ intervention versus active control

The certainty of evidence in this comparison ranged from very low to low (summary of findings Table 2). Functional status, 6MWD, was rated as very low certainty due to imprecision (small sample size and a confidence interval crossing the line of no effect) and risk of bias (lack of blinding and attrition). HADS‐A evidence was rated as low certainty due to imprecision (small sample size) and lack of blinding of participants and those providing the intervention. Similarly, evidence for HADS‐D was rated as very low certainty due to imprecision (small sample size and a confidence interval crossing the line of no effect) and risk of bias (lack of blinding of participants and personnel).

Qualitative

The certainty of the qualitative evidence was assessed as very low (summary of findings Table 3).  There were some concerns with methodological limitations for all outcomes because of the research design, recruitment strategy, and how the data were collected.  Coherence and adequacy of data were of major concern as the evidence was based on one study, and we could not be certain of any issues about the qualitative data fitting the findings of the review. The relevance of the findings from the evidence was of some concern, because the study did not answer all aspects of the context specified in the review question.

Potential biases in the review process

This review was based on a published protocol (Janjua 2019). The terminology used to describe comorbidity and multimorbidity varies across studies and the terms are sometimes used synonymously (Smith 2016). We therefore included both terms and their variants in our search strategy and conducted a broad search across multiple databases. Despite this, it is possible that we could have missed relevant studies that did not describe the population with our included search terms. To try and mitigate this, we conducted supplementary forward‐and‐backward citation searches of our included studies.

This multimorbidity review is the first of its kind in Cochrane Airways. We wanted to conduct a review to explore multimorbid COPD, and there are a number of ways that this could have been done. Mindful of taking on too large a project for the time and money we had available, we decided to focus on interventions for COPD that were tailored to take account of the multimorbidity. We also decided to exclude populations where anxiety or depression were the sole comorbidity, and also because of potential overlap with other Cochrane Reviews on depression and anxiety in people with COPD. This decision may have been misguided, owing to the high prevalence of clinically relevant anxiety and depression in people with COPD — 40% compared to less than 10% in the general population (Atlantis 2013). Furthermore, as explained by Atlantis 2013 "Depression or anxiety comorbidity in patients with COPD predicts poor adherence to pulmonary rehabilitation and COPD‐related medication; decreased exercise capacity and health‐related quality of life; lost productivity; and increased health resource use, functional disability, and risk of exacerbation and mortality." Furthermore, there is a complex relationship between smoking, COPD and depression and anxiety — smoking is a risk factor for COPD, and depression predicts smoking initiation and deceases physical activity (Atlantis 2013). This limitation may have meant we missed studies that provided COPD interventions tailored to take account of people's depression and anxiety that would benefit 40% of the COPD population. Furthermore, because of these exclusions is possible that the level of anxiety and depression in the study populations may be lower than in the general COPD population and therefore the scope for improvement in these outcomes may have decreased.

We decided not to contact the authors of included studies for further information. This was a pragmatic decision, as we felt that the future information requested would not ultimately allow us to draw a firmer conclusion, even if greater accuracy was achieved.

Agreements and disagreements with other studies or reviews

A review by Kastner 2018 investigated interventions aimed at managing multiple chronic diseases in older people. They included studies of patients who had the same combination of chronic diseases, rather than studies where all the participants had an index condition with any comorbidity. The interventions were mainly co‐ordinated care or health technology involving multiple components. They found that co‐ordinated care interventions had the greatest potential for improving outcomes in older adults. They highlighted a general lack of evidence around interventions aimed at managing multimorbidity.

A Cochrane Review of interventions for people with multimorbidity in primary care or community settings (Smith 2016) included a mix of studies in which participants either had the same combination of chronic diseases or a broad range of conditions. The included interventions were mainly organisation of care, or self‐management, and again involved multiple components. They found mixed evidence on the effectiveness of interventions, with no clear improvements seen in clinical outcomes, healthcare use, medication adherence, patient or healthcare professional behaviour or cost. However, they observed an improvement in mental health and functional outcomes.

The population included in this review differs from the above reviews,  in that we included participants with an index condition (COPD) and one or more comorbidities. However, we found a similar paucity of evidence on interventions for managing patients with comorbidities. Because the number of included studies was small, it is difficult to draw direct comparisons between the efficacy of interventions in people with COPD and multimorbidity and those without multimorbidity. However limited evidence indicates the effects of pulmonary rehabilitation interventions were similar to or better than those seen in previous COPD trials.

Study flow diagram for randomised controlled trials

Figuras y tablas -
Figure 1

Study flow diagram for randomised controlled trials

Study flow diagram for qualitative studies

Figuras y tablas -
Figure 2

Study flow diagram for qualitative studies

Risk of bias summary

Figuras y tablas -
Figure 3

Risk of bias summary

Comparison 1: Intervention versus usual care, Outcome 1: Quality of life ‐ SGRQ total

Figuras y tablas -
Analysis 1.1

Comparison 1: Intervention versus usual care, Outcome 1: Quality of life ‐ SGRQ total

Comparison 1: Intervention versus usual care, Outcome 2: Quality of life ‐ CAT total

Figuras y tablas -
Analysis 1.2

Comparison 1: Intervention versus usual care, Outcome 2: Quality of life ‐ CAT total

Comparison 1: Intervention versus usual care, Outcome 3: Quality of life ‐ CRQ domains

Figuras y tablas -
Analysis 1.3

Comparison 1: Intervention versus usual care, Outcome 3: Quality of life ‐ CRQ domains

Comparison 1: Intervention versus usual care, Outcome 4: Quality of life ‐ MLHFQ

Figuras y tablas -
Analysis 1.4

Comparison 1: Intervention versus usual care, Outcome 4: Quality of life ‐ MLHFQ

Comparison 1: Intervention versus usual care, Outcome 5: Quality of life ‐ EQ‐5D

Figuras y tablas -
Analysis 1.5

Comparison 1: Intervention versus usual care, Outcome 5: Quality of life ‐ EQ‐5D

Comparison 1: Intervention versus usual care, Outcome 6: Exacerbations ‐ people experiencing one or more

Figuras y tablas -
Analysis 1.6

Comparison 1: Intervention versus usual care, Outcome 6: Exacerbations ‐ people experiencing one or more

Comparison 1: Intervention versus usual care, Outcome 7: Exacerbations ‐ mean number per person

Figuras y tablas -
Analysis 1.7

Comparison 1: Intervention versus usual care, Outcome 7: Exacerbations ‐ mean number per person

Comparison 1: Intervention versus usual care, Outcome 8: Functional status ‐ 6MWT

Figuras y tablas -
Analysis 1.8

Comparison 1: Intervention versus usual care, Outcome 8: Functional status ‐ 6MWT

Comparison 1: Intervention versus usual care, Outcome 9: Functional status ‐ ISWT

Figuras y tablas -
Analysis 1.9

Comparison 1: Intervention versus usual care, Outcome 9: Functional status ‐ ISWT

Comparison 1: Intervention versus usual care, Outcome 10: Functional status ‐ ESWT

Figuras y tablas -
Analysis 1.10

Comparison 1: Intervention versus usual care, Outcome 10: Functional status ‐ ESWT

Comparison 1: Intervention versus usual care, Outcome 11: All‐cause hospital admissions ‐ people experiencing one or more

Figuras y tablas -
Analysis 1.11

Comparison 1: Intervention versus usual care, Outcome 11: All‐cause hospital admissions ‐ people experiencing one or more

Comparison 1: Intervention versus usual care, Outcome 12: All‐cause hospital admissions ‐ mean number per person

Figuras y tablas -
Analysis 1.12

Comparison 1: Intervention versus usual care, Outcome 12: All‐cause hospital admissions ‐ mean number per person

Comparison 1: Intervention versus usual care, Outcome 13: Respiratory‐related hospital admissions

Figuras y tablas -
Analysis 1.13

Comparison 1: Intervention versus usual care, Outcome 13: Respiratory‐related hospital admissions

Comparison 1: Intervention versus usual care, Outcome 14: All‐cause mortality (deaths)

Figuras y tablas -
Analysis 1.14

Comparison 1: Intervention versus usual care, Outcome 14: All‐cause mortality (deaths)

Comparison 1: Intervention versus usual care, Outcome 15: Anxiety HADS‐A

Figuras y tablas -
Analysis 1.15

Comparison 1: Intervention versus usual care, Outcome 15: Anxiety HADS‐A

Comparison 1: Intervention versus usual care, Outcome 16: Depression HADS‐D

Figuras y tablas -
Analysis 1.16

Comparison 1: Intervention versus usual care, Outcome 16: Depression HADS‐D

Comparison 2: Intervention versus active comparison, Outcome 1: Quality of life ‐ CRQ domains

Figuras y tablas -
Analysis 2.1

Comparison 2: Intervention versus active comparison, Outcome 1: Quality of life ‐ CRQ domains

Comparison 2: Intervention versus active comparison, Outcome 2: Functional status ‐ 6MWT

Figuras y tablas -
Analysis 2.2

Comparison 2: Intervention versus active comparison, Outcome 2: Functional status ‐ 6MWT

Comparison 2: Intervention versus active comparison, Outcome 3: Functional status ‐ ESWT

Figuras y tablas -
Analysis 2.3

Comparison 2: Intervention versus active comparison, Outcome 3: Functional status ‐ ESWT

Comparison 2: Intervention versus active comparison, Outcome 4: Functional status ‐ ISWT

Figuras y tablas -
Analysis 2.4

Comparison 2: Intervention versus active comparison, Outcome 4: Functional status ‐ ISWT

Comparison 2: Intervention versus active comparison, Outcome 5: Anxiety HADS‐A

Figuras y tablas -
Analysis 2.5

Comparison 2: Intervention versus active comparison, Outcome 5: Anxiety HADS‐A

Comparison 2: Intervention versus active comparison, Outcome 6: Depression HADS‐D

Figuras y tablas -
Analysis 2.6

Comparison 2: Intervention versus active comparison, Outcome 6: Depression HADS‐D

Summary of findings 1. Intervention compared to usual care for COPD and at least one other long‐term condition

Intervention compared to usual care for COPD and at least one other long‐term condition

Patient or population: COPD and at least one other long‐term condition
Setting: community teaching hospital (1), hospital outpatient clinic (1), university hospital (1), tertiary public hospital (1), multi‐centre (3), single hospital (1)
Intervention: Intervention (rehabilitation, organisation of care, pharmacotherapy, multicomponent intervention)
Comparison: Usual care

Outcome domain

Intervention group

(follow‐up)

Anticipated absolute effects* (95% CI)

Relative effect

No of participants

(studies)

Certainty of the evidence

(GRADE)

Comments

Risk with usual care

Risk with intervention

Quality of life ‐ SGRQ total
Scale from: 0 to 100 (lower scores better)

 

Rehabilitation (pulmonary rehab)

(follow‐up 52 weeks)

The mean SGRQ total score was 70

MD 10.85 lower (12.66 lower to 9.04 lower)

70
(1 RCT)

(95% CI)⊕⊝⊝
LOWa,b
 

MCID for SGRQ is a change of 4 points (Jones 2005)

 

Quality of life ‐ CAT total

Scale from 0 to 40 (lower scores better)

 

 

 

Pharmacotherapy (optimised COPD treatment)

(follow‐up 52 weeks)

The mean CAT total score was 0.4

MD 0.00
(3.40 lower to 3.40 higher)

77
(1 RCT)

⊕⊕⊝⊝
LOWa,c,d

MCID for CAT total is 2 points (Kon 2014)

 

 

 

 

Rehabilitation (pulmonary rehab)

(follow‐up 52 weeks)

The mean CAT total score was 24.34

MD 8.02 lower
(9.44 lower to 6.6 lower)

70
(1 RCT)

⊕⊕⊕⊝
MODERATEa,b

Organisation of care (telemonitoring)

(follow‐up 39 weeks)

The mean CAT total score was 17.17

MD 0.41 lower
(2.19 lower to 1.37 higher)

312
(1 RCT)

⊕⊕⊝⊝
LOWc,d

Multicomponent intervention

(follow‐up 52 weeks)

The mean CAT total score was 1.6

MD 6.9 lower
(9.56 lower to 4.24 lower)

80
(1 RCT)

⊕⊕⊕⊝
MODERATEa,c

Exacerbations ‐ number of people experiencing one or more

 

Rehabilitation (case management)
(follow‐up 52 weeks)

 

 

573 per 1000

594 per 1000 (501 to 678)

OR 1.09
(0.75 to 1.57)

470
(1 RCT)

⊕⊝⊝⊝
VERY LOWe,f

Functional status‐6MWD (metres)

 

 

Rehabilitation (pulmonary rehab and water‐based exercise)

(follow‐up 38.8 weeks**)

The mean distance walked in 6 minutes was 344 metres

MD 60.4 metres higher
(44.26 higher to 76.54 higher)

100
(2 RCTs)

⊕⊕⊝⊝
LOWa,c

MCID for the 6MWT is 25 to 35 metres (Holland 2013)

 

 

Multicomponent intervention

(follow‐up 52 weeks)

The mean distance walked in six minutes was ‐15

MD 75 higher
(28.06 higher to 121.94 higher)

80
(1 RCT)

⊕⊕⊝⊝
LOWa,c

All‐cause hospital admissions ‐ people experiencing one or more

 

Organisation of care (telemonitoring)

(follow‐up 39 weeks)

 

292 per 1000

 

273 per 1000
(185 to 382)

OR 0.91 (0.55 to 1.50)

312
(1 RCT)

⊕⊕⊝⊝
LOWc

Multicomponent intervention

(follow‐up 17 weeks)

732 per 1000

 

459 per 1000
(277 to 647)

OR 0.31
(0.14 to 0.67)

112
(1 RCT)

⊕⊕⊝⊝
LOWa,c

All‐cause mortality (deaths)

 

 

 

Pharmacotherapy (optimised COPD treatment)
(follow‐up 17.6 weeks**)

170 per 1000

102 per 1000
(45 to 217)

OR 0.55
(0.23 to 1.35)

177
(2 RCTs)

⊕⊝⊝⊝
VERY LOWa,e,g

Organisation of care (case management and telemonitoring)

(follow‐up 46.7 weeks**)

102 per 1000

60 per 1000
(36 to 98)

OR 0.56
(0.33 to 0.96)

782
(2 RCTs)

⊕⊕⊝⊝
LOWe

Multicomponent intervention

(follow‐up 52 weeks)

18 per 1000

18 per 1000
(1 to 230)

OR 1.00
(0.06 to 16.39)

112
(1 RCT)

⊕⊝⊝⊝
VERY LOWa,c,h

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

**Weighted mean duration of follow‐up

6MWD: 6‐minute walk distance; CAT: COPD assessment test; CI: Confidence interval; COPD: chronic obstructive pulmonary disease; GIV: generic inverse variance; HADS: Hospital Anxiety and Depression Scale; MD: mean difference; MCID: minimally clinically important difference; OR: Odds ratio; RCT: randomised controlled trial;RR: Risk ratio; SGRQ: St George's Respiratory Questionnaire.

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

aThe evidence was downgraded by 1 for imprecision due to the optimal information size of less than 200 participants.
bThe evidence was downgraded by 1 for risk of performance bias.
cThe evidence was downgraded by 1 for risk of performance and attrition bias.
dThe evidence was downgraded by 1 for imprecision due to wide confidence intervals.
eThe evidence was downgraded by 2 for risk of performance and detection bias.
fThe evidence was downgraded by 1 for imprecision as the lower confidence interval crossed the line of no effect.
gThe evidence was downgraded by 1 for risk of attrition due to high dropout rate in the study.
hThe evidence for this outcome was downgraded by 2 for imprecision due to very wide confidence intervals.

Figuras y tablas -
Summary of findings 1. Intervention compared to usual care for COPD and at least one other long‐term condition
Summary of findings 2. Intervention compared to active comparison for COPD and at least one other long‐term condition

Intervention compared to active comparison for COPD and at least one other long‐term condition

Patient or population: COPD and at least one other long‐term condition
Setting: outpatient respiratory departments from 5 hospitals (multicentre), hospital outpatient clinic (1), tertiary public hospital (1)
Intervention: Intervention (rehabilitation water‐based versus land‐based exercise programme)
Comparison: active comparison

Outcome domain

Intervention group (follow‐up)

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with active comparison

Risk with Intervention

Quality of life ‐ SGRQ total
 

Not reported
 

Quality of life ‐ CAT total
 

Not reported
 

Exacerbations
 

Not reported
 

Functional status ‐ 6MWD (metres)

Rehabilitation

(follow‐up 8 weeks)

The mean distance walked in 6 minutes was 43 metres

MD 5 metres higher
(22.21 lower to 32.21 higher)

30
(1 RCT)

⊕⊝⊝⊝
VERY LOWa,b,c

The MCID for 6MWT is 25 to 35 metres (Holland 2013)

All‐cause hospital admissions
 

Not reported
 

All‐cause mortality
 

Not reported
 

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

6MWD: 6‐minute walk distance; CAT: COPD assessment test; CI: Confidence interval; COPD: chronic obstructive pulmonary disease; MD: mean difference; MCID: minimally clinically important difference; RCT: randomised controlled trial;SGRQ: St George's Respiratory Questionnaire.

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

aThe evidence was downgraded by 1 for imprecision due to the lower confidence interval crossing 1.0.
bThe evidence was downgraded by 1 for imprecision as the optimal information size was less than 200 participants.
cThe evidence was downgraded by 2 for risk of performance and attrition bias.

Figuras y tablas -
Summary of findings 2. Intervention compared to active comparison for COPD and at least one other long‐term condition
Summary of findings 3. Summary of findings table for qualitative GRADE CERqual assessment

Theme

Methodological limitations

Coherence

Adequacy of data

Relevance

Dissemination bias

Overall assessment

Explanation of

CERQual assessment

Studies contributing to the review finding

Health status, beliefs and concerns

The findings were graded

as very low confidence

because of minor

concerns regarding

methodological limitations

and relevance. There were major concerns regarding

coherence and adequacy of data

Middlemass 2017

Unchanging nature of condition

Some

concernsa

Major concernsb

Major concernsb

Some

Concernsc

No concerns

Very lowc

Withdrawal of face to face communication

Some

concernsa

Major concernsb

Major concernsb

Some

Concernsc

No concerns

Very lowc

Reminder of illness and anxiety

Some

concernsa

Major concernsb

Major concernsb

Some

Concernsc

No concerns

Very lowc

Information

Subjective norms

Some

concernsa

Major concernsb

Major concernsb

Some

Concernsc

No concerns

Very lowc

Technology

Unreliable technology

Some

concernsa

Major concernsb

Major concernsb

Some

Concernsc

No concerns

Very lowc

HIT Self‐efficacy

Some

concernsa

Major concernsb

Major concernsb

Some

Concernsc

No concerns

Very lowc

Perceived usefulness

Daily monitoring of conditions

Some

concernsa

Major concernsb

Major concernsb

Some

Concernsc

No concerns

Very lowc

Factors affecting usefulness

Lack of feedback

Some

concernsa

Major concernsb

Major concernsb

Some

Concernsc

No concerns

Very lowc

Behaviour

Self‐management and healthcare utilisation

Some

concernsa

Major concernsb

No concerns

No concerns

No concerns

Very lowc

Criteria for assessment (https://www.cerqual.org/publications/)

Methodological limitations: risk to rigour see Table 1.

Coherence: how clear and cogent the fit is between the data from the primary studies and the review finding that synthesises that data. ‘Findings’ are ‘transformations’ of the underlying data into descriptions, interpretations and /or explanations of the phenomenon of interest. Findings are developed by identifying patterns in the data across primary studies

Adequacy: overall determination of the degree of richness as well as the quantity of data supporting the review finding i.e., extent to which information that the study authors provide is detailed enough to interpret the meaning and context of what is being researched.

Relevance: refers to the extent to which the body of data from primary studies supports the review finding in terms of applicability to the review question.

Dissemination bias: a systematic distortion of the phenomenon of interest due to selective dissemination of qualitative studies or the findings of qualitative studies

aThere were some concerns about the research design, and recruitment strategy to address the aims. It was not clear how the data was collected.
bThe evidence mainly because the evidence is based on one study. Due to the limited number of studies, we cannot be certain that there are any issues about whether the data fit the finding of the review. Findings do not support quantitative data.
cThe findings from the study did not answer all aspects of context specified in the review question.

Figuras y tablas -
Summary of findings 3. Summary of findings table for qualitative GRADE CERqual assessment
Table 1. Risk of bias assessment for qualitative studies

Study ID

Is there a statement of research aims?

Is a qualitative approach justified?

Was the research design appropriate to address the aims?

Was the recruitment strategy appropriate to address the aims?

Were data collected in a way that addressed the research issue?

Was the researcher/participant relationship adequately considered?

Have ethical issues been taken into consideration?

Was the data analysis sufficiently rigorous?

Was there a clear statement of findings?

Overall assessment

Middlemass 2017

Yes

Yes

Can't tell

Yes

Can't tell

No

Yes

Yes

Yes

Some concerns

To test the HITAM and see if it could be used to increase the adoption of HIT. HIT has been shown to reduce mortality. Many with LTC are over 60 and HIT is not always accepted.

Pre‐ and post‐interviews to ascertain perceptions/experience on the HITAM elements. However, no rationale for adopting a qualitative and framework approach.

The study design (instrumental, collective case design) is not justified.

Selection process for interviews included those who had taken part in the pilot and those assigned to the Rx. One was not interviewed and was unwell.

Setting of interviews (home or telephone) was not justified. Not clear if interviewers were in‐depth or semi‐structured. Not justified why interviews were chosen rather than focus groups. Interview schedule was used and is made available. Interviews were audio‐recorded. Data saturation is not mentioned.

The interviewer's role is not described or examined.

Ethical approval obtained and informed written consent to take part in two taped interviewers to ascertain views if using the equipment.

Process of analysis is well described, although unsure how the framework was formed. Quotes provided to support themes. Contrasting data are taken into account and described.

Explicit well organised findings and multiple researchers involved in the process.

Abbreviations: HITAM: Health Information Technology Acceptance Model; LTC: long term condition.

Figuras y tablas -
Table 1. Risk of bias assessment for qualitative studies
Table 2. Framework and map of interventions identified from included studies

Interventions (identified fromGOLD 2021 guideline andCochrane Airways subtopic list.)

Study

Interventions

Evidence type (quantitative, qualitative or mixed methods)

Comorbidities

  • Reducing risk factors

    • Smoking cessation

  • Vaccination

    • Pneumococcal

    • Influenza

  • Pharmacotherapies

    • Short‐acting inhalers (e.g. SABA)

    • Long‐acting inhalers (e.g. LABA, LAMA, ICS)

    • Phosphodiesterase‐4 (PDE4) inhibitors (e.g. Roflumilast)

    • Mucolytic agents

    • Combination inhalers and triple therapy

    • Methyl xanthines

    • Oral corticosteroids

    • Antibiotics

    • Statins

    • Alpha‐1 antitrypsin augmentation therapy

    • Biomarker mediated therapy

EUCTR2010‐021412‐42‐GB

Inhaler optimisation and best supportive care vs UC

Quantitative

All had lung cancer

Gottlieb 2020

Management: optimising COPD treatment vs UC

Quantitative

All had lung cancer or head and neck cancer, and some had other comorbidities including ischaemic heart diseases, heart failure, depression/anxiety, osteoporosis, cerebrovascular disease, diabetes

  • Rehabilitation

    • Pulmonary rehabilitation

    • Exercise therapy (e.g. upper limb exercise, ongoing physical exercise after pulmonary rehabilitation)

    • Complementary therapies (e.g. active mind‐body therapy, Tai chi, singing)

Budnevskiy 2015

Pulmonary rehabilitation vs UC

Quantitative

All had metabolic syndrome

McNamara 2013b

Water‐based exercise training vs land‐based exercise training vs UC

Quantitative

One or more physical comorbidities: musculoskeletal, or neurological, or obesity

  • Self‐management

  • Organisation of care

    • Support services (e.g. social care, specialist respiratory nurse)

    • Integrated care

    • Telehealthcare

    • Digital management interventions

    • Home care

    • Integrated disease management (e.g. disease management programming)

    • Interventions to promote or increase adherence to PR or other treatments

Rose 2018

Case management vs UC

Quantitative

Two or more comorbidities: cardiovascular disease, including coronary artery disease, hypertension and congestive heart failure, diabetes, depression, osteopenia, osteoporosis, and gastro‐oesophageal reflux disease.

Walker 2018

Middlemass 2017

Telemonitoring vs UC

Quantitative

Qualitative

One or more non‐pulmonary comorbidity: congestive heart failure or ischaemic heart disease or both, hypertension, osteoporosis, hyperlipidaemia, osteoporosis, sleep‐related disordered breathing.

  • Other treatments

    • Oxygen therapy and ventilatory support (e.g. NIV, ambulatory oxygen)

    • Nutritional support

    • Lung volume reduction surgery

    • Lung transplantation

    • Supportive, palliative, end of life and hospice care

    • Psychotherapy

    • interventions for sexual dysfunction

    • Self‐help groups

  • Multicomponent intervention

Bernocchi 2018

Maintenance rehabilitation and telemonitoring vs UC. Following inpatient rehabilitation participants randomised to personalised discharge plus nurse telephone support and telemonitoring, plus physiotherapist personalised rehabilitation vs UC

Quantitative

All had cardiovascular disease

UC: usual care

Figuras y tablas -
Table 2. Framework and map of interventions identified from included studies
Table 3. Summary of interventions

Study

Duration (weeks)

Intervention

Comparison

Setting

Provider

Materials/method

Delivery

Tailoring

Pharmacotherapy

EUCTR2010‐021412‐42‐GB

4

Pharmacotherapy: inhaler optimisation and best supportive care

UC

Outpatient

NR

Evohaler 100 μg, Spiriva 18 mg via Handihaler, fluticasone propionate 500 μg

Inhaler

Considered the poor prognosis of patients with lung cancer: intervention group treated with maximum inhaled therapy

Gottlieb 2020

25

Management: optimising COPD treatment

UC

Outpatient clinic

Pulmonary physician

Two visits at 12 and 24 weeks in an outpatient clinic where the adjustment of COPD treatment was considered

Dialogue with the participants

Adjustment of COPD treatment considered at each visit

Rehabilitation

Budnevskiy 2015

52

Pulmonary rehabilitation

UC

NR

NR

Series of seminars covering treatment and prevention of COPD and MS. Physical training: therapeutic exercises.

Group seminars

Exercises took into account concomitant MS

Gurgun 2013

8

Pulmonary rehabilitation and nutritional support

PR or UC

Outpatient clinic

Dietician

Dietary counselling and oral supplementation in combination with exercise training

Supervised exercise training, dietary counselling

Daily nutrition intake of the patient was checked

McNamara 2013b

8

Water‐based exercise training

Land‐based exercise training or UC

Hospital hydrotherapy pool

Physiotherapist

Exercise in a hydrotherapy pool: warm‐up, lower limb endurance, upper limb endurance, and cool down

Supervised exercise

Training intensity measured, participants chose the most comfortable level of water immersion

Organisation of care

Rose 2018

52

Case management

UC

Outpatient

Case‐manager

Education based on 'Living Well with COPD', ongoing communication with physician and hospital specialist

Education session at enrolment, telephone consultations

Individualised action plan

Walker 2018

39

Telemonitoring

UC

Outpatient

Study nurse

CHROMED monitoring platform

Wearable device

Respiratory alert triggered contact with the nurse if worsening was detected

Multicomponent intervention

Bernocchi 2018

17

Inpatient rehabilitation, personalised discharge, nurse telephone support and telemonitoring, physiotherapist personalised maintenance rehabilitation

Inpatient rehabilitation, UC

Participants' home

Nurse; physiotherapist

Exercise programme, mini‐ergometer, pedometer and diary. Participants provided with a pulse oximeter, and a portable one‐lead ECG

PT instruction; weekly phone call with NT; weekly phone call with PT

Personalised exercise programme

Abbreviations: COPD: Chronic obstructive pulmonary disease; MS: metabolic syndrome; NT: Nurse Tutor; PT:Physiotherapist tutor; UC: usual care

Figuras y tablas -
Table 3. Summary of interventions
Table 4. Summary of baseline characteristics

Study

COPD severity

Comorbidities (%)

Intervention

Comorbidities (%)

Control

Ethnicity

Male (%)

Age (Mean, SD)

Intervention

Control

Intervention

Control

Bernocchi 2018

Mild to very severe

Cardiovascular disease (100%)

Cardiovascular disease (100%)

NR

88

75

71 (9)

70 (9.5)

Budnevskiy 2015

Moderate

Metabolic syndrome (100%)

Metabolic syndrome (100%)

NR

66

71

NR

NR

EUCTR2010‐021412‐42‐GB

NR

Lung cancer (100%)

Lung cancer (100%)

NR

34

38

68 (59 to 75)*

67 (61 to 71)*

Gottlieb 2020

Mild to severe

Lung cancer (84%); head‐and‐neck cancer (16%); Ischaemic heart disease/heart failure (12.3%); Depression/anxiety (3.5%); Osteoporosis (12.3%); Cerebrovascular disease (14%); Diabetes (14%)

Lung cancer (82.5%); head‐and‐neck cancer (17.5%); ischaemic heart disease/heart failure (10.5%); Depression/anxiety (3.5%); Osteoporosis (10.5%); Cerebrovascular disease (8.8%);
Diabetes (8.8%)

NR

58

69

67.8 (8.3)

67.2 (8.1)

McNamara 2013b

NR

Water‐based group:

Musculoskeletal (50%);
neurological (5.5%);
obesity (44.5%)

Land‐based group:

Musculoskeletal (65%);
neurological (5%);
obesity (30%)

Musculoskeletal (47%);
neurological (0%);
obesity (53%)

NR

Water‐based group: 28

Land‐based group: 50

47

Water‐based group: 72 (10)

Land‐based group: 73(7)

70 (9)

Rose 2018

Moderate to severe

Two or more comorbidities: cardiovascular disease (75%); diabetes (18%); depression (17%); Osteopenia and osteoporosis (30%); Gastro‐oesophageal reflux disease (14%); Hypothyroidism (9%); Osteoarthritis (9%); Glaucoma and cataracts (9%); Cachexia and malnutrition (10%); Chronic kidney disease (7%); Anxiety (6%); Peripheral muscle dysfunction (6%); Obstructive sleep apnoea (5%); Lung cancer (6); Cerebrovascular accident (3%)

Two or more comorbidities: cardiovascular disease (76%); diabetes (22%); depression (20%); Osteopenia and osteoporosis (29%); Gastro‐oesophageal reflux disease (12%); Hypothyroidism (9%); Osteoarthritis (9%); Glaucoma and cataracts (9%); Cachexia and malnutrition (8%); Chronic kidney disease (7%); Anxiety (7%); Peripheral muscle dysfunction (6%); Obstructive sleep apnoea (6%); Lung cancer (6%); Cerebrovascular accident (4%)

NR

50

44

71 (9.2)

71 (9.7)

Walker 2018

Mild to very severe

One or more non‐pulmonary comorbidities: Congestive heart failure (12%); ischaemic heart disease (25%; Congestive heart failure plus ischaemic heart disease (12%); hypertension (72%); Sleep‐related disordered breathing (11%); Osteoporosis (17%); Hyperlipidemia (53%)

One or more non‐pulmonary comorbidities: Congestive heart failure (8%); ischaemic heart disease (23%; Congestive heart failure plus ischaemic heart disease (13%); hypertension (68%); Sleep‐related disordered breathing (6%); Osteoporosis (15%); Hyperlipidemia (58%)

NR

66

66

71 (66.0 to 75.8)*

71 (65.3 to 76.0)*

*median, interquartile range

Abbreviations: NR: not reported

Figuras y tablas -
Table 4. Summary of baseline characteristics
Table 5. Summary of relevant quantitative study outcomes

Study ID

Outcome domain

Outcome measure

End point time (weeks)

Bernocchi 2018

All‐cause hospital admissions

Number of events

17

Functional status

6MWT

Functional status

PASE

Mortality (all‐causes)

Number of events

Quality of life

MLHFQ

Quality of life

CAT

Quality of life

Dyspnoea MRC

Respiratory hospital admissions

Number of events

Functional status

6MWT

Quality of life

CAT (total)

Quality of life

SGRQ (total)

Quality of life

Dyspnoea MRC

Budnevskiy 2015

Functional status

6MWT

52 weeks

Quality of life

CCQ

Quality of life

CAT (total)

Quality of life

SGRQ (total)

EUCTR2010‐021412‐42‐GB

Adverse events

Number of events

4

Mortality (all‐causes)

Number of events

Quality of life

Dyspnoea

Gottlieb 2020

Mortality (all‐causes)

Number of events

25

Quality of life

CAT (total)

McNamara 2013b

Anxiety symptoms

HADS‐A (anxiety)

8

Depression symptoms

HADS‐D (depression)

Functional status

ESWT

Functional status

6MWT

Functional status

ISWT

Functional status

ESWT

Quality of life

CRDQ dyspnoea

Rose 2018

All‐cause hospital admissions

Mean & SD

All‐cause hospital admissions

Risk difference

52

Anxiety symptoms

HADS‐A (anxiety)

Depression symptoms

HADS‐D (depression)

Exacerbations

Exacerbation (ED visit)

Mortality (all‐causes)

Number of events

Quality of life

SGRQ (total)

Walker 2018

All‐cause hospital admissions

Hospitalisation rate

39

All‐cause hospital admissions

Number of people experiencing one or more events

Exacerbations

Exacerbation rate moderate exacerbations

Mortality (all‐causes)

Number of events

Quality of life

CAT (total)

Quality of life

PHQ‐9

Quality of life

EQ‐5D utility

Quality of life

EQ‐5D VAS

6MWT: 6‐Minute Walk Test; CAT: COPD Assessment Test; CCQ: COPD clinical questionnaire; CRQ: Chronic Respiratory Disease Questionnaire; CSES: Coping Self‐Efficacy Scale; EQ‐5D: EuroQuol‐5D; ESWT: Endurance Shuttle Walk Test; HADS: Hospital Anxiety and Depression Scale; ICFS: Identity‐Consequence Fatigue Score; ISWT: Incremental Shuttle Walk Test; MLHFQ: Minnesota Living with Heart Failure Questionnaire; MRC: Medical Research Council; PASE: Physical Activity Profile; PHQ‐9: Patient Health Questionnaire‐9; PIH: Partners in Health scale; SGRQ: St George's Respiratory Questionnaire; VAS: Visual Analogue Scale.

Figuras y tablas -
Table 5. Summary of relevant quantitative study outcomes
Table 6. First and second order constructs of qualitative studies

Study

Aims

Main themes and example quotes provided in study report

Author comments provided in the study report

Conclusions ‐ review author team interpretation of study results

Middlemass 2017, UK

To explore the usefulness of the HITAM for understanding acceptance of HIT in older people (≥ 60 years age) with COPD and associated heart diseases

Health status, beliefs and concerns

Unchanging nature of condition: Patients accepted that their chronic condition was unlikely to change and that ageing and (eventual) death was inevitable: "I'm getting older and I'm not going to get any better. I haven't got young genes to repair everything. So, if I can pummel along the way I am, I'll accept it".

Acceptance of illness: Some patients had accepted the life‐restricting (and sometimes life‐threatening) limitations of their LTCs

Beliefs about condition influence motivation to engage in HIT

Withdrawal of face‐to‐face communication: "But I would hope they would still do their person‐ to‐person contact [and] that they wouldn't just forget."

Concern of losing face to face contact: Patients were concerned that they would lose face‐to‐face communication with their HCP when using TM

HCP face to face interaction is re‐assuring that their condition is actively being monitored

Reminder of illness and anxiety :"This is reminding me every day, then I should think I wonder what my reading is, how good it is or how bad it is and I thought no, get away from illness you know. Every time as soon I started thinking about it, I started thinking about my illness..."

Fear of illness: Patients perception of telemonitoring was linked to fear of reminders of how serious their condition was, which led to them not continuing with home monitoring

Belief that HIT causes anxiety about condition which can lead to non‐adherence/

Information

Subjective norms:"I think if my very close relatives…and if the GP said it is essential… I would say I'm definitely going ahead with it"

Increased motivation to comply with HIT: Close relatives and GP influenced and increased individuals' perceptions of using HIT

Input from HCP and relatives increases acceptance to use HIT

Technology

Unreliable technology:"...a couple of times it didn't go through very well, but that was an internet problem".

Unreliable technology: Poor internet connectivity and data transmission in rural areas led to generation of technical alerts, which led to the study research nurse visiting the patient to find out what the problem was.

HIT can be beneficial for those who cannot visit HCP face to face, however, this can be limited by Internet connectivity

HIT Self‐efficacy:"The very first time I really got panicked. But then the next day when I did it, it was easier, but I was at the start of a chest infection, which did affect me… It helped my husband stood beside me and was chatting saying yeah you're doing fine, not long to go, just a little bit of encouragement"

Increased self‐efficacy: Both HCPs and patients' significant others were key to them using the TM equipment.

Patients' relatives and HCPs can help to reduce apprehension of using HIT in the initial stages

Perceived usefulness

Daily monitoring of conditions: "...I feel more comfortable knowing that somebody's checking it all the time, you know they're looking at it every day..."

Confidence of daily monitoring: Patients perception of being linked to a HCP checking data and ready to act on change in health status led them to feel safe about using HIT.

Patients' perceptions are dependent on knowing that HCP involvement is linked to HIT

Factors affecting usefulness

Lack of feedback: "I'm in a vacuum. I'm doing something, I'm sending it off to you, [but] there's no feedback..."

Lack of feedback: there was lack of two‐way communication between the patient and HCP

Lack of feedback from HCP resulted in reduced perception of usefulness of HIT

Behaviour

Self‐management and health care utilisation: Patients felt that their condition stabilised after joining the study and did not need to go to the GP so often: "I've been less to the surgery... Because I think it's helped me sort everything out. I'm much better on the medication I'm on now for my blood pressure."

Improved self‐management and reduced need to see the GP: Patients' condition stabilised whilst enrolled in the study, and GP visits also declined.

HIT may lead to changes in behaviour towards improving patients' self‐management and a reduced need to visit the GP

Abbreviations: HITAM: Health Information Technology Acceptance Model

Figuras y tablas -
Table 6. First and second order constructs of qualitative studies
Comparison 1. Intervention versus usual care

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Quality of life ‐ SGRQ total Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.1.1 Rehabilitation

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.2 Quality of life ‐ CAT total Show forest plot

4

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.2.1 Pharmacotherapy

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.2.2 Rehabilitation

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.2.3 Organisation of care

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.2.4 Multicomponent intervention

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.3 Quality of life ‐ CRQ domains Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.3.1 Dyspnoea

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.3.2 Fatigue

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.3.3 Emotion

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.3.4 Mastery

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.4 Quality of life ‐ MLHFQ Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.4.1 Multicomponent intervention

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.5 Quality of life ‐ EQ‐5D Show forest plot

1

Std. Mean Difference (IV, Random, 95% CI)

Totals not selected

1.5.1 VAS

1

Std. Mean Difference (IV, Random, 95% CI)

Totals not selected

1.5.2 EQ‐5D utility domain

1

Std. Mean Difference (IV, Random, 95% CI)

Totals not selected

1.6 Exacerbations ‐ people experiencing one or more Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Totals not selected

1.6.1 Rehabilitation

1

Odds Ratio (M‐H, Random, 95% CI)

Totals not selected

1.7 Exacerbations ‐ mean number per person Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.7.1 Rehabilitation

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.8 Functional status ‐ 6MWT Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

1.8.1 Rehabilitation

2

100

Mean Difference (IV, Random, 95% CI)

60.40 [44.26, 76.54]

1.8.2 Multicomponent intervention

1

80

Mean Difference (IV, Random, 95% CI)

75.00 [28.06, 121.94]

1.9 Functional status ‐ ISWT Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.9.1 Rehabilitation

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.10 Functional status ‐ ESWT Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.10.1 Rehabilitation

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.11 All‐cause hospital admissions ‐ people experiencing one or more Show forest plot

2

Odds Ratio (M‐H, Random, 95% CI)

Totals not selected

1.11.1 Organisation of care

1

Odds Ratio (M‐H, Random, 95% CI)

Totals not selected

1.11.2 Multicomponent intervention

1

Odds Ratio (M‐H, Random, 95% CI)

Totals not selected

1.12 All‐cause hospital admissions ‐ mean number per person Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.12.1 Organisation of care

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.13 Respiratory‐related hospital admissions Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Totals not selected

1.13.1 Multicomponent intervention

1

Odds Ratio (M‐H, Random, 95% CI)

Totals not selected

1.14 All‐cause mortality (deaths) Show forest plot

5

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

1.14.1 Pharmacotherapy

2

177

Odds Ratio (M‐H, Random, 95% CI)

0.55 [0.23, 1.35]

1.14.2 Organisation of care

2

782

Odds Ratio (M‐H, Random, 95% CI)

0.56 [0.33, 0.96]

1.14.3 Multicomponent intervention

1

112

Odds Ratio (M‐H, Random, 95% CI)

1.00 [0.06, 16.39]

1.15 Anxiety HADS‐A Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.15.1 Rehabilitation

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.16 Depression HADS‐D Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.16.1 Rehabilitation

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.16.2 Organisation of care

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 1. Intervention versus usual care
Comparison 2. Intervention versus active comparison

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

2.1 Quality of life ‐ CRQ domains Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.1.1 Dyspnoea

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.1.2 Fatigue

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.1.3 Emotion

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.1.4 Mastery

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.2 Functional status ‐ 6MWT Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.2.1 Rehabilitation

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.3 Functional status ‐ ESWT Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.3.1 Rehabilitation

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.4 Functional status ‐ ISWT Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.4.1 Rehabilitation

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.5 Anxiety HADS‐A Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.5.1 Rehabilitation

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.6 Depression HADS‐D Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2.6.1 Rehabilitation

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 2. Intervention versus active comparison