Scolaris Content Display Scolaris Content Display

Carbamazepine versus phenobarbitone monotherapy for epilepsy: an individual participant data review

Esta versión no es la más reciente

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Banu 2007 {published and unpublished data}

Banu SH, Jahan M, Koli UK, Ferdousi S, Khan NZ, Neville B. Side effects of phenobarbital and carbamazepine in childhood epilepsy: randomised controlled trial. BMJ 2007;334(7605):1207. CENTRAL

Bidabadi 2009 {unpublished data only}

Bidabadi E. Comparison of the effects of phenobarbital versus carbamazepine as single drug therapy in partial seizure with secondary generalization in children. Epilepsia 2009;50 Suppl 10:167. CENTRAL

Cereghino 1974 {published data only}

Cereghino JJ, Brock JT, Van Meter JC, Penry JK, Smith LD, White BG. Carbamazepine for epilepsy. A controlled prospective evaluation. Neurology 1974;24(5):401‐10. CENTRAL

Chen 1996 {published data only}

Chen YJ, Kang WM, So WCM. Comparison of antiepileptic drugs on cognitive function in newly diagnosed epileptic children: a psychometric and neurophysiological study. Epilepsia 1996;37(1):81‐6. CENTRAL

Cossu 1984 {published data only}

Cossu G, Monaco F, Piras MR, Grossi E. Short‐term therapy with carbamazepine and phenobarbital: effects on cognitive functioning in temporal lobe epilepsy [Trattamento a breve termine con carbamazepina e fenobarbital: efftto sulle funzioni mnesiche nell'epilessia temporale]. Bollettino Lega Italiana contro l'Epilessia 1984;45/46:377‐9. CENTRAL

Czapinski 1997 {unpublished data only}

Czapinski P, Terczynski A, Czapinska E. Randomised 36‐month comparative study of valproic acid, phenytoin, phenobarbital and carbamazepine efficacy in patients with newly diagnosed epilepsy with partial complex seizures. Epilepsia 1997;38 Suppl(3):42. CENTRAL

de Silva 1996 {published and unpublished data}

de Silva M, MacArdle B, McGowan M, Hughes E, Stewart J, Neville BG, et al. Randomised comparative monotherapy trial of phenobarbitone, phenytoin, carbamazepine, or sodium valproate for newly diagnosed childhood epilepsy. Lancet 1996;347(9003):709‐13. CENTRAL

Feksi 1991 {published and unpublished data}

Feksi AT, Kaamugisha J, Sander JW, Gatiti S, Shorvon SD. Comprehensive primary health care antiepileptic drug treatment programme in rural and semi‐urban Kenya. ICBERG (International Community‐based Epilepsy Research Group). Lancet 1991;337(16):406‐9. CENTRAL

Heller 1995 {published and unpublished data}

Heller AJ, Chesterman P, Elwes RD, Crawford P, Chadwick D, Johnson AL, et al. Phenobarbitone, phenytoin, or sodium valproate for newly diagnosed epilepsy: a randomized comparative monotherapy trial. Journal of Neurology, Neurosurgery, and Psychiatry 1995;58(1):44‐50. CENTRAL

Mattson 1985 {published and unpublished data}

Mattson RH, Cramer JA, Collins JF, Smith DB, Delgado‐Escueta AV, Browne TR, et al. Comparison of carbamazepine, phenobarbital, phenytoin, and primidone in partial and secondarily generalized tonic‐clonic seizures. New England Journal of Medicine 1985;313(3):145‐51. CENTRAL

Mitchell 1987 {published data only}

Mitchell WG, Chavez JM. Carbamazepine versus phenobarbital for partial onset seizures in children. Epilepsia 1987;28(1):56‐60. CENTRAL

Ogunrin 2005 {published and unpublished data}

Ogunrin O, Adamolekun B, Ogunniyi A. Cognitive effects of anti‐epileptic drugs in Nigerians with epilepsy. African Journal of Neurological Sciences 2005;24(1):18‐24. CENTRAL

Placencia 1993 {published and unpublished data}

Placencia M, Sander JW, Shorvon SD, Roman M, Alarcon F, Bimos C, et al. Antiepileptic drug treatment in a community health care setting in northern Ecuador: a prospective 12‐month assessment. Epilepsy Research 1993;14(3):237‐44. CENTRAL

References to studies excluded from this review

Bird 1966 {published data only}

Bird CAK, Griffin BP, Miklaszewska JM, Galbraith AW. Tegretol (carbamazepine): a controlled trial of a new anti‐convulsant. British Journal of Psychiatry 1966;112:737‐42. CENTRAL

Castro‐Gago 1998 {published data only}

Castro‐Gago M, Eiris‐Punal J, Novo‐Rodriguez MI, Couceiro J, Camina F, Rodriguez‐Segade S. Serum carnitine levels in epileptic children before and during treatment with valproic acid, carbamazepine, and phenobarbital. Journal of Child Neurology 1998;13(11):546‐9. CENTRAL

Cereghino 1973 {published data only}

Cereghino JJ, Brock JT, White BG, Penry JK. Evaluation of carbamazepine in epileptic patients. Neurology 1973;23:433. CENTRAL

Hansen 1980 {published data only}

Hansen BS, Dam M, Brandt J, Hvidberg EF, Angelo H, Christensen JM, et al. Influence of dextropropoxyphene on steady state serum levels and protein binding of three anti‐epileptic drugs in man. Acta Neurologica Scandinavica 1980;61(6):357‐67. CENTRAL

Kuzuya 1993 {published data only}

Kuzuya T, Hasegawa T, Shimizu K, Nabeshima T. Effect of anti‐epileptic drugs on serum zinc and copper concentrations in epileptic patients. International Journal of Clinical Pharmacology, Therapy, & Toxicology 1993;31(2):61‐5. CENTRAL

Marjerrison 1968 {published data only}

Marjerrison G, Jedlicki SM, Keogh RP, Hrychuk W, Poulakakis GM. Carbamazepine: behavioral, anticonvulsant and EEG effects in chronically‐hospitalized epileptics. Diseases of the Nervous System 1968;29(2):133‐6. CENTRAL

Meador 1990 {published data only}

Meador KJ, Loring DW, Huh K, Gallagher BB, King DW. Comparative cognitive effects of anticonvulsants. Neurology 1990;40:391‐4. CENTRAL

Sabers 1995 {published data only}

Sabers A, Moller A, Dam M, Smed A, Arlien‐Soborg P, Buchman J, et al. Cognitive function and anticonvulsant therapy: effect of monotherapy in epilepsy. Acta Neurologica Scandinavica 1995;92(1):19‐27. CENTRAL

Smith 1987 {published data only}

Smith DB, Mattson RH, Cramer JA, Collins JF, Novelly RA, Craft B. Results of a nationwide Veterans Administration Cooperative Study comparing the efficacy and toxicity of carbamazepine, phenobarbital, phenytoin, and primidone. Epilepsia 1987;28(Suppl 3):S50‐8. CENTRAL

Annegers 1999

Annegers JF, Dubinsky S, Coan SP, Newmark ME, Roht L. The incidence of epilepsy and unprovoked seizures in multiethnic, urban health maintenance organizations. Epilepsia 1999;40(4):502‐6.

Atkins 2004

Atkins D, Best D, Briss PA, Eccles M, Falck‐Ytter Y, Flottorp S, et al. GRADE Working Group. Grading quality of evidence and strength of recommendations. BMJ 2004;328(7454):1490.

Baulac 2002

Baulac M, Cramer JA, Mattson RH. Phenobarbital and other barbiturates: adverse effects. In: Levy RH, Mattson RH, Meldrum BS, et al. editor(s). Antiepileptic Drugs. 5th Edition. Philadelphia: Lippincott Williams & Wilkins, 2002:528‐40.

Cockerell 1995

Cockerell OC, Johnson AL, Sander JW, Hart YM, Shorvon SD. Remission of epilepsy: results from the National General Practice Study of Epilepsy. Lancet 1995;346(8968):140‐4.

Commission 1981

Commission on Classification and Terminology of the International League Against Epilepsy. Proposal for revised clinical and electroencephalographic classification of epileptic seizures. Epilepsia 1981;22(4):489‐501.

Commission 1989

Commission on Classification and Terminology of the International League Against Epilepsy. Proposal for revised classification of epilepsies and epileptic syndromes. Epilepsia 1989;30(4):389‐99.

Gruber 1962

Gruber CM, Brock JT, Dyken MD. Comparison of the effectiveness of phenobarbital, mephobarbital, primidone, dipheylhydantoin, ethotoin, metharbital, and methylphenylhydantion in motor seizures. Clinical Pharmacology and Therapeutics 1962;3:23‐8.

Hauser 1993

Hauser WA, Annegers JF, Kurland LT. Incidence of epilepsy and unprovoked seizures in Rochester, Minnesota 1935 ‐ 1984. Epilepsia 1993;34:453‐68.

Higgins 2003

Higgins JPT, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta‐analyses. BMJ 2003;327:557‐60.

Higgins 2011

Higgins JPT, Green S (editors). Cochrane Handbookfor Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org..

Hirtz 2007

Hirtz D, Thurman DJ, Gwinn‐Hardy K, Mohamed M, Chaudhuri AR, Zalutsky R. How common are the "common" neurologic disorders?. Neurology 2007;68:326‐37.

ILAE 1998

ILAE Commission on Antiepileptic Drugs. Considerations on designing clinical trials to evaluate the place of new antiepileptic drugs in the treatment of newly diagnosed and chronic patients with epilepsy. Epilepsia 1998;39(7):799‐803.

ILAE 2006

Glauser T, Ben‐Menachem E, Bourgeois B, Cnaan A, Chadwick D, Guerreiro C, et al. ILAE treatment guidelines: evidence based analysis of antiepileptic drug efficacy and effectiveness as initial monotherapy for epileptic seizures and syndromes. Epilepsia 2006;47(7):1094‐120.

Jones 1996

Jones B, Jarvis P, Lewis JA, Ebbutt AF. Trials to assess equivalence: the importance of rigorous methods. BMJ 1996;313(7048):36‐9.

Juul‐Jenson 1983

Juul‐Jenson P, Foldspang A. Natural history of epileptic seizures. Epilepsia 1983;24:297‐312.

Kirkham 2010

Kirkham JJ, Dwan KM, Altman DG, Gamble C, Dodd S, Smyth R, et al. The impact of outcome reporting bias in randomised controlled trials on a cohort of systematic reviews. BMJ 2010;340:c365.

Kwan 2000

Kwan P, Brodie MJ. Early identification of refractory epilepsy. New England Journal of Medicine 2000;342:314‐9.

Lefebvre 2011

Lefebvre C, Manheimer E, Glanville J. Chapter 6: Searching for studies. In: 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 handbook.cochrane.org.

Liporace 1994

Liporace JD, Sperling MR, Dichter MA. Absence seizures and carbamazepine in adults. Epilepsia 1994;35(5):1026‐8.

MacDonald 1995

MacDonald RL, Kelly KM. Antiepileptic drug mechanisms of action. Epilepsia 1995;36(Suppl 2):S2‐12.

MacDonald 2000

MacDonald BK, Johnson AL, Goodridge DM, Cockerell OC, Sander JWA, Shorvon SD. Factors predicting prognosis of epilepsy after presentation with seizures. Annals of Neurology 2000;48:833‐41.

Malafosse 1994

Malfosse A, Genton P, Hirsch E, Marescaux C, Broglin D, Bernasconi R. Idiopathic Generalised Epilepsies: Clinical, Experimental and Genetic. Eastleigh: John Libbey and Company, 1994.

Marson 2000

Marson AG, Williamson PR, Hutton JL, Clough HE, Chadwick DW. Carbamazepine versus valproate monotherapy for epilepsy. Cochrane Database of Systematic Reviews 2000, Issue 3. [DOI: 10.1002/14651858.CD001030]

Matlow 2012

Matlow J, Koren G. Is carbamazepine safe to take during pregnancy?. Canadian Family Physician 2012;58:163‐4.

Meador 2008

Meador K, Reynolds M, Crean S, Fahrbach K, Probst C. Pregnancy outcomes in women with epilepsy: a systematic reviews and meta‐analysis of published pregnancy registries and cohorts. Epilepsy Research 2008;81:1‐13.

Moher 2009

Moher D, Liberati A, Tetzlaff J, Altman DG, The PRISMA Group. Preferred Reporting Items for Systematic Reviews and Meta‐Analyses: The PRISMA Statement. BMJ 2009;339:2535.

Morrow 2006

Morrow J, Russel A, Guthrie E, Parsons L, Robertson I, Waddell R, et al. Malformation risks of antiepileptic drugs in pregnancy: a prospective study from the UK Epilepsy and Pregnancy Register. Journal of Neurology, Neurosurgery, and Neuropsychiatry 2006;77(2):193‐8.

Murray 1994

Murray CJL, Lopez AD. Global comparative assessments in the health sector. World Health Organization. Geneva, 1994.

Ngugi 2010

Ngugi AK, Bottomley C, Kleinschmidt I, Sander JW, Newton CR. Estimation of the burden of active and life‐time epilepsy: a meta‐analytic approach. Epilepsia 2010;51:883‐90.

NICE 2012

National Institute for Health and Care Excellence. The epilepsies: the diagnosis and management of the epilepsies in adults and children in primary and secondary care; Clinical Guidance 137. London: National Institute for Health and Care Excellence, 2012.

Nolan 2013a

Nolan SJ, Sutton L, Marson A, Tudur Smith C. Consistency of outcome and statistical reporting of time‐to‐event data: the impact on Cochrane Reviews and meta‐analyses in epilepsy. 21st Cochrane Colloquium: Better Knowledge for Better Health. Quebec City, 2013:114‐5.

Nolan 2013b

Nolan SJ, Marson AG, Pulman J, Tudur Smith C. Phenytoin versus valproate monotherapy for partial onset seizures and generalised onset tonic‐clonic seizures. Cochrane Database of Systematic Reviews 2013, Issue 8. [DOI: 10.1002/14651858.CD001769.pub2]

Olafsson 2005

Olafsson E, Ludvigsson P, Gudmundsson G, Hesdorfer D, Kjartansson O, Hauser WA. Incidence of unprovoked seizures and epilepsy in Iceland and assessment of the epilepsy syndrome classification: a prospective study. Lancet Neurology 2005;4:627‐34.

Pal 1998

Pal DK, Das T, Chaudhury G, Johnson AL, Neville BG. Randomised controlled trial to assess acceptability of phenobarbital for childhood epilepsy in rural India. Lancet 1998;351(9095):19‐23.

Parmar 1998

Parmar MK, Torri V, Stewart L. Extracting summary statistics to perform meta‐analyses of the published literature for survival endpoints. Statistics in Medicine 1998;17(24):2815‐34.

Ragsdale 1991

Ragsdale DS, Scheuer T, Catterall WA. Frequency and voltage dependent inhibition of type hA Naı channels, expressed in a mammalian cell line, by local anesthetic, antiarrhythmic, and anticonvulsant drugs. Molecular Pharmacology 1991;40:756‐65.

Rho 1996

Rho JM, Donevan SD, Rogawski MA. Direct activation of GABAA receptors by barbiturates in cultured rat hippocampal neurons. The Journal of Physiology 1996;497(2):509‐22.

Sander 1996

Sander JW, Shorvon SD. Epidemiology of the epilepsies. Journal of Neurology, Neurosurgery, and Psychiatry 1996;61(5):433‐43.

Sander 2004

Sander JW. The use of anti‐epileptic drugs ‐ principles and practice. Epilepsia 2004;45(6):28‐34.

Shakir 1980

Shakir RA. Sodium valproate, phenytoin and carbamazepine as sole anticonvulsants. The place of sodium valproate in the treatment of epilepsy. London: Academic Press Inc (London) Ltd and the Royal Society of Medicine, 1980:7‐16.

Shields 1983

Shields WD, Saslow E. Myoclonic, atonic, and absence seizures following institution of carbamazepine therapy in children. Neurology 1983;33:1487‐9.

Snead 1985

Snead OC, Hosey LC. Exacerbation of seizures in children by carbamazepine. New England Journal of Medicine 1985;313:916‐21.

Stata 2009 [Computer program]

StataCorp. Stata Statistical Software: Release 11. CollegeStation, TX: StataCorp LP, 2009.

Trimble 1988

Trimble MR, Cull C. Children of school age: the influence of antiepileptic drugs on behavior and intellect. Epilepsia 1988;29(Suppl 3):S15‐19.

Tudur Smith 2007

Tudur Smith C, Marson AG, Chadwick DW, Williamson PR. Multiple treatment comparisons in epilepsy monotherapy trials. Trials 2007;5(8):34.

Wallace 1997

Wallace H, Shorvon SD, Hopkins A, O'Donoghue M. Guidelines for the Clinical Management of Adults with Poorly Controlled Epilepsy. London: Royal College of Physicians, 1997.

Williamson 2002

Williamson PR, Tudur Smith C, Hutton JL, Marson AG. Aggregate data meta‐analysis with time‐to‐event outcomes. Statistics in Medicine 2002;21(11):3337‐51.

References to other published versions of this review

Nolan 2015

Nolan SJ, Marson AG, Weston J, Tudur Smith C. Carbamazepine versus phenobarbitone monotherapy for epilepsy: an individual participant data review. Cochrane Database of Systematic Reviews 2015, Issue 7. [DOI: 10.1002/14651858.CD001904.pub2]

Tudur 2000

Tudur C, Marson AG, Williamson PR, Hutton JL, Chadwick DW. Carbamazepine vs phenobarbitone monotherapy for epilepsy. Cochrane Database of Systematic Reviews 2000, Issue 1. [DOI: 10.1002/14651858.CD001904]

Tudur Smith 2003

Tudur Smith C, Marson AG, Williamson PR. Carbamazepine versus phenobarbitone monotherapy for epilepsy. Cochrane Database of Systematic Reviews 2003, Issue 1. [DOI: 10.1002/14651858.CD001904]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Banu 2007

Methods

Single‐centre, double‐blind randomised controlled trial of participants recruited from clinical referral to a multidisciplinary child development centre at a children's hospital in Dhaka, Bangladesh

2 treatment arms: CBZ and PB

Participants

108 children between the ages of 2 to 15 with 2 or more generalised tonic‐clonic, partial, or secondarily generalised seizures in the previous year

Number randomised: CBZ = 54, PB = 54

61 male children (56%)

59 with partial seizures (55%)

26 had previous AED treatment (24%)

Mean age (range): 6 (2 to 15 years)

Study duration: 12 months

Range of follow‐up: 0 to 20.5 months

Interventions

Monotherapy with CBZ (immediate release) or PB

Starting daily dose: CBZ = 1.5 mg/kg/day, PB = 5 mg/kg/day

Maximum daily dose: CBZ = 4 mg/kg/day, PB = 16 mg/kg/day

Outcomes

  • Seizure control: seizure freedom during the last quarter of the 12‐month follow‐up

  • Time to first seizure after randomisation

  • Time to treatment withdrawal due to adverse events

  • Change in behaviour from baseline according to age‐appropriate questionnaire

  • Incidence of behavioural side‐effects

Notes

We received IPD for all randomised participants. We received reasons for withdrawal of allocated treatment as well as the date of the last follow‐up visit, but withdrawal of allocated treatment did not always coincide with the date of the last follow‐up visit (i.e. several participants had the allocated treatment substituted for the other trial drug and continued to be followed up). Dates of withdrawal of allocated treatment could not be provided; therefore, we could not calculate 'time to withdrawal of allocated treatment'. We received the date of first seizure after randomisation, but dates of other seizures in the follow‐up time could not be provided; therefore, we calculated 'time to first seizure' for all participants, but we could not calculate the time to six‐ and 12‐month remission.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Participants were 'randomly assigned to treatment'; the method of randomisation was not stated and not provided by the authors.

Allocation concealment (selection bias)

Low risk

Allocation was concealed by sealed envelopes prepared on a different site to the site of recruitment of participants.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants, a psychologist, and a therapist were blinded throughout the trial. The treating physician was unblinded for practical and ethical reasons.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

A researcher performing outcome assessment was blinded throughout the trial but unblinded for analysis. It was unclear if this could have influenced the results.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Attrition rates were reported. We analysed all randomised participants from the IPD provided².

Selective reporting (reporting bias)

Low risk

We calculated 1 outcome for this review from the IPD provided². We could not calculate other outcomes for this review as the appropriate data were not recorded/not available. All cognitive outcomes from the study were well reported.

Other bias

High risk

There were inconsistencies between rates of seizure recurrence between the data provided and the published paper, which the authors could not resolve (see Sensitivity analysis).

Bidabadi 2009

Methods

Six‐month, systematic, simple randomised trial of children referred to a child neurology clinic (the author was from Guilan University of Medical Sciences, Iran, so it was likely that the study was also conducted there)

2‐arm trial: CBZ and PB

Participants

Children aged 2 to 12 years with partial seizures with secondary generalisation

Number randomised: CBZ = 36, PB = 35

36 male children (53%)

100% partial seizures,

the per cent newly diagnosed was not stated

Age range: 2 to 12 years

Study duration: 6 months

Mean follow‐up: not stated

Interventions

Monotherapy with PB or CBZ. Doses started or achieved not stated

Outcomes

  • Proportion seizure‐free

  • Response rate and rate of side‐effects

  • Seizure frequency and seizure duration

Notes

The trial was reported in abstract form only with very limited information. Outcomes chosen for this review were not reported; IPD were not available.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The trial was described as a 'systematic simple randomised study'; no further information was provided.

Allocation concealment (selection bias)

Unclear risk

No information was provided.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No information was provided on blinding.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information was provided on blinding.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No attrition rates were reported; it was unclear if all participants were analysed.

Selective reporting (reporting bias)

Unclear risk

There was no protocol available; the study was available in abstract format only. Outcomes for this review were not available.

Other bias

Low risk

We detected no other bias.

Cereghino 1974

Methods

Randomised, double‐blind cross‐over trial with 3, 21‐day treatment periods and a 2‐week washout period (regular medications used)

3 treatment arms: CBZ, phenytoin, and PB

Participants

Institutionalised adult participants with uncontrolled seizures on current medication

Number randomised: PB = 45, CBZ = 45

41 participants (91%) with partial epilepsy

28 (62%) male participants 

Age range: 18 to 51 years

Study duration: 13 weeks (3 x 21‐day treatment periods plus 2 x 2‐week washout periods)

Interventions

Monotherapy with PB or CBZ
Daily dose: PB = 300 mg/day or CBZ = 1200 mg/day

Outcomes

  • Behaviour outcomes

  • Adverse effects

  • Seizure frequency

  • Time to treatment withdrawal due to poor seizure control

Notes

The outcomes chosen for this review were not reported due to the cross‐over design of the trial.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation of groups from random number tables (confirmed by author).

Allocation concealment (selection bias)

Unclear risk

No information provided.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No information provided on blinding.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information provided on blinding.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Withdrawal rates reported, no further information provided.

Selective reporting (reporting bias)

Low risk

All efficacy and tolerability outcomes specified in the methods sections reported well in the results section. No protocol available, outcomes for this review not available due to trial cross‐over design.

Other bias

High risk

Cross‐over design may not be appropriate for monotherapy designs, likely carryover effects from one period to another so the comparison may not be entirely monotherapy.

Chen 1996

Methods

Randomised, parallel group study conducted in Taiwan

3 treatment arms: CBZ, PB, sodium valproate

Participants

Children with 2 or more previously untreated unprovoked epileptic seizures

Number randomised: PB = 25, CBZ = 26; number analysed: PB = 23, CBZ = 25 (see notes)

Mean age (range): PB = 9.9 (7 to 15 years), CBZ = 10.8 (7 to 15 years)

CBZ versus PB: 26 (54%) participants with partial epilepsy

25 (52%) male participants

Study duration: 12 months

Range of follow‐up: not stated

Interventions

Monotherapy with PB or CBZ. Dose started or achieved not stated

Outcomes

  • Cognitive/psychometric outcomes: IQ (WISC‐R scale) and developmental delay (Bender‐Gestalt test)

  • Auditory event‐related potentials (neurophysiological outcome)

  • Incidence of allergic reactions

  • Seizure control

Notes

2 children from the PB group and 1 child from the CBZ group withdrew from the study because of allergic reactions. Published results were presented for children who completed the study only. Outcomes chosen for this review were not reported; IPD were not available.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were allocated with "simple randomisation of block size 3."

Allocation concealment (selection bias)

Unclear risk

No information was provided.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The cognitive assessor was 'single‐blinded', implying that participants and personnel were unblinded, but no further information was provided.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The cognitive assessor was single‐blinded.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Withdrawal rates were reported; results were presented only for those who completed the study (CBZ versus PB: 3/51 (6%) excluded from analysis). An ITT approach was not taken.

Selective reporting (reporting bias)

Low risk

All cognitive, efficacy, and tolerability outcomes specified in the methods sections were reported well in the results section. No protocol was available. Outcomes chosen for this review were not reported.

Other bias

Low risk

We detected no other bias

Cossu 1984

Methods

Randomised, double‐blind study to assess short‐term therapy of CBZ and PB on cognitive and memory function conducted in Italy 3 treatment arms: CBZ, PB, and placebo

Participants

Participants with newly diagnosed and untreated temporal lobe epilepsy with no seizures in the previous month

Number randomised: CBZ = 6, PB = 6

100% partial (temporal lobe epilepsy), 100% newly diagnosed

Mean age (SD): CBZ = 26.33 (9.73) years, PB = 18.5 (2.56) years

Age range: 15 to 45 years

1 male and 5 females in each group

Study duration: 3 weeks; all participants completed in 3 weeks

Interventions

Monotherapy with CBZ or PB, Dose started and achieved not stated

Outcomes

  • Changes in memory function from baseline after 3 weeks of treatment (verbal, visual, (visual‐verbal and visual‐non‐verbal), acoustic, tactile, and spatial)

Notes

The trial was published in Italian; the characteristics and outcomes were translated. Outcomes chosen for this review were not reported; IPD were not available.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The trial was described as randomised ('randomizzazione' in Italian); no further information was available.

Allocation concealment (selection bias)

Unclear risk

No information provided.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Trial is described as double‐blind ('condizioni di doppia cecità' in Italian), we assume this refers to participants and personnel.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information provided on blinding of outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants completed this short study and contribute to analysis.

Selective reporting (reporting bias)

Unclear risk

Cognitive and memory outcomes described in methods section well reported in results section. No seizure outcomes or adverse events reported and outcomes chosen for this review not reported. No protocol available so unclear if seizure outcomes were planned a priori.

Other bias

High risk

Very small participant numbers and very short‐term follow‐up. Unclear if this study was adequately powered and of sufficient duration to detect differences.

Czapinski 1997

Methods

36‐month randomised comparative study

4 treatment arms: CBZ, sodium valproate, phenytoin, PB

Participants

Adults with newly diagnosed epilepsy with partial complex seizures

Number randomised: PB = 30, CBZ = 30

100% partial epilepsy (partial complex seizures)

Age range: 18 to 40 years

Percentage male and range of follow‐up: not mentioned

Interventions

Monotherapy with PB or CBZ

Starting doses CBZ = 400 mg/day, PB = 100 mg/day. Dose achieved not stated

Outcomes

  • Proportion achieving 24‐month remission at 3 years and exclusions after randomisation due to adverse effects or no efficacy

Notes

This was an abstract only. Outcomes chosen for this review were not reported. IPD were pledged but not received.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The study was randomised, but no further information was provided.

Allocation concealment (selection bias)

Unclear risk

No information was provided.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No information was provided.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information was provided.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

"Exclusion rates" were reported for all treatment groups; no further information was provided.

Selective reporting (reporting bias)

Unclear risk

No protocol was available; the study was available in abstract format only. Outcomes for this review were not available.

Other bias

Low risk

We detected no other bias.

de Silva 1996

Methods

Randomised, parallel group, open‐label paediatric study conducted in 2 centres in the UK

4 treatment arms: CBZ, sodium valproate, phenytoin, PB

Participants

Children with newly diagnosed epilepsy (2 or more untreated partial or generalised tonic‐clonic seizures in the 12 months preceding the study)

Number randomised: PB = 10, CBZ = 54 (see notes)

35 children (55%) with partial epilepsy

34 (53%) male children

Mean age (range): 9 (3 to 16) years

Range of follow‐up: 3 to 88 (months)

Interventions

Monotherapy with PB or CBZ

Median daily dose achieved: PB = not stated; CBZ = 400 mg/day

Outcomes

  • Time to first seizure recurrence after start of therapy

  • Time to 12‐month remission from all seizures

  • Adverse effects and withdrawals due to adverse events

Notes

6 of the first 10 children assigned to PB had unacceptable adverse effects, so no further children were assigned to PB. The 10 children randomised to PB were retained in analysis. We received IPD for all outcomes of this review.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A randomisation list was generated using permuted blocks of size 8 or 16 with stratification for centre, seizure type, and presence of neurological signs.

Allocation concealment (selection bias)

Low risk

Allocation was concealed via 4 batches of concealed opaque envelopes.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinded ‐ the authors stated that masking of treatment would not have been "practicable or ethical" and would have "undermine[d] compliance". Lack of masking could have led to early withdrawal of the PB arm from the trial.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinded ‐ the authors stated masking of treatment would not have been "practicable or ethical" and would have "undermine[d] compliance". Lack of masking could have led to early withdrawal of the PB arm from the trial, which was likely to have influenced the overall results.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Attrition rates were reported; we analysed all randomised participants from the IPD provided²

Selective reporting (reporting bias)

Low risk

All outcomes were reported or calculated with the IPD provided²

Other bias

Low risk

We detected no other bias

Feksi 1991

Methods

Randomised parallel group trial conducted among residents of the Nakuru district, a semi‐urban population of rural Kenya

2 treatment arms: CBZ and PB

Participants

Participants had a history of generalised tonic‐clonic seizures and at least 2 generalised tonic‐clonic seizures within the preceding year (with or without other seizure types) and untreated in the 3 months prior to the study. 79 (26%) participants had been treated in the past with AEDs

Number randomised: PB = 150, CBZ = 152

115 (38%) of participants had experienced partial seizures

173 (57%) male participants

Mean age (range): 21 (6 to 65 years)

Range of follow‐up: participants followed up for up to 1 year

Interventions

Monotherapy with CBZ or PB

Starting doses: PB: 6 to 10 years of age: 30 mg/day, 11 to 15 years of age: 45 mg/day, 16+ years of age: 60 mg/day

CBZ: 6 to 10 years of age: 400 mg/day, 11 to 15 years of age: 500 mg/day, 16+ years of age: 600 mg/day

Dose achieved not stated

Outcomes

  • Adverse effects

  • Withdrawals from allocated treatment

  • Seizure frequency (during second 6 months of study)

Notes

IPD were made available but not used because of inconsistencies and problems with the data provided (see Included studies for further details).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Participants randomised with random number list, no information provided on method of generating random list.

Allocation concealment (selection bias)

Low risk

Allocation concealed via sealed opaque envelopes (information provided by study author).

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No information provided.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information provided.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Attrition rates reported, results presented only for participants completing 12 months follow‐up (results not presented for 53 (17.5%) participants out of 302 who withdrew from treatment), approach is not ITT.

Selective reporting (reporting bias)

Low risk

No protocol available, outcomes chosen for this review not reported. Seizure outcomes and adverse events well reported.

Other bias

High risk

Inconsistencies with IPD and published results so IPD could not be used (see Included studies for further details).

Heller 1995

Methods

Randomised, parallel group, open‐label study conducted in 2 centres in the UK

4 treatment arms: CBZ, sodium valproate, phenytoin, PB

Participants

Adults with newly diagnosed epilepsy (2 or more untreated partial or generalised tonic‐clonic seizures in the 12 months preceding the study)

Number randomised: PB = 58, CBZ = 61

49 participants (41%) with partial epilepsy

55 (46%) male participants

Mean age (range): 32 (13 to 77) years

Range of follow‐up: 1 to 91 months

Interventions

Monotherapy with PB or CBZ. Median daily dose achieved: PB = 105 mg/day; CBZ = 600 mg/day

Outcomes

  • Time to first seizure recurrence after start of therapy

  • Time to 12‐month remission from all seizures

  • Adverse effects and withdrawals due to adverse events

Notes

We received IPD for all outcomes of this review.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation list generated using permuted blocks of size 8 or 16 with stratification for centre, seizure type and presence of neurological signs.

Allocation concealment (selection bias)

Low risk

Allocation concealed via 4 batches of concealed opaque envelopes.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinded, authors state masking of treatment would not be “practical” and would have “introduced bias due to a very large dropout rate.” Lack of blinding may have lead to more withdrawals of PB.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinded, authors state masking of treatment would not be “practical” and would have “introduced bias due to a very large dropout rate.” Lack of blinding may have lead to more withdrawals of PB which is likely to have influenced the overall results.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Attrition rates reported, all randomised participants analyses from IPD provided²

Selective reporting (reporting bias)

Low risk

All outcomes reported or calculated with IPD provided²

Other bias

Low risk

No other bias detected

Mattson 1985

Methods

Multicentre, randomised, parallel group, double‐blinded study over 10 centres in the USA with separate randomisation schemes used for each seizure type

4 treatments: CBZ, phenytoin, PB, primidone

Participants

Adults with previously untreated or under‐treated simple or complex partial or secondary generalised tonic‐clonic seizures

Number randomised: CBZ = 155, PB = 155

100% partial epilepsy

268 (88%) male participants

Mean age (range): 41 (18 to 82) years

Range of follow‐up: 1 to 177 months

Interventions

Monotherapy with PB or CBZ

Median daily dose achieved: PB = 160 mg/day; CBZ = 800 mg/day

Outcomes

  • Participant retention/time to drug failure (length of time participant continued to take randomised drug)

  • Composite scores of seizure frequency (seizure rates and total seizure control) and toxicity

  • Incidence of side‐effects

Notes

We received IPD for all outcomes of this review

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Participants were randomised with stratification for seizure type. The method of randomisation was not stated and not provided by the authors.

Allocation concealment (selection bias)

Unclear risk

No information was provided in the publication or by the study authors.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The trial was double‐blind (participants and personnel), which was achieved using an additional blank tablet.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It was unclear if outcome assessment was blinded; no information was provided.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Attrition rates were reported; we analysed all randomised participants from the IPD provided².

Selective reporting (reporting bias)

Low risk

All outcomes were reported or calculated with the IPD provided².

Other bias

Unclear risk

We detected no other bias.

Mitchell 1987

Methods

Randomised, double‐blind, single‐centre, parallel paediatric study conducted in Los Angeles, USA

2 treatment arms: CBZ and PB

Participants

Children with newly diagnosed epilepsy

Number randomised: PB = 18, CBZ = 15

100% partial epilepsy, 100% newly diagnosed

20 (61%) male children

Mean age (range): PB = 7.89 (2 to 12 years), CBZ = 6.07 (2 to 12 years)

Study duration: 12 months

Range of follow‐up: not reported

Interventions

Monotherapy with PB or CBZ. Doses started and achieved not stated

Outcomes

  • Change in cognitive, intelligence (IQ), behavioural, and psychometric scores between baseline, 6 months, and 12 months

  • Compliance, drug changes, and withdrawal rates

  • Seizure control at 6 and 12 months (excellent/good/fair/poor)

Notes

33 participants were randomised to PB (18) and CBZ (15) in this study; 6 children were enrolled into a six‐month pilot study (PB (4) CBZ (2)) prior to the randomised study. The 6 children were included in six‐month follow‐up psychometric data.

Outcomes for this review were not reported; IPD were not available.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

33 children were "randomised using a scheme that balanced drug distribution by age and sex"; no further details were provided on the randomisation scheme. 6 non‐randomised children were also used in some analyses.

Allocation concealment (selection bias)

Unclear risk

No information was provided.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The trial blinded participants (and parents); clinicians were unblinded for clinical follow‐up.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The trial blinded psychometric (cognitive) testers blinded for clinical follow‐up.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Attrition rates were reported; results were reported for all children who completed each stage of follow‐up.

Selective reporting (reporting bias)

Low risk

Cognitive/behavioural outcomes, seizure control outcomes, and adverse events were all well reported. No protocol was available; outcomes for this review were not reported.

Other bias

High risk

There was evidence that the study may have been underpowered to detect differences (e.g. 55% power to find a 5‐point difference in IQ score). The behavioural questionnaire was not fully validated. Non‐randomised children from a pilot study were included in the results for psychometric outcomes and medical outcomes.

Ogunrin 2005

Methods

Double‐blinded, parallel group, randomised study conducted in a single‐centre in Nigeria. 3 treatment arms: carbamazepine, phenytoin, phenobarbitone

Participants

Consectuive newly diagnosed participants aged 14 or over presenting at the outpatient neurology clinic of the University Teaching Hopsital, Benin City, Nigeria, with recurrent, untreated afebrile seizures
Number randomised: PB = 18, CBZ = 19
7 participants with partial seizures (19%)

22 male participants (59%)

Mean age (range): 23.62 years (14 to 38 years)
Range of follow‐up: all participants followed up for 12 weeks

Interventions

Monotherapy with PB or CBZ. Median daily dose (range): PB = 120 mg (60 to 180 mg), CBZ = 600 mg (400 mg to 1200 mg)

Outcomes

  • Cognitive measures (reaction times, mental speed, memory, attention)

Notes

We received IPD for all randomised participants. The study duration was 12 weeks; all participants completed the study without withdrawing; therefore, we could not calculate the outcomes 'time to withdrawal of allocated drug', 'time to six‐month remission', and 'time to 12‐month remission'. We calculated 'time to first seizure' from the IPD provided

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The study randomised participants using simple randomisation: Each participant was asked to pick 1 from a table of numbers (1 to 60); the numbers corresponded to allocation of 1 of 3 drugs (the author provided information).

Allocation concealment (selection bias)

Low risk

Recruitment/randomisation of participants and allocations of treatments took place on different sites (the author provided information).

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants were single‐blinded. The study did not blind the research assistant recruiting participants and counselling on medication adherence.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Investigators performing cognitive assessments were single‐blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants completed the study. We analysed all randomised participants from the IPD provided².

Selective reporting (reporting bias)

Low risk

We calculated 1 outcome for this review from the IPD provided². Other outcomes for this review were not available because of short study length. All cognitive outcomes from the study were well reported.

Other bias

Low risk

We detected no other bias.

Placencia 1993

Methods

Randomised parallel group study conducted in the context of existing community health care in a rural highland area of a developing country (Ecuador)

Participants

Participants with a history of at least 2 afebrile seizures and no previous AED treatment in the 4 weeks preceding the study were eligible

Number randomised: PB = 97, CBZ = 95

133 participants (69%) with partial epilepsy

67 (35%) male participants

Mean age (range): PB = 28.6 (2 to 68 years), CBZ = 29.2 (2 to 68 years)

Study duration: 12 months

Range of follow‐up: 0 to 53.4 months

Interventions

Monotherapy with PB or CBZ. Minimum maintenance doses by age groups:

2 to 5 years: PB: 15 mg/day, CBZ: 150 mg/day; 6 to 10 years: PB: 30 mg/day, CBZ: 300 mg/day; 11 to 15 years: PB: 45 mg/day, CBZ: 500 mg/day; > 16 PB: 60 mg/day, CBZ: 600 mg/day. Doses gradually increased

Doses achieved not stated

Outcomes

  • Proportion seizure‐free at 3‐, 6‐, and 12‐month follow‐ups

  • Proportion seizure‐free, with more than 50% seizure reduction and no change in seizure frequency in 6‐ to 12‐month follow‐up period

  • Incidence of adverse effects

Notes

We received IPD for all outcomes used in this review. Results in the published paper were given for 139 participants who completed 6 months' follow‐up, but we received IPD for all 192 participants randomised.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants randomised with random number list, no information provided on method of generating random list.

Allocation concealment (selection bias)

High risk

Allocation concealed used sealed opaque envelopes but method not used for all participants (information provided by study author).

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No information provided.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information provided.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Attrition rates reported, all randomised participants analysed from IPD provided².

Selective reporting (reporting bias)

Low risk

All outcomes were reported or calculated with the IPD provided².

Other bias

High risk

Inconsistencies between number and reasons of withdrawals between the data and the published paper which could not be resolved by the authors (see Sensitivity analysis).

AED: antiepileptic drug
CBZ: carbamazepine
IPD: individual participant data
IQ: intelligence quotient
ITT: intention‐to‐treat
PB: phenobarbitone
WISC‐R scale: the Wechsler Intelligence Scale for Children

²For studies for which we received IPD (Banu 2007; de Silva 1996; Heller 1995; Mattson 1985; Ogunrin 2005; Placencia 1993), attrition and reporting bias were reduced as we requested attrition rates and unpublished outcome data.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Bird 1966

It was unclear whether this trial was randomised and whether participants received either CBZ or PB as monotherapy.

Castro‐Gago 1998

The trial was not randomised, and the treatment choice was made based on types of seizures.

Cereghino 1973

This was a preliminary analysis of Cereghino 1974.

Hansen 1980

The trial was not randomised; participants were already on CBZ or PB monotherapy upon entry into the study.

Kuzuya 1993

The trial was not randomised; participants were already on CBZ or PB monotherapy upon entry into the study.

Marjerrison 1968

CBZ or PB therapy were added to current treatment. We could not make a comparison between CBZ monotherapy and PB monotherapy.

Meador 1990

We could not make a comparison between CBZ monotherapy and PB monotherapy. This was a cross‐over trial, but some participants were receiving treatment at the start of the first period, which had to be withdrawn slowly.

Sabers 1995

The trial was not fully randomised: "The treatment was chosen at random unless the individual diagnoses required a specific drug."

Smith 1987

This reported the same trial as Mattson 1985, and Mattson 1985 gave more relevant information.

CBZ: carbamazepine
PB: phenobarbitone

Data and analyses

Open in table viewer
Comparison 1. Carbamazepine versus phenobarbitone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Time to withdrawal of allocated treatment Show forest plot

4

676

Hazard Ratio (Fixed, 95% CI)

1.49 [1.15, 1.94]

Analysis 1.1

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 1 Time to withdrawal of allocated treatment.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 1 Time to withdrawal of allocated treatment.

2 Time to withdrawal of allocated treatment ‐ stratified by epilepsy type Show forest plot

4

676

Hazard Ratio (Fixed, 95% CI)

1.50 [1.15, 1.95]

Analysis 1.2

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 2 Time to withdrawal of allocated treatment ‐ stratified by epilepsy type.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 2 Time to withdrawal of allocated treatment ‐ stratified by epilepsy type.

2.1 Generalised onset

3

156

Hazard Ratio (Fixed, 95% CI)

1.53 [0.81, 2.88]

2.2 Partial onset

4

520

Hazard Ratio (Fixed, 95% CI)

1.49 [1.12, 2.00]

3 Time to 12‐month remission Show forest plot

4

683

Hazard Ratio (Fixed, 95% CI)

0.93 [0.72, 1.19]

Analysis 1.3

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 3 Time to 12‐month remission.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 3 Time to 12‐month remission.

4 Time to 12‐month remission ‐ stratified by epilepsy type Show forest plot

4

683

Hazard Ratio (Fixed, 95% CI)

0.93 [0.72, 1.20]

Analysis 1.4

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 4 Time to 12‐month remission ‐ stratified by epilepsy type.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 4 Time to 12‐month remission ‐ stratified by epilepsy type.

4.1 Generalised onset

3

158

Hazard Ratio (Fixed, 95% CI)

0.64 [0.41, 1.01]

4.2 Partial onset

4

525

Hazard Ratio (Fixed, 95% CI)

1.11 [0.81, 1.51]

5 Time to six‐month remission Show forest plot

4

683

Hazard Ratio (Fixed, 95% CI)

1.02 [0.83, 1.26]

Analysis 1.5

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 5 Time to six‐month remission.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 5 Time to six‐month remission.

6 Time to six‐month remission ‐ stratified by epilepsy type Show forest plot

4

683

Hazard Ratio (Fixed, 95% CI)

0.99 [0.80, 1.23]

Analysis 1.6

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 6 Time to six‐month remission ‐ stratified by epilepsy type.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 6 Time to six‐month remission ‐ stratified by epilepsy type.

6.1 Generalised onset

3

158

Hazard Ratio (Fixed, 95% CI)

0.69 [0.47, 1.01]

6.2 Partial onset

4

525

Hazard Ratio (Fixed, 95% CI)

1.17 [0.90, 1.50]

7 Time to first seizure Show forest plot

6

822

Hazard Ratio (Fixed, 95% CI)

0.86 [0.71, 1.04]

Analysis 1.7

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 7 Time to first seizure.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 7 Time to first seizure.

8 Time to first seizure ‐ stratified by epilepsy type Show forest plot

6

822

Hazard Ratio (Fixed, 95% CI)

0.87 [0.72, 1.06]

Analysis 1.8

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 8 Time to first seizure ‐ stratified by epilepsy type.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 8 Time to first seizure ‐ stratified by epilepsy type.

8.1 Generalised onset

5

238

Hazard Ratio (Fixed, 95% CI)

1.23 [0.86, 1.77]

8.2 Partial onset

6

584

Hazard Ratio (Fixed, 95% CI)

0.76 [0.60, 0.96]

9 Time to first seizure ‐ sensitivity analysis Show forest plot

6

822

Hazard Ratio (Fixed, 95% CI)

0.89 [0.73, 1.09]

Analysis 1.9

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 9 Time to first seizure ‐ sensitivity analysis.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 9 Time to first seizure ‐ sensitivity analysis.

9.1 Generalised onset

5

173

Hazard Ratio (Fixed, 95% CI)

1.39 [0.90, 2.13]

9.2 Partial onset

6

584

Hazard Ratio (Fixed, 95% CI)

0.76 [0.60, 0.96]

9.3 Uncertain seizure type

3

65

Hazard Ratio (Fixed, 95% CI)

1.22 [0.59, 2.51]

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

'Risk of bias' graph: review authors' judgements about each 'Risk of bias' item presented as percentages across all included studies
Figuras y tablas -
Figure 2

'Risk of bias' graph: review authors' judgements about each 'Risk of bias' item presented as percentages across all included studies

'Risk of bias' summary: review authors' judgements about each 'Risk of bias' item for each included study
Figuras y tablas -
Figure 3

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

Time to withdrawal of allocated treatment
Figuras y tablas -
Figure 4

Time to withdrawal of allocated treatment

Time to withdrawal of allocated treatment ‐ stratified by epilepsy type
Figuras y tablas -
Figure 5

Time to withdrawal of allocated treatment ‐ stratified by epilepsy type

Time to 12‐month remission
Figuras y tablas -
Figure 6

Time to 12‐month remission

Time to 12‐month remission ‐ stratified by epilepsy type
Figuras y tablas -
Figure 7

Time to 12‐month remission ‐ stratified by epilepsy type

Time to six‐month remission
Figuras y tablas -
Figure 8

Time to six‐month remission

Time to six‐month remission ‐ stratified by epilepsy type
Figuras y tablas -
Figure 9

Time to six‐month remission ‐ stratified by epilepsy type

Time to first seizure
Figuras y tablas -
Figure 10

Time to first seizure

Time to first seizure ‐ stratified by epilepsy type
Figuras y tablas -
Figure 11

Time to first seizure ‐ stratified by epilepsy type

Time to six‐month remission ‐ Mattson 1985
Figuras y tablas -
Figure 12

Time to six‐month remission ‐ Mattson 1985

Time to first seizure ‐ de Silva 1996
Figuras y tablas -
Figure 13

Time to first seizure ‐ de Silva 1996

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 1 Time to withdrawal of allocated treatment.
Figuras y tablas -
Analysis 1.1

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 1 Time to withdrawal of allocated treatment.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 2 Time to withdrawal of allocated treatment ‐ stratified by epilepsy type.
Figuras y tablas -
Analysis 1.2

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 2 Time to withdrawal of allocated treatment ‐ stratified by epilepsy type.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 3 Time to 12‐month remission.
Figuras y tablas -
Analysis 1.3

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 3 Time to 12‐month remission.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 4 Time to 12‐month remission ‐ stratified by epilepsy type.
Figuras y tablas -
Analysis 1.4

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 4 Time to 12‐month remission ‐ stratified by epilepsy type.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 5 Time to six‐month remission.
Figuras y tablas -
Analysis 1.5

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 5 Time to six‐month remission.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 6 Time to six‐month remission ‐ stratified by epilepsy type.
Figuras y tablas -
Analysis 1.6

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 6 Time to six‐month remission ‐ stratified by epilepsy type.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 7 Time to first seizure.
Figuras y tablas -
Analysis 1.7

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 7 Time to first seizure.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 8 Time to first seizure ‐ stratified by epilepsy type.
Figuras y tablas -
Analysis 1.8

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 8 Time to first seizure ‐ stratified by epilepsy type.

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 9 Time to first seizure ‐ sensitivity analysis.
Figuras y tablas -
Analysis 1.9

Comparison 1 Carbamazepine versus phenobarbitone, Outcome 9 Time to first seizure ‐ sensitivity analysis.

Summary of findings for the main comparison. Summary of findings ‐ Carbamazepine compared with phenobarbitone for epilepsy (primary outcome)

Carbamazepine compared with phenobarbitone for epilepsy

Patient or population: adults and children with newly onset partial or generalised epilepsy

Settings: outpatients

Intervention: carbamazepine

Comparison: phenobarbitone

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)¹

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Phenobarbitone

Carbamazepine

Time to withdrawal of allocated treatment ‐ all participants, stratified by epilepsy type

Range of follow‐up (all participants): 0 to 4653 days

390 per 1000

281 per 1000
(224 to 350)

HR 1.50 (1.15 to 1.95)

676

(4 studies)

⊕⊕⊝⊝
low2,3

HR > 1 indicates a
clinical advantage for
carbamazepine

Time to withdrawal of allocated treatment

Subgroup: generalised onset seizures

Range of follow‐up (all participants): 0 to 4653 days

286 per 1000

197 per 1000
(110 to 340)

HR 1.53 (0.81 to 2.88)

156

(3 studies)

⊕⊕⊝⊝
low2,3

HR > 1 indicates a
clinical advantage for
carbamazepine

Time to withdrawal of allocated treatment

Subgroup: partial onset seizures

Range of follow‐up (all participants): 0 to 4272 days

420 per 1000

307 per 1000
(239 to 385)

HR 1.49 (1.12 to 2.00)

520

(4 studies)

⊕⊕⊝⊝
low2,3

HR > 1 indicates a
clinical advantage for
carbamazepine

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The assumed risk is calculated as the event rate in the phenobarbitone treatment group. The corresponding risk in the carbamazepine treatment group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
The corresponding risk is calculated as the assumed risk x the relative risk of the intervention where relative risk = (1 ‐ exp(HR x ln(1 ‐ assumed risk)) )/assumed risk.
CI: confidence interval; RR: risk ratio; HR: hazard ratio; exp: exponential.

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

1Pooled HR for all participants adjusted for seizure type.
2There was high risk of bias for at least one element of three studies included in the analysis; de Silva 1996 and Heller 1995 were open‐label, and the lack of masking may have influenced the withdrawal rates in the study. Placencia 1993 did not adequately conceal allocation for all participants, which may have influenced the withdrawal rates in the study. There were inconsistencies in Placencia 1993 between published data and IPD, which the authors could not resolve.
3Substantial heterogeneity was present between studies; sensitivity analyses showed that Placencia 1993 contributed the largest amount of variability to analysis.

Figuras y tablas -
Summary of findings for the main comparison. Summary of findings ‐ Carbamazepine compared with phenobarbitone for epilepsy (primary outcome)
Summary of findings 2. Summary of findings ‐ Carbamazepine compared with phenobarbitone for epilepsy (secondary outcome)

Carbamazepine compared with phenobarbitone for epilepsy

Patient or population: adults and children with newly onset partial or generalised epilepsy

Settings: outpatients

Intervention: carbamazepine

Comparison: phenobarbitone

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)¹

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Phenobarbitone

Carbamazepine

Time to achieve 12‐month remission ‐ all participants, stratified by epilepsy type

Range of follow‐up (all participants): 0 to 4222 days

367 per 1000

346 per 1000
(280 to 422)

HR 0.93

(0.72 to 1.20)

683
(4 studies)

⊕⊕⊝⊝
low2,3

HR > 1 indicates a
clinical advantage for
phenobarbitone

Time to achieve 12‐month remission

Subgroup: generalised onset seizures

Range of follow‐up (all participants): 0 to 4163 days

500 per 1000

358 per 1000
(247 to 503)

HR 0.64

(0.41 to 1.01)

158
(3 studies)

⊕⊕⊝⊝
low2,3

HR > 1 indicates a
clinical advantage for
phenobarbitone

Time to achieve 12‐month remission

Subgroup: partial onset seizures

Range of follow‐up (all participants): 0 to 4222 days

329 per 1000

358 per 1000
(276 to 453)

HR 1.11

(0.81 to 1.51)

525
(4 studies)

⊕⊕⊝⊝
low2,3

HR > 1 indicates a
clinical advantage for
phenobarbitone

Time to first seizure ‐ all participants, stratified by epilepsy type

Range of follow‐up (all participants): 0 to 4108 days

487 per 1000

536 per 1000
(467 to 604)

HR 0.87

(0.72 to 1.06)

822

(6 studies)

⊕⊕⊝⊝
low4,5,6

HR > 1 indicates a
clinical advantage for
carbamazepine

Time to first seizure ‐ Subgroup: generalised onset seizures

Range of follow‐up (all participants): 0 to 4108 days

548 per 1000

475 per 1000
(361 to 602)

HR 1.23

(0.86 to 1.77)

238

(5 studies)

⊕⊕⊝⊝
low4,5,6

HR > 1 indicates a
clinical advantage for
carbamazepine

Time to first seizure ‐ Subgroup: partial onset seizures

Range of follow‐up (all participants): 0 to 4108 days

462 per 1000

557 per 1000
(475 to 644)

HR 0.76

(0.60 to 0.96)

584

(6 studies)

⊕⊕⊝⊝
low4,5,6

HR > 1 indicates a
clinical advantage for
carbamazepine

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The assumed risk is calculated as the event rate in the phenobarbitone treatment group. The corresponding risk in the carbamazepine treatment group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
The corresponding risk is calculated as the assumed risk x the relative risk of the intervention where relative risk = (1 ‐ exp(HR x ln(1 ‐ assumed risk)) )/assumed risk.
CI: confidence interval; RR: risk ratio; HR: hazard ratio; exp: exponential.

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

1Pooled HR for all participants adjusted for seizure type.
2There was high risk of bias for at least one element of three studies included in the analysis; de Silva 1996 and Heller 1995 were open‐label, and the lack of masking may have influenced the withdrawal rates in the study. Placencia 1993 did not adequately conceal allocation for all participants, which may have influenced the withdrawal rates in the study and therefore the remission rates in the study.
3Substantial heterogeneity was present between studies; sensitivity analyses showed that Placencia 1993 contributed the largest amount of variability to the analysis.
4There was high risk of bias for at least one element of four studies included in the analysis; de Silva 1996 and Heller 1995 were open‐label, and the lack of masking may have influenced the withdrawal rates in the study. Placencia 1993 was not adequately concealed for all participants, which may have influenced the withdrawal rates in the study and therefore the seizure recurrence rates in the trial. There were inconsistencies between published data and IPD, which the authors could not resolve in Banu 2007.
5Substantial heterogeneity was present between studies; sensitivity analyses showed that Placencia 1993 and Ogunrin 2005 contributed the largest amount of variability to the analysis.
6Misclassification of seizure type in Ogunrin 2005 for 19 individuals may have impacted on the trial result. Sensitivity analysis to adjust for misclassification reduced the amount of heterogeneity in the analysis.

Figuras y tablas -
Summary of findings 2. Summary of findings ‐ Carbamazepine compared with phenobarbitone for epilepsy (secondary outcome)
Table 1. Outcomes considered and summary of results for trials with no IPD

Trial

Outcomes reported

Summary of results

Bidabadi 2009

  1. Proportion seizure‐free

  2. Response rate

  3. Rate of side‐effects

  4. Mean seizure frequency per month

  5. Mean seizure duration

  1. CBZ: 23/36 (64%), PB: 22/35 (63%)

  2. No statistically significant difference between groups

  3. No statistically significant difference between groups

  4. CBZ: 0.66, PB: 0.8

  5. CBZ: 12.63 seconds, PB: 15 seconds

Cereghino 1974

  1. Behaviour measured with rating scale modified from the Ward Behavior Rating Scale

  2. Seizure control

  3. Side‐effects

  4. Withdrawals

  1. No change or improvement in behaviour was more common on PB than CBZ (40% versus 12%); predominant improvement with some deterioration was more common on CBZ than PB (36% versus 12%)

  2. No difference between PB and CBZ in terms of seizure control

  3. Gastrointestinal and "impaired function" side‐effects were more common on CBZ than PB in the first few study days. Side‐effects of both drugs were minimal in later stages of the study

  4. PB: 26/44 (59%), CBZ: 27/45 (60%)

Chen 1996

  1. IQ scores measured with WISC‐R scale

  2. Time to complete the Bender‐Gestalt test

  3. Auditory event‐related potentials

  4. Incidence of allergic reactions

  5. Seizure control

  1. No significant difference between groups

  2. No significant difference between groups

  3. No significant difference between groups

  4. 2 children from PB group and 1 child from CBZ group withdrew from the study because of allergic reactions

  5. No significant difference between groups

Cossu 1984

Changes in memory function from baseline after 3 weeks of treatment (verbal, visual, (visual‐verbal and visual‐non‐verbal), acoustic, tactile, and spatial)

  1. Significant decrease in visual‐verbal memory for CBZ and acoustic memory for PB

  2. No significant differences for other tests

Czapinski 1997

  1. Proportion achieving 24‐month remission at 3 years

  2. Proportion excluded after randomisation due to adverse effects or no efficacy

  1. PB: 60%, CBZ: 62%

  2. PB: 33%, CBZ: 30%

Feksi 1991

  1. Adverse effects

  2. Withdrawals from allocated treatment

  3. Seizure frequency (during second 6 months of study, participants completing the study only)

PB (n = 123), CBZ (n = 126)

  1. Minor adverse effects reported in PB: 58 participants (39%) reported 86 adverse events, CBZ: 46 participants (30%) reported 68 adverse events

  2. PB: all withdrawals: PB: 27 (18%), CBZ: 26 (17%); withdrawals due to side‐effects: PB: 8 (5%), CBZ: 5 (3%)

  3. Seizure‐free: PB: 67 (54%), CBZ: 65 (52%); > 50% reduction of seizures from baseline: PB: 28 (23%), CBZ: 37 (29%); between 50% reduction to 50% increase of seizures: PB: 18 (15%), CBZ: 17 (13%); > 50% increase in seizures: PB: 10 (8%), CBZ: 7 (6%)

Mitchell 1987

  1. Cognitive/behavioural outcomes at 1, 2, 6, and 12 months

  2. Compliance, drug changes, and withdrawal rates

  3. Seizure control at 6 and 12 months (excellent/good/fair/poor)

  1. No significant differences between treatment groups (children from pilot study included for 6 and 12 months)

  2. Compliance (children from pilot study included): trend towards better compliance in CBZ group (not significant)

    • Randomised participants only: trend towards higher rate withdrawal from treatment in PB group (not significant). More mild systemic side‐effects in CBZ group (significant). 3 children switched from CBZ to PB and 1 from PB to CB following adverse reactions

  3. Seizure control at 6 months: excellent/good: PB = 15, CBZ = 13 (children from pilot study included) fair/poor PB = 5, CBZ = 3; seizure control at 12 months: excellent/good: PB = 13, CBZ = 9 (children from pilot study included) fair/poor PB = 4, CBZ = 4

CBZ: carbamazepine

IQ: intelligence quotient
PB: phenobarbitone

WISC‐R scale: the Wechsler Intelligence Scale for Children

Figuras y tablas -
Table 1. Outcomes considered and summary of results for trials with no IPD
Table 2. Number of participants contributing to each analysis

Trial

Number randomised

Time to withdrawal of

allocated treatment

Time to 12‐month

remission

Time to six‐month

remission

Time to first seizure

CBZ

PB

Total

CBZ

PB

Total

CBZ

PB

Total

CBZ

PB

Total

CBZ

PB

Total

Banu 2007¹

54

54

108

Information not available

Information not available

Information not available

54

54

108

de Silva 1996²

54

10

64

53

10

63

54

10

64

54

10

64

54

10

64

Heller 1995³

61

58

119

60

55

115

61

58

119

61

58

119

61

58

119

Mattson 1985

155

155

310

154

155

309

154

155

309

154

155

309

151

151

302

Ogunrin 2005

19

18

37

Information not available

Information not available

Information not available

19

18

37

Placencia 1993

95

97

192

94

95

189

95

96

191

95

96

191

95

97

192

Total

438

392

830

361

315

676

364

319

683

364

319

683

434

388

822

CBZ: carbamazepine
PB: phenobarbitone
¹The date of withdrawal of allocated treatment was not recorded in all cases for Banu 2007, so we could not calculate 'time to withdrawal of allocated treatment'. The date of first seizure after randomisation was recorded, but all dates of subsequent seizures were not recorded; therefore, we could calculate 'time to first seizure', but we could not calculate 'time to six‐month remission' and 'time to 12‐month remission'.
²We received IPD for 70 participants recruited in de Silva 1996; the randomised drug was not recorded in six participants. Reasons for treatment withdrawal were not available for one participant randomised to CBZ; we did not include this participant in the analysis of time to treatment withdrawal.
³Reasons for treatment withdrawal were not available for four participants (one randomised to CBZ and three to PB) in Heller 1995; we did not include these participants in the analysis of time to treatment withdrawal.
⁴No follow‐up data after randomisation were available for one participant randomised to CBZ in Mattson 1985. Dates of seizure recurrence were not available for seven participants (three randomised to CBZ and four to PB); we did not include these participants in the analysis of time to first seizure.
⁵The study duration of Ogunrin 2005 was 12 weeks; therefore, six‐ and 12‐month remission of seizures could not be achieved, so we could not calculate these outcomes. All randomised participants completed the study without withdrawing from treatment, so we could not analyse the time to treatment withdrawal.
⁶Reasons for treatment withdrawal were not available for three participants (one randomised to CBZ and two randomised to PB) in Placencia 1993. We did not include these participants in the analysis of time to treatment withdrawal. Seizure data after occurrence of first seizure were not available for one participant randomised to PB, so we did not include this participant in the analyses of time to six‐month and time to 12‐month remission.

Figuras y tablas -
Table 2. Number of participants contributing to each analysis
Table 3. Reasons for premature discontinuation (withdrawal of allocated treatment)

Reason for early termination

Classification

de Silva 1996 ¹

Heller 1995 ¹

Mattson 1985

Placencia 1993 ²

Banu 2007 ³

Total⁴

CBZ n = 53

PB = 10

CBZ n = 60

PB = 55

CBZ n = 154

PB = 155

CBZ = 94

PB = 95

CBZ = 54

PB = 54

CBZ = 415

PB = 369

Adverse events

Event

3

2

8

12

11

5

5

5

0

0

27

24

Seizure recurrence

Event

12

2

5

7

3

7

0

0

1

2

21

18

Both seizure recurrence and adverse events

Event

6

4

4

3

30

26

0

0

0

0

40

33

Non‐compliance/participant choice

Event

0

0

0

0

11

19

13

9

6

0

30

28

Another AED added/AED changed

Event

0

0

0

0

0

3

0

0

7

4

7

7

Participant went into remission

Censored

18

1

6

3

0

0

0

0

0

2

24

6

Lost to follow‐up

Censored

0

0

0

0

26

26

11

5

7

15

44

46

Death⁵

Censored

0

0

0

0

4

2

2

1

0

0

6

3

Other⁶

Censored

0

0

0

0

16

13

0

0

0

0

16

13

Completed the study (did not withdraw)

Censored

14

1

37

30

53

54

63

75

33

31

200

191

AED: antiepileptic drug
CBZ: carbamazepine
n: number of individuals contributing to the outcome 'time to treatment withdrawal'
PB: phenobarbitone
¹Four participants for Heller 1995 (one on CBZ and three on PB) and one for de Silva 1996 (CBZ) had missing reasons for treatment withdrawal.
²There were inconsistencies between IPD and the publication of Placencia 1993; we performed sensitivity analysis (see Effects of interventions). There were missing reasons for treatment withdrawal for three participants (one on CBZ and two on PB); we did not include these participants in the analysis.
³Banu 2007 provided reasons for treatment withdrawal, but dates of treatment withdrawal could not be provided for all participants, so we could not calculate 'time to withdrawal of allocated treatment'.
⁴All participants in Ogunrin 2005 completed the study without withdrawing; therefore, this study did not contribute to 'time to withdrawal of allocated treatment'.
⁵Death was due to reasons not related to the study drug.
⁶Other reasons from Mattson 1985: participants developed other medical disorders including neurological and psychiatric disorders.

Figuras y tablas -
Table 3. Reasons for premature discontinuation (withdrawal of allocated treatment)
Table 4. Sensitivity analyses

Analysis

Time to withdrawal of

allocated treatment

Time to 12‐month

remission

Time to six‐month

remission

Time to first seizure¹

Original analysis

Participants

676 (Analysis 1.2)

683 (Analysis 1.4)

683 (Analysis 1.6)

822 (Analysis 1.8)

Pooled HR (95% CI)

P value

1.50 (1.15 to 1.95)

P = 0.003

0.93 (0.72 to 1.20)

P = 0.57

0.99 (0.80 to 1.23)

P = 0.95

0.87 (0.72 to 1.06)

P = 0.18

Heterogeneity

I² statistic = 35%

I² statistic = 55%

I² statistic = 58%

I² statistic = 44%

Sensitivity analysis

for Placencia 1993²

Participants

487

492

492

630

Pooled HR (95% CI)

P value

1.66 (1.25 to 2.20)

P = 0.0005

0.82 (0.61 to 1.09)

P = 0.15

0.88 (0.68 to 1.14)

P = 0.34

0.87 (0.71 to 1.08)

P = 0.22

Heterogeneity

I² statistic = 35%

I² statistic = 0%

I² statistic = 0%

I² statistic = 34%

Sensitivity analysis

for de Silva 1996³

Participants

633

640

640

779

Pooled HR (95% CI)

P value

1.42 (1.08 to 1.86)

P = 0.01

0.90 (0.69 to 1.17)

P = 0.42

0.97 (0.78 to 1.21)

P = 0.79

0.87 (0.71 to 1.06)

P = 0.17

Heterogeneity

I² statistic = 0%

I² statistic = 57%

I² statistic = 60%

I² statistic = 39%

CI: confidence interval
HR: hazard ratio
¹We performed sensitivity analyses for potential misclassification of seizure type (see Analysis 1.9) and because of inconsistencies between published data and IPD for Banu 2007 (see Sensitivity analysis and Effects of interventions for full details).
²We performed sensitivity analysis excluding all randomised participants in Placencia 1993 because of inadequate allocation concealment in the study. We performed further sensitivity analysis for the outcome 'time to withdrawal of allocation concealment' because of inconsistencies between published data and IPD for Placencia 1993 (see Sensitivity analysis and Effects of interventions for full details).
³We performed sensitivity analysis including only the participants in de Silva 1996, which were randomised before the phenobarbitone arm was withdrawn (see Sensitivity analysis and Effects of interventions for full details).

Figuras y tablas -
Table 4. Sensitivity analyses
Table 5. Adverse event data (narrative report)

Trial

Adverse event data¹

Summary of reported results

Carbamazepine (CBZ)

Phenobarbitone (PB)

Banu 2007²

Reported list of 'problems' at the last visit (provided as IPD)

CBZ (n = 54): speech/learning delay (n = 6), headaches (n = 3), restlessness/hyperactivity/poor attention/irritability (n = 6), psychomotor deterioration/delay (n = 2), sleep disturbances (n = 2), fatigue (n = 1), hydrocephalus (build up of fluid on the brain) (n = 1), CBZ hypersensitivity (n = 1), aggression (n = 1), temper tantrums (n = 1), other behavioural problems (n = 5), poor cognition (n = 1), mild stroke (n = 1), mild right‐sided weakness (n = 1), intolerable behavioural problems (n = 6)

PB (n = 54): speech/learning delay (n = 7), restlessness/hyperactivity/poor attention/irritability (n = 8), sleep disturbances (n = 1), fatigue (n = 1), poor cognition (n = 2), aggression (n = 1), temper tantrums (n = 3), breath‐holding attacks (n = 1), other behavioural problems (n = 3), facial twitching (n = 1), left‐sided weakness (n = 1), leg pain (n = 1), vomiting (n = 1), intolerable behavioural problems (n = 4)

Bidabadi 2009³

Rate of drug side‐effects

No statistical significant difference was seen after treatment between 2 groups in the rate of drug side‐effects

No statistical significant difference was seen after treatment between 2 groups in the rate of drug side‐effects

Cereghino 1974²,

Most frequently observed side‐effects

Gastrointestinal side‐effects and "impaired function" (general malaise). Frequency not clearly stated

Gastrointestinal side‐effects and "impaired function" (general malaise). Frequency not clearly stated

Chen 1996

Withdrawal from the study due to 'allergic reactions'

CBZ (n = 24): 1 participant withdrew due to an allergic reaction

PB (n = 23): 2 participants withdrew due to allergic reactions

Cossu 1984

No adverse events reported

Not reported

Not reported

Czapinski 1997³

"Exclusions due to adverse events or no efficacy"

Proportion "excluded": 30% (out of 30 randomised to CBZ)

Proportion "excluded": 33.3% (out of 30 randomised to PB)

de Silva 1996,

"Unacceptable" adverse events leading to drug withdrawal

CBZ (n = 54): drowsiness (n = 1), blood dyscrasia (n = 1)

PB (n = 10): drowsiness (n = 1), behavioural (n = 5)

Feksi 1991

Reports of minor adverse events and side‐effects leading to drug withdrawal

CBZ (n = 150): withdrawals due to side‐effects: skin rash (n = 4), psychosis (n = 1), aggressive behaviour (n = 1).

Minor adverse events: CBZ: 46 participants reported 68 adverse events

PB (n = 152): withdrawals due to side‐effects: skin rash (n = 1), psychosis (n = 1), hyperactivity (n = 3).

Minor adverse events: 58 participants reported 86 adverse events

Heller 1995

"Unacceptable" adverse events

leading to drug withdrawal

CBZ (n = 61): drowsiness (n = 3), rash (n = 2), headache (n = 1), depression (n = 1)

PB (n = 58): drowsiness (n = 4), lethargy (n = 4), rash (n = 1), dizziness (n = 2), headaches (n = 1), nausea and vomiting (n = 1)

Mattson 1985²

Narrative report of 'adverse effects' and 'serious side‐effects'

CBZ (n = 155): motor disturbance (ataxia, incoordination, nystagmus, tremor ‐ 33%), dysmorphic and idiosyncratic side‐effects (gum hypertrophy, hirsutism, acne, and rash ‐ 14%), gastrointestinal problems (27%), decreased libido or impotence (13%). No serious side‐effects

PB (n = 155): motor disturbance (ataxia, incoordination, nystagmus, tremor ‐ 24%), dysmorphic and idiosyncratic side‐effects (gum hypertrophy, hirsutism, acne, and rash ‐11 %), gastrointestinal problems (13%), decreased libido or impotence (16%). No serious side‐effects

Mitchell 1987

Systemic side‐effects and side‐effects leading to drug change

CBZ (n = 15): 4 participants switched from CBZ to PB; 3 due to systemic side‐effects (1 with persistent rashes and 1 with marked granulocytopenia (decrease of granulocytes (white blood cells)) and 1 due to behavioural changes

PB (n = 18): 1 participant switched from PB to CBZ due to substantial behavioural side‐effects

Ogunrin 2005²

Participant‐reported symptomatic complaints (provided as IPD)

CBZ (n = 19), memory impairment (n = 9), psychomotor retardation (n = 1), inattention (n = 1), transient rash (n = 1), CBZ‐induced cough (n = 1)

PB (n = 18), memory impairment (n = 13), psychomotor retardation (n = 8), inattention (n = 9)

Placencia 1993

Number of participants reporting side‐effects

CBZ (n = 95): 53 participants reported at least 1 side‐effect

PB (n = 97): 50 participants reported at least 1 side‐effect

CBZ: carbamazepine; PB: phenobarbitone
¹We recorded adverse event data as reported narratively in the publications; therefore, exact definition of a symptom may vary. Adverse event data were supplied as IPD for Banu 2007 and Ogunrin 2005. Adverse event data were not requested in original IPD requests (de Silva 1996; Heller 1995; Mattson 1985; Placencia 1993), but will be for all future IPD requests. For numbers of withdrawals due to adverse events in studies for which we received IPD (Banu 2007; de Silva 1996; Heller 1995; Mattson 1985; Placencia 1993), see Table 3.
²Bidabadi 2009 and Czapinski 1997 are abstracts only so very little information was reported.
³Participants may report more than one adverse event.
⁴Note that the recruited participants in this study were institutionalised; therefore, the "precise nature of side‐effects was not always determinable". The two most frequently occurring side‐effects were reported as the frequency of participants reporting the side‐effect on each day of the treatment period; however, overall totals of participants reporting each side‐effect were not reported.
⁵Participants may have withdrawn due to adverse event alone or a combination of adverse events and poor efficacy (seizures).
⁶The phenobarbitone arm of de Silva 1996 was stopped prematurely after 10 children were randomised to this arm because of concerns over behavioural adverse events (see the 'Characteristics of included studies' tables).

Figuras y tablas -
Table 5. Adverse event data (narrative report)
Comparison 1. Carbamazepine versus phenobarbitone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Time to withdrawal of allocated treatment Show forest plot

4

676

Hazard Ratio (Fixed, 95% CI)

1.49 [1.15, 1.94]

2 Time to withdrawal of allocated treatment ‐ stratified by epilepsy type Show forest plot

4

676

Hazard Ratio (Fixed, 95% CI)

1.50 [1.15, 1.95]

2.1 Generalised onset

3

156

Hazard Ratio (Fixed, 95% CI)

1.53 [0.81, 2.88]

2.2 Partial onset

4

520

Hazard Ratio (Fixed, 95% CI)

1.49 [1.12, 2.00]

3 Time to 12‐month remission Show forest plot

4

683

Hazard Ratio (Fixed, 95% CI)

0.93 [0.72, 1.19]

4 Time to 12‐month remission ‐ stratified by epilepsy type Show forest plot

4

683

Hazard Ratio (Fixed, 95% CI)

0.93 [0.72, 1.20]

4.1 Generalised onset

3

158

Hazard Ratio (Fixed, 95% CI)

0.64 [0.41, 1.01]

4.2 Partial onset

4

525

Hazard Ratio (Fixed, 95% CI)

1.11 [0.81, 1.51]

5 Time to six‐month remission Show forest plot

4

683

Hazard Ratio (Fixed, 95% CI)

1.02 [0.83, 1.26]

6 Time to six‐month remission ‐ stratified by epilepsy type Show forest plot

4

683

Hazard Ratio (Fixed, 95% CI)

0.99 [0.80, 1.23]

6.1 Generalised onset

3

158

Hazard Ratio (Fixed, 95% CI)

0.69 [0.47, 1.01]

6.2 Partial onset

4

525

Hazard Ratio (Fixed, 95% CI)

1.17 [0.90, 1.50]

7 Time to first seizure Show forest plot

6

822

Hazard Ratio (Fixed, 95% CI)

0.86 [0.71, 1.04]

8 Time to first seizure ‐ stratified by epilepsy type Show forest plot

6

822

Hazard Ratio (Fixed, 95% CI)

0.87 [0.72, 1.06]

8.1 Generalised onset

5

238

Hazard Ratio (Fixed, 95% CI)

1.23 [0.86, 1.77]

8.2 Partial onset

6

584

Hazard Ratio (Fixed, 95% CI)

0.76 [0.60, 0.96]

9 Time to first seizure ‐ sensitivity analysis Show forest plot

6

822

Hazard Ratio (Fixed, 95% CI)

0.89 [0.73, 1.09]

9.1 Generalised onset

5

173

Hazard Ratio (Fixed, 95% CI)

1.39 [0.90, 2.13]

9.2 Partial onset

6

584

Hazard Ratio (Fixed, 95% CI)

0.76 [0.60, 0.96]

9.3 Uncertain seizure type

3

65

Hazard Ratio (Fixed, 95% CI)

1.22 [0.59, 2.51]

Figuras y tablas -
Comparison 1. Carbamazepine versus phenobarbitone