Scolaris Content Display Scolaris Content Display

Intervenciones para el tratamiento de la fatiga en adultos con un tumor cerebral primario

Contraer todo Desplegar todo

Referencias

Boele 2013 {published data only}

Boele FW, Douw L, de Groot M, van Thuijl HF, Cleijne W, Heimans JJ, et al. The effect of modafinil on fatigue, cognitive functioning, and mood in primary brain tumor patients: a multicenter randomized controlled trial. Neuro-oncology 2013;15(10):1420-8. CENTRAL

Laigle‐Donadey 2019 {published data only}10.1093/noajnl/vdz043

Laigle-Donadey F, Ducray F, Boone M, Diallo MH, Hajage D, Ramirez C, et al. A phase III double-blind placebo-controlled randomized study of dexamphetamine sulfate for fatigue in primary brain tumors patients: an ANOCEF trial (DXA). Neuro-oncology Advances 2019;1(1):1-9. CENTRAL

Porter 2020 {published data only}

Porter AB, Liu H, Kohli S, Cerhan JH, Sloan JA, McMurray R, et al. A phase III randomized, double-blind placebo controlled study of armodafinil (Nuvigil) to reduce cancer-related fatigue in patients with high-grade glioma (Alliance A221101). Journal of Clinical Oncology 2020;38(Suppl 15):12007. CENTRAL

Bigatão 2016 {published data only}

Bigatão MD, Peria FM, Tirapelli DP, Carlotti Junior CG. Educational program on fatigue for brain tumor patients: possibility strategy? Arquivos de Neuro-psiquiatria 2016;74(2):155-60. CENTRAL [DOI: 10.1590/0004-282X20160007]

Boele 2018 {published data only}

Boele FW, Klein M, Verdonck-de Leeuw IM, Cuijpers P, Heimans JJ, Snijders TJ, et al. Internet-based guided self-help for glioma patients with depressive symptoms: a randomized controlled trial. Journal of Neuro-oncology 2018;137(1):191-203. CENTRAL [DOI: 10.1007/s11060-017-2712-5]

Butler 2007 {published data only}

Butler JM, Case LM, Atkins J, Frizzell B, Sanders G, Griffin P, et al. A phase III, double-blind, placebo-controlled prospective randomized clinical trial of d-threo-methylphenidate HCl in brain tumor patients receiving radiation therapy. International Journal of Radiation Oncology, Biology, Physics 2007;69(5):1496-501. CENTRAL

Gehring 2009 {published and unpublished data}

Gehring K, Sitskoorn MM, Gundy CM, Sikkes SA, Klein M, Postma TJ, et al. Cognitive rehabilitation in patients with gliomas: a randomized, controlled trial. Journal of Clinical Oncology 2009;27(22):3712-22. CENTRAL

Gehring 2012 {published data only}

Gehring K, Patwardhan SY, Collins R, Groves MD, Etzel CJ, Meyers CA, et al. A randomised trial on the efficacy of methylphenidate and modafinil for improving cognitive functioning and symptoms in patients with a primary brain tumor. Journal of Neuro-oncology 2012;107(1):165-74. CENTRAL [DOI: 10.1007/s11060-011-0723-1]

Gehring 2020 {published data only}

Gehring K, Stuiver MM, Visser E, Kloek C, van den Bent M, Hanse M, et al. A pilot randomized controlled trial of exercise to improve cognitive performance in patients with stable glioma: a proof of concept. Neuro-oncology 2020;22(1):103-15. CENTRAL [DOI: 10.1093/neuonc/noz178]

Hansen 2020 {published data only}

Hansen A, Pedersen CB, Jarden JO, Beier D, Minet LR, Søgaard K. Effectiveness of physical therapy–and occupational therapy-based rehabilitation in people who have glioma and are undergoing active anticancer treatment: single-blind, randomized controlled trial. Physical Therapy 2020;100(3):564-74. CENTRAL [DOI: 10.1093/ptj/pzz180]

Kaleita 2006 {published data only}

Kaleita TA, Wellisch DK, Graham CA, Steh B, Nghiemphu P, Ford JM, et al. Pilot study of modafinil for treatment of neurobehavioral dysfunction and fatigue in adult patients with brain tumors. Journal of Clinical Oncology 2006;24(18S):1503. CENTRAL

Lee 2016 {published data only}

Lee EQ, Muzikansky A, Drappatz J, Kesari S, Wong ET, Fadul CE, et al. A randomized, placebo-controlled pilot trial of armodafinil for fatigue in patients with gliomas undergoing radiotherapy. Neuro-oncology 2016;18(6):849-54. CENTRAL [DOI: 10.1093/neuonc/now007]

Locke 2008 {published data only}

Locke DE, Cerhan JH, Wu W, Malec JF, Clark MM, Rummans TA, et al. Cognitive rehabilitation and problem-solving to improve quality of life of patients with primary brain tumors: a pilot study. Journal of Supportive Oncology 2008;6(8):383-91. CENTRAL

Naughton 2018 {published data only}

Naughton MJ, Case LD, Peiffer A, Chan M, Stieber V, Moore D, et al. Quality of life of irradiated brain tumor survivors treated with donepezil or placebo: results of the WFU CCOP research base protocol 91105. Neuro-oncology Practice 2018;5(2):114-21. CENTRAL [DOI: 10.1093/nop/npx016]

Page 2015 {published and unpublished data}

Page BR, Shaw EG, Lu L, Bryant D, Grisell D, Lesser GJ et al. Phase II double-blind placebo-controlled randomized study of armodafinil for brain radiation-induced fatigue. Neuro-oncology 2015;17(10):1393-401. CENTRAL

Shaw 2006 {published data only}

Shaw EG, Rosdhal R, D'Agostino RB, Lovato J, Naughton MJ, Robbins ME, et al. Phase II study of donepezil in irradiated brain tumor patients: effect on cognitive function, mood, and quality of life. Journal of Clinical Oncology 2006;24(9):1415-20. CENTRAL

Referencias de los estudios en espera de evaluación

Rooney 2020 {published data only}

Rooney AG, Hewins W, Walker A, Withington L, Mackinnon M, Robson S, et al. Innv-27. BT-LIFE (brain tumours, lifestyle interventions, and fatigue evaluation): lessons learned from running a novel multi-sectoral research trial. Neuro-oncology 2020;22(Suppl 2):ii122. CENTRAL

Cordier 2019 {published data only}

Cordier D, Gerber M, Brand S. Effects of two types of exercise training on psychological well-being, sleep, quality of life and physical fitness in patients with high-grade glioma (WHO III and IV): study protocol for a randomized controlled trial. Cancer Communications 2019;39(1):46. CENTRAL [DOI: 10.1186/s40880-019-0390-8]

NCT03259438 {published data only}

NCT03259438. The vitality project for fatigued female cancer survivors. clinicaltrials.gov/ct2/show/NCT03259438 (first received 23 August 2017). CENTRAL

Röttgering 2020 {published data only}

Röttgering J. Cognitive behavioural therapy in treating severe fatigue in patients with primary brain tumors – a randomized controlled clinical trial. trialsearch.who.int/Trial2.aspx?TrialID=NL8711 (first received 14 June 2020). CENTRAL [trialregister.nl/trial/8711]

Arlt 1997

Arlt W, Hove U, Müller B, Reincke M, Berwiler U, Schwab F, et al. Frequency and frequently overlooked: treatment-induced endocrine dysfunction in adult long-term survivors or primary brain tumours. Neurology 1997;49(2):498-506. [PMID: 9270585]

Armstrong 2006

Armstrong TS, Mendoza T, Gring I, Coco C, Cohen MZ, Eriksen L, et al. Validation of the M.D. Anderson Symptom Inventory Brain Tumour Module (MDASI-BT). Journal of Neuro-oncology 2006;80(1):27-35. [PMID: 16598415]

Armstrong 2010

Armstrong TS, Cron SG, Boloanos EV, Gilber MR, Kang DH. Risk factors for fatigue severity in primary brain tumour patients. Cancer 2010;116(11):2707-15. [DOI: 10.1002/cncr.25018]

Armstrong 2012

Armstrong TS, Gilbert MR. Practical strategies for management of fatigue and sleep disorders in people with brain tumors. Neuro-oncology 2012;14(Suppl 4):iv65-iv72. [DOI: 10.1093/neuonc/nos210]

Beck 1979

Beck AT, Rush AJ, Shaw BF, Emery G. Cognitive Therapy of Depression. New York (NY): Guildford Press, 1979.

Bowe 2012

Bowe JE. Fatigue, brain, behavior, and immunity: summary of the 2012 Named Series on fatigue. Behavior and Immunity 2012;26(8):1220-3. [10.1016/j.bbi.2012.08.009]

Cella 1993

Cella DF, Tulsky DS, Gray G, Sarafian B, Linn E, Bonomi A, et al. The Functional Assessment of Cancer Therapy Scale: development and validation of the general measure. Journal of Clinical Oncology 1993;11(3):570-9. [PMID: 8445433]

Coomans 2019

Coomans MB, Dirven L, Aaronson NK, Baumert BG, van den Bent M, Bottomley A, et al. Symptom clusters in newly diagnosed glioma patients: which symptom clusters are independently associated with functioning and global health status? Neuro-oncology 2019;21(11):1447-57.

Cramp 2012

Cramp F, Byron-Daniel J. Exercise for the management of cancer-related fatigue in adults. Cochrane Database of Systematic Reviews 2012, Issue 11. Art. No: CD006145. [DOI: 10.1002/14651858.CD006145.pub2]

Deeks 2001

Deeks JJ. Systematic reviews in health care: systematic reviews of evaluations of diagnostic and screening tests. BMJ 2001;323(7305):157-62. [DOI: 10.1136/bmj.323.7305.157]

DerSimonian 1986

DerSimonian R, Laird N. Meta-analysis in clinical trials. Controlled Clinical Trials 1986;7(3):177-88.

Dimeo 2001

Dimeo FC. Effects of exercise on cancer-related fatigue. Cancer 2001;92(6 Suppl):1689-93.

Drappatz 2007

Drappatz J, Schiff D, Kesari S, Norden AD, Wen P. Medical management of brain tumour patients. Radiotherapy Oncology 2007;100(1):131-6. [DOI: 10.1016/j.ncl.2007.07.015]

Dun 2021

Dun L, Xian-Yi W, Si-Ting H, Xin-Yuan Y. Effects of sleep interventions on cancer-related fatigue and quality of life in cancer patients: a systematic review and meta-analysis. Supportive Care in Cancer 2021;6:1-13. [DOI: 10.1007/s00520-021-06563-5]

Fox 2007

Fox SW, Lyon D, Ferace E. Symptom clusters in patients with high-grade glioma. Journal of Nursing Scholarship 2007;39(1):61-7. [PMID: 17393967]

Glaus 1998

Glaus A. Fatigue in patients with cancer. Analysis and assessment. Recent Results Cancer Research 1998;I44(I-XI):1-172. [PMID: 9551500]

Goedendorp 2009

Goedendorp MM, Gielissen MF, Verhagen CA, Bleijenberg G. Psychosocial interventions for reducing fatigue during cancer treatment in adults. Cochrane Database of Systematic Reviews 2009, Issue 1. Art. No: CD006953. [DOI: 10.1002/14651858.CD006953.pub2]

Grant 2016

Grant R, Brown D. Fatigue randomized controlled trials – how tired is "too tired" in patients undergoing glioma treatment? Neuro-oncology 2016;18(6):759-60. [DOI: 10.1093/neuonc/now052]

Higgins 2011

Higgins JP, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions. Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from training.cochrane.org/handbook/archive/v5.1/.

Hinds 2007

Hinds PS, Hockenberry MJ, Gattuso JS, Srivastava DK, Tong X, Jones H, et al. Dexamethasone alters sleep and fatigue in pediatric patients with acute lymphoblastic leukemia. Cancer 2007;110(10):2321-30. [PMID: 17926333]

Holley 2000

Holley SK. Evaluating patient distress from cancer-related fatigue: an instrument development study. Oncology Nursing Forum 2000;27(9):1425-31. [PMID: 11058974]

ICH‐GCP 1997

International Conference on Harmonisation Expert Working Group. International Conference on Harmonisation of technical requirements for registration of pharmaceuticals for human use. ICH harmonised tripartite guideline. CFR & ICH Guidelines 1997;1(1):19063-2043.

Jean‐Pierre 2007

Jean-Pierre P, Figueroa-Moseley CD, Kohli S, Fiscella K, Palesh OG, Morrow GR. Assessment of cancer-related fatigue: implications for clinical diagnosis and treatment. Oncologist 2007;12(Suppl 1):11-21. [PMID: 17573452]

Katz 2012

Katz LH, Goldvaser H, Gafter-Gvili A, Tur-Kaspa R. Extended peginterferon plus ribavirin treatment for 72 weeks versus standard peginterferon plus ribavirin treatment for 48 weeks in chronic hepatitis C genotype 1 infected slow-responder adult patients. Cochrane Database of Systematic Reviews 2012, Issue 9. Art. No: CD008516. [DOI: 10.1002/14651858.CD008516.pub2]

Kim 2012

Kim BR, Chun MH, Han EY, Kim D-K. Fatigue assessment and rehabilitation outcomes in patients with brain tumours. Supportive Care in Cancer 2012;20(4):805-12. [DOI: 10.1007/s00520-011-1153-5]

Kvale 2009

Kvale EA, Murthy R, Taylor R, Lee JY, Nabors LB. Distress and quality of life in primary high-grade brain tumour patients. Supportive Care in Cancer 2009;17(7):793-9. [DOI: 10.1007/s00520-008-0551-9]

Lawrence 2004

Lawrence DP, Kupelnick B, Miller K, Devine D, Lau J. Evidence report on the occurrence, assessment, and treatment of fatigue in cancer patients. Journal of the National Cancer Institute. Monographs 2004;32:40-50.

Louis 2021

Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. 2021 WHO classification of tumours of the central nervous system. Neuro-Oncology 2021;23(8):1231-51.

Lovely 1999

Lovely MP, Miaskowski C, Dodd M. Relationship between fatigue and quality of life in patients with glioblastoma multiformae. Oncology Nursing Forum 1999;26(5):921-5. [PMID: 10382191]

Lu 2009

Lu Y, Yu W, Wang X. Efficacy of topiramate in adult patients with symptomatic epilepsy: an open-label, long-term, retrospective observation. CNS Drugs 2009;23(4):351-9. [PMID: 19374462]

Maschio 2008

Maschio M, Dinapoli L, Zarabla A, Pompili A, Carapella C, Pace A, et al. Outcome and tolerability of topiramate in brain tumour associated epilepsy. Journal of Neuro-oncology 2008;86(1):61-70. [PMID: 17598071]

Mendoza 1999

Mendoza TR, Wang XS, Cleeland CS, Morrissey M, Johnson BA, Wendt JK, et al. The rapid assessment of fatigue severity in cancer patients: use of the Brief Fatigue Inventory. Cancer 1999;85(5):1186-96. [PMID: 10091805]

Miaskowski 2011

Miaskowski C, Lee K, Dunn L, Dodd M, Aouizerat BE, West C, et al. Sleep–wake circadian activity rhythm parameters and fatigue in oncology patients before the initiation of radiation therapy. Cancer Nursing 2011;34(4):255-68. [DOI: 10.1097/NCC.0b013e3181f65d9b]

Minton 2010

Minton O, Richardson A, Sharpe M, Stone P. Drug therapy for the management of cancer-related fatigue. Cochrane Database of Systematic Reviews 2010, Issue 7. Art. No: CD006704. [DOI: 10.1002/14651858.CD006704.pub3]

Minton 2013

Minton O, Berger A, Barsevick A, Cramp F, Goedendorp M, Mitchell SA, et al. Cancer-related fatigue and its impact on functioning. Cancer 2013;119(S11):2124-30.

Molassiotis 2010

Molassiotis A, Wilson B, Brunton L, Chaudhary H, Gattamaneni R, McBain C. Symptom experience in patients with primary brain tumours: a longitudinal exploratory study. European Journal of Oncology Nursing 2010;14(5):410-6. [DOI: 10.1016/j.ejon.2010.03.001]

NCCN 2018

National Comprehensive Cancer Network (NCCN). Cancer-related fatigue. NCCN Clinical Practice Guidelines in Oncology2018;Version 2.

Pelletier 2002

Pelletier G, Verhoef MJ, Khatri N, Hagen N. Quality of life in brain tumour patients: the relative contribution of depression, fatigue, emotional distress and existential issues. Journal of Neuro-oncology 2002;57(1):41-9. [PMID: 12125966]

Piper 1998

Piper BF, Dibble SL, Dodd MJ, Weiss MC, Slaughter RE, Paul SM. The revised Piper Fatigue Sale: psychometric evaluation in women with breast cancer. Oncology Nursing Forum 1998;25(4):677-87. [PMID: 9599351]

Review Manager 2020 [Computer program]

Nordic Cochrane Centre, The Cochrane CollaborationReview Manager 5 (RevMan 5). Version 5.4. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2020.

Ringdal 1993

Ringdal GI, Ringdal K. Testing the EORTC Quality of Life Questionnaire on cancer patients with heterogeneous diagnoses. Quality of Life Research 1993;2(2):129-40.

Rooney 2011

Rooney AG, Carson A, Grant R. Depression in cerebral glioma patients: a systematic review of observational studies. Journal of the National Cancer Institute 2011;103(1):61-76. [DOI: 10.1093/jnci/djq458]

Roscoe 2002

Roscoe JA, Morrow GR, Hickok JT, Bushunow P, Matteson S, Rakita D, et al. Temporal interrelationships among fatigue, circadian rhythm and depression in breast cancer patients undergoing chemotherapy treatment. Support Care Cancer 2002;10(4):329-36. [PMID: 12029433]

Salo 2002

Salo J, Neimela A, Joukamaa M, Koivukangas J. Effect of brain tumour laterality on patients' perceived quality of life. Journal of Neurology Neurosurgery and Psychiatry 2002;72(3):373-7. [PMID: 11861699]

Schiff 2015

Schiff D, Lee EQ, Nayak L, Norden AD, Reardon D, Wen PY. Medical management of brain tumors and the sequelae of treatment. Neuro-oncology 2015;17(4):488-504.

Servaes 2007

Servaes P, Gielissen M, Verhagen S, Bleijenberg G. The course of severe fatigue in disease-free breast cancer patients: a longitudinal study. Psycho-oncology 2007;16(9):787-95. [PMID: 17086555]

Stein 2004

Stein KD, Jacobsen PB, Blanchard CM, Thors C. Further validation of the Multidimensional Fatigue Symptoms Inventory – Short Form. Journal of Pain and Symptom Management 2004;27(1):14-23. [DOI: 10.1016/j.jpainsymman.2003.06.003]

Stone 2000

Stone P, Richardson A, Ream E, Smith AG, Kerr DJ, Kearney N. Cancer-related fatigue: inevitable, unimportant and untreatable? Results of a multi-centre patient survey. Cancer Fatigue Forum. Annals of Oncology 2000;11(8):971-5. [PMID: 11038033]

Struik 2009

Struik K, Klein M, Heimans JJ, Gielissen MF, Bleijenberg G, Taphoorn MJ, et al. Fatigue in low-grade glioma. Journal of Neuro-oncology 2009;92(1):73-8. [DOI: 10.1007/s11060-008-9738-7]

Taphoorn 1995

Taphoorn MJ, Heimans JJ, van der Veen EA, Karim AB. Endocrine functions in long-term survivors of low-grade supratentorial glioma treated with radiation therapy. Journal of Neuro-oncology 1995;25(2):97-102. [PMID: 8543975]

van Coevorden‐van Loon 2017

van Coevorden-van Loon, Ellen MP, Coomans MB, Heijenbrok-Kal MH, Ribbers GM, van den Bent MJ. Fatigue in patients with low grade glioma: systematic evaluation of assessment and prevalence. Journal of Neuro-oncology 2017;133(2):237-46.

Volkow 2002

Volkow ND, Fowler JS, Wang G, Ding Y, Gatley SJ. Mechanism of action of methylphenidate: insights from PET imaging studies. Journal of Attention Disorders 2002;6(Suppl 1):S31-43. [PMID: 12685517]

WHOQOL Group 1995

The WHOQOL Group. The World Health Organization Quality of Life Assessment (WHOQOL): position paper from the World Health Organization. Social Science and Medicine 1995;41(10):1403-9. [PMID: 8560308]

Winningham 1992

Winningham ML. How exercise mitigates fatigue: implications for people receiving cancer therapy. In: Carroll-Johnson RM, editors(s). The Biotherapy of Cancer V. Pittsburgh (PA): Oncology Nursing Press, 1992:16-21.

Yellen 1997

Yellen SB, Cella DF, Webster K, Blendowski C, Kaplan E. Measuring fatigue and other anemia-related symptoms with the Functional Assessment of Cancer Therapy (FACT) measurement system. Journal of Pain and Symptom Management 1997;13(2):63-74. [PMID: 9095563]

Young 2010

Young JW, Greyer MA. Action of modafinil-increased motivation via the dopamine transporter inhibition and D1 receptors? Biological Psychiatry 2010;67(8):784-7. [DOI: 10.1016/j.biopsych.2009.12.015]

Referencias de otras versiones publicadas de esta revisión

Day 2014

Day J, Yust-Katz S, Cachia D, Rooney A, Katz LH, Wefel J, et al. Interventions for the management of fatigue in adults with a primary brain tumour. Cochrane Database of Systematic Reviews 2014, Issue 11. Art. No: CD011376. [DOI: 10.1002/14651858.CD011376]

Day 2016

Day J, Yust-Katz S, Cachia D, Wefel J, Katz LH, Tremont Lukats IW, et al. Interventions for the management of fatigue in adults with a primary brain tumour. Cochrane Database of Systematic Reviews 2016, Issue 4. Art. No: CD011376. [DOI: 10.1002/14651858.CD011376.pub2]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Boele 2013

Study characteristics

Methods

Double‐blind randomised placebo‐controlled trial

Participants

Inclusion criteria: aged ≥ 18 years; diagnosed with a histologically confirmed glioma or meningioma; no signs of tumour recurrence in last 6 months; fatigue self‐reported CIS score > 27

Exclusion criteria: history of psychiatric disease or symptoms; expected adverse interactions between prescribed medications and modafinil; unable to communicate in Dutch

Number randomised: modafinil: 20; placebo: 17

Age: 48.16 (SD 12.02) years

Sex: 62.2% women; 37.8% men

Tumour type: meningioma (32.4%), low‐grade glioma (37.8%) or high‐grade glioma (29.7%)

Previous treatment: surgery (94.6%), without further radiotherapy (56.8%) or chemotherapy (78.4%)

Follow‐up: 6 weeks and 12 weeks

Setting: 3 centres in the Netherlands

Interventions

2 treatment arms for 6 weeks' duration

Arm I treatment schedule

Week 1: oral modafinil 200 mg per day in 2 divided does (100 mg upon waking, 100 mg at lunchtime)

Week 2–6: oral modafinil 400 mg per day in 2 divided doses (200 mg upon waking, 200 mg at lunchtime)

Week 7: washout period

Week 8–12: matched placebo

Arm II treatment schedule

Week 1–6: matched placebo

Week 7: washout period

Week 8: oral modafinil 200 mg per day in 2 divided does (100 mg upon waking, 100 mg at lunchtime)

Week 9–12: oral modafinil 400 mg per day in 2 divided doses (200 mg upon waking, 200 mg at lunchtime)

Outcomes

Fatigue (CIS)

Depression (Center for Epidemiologic Studies Depression Scale)

Health‐related quality of life (Medical Outcomes Study Short‐Form Health Survey)

Subjective cognitive functioning (Medical Outcomes Study subjective cognitive functioning scale)

Objective cognitive functioning (Rey Auditory Verbal Learning Test, Memory Comparison Test, Stroop Colour Word Test, Letter Digit Substitution Test, Concept Shifting Test, Categorical Word Fluency Test, Concept Shifting Test)

Notes

Mean imputation used where missing values were present in questionnaires or neuropsychological assessments.

No corrections for multiple statistical testing carried out.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A pharmacy randomization system was used to assign participants".

Comment: confirmed via correspondence.

Allocation concealment (selection bias)

Low risk

Quote: "A pharmacy randomization system was used to assign participants".

Comment: confirmed via correspondence.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Patients, treating physicians, and researchers were blind to treatment allocation".

Comment: confirmed via correspondence.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "Patients, treating physicians, and researchers were blind to treatment allocation".

Comment: confirmed via correspondence.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear how much imputation may have affected the result as sensitivity analysis was not carried out to determine whether missing data altered the results of the review.

Similar number of participants dropped out between time points baseline and 6‐week follow‐up (modafinil arm: 4 participants; placebo arm: 3 participants), more participants dropped out of placebo arm (4 participants) than modafinil arm (1 participant) between 6‐week follow‐up and 12‐week follow‐up. Mean imputation was used where missing values were present in questionnaires or neuropsychological assessments. Details of adverse events per group confirmed through correspondence with the lead author.

5 participants dropped out due to adverse events while receiving modafinil. Details per participant were:

  • tingling sensations, depressive feelings, feeling nervous/fidgety, dizziness;

  • depressive feelings, crying without a clear cause;

  • vertigo, feeling as if about to get a seizure, 'light feeling' in head;

  • increased headaches, feeling nervous, tingling sensations, feeling anxious;

  • reduced appetite, nausea, sometimes vomiting, stuffy feeling in head, feeling fidgety.

2 participants dropped out of the trial due to adverse events while receiving placebo. Details per participant were:

  • vertigo, nausea;

  • seizures, fatigue.

Selective reporting (reporting bias)

Low risk

All outcomes reported.

Other bias

Low risk

None.

Laigle‐Donadey 2019

Study characteristics

Methods

Double‐blind, randomised placebo‐controlled trial

Participants

Inclusion criteria: aged ≥ 18 years; KPS > 60; diagnosed with a histologically confirmed and stable PBT; ≥ 3 months since radiotherapy with or without (concurrent) chemotherapy; severe self‐reported fatigue on the 20‐item Multidimensional Fatigue Inventory (score ≥ 60) 

Exclusion criteria: treatable fatigue; other symptoms impacting test completion (e.g. severe aphasia); contraindications for amphetamines; co‐occurring depression (assessed as a HADS score > 8) 

Number randomised: dexamfetamine: 23; placebo: 23 

Age (mean): intervention: 55 years; control: 49 years

Sex: women (48.8%); men (51.2%)

Tumour type: high‐grade glioma (75.6%), low‐grade glioma (7.3%), CNS lymphoma (9.7%) and medulloblastoma (7.3%)

Previous treatment: radiotherapy + chemotherapy: 34 participants; radiotherapy alone: 3 participants; chemotherapy alone: 4 participants

Follow‐up: 3 months 

Setting: multi‐institutional in France

Interventions

2 arms for 3 months' duration

Arm 1 treatment schedule

Day 1–10: dexamfetamine 10 mg in divided doses (5 mg in the morning, 5 mg at noon)

Day 11–20: if previous dose tolerated, dexamfetamine 20 mg in divided doses (2 × 5 mg in the morning, 2 × 5 mg at noon) 

Day 21–30: if previous dose tolerated, dexamfetamine 30 mg taken in divided doses (3 × 5 mg in the morning, 3 × 5 mg at noon) 

Day 31: if previous dose tolerated, dexamfetamine 30 mg dose continued to 3‐month endpoint. If not tolerated, dexamfetamine 10 mg taken in divided doses until 3‐month endpoint

Arm II placebo schedule

Day 1–10: 2 × identical‐appearing tablets (1 × in the morning, 1 at noon) 

Day 11–20: if previous dose tolerated, 4 × identical‐appearing tablets (2 × in the morning, 2 at noon)  

Day 21–30: if previous dose tolerated, 6 × identical‐appearing tablets (3 × in the morning, 3 at noon)  

Day 31: if previous dose tolerated, 6 × identical‐appearing tablets continued to 3‐month endpoint

Outcomes

Fatigue (20‐item Multidimensional Fatigue Inventory) 

Depression (HADS) 

Quality of Life (EORTC QLQ‐C30/EORTC QLQ‐BN 20) 

Cognition (Mattis Dementia Rating Scale, Verbal Fluency‐Category test, Trail Making Test Parts A and B, Wisconsin Card Sorting Test, Marin Apathy Scale, Grober and Buschke test 

Toxicity and adverse events (Norris Scale)

Notes

Did not state any use of imputation for missing data.

No corrections for multiple statistical testing carried out.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Manuscript did not mention how the randomisation was performed and there was no reported analysis to assess whether the 2 groups were different at baseline.

Allocation concealment (selection bias)

Unclear risk

No details provided.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Identical‐appearing tablets and dose schedule.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Stated double‐blind methodology used. Probably done. 

Incomplete outcome data (attrition bias)
All outcomes

Low risk

High retention rate 89% with reasons for drop‐out specified:

  • "progression of disease (n = 1 in the placebo arm),

  • refusal of further therapy (n = 1 in the placebo arm), 

  • exclusion criteria discovered only after inclusion because they had previously been masked by the patients themselves (depression and hyperthyroid disease, 2 patients in the placebo arm), 

  • and loss to follow‐up (1 participant in the DXA [dexamfetamine] arm)"

Selective reporting (reporting bias)

Low risk

Data not analysed for 1 cognitive test due to about 50% missing data. All other outcomes reported.

Other bias

Low risk

None.

Porter 2020

Study characteristics

Methods

Double‐blind randomised placebo‐controlled trial

Participants

Inclusion criteria: adults diagnosed with a high‐grade glioma; ≥ 4 weeks following radiotherapy; previous surgery or biopsy with concurrent radiotherapy and chemotherapy; clinically stable or improved KPS compared to previous month; stable dose of corticosteroids; fatigue self‐reported > 6 on the 'worst' item of the BFI

Exclusion criteria: pregnancy

Number randomised: armodafinil 150 mg: 103; armodafinil 250 mg: 97; placebo: 97

Age: 57.3 (SD 12.3) years

Sex: women: 41.2%; men: 58.8%

Tumour type: clinically stable high‐grade glioma; no further details

Previous treatment: completed radiotherapy but ongoing chemotherapy was allowed; no further details

Follow‐up: 4 weeks and 8 weeks

Setting: USA

Interventions

2 treatment arms, 1 placebo arm, for 8 weeks' duration

Arm I treatment schedule

Oral armodafinil 150 mg taken in the morning for 8 weeks

Arm II treatment schedule

Oral armodafinil 250 mg taken in the morning for 8 weeks

Arm III placebo schedule

Oral placebo taken in the morning for 8 weeks

Outcomes

Fatigue (BFI)

Health‐related quality of life (Linear Analogue Self‐Assessment)

Objective cognitive functioning (Symbol Digit Modalities Test)

Adverse events (Common Terminology Criteria for Adverse Events Version 4.0)

Notes

Data not yet published – data available via ClinicalTrials.gov

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

There was no mention how the randomisation was performed and there was no reported analysis to assess whether the 2 groups were different at baseline.

Allocation concealment (selection bias)

Unclear risk

No details provided.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind methodology reported. Probably done.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details provided.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No details of reasons missing data.

Selective reporting (reporting bias)

Low risk

All outcomes reported.

Other bias

Low risk

None.

BFI: Brief Fatigue Inventory; CIS: Checklist Individual Strength; EORTC QLQ‐BN 20: European Organization for Research and Treatment of Cancer Quality of Life Questionnaire–Brain Neoplasm; EORTC QLQ‐C30: European Organization for Research and Treatment of Cancer Quality of Life Questionnaire‐C30; HADS: Hospital Anxiety and Depression Scale; KPS: Karnofsky Performance Scale; PBT: primary brain tumour.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Bigatão 2016

Fatigue was not a necessary inclusion criterion.

Boele 2018

Fatigue was not a necessary inclusion criterion.

Butler 2007

Fatigue was not a necessary inclusion criterion.

Gehring 2009

Fatigue was not a necessary inclusion criterion.

Gehring 2012

Fatigue was not a necessary inclusion criterion.

Gehring 2020

Fatigue was not a necessary inclusion criterion.

Hansen 2020

Fatigue was not a necessary inclusion criterion.

Kaleita 2006

Fatigue was not a necessary inclusion criterion.

Lee 2016

Fatigue was not a necessary inclusion criterion.

Locke 2008

Fatigue was not a necessary inclusion criterion.

Naughton 2018

Fatigue was not a necessary inclusion criterion. No control group.

Page 2015

Fatigue was not a necessary inclusion criterion.

Shaw 2006

Fatigue was not a necessary inclusion criterion. No control group.

Characteristics of studies awaiting classification [ordered by study ID]

Rooney 2020

Methods

Randomised controlled feasibility trial

Participants

Inclusion criteria: adults aged ≥ 18 years diagnosed with a primary brain tumour; ≥ 3 months since radiotherapy or chemotherapy (or both); clinically and radiologically stable; self‐reported fatigue > 4 on Brief Fatigue Inventory

Exclusion criteria: inability to give informed consent; clinically unstable; radiological progression; significant cognitive or sensory impairment

Number randomised: 46

Follow‐up: after interventions and at 16 weeks

Setting: 4 centres across the UK

Interventions

2 treatment arms and 1 self‐guided support arm, of 16 weeks

Arm I 'Health Coaching' schedule

Participants received an information leaflet and 'Health Coaching', which involved an individualised programme based on physical measurements and lifestyle information. Participants wore an activity monitoring device and recorded information in a diary. Participants were offered 5 × 45‐minute Health Coaching sessions (delivered remotely or in person) focused on setting personal goals to change lifestyle factors contributing to fatigue.

Arm II 'Health Coaching plus Patient Activation' schedule

Participants received the same intervention as in arm I plus Patient Activation. Participants were offered 2 × 1‐hour Patient Activation sessions, delivered by a trained coach, focused on improving their approach to managing fatigue.

Arm III control schedule

Participants were given fatigue management information alongside treatment as usual.

Outcomes

Feasibility (participant recruitment and retention, acceptability, manageability)

Fatigue (Brief Fatigue Inventory)

Depression (Hospital Anxiety and Depression Scale)

Quality of Life (Psychological Outcome Profiles)

Notes

Data not yet published

Characteristics of ongoing studies [ordered by study ID]

Cordier 2019

Study name

Effects of two types of exercise training on psychological well‐being, sleep, quality of life and physical fitness in patients with high‐grade glioma (WHO III and IV): study protocol for a randomized controlled trial

Methods

Randomised controlled clinical trial

Participants randomly assigned to receive 1 of 2 exercise courses or to a control group

Participants

Inclusion criteria: adults aged 18–75 years with a diagnosis of high‐grade glioma, following treatment (surgery with radiotherapy or chemotherapy, or both)

Exclusion criteria: diagnosis of severe psychiatric disorder or severe health comorbidities (e.g. severe diabetes, severe cardiovascular disease)

Interventions

Arm 1: endurance training for 2 × 40‐ to 60‐minute sessions per week for 6 weeks

Arm 2: resistance training for 2 × 40‐ to 60‐minute sessions per week for 6 weeks

Arm 3: control group – individualised counselling "not intended to actively improve participants' well‐being" (taken from clinicaltrials.gov/ct2/show/NCT03775369)

Outcomes

Self‐reported fatigue (Functional Assessment of Cancer Therapy – Fatigue, Fatigue Severity Scale)

Self‐reported depression (Hamilton Depression Rating Scale)

Self‐reported intolerance of uncertainty (Intolerance of Uncertainty Scale)

Self‐reported measure of insomnia (Insomnia Severity Index)

Self‐reported stress (Perceived Stress Scale)

Self‐reported mental toughness (Mental Toughness Questionnaire)

Self‐reported physical activity (International Physical Activity Questionnaire)

Objective walking capacity (6‐minute walking test)

Objective grip strength (grip force)

Other measures:

Change in sleep continuity and sleep architecture (assessed by electroencephalogram)

Change in C reactive protein (assessed by blood test)

Starting date

2018

Contact information

Serge Brand: [email protected]

Dominik Cordier: [email protected]

Tel: 161‐2074‐782

Notes

ClinicalTrials.gov identifier: NCT03775369

Current status: recruiting

NCT03259438

Study name

The Vitality Project for fatigued female cancer survivors

Methods

Double‐blind randomised controlled trial

Participants randomly assigned to receive 1 of 2 courses: 2 hours and 15 minutes twice weekly of qigong (mind–body movement) or healthy living (nutrition and exercise) classes

Participants

Inclusion criteria: women aged 18–70 years; cancer treatment completed ≥ 8 weeks prior to enrolment, including chemotherapy, radiotherapy, surgery, or a combination of these; ≥ 3/10 on self‐reported measure of fatigue interference in daily life; understands English; willingness to complete EEG, ECG, EMG and fMRI, and questionnaires, and have blood drawn; able to pass basic physical movement assessment to verify safety for enrolment into the study

Exclusion criteria: history of coronary artery or coronary heart disease, myocardial infarction or heart murmur; pacemaker implant; peripheral neuropathy in hands; history of a major psychiatric disorder, not including depression or anxiety; active substance misuse; tobacco use; pregnancy; consumption of caffeine or cocoa products within 2 hours prior to data collection; inability to complete gentle exercise

Follow‐up: after 10‐week intervention, and 3 months after intervention

Setting: USA

Interventions

Arm 1: 2 hours and 15 minutes twice weekly qigong classes, focusing on mind–body movement, for 10 weeks

Arm 2: 2 hours and 15 minutes twice weekly healthy living counselling focusing on nutrition, and aerobic and strengthening exercise, for 10 weeks

Outcomes

Self‐reported fatigue (FACIT‐Fatigue Scale, Fatigue Symptom inventory)

Physical measurements (ECG, impedance cardiography, EEG, tactile acuity, EMG, precision grip, electrodermal activity, mechanical lung function, inflammatory cytokines, resting state fMRI, muscle strength, 6‐minute walk test) 

Objective measure of cognition (short‐form OSpan (operation span))

Self‐reported anxiety and depression (Patient Health Questionnaire, Profile of Mood States Questionnaire)

Self‐reported emotional dysregulation (Difficulties in Emotion Regulation Scale)

Self‐reported physical, social, emotional and functional well‐being (FACT‐G)

Self‐reported quality of life (Rand 36‐item Short Form Health Survey)

Self‐reported sleep quality, habits and patterns (Pittsburgh Sleep Quality Index)

Heart rate and step count (via Apple watch)

Self‐reported stress (Perceived Stress Scale)

Self‐reported social support (Multidimensional Scale of Perceived Social Support)

Self‐reported tendency to care for others before themselves (Unmitigated Communion Scale)

Self‐reported quantity of exercising and relaxing (Godin Leisure time questionnaire)

Starting date

2017

Contact information

Ellen Flynn

Miriam Hospital Outpatient, Providence, RI, USA

Tel: +1 401‐793‐7020; email: [email protected]   

Notes

ClinicalTrials.gov Identifier: NCT03259438

Current status: unknown as of November 2021

Röttgering 2020

Study name

Cognitive behavioral therapy in treating severe fatigue in patients with primary brain tumors – a randomized controlled clinical trial

Methods

Randomised controlled trial

Psychological intervention or control group

Participants

Inclusion criteria: adults aged > 18 years with a histological diagnosis of PBT; with stable disease; ≥ 2 months since active treatment; expected survival ≥ 3 months; able to read, write and speak Dutch; and scoring over a clinical cut‐off score for severe fatigue on the CIS‐20

Exclusion criteria: other causes of fatigue not related to brain tumour treatment or receiving pharmacological treatment for fatigue in the past 3 months. Those experiencing depression, primary sleep disorders, receiving psychological intervention for a psychiatric disorder, currently (or within 3 months of being) pregnant or a KPS score < 70

Interventions

Arm 1: cognitive behaviour therapy involving therapist sessions and online modules, for 12 weeks

Arm 2: control group (details unknown)

Outcomes

Self‐reported fatigue (CIS – Fatigue score)

Self‐reported depression, anxiety and health‐related quality of life

Objective measure of neurocognitive functioning

Starting date

2020

Contact information

Jantine Röttgering

Tel: +31 020‐4448‐453; email: [email protected]

Notes

Netherlands Trial Register identifier: NL8711

Current status: unknown

CIS‐20: 20‐item Checklist Individual Strength; ECG: electrocardiogram; EEG: electroencephalogram; EMG: electromyography; FACIT‐F: Functional Assessment of Chronic Illness Therapy – Fatigue; FACT‐G: Functional Assessment of Cancer Therapy – General; fMRI: functional magnetic resonance imaging; KPS: Karnofsky Performance Scale; PBT: primary brain tumour; WHO: World Health Organization.

Data and analyses

Open in table viewer
Comparison 1. Boele 2013: modafinil versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Fatigue Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 1.1

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 1: Fatigue

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 1: Fatigue

1.1.1 Concentration problems

1

55

Mean Difference (IV, Fixed, 95% CI)

‐1.06 [‐3.18, 1.06]

1.1.2 Reduced motivation

1

55

Mean Difference (IV, Fixed, 95% CI)

‐0.48 [‐2.93, 1.97]

1.1.3 Reduced activity

1

55

Mean Difference (IV, Fixed, 95% CI)

‐1.01 [‐5.64, 3.62]

1.1.4 Fatigue severity

1

55

Mean Difference (IV, Fixed, 95% CI)

‐0.22 [‐0.79, 0.35]

1.2 Objective cognitive functioning Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 2: Objective cognitive functioning

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 2: Objective cognitive functioning

1.2.1 Verbal memory

1

53

Mean Difference (IV, Fixed, 95% CI)

0.26 [‐0.05, 0.57]

1.2.2 Working memory

1

53

Mean Difference (IV, Fixed, 95% CI)

0.07 [‐0.04, 0.18]

1.2.3 Attentional functioning

1

53

Mean Difference (IV, Fixed, 95% CI)

‐0.03 [‐0.05, ‐0.01]

1.2.4 Information processing

1

53

Mean Difference (IV, Fixed, 95% CI)

0.17 [‐1.19, 1.53]

1.2.5 Executive functioning

1

53

Mean Difference (IV, Fixed, 95% CI)

0.14 [‐9.33, 9.61]

1.2.6 Psychomotor speed

1

53

Mean Difference (IV, Fixed, 95% CI)

0.10 [‐0.26, 0.46]

1.3 Subjective cognitive functioning Show forest plot

1

55

Mean Difference (IV, Fixed, 95% CI)

1.62 [‐0.74, 3.98]

Analysis 1.3

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 3: Subjective cognitive functioning

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 3: Subjective cognitive functioning

1.4 Depression Show forest plot

0

0

Mean Difference (IV, Fixed, 95% CI)

Not estimable

Analysis 1.4

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 4: Depression

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 4: Depression

1.5 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 1.5

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 5: Health‐related quality of life

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 5: Health‐related quality of life

1.5.1 Physical

1

55

Mean Difference (IV, Fixed, 95% CI)

1.34 [‐20.11, 22.79]

1.5.2 Mental

1

55

Mean Difference (IV, Fixed, 95% CI)

‐1.40 [‐4.84, 2.04]

1.6 Adverse events Show forest plot

1

55

Risk Ratio (M‐H, Fixed, 95% CI)

2.79 [0.59, 13.16]

Analysis 1.6

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 6: Adverse events

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 6: Adverse events

Study flow diagram. RCT: randomised controlled trial.

Figuras y tablas -
Figure 1

Study flow diagram. RCT: randomised controlled trial.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Figuras y tablas -
Figure 2

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 1: Fatigue

Figuras y tablas -
Analysis 1.1

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 1: Fatigue

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 2: Objective cognitive functioning

Figuras y tablas -
Analysis 1.2

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 2: Objective cognitive functioning

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 3: Subjective cognitive functioning

Figuras y tablas -
Analysis 1.3

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 3: Subjective cognitive functioning

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 4: Depression

Figuras y tablas -
Analysis 1.4

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 4: Depression

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 5: Health‐related quality of life

Figuras y tablas -
Analysis 1.5

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 5: Health‐related quality of life

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 6: Adverse events

Figuras y tablas -
Analysis 1.6

Comparison 1: Boele 2013: modafinil versus placebo, Outcome 6: Adverse events

Summary of findings 1. Boele 2013: modafinil compared with placebo for fatigue in people with a primary brain tumour

Modafinil compared with placebo for fatigue in people with a primary brain tumour

Patient or population: people with a primary brain tumour

Settings: hospital, outpatient

Intervention: modafinil

Comparison: placebo

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No. of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Placebo

Modafinil

Fatigueconcentration problems

Subscale from CIS range: 0–35

(follow‐up: 6 weeks)

The mean concentration problem score ranged across control groups from 15.91 to 23.91 points

The mean concentration problem score in the intervention groups was 1.06 lower

(3.18 lower to 1.06 higher)

37 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicate a higher level of concentration problems.

Fatiguereduced motivation

Subscale from CIS range: 0–28

(follow‐up: 6 weeks)

The mean reduced motivation score ranged across control groups from 10.22 to 19.48 points

The mean reduced motivation score in the intervention groups was 0.48 lower

(2.93 lower to 1.97 higher)

37 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicate lower motivation.

Fatiguereduced activity

Subscale from CIS range: 0–21

(follow‐up: 6 weeks)

The mean reduced activity score ranged across control groups from 3.84 to 21.34 points

The mean reduced activity score in the intervention groups was 1.01 lower

(5.64 lower to 3.62 higher)

37 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicate lower activity.

Fatigueseverity

Subscale from CIS range: 0–56

The mean fatigue severity score ranged across control groups from 34.06 to 36.22 points

The mean fatigue severity score in the intervention groups was 0.22 lower

(0.79 lower to 0.35 higher)

37 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicate a higher level of fatigue.

Adverse events

(follow‐up: 6 weeks)

Low‐risk population

RR 2.79 (0.59 to 13.16)

37 (1)

⊕⊝⊝⊝
Very lowa

30 per 100
(1 to 180)

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (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).

CIS: Checklist Individual Strength; CI: confidence interval; RR: risk ratio.

GRADE Working Group grades of evidence
High certainty: further research is very unlikely to change our confidence in the estimate of effect.
Moderate certainty: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low certainty: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low certainty: we are very uncertain about the estimate.

aDowngraded three levels; there was very low‐certainty evidence due to low accrual, such that recruitment did not meet the estimated power requirement to detect a true effect.

Figuras y tablas -
Summary of findings 1. Boele 2013: modafinil compared with placebo for fatigue in people with a primary brain tumour
Summary of findings 2. Laigle‐Donadey 2019: dexamfetamine sulfate compared with placebo for fatigue in primary brain tumour

Dexamfetamine sulfate compared with placebo for fatigue in primary brain tumour

Patient or population: people with a primary brain tumour

Settings: hospital, outpatient

Intervention: dexamfetamine sulfate

Comparison: placebo

Outcomes

Median change from baseline (IQR)

Between‐group comparison

No. of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Placebo

dexamfetamine sulfate

Fatigue

MFI‐20 scale

(follow‐up: 3 months)

The median difference in fatigue score between baseline and 3‐month follow‐up in the control group was 14 points (IQR 4.5 to 24.25)

The median difference in fatigue score between baseline and 3‐month follow‐up in the dexamfetamine sulfate group was 4 points lower (IQR 4 to 13)

41 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicate a higher level of fatigue.

Fatigue ('asthenia')

Norris Scale

The median difference in fatigue score between baseline and 3‐month follow‐up in the control group was 4.25 points (IQR –0.06 to 12.16)

The median difference in fatigue score between baseline and 3‐month follow‐up in the dexamfetamine sulfate group was 2.31 points lower (IQR –10.88 to 13.96)

41 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicate a higher level of fatigue.

Cognition

Trail Making Test A

The median difference in score between baseline and 3‐month follow‐up in the control group was 6.5 points (IQR –0.75 to 17.5)

The median difference in score between baseline and 3‐month follow‐up in the dexamfetamine sulfate group was 7.5 points lower (IQR –4 to 10)

41 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicate poorer cognition.

Cognition

Trail Making Test B

The median difference in score between baseline and 3‐month follow‐up in the control group was –2 points (IQR –16 to 21.75)

The median difference in score between baseline and 3‐month follow‐up in the dexamfetamine sulfate group was 18 points higher (IQR –37 to 39)

41 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicate poorer cognitive performance.

Cognition

Semantic Fluency

The median difference in score between baseline and 3‐month follow‐up in the control group was 1 points (IQR –1 to 3)

The median difference in score between baseline and 3‐month follow‐up in the dexamfetamine sulfate group was 1 point lower (IQR –5 to 3)

41 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicate better cognitive performance.

Cognition Lexical Fluency

The median difference in score between baseline and 3‐month follow‐up in the control group was 1 point (IQR –0.75 to 2.75)

The median difference in score between baseline and 3‐month follow‐up in the dexamfetamine sulfate group was 2 points lower (IQR –4 to 4)

41 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicate better cognitive performance.

Cognition Episodic memory (Grober and Buschke test)

The median difference in score between baseline and 3‐month follow‐up in the control group was –0.5 points (IQR –4.25 to 2.5)

The median difference in score between baseline and 3‐month follow‐up in the dexamfetamine sulfate group was 0.5 points higher (IQR –4.5 to 4)

41 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicate better cognitive performance.

Cognition Mattis Scale

The median difference in score between baseline and 3‐month follow‐up in the control group was –2 (IQR –7 to 1)

The median difference in fatigue score between baseline and 3‐month follow‐up in the dexamfetamine sulfate group was 1point higher (IQR –4 to 0)

41 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicate better cognitive performance.

Affectivity Norris Scale

The median difference in score between baseline and 3‐month follow‐up in the control group was 8.25 (IQR 2.34 to 18.09)

The median difference in score between baseline and 3‐month follow‐up in the dexamfetamine sulfate group was 5.87 points lower (IQR –10.78 to 10)

41 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicate higher affectivity.

Apathy Marin Scale

The median difference in score between baseline and 3‐month follow‐up in the control group was 0.5 (IQR –6 to 2.75)

The median difference in score between baseline and 3‐month follow‐up in the dexamfetamine sulfate group was 1.5 points higher (IQR –2 to 11)

41 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicated higher apathy.

Depression HADS

41 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicate more severe depression symptoms.

Quality of life EORTC QLQ‐C30

41 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicate better quality of life.

Quality of life EORTC QLQ‐BN 20

41 (1)

⊕⊝⊝⊝
Very lowa

Higher scores indicate better quality of life.

EORTC QLQ‐BN 20: European Organization for Research and Treatment of Cancer Quality of Life Questionnaire–Brain Neoplasm; EORTC QLQ‐C30: European Organization for Research and Treatment of Cancer Quality of Life Questionnaire‐C30; HADS: Hospital Anxiety and Depression Scale; IQR: interquartile range; MFI‐20: 20‐item Multidimensional Fatigue Inventory.

GRADE Working Group grades of evidence
High certainty: further research is very unlikely to change our confidence in the estimate of effect.
Moderate certainty: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low certainty: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low certainty: we are very uncertain about the estimate.

aDowngraded three levels; there was very low‐certainty evidence due to low accrual, such that recruitment did not meet the estimated power requirement to detect a true effect.

Figuras y tablas -
Summary of findings 2. Laigle‐Donadey 2019: dexamfetamine sulfate compared with placebo for fatigue in primary brain tumour
Summary of findings 3. Porter 2020: armodafinil compared with placebo for fatigue in high‐grade glioma

Armodafinil (150 mg and 250 mg) compared with placebo for fatigue

Patient or population: people with primary brain tumour

Settings: hospital, outpatient

Intervention: armodafinil (150 mg and 250 mg)

Comparison: placebo

Outcomes

Group comparisons

No. of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Placebo

Armodafinil 150 mg

Armodafinil 250 mg

Fatigue Brief Fatigue Inventory (number of participants with an improvement in 2 points from baseline at 8 weeks (%))

29 (29.9%)

27 (27.8%)

29 (28.2%)

297 (1)

⊕⊕⊝⊝
Lowa

Higher scores indicate higher fatigue.

Cognition Symbol Digit Modalities Test (median (range))

0.0 (–3.4 to 4.9)

0.0 (–1.6 to 2.5)

0.3 (–3.4 to 2.5)

180 (1)

⊕⊕⊝⊝
Lowa

Higher scores indicate better performance.

Quality of life Linear Analogue Self Assessment (median (range))

2.0 (–15.0 to 27.0

4.0 (–32.0 to 26.0)

3.0 (–16.0 to 27.0)

232 (1)

⊕⊕⊝⊝
Lowa

Higher scores indicate better quality of life.

Adverse events Grade 3 or higher (number of participants (%))

3 (2.8%)

6 (5.5%)

8 (7.3%)

328 (1)

⊕⊕⊝⊝
Lowa

GRADE Working Group grades of evidence
High certainty: further research is very unlikely to change our confidence in the estimate of effect.
Moderate certainty: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low certainty: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low certainty: we are very uncertain about the estimate.

aDowngraded two levels; there was low‐certainty evidence due to the level of unclear risk of bias relating to selection, detection and attrition.

Figuras y tablas -
Summary of findings 3. Porter 2020: armodafinil compared with placebo for fatigue in high‐grade glioma
Comparison 1. Boele 2013: modafinil versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Fatigue Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

1.1.1 Concentration problems

1

55

Mean Difference (IV, Fixed, 95% CI)

‐1.06 [‐3.18, 1.06]

1.1.2 Reduced motivation

1

55

Mean Difference (IV, Fixed, 95% CI)

‐0.48 [‐2.93, 1.97]

1.1.3 Reduced activity

1

55

Mean Difference (IV, Fixed, 95% CI)

‐1.01 [‐5.64, 3.62]

1.1.4 Fatigue severity

1

55

Mean Difference (IV, Fixed, 95% CI)

‐0.22 [‐0.79, 0.35]

1.2 Objective cognitive functioning Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

1.2.1 Verbal memory

1

53

Mean Difference (IV, Fixed, 95% CI)

0.26 [‐0.05, 0.57]

1.2.2 Working memory

1

53

Mean Difference (IV, Fixed, 95% CI)

0.07 [‐0.04, 0.18]

1.2.3 Attentional functioning

1

53

Mean Difference (IV, Fixed, 95% CI)

‐0.03 [‐0.05, ‐0.01]

1.2.4 Information processing

1

53

Mean Difference (IV, Fixed, 95% CI)

0.17 [‐1.19, 1.53]

1.2.5 Executive functioning

1

53

Mean Difference (IV, Fixed, 95% CI)

0.14 [‐9.33, 9.61]

1.2.6 Psychomotor speed

1

53

Mean Difference (IV, Fixed, 95% CI)

0.10 [‐0.26, 0.46]

1.3 Subjective cognitive functioning Show forest plot

1

55

Mean Difference (IV, Fixed, 95% CI)

1.62 [‐0.74, 3.98]

1.4 Depression Show forest plot

0

0

Mean Difference (IV, Fixed, 95% CI)

Not estimable

1.5 Health‐related quality of life Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

1.5.1 Physical

1

55

Mean Difference (IV, Fixed, 95% CI)

1.34 [‐20.11, 22.79]

1.5.2 Mental

1

55

Mean Difference (IV, Fixed, 95% CI)

‐1.40 [‐4.84, 2.04]

1.6 Adverse events Show forest plot

1

55

Risk Ratio (M‐H, Fixed, 95% CI)

2.79 [0.59, 13.16]

Figuras y tablas -
Comparison 1. Boele 2013: modafinil versus placebo